1
|
Nicoletti A, Paratore M, Vitale F, Negri M, Quero G, Esposto G, Mignini I, Alfieri S, Gasbarrini A, Zocco MA, Zileri Dal Verme L. Understanding the Conundrum of Pancreatic Cancer in the Omics Sciences Era. Int J Mol Sci 2024; 25:7623. [PMID: 39062863 PMCID: PMC11276793 DOI: 10.3390/ijms25147623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Pancreatic cancer (PC) is an increasing cause of cancer-related death, with a dismal prognosis caused by its aggressive biology, the lack of clinical symptoms in the early phases of the disease, and the inefficacy of treatments. PC is characterized by a complex tumor microenvironment. The interaction of its cellular components plays a crucial role in tumor development and progression, contributing to the alteration of metabolism and cellular hyperproliferation, as well as to metastatic evolution and abnormal tumor-associated immunity. Furthermore, in response to intrinsic oncogenic alterations and the influence of the tumor microenvironment, cancer cells undergo a complex oncogene-directed metabolic reprogramming that includes changes in glucose utilization, lipid and amino acid metabolism, redox balance, and activation of recycling and scavenging pathways. The advent of omics sciences is revolutionizing the comprehension of the pathogenetic conundrum of pancreatic carcinogenesis. In particular, metabolomics and genomics has led to a more precise classification of PC into subtypes that show different biological behaviors and responses to treatments. The identification of molecular targets through the pharmacogenomic approach may help to personalize treatments. Novel specific biomarkers have been discovered using proteomics and metabolomics analyses. Radiomics allows for an earlier diagnosis through the computational analysis of imaging. However, the complexity, high expertise required, and costs of the omics approach are the main limitations for its use in clinical practice at present. In addition, the studies of extracellular vesicles (EVs), the use of organoids, the understanding of host-microbiota interactions, and more recently the advent of artificial intelligence are helping to make further steps towards precision and personalized medicine. This present review summarizes the main evidence for the application of omics sciences to the study of PC and the identification of future perspectives.
Collapse
Affiliation(s)
- Alberto Nicoletti
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Mattia Paratore
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Federica Vitale
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Marcantonio Negri
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Giuseppe Quero
- Centro Pancreas, Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (S.A.)
| | - Giorgio Esposto
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Irene Mignini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Sergio Alfieri
- Centro Pancreas, Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (S.A.)
| | - Antonio Gasbarrini
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Maria Assunta Zocco
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| | - Lorenzo Zileri Dal Verme
- CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (M.P.); (F.V.); (M.N.); (G.E.); (I.M.); (A.G.); (L.Z.D.V.)
| |
Collapse
|
2
|
Mokhtari A, Casale R, Salahuddin Z, Paquier Z, Guiot T, Woodruff HC, Lambin P, Van Laethem JL, Hendlisz A, Bali MA. Development of Clinical Radiomics-Based Models to Predict Survival Outcome in Pancreatic Ductal Adenocarcinoma: A Multicenter Retrospective Study. Diagnostics (Basel) 2024; 14:712. [PMID: 38611625 PMCID: PMC11011556 DOI: 10.3390/diagnostics14070712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
PURPOSE This multicenter retrospective study aims to identify reliable clinical and radiomic features to build machine learning models that predict progression-free survival (PFS) and overall survival (OS) in pancreatic ductal adenocarcinoma (PDAC) patients. METHODS Between 2010 and 2020 pre-treatment contrast-enhanced CT scans of 287 pathology-confirmed PDAC patients from two sites of the Hopital Universitaire de Bruxelles (HUB) and from 47 hospitals within the HUB network were retrospectively analysed. Demographic, clinical, and survival data were also collected. Gross tumour volume (GTV) and non-tumoral pancreas (RPV) were semi-manually segmented and radiomics features were extracted. Patients from two HUB sites comprised the training dataset, while those from the remaining 47 hospitals of the HUB network constituted the testing dataset. A three-step method was used for feature selection. Based on the GradientBoostingSurvivalAnalysis classifier, different machine learning models were trained and tested to predict OS and PFS. Model performances were assessed using the C-index and Kaplan-Meier curves. SHAP analysis was applied to allow for post hoc interpretability. RESULTS A total of 107 radiomics features were extracted from each of the GTV and RPV. Fourteen subgroups of features were selected: clinical, GTV, RPV, clinical & GTV, clinical & GTV & RPV, GTV-volume and RPV-volume both for OS and PFS. Subsequently, 14 Gradient Boosting Survival Analysis models were trained and tested. In the testing dataset, the clinical & GTV model demonstrated the highest performance for OS (C-index: 0.72) among all other models, while for PFS, the clinical model exhibited a superior performance (C-index: 0.70). CONCLUSIONS An integrated approach, combining clinical and radiomics features, excels in predicting OS, whereas clinical features demonstrate strong performance in PFS prediction.
Collapse
Affiliation(s)
- Ayoub Mokhtari
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Roberto Casale
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Zohaib Salahuddin
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
| | - Zelda Paquier
- Medical Physics Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Thomas Guiot
- Medical Physics Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Henry C. Woodruff
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Centre+, 6229HX Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Centre+, 6229HX Maastricht, The Netherlands
| | - Jean-Luc Van Laethem
- Department of Gastroenterology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Alain Hendlisz
- Department of Gastroenterology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Maria Antonietta Bali
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
3
|
Litjens G, Broekmans JPEA, Boers T, Caballo M, van den Hurk MHF, Ozdemir D, van Schaik CJ, Janse MHA, van Geenen EJM, van Laarhoven CJHM, Prokop M, de With PHN, van der Sommen F, Hermans JJ. Computed Tomography-Based Radiomics Using Tumor and Vessel Features to Assess Resectability in Cancer of the Pancreatic Head. Diagnostics (Basel) 2023; 13:3198. [PMID: 37892019 PMCID: PMC10606005 DOI: 10.3390/diagnostics13203198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The preoperative prediction of resectability pancreatic ductal adenocarcinoma (PDAC) is challenging. This retrospective single-center study examined tumor and vessel radiomics to predict the resectability of PDAC in chemo-naïve patients. The tumor and adjacent arteries and veins were segmented in the portal-venous phase of contrast-enhanced CT scans, and radiomic features were extracted. Features were selected via stability and collinearity testing, and least absolute shrinkage and selection operator application (LASSO). Three models, using tumor features, vessel features, and a combination of both, were trained with the training set (N = 86) to predict resectability. The results were validated with the test set (N = 15) and compared to the multidisciplinary team's (MDT) performance. The vessel-features-only model performed best, with an AUC of 0.92 and sensitivity and specificity of 97% and 73%, respectively. Test set validation showed a sensitivity and specificity of 100% and 88%, respectively. The combined model was as good as the vessel model (AUC = 0.91), whereas the tumor model showed poor performance (AUC = 0.76). The MDT's prediction reached a sensitivity and specificity of 97% and 84% for the training set and 88% and 100% for the test set, respectively. Our clinician-independent vessel-based radiomics model can aid in predicting resectability and shows performance comparable to that of the MDT. With these encouraging results, improved, automated, and generalizable models can be developed that reduce workload and can be applied in non-expert hospitals.
Collapse
Affiliation(s)
- Geke Litjens
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Joris P. E. A. Broekmans
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Tim Boers
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Marco Caballo
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Maud H. F. van den Hurk
- Department of Plastic and Reconstructive Surgery, Saint Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
| | - Dilek Ozdemir
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Caroline J. van Schaik
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Markus H. A. Janse
- Image Sciences Institute, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Erwin J. M. van Geenen
- Department of Gastroenterology and Hepatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Cees J. H. M. van Laarhoven
- Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Mathias Prokop
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Peter H. N. de With
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - John J. Hermans
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
4
|
Ahmed TM, Kawamoto S, Hruban RH, Fishman EK, Soyer P, Chu LC. A primer on artificial intelligence in pancreatic imaging. Diagn Interv Imaging 2023; 104:435-447. [PMID: 36967355 DOI: 10.1016/j.diii.2023.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
Artificial Intelligence (AI) is set to transform medical imaging by leveraging the vast data contained in medical images. Deep learning and radiomics are the two main AI methods currently being applied within radiology. Deep learning uses a layered set of self-correcting algorithms to develop a mathematical model that best fits the data. Radiomics converts imaging data into mineable features such as signal intensity, shape, texture, and higher-order features. Both methods have the potential to improve disease detection, characterization, and prognostication. This article reviews the current status of artificial intelligence in pancreatic imaging and critically appraises the quality of existing evidence using the radiomics quality score.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Research Center, Department of Pathology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, Department of Radiology, Hôpital Cochin-APHP, 75014, 75006, Paris, France, 7501475006
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
5
|
Li J, Du J, Li Y, Meng M, Hang J, Shi H. A nomogram based on CT texture features to predict the response of patients with advanced pancreatic cancer treated with chemotherapy. BMC Gastroenterol 2023; 23:274. [PMID: 37563572 PMCID: PMC10416463 DOI: 10.1186/s12876-023-02902-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the predictive value of computed tomography (CT) texture features in the treatment response of patients with advanced pancreatic cancer (APC) receiving palliative chemotherapy. METHODS This study enrolled 84 patients with APC treated with first-line chemotherapy and conducted texture analysis on primary pancreatic tumors. 59 patients and 25 were randomly assigned to the training and validation cohorts at a ratio of 7:3. The treatment response to chemotherapy was evaluated according to the Response Evaluation Criteria in Solid Tumors (RECIST1.1). The patients were divided into progressive and non-progressive groups. The least absolute shrinkage selection operator (LASSO) was applied for feature selection in the training cohort and a radiomics signature (RS) was calculated. A nomogram was developed based on a multivariate logistic regression model incorporating the RS and carbohydrate antigen 19-9 (CA19-9), and was internally validated using the C-index and calibration plot. We performed the decision curve analysis (DCA) and clinical impact curve analysis to reflect the clinical utility of the nomogram. The nomogram was further externally confirmed in the validation cohort. RESULTS The multivariate logistic regression analysis indicated that the RS and CA19-9 were independent predictors (P < 0.05), and a trend was found for chemotherapy between progressive and non-progressive groups. The nomogram incorporating RS, CA19-9 and chemotherapy showed favorable discriminative ability in the training (C-index = 0.802) and validation (C-index = 0.920) cohorts. The nomogram demonstrated favorable clinical utility. CONCLUSION The RS of significant texture features was significantly associated with the early treatment effect of patients with APC treated with chemotherapy. Based on the RS, CA19-9 and chemotherapy, the nomogram provided a promising way to predict chemotherapeutic effects for APC patients.
Collapse
Affiliation(s)
- Jingjing Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Jiadi Du
- Department of Computer Science, Missouri University of Science and Technology, Rolla, MO, U.S
| | - Yuying Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Mingzhu Meng
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Junjie Hang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 518116, Shenzhen, China.
- Department of Oncology, the Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, China.
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China.
| |
Collapse
|
6
|
Amaral MJ, Oliveira RC, Donato P, Tralhão JG. Pancreatic Cancer Biomarkers: Oncogenic Mutations, Tissue and Liquid Biopsies, and Radiomics-A Review. Dig Dis Sci 2023:10.1007/s10620-023-07904-6. [PMID: 36988759 DOI: 10.1007/s10620-023-07904-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/24/2023] [Indexed: 03/30/2023]
Abstract
Pancreatic cancer is one of the most fatal malignancies, as approximately 80% of patients are at advanced stages by the time of diagnosis. The main reason for the poor overall survival is late diagnosis that is partially due to the lack of tools for early-stage detection. In addition, there are several challenges in evaluating response to treatment and predicting prognosis. In this article, we do a review of the most common pancreatic cancer biomarkers with emphasis in new and promising approaches. Liquid biopsies seem to have important clinical applications in early detection, screening, prognosis, and longitudinal monitoring of on-treatment patients. Together with biomarkers in imaging, can represent valuable alternative non-invasive tools in order to achieve a more effective management of pancreatic cancer patients.
Collapse
Affiliation(s)
- Maria João Amaral
- General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Praceta Mota Pinto, 3000-075, Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Rui Caetano Oliveira
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paulo Donato
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Radiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - José Guilherme Tralhão
- General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Praceta Mota Pinto, 3000-075, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Biophysics Institute, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
7
|
Liao J, Li X, Gan Y, Han S, Rong P, Wang W, Li W, Zhou L. Artificial intelligence assists precision medicine in cancer treatment. Front Oncol 2023; 12:998222. [PMID: 36686757 PMCID: PMC9846804 DOI: 10.3389/fonc.2022.998222] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/22/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer is a major medical problem worldwide. Due to its high heterogeneity, the use of the same drugs or surgical methods in patients with the same tumor may have different curative effects, leading to the need for more accurate treatment methods for tumors and personalized treatments for patients. The precise treatment of tumors is essential, which renders obtaining an in-depth understanding of the changes that tumors undergo urgent, including changes in their genes, proteins and cancer cell phenotypes, in order to develop targeted treatment strategies for patients. Artificial intelligence (AI) based on big data can extract the hidden patterns, important information, and corresponding knowledge behind the enormous amount of data. For example, the ML and deep learning of subsets of AI can be used to mine the deep-level information in genomics, transcriptomics, proteomics, radiomics, digital pathological images, and other data, which can make clinicians synthetically and comprehensively understand tumors. In addition, AI can find new biomarkers from data to assist tumor screening, detection, diagnosis, treatment and prognosis prediction, so as to providing the best treatment for individual patients and improving their clinical outcomes.
Collapse
Affiliation(s)
- Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Zhou
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, The Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Shao C, Zhang J, Guo J, Zhang L, Zhang Y, Ma L, Gong C, Tian Y, Chen J, Yu N. A radiomics nomogram model for predicting prognosis of pancreatic ductal adenocarcinoma after high-intensity focused ultrasound surgery. Int J Hyperthermia 2023; 40:2184397. [PMID: 36888994 DOI: 10.1080/02656736.2023.2184397] [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: 03/10/2023] Open
Abstract
OBJECTIVE To develop and validate a radiomics nomogram for predicting the survival of patients with pancreatic ductal adenocarcinoma (PDAC) after receiving high-intensity focused ultrasound (HIFU) treatment. METHODS A total of 52 patients with PDAC were enrolled. To select features, the least absolute shrinkage and selection operator algorithm were applied, and the radiomics score (Rad-Score) was obtained. Radiomics model, clinics model, and radiomics nomogram model were constructed by multivariate regression analysis. The identification, calibration, and clinical application of nomogram were evaluated. Survival analysis was performed using Kaplan-Meier (K-M) method. RESULTS According to conclusions made from the multivariate Cox model, Rad-Score, and tumor size were independent risk factors for OS. Compared with the clinical model and radiomics model, the combination of Rad-Score and clinicopathological factors could better predict the survival of patients. Patients were divided into high-risk and low-risk groups according to Rad-Score. K-M analysis showed that the difference between the two groups was statistically significant (p < 0.05). In addition, the radiomics nomogram model indicated better discrimination, calibration, and clinical practicability in training and validation cohorts. CONCLUSION The radiomics nomogram effectively evaluates the prognosis of patients with advanced pancreatic cancer after HIFU surgery, which could potentially improve treatment strategies and promote individualized treatment of advanced pancreatic cancer.
Collapse
Affiliation(s)
- Changjie Shao
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Jing Guo
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liang Zhang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuhan Zhang
- University of Southern California, Los Angeles, CA, USA
| | - Leiyuan Ma
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuanxin Gong
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqi Tian
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Chen
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Yang J, Liu Y, Liu S. Comment on "Prognostic value of preoperative enhanced computed tomography as a quantitative imaging biomarker in pancreatic cancer". World J Gastroenterol 2022; 28:6310-6313. [PMID: 36504551 PMCID: PMC9730437 DOI: 10.3748/wjg.v28.i44.6310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 11/16/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies because of its high invasiveness and metastatic potential. Computed tomography (CT) is often used as a preliminary diagnostic tool for pancreatic cancer, and it is increasingly used to predict treatment response and disease stage. Recently, a study published in World Journal of Gastroenterology reported that quantitative analysis of preoperative enhanced CT data can be used to predict postoperative overall survival in patients with PDAC. A tumor relative enhancement ratio of ≤ 0.7 indicates a higher tumor stage and poor prognosis.
Collapse
Affiliation(s)
- Jian Yang
- Central Laboratory, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar 161000, Heilongjiang Province, China
| | - Ying Liu
- Department of Medical Oncology, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar 161000, Heilongjiang Province, China
| | - Shi Liu
- Central Laboratory, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar 161000, Heilongjiang Province, China
| |
Collapse
|
10
|
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
|
11
|
Liver metastases in pancreatic ductal adenocarcinoma: a predictive model based on CT texture analysis. Radiol Med 2022; 127:1079-1084. [PMID: 36057929 DOI: 10.1007/s11547-022-01548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
|
12
|
Marti-Bonmati L, Cerdá-Alberich L, Pérez-Girbés A, Díaz Beveridge R, Montalvá Orón E, Pérez Rojas J, Alberich-Bayarri A. Pancreatic cancer, radiomics and artificial intelligence. Br J Radiol 2022; 95:20220072. [PMID: 35687700 PMCID: PMC10996946 DOI: 10.1259/bjr.20220072] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 11/05/2022] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) are generally classified into four categories based on contrast-enhanced CT at diagnosis: resectable, borderline resectable, unresectable, and metastatic disease. In the initial grading and staging of PDAC, structured radiological templates are useful but limited, as there is a need to define the aggressiveness and microscopic disease stage of these tumours to ensure adequate treatment allocation. Quantitative imaging analysis allows radiomics and dynamic imaging features to provide information of clinical outcomes, and to construct clinical models based on radiomics signatures or imaging phenotypes. These quantitative features may be used as prognostic and predictive biomarkers in clinical decision-making, enabling personalised management of advanced PDAC. Deep learning and convolutional neural networks also provide high level bioinformatics tools that can help define features associated with a given aspect of PDAC biology and aggressiveness, paving the way to define outcomes based on these features. Thus, the prediction of tumour phenotype, treatment response and patient prognosis may be feasible by using such comprehensive and integrated radiomics models. Despite these promising results, quantitative imaging is not ready for clinical implementation in PDAC. Limitations include the instability of metrics and lack of external validation. Large properly annotated datasets, including relevant semantic features (demographics, blood markers, genomics), image harmonisation, robust radiomics analysis, clinically significant tasks as outputs, comparisons with gold-standards (such as TNM or pretreatment classifications) and fully independent validation cohorts, will be required for the development of trustworthy radiomics and artificial intelligence solutions to predict PDAC aggressiveness in a clinical setting.
Collapse
Affiliation(s)
- Luis Marti-Bonmati
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
- Department of Radiology, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Leonor Cerdá-Alberich
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
| | | | | | - Eva Montalvá Orón
- Department of Surgery, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Judith Pérez Rojas
- Department of Pathology, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Angel Alberich-Bayarri
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
- Quantitative Imaging Biomarkers in Medicine, Quibim
SL, Valencia,
Spain
| |
Collapse
|
13
|
Update on quantitative radiomics of pancreatic tumors. Abdom Radiol (NY) 2022; 47:3118-3160. [PMID: 34292365 DOI: 10.1007/s00261-021-03216-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Radiomics is a newer approach for analyzing radiological images obtained from conventional imaging modalities such as computed tomography, magnetic resonance imaging, endoscopic ultrasonography, and positron emission tomography. Radiomics involves extracting quantitative data from the images and assessing them to identify diagnostic or prognostic features such as tumor grade, resectability, tumor response to neoadjuvant therapy, and survival. The purpose of this review is to discuss the basic principles of radiomics and provide an overview of the current clinical applications of radiomics in the field of pancreatic tumors.
Collapse
|
14
|
Keyl J, Kasper S, Wiesweg M, Götze J, Schönrock M, Sinn M, Berger A, Nasca E, Kostbade K, Schumacher B, Markus P, Albers D, Treckmann J, Schmid KW, Schildhaus HU, Siveke JT, Schuler M, Kleesiek J. Multimodal survival prediction in advanced pancreatic cancer using machine learning. ESMO Open 2022; 7:100555. [PMID: 35988455 PMCID: PMC9588888 DOI: 10.1016/j.esmoop.2022.100555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background Existing risk scores appear insufficient to assess the individual survival risk of patients with advanced pancreatic ductal adenocarcinoma (PDAC) and do not take advantage of the variety of parameters that are collected during clinical care. Methods In this retrospective study, we built a random survival forest model from clinical data of 203 patients with advanced PDAC. The parameters were assessed before initiation of systemic treatment and included age, CA19-9, C-reactive protein, metastatic status, neutrophil-to-lymphocyte ratio and total serum protein level. Separate models including imaging and molecular parameters were built for subgroups. Results Over the entire cohort, a model based on clinical parameters achieved a c-index of 0.71. Our approach outperformed the American Joint Committee on Cancer (AJCC) staging system and the modified Glasgow Prognostic Score (mGPS) in the identification of high- and low-risk subgroups. Inclusion of the KRAS p.G12D mutational status could further improve the prediction, whereas radiomics data of the primary tumor only showed little benefit. In an external validation cohort of PDAC patients with liver metastases, our model achieved a c-index of 0.67 (mGPS: 0.59). Conclusions The combination of multimodal data and machine-learning algorithms holds potential for personalized prognostication in advanced PDAC already at diagnosis. We developed a machine-learning-based prediction model that outperforms the AJCC staging system and mGPS. Applying our model to an external validation cohort demonstrates generalizability. Explainable machine learning enables to understand the decision making of our model and identifies relevant parameters. Combining clinical, imaging and genetic data holds potential for personalized prognostication in advanced PDAC.
Collapse
Affiliation(s)
- J Keyl
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany.
| | - S Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - M Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - J Götze
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Schönrock
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Sinn
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Berger
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - E Nasca
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - K Kostbade
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - B Schumacher
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - P Markus
- Department of General Surgery and Traumatology, Elisabeth Hospital Essen, Essen, Germany
| | - D Albers
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - J Treckmann
- Department of General, Visceral and Transplant Surgery, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - K W Schmid
- Medical Faculty, University of Duisburg-Essen, Essen, Germany; Institute of Pathology, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - H-U Schildhaus
- Medical Faculty, University of Duisburg-Essen, Essen, Germany; Institute of Pathology, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - J T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany; Bridge Institute of Experimental Tumor Therapy (BIT), West German Cancer Center, University Hospital Essen (AöR), Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK) Partner site Essen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - J Kleesiek
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
15
|
A systematic review of prognosis predictive role of radiomics in pancreatic cancer: heterogeneity markers or statistical tricks? Eur Radiol 2022; 32:8443-8452. [PMID: 35904618 DOI: 10.1007/s00330-022-08922-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/07/2022] [Accepted: 05/30/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES We aimed to systematically evaluate the prognostic prediction accuracy of radiomics features extracted from pre-treatment imaging in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Radiomics literature on overall survival (OS) prediction of PDAC were all included in this systematic review. A further meta-analysis was performed on the effect size of first-order entropy. Methodological quality and risk of bias of the included studies were assessed by the radiomics quality score (RQS) and prediction model risk of bias assessment tool (PROBAST). RESULTS Twenty-three studies were finally identified in this review. Two (8.7%) studies compared prognosis prediction ability between radiomics model and TNM staging model by C-index, and both showed a better performance of the radiomics. Twenty-one (91.3%) studies reported significant predictive values of radiomics features. Nine (39.1%) studies were included in the meta-analysis, and it showed a significant correlation between first-order entropy and OS (HR 1.66, 95%CI 1.18-2.34). RQS assessment revealed validation was only performed in 5 (21.7%) studies on internal datasets and 2 (8.7%) studies on external datasets. PROBAST showed that 22 (95.7%) studies have a high risk of bias in participants because of the retrospective study design. CONCLUSION First-order entropy was significantly associated with OS and might improve the accuracy of PDAC prognosis prediction. Existing studies were poorly validated, and it should be noted in future studies. Modification of PROBAST for radiomics studies is necessary since the strict requirements of prospective study design may not be applicable to the demand for a large sample size in the model construction stage. KEY POINTS • Radiomics based on the primary lesion holds great potential for prognosis prediction. First-order entropy was significantly associated with the overall survival of PDAC and might improve the accuracy of current PDAC prognosis prediction. • We strongly recommend that at least an internal validation should be conducted in any radiomics study. Attention should be paid to the complex relationships between radiomics features. • Due to the close relationship between radiomics and big data, the strict requirement of prospective study design in PROABST may not be appropriate for radiomics studies. A balance between study types and sample sizes for radiomics studies needs to be found in the model construction stage.
Collapse
|
16
|
Cheng S, Yu X, Chen X, Jin Z, Xue H, Wang Z, Xie P. CT-based radiomics model for preoperative prediction of hepatic encephalopathy after transjugular intrahepatic portosystemic shunt. Br J Radiol 2022; 95:20210792. [PMID: 35019776 PMCID: PMC9153699 DOI: 10.1259/bjr.20210792] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To develop and evaluate a machine learning-based CT radiomics model for the prediction of hepatic encephalopathy (HE) after transjugular intrahepatic portosystemic shunt (TIPS). METHODS A total of 106 patients who underwent TIPS placement were consecutively enrolled in this retrospective study. Regions of interest (ROIs) were drawn on unenhanced, arterial phase, and portal venous phase CT images, and radiomics features were extracted, respectively. A radiomics model was established to predict the occurrence of HE after TIPS by using random forest algorithm and 10-fold cross-validation. Receiver operating characteristic (ROC) curves were performed to validate the capability of the radiomics model and clinical model on the training, test and original data sets, respectively. RESULTS The radiomics model showed favorable discriminatory ability in the training cohort with an area under the curve (AUC) of 0.899 (95% CI, 0.848 to 0.951), while in the test cohort, it was confirmed with an AUC of 0.887 (95% CI, 0.760 to 1.00). After applying this model to original data set, it had an AUC of 0.955 (95% CI, 0.896 to 1.00). A clinical model was also built with an AUC of 0.649 (95% CI, 0.530 to 0.767) in the original data set, and a Delong test demonstrated its relative lower efficiency when compared with the radiomics model (p < 0.05). CONCLUSION Machine learning-based CT radiomics model performed better than traditional clinical parameter-based models in the prediction of post-TIPS HE. ADVANCES IN KNOWLEDGE Radiomics model for the prediction of post-TIPS HE was built based on feature extraction from routine acquired pre-operative CT images and feature selection by random forest algorithm, which showed satisfied performance and proved the advantages of machine learning in this field.
Collapse
Affiliation(s)
- Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Department of Radiology, Sichuan Provincial People's Hospital, Sichuan Academy of Medical Sciences, Chengdu, China
| | - Xiang Yu
- Department of Radiology, Sichuan Provincial People's Hospital, Sichuan Academy of Medical Sciences, Chengdu, China
| | - Xinyue Chen
- CT Collaboration, Siemens-Healthineers, Beijing, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiwei Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ping Xie
- Department of Radiology, Sichuan Provincial People's Hospital, Sichuan Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
17
|
Yang Q, Mao Y, Xie H, Qin T, Mai Z, Cai Q, Wen H, Li Y, Zhang R, Liu L. Identifying Outcomes of Patients With Advanced Pancreatic Adenocarcinoma and RECIST Stable Disease Using Radiomics Analysis. JCO Precis Oncol 2022; 6:e2100362. [PMID: 35319966 PMCID: PMC8966975 DOI: 10.1200/po.21.00362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Few studies have explored the biomarkers for predicting the heterogeneous outcomes of patients with advanced pancreatic adenocarcinoma showing stable disease (SD) on the initial postchemotherapy computed tomography. We aimed to devise a radiomics signature (RS) to predict these outcomes for further risk stratification.
Collapse
Affiliation(s)
- Qiuxia Yang
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yize Mao
- Department of Pancreatic-Biliary Surgical Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hui Xie
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tao Qin
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhijun Mai
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qian Cai
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hailin Wen
- Cancer Hospital Chinese Academy of Medical Science, Shenzhen Center, Shenzhen, China
| | - Yong Li
- Department of Medical Imaging Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Zhang
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lizhi Liu
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
18
|
Preuss K, Thach N, Liang X, Baine M, Chen J, Zhang C, Du H, Yu H, Lin C, Hollingsworth MA, Zheng D. Using Quantitative Imaging for Personalized Medicine in Pancreatic Cancer: A Review of Radiomics and Deep Learning Applications. Cancers (Basel) 2022; 14:cancers14071654. [PMID: 35406426 PMCID: PMC8997008 DOI: 10.3390/cancers14071654] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary With a five-year survival rate of only 3% for the majority of patients, pancreatic cancer is a global healthcare challenge. Radiomics and deep learning, two novel quantitative imaging methods that treat medical images as minable data instead of just pictures, have shown promise in advancing personalized management of pancreatic cancer through diagnosing precursor diseases, early detection, accurate diagnosis, and treatment personalization. Radiomics and deep learning methods aim to collect hidden information in medical images that is missed by conventional radiology practices through expanding the data search and comparing information across different patients. Both methods have been studied and applied in pancreatic cancer. In this review, we focus on the current progress of these two methods in pancreatic cancer and provide a comprehensive narrative review on the topic. With better regulation, enhanced workflow, and larger prospective patient datasets, radiomics and deep learning methods could show real hope in the battle against pancreatic cancer through personalized precision medicine. Abstract As the most lethal major cancer, pancreatic cancer is a global healthcare challenge. Personalized medicine utilizing cutting-edge multi-omics data holds potential for major breakthroughs in tackling this critical problem. Radiomics and deep learning, two trendy quantitative imaging methods that take advantage of data science and modern medical imaging, have shown increasing promise in advancing the precision management of pancreatic cancer via diagnosing of precursor diseases, early detection, accurate diagnosis, and treatment personalization and optimization. Radiomics employs manually-crafted features, while deep learning applies computer-generated automatic features. These two methods aim to mine hidden information in medical images that is missed by conventional radiology and gain insights by systematically comparing the quantitative image information across different patients in order to characterize unique imaging phenotypes. Both methods have been studied and applied in various pancreatic cancer clinical applications. In this review, we begin with an introduction to the clinical problems and the technology. After providing technical overviews of the two methods, this review focuses on the current progress of clinical applications in precancerous lesion diagnosis, pancreatic cancer detection and diagnosis, prognosis prediction, treatment stratification, and radiogenomics. The limitations of current studies and methods are discussed, along with future directions. With better standardization and optimization of the workflow from image acquisition to analysis and with larger and especially prospective high-quality datasets, radiomics and deep learning methods could show real hope in the battle against pancreatic cancer through big data-based high-precision personalization.
Collapse
Affiliation(s)
- Kiersten Preuss
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Nutrition and Health Sciences, University of Nebraska Lincoln, Lincoln, NE 68588, USA
| | - Nate Thach
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Computer Science, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Xiaoying Liang
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Michael Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
| | - Justin Chen
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Naperville North High School, Naperville, IL 60563, USA
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Huijing Du
- Department of Mathematics, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Hongfeng Yu
- Department of Computer Science, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Dandan Zheng
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14626, USA
- Correspondence: ; Tel.: +1-(585)-276-3255
| |
Collapse
|
19
|
Chen X, Fu R, Shao Q, Chen Y, Ye Q, Li S, He X, Zhu J. Application of artificial intelligence to pancreatic adenocarcinoma. Front Oncol 2022; 12:960056. [PMID: 35936738 PMCID: PMC9353734 DOI: 10.3389/fonc.2022.960056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Pancreatic cancer (PC) is one of the deadliest cancers worldwide although substantial advancement has been made in its comprehensive treatment. The development of artificial intelligence (AI) technology has allowed its clinical applications to expand remarkably in recent years. Diverse methods and algorithms are employed by AI to extrapolate new data from clinical records to aid in the treatment of PC. In this review, we will summarize AI's use in several aspects of PC diagnosis and therapy, as well as its limits and potential future research avenues. METHODS We examine the most recent research on the use of AI in PC. The articles are categorized and examined according to the medical task of their algorithm. Two search engines, PubMed and Google Scholar, were used to screen the articles. RESULTS Overall, 66 papers published in 2001 and after were selected. Of the four medical tasks (risk assessment, diagnosis, treatment, and prognosis prediction), diagnosis was the most frequently researched, and retrospective single-center studies were the most prevalent. We found that the different medical tasks and algorithms included in the reviewed studies caused the performance of their models to vary greatly. Deep learning algorithms, on the other hand, produced excellent results in all of the subdivisions studied. CONCLUSIONS AI is a promising tool for helping PC patients and may contribute to improved patient outcomes. The integration of humans and AI in clinical medicine is still in its infancy and requires the in-depth cooperation of multidisciplinary personnel.
Collapse
Affiliation(s)
- Xi Chen
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Ruibiao Fu
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Shao
- Department of Surgical Ward 1, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Yan Chen
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qinghuang Ye
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Li
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiongxiong He
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Jinhui Zhu
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Jinhui Zhu,
| |
Collapse
|
20
|
Nardone V, Reginelli A, Grassi R, Boldrini L, Vacca G, D'Ippolito E, Annunziata S, Farchione A, Belfiore MP, Desideri I, Cappabianca S. Delta radiomics: a systematic review. Radiol Med 2021; 126:1571-1583. [PMID: 34865190 DOI: 10.1007/s11547-021-01436-7] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/18/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Radiomics can provide quantitative features from medical imaging that can be correlated with various biological features and clinical endpoints. Delta radiomics, on the other hand, consists in the analysis of feature variation at different acquisition time points, usually before and after therapy. The aim of this study was to provide a systematic review of the different delta radiomics approaches. METHODS Eligible articles were searched in Embase, PubMed, and ScienceDirect using a search string that included free text and/or Medical Subject Headings (MeSH) with three key search terms: "radiomics", "texture", and "delta". Studies were analysed using QUADAS-2 and the RQS tool. RESULTS Forty-eight studies were finally included. The studies were divided into preclinical/methodological (five studies, 10.4%); rectal cancer (six studies, 12.5%); lung cancer (twelve studies, 25%); sarcoma (five studies, 10.4%); prostate cancer (three studies, 6.3%), head and neck cancer (six studies, 12.5%); gastrointestinal malignancies excluding rectum (seven studies, 14.6%), and other disease sites (four studies, 8.3%). The median RQS of all studies was 25% (mean 21% ± 12%), with 13 studies (30.2%) achieving a quality score < 10% and 22 studies (51.2%) < 25%. CONCLUSIONS Delta radiomics shows potential benefit for several clinical endpoints in oncology (differential diagnosis, prognosis and prediction of treatment response, and evaluation of side effects). Nevertheless, the studies included in this systematic review suffer from the bias of overall low quality, so that the conclusions are currently heterogeneous, not robust, and not replicable. Further research with prospective and multicentre studies is needed for the clinical validation of delta radiomics approaches.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy.
| | - Roberta Grassi
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Luca Boldrini
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giovanna Vacca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Emma D'Ippolito
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Salvatore Annunziata
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Alessandra Farchione
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Maria Paola Belfiore
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
21
|
Cheng S, Jin Z, Xue H. Assessment of Response to Chemotherapy in Pancreatic Cancer with Liver Metastasis: CT Texture as a Predictive Biomarker. Diagnostics (Basel) 2021; 11:diagnostics11122252. [PMID: 34943489 PMCID: PMC8700536 DOI: 10.3390/diagnostics11122252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/21/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
In this paper, we assess changes in CT texture of metastatic liver lesions after treatment with chemotherapy in patients with pancreatic cancer and determine if texture parameters correlate with measured time to progression (TTP). This retrospective study included 110 patients with pancreatic cancer with liver metastasis, and mean, entropy, kurtosis, skewness, mean of positive pixels, and standard deviation (SD) values were extracted during texture analysis. Response assessment was also obtained by using RECIST 1.1, Choi and modified Choi criteria, respectively. The correlation of texture parameters and existing assessment criteria with TTP were evaluated using Kaplan-Meier and Cox regression analyses in the training cohort. Kaplan-Meier curves of the proportion of patients without disease progression were significantly different for several texture parameters, and were better than those for RECIST 1.1-, Choi-, and modified Choi-defined response (p < 0.05 vs. p = 0.398, p = 0.142, and p = 0.536, respectively). Cox regression analysis showed that percentage change in SD was an independent predictor of TTP (p = 0.016) and confirmed in the validation cohort (p = 0.019). In conclusion, CT texture parameters have the potential to become predictive imaging biomarkers for response evaluation in pancreatic cancer with liver metastasis.
Collapse
|
22
|
Gebauer L, Moltz JH, Mühlberg A, Holch JW, Huber T, Enke J, Jäger N, Haas M, Kruger S, Boeck S, Sühling M, Katzmann A, Hahn H, Kunz WG, Heinemann V, Nörenberg D, Maurus S. Quantitative Imaging Biomarkers of the Whole Liver Tumor Burden Improve Survival Prediction in Metastatic Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13225732. [PMID: 34830885 PMCID: PMC8616514 DOI: 10.3390/cancers13225732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Finding prognostic biomarkers and associated models with high accuracy in patients with pancreatic cancer remains a challenge. The aim of this study was to analyze whether the combination of quantitative imaging biomarkers based on geometric and radiomics analysis of whole liver tumor burden and established clinical parameters improves the prediction of survival in patients with metastatic pancreatic cancer. In this retrospective study a total of 75 patients with pancreatic cancer and liver metastases were analyzed. Segmentations of whole liver tumor burden from baseline contrast-enhanced CT images were used to derive different quantitative imaging biomarkers. For comparison, we chose two clinical prognostic models from the literature. We found that a combined clinical and imaging-based model has a significantly higher predictive performance to discriminate survival than the underlying clinical models alone (p < 0.003). Abstract Finding prognostic biomarkers with high accuracy in patients with pancreatic cancer (PC) remains a challenging problem. To improve the prediction of survival and to investigate the relevance of quantitative imaging biomarkers (QIB) we combined QIB with established clinical parameters. In this retrospective study a total of 75 patients with metastatic PC and liver metastases were analyzed. Segmentations of whole liver tumor burden (WLTB) from baseline contrast-enhanced CT images were used to derive QIBs. The benefits of QIBs in multivariable Cox models were analyzed in comparison with two clinical prognostic models from the literature. To discriminate survival, the two clinical models had concordance indices of 0.61 and 0.62 in a statistical setting. Combined clinical and imaging-based models achieved concordance indices of 0.74 and 0.70 with WLTB volume, tumor burden score (TBS), and bilobar disease being the three WLTB parameters that were kept by backward elimination. These combined clinical and imaging-based models have significantly higher predictive performance in discriminating survival than the underlying clinical models alone (p < 0.003). Radiomics and geometric WLTB analysis of patients with metastatic PC with liver metastases enhances the modeling of survival compared with models based on clinical parameters alone.
Collapse
Affiliation(s)
- Leonie Gebauer
- Department of Medicine III, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.W.H.); (M.H.); (S.K.); (S.B.); (V.H.)
- Correspondence:
| | - Jan H. Moltz
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Str. 2, 28359 Bremen, Germany; (J.H.M.); (H.H.)
| | - Alexander Mühlberg
- CT R&D Image Analytics, Siemens Healthineers, Siemensstr. 1, 91301 Forchheim, Germany; (A.M.); (M.S.); (A.K.)
| | - Julian W. Holch
- Department of Medicine III, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.W.H.); (M.H.); (S.K.); (S.B.); (V.H.)
| | - Thomas Huber
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (T.H.); (J.E.); (N.J.); (W.G.K.); (D.N.); (S.M.)
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Johanna Enke
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (T.H.); (J.E.); (N.J.); (W.G.K.); (D.N.); (S.M.)
| | - Nils Jäger
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (T.H.); (J.E.); (N.J.); (W.G.K.); (D.N.); (S.M.)
| | - Michael Haas
- Department of Medicine III, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.W.H.); (M.H.); (S.K.); (S.B.); (V.H.)
| | - Stephan Kruger
- Department of Medicine III, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.W.H.); (M.H.); (S.K.); (S.B.); (V.H.)
| | - Stefan Boeck
- Department of Medicine III, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.W.H.); (M.H.); (S.K.); (S.B.); (V.H.)
| | - Michael Sühling
- CT R&D Image Analytics, Siemens Healthineers, Siemensstr. 1, 91301 Forchheim, Germany; (A.M.); (M.S.); (A.K.)
| | - Alexander Katzmann
- CT R&D Image Analytics, Siemens Healthineers, Siemensstr. 1, 91301 Forchheim, Germany; (A.M.); (M.S.); (A.K.)
| | - Horst Hahn
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Str. 2, 28359 Bremen, Germany; (J.H.M.); (H.H.)
| | - Wolfgang G. Kunz
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (T.H.); (J.E.); (N.J.); (W.G.K.); (D.N.); (S.M.)
| | - Volker Heinemann
- Department of Medicine III, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.W.H.); (M.H.); (S.K.); (S.B.); (V.H.)
| | - Dominik Nörenberg
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (T.H.); (J.E.); (N.J.); (W.G.K.); (D.N.); (S.M.)
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Stefan Maurus
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (T.H.); (J.E.); (N.J.); (W.G.K.); (D.N.); (S.M.)
| |
Collapse
|
23
|
Sofue K, Ueshima E, Masuda A, Shirakawa S, Zen Y, Ueno Y, Tsujita Y, Yamaguchi T, Yabe S, Tanaka T, Inomata N, Toyama H, Fukumoto T, Kodama Y, Murakami T. Estimation of pancreatic fibrosis and prediction of postoperative pancreatic fistula using extracellular volume fraction in multiphasic contrast-enhanced CT. Eur Radiol 2021; 32:1770-1780. [PMID: 34636963 DOI: 10.1007/s00330-021-08255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/30/2021] [Accepted: 08/07/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To investigate the diagnostic performance of the extracellular volume (ECV) fraction in multiphasic contrast-enhanced computed tomography (CE-CT) for estimating histologic pancreatic fibrosis and predicting postoperative pancreatic fistula (POPF). METHODS Eighty-five patients (49 men; mean age, 69 years) who underwent multiphasic CE-CT followed by pancreaticoduodenectomy with pancreaticojejunal anastomosis between January 2012 and December 2018 were retrospectively included. The ECV fraction was calculated from absolute enhancements of the pancreas and aorta between the precontrast and equilibrium-phase images, followed by comparisons among histologic pancreatic fibrosis grades (F0‒F3). The diagnostic performance of the ECV fraction in advanced fibrosis (F2‒F3) was evaluated using receiver operating characteristic curve analysis. Multivariate logistic regression analysis was used to evaluate the associations of the risk of POPF development with patient characteristics, histologic findings, and CT imaging parameters. RESULTS The mean ECV fraction of the pancreas was 34.4% ± 9.5, with an excellent intrareader agreement of 0.811 and a moderate positive correlation with pancreatic fibrosis (r = 0.476; p < 0.001). The mean ECV fraction in advanced fibrosis was significantly higher than that in no/mild fibrosis (44.4% ± 10.8 vs. 31.7% ± 6.7; p < 0.001), and the area under the receiver operating characteristic curve for the diagnosis of advanced fibrosis was 0.837. Twenty-two patients (25.9%) developed clinically relevant POPF. Multivariate logistic regression analysis demonstrated that the ECV fraction was a significant predictor of POPF. CONCLUSIONS The ECV fraction can offer quantitative information for assessing pancreatic fibrosis and POPF after pancreaticojejunal anastomosis. KEY POINTS • There was a moderate positive correlation of the extracellular volume (ECV) fraction of the pancreas in contrast-enhanced CT with the histologic grade of pancreatic fibrosis (r = 0.476; p < 0.001). • The ECV fraction was higher in advanced fibrosis (F2‒F3) than in no/mild fibrosis (F0‒F1) (p < 0.001), with an AUC of 0.837 for detecting advanced fibrosis. • The ECV fraction was an independent risk factor for predicting subclinical (odds ratio, 0.81) and clinical (odds ratio, 0.80) postoperative pancreatic fistula.
Collapse
Affiliation(s)
- 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
| | - Atsuhiro Masuda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sachiyo Shirakawa
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital & King's College London, London, UK
| | - Yoshiko Ueno
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yushi Tsujita
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takeru Yamaguchi
- 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
| | - Takeshi Tanaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriko Inomata
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hirochika Toyama
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Fukumoto
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
24
|
Chen X, Liu F, Xue Q, Weng X, Xu F. Metastatic pancreatic cancer: Mechanisms and detection (Review). Oncol Rep 2021; 46:231. [PMID: 34498718 PMCID: PMC8444192 DOI: 10.3892/or.2021.8182] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal malignancy. Its prevalence rate remains low but continues to grow each year. Among all stages of PC, metastatic PC is defined as late-stage (stage IV) PC and has an even higher fatality rate. Patients with PC do not have any specific clinical manifestations. Most cases are inoperable at the time-point of diagnosis. Prognosis is also poor even with curative-intent surgery. Complications during surgery, postoperative pancreatic fistula and recurrence with metastatic foci make the management of metastatic PC difficult. While extensive efforts were made to improve survival outcomes, further elucidation of the molecular mechanisms of metastasis poses a formidable challenge. The present review provided an overview of the mechanisms of metastatic PC, summarizing currently known signaling pathways (e.g. epithelial-mesenchymal transition, NF-κB and KRAS), imaging that may be utilized for early detection and biomarkers (e.g. carbohydrate antigen 19-9, prostate cancer-associated transcript-1, F-box/LRR-repeat protein 7 and tumor stroma), giving insight into promising therapeutic targets.
Collapse
Affiliation(s)
- Xiangling Chen
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Fangfang Liu
- Department of Art, Art College, Southwest Minzu University, Chengdu, Sichuan 610041, P.R. China
| | - Qingping Xue
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiechuan Weng
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
25
|
Vincent P, Maeder ME, Hunt B, Linn B, Mangels-Dick T, Hasan T, Wang KK, Pogue BW. CT radiomic features of photodynamic priming in clinical pancreatic adenocarcinoma treatment. Phys Med Biol 2021; 66. [PMID: 34261044 DOI: 10.1088/1361-6560/ac1458] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022]
Abstract
Photodynamic therapy (PDT) offers localized focal ablation in unresectable pancreatic tumors while tissues surrounding the treatment volume experience a lower light dose, termed photodynamic priming (PDP). While PDP does not cause tissue damage, it has been demonstrated to promote vascular permeability, improve drug delivery, alleviate tumor cell density, and reduce desmoplasia and the resultant internal pressure in pre-clinical evaluation. Preclinical data supports PDP as a neoadjuvant therapy beneficial to subsequent chemotherapy or immunotherapy, yet it is challenging to quantify PDP effects in clinical treatment without additional imaging and testing. This study investigated the potential of radiomic analysis using CT scans acquired before and after PDT to identify areas experiencing PDT-induced necrosis as well as quantify PDP effects in the surrounding tissues. A total of 235 CT tumor slices from seven patients undergoing PDT for pancreatic tumors were examined. Radiomic features assessed included intensity metrics (CT number in Hounsfield Units) and texture analysis using several gray-level co-occurrence matrix (GLCM) parameters. Pre-treatment scans of tumor areas that resulted in PDT-induced necrosis showed statistically significant differences in intensity and texture-based features that could be used to predict the regions that did respond (paired t-test, response versus no response,p < 0.001). Evaluation of PDP effects on the surrounding tissues also demonstrated statistically significant differences, in tumor mean value, standard deviation, and GLCM parameters of contrast, dissimilarity and homogeneity (t-test, pre versus post,p < 0.001). Using leave-one-out cross validation, six intensity and texture-based features were combined into a support-vector machine model which demonstrated reliable prediction of treatment effects for six out of seven patients (ROC curve, AUC = 0.93). This study provides pilot evidence that texture features extracted from CT scans could be utilized as an effective clinical diagnostic prediction and assessment of PDT and PDP effects in pancreatic tumors. (clinical trial NCT03033225).
Collapse
Affiliation(s)
- Phuong Vincent
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, United States of America
| | - Matthew E Maeder
- Dartmouth-Hitchcock Department of Radiology, Lebanon NH 03756, United States of America
| | - Brady Hunt
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, United States of America
| | - Bryan Linn
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55902, United States of America
| | - Tiffany Mangels-Dick
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55902, United States of America
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114, United States of America
| | - Kenneth K Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55902, United States of America
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, United States of America
| |
Collapse
|
26
|
Salinas-Miranda E, Deniffel D, Dong X, Healy GM, Khalvati F, O'Kane GM, Knox J, Bathe OF, Baracos VE, Gallinger S, Haider MA. Prognostic value of early changes in CT-measured body composition in patients receiving chemotherapy for unresectable pancreatic cancer. Eur Radiol 2021; 31:8662-8670. [PMID: 33934171 DOI: 10.1007/s00330-021-07899-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Skeletal muscle mass is a prognostic factor in pancreatic ductal adenocarcinoma (PDAC). However, it remains unclear whether changes in body composition provide an incremental prognostic value to established risk factors, especially the Response Evaluation Criteria in Solid Tumors version 1.1 (RECISTv1.1). The aim of this study was to determine the prognostic value of CT-quantified body composition changes in patients with unresectable PDAC starting chemotherapy. METHODS We retrospectively evaluated 105 patients with unresectable (locally advanced or metastatic) PDAC treated with FOLFIRINOX (n = 64) or gemcitabine-based (n = 41) first-line chemotherapy within a multicenter prospective trial. Changes (Δ) in skeletal muscle index (SMI), subcutaneous (SATI), and visceral adipose tissue index (VATI) between pre-chemotherapy and first follow-up CT were assessed. Cox regression models and covariate-adjusted survival curves were used to identify predictors of overall survival (OS). RESULTS At multivariable analysis, adjusting for RECISTv1.1-response at first follow-up, ΔSMI was prognostic for OS with a hazard ratio (HR) of 1.2 (95% CI: 1.08-1.33, p = 0.001). No significant association with OS was observed for ΔSATI (HR: 1, 95% CI: 0.97-1.04, p = 0.88) and ΔVATI (HR: 1.01, 95% CI: 0.99-1.04, p = 0.33). At an optimal cutoff of 2.8 cm2/m2 per 30 days, the median survival of patients with high versus low ΔSMI was 143 versus 233 days (p < 0.001). CONCLUSIONS Patients with a lower rate of skeletal muscle loss at first follow-up demonstrated improved survival for unresectable PDAC, regardless of their RECISTv1.1-category. Assessing ΔSMI at the first follow-up CT may be useful for prognostication, in addition to routine radiological assessment. KEY POINTS • In patients with unresectable pancreatic ductal adenocarcinoma, change of skeletal muscle index (ΔSMI) in the early phase of chemotherapy is prognostic for overall survival, even after adjusting for Response Evaluation Criteria in Solid Tumors version 1.1 (RECISTv1.1) assessment at first follow-up. • Changes in adipose tissue compartments at first follow-up demonstrated no significant association with overall survival. • Integrating ΔSMI into routine radiological assessment may improve prognostic stratification and impact treatment decision-making at the first follow-up.
Collapse
Affiliation(s)
- Emmanuel Salinas-Miranda
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada
| | - Dominik Deniffel
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada.,Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Xin Dong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Gerard M Healy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada
| | - Farzad Khalvati
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada
| | - Grainne M O'Kane
- Department of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Jennifer Knox
- Department of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.,Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Oliver F Bathe
- Departments of Surgery and Oncology, University of Calgary, Calgary, AB, Canada
| | - Vickie E Baracos
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Steven Gallinger
- Ontario Institute for Cancer Research, Toronto, ON, Canada.,Hepatobiliary Pancreatic Surgical Oncology Program, University Health Network, Toronto, ON, Canada
| | - Masoom A Haider
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada. .,Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada. .,Ontario Institute for Cancer Research, Toronto, ON, Canada.
| |
Collapse
|
27
|
Abunahel BM, Pontre B, Kumar H, Petrov MS. Pancreas image mining: a systematic review of radiomics. Eur Radiol 2021; 31:3447-3467. [PMID: 33151391 DOI: 10.1007/s00330-020-07376-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/25/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To systematically review published studies on the use of radiomics of the pancreas. METHODS The search was conducted in the MEDLINE database. Human studies that investigated the applications of radiomics in diseases of the pancreas were included. The radiomics quality score was calculated for each included study. RESULTS A total of 72 studies encompassing 8863 participants were included. Of them, 66 investigated focal pancreatic lesions (pancreatic cancer, precancerous lesions, or benign lesions); 4, pancreatitis; and 2, diabetes mellitus. The principal applications of radiomics were differential diagnosis between various types of focal pancreatic lesions (n = 19), classification of pancreatic diseases (n = 23), and prediction of prognosis or treatment response (n = 30). Second-order texture features were most useful for the purpose of differential diagnosis of diseases of the pancreas (with 100% of studies investigating them found a statistically significant feature), whereas filtered image features were most useful for the purpose of classification of diseases of the pancreas and prediction of diseases of the pancreas (with 100% of studies investigating them found a statistically significant feature). The median radiomics quality score of the included studies was 28%, with the interquartile range of 22% to 36%. The radiomics quality score was significantly correlated with the number of extracted radiomics features (r = 0.52, p < 0.001) and the study sample size (r = 0.34, p = 0.003). CONCLUSIONS Radiomics of the pancreas holds promise as a quantitative imaging biomarker of both focal pancreatic lesions and diffuse changes of the pancreas. The usefulness of radiomics features may vary depending on the purpose of their application. Standardisation of image acquisition protocols and image pre-processing is warranted prior to considering the use of radiomics of the pancreas in routine clinical practice. KEY POINTS • Methodologically sound studies on radiomics of the pancreas are characterised by a large sample size and a large number of extracted features. • Optimisation of the radiomics pipeline will increase the clinical utility of mineable pancreas imaging data. • Radiomics of the pancreas is a promising personalised medicine tool in diseases of the pancreas.
Collapse
Affiliation(s)
| | - Beau Pontre
- School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Haribalan Kumar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
28
|
Hang J, Xu K, Yin R, Shao Y, Liu M, Shi H, Wang X, Wu L. Role of CT texture features for predicting outcome of pancreatic cancer patients with liver metastases. J Cancer 2021; 12:2351-2358. [PMID: 33758611 PMCID: PMC7974874 DOI: 10.7150/jca.49569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: The purpose of this study was to evaluate the prognostic value of computed tomography (CT) texture features of pancreatic cancer with liver metastases. Methods: We included 39 patients with metastatic pancreatic cancer (MPC) with liver metastases and performed texture analysis on primary tumors and metastases. The correlations between texture parameters were assessed using Pearson's correlation. Univariate Cox proportional hazards model was used to assess the correlations between clinicopathological characteristics, texture features and overall survival (OS). The univariate Cox regression model revealed four texture features potentially correlated with OS (P<0.1). A radiomics score (RS) was determined using a sequential combination of four texture features with potential prognostic value that were weighted according to their β-coefficients. Furthermore, all variables with P<0.1 were included in the multivariate analysis. A nomogram,which was developed to predict OS according to independent prognostic factors, was internally validated using the C-index and calibration plots. Kaplan-Meier analysis and the log-rank test were performed to stratify OS according to the RS and nomogram total points (NTP). Results: Few significant correlations were found between texture features of primary tumors and those of liver metastases. However, texture features within primary tumors or liver metastases were significantly associated. Multivariate analysis showed that Eastern Cooperative Oncology Group performance status (ECOG PS), chemotherapy, Carbohydrate antigen 19-9 (CA19-9), and the RS were independent prognostic factors (P<0.05). The nomogram incorporating these factors showed good discriminative ability (C-index = 0.754). RS and NTP stratified patients into two potential risk groups (P<0.01). Conclusion: The RS derived from significant texture features of primary tumors and metastases shows promise as a prognostic biomarker of OS of patients with MPC. A nomogram based on the RS and other independent prognostic clinicopathological factors accurately predicts OS.
Collapse
Affiliation(s)
- Junjie Hang
- Department of Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Kequn Xu
- Department of Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Ruohan Yin
- Department of Medical Imaging, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Yueting Shao
- Department of Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Muhan Liu
- Department of Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Haifeng Shi
- Department of Medical Imaging, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Xiaoyong Wang
- Department of Gastroenterology, Changzhou No.2 People's Hospital, Nanjing Medical University, Xinglong Road 19, Changzhou 213000, China
| | - Lixia Wu
- Department of Oncology, Shanghai JingAn District ZhaBei Central Hospital, Zhonghuaxin Road 619, Shanghai 200040, China
| |
Collapse
|
29
|
The potential role of MR based radiomic biomarkers in the characterization of focal testicular lesions. Sci Rep 2021; 11:3456. [PMID: 33568713 PMCID: PMC7875983 DOI: 10.1038/s41598-021-83023-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 01/28/2021] [Indexed: 12/18/2022] Open
Abstract
How to differentiate with MRI-based techniques testicular germ (TGCTs) and testicular non-germ cell tumors (TNGCTs) is still under debate and Radiomics may be the turning key. Our purpose is to investigate the performance of MRI-based Radiomics signatures for the preoperative prediction of testicular neoplasm histology. The aim is twofold: (i), differentiating TGCTs and TNGCTs status and (ii) differentiating seminomas (SGCTs) from non-seminomatous (NSGCTs). Forty-two patients with pathology-proven testicular neoplasms and referred for pre-treatment MRI, were retrospectively enrolled. Thirty-two out of 44 lesions were TGCTs. Twelve out of 44 were TNGCTs or other histologies. Two radiologists segmented the volume of interest on T2-weighted images. Approximately 500 imaging features were extracted. Least Absolute Shrinkage and Selection Operator (LASSO) was applied as method for variable selection. A linear model and a linear support vector machine (SVM) were trained with selected features to assess discrimination scores for the two endpoints. LASSO identified 3 features that were employed to build fivefold validated linear discriminant and linear SVM classifiers for the TGCT-TNGCT endpoint giving an overall accuracy of 89%. Four features were employed to build another SVM for the SGCT-SNGCT endpoint with an overall accuracy of 86%. The data obtained proved that T2-weighted-based Radiomics is a promising tool in the diagnostic workup of testicular neoplasms by discriminating germ cell from non-gem cell tumors, and seminomas from non-seminomas.
Collapse
|
30
|
Chen F, Zhou Y, Qi X, Xia W, Zhang R, Zhang J, Gao X, Zhang L. CT texture analysis for the presurgical prediction of superior mesenteric-portal vein invasion in pancreatic ductal adenocarcinoma: comparison with CT imaging features. Clin Radiol 2021; 76:358-366. [PMID: 33581837 DOI: 10.1016/j.crad.2021.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022]
Abstract
AIM To investigate the value of computed tomography (CT) texture analysis (TA) and imaging features for evaluating suspected surgical superior mesenteric-portal vein (SMPV) invasion in patients with pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Fifty-four patients with PDAC in the pancreatic head or uncinate process with suspected SMPV involvement were analysed retrospectively. SMPV invasion status was identified by surgical exploration. For each patient, 396 texture features were extracted on pretreatment CT. Non-parametric tests and minimum redundancy maximum relevance were used for feature selection. A CTTA model was constructed using multivariate logistic regression, and the area under the receiver operating characteristic (AUROC) of the model was calculated. Two reviewers evaluated qualitative imaging features independently for SMPV invasion and interobserver agreement was investigated. The diagnostic performance of the imaging features and the CTTA model for SMPV invasion was compared using the McNemar test. RESULTS Of the 54 patients with PDAC, SMPV invasion was detected in 23 (42.6%). The CTTA model yielded an AUROC of 0.88 (95% confidence interval, 0.76-0.97) and achieved significantly higher specificity (0.90) than the two reviewers (0.61 and 0.65; p=0.027 and 0.043). Interobserver agreement was moderate between the two reviewers (κ = 0.517). Of the 13 cases with disagreement between the two reviewers, 11 cases were predicted accurately by the CTTA model. CONCLUSION CTTA can predict suspected SMPV invasion in PDAC and may be a beneficial addition for qualitative imaging evaluation.
Collapse
Affiliation(s)
- F Chen
- Department of Radiology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Y Zhou
- Department of Hepatobiliary Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - X Qi
- Department of Pathology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - W Xia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - R Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - J Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - X Gao
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - L Zhang
- Department of Radiology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China.
| |
Collapse
|
31
|
Qiu JJ, Yin J, Qian W, Liu JH, Huang ZX, Yu HP, Ji L, Zeng XX. A Novel Multiresolution-Statistical Texture Analysis Architecture: Radiomics-Aided Diagnosis of PDAC Based on Plain CT Images. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:12-25. [PMID: 32877335 DOI: 10.1109/tmi.2020.3021254] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Early screening of PDAC (pancreatic ductal adenocarcinoma) based on plain CT (computed tomography) images is of great significance. Therefore, this work conducted a radiomics-aided diagnosis analysis of PDAC based on plain CT images. We explored a novel MSTA (multiresolution-statistical texture analysis) architecture to extract texture features and built machine learning models to classify PDACs and HPs (healthy pancreases). We also performed significance tests of differences to analyze the relationships between histopathological characteristics and texture features. The MSTA architecture originates from the analysis of histopathological characteristics and combines multiresolution analysis and statistical analysis to extract texture features. The MSTA architecture achieved better experimental results than the traditional architecture that scales the coefficient matrices of the multiresolution analysis. In the validation of the classifications, the MSTA architecture achieved an accuracy of 81.19% and an AUC (area under the ROC (receiver operating characteristic) curve) of 0.88 (95% confidence interval: 0.84-0.92). In the test of the classifications, it achieved an accuracy of 77.66% and an AUC of 0.79 (95% confidence interval: 0.71-0.87). Moreover, the significance tests of differences showed that the extracted texture features may be relevant to the histopathological characteristics. The MSTA architecture is beneficial for the radiomics-aided diagnosis of PDAC based on plain CT images. Its texture features can potentially enhance radiologists' imaging interpretation abilities.
Collapse
|
32
|
|
33
|
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
|
34
|
Nie K, Zhang L, You Y, Li H, Guo X, Zhang Z, Zhang C, Ji Y. Irinotecan combined with oxaliplatin and S-1 in patients with metastatic pancreatic adenocarcinoma: a single-arm, three-centre, prospective study. Ther Adv Med Oncol 2020; 12:1758835920970843. [PMID: 33240399 PMCID: PMC7675906 DOI: 10.1177/1758835920970843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/09/2020] [Indexed: 01/05/2023] Open
Abstract
Objective To study the efficacy and toxicity of irinotecan combined with oxaliplatin and S-1 in patients with metastatic pancreatic adenocarcinoma. Patients and methods Previously untreated patients with cytologically or histologically confirmed metastatic pancreatic adenocarcinoma underwent a treatment regimen consisting of an intravenous infusion of irinotecan 165 mg/m2 and oxaliplatin 85 mg/m2 on day 1, and oral S-1 40 mg/m2 twice daily on days 1-14, repeating the regimen every 21 days until one of the following occurred: disease progression, intolerable toxicity, or patient death. The primary endpoint was overall survival (OS), and the secondary endpoints were progression-free survival (PFS), response rate, toxicity, and quality of life. This ongoing study had been registered on ClinicalTrials.gov, NCT03726021. Results A total of 41 patients were enrolled in this study, 18 men and 23 women. The median PFS was 4.33 months [95% confidence interval (CI): 2.83-5.88] and the median OS was 11.00 months (95% CI: 9.16-12.84). There were no instances of a complete response; the partial response, stable disease, and disease progression rates were 39.02% (16/41), 29.27% (12/41), and 31.71% (13/41), respectively.The most common adverse side effects were mild to moderate nausea, vomiting, neutropenia, and thrombocytopenia. Grade 3 or 4 neutropenia and thrombocytopenia were observed in 29.27% (12/41) and 12.20% (5/41) of the patients, respectively. No treatment-related death was observed. Conclusion Irinotecan combined with oxaliplatin and S-1 is a safe and effective treatment for metastatic pancreatic adenocarcinoma, and any toxicities are mild to moderate and tolerable. A larger study population is needed for further evaluation.
Collapse
Affiliation(s)
- Keke Nie
- Department of Oncology, Qingdao Central Hospital, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Ling Zhang
- Department of Oncology, Qingdao Central Hospital, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Yunhong You
- Department of Oncology, Qingdao Central Hospital, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Hongmei Li
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiuhui Guo
- Department of Haematology & Oncology, Pingdu People's Hospital, Shandong, China
| | - Zhongfa Zhang
- Department of Oncology, Qingdao Central Hospital, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Chunling Zhang
- Department of Oncology, Qingdao Central Hospital, The Affiliated Qingdao Central Hospital of Qingdao University, 127 Si Liu South Road, Qingdao City, 266042, China
| | - Youxin Ji
- Department of Oncology, Qingdao Central Hospital, The Affiliated Qingdao Central Hospital of Qingdao University, 127 Si Liu South Road, 266042, Qingdao City, China
| |
Collapse
|
35
|
Walma MS, Brada LJ, Patuleia SIS, Blomjous JG, Bollen TL, Bosscha K, Bruijnen RC, Busch OR, Creemers GJ, Daams F, van Dam R, Festen S, Jan de Groot D, Willem de Groot J, Mohammad NH, Hermans JJ, de Hingh IH, Kerver ED, van Leeuwen MS, van der Leij C, Liem MS, van Lienden KP, Los M, de Meijer VE, Meijerink MR, Mekenkamp LJ, Nederend J, Nio CY, Patijn GA, Polée MB, Pruijt JF, Renken NS, Rombouts SJ, Schouten TJ, Stommel MWJ, Verweij ME, de Vos-Geelen J, de Vries JJJ, Vulink A, Wessels FJ, Wilmink JW, van Santvoort HC, Besselink MG, Molenaar IQ. Treatment strategies and clinical outcomes in consecutive patients with locally advanced pancreatic cancer: A multicenter prospective cohort. Eur J Surg Oncol 2020; 47:699-707. [PMID: 33280952 DOI: 10.1016/j.ejso.2020.11.137] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/10/2020] [Accepted: 11/21/2020] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Since current studies on locally advanced pancreatic cancer (LAPC) mainly report from single, high-volume centers, it is unclear if outcomes can be translated to daily clinical practice. This study provides treatment strategies and clinical outcomes within a multicenter cohort of unselected patients with LAPC. MATERIALS AND METHODS Consecutive patients with LAPC according to Dutch Pancreatic Cancer Group criteria, were prospectively included in 14 centers from April 2015 until December 2017. A centralized expert panel reviewed response according to RECIST v1.1 and potential surgical resectability. Primary outcome was median overall survival (mOS), stratified for primary treatment strategy. RESULTS Overall, 422 patients were included, of whom 77% (n = 326) received chemotherapy. The majority started with FOLFIRINOX (77%, 252/326) with a median of six cycles (IQR 4-10). Gemcitabine monotherapy was given to 13% (41/326) of patients and nab-paclitaxel/gemcitabine to 10% (33/326), with a median of two (IQR 3-5) and three (IQR 3-5) cycles respectively. The mOS of the entire cohort was 10 months (95%CI 9-11). In patients treated with FOLFIRINOX, gemcitabine monotherapy, or nab-paclitaxel/gemcitabine, mOS was 14 (95%CI 13-15), 9 (95%CI 8-10), and 9 months (95%CI 8-10), respectively. A resection was performed in 13% (32/252) of patients after FOLFIRINOX, resulting in a mOS of 23 months (95%CI 12-34). CONCLUSION This multicenter unselected cohort of patients with LAPC resulted in a 14 month mOS and a 13% resection rate after FOLFIRINOX. These data put previous results in perspective, enable us to inform patients with more accurate survival numbers and will support decision-making in clinical practice.
Collapse
Affiliation(s)
- Marieke S Walma
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands; Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Lilly J Brada
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands; Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Susana I S Patuleia
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | | | - Thomas L Bollen
- Dept. of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - Koop Bosscha
- Dept. of Surgery, Jeroen Bosch Hospital, 's-Hertogenbosch, the Netherlands
| | - Rutger C Bruijnen
- Dept. of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - Olivier R Busch
- Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Geert-Jan Creemers
- Dept. of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | - Freek Daams
- Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ronald van Dam
- Dept. of Surgery, Maastricht UMC+, Maastricht, the Netherlands
| | | | - Derk Jan de Groot
- Dept. of Medical Oncology, UMC Groningen, Groningen, the Netherlands
| | | | - Nadia Haj Mohammad
- Dept. of Medical Oncology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - John J Hermans
- Dept. of Radiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Emile D Kerver
- Dept. of Medical Oncology, OLVG, Amsterdam, the Netherlands
| | - Maarten S van Leeuwen
- Dept. of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | | | - Mike S Liem
- Dept. of Surgery, Medical Spectrum Twente, Enschede, the Netherlands
| | - Krijn P van Lienden
- Dept. of Radiology Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amstrdam, the Netherlands
| | - Maartje Los
- Dept. of Medical Oncology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | | | - Martijn R Meijerink
- Dept. of Radiology Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amstrdam, the Netherlands
| | - Leonie J Mekenkamp
- Dept. of Medical Oncology, Medical Spectrum Twente, Enschede, the Netherlands
| | - Joost Nederend
- Dept. of Radiology, Catharina Hospital, Eindhoven, the Netherlands
| | - C Yung Nio
- Dept. of Radiology Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amstrdam, the Netherlands
| | - Gijs A Patijn
- Dept. of Surgery, Isala Clinics, Zwolle, the Netherlands
| | - Marco B Polée
- Dept. of Medical Oncology, Medical Center Leeuwarden, Leeuwarden, the Netherlands
| | - Johannes F Pruijt
- Dept. of Medical Oncology, Jeroen Bosch Hospital, 's-Hertogenbosch, the Netherlands
| | - Nomdo S Renken
- Dept. of Radiology, Reinier de Graaf Hospital, Delft, the Netherlands
| | - Steffi J Rombouts
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands; Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Thijs J Schouten
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - Martijn W J Stommel
- Dept. of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Maaike E Verweij
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - Judith de Vos-Geelen
- Dept. of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, the Netherlands
| | - Jan J J de Vries
- Dept. of Radiology Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amstrdam, the Netherlands
| | - Annelie Vulink
- Dept. of Medical Oncology, Reinier de Graaf Hospital, Delft, the Netherlands
| | - Frank J Wessels
- Dept. of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - Johanna W Wilmink
- Dept. of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam; the Netherlands
| | - Hjalmar C van Santvoort
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | - Marc G Besselink
- Dept. of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - I Quintus Molenaar
- Dept. of Surgery, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
36
|
Salinas-Miranda E, Khalvati F, Namdar K, Deniffel D, Dong X, Abbas E, Wilson JM, O'Kane GM, Knox J, Gallinger S, Haider MA. Validation of Prognostic Radiomic Features From Resectable Pancreatic Ductal Adenocarcinoma in Patients With Advanced Disease Undergoing Chemotherapy. Can Assoc Radiol J 2020; 72:605-613. [PMID: 33151087 DOI: 10.1177/0846537120968782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Radiomic features in pancreatic ductal adenocarcinoma (PDAC) often lack validation in independent test sets or are limited to early or late stage disease. Given the lethal nature of PDAC it is possible that there are similarities in radiomic features of both early and advanced disease reflective of aggressive biology. PURPOSE To assess the performance of prognostic radiomic features previously published in patients with resectable PDAC in a test set of patients with unresectable PDAC undergoing chemotherapy. METHODS The pre-treatment CT of 108 patients enrolled in a prospective chemotherapy trial were used as a test cohort for 2 previously published prognostic radiomic features in resectable PDAC (Sum Entropy and Cluster Tendency with square-root filter[Sqrt]). We assessed the performance of these 2 radiomic features for the prediction of overall survival (OS) and time to progression (TTP) using Cox proportional-hazard models. RESULTS Sqrt Cluster Tendency was significantly associated with outcome with a hazard ratio (HR) of 1.27(for primary pancreatic tumor plus local nodes), (Confidence Interval(CI):1.01 -1.6, P-value = 0.039) for OS and a HR of 1.25(CI:1.00 -1.55, P-value = 0.047) for TTP. Sum entropy was not associated with outcomes. Sqrt Cluster Tendency remained significant in multivariate analysis. CONCLUSION The CT radiomic feature Sqrt Cluster Tendency, previously demonstrated to be prognostic in resectable PDAC, remained a significant prognostic factor for OS and TTP in a test set of unresectable PDAC patients. This radiomic feature warrants further investigation to understand its biologic correlates and CT applicability in PDAC patients.
Collapse
Affiliation(s)
- Emmanuel Salinas-Miranda
- 90755Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, Toronto, Ontario, Canada.,PanCuRx Translational Research Initiative, 90755Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Farzad Khalvati
- 90755Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, Toronto, Ontario, Canada.,Joint Department of Medical Imaging, University Health Network/Sinai Health System, Toronto, Ontario, Canada
| | - Kashayar Namdar
- 90755Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, Toronto, Ontario, Canada
| | - Dominik Deniffel
- 90755Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, Toronto, Ontario, Canada
| | - Xin Dong
- 90755Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, Toronto, Ontario, Canada
| | - Engy Abbas
- Joint Department of Medical Imaging, University Health Network/Sinai Health System, Toronto, Ontario, Canada
| | - Julie M Wilson
- PanCuRx Translational Research Initiative, 90755Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Grainne M O'Kane
- PanCuRx Translational Research Initiative, 90755Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jennifer Knox
- PanCuRx Translational Research Initiative, 90755Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, 90755Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Hepatobiliary Pancreatic Surgical Oncology Program, University Health Network, Toronto, Ontario, Canada
| | - Masoom A Haider
- 90755Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, Toronto, Ontario, Canada.,PanCuRx Translational Research Initiative, 90755Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Joint Department of Medical Imaging, University Health Network/Sinai Health System, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Feng P, Wang ZD, Fan W, Liu H, Pan JJ. Diagnostic advances of artificial intelligence and radiomics in gastroenterology. Artif Intell Gastroenterol 2020; 1:37-50. [DOI: 10.35712/aig.v1.i2.37] [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: 05/27/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Traditional medical imaging, including ultrasound, computed tomography, magnetic resonance imaging, or positron emission tomography, remains widely used diagnostic modalities for gastrointestinal diseases at present. These modalities are used to assess changes in morphology, attenuation, signal intensity, and enhancement characteristics. Gastrointestinal tumors, especially malignant tumors, are commonly seen in clinical practice with an increasing number of deaths each year. Because the imaging manifestations of different diseases usually overlap, accurate early diagnosis of tumor lesions, noninvasive and effective evaluation of tumor staging, and prediction of prognosis remain challenging. Fortunately, traditional medical images contain a great deal of important information that cannot be recognized by human eyes but can be extracted by artificial intelligence (AI) technology, which can quantitatively assess the heterogeneity of lesions and provide valuable information, including therapeutic effects and patient prognosis. With the development of computer technology, the combination of medical imaging and AI technology is considered to represent a promising field in medical image analysis. This new emerging field is called “radiomics”, which makes big data mining and extraction from medical imagery possible and can help clinicians make effective decisions and develop personalized treatment plans. Recently, AI and radiomics have been gradually applied to lesion detection, qualitative and quantitative diagnosis, histopathological grading and staging of tumors, therapeutic efficacy assessment, and prognosis evaluation. In this minireview, we briefly introduce the basic principles and technology of radiomics. Then, we review the research and application of AI and radiomics in gastrointestinal diseases, especially diagnostic advancements of radiomics in the differential diagnosis, treatment option, assessment of therapeutic efficacy, and prognosis evaluation of esophageal, gastric, hepatic, pancreatic, and colorectal diseases.
Collapse
Affiliation(s)
- Pei Feng
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Zhen-Dong Wang
- Department of Ultrasound, Beijing Sihui Hospital of Traditional Chinese Medicine, Beijing 100022, China
| | - Wei Fan
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Jing-Jing Pan
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| |
Collapse
|
38
|
Chu LC, Park S, Kawamoto S, Yuille AL, Hruban RH, Fishman EK. Pancreatic Cancer Imaging: A New Look at an Old Problem. Curr Probl Diagn Radiol 2020; 50:540-550. [PMID: 32988674 DOI: 10.1067/j.cpradiol.2020.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022]
Abstract
Computed tomography is the most commonly used imaging modality to detect and stage pancreatic cancer. Previous advances in pancreatic cancer imaging have focused on optimizing image acquisition parameters and reporting standards. However, current state-of-the-art imaging approaches still misdiagnose some potentially curable pancreatic cancers and do not provide prognostic information or inform optimal management strategies beyond stage. Several recent developments in pancreatic cancer imaging, including artificial intelligence and advanced visualization techniques, are rapidly changing the field. The purpose of this article is to review how these recent advances have the potential to revolutionize pancreatic cancer imaging.
Collapse
Affiliation(s)
- Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Seyoun Park
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alan L Yuille
- Department of Computer Science, Johns Hopkins University, Baltimore, MD
| | - Ralph H Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
39
|
Miyata T, Hayashi H, Yamashita YI, Matsumura K, Nakao Y, Itoyama R, Yamao T, Tsukamoto M, Okabe H, Imai K, Chikamoto A, Ishiko T, Baba H. Prognostic Value of the Preoperative Tumor Marker Index in Resected Pancreatic Ductal Adenocarcinoma: A Retrospective Single-Institution Study. Ann Surg Oncol 2020; 28:1572-1580. [PMID: 32804325 DOI: 10.1245/s10434-020-09022-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 07/29/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND The prediction of prognostic outcomes can provide the most suitable strategy for patients with pancreatic ductal adenocarcinoma (PDAC). This study aimed to evaluate the clinical value of the preoperative tumor marker index (pre-TI) in predicting prognostic outcomes after resection for PDAC. METHODS For 183 patients who underwent pancreatic resection of PDAC, adjusted carbohydrate antigen 19-9 (CA19-9), carcinoembryonic antigen (CEA), pancreatic cancer-associated antigen-2 (DUpan-2), and s-pancreas-1 antigen (SPan-1) were retrospectively evaluated, and the positive number of these markers was scored as the pre-TI. RESULTS A high pre-TI (≥ 2) was significantly associated with a larger tumor and lymph node metastases, and the patients with a high pre-TI had worse prognostic outcomes in terms of both relapse-free survival (RFS) (P < 0.0001, log-rank) and overall survival (OS) (P < 0.0001, Λlog-rank) than the patients with a low pre-TI. The pre-TI was one of the independent factors of a poor prognosis for RFS (hazard ratio [HR], 2.36; P < 0.0001) and OS (HR, 2.27; P < 0.0001). In addition, even for the patients with normal adjusted CA19-9 values (n = 74, 40.4%), those with the high pre-TI had a significantly poorer prognosis than those with a low pre-TI (RFS: P = 0.002, log-rank; OS: P = 0.031, log-rank). CONCLUSIONS The pre-TI could be a potent predictive marker of prognostic outcomes for patients with resections for PDAC. Patients with a high pre-TI may need additional strategies to improve their prognosis.
Collapse
Affiliation(s)
- Tatsunori Miyata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Nakao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Rumi Itoyama
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takanobu Yamao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masayo Tsukamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirohisa Okabe
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Akira Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatoshi Ishiko
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
40
|
Cheng SH, Liu D, Hou B, Hu Y, Huo L, Xing H, Jin ZY, Xue HD. PET-MR Imaging and MR Texture Analysis in the Diagnosis of Pancreatic Cysts: A Prospective Preliminary Study. Acad Radiol 2020; 27:996-1005. [PMID: 31606313 DOI: 10.1016/j.acra.2019.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/24/2019] [Accepted: 09/02/2019] [Indexed: 02/05/2023]
Abstract
RATIONALE AND OBJECTIVES Our aim was to evaluate the capability of textural and metabolic parameters measured at pretreatment 18F-fluorodeoxyglucose Positron emission tomography (PET)-MR in differentiating malignant from benign pancreatic cystic lesions. MATERIALS AND METHOD Forty consecutive patients were prospectively enrolled in this study. They underwent simultaneous PET-MR for the diagnosis of pancreatic cysts. Thirty texture parameters were extracted from manually contoured axial T2-weighted imaging with fat suppression (T2FS) and apparent diffusion coefficient images, respectively. Maximal and mean standardized uptake values (SUVmax and SUVmean, respectively) of pancreatic cysts were measured at PET-MR imaging. The Mann-Whitney test was used to compare both textural and metabolic parameters between benign and malignant group. RESULTS FDG uptake was significantly higher in patients with malignant pancreatic cysts (SUVmaxp = 0.002, SUVmeanp < 0.001). Malignant cysts showed significantly lower standard deviation for spatial scaling factor at 3-6mm on T2FS images and lower skewness for spatial scaling factor at 2-4mm on apparent diffusion coefficient images (p < 0.01). SUVmean had the highest Area under the curve of 0.892 on receiver-operating characteristic analysis with a sensitivity, specificity, and accuracy of 88.9%, 87.1%, and 87.6%, respectively. When metabolic and textural features were combined into a single diagnostic model, the AUC increased to 0.961, with a sensitivity, specificity, and accuracy of 88.9%, 96.8%, and 95.0%, respectively. CONCLUSION Our study implied that PET-MR showed no obvious advantages over traditional PET-related imaging in differentiating malignant from benign pancreatic cystic lesions. Diagnostic model based on the combination of metabolic and textural parameters showed satisfactory performance.
Collapse
Affiliation(s)
- Si-Hang Cheng
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Dong Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan No.1, Wangfujing Street, Dongcheng District, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan No.1, Wangfujing Street, Dongcheng District, Beijing, China
| | - Ya Hu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Haiqun Xing
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan No.1, Wangfujing Street, Dongcheng District, Beijing, China
| | - Hua-Dan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan No.1, Wangfujing Street, Dongcheng District, Beijing, China.
| |
Collapse
|
41
|
Chen BB. Artificial intelligence in pancreatic disease. Artif Intell Med Imaging 2020; 1:19-30. [DOI: 10.35711/aimi.v1.i1.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Bang-Bin Chen
- Department of Medical Imaging, National Taiwan University Hospital, Taipei 10016, Taiwan
- Department of Radiology, College of Medicine, National Taiwan University, Taipei 10016, Taiwan
| |
Collapse
|
42
|
Abstract
MRI and MRCP play an important role in the diagnosis of chronic pancreatitis (CP) by imaging pancreatic parenchyma and ducts. MRI/MRCP is more widely used than computed tomography (CT) for mild to moderate CP due to its increased sensitivity for pancreatic ductal and gland changes; however, it does not detect the calcifications seen in advanced CP. Quantitative MR imaging offers potential advantages over conventional qualitative imaging, including simplicity of analysis, quantitative and population-based comparisons, and more direct interpretation of detected changes. These techniques may provide quantitative metrics for determining the presence and severity of acinar cell loss and aid in the diagnosis of chronic pancreatitis. Given the fact that the parenchymal changes of CP precede the ductal involvement, there would be a significant benefit from developing MRI/MRCP-based, more robust diagnostic criteria combining ductal and parenchymal findings. Among cross-sectional imaging modalities, multi-detector CT (MDCT) has been a cornerstone for evaluating chronic pancreatitis (CP) since it is ubiquitous, assesses primary disease process, identifies complications like pseudocyst or vascular thrombosis with high sensitivity and specificity, guides therapeutic management decisions, and provides images with isotropic resolution within seconds. Conventional MDCT has certain limitations and is reserved to provide predominantly morphological (e.g., calcifications, organ size) rather than functional information. The emerging applications of radiomics and artificial intelligence are poised to extend the current capabilities of MDCT. In this review article, we will review advanced imaging techniques by MRI, MRCP, CT, and ultrasound.
Collapse
|
43
|
Identification of Pancreaticoduodenectomy Resection for Pancreatic Head Adenocarcinoma: A Preliminary Study of Radiomics. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:2761627. [PMID: 32377222 PMCID: PMC7182967 DOI: 10.1155/2020/2761627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
Background In a pathological examination of pancreaticoduodenectomy for pancreatic head adenocarcinoma, a resection margin without cancer cells in 1 mm is recognized as R0; a resection margin with cancer cells in 1 mm is recognized as R1. The preoperative identification of R0 and R1 is of great significance for surgical decision and prognosis. We conducted a preliminary radiomics study based on preoperative CT (computer tomography) images to evaluate a resection margin which was R0 or R1. Methods We retrospectively analyzed 258 preoperative CT images of 86 patients (34 cases of R0 and 52 cases of R1) who were diagnosed as pancreatic head adenocarcinoma and underwent pancreaticoduodenectomy. The radiomics study consists of five stages: (i) delineate and segment regions of interest (ROIs); (ii) by solving discrete Laplacian equations with Dirichlet boundary conditions, fit the ROIs to rectangular regions; (iii) enhance the textures of the fitted ROIs combining wavelet transform and fractional differential; (iv) extract texture features from the enhanced ROIs combining wavelet transform and statistical analysis methods; and (v) reduce features using principal component analysis (PCA) and classify the resection margins using the support vector machine (SVM), and then investigate the associations between texture features and histopathological characteristics using the Mann-Whitney U-test. To reduce overfitting, the SVM classifier embedded a linear kernel and adopted the leave-one-out cross-validation. Results It achieved an AUC (area under receiver operating characteristic curve) of 0.8614 and an accuracy of 84.88%. Setting p ≤ 0.01 in the Mann-Whitney U-test, two features of the run-length matrix, which are derived from diagonal sub-bands in wavelet decomposition, showed statistically significant differences between R0 and R1. Conclusions It indicates that the radiomics study is rewarding for the aided diagnosis of R0 and R1. Texture features can potentially enhance physicians' diagnostic ability.
Collapse
|