1
|
Dogru GD, Tugcu AO, Dursun CU. Enhancing diagnostic frameworks in pancreatic cancer imaging: A critical appraisal. World J Radiol 2025; 17:104818. [PMID: 40176956 PMCID: PMC11959621 DOI: 10.4329/wjr.v17.i3.104818] [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: 01/07/2025] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
This letter to the editor critically appraises the study by Luo et al. While the study provides valuable insights into imaging-pathology correlations in pancreatic cancer, we identify several opportunities for enhancing its clinical relevance. Notably, the exclusion of magnetic resonance cholangiopancreatography and positron emission tomography/computed tomography imaging limits the study's diagnostic scope, as these modalities offer superior capabilities in differentiating benign from malignant lesions and assessing metabolic tumor activity. Additionally, the retrospective, cross-sectional design restricts the potential for dynamic insights into disease progression. We also highlight the untapped potential of radiomics-based analyses, which could significantly improve diagnostic accuracy and prognostic assessments. We recommend integrating these advanced imaging modalities, adopting longitudinal study designs, and leveraging radiomics approaches in future research to enhance the diagnostic frameworks in pancreatic cancer imaging.
Collapse
Affiliation(s)
- Galip Dogukan Dogru
- Department of Radiation Oncology, Gulhane Training and Research Hospital, Ankara 06010, Türkiye
| | - Ahmet Oguz Tugcu
- Department of Radiation Oncology, Gulhane Training and Research Hospital, Ankara 06010, Türkiye
| | - Cemal Ugur Dursun
- Department of Radiation Oncology, Kartal Dr. Lutfi Kirdar City Hospital, İstanbul 34865, Türkiye
| |
Collapse
|
2
|
Vitale F, Zileri Dal Verme L, Paratore M, Negri M, Nista EC, Ainora ME, Esposto G, Mignini I, Borriello R, Galasso L, Alfieri S, Gasbarrini A, Zocco MA, Nicoletti A. The Past, Present, and Future of Biomarkers for the Early Diagnosis of Pancreatic Cancer. Biomedicines 2024; 12:2840. [PMID: 39767746 PMCID: PMC11673965 DOI: 10.3390/biomedicines12122840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/30/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Pancreatic cancer is one of the most aggressive cancers with a very poor 5-year survival rate and reduced therapeutic options when diagnosed in an advanced stage. The dismal prognosis of pancreatic cancer has guided significant efforts to discover novel biomarkers in order to anticipate diagnosis, increasing the population of patients who can benefit from curative surgical treatment. CA 19-9 is the reference biomarker that supports the diagnosis and guides the response to treatments. However, it has significant limitations, a low specificity, and is inefficient as a screening tool. Several potential biomarkers have been discovered in the serum, urine, feces, and pancreatic juice of patients. However, most of this evidence needs further validation in larger cohorts. The advent of advanced omics sciences and liquid biopsy techniques has further enhanced this field of research. The aim of this review is to analyze the historical evolution of the research on novel biomarkers for the early diagnosis of pancreatic cancer, focusing on the current evidence for the most promising biomarkers from different body fluids and the novel trends in research, such as omics sciences and liquid biopsy, in order to favor the application of modern personalized medicine.
Collapse
Affiliation(s)
- 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Enrico Celestino Nista
- 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Maria Elena Ainora
- 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Raffaele Borriello
- 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - Linda Galasso
- 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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;
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| | - 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; (F.V.); (L.Z.D.V.); (M.P.); (M.N.); (E.C.N.); (M.E.A.); (G.E.); (I.M.); (R.B.); (L.G.); (A.G.); (A.N.)
| |
Collapse
|
3
|
Zaccaria GM, Berloco F, Buongiorno D, Brunetti A, Altini N, Bevilacqua V. A time-dependent explainable radiomic analysis from the multi-omic cohort of CPTAC-Pancreatic Ductal Adenocarcinoma. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108408. [PMID: 39342876 DOI: 10.1016/j.cmpb.2024.108408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND AND OBJECTIVE In Pancreatic Ductal Adenocarcinoma (PDA), multi-omic models are emerging to answer unmet clinical needs to derive novel quantitative prognostic factors. We realized a pipeline that relies on survival machine-learning (SML) classifiers and explainability based on patients' follow-up (FU) to stratify prognosis from the public-available multi-omic datasets of the CPTAC-PDA project. MATERIALS AND METHODS Analyzed datasets included tumor-annotated radiologic images, clinical, and mutational data. A feature selection was based on univariate (UV) and multivariate (MV) survival analyses according to Overall Survival (OS) and recurrence (REC). In this study, we considered seven multi-omic datasets and compared four SML classifiers: Cox, survival random forest, generalized boosted, and support vector machines (SVM). For each classifier, we assessed the concordance (C) index on the validation set. The best classifiers for the validation set on both OS and REC underwent explainability analyses using SurvSHAP(t), which extends SHapley Additive exPlanations (SHAP). RESULTS According to OS, after UV and MV analyses we selected 18/37 and 10/37 multi-omic features, respectively. According to REC, based on UV and MV analyses we selected 10/35 and 5/35 determinants, respectively. Generally, SML classifiers including radiomics outperformed those modelled on clinical or mutational predictors. For OS, the Cox model encompassing radiomic, clinical, and mutational features reached 75 % of C index, outperforming other classifiers. On the other hand, for REC, the SVM model including only radiomics emerged as the best-performing, with 68 % of C index. For OS, SurvSHAP(t) identified the first order Median Gray Level (GL) intensities, the gender, the tumor grade, the Joint Energy GL Co-occurrence Matrix (GLCM), and the GLCM Informational Measures of Correlations of type 1 as the most important features. For REC, the first order Median GL intensities, the GL size zone matrix Small Area Low GL Emphasis, and first order variance of GL intensities emerged as the most discriminative. CONCLUSIONS In this work, radiomics showed the potential for improving patients' risk stratification in PDA. Furthermore, a deeper understanding of how radiomics can contribute to prognosis in PDA was achieved with a time-dependent explainability of the top multi-omic predictors.
Collapse
Affiliation(s)
- Gian Maria Zaccaria
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona, 4, Bari, 70126, Italy
| | - Francesco Berloco
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona, 4, Bari, 70126, Italy.
| | - Domenico Buongiorno
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona, 4, Bari, 70126, Italy; Apulian Bioengineering srl, Via delle Violette, 14, Modugno, 70026, Italy
| | - Antonio Brunetti
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona, 4, Bari, 70126, Italy; Apulian Bioengineering srl, Via delle Violette, 14, Modugno, 70026, Italy
| | - Nicola Altini
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona, 4, Bari, 70126, Italy
| | - Vitoantonio Bevilacqua
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona, 4, Bari, 70126, Italy; Apulian Bioengineering srl, Via delle Violette, 14, Modugno, 70026, Italy
| |
Collapse
|
4
|
Jiang L, Ye Y, Feng Z, Liu W, Cao Y, Zhao X, Zhu X, Zhang H. Stereotactic body radiation therapy for the primary tumor and oligometastases versus the primary tumor alone in patients with metastatic pancreatic cancer. Radiat Oncol 2024; 19:111. [PMID: 39160547 PMCID: PMC11334573 DOI: 10.1186/s13014-024-02493-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/19/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Local therapies may benefit patients with oligometastatic cancer. However, there were limited data about pancreatic cancer. Here, we compared the efficacy and safety of stereotactic body radiation therapy (SBRT) to the primary tumor and all oligometastases with SBRT to the primary tumor alone in patients with metastatic pancreatic cancer. METHODS A retrospective review of patients with synchronous oligometastatic pancreatic cancer (up to 5 lesions) receiving SBRT to all lesions (including all oligometastases and the primary tumor) were performed. Another comparable group of patients with similar baseline characteristics, including metastatic burden, SBRT doses, and chemotherapy regimens, receiving SBRT to the primary tumor alone were identified. The primary endpoint was overall survival (OS). The secondary endpoints were progression frees survival (PFS), polyprogression free survival (PPFS) and adverse events. RESULTS There were 59 and 158 patients receiving SBRT to all lesions and to the primary tumor alone. The median OS of patients with SBRT to all lesions and the primary tumor alone was 10.9 months (95% CI 10.2-11.6 months) and 9.3 months (95% CI 8.8-9.8 months) (P < 0.001). The median PFS of two groups was 6.5 months (95% CI 5.6-7.4 months) and 4.1 months (95% CI 3.8-4.4 months) (P < 0.001). The median PPFS of two groups was 9.8 months (95% CI 8.9-10.7 months) and 7.8 months (95% CI 7.2-8.4 months) (P < 0.001). Additionally, 14 (23.7%) and 32 (20.2%) patients in two groups had grade 3 or 4 treatment-related toxicity. CONCLUSIONS SBRT to all oligometastases and the primary tumor in patients with pancreatic cancer may improve survival, which needs prospective verification.
Collapse
Affiliation(s)
- Lingong Jiang
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yusheng Ye
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhiru Feng
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Wenyu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Changhai Hospital affiliated to Naval Medical University, Shanghai, China
| | - Yangsen Cao
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xianzhi Zhao
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaofei Zhu
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
5
|
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
|
6
|
Jiang W, Pan X, Luo Q, Huang S, Liang Y, Zhong X, Zhang X, Deng W, Lv Y, Chen L. Radiomics analysis of pancreas based on dual-energy computed tomography for the detection of type 2 diabetes mellitus. Front Med (Lausanne) 2024; 11:1328687. [PMID: 38707184 PMCID: PMC11069320 DOI: 10.3389/fmed.2024.1328687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/03/2024] [Indexed: 05/07/2024] Open
Abstract
Objective To utilize radiomics analysis on dual-energy CT images of the pancreas to establish a quantitative imaging biomarker for type 2 diabetes mellitus. Materials and methods In this retrospective study, 78 participants (45 with type 2 diabetes mellitus, 33 without) underwent a dual energy CT exam. Pancreas regions were segmented automatically using a deep learning algorithm. From these regions, radiomics features were extracted. Additionally, 24 clinical features were collected for each patient. Both radiomics and clinical features were then selected using the least absolute shrinkage and selection operator (LASSO) technique and then build classifies with random forest (RF), support vector machines (SVM) and Logistic. Three models were built: one using radiomics features, one using clinical features, and a combined model. Results Seven radiomic features were selected from the segmented pancreas regions, while eight clinical features were chosen from a pool of 24 using the LASSO method. These features were used to build a combined model, and its performance was evaluated using five-fold cross-validation. The best classifier type is Logistic and the reported area under the curve (AUC) values on the test dataset were 0.887 (0.73-1), 0.881 (0.715-1), and 0.922 (0.804-1) for the respective models. Conclusion Radiomics analysis of the pancreas on dual-energy CT images offers potential as a quantitative imaging biomarker in the detection of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Radiology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Xianpan Pan
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Qunzhi Luo
- Department of Radiology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Shiqi Huang
- Department of Radiology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Yuhong Liang
- Department of Radiology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Xixi Zhong
- Department of Radiology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Xianjie Zhang
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Wei Deng
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Yaping Lv
- Department of Radiology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Lei Chen
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| |
Collapse
|
7
|
Grewal M, Ahmed T, Javed AA. Current state of radiomics in hepatobiliary and pancreatic malignancies. ARTIFICIAL INTELLIGENCE SURGERY 2023; 3:217-32. [DOI: 10.20517/ais.2023.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Rising in incidence, hepatobiliary and pancreatic (HPB) cancers continue to exhibit dismal long-term survival. The overall poor prognosis of HPB cancers is reflective of the advanced stage at which most patients are diagnosed. Late diagnosis is driven by the often-asymptomatic nature of these diseases, as well as a dearth of screening modalities. Additionally, standard imaging modalities fall short of providing accurate and detailed information regarding specific tumor characteristics, which can better inform surgical planning and sequencing of systemic therapy. Therefore, precise therapeutic planning must be delayed until histopathological examination is performed at the time of resection. Given the current shortcomings in the management of HPB cancers, investigations of numerous noninvasive biomarkers, including circulating tumor cells and DNA, proteomics, immunolomics, and radiomics, are underway. Radiomics encompasses the extraction and analysis of quantitative imaging features. Along with summarizing the general framework of radiomics, this review synthesizes the state of radiomics in HPB cancers, outlining its role in various aspects of management, present limitations, and future applications for clinical integration. Current literature underscores the utility of radiomics in early detection, tumor characterization, therapeutic selection, and prognostication for HPB cancers. Seeing as single-center, small studies constitute the majority of radiomics literature, there is considerable heterogeneity with respect to steps of the radiomics workflow such as segmentation, or delineation of the region of interest on a scan. Nonetheless, the introduction of the radiomics quality score (RQS) demonstrates a step towards greater standardization and reproducibility in the young field of radiomics. Altogether, in the setting of continually improving artificial intelligence algorithms, radiomics represents a promising biomarker avenue for promoting enhanced and tailored management of HPB cancers, with the potential to improve long-term outcomes for patients.
Collapse
|
8
|
Ramaekers M, Viviers CGA, Janssen BV, Hellström TAE, Ewals L, van der Wulp K, Nederend J, Jacobs I, Pluyter JR, Mavroeidis D, van der Sommen F, Besselink MG, Luyer MDP. Computer-Aided Detection for Pancreatic Cancer Diagnosis: Radiological Challenges and Future Directions. J Clin Med 2023; 12:4209. [PMID: 37445243 PMCID: PMC10342462 DOI: 10.3390/jcm12134209] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Radiological imaging plays a crucial role in the detection and treatment of pancreatic ductal adenocarcinoma (PDAC). However, there are several challenges associated with the use of these techniques in daily clinical practice. Determination of the presence or absence of cancer using radiological imaging is difficult and requires specific expertise, especially after neoadjuvant therapy. Early detection and characterization of tumors would potentially increase the number of patients who are eligible for curative treatment. Over the last decades, artificial intelligence (AI)-based computer-aided detection (CAD) has rapidly evolved as a means for improving the radiological detection of cancer and the assessment of the extent of disease. Although the results of AI applications seem promising, widespread adoption in clinical practice has not taken place. This narrative review provides an overview of current radiological CAD systems in pancreatic cancer, highlights challenges that are pertinent to clinical practice, and discusses potential solutions for these challenges.
Collapse
Affiliation(s)
- Mark Ramaekers
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands;
| | - Christiaan G. A. Viviers
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands; (C.G.A.V.); (T.A.E.H.); (F.v.d.S.)
| | - Boris V. Janssen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (B.V.J.); (M.G.B.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Terese A. E. Hellström
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands; (C.G.A.V.); (T.A.E.H.); (F.v.d.S.)
| | - Lotte Ewals
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (L.E.); (K.v.d.W.); (J.N.)
| | - Kasper van der Wulp
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (L.E.); (K.v.d.W.); (J.N.)
| | - Joost Nederend
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands; (L.E.); (K.v.d.W.); (J.N.)
| | - Igor Jacobs
- Department of Hospital Services and Informatics, Philips Research, 5656 AE Eindhoven, The Netherlands;
| | - Jon R. Pluyter
- Department of Experience Design, Philips Design, 5656 AE Eindhoven, The Netherlands;
| | - Dimitrios Mavroeidis
- Department of Data Science, Philips Research, 5656 AE Eindhoven, The Netherlands;
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands; (C.G.A.V.); (T.A.E.H.); (F.v.d.S.)
| | - Marc G. Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (B.V.J.); (M.G.B.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Misha D. P. Luyer
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands;
| |
Collapse
|
9
|
Taha A, Taha-Mehlitz S, Ortlieb N, Ochs V, Honaker MD, Rosenberg R, Lock JF, Bolli M, Cattin PC. Machine learning in pancreas surgery, what is new? literature review. Front Surg 2023; 10:1142585. [PMID: 37383385 PMCID: PMC10293756 DOI: 10.3389/fsurg.2023.1142585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/19/2023] [Indexed: 06/30/2023] Open
Abstract
Background Machine learning (ML) is an inquiry domain that aims to establish methodologies that leverage information to enhance performance of various applications. In the healthcare domain, the ML concept has gained prominence over the years. As a result, the adoption of ML algorithms has become expansive. The aim of this scoping review is to evaluate the application of ML in pancreatic surgery. Methods We integrated the preferred reporting items for systematic reviews and meta-analyses for scoping reviews. Articles that contained relevant data specializing in ML in pancreas surgery were included. Results A search of the following four databases PubMed, Cochrane, EMBASE, and IEEE and files adopted from Google and Google Scholar was 21. The main features of included studies revolved around the year of publication, the country, and the type of article. Additionally, all the included articles were published within January 2019 to May 2022. Conclusion The integration of ML in pancreas surgery has gained much attention in previous years. The outcomes derived from this study indicate an extensive literature gap on the topic despite efforts by various researchers. Hence, future studies exploring how pancreas surgeons can apply different learning algorithms to perform essential practices may ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Anas Taha
- Department of Biomedical Engineering, Faculty of Medicine, University of Basel, Allschwil, Switzerland
| | - Stephanie Taha-Mehlitz
- Clarunis, Department of Visceral Surgery, University Center for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital, Basel, Switzerland
| | - Niklas Ortlieb
- Goethe University Frankfurt, Faculty of Business and Economics, Frankfurt am Main, Germany
| | - Vincent Ochs
- Department of Biomedical Engineering, Faculty of Medicine, University of Basel, Allschwil, Switzerland
| | - Michael Drew Honaker
- Department of Surgery, East Carolina University, Brody School of Medicine, Greenville, NC, United States
| | - Robert Rosenberg
- Cantonal Hospital Basel-Landschaft, Centre for Gastrointestinal and Liver Diseases, Liestal, Switzerland
| | - Johan F. Lock
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Martin Bolli
- Clarunis, Department of Visceral Surgery, University Center for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital, Basel, Switzerland
| | - Philippe C. Cattin
- Department of Biomedical Engineering, Faculty of Medicine, University of Basel, Allschwil, Switzerland
| |
Collapse
|
10
|
Jia X, Liang J, Ma X, Wang W, Lai C. Radiomic-based machine learning model for predicting the surgical risk in children with abdominal neuroblastoma. WORLD JOURNAL OF PEDIATRIC SURGERY 2023; 6:e000531. [PMID: 37223779 PMCID: PMC10201264 DOI: 10.1136/wjps-2022-000531] [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: 11/18/2022] [Accepted: 03/27/2023] [Indexed: 05/25/2023] Open
Abstract
Background Preoperative imaging assessment of surgical risk is very important for the prognosis of these children. To develop and validate a radiomics-based machine learning model based on the analysis of radiomics features to predict surgical risk in children with abdominal neuroblastoma (NB). Methods A retrospective study was conducted from April 2019 to March 2021 among 74 children with abdominal NB. A total of 1874 radiomic features in MR images were extracted from each patient. Support vector machines (SVMs) were used to establish the model. Eighty percent of the data were used as the training set to optimize the model, and 20% of the data were used to validate its accuracy, sensitivity, specificity and area under the curve (AUC) to verify its effectiveness. Results Among the 74 children with abdominal NB, 55 (65%) had surgical risk and 19 (35%) had no surgical risk. A t test and Lasso identified that 28 radiomic features were associated with surgical risk. After developing an SVM-based model using these features, predictions were made about whether children with abdominal NB had surgical risk. The model achieved an AUC of 0.94 (a sensitivity of 0.83 and a specificity of 0.80) with 0.890 accuracy in the training set and an AUC of 0.81 (a sensitivity of 0.73 and a specificity of 0.82) with 0.838 accuracy in the test set. Conclusions Radiomics and machine learning can be used to predict the surgical risk in children with abdominal NB. The model based on 28 radiomic features established by SVM showed good diagnostic efficiency.
Collapse
Affiliation(s)
- Xuan Jia
- Department of Radiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiawei Liang
- Department of Radiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohui Ma
- Department of Radiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenqi Wang
- Department of Radiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Can Lai
- Department of Radiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Jan Z, El Assadi F, Abd-Alrazaq A, Jithesh PV. Artificial Intelligence for the Prediction and Early Diagnosis of Pancreatic Cancer: Scoping Review. J Med Internet Res 2023; 25:e44248. [PMID: 37000507 PMCID: PMC10131763 DOI: 10.2196/44248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/21/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Pancreatic cancer is the 12th most common cancer worldwide, with an overall survival rate of 4.9%. Early diagnosis of pancreatic cancer is essential for timely treatment and survival. Artificial intelligence (AI) provides advanced models and algorithms for better diagnosis of pancreatic cancer. OBJECTIVE This study aims to explore AI models used for the prediction and early diagnosis of pancreatic cancers as reported in the literature. METHODS A scoping review was conducted and reported in line with the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews) guidelines. PubMed, Google Scholar, Science Direct, BioRXiv, and MedRxiv were explored to identify relevant articles. Study selection and data extraction were independently conducted by 2 reviewers. Data extracted from the included studies were synthesized narratively. RESULTS Of the 1185 publications, 30 studies were included in the scoping review. The included articles reported the use of AI for 6 different purposes. Of these included articles, AI techniques were mostly used for the diagnosis of pancreatic cancer (14/30, 47%). Radiological images (14/30, 47%) were the most frequently used data in the included articles. Most of the included articles used data sets with a size of <1000 samples (11/30, 37%). Deep learning models were the most prominent branch of AI used for pancreatic cancer diagnosis in the studies, and the convolutional neural network was the most used algorithm (18/30, 60%). Six validation approaches were used in the included studies, of which the most frequently used approaches were k-fold cross-validation (10/30, 33%) and external validation (10/30, 33%). A higher level of accuracy (99%) was found in studies that used support vector machine, decision trees, and k-means clustering algorithms. CONCLUSIONS This review presents an overview of studies based on AI models and algorithms used to predict and diagnose pancreatic cancer patients. AI is expected to play a vital role in advancing pancreatic cancer prediction and diagnosis. Further research is required to provide data that support clinical decisions in health care.
Collapse
Affiliation(s)
- Zainab Jan
- College of Health & Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Farah El Assadi
- College of Health & Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Alaa Abd-Alrazaq
- AI Center for Precision Health, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | |
Collapse
|
12
|
Jan Z, El Assadi F, Abd-alrazaq A, Jithesh PV. Artificial Intelligence for the Prediction and Early Diagnosis of Pancreatic Cancer: Scoping Review (Preprint).. [DOI: 10.2196/preprints.44248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
BACKGROUND
Pancreatic cancer is the 12th most common cancer worldwide, with an overall survival rate of 4.9%. Early diagnosis of pancreatic cancer is essential for timely treatment and survival. Artificial intelligence (AI) provides advanced models and algorithms for better diagnosis of pancreatic cancer.
OBJECTIVE
This study aims to explore AI models used for the prediction and early diagnosis of pancreatic cancers as reported in the literature.
METHODS
A scoping review was conducted and reported in line with the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews) guidelines. PubMed, Google Scholar, Science Direct, BioRXiv, and MedRxiv were explored to identify relevant articles. Study selection and data extraction were independently conducted by 2 reviewers. Data extracted from the included studies were synthesized narratively.
RESULTS
Of the 1185 publications, 30 studies were included in the scoping review. The included articles reported the use of AI for 6 different purposes. Of these included articles, AI techniques were mostly used for the diagnosis of pancreatic cancer (14/30, 47%). Radiological images (14/30, 47%) were the most frequently used data in the included articles. Most of the included articles used data sets with a size of <1000 samples (11/30, 37%). Deep learning models were the most prominent branch of AI used for pancreatic cancer diagnosis in the studies, and the convolutional neural network was the most used algorithm (18/30, 60%). Six validation approaches were used in the included studies, of which the most frequently used approaches were k-fold cross-validation (10/30, 33%) and external validation (10/30, 33%). A higher level of accuracy (99%) was found in studies that used support vector machine, decision trees, and k-means clustering algorithms.
CONCLUSIONS
This review presents an overview of studies based on AI models and algorithms used to predict and diagnose pancreatic cancer patients. AI is expected to play a vital role in advancing pancreatic cancer prediction and diagnosis. Further research is required to provide data that support clinical decisions in health care.
Collapse
|
13
|
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
|
14
|
Salinas-Miranda E, Healy GM, Grünwald B, Jain R, Deniffel D, O'Kane GM, Grant R, Wilson J, Knox J, Gallinger S, Fischer S, Khokha R, Haider MA. Correlation of transcriptional subtypes with a validated CT radiomics score in resectable pancreatic ductal adenocarcinoma. Eur Radiol 2022; 32:6712-6722. [PMID: 36006427 DOI: 10.1007/s00330-022-09057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/14/2022] [Accepted: 07/24/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Transcriptional classifiers (Bailey, Moffitt and Collison) are key prognostic factors of pancreatic ductal adenocarcinoma (PDAC). Among these classifiers, the squamous, basal-like, and quasimesenchymal subtypes overlap and have inferior survival. Currently, only an invasive biopsy can determine these subtypes, possibly resulting in treatment delay. This study aimed to investigate the association between transcriptional subtypes and an externally validated preoperative CT-based radiomic prognostic score (Rad-score). METHODS We retrospectively evaluated 122 patients who underwent resection for PDAC. All treatment decisions were determined at multidisciplinary tumor boards. Tumor Rad-score values from preoperative CT were dichotomized into high or llow categories. The primary endpoint was the correlation between the transcriptional subtypes and the Rad-score using multivariable linear regression, adjusting for clinical and histopathological variables (i.e., tumor size). Prediction of overall survival (OS) was secondary endpoint. RESULTS The Bailey transcriptional classifier significantly associated with the Rad-score (coefficient = 0.31, 95% confidence interval [CI]: 0.13-0.44, p = 0.001). Squamous subtype was associated with high Rad-scores while non-squamous subtype was associated with low Rad-scores (adjusted p = 0.03). Squamous subtype and high Rad-score were both prognostic for OS at multivariable analysis with hazard ratios (HR) of 2.79 (95% CI: 1.12-6.92, p = 0.03) and 4.03 (95% CI: 1.42-11.39, p = 0.01), respectively. CONCLUSIONS In patients with resectable PDAC, an externally validated prognostic radiomic model derived from preoperative CT is associated with the Bailey transcriptional classifier. Higher Rad-scores were correlated with the squamous subtype, while lower Rad-scores were associated with the less lethal subtypes (immunogenic, ADEX, pancreatic progenitor). KEY POINTS • The transcriptional subtypes of PDAC have been shown to have prognostic importance but they require invasive biopsy to be assessed. • The Rad-score radiomic biomarker, which is obtained non-invasively from preoperative CT, correlates with the Bailey squamous transcriptional subtype and both are negative prognostic biomarkers. • The Rad-score is a promising non-invasive imaging biomarker for personalizing neoadjuvant approaches in patients undergoing resection for PDAC, although additional validation studies are required.
Collapse
Affiliation(s)
- Emmanuel Salinas-Miranda
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, 6th Floor, Office 6 200, Toronto, ON, M5G 1X5, Canada.,Joint Department of Medical Imaging, University Health Network/Sinai Health System, 600 University Ave, 5th Floor, Toronto, ON, M5G1X5, Canada
| | - Gerard M Healy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, 6th Floor, Office 6 200, Toronto, ON, M5G 1X5, Canada.,Joint Department of Medical Imaging, University Health Network/Sinai Health System, 600 University Ave, 5th Floor, Toronto, ON, M5G1X5, Canada.,Department of Medical Imaging, University of Toronto, 263 McCaul St 4th Floor, Toronto, ON, M5T 1W5, Canada
| | - Barbara Grünwald
- Department of Pathology, University Health Network, 610 University Ave, Toronto, ON, M5G 2C1, Canada.,PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Rahi Jain
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, 610 University Ave, Toronto, ON, M5G 2C1, Canada
| | - Dominik Deniffel
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, 6th Floor, Office 6 200, Toronto, ON, M5G 1X5, Canada
| | - Grainne M O'Kane
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada.,Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, 610 University Ave, Toronto, ON, M5G 2C1, Canada
| | - Robert Grant
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada.,Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, 610 University Ave, Toronto, ON, M5G 2C1, Canada
| | - Julie Wilson
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada
| | - Jennifer Knox
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada.,Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, 610 University Ave, Toronto, ON, M5G 2C1, Canada
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada.,Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, 610 University Ave, Toronto, ON, M5G 2C1, Canada.,Hepatobiliary Pancreatic Surgical Oncology Program, University Health Network, 190 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Sandra Fischer
- Department of Pathology, University Health Network, 610 University Ave, Toronto, ON, M5G 2C1, Canada
| | - Rama Khokha
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, 610 University Ave, Toronto, ON, M5G 2C1, Canada
| | - Masoom A Haider
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, 6th Floor, Office 6 200, Toronto, ON, M5G 1X5, Canada. .,Joint Department of Medical Imaging, University Health Network/Sinai Health System, 600 University Ave, 5th Floor, Toronto, ON, M5G1X5, Canada. .,Department of Medical Imaging, University of Toronto, 263 McCaul St 4th Floor, Toronto, ON, M5T 1W5, Canada. .,PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, ON, M5G 0A3, Canada.
| |
Collapse
|
15
|
Laino ME, Ammirabile A, Lofino L, Mannelli L, Fiz F, Francone M, Chiti A, Saba L, Orlandi MA, Savevski V. Artificial Intelligence Applied to Pancreatic Imaging: A Narrative Review. Healthcare (Basel) 2022; 10:1511. [PMID: 36011168 PMCID: PMC9408381 DOI: 10.3390/healthcare10081511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 12/19/2022] Open
Abstract
The diagnosis, evaluation, and treatment planning of pancreatic pathologies usually require the combined use of different imaging modalities, mainly, computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). Artificial intelligence (AI) has the potential to transform the clinical practice of medical imaging and has been applied to various radiological techniques for different purposes, such as segmentation, lesion detection, characterization, risk stratification, or prediction of response to treatments. The aim of the present narrative review is to assess the available literature on the role of AI applied to pancreatic imaging. Up to now, the use of computer-aided diagnosis (CAD) and radiomics in pancreatic imaging has proven to be useful for both non-oncological and oncological purposes and represents a promising tool for personalized approaches to patients. Although great developments have occurred in recent years, it is important to address the obstacles that still need to be overcome before these technologies can be implemented into our clinical routine, mainly considering the heterogeneity among studies.
Collapse
Affiliation(s)
- Maria Elena Laino
- Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Angela Ammirabile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Ludovica Lofino
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | | | - Francesco Fiz
- Nuclear Medicine Unit, Department of Diagnostic Imaging, E.O. Ospedali Galliera, 56321 Genoa, Italy
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital, 72074 Tübingen, Germany
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Nuclear Medicine, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09124 Cagliari, Italy
| | | | - Victor Savevski
- Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
16
|
Schuurmans M, Alves N, Vendittelli P, Huisman H, Hermans J. Setting the Research Agenda for Clinical Artificial Intelligence in Pancreatic Adenocarcinoma Imaging. Cancers (Basel) 2022; 14:cancers14143498. [PMID: 35884559 PMCID: PMC9316850 DOI: 10.3390/cancers14143498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, associated with a 98% loss of life expectancy and a 30% increase in disability-adjusted life years. Image-based artificial intelligence (AI) can help improve outcomes for PDAC given that current clinical guidelines are non-uniform and lack evidence-based consensus. However, research on image-based AI for PDAC is too scattered and lacking in sufficient quality to be incorporated into clinical workflows. In this review, an international, multi-disciplinary team of the world’s leading experts in pancreatic cancer breaks down the patient pathway and pinpoints the current clinical touchpoints in each stage. The available PDAC imaging AI literature addressing each pathway stage is then rigorously analyzed, and current performance and pitfalls are identified in a comprehensive overview. Finally, the future research agenda for clinically relevant, image-driven AI in PDAC is proposed. Abstract Pancreatic ductal adenocarcinoma (PDAC), estimated to become the second leading cause of cancer deaths in western societies by 2030, was flagged as a neglected cancer by the European Commission and the United States Congress. Due to lack of investment in research and development, combined with a complex and aggressive tumour biology, PDAC overall survival has not significantly improved the past decades. Cross-sectional imaging and histopathology play a crucial role throughout the patient pathway. However, current clinical guidelines for diagnostic workup, patient stratification, treatment response assessment, and follow-up are non-uniform and lack evidence-based consensus. Artificial Intelligence (AI) can leverage multimodal data to improve patient outcomes, but PDAC AI research is too scattered and lacking in quality to be incorporated into clinical workflows. This review describes the patient pathway and derives touchpoints for image-based AI research in collaboration with a multi-disciplinary, multi-institutional expert panel. The literature exploring AI to address these touchpoints is thoroughly retrieved and analysed to identify the existing trends and knowledge gaps. The results show absence of multi-institutional, well-curated datasets, an essential building block for robust AI applications. Furthermore, most research is unimodal, does not use state-of-the-art AI techniques, and lacks reliable ground truth. Based on this, the future research agenda for clinically relevant, image-driven AI in PDAC is proposed.
Collapse
Affiliation(s)
- Megan Schuurmans
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
- Correspondence: (M.S.); (N.A.)
| | - Natália Alves
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
- Correspondence: (M.S.); (N.A.)
| | - Pierpaolo Vendittelli
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
| | - Henkjan Huisman
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
| | - John Hermans
- Department of Medical Imaging, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands;
| |
Collapse
|
17
|
Eroğlu O, Eroğlu Y, Yıldırım M, Karlıdag T, Çınar A, Akyiğit A, Kaygusuz İ, Yıldırım H, Keleş E, Yalçın Ş. Is it useful to use computerized tomography image-based artificial intelligence modelling in the differential diagnosis of chronic otitis media with and without cholesteatoma? Am J Otolaryngol 2022; 43:103395. [PMID: 35241288 DOI: 10.1016/j.amjoto.2022.103395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/01/2022] [Accepted: 02/13/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Cholesteatoma is an aggressive form of chronic otitis media (COM). For this reason, it is important to distinguish between COM with and without cholesteatoma. In this study, the role of artificial intelligence modelling in differentiating COM with and without cholesteatoma on computed tomography images was evaluated. METHODS The files of 200 patients who underwent mastoidectomy and/or tympanoplasty for COM in our clinic between January 2016 and January 2021 were retrospectively reviewed. According to the presence of cholesteatoma, the patients were divided into two groups as chronic otitis with cholesteatoma (n = 100) and chronic otitis without cholesteatoma (n = 100). The control group (n = 100) consisted of patients who did not have any previous ear disease and did not have any active complaints about the ear. Temporal bone computed tomography (CT) images of all patients were analyzed. The distinction between cholesteatoma and COM was evaluated by using 80% of the CT images obtained for the training of artificial intelligence modelling and the remaining 20% for testing purposes. RESULTS The accuracy rate obtained in the hybrid model we used in our study was 95.4%. The proposed model correctly predicted 2952 out of 3093 CT images, while it predicted 141 incorrectly. It correctly predicted 936 (93.78%) of 998 images in the COM group with cholesteatoma, 835 (92.77%) of 900 images in the COM group without cholesteatoma, and 1181 (98.82%) of 1195 images in the normal group. CONCLUSION In our study, it has been shown that the differentiation of COM with and without cholesteatoma with artificial intelligence modelling can be made with highly accurate diagnosis rates by using CT images. With the deep learning modelling we proposed, the highest correct diagnosis rate in the literature was obtained. According to the results of our study, we think that with the use of artificial intelligence in practice, the diagnosis of cholesteatoma can be made earlier, it will help in the selection of the most appropriate treatment approach, and the complications can be reduced.
Collapse
Affiliation(s)
- Orkun Eroğlu
- Fırat University, School of Medicine, Department of Otorhinolaryngology, Elazig, Turkey.
| | - Yeşim Eroğlu
- Fırat University, School of Medicine, Department of Radiology, Elazig, Turkey
| | - Muhammed Yıldırım
- Malatya Turgut Ozal University, Faculty of Engineering and Natural Sciences, Department of Computer Engineering, Malatya, Turkey
| | - Turgut Karlıdag
- Fırat University, School of Medicine, Department of Otorhinolaryngology, Elazig, Turkey
| | - Ahmet Çınar
- Fırat University, School of Engineering, Department of Computer Engineering, Elazig, Turkey.
| | - Abdulvahap Akyiğit
- Fırat University, School of Medicine, Department of Otorhinolaryngology, Elazig, Turkey
| | - İrfan Kaygusuz
- Fırat University, School of Medicine, Department of Otorhinolaryngology, Elazig, Turkey
| | - Hanefi Yıldırım
- Fırat University, School of Medicine, Department of Radiology, Elazig, Turkey.
| | - Erol Keleş
- Fırat University, School of Medicine, Department of Otorhinolaryngology, Elazig, Turkey
| | - Şinasi Yalçın
- Fırat University, School of Medicine, Department of Otorhinolaryngology, Elazig, Turkey
| |
Collapse
|
18
|
Meng Y, Zhang H, Li Q, Liu F, Fang X, Li J, Yu J, Feng X, Lu J, Bian Y, Shao C. Magnetic Resonance Radiomics and Machine-learning Models: An Approach for Evaluating Tumor-stroma Ratio in Patients with Pancreatic Ductal Adenocarcinoma. Acad Radiol 2022; 29:523-535. [PMID: 34563443 DOI: 10.1016/j.acra.2021.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To develop and validate a magnetic resonance imaging (MRI)-based machine learning classifier for evaluating the tumor-stroma ratio (TSR) in patients with pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS In this retrospective study, 148 patients with PDAC underwent an MR scan and surgical resection. We used hematoxylin and eosin to quantify the TSR. For each patient, we extracted 1,409 radiomics features and reduced them using the least absolute shrinkage and selection operator logistic regression algorithm. The extreme gradient boosting (XGBoost) classifier was developed using a training set comprising 110 consecutive patients, admitted between December 2016 and December 2017. The model was validated in 38 consecutive patients, admitted between January 2018 and April 2018. We determined the performance of the XGBoost classifier based on its discriminative ability, calibration, and clinical utility. RESULTS A log-rank test revealed significantly longer survival in the TSR-low group. The prediction model displayed good discrimination in the training (area under the curve [AUC], 0.82) and validation set (AUC, 0.78). While the sensitivity, specificity, accuracy, positive predictive value, and negative predictive value for the training set were 77.14%, 75.00%, 0.76%, 0.84%, and 0.65%, respectively, those for the validation set were 58.33%, 92.86%, 0.71%, 0.93%, and 0.57%, respectively. CONCLUSION We developed an XGBoost classifier based on MRI radiomics features, a non-invasive prediction tool that can evaluate the TSR of patients with PDAC. Moreover, it will provide a basis for interstitial targeted therapy selection and monitoring.
Collapse
|
19
|
Tharmaseelan H, Hertel A, Tollens F, Rink J, Woźnicki P, Haselmann V, Ayx I, Nörenberg D, Schoenberg SO, Froelich MF. Identification of CT Imaging Phenotypes of Colorectal Liver Metastases from Radiomics Signatures-Towards Assessment of Interlesional Tumor Heterogeneity. Cancers (Basel) 2022; 14:cancers14071646. [PMID: 35406418 PMCID: PMC8997087 DOI: 10.3390/cancers14071646] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Tumoral heterogeneity (TH) is a major challenge in the treatment of metastatic colorectal cancer (mCRC) and is associated with inferior response. Therefore, the identification of TH would be beneficial for treatment planning. TH can be assessed by identifying genetic alterations. In this work, a radiomics-based approach for assessment of TH in colorectal liver metastases (CRLM) in CT scans is demonstrated. (2) Methods: In this retrospective study, CRLM of mCRC were segmented and radiomics features extracted using pyradiomics. Unsupervised k-means clustering was applied to features and lesions. Feature redundancy was evaluated by principal component analysis and reduced by Pearson correlation coefficient cutoff. Feature selection was conducted by LASSO regression and visual analysis of the clusters by radiologists. (3) Results: A total of 47 patients’ (36% female, median age 64) CTs with 261 lesions were included. Five clusters were identified, and the categories small disseminated (n = 31), heterogeneous (n = 105), homogeneous (n = 64), mixed (n = 59), and very large type (n = 2) were assigned based on visual characteristics. Further statistical analysis showed correlation (p < 0.01) of clusters with sex, primary location, T- and N-status, and mutational status. Feature reduction and selection resulted in the identification of four features as a final set for cluster definition. (4) Conclusions: Radiomics features can characterize TH in liver metastases of mCRC in CT scans, and may be suitable for a better pretherapeutic classification of liver lesion phenotypes.
Collapse
Affiliation(s)
- Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Alexander Hertel
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Fabian Tollens
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Johann Rink
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Piotr Woźnicki
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Verena Haselmann
- Institute of Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany;
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Dominik Nörenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Stefan O. Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Matthias F. Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
- Correspondence:
| |
Collapse
|
20
|
Janssen BV, Verhoef S, Wesdorp NJ, Huiskens J, de Boer OJ, Marquering H, Stoker J, Kazemier G, Besselink MG. Imaging-based Machine-learning Models to Predict Clinical Outcomes and Identify Biomarkers in Pancreatic Cancer: A Scoping Review. Ann Surg 2022; 275:560-567. [PMID: 34954758 DOI: 10.1097/sla.0000000000005349] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To perform a scoping review of imaging-based machine-learning models to predict clinical outcomes and identify biomarkers in patients with PDAC. SUMMARY OF BACKGROUND DATA Patients with PDAC could benefit from better selection for systemic and surgical therapy. Imaging-based machine-learning models may improve treatment selection. METHODS A scoping review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses-scoping review guidelines in the PubMed and Embase databases (inception-October 2020). The review protocol was prospectively registered (open science framework registration: m4cyx). Included were studies on imaging-based machine-learning models for predicting clinical outcomes and identifying biomarkers for PDAC. The primary outcome was model performance. An area under the curve (AUC) of ≥0.75, or a P-value of ≤0.05, was considered adequate model performance. Methodological study quality was assessed using the modified radiomics quality score. RESULTS After screening 1619 studies, 25 studies with 2305 patients fulfilled the eligibility criteria. All but 1 study was published in 2019 and 2020. Overall, 23/25 studies created models using radiomics features, 1 study quantified vascular invasion on computed tomography, and one used histopathological data. Nine models predicted clinical outcomes with AUC measures of 0.78-0.95, and C-indices of 0.65-0.76. Seventeen models identified biomarkers with AUC measures of 0.68-0.95. Adequate model performance was reported in 23/25 studies. The methodological quality of the included studies was suboptimal, with a median modified radiomics quality score score of 7/36. CONCLUSIONS The use of imaging-based machine-learning models to predict clinical outcomes and identify biomarkers in patients with PDAC is increasingly rapidly. Although these models mostly have good performance scores, their methodological quality should be improved.
Collapse
Affiliation(s)
- Boris V Janssen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Severano Verhoef
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Nina J Wesdorp
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Onno J de Boer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Henk Marquering
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap Stoker
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert Kazemier
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Yu J, Li Q, Zhang H, Meng Y, Liu YF, Jiang H, Ma C, Liu F, Fang X, Li J, Feng X, Shao C, Bian Y, Lu J. Contrast-enhanced computed tomography radiomics and multilayer perceptron network classifier: an approach for predicting CD20 + B cells in patients with pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2022; 47:242-253. [PMID: 34708252 DOI: 10.1007/s00261-021-03285-4] [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: 06/19/2021] [Revised: 09/11/2021] [Accepted: 09/11/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE To develop and validate a machine-learning classifier based on contrast-enhanced computed tomography (CT) for the preoperative prediction of CD20+ B lymphocyte expression in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Overall, 189 patients with PDAC (n = 132 and n = 57 in the training and validation sets, respectively) underwent immunohistochemistry and radiomics feature extraction. The X-tile software was used to stratify them into groups with 'high' and 'low' CD20+ B lymphocyte expression levels. For each patient, 1409 radiomic features were extracted from volumes of interest and reduced using variance analysis and Spearman correlation analysis. A multilayer perceptron (MLP) network classifier was developed using the training and validation set. Model performance was determined by its discriminative ability, calibration, and clinical utility. RESULTS A log-rank test showed that the patients with high CD20+ B expression had significantly longer survival than those with low CD20+ B expression. The prediction model showed good discrimination in both the training and validation sets. For the training set, the area under the curve (AUC), sensitivity, specificity, accuracy, positive predictive value, and negative predictive value were 0.82 (95% CI 0.74-0.89), 92.42%, 57.58%, 0.75, 0.69, and 0.88, respectively; whereas these values for the validation set were 0.84 (95% CI 0.72-0.93), 86.21%, 78.57%, 0.83, 0.81, and 0.85, respectively. CONCLUSION The MLP network classifier based on contrast-enhanced CT can accurately predict CD20+ B expression in patients with PDAC.
Collapse
Affiliation(s)
- Jieyu Yu
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Qi Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Yinghao Meng
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Yan Fang Liu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chao Ma
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Xiaochen Feng
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China.
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai, 200434, China.
| |
Collapse
|
22
|
Casà C, Piras A, D’Aviero A, Preziosi F, Mariani S, Cusumano D, Romano A, Boskoski I, Lenkowicz J, Dinapoli N, Cellini F, Gambacorta MA, Valentini V, Mattiucci GC, Boldrini L. The impact of radiomics in diagnosis and staging of pancreatic cancer. Ther Adv Gastrointest Endosc 2022; 15:26317745221081596. [PMID: 35342883 PMCID: PMC8943316 DOI: 10.1177/26317745221081596] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) is one of the most aggressive tumours, and better risk stratification among patients is required to provide tailored treatment. The meaning of radiomics and texture analysis as predictive techniques are not already systematically assessed. The aim of this study is to assess the role of radiomics in PC. METHODS A PubMed/MEDLINE and Embase systematic review was conducted to assess the role of radiomics in PC. The search strategy was 'radiomics [All Fields] AND ("pancreas" [MeSH Terms] OR "pancreas" [All Fields] OR "pancreatic" [All Fields])' and only original articles referred to PC in humans in the English language were considered. RESULTS A total of 123 studies and 183 studies were obtained using the mentioned search strategy on PubMed and Embase, respectively. After the complete selection process, a total of 56 papers were considered eligible for the analysis of the results. Radiomics methods were applied in PC for assessment technical feasibility and reproducibility aspects analysis, risk stratification, biologic or genomic status prediction and treatment response prediction. DISCUSSION Radiomics seems to be a promising approach to evaluate PC from diagnosis to treatment response prediction. Further and larger studies are required to confirm the role and allowed to include radiomics parameter in a comprehensive decision support system.
Collapse
Affiliation(s)
- Calogero Casà
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Andrea D’Aviero
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Preziosi
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Mariani
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Cusumano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angela Romano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ivo Boskoski
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Jacopo Lenkowicz
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Nicola Dinapoli
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Cellini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Antonietta Gambacorta
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Carlo Mattiucci
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Boldrini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
23
|
Hayashi H, Uemura N, Matsumura K, Zhao L, Sato H, Shiraishi Y, Yamashita YI, Baba H. Recent advances in artificial intelligence for pancreatic ductal adenocarcinoma. World J Gastroenterol 2021; 27:7480-7496. [PMID: 34887644 PMCID: PMC8613738 DOI: 10.3748/wjg.v27.i43.7480] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/02/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains the most lethal type of cancer. The 5-year survival rate for patients with early-stage diagnosis can be as high as 20%, suggesting that early diagnosis plays a pivotal role in the prognostic improvement of PDAC cases. In the medical field, the broad availability of biomedical data has led to the advent of the "big data" era. To overcome this deadly disease, how to fully exploit big data is a new challenge in the era of precision medicine. Artificial intelligence (AI) is the ability of a machine to learn and display intelligence to solve problems. AI can help to transform big data into clinically actionable insights more efficiently, reduce inevitable errors to improve diagnostic accuracy, and make real-time predictions. AI-based omics analyses will become the next alterative approach to overcome this poor-prognostic disease by discovering biomarkers for early detection, providing molecular/genomic subtyping, offering treatment guidance, and predicting recurrence and survival. Advances in AI may therefore improve PDAC survival outcomes in the near future. The present review mainly focuses on recent advances of AI in PDAC for clinicians. We believe that breakthroughs will soon emerge to fight this deadly disease using AI-navigated precision medicine.
Collapse
Affiliation(s)
- Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Liu Zhao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yuta Shiraishi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yo-ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
24
|
Goyal H, Sherazi SAA, Mann R, Gandhi Z, Perisetti A, Aziz M, Chandan S, Kopel J, Tharian B, Sharma N, Thosani N. Scope of Artificial Intelligence in Gastrointestinal Oncology. Cancers (Basel) 2021; 13:5494. [PMID: 34771658 PMCID: PMC8582733 DOI: 10.3390/cancers13215494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal cancers are among the leading causes of death worldwide, with over 2.8 million deaths annually. Over the last few decades, advancements in artificial intelligence technologies have led to their application in medicine. The use of artificial intelligence in endoscopic procedures is a significant breakthrough in modern medicine. Currently, the diagnosis of various gastrointestinal cancer relies on the manual interpretation of radiographic images by radiologists and various endoscopic images by endoscopists. This can lead to diagnostic variabilities as it requires concentration and clinical experience in the field. Artificial intelligence using machine or deep learning algorithms can provide automatic and accurate image analysis and thus assist in diagnosis. In the field of gastroenterology, the application of artificial intelligence can be vast from diagnosis, predicting tumor histology, polyp characterization, metastatic potential, prognosis, and treatment response. It can also provide accurate prediction models to determine the need for intervention with computer-aided diagnosis. The number of research studies on artificial intelligence in gastrointestinal cancer has been increasing rapidly over the last decade due to immense interest in the field. This review aims to review the impact, limitations, and future potentials of artificial intelligence in screening, diagnosis, tumor staging, treatment modalities, and prediction models for the prognosis of various gastrointestinal cancers.
Collapse
Affiliation(s)
- Hemant Goyal
- Department of Internal Medicine, The Wright Center for Graduate Medical Education, 501 S. Washington Avenue, Scranton, PA 18505, USA
| | - Syed A. A. Sherazi
- Department of Medicine, John H Stroger Jr Hospital of Cook County, 1950 W Polk St, Chicago, IL 60612, USA;
| | - Rupinder Mann
- Department of Medicine, Saint Agnes Medical Center, 1303 E. Herndon Ave, Fresno, CA 93720, USA;
| | - Zainab Gandhi
- Department of Medicine, Geisinger Wyoming Valley Medical Center, 1000 E Mountain Dr, Wilkes-Barre, PA 18711, USA;
| | - Abhilash Perisetti
- Division of Interventional Oncology & Surgical Endoscopy (IOSE), Parkview Cancer Institute, 11050 Parkview Circle, Fort Wayne, IN 46845, USA; (A.P.); (N.S.)
| | - Muhammad Aziz
- Department of Gastroenterology and Hepatology, University of Toledo Medical Center, 3000 Arlington Avenue, Toledo, OH 43614, USA;
| | - Saurabh Chandan
- Division of Gastroenterology and Hepatology, CHI Health Creighton University Medical Center, 7500 Mercy Rd, Omaha, NE 68124, USA;
| | - Jonathan Kopel
- Department of Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX 79430, USA;
| | - Benjamin Tharian
- Department of Gastroenterology and Hepatology, The University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA;
| | - Neil Sharma
- Division of Interventional Oncology & Surgical Endoscopy (IOSE), Parkview Cancer Institute, 11050 Parkview Circle, Fort Wayne, IN 46845, USA; (A.P.); (N.S.)
| | - Nirav Thosani
- Division of Gastroenterology, Hepatology & Nutrition, McGovern Medical School, UTHealth, 6410 Fannin, St #1014, Houston, TX 77030, USA;
| |
Collapse
|
25
|
Kröner PT, Engels MML, Glicksberg BS, Johnson KW, Mzaik O, van Hooft JE, Wallace MB, El-Serag HB, Krittanawong C. Artificial intelligence in gastroenterology: A state-of-the-art review. World J Gastroenterol 2021; 27:6794-6824. [PMID: 34790008 PMCID: PMC8567482 DOI: 10.3748/wjg.v27.i40.6794] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/15/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
The development of artificial intelligence (AI) has increased dramatically in the last 20 years, with clinical applications progressively being explored for most of the medical specialties. The field of gastroenterology and hepatology, substantially reliant on vast amounts of imaging studies, is not an exception. The clinical applications of AI systems in this field include the identification of premalignant or malignant lesions (e.g., identification of dysplasia or esophageal adenocarcinoma in Barrett’s esophagus, pancreatic malignancies), detection of lesions (e.g., polyp identification and classification, small-bowel bleeding lesion on capsule endoscopy, pancreatic cystic lesions), development of objective scoring systems for risk stratification, predicting disease prognosis or treatment response [e.g., determining survival in patients post-resection of hepatocellular carcinoma), determining which patients with inflammatory bowel disease (IBD) will benefit from biologic therapy], or evaluation of metrics such as bowel preparation score or quality of endoscopic examination. The objective of this comprehensive review is to analyze the available AI-related studies pertaining to the entirety of the gastrointestinal tract, including the upper, middle and lower tracts; IBD; the hepatobiliary system; and the pancreas, discussing the findings and clinical applications, as well as outlining the current limitations and future directions in this field.
Collapse
Affiliation(s)
- Paul T Kröner
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Megan ML Engels
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
- Cancer Center Amsterdam, Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam 1105, The Netherlands
| | - Benjamin S Glicksberg
- The Hasso Plattner Institute for Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Kipp W Johnson
- The Hasso Plattner Institute for Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Obaie Mzaik
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Amsterdam 2300, The Netherlands
| | - Michael B Wallace
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
- Division of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, Abu Dhabi 11001, United Arab Emirates
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX 77030, United States
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX 77030, United States
| | - Chayakrit Krittanawong
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX 77030, United States
- Section of Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States
| |
Collapse
|
26
|
Gutiérrez ML, Muñoz-Bellvís L, Orfao A. Genomic Heterogeneity of Pancreatic Ductal Adenocarcinoma and Its Clinical Impact. Cancers (Basel) 2021; 13:4451. [PMID: 34503261 PMCID: PMC8430663 DOI: 10.3390/cancers13174451] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer death due to limited advances in recent years in early diagnosis and personalized therapy capable of overcoming tumor resistance to chemotherapy. In the last decades, significant advances have been achieved in the identification of recurrent genetic and molecular alterations of PDAC including those involving the KRAS, CDKN2A, SMAD4, and TP53 driver genes. Despite these common genetic traits, PDAC are highly heterogeneous tumors at both the inter- and intra-tumoral genomic level, which might contribute to distinct tumor behavior and response to therapy, with variable patient outcomes. Despite this, genetic and genomic data on PDAC has had a limited impact on the clinical management of patients. Integration of genomic data for classification of PDAC into clinically defined entities-i.e., classical vs. squamous subtypes of PDAC-leading to different treatment approaches has the potential for significantly improving patient outcomes. In this review, we summarize current knowledge about the most relevant genomic subtypes of PDAC including the impact of distinct patterns of intra-tumoral genomic heterogeneity on the classification and clinical and therapeutic management of PDAC.
Collapse
Affiliation(s)
- María Laura Gutiérrez
- Department of Medicine and Cytometry Service (NUCLEUS), Universidad de Salamanca, 37007 Salamanca, Spain;
- Cancer Research Center (IBMCC-CSIC/USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, 28029 Madrid, Spain
| | - Luis Muñoz-Bellvís
- Cancer Research Center (IBMCC-CSIC/USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, 28029 Madrid, Spain
- Service of General and Gastrointestinal Surgery, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service (NUCLEUS), Universidad de Salamanca, 37007 Salamanca, Spain;
- Cancer Research Center (IBMCC-CSIC/USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium-CIBER-CIBERONC, 28029 Madrid, Spain
| |
Collapse
|
27
|
Knolle M, Kaissis G, Jungmann F, Ziegelmayer S, Sasse D, Makowski M, Rueckert D, Braren R. Efficient, high-performance semantic segmentation using multi-scale feature extraction. PLoS One 2021; 16:e0255397. [PMID: 34411138 PMCID: PMC8375977 DOI: 10.1371/journal.pone.0255397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/15/2021] [Indexed: 11/19/2022] Open
Abstract
The success of deep learning in recent years has arguably been driven by the availability of large datasets for training powerful predictive algorithms. In medical applications however, the sensitive nature of the data limits the collection and exchange of large-scale datasets. Privacy-preserving and collaborative learning systems can enable the successful application of machine learning in medicine. However, collaborative protocols such as federated learning require the frequent transfer of parameter updates over a network. To enable the deployment of such protocols to a wide range of systems with varying computational performance, efficient deep learning architectures for resource-constrained environments are required. Here we present MoNet, a small, highly optimized neural-network-based segmentation algorithm leveraging efficient multi-scale image features. MoNet is a shallow, U-Net-like architecture based on repeated, dilated convolutions with decreasing dilation rates. We apply and test our architecture on the challenging clinical tasks of pancreatic segmentation in computed tomography (CT) images as well as brain tumor segmentation in magnetic resonance imaging (MRI) data. We assess our model's segmentation performance and demonstrate that it provides performance on par with compared architectures while providing superior out-of-sample generalization performance, outperforming larger architectures on an independent validation set, while utilizing significantly fewer parameters. We furthermore confirm the suitability of our architecture for federated learning applications by demonstrating a substantial reduction in serialized model storage requirement as a surrogate for network data transfer. Finally, we evaluate MoNet's inference latency on the central processing unit (CPU) to determine its utility in environments without access to graphics processing units. Our implementation is publicly available as free and open-source software.
Collapse
Affiliation(s)
- Moritz Knolle
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Institute for Artificial Intelligence and Informatics in Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Georgios Kaissis
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Institute for Artificial Intelligence and Informatics in Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- OpenMined
- Department of Computing, Imperial College London, London, United Kingdom
| | - Friederike Jungmann
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sebastian Ziegelmayer
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Daniel Sasse
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Marcus Makowski
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Daniel Rueckert
- Institute for Artificial Intelligence and Informatics in Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Computing, Imperial College London, London, United Kingdom
| | - Rickmer Braren
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
28
|
Wang S, Zheng Y, Yang F, Zhu L, Zhu XQ, Wang ZF, Wu XL, Zhou CH, Yan JY, Hu BY, Kong B, Fu DL, Bruns C, Zhao Y, Qin LX, Dong QZ. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther 2021; 6:249. [PMID: 34219130 PMCID: PMC8255319 DOI: 10.1038/s41392-021-00659-4] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an increasingly common cause of cancer mortality with a tight correspondence between disease mortality and incidence. Furthermore, it is usually diagnosed at an advanced stage with a very dismal prognosis. Due to the high heterogeneity, metabolic reprogramming, and dense stromal environment associated with pancreatic cancer, patients benefit little from current conventional therapy. Recent insight into the biology and genetics of pancreatic cancer has supported its molecular classification, thus expanding clinical therapeutic options. In this review, we summarize how the biological features of pancreatic cancer and its metabolic reprogramming as well as the tumor microenvironment regulate its development and progression. We further discuss potential biomarkers for pancreatic cancer diagnosis, prediction, and surveillance based on novel liquid biopsies. We also outline recent advances in defining pancreatic cancer subtypes and subtype-specific therapeutic responses and current preclinical therapeutic models. Finally, we discuss prospects and challenges in the clinical development of pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Feng Yang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiao-Qiang Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhe-Fang Wang
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Xiao-Lin Wu
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Cheng-Hui Zhou
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Jia-Yan Yan
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - De-Liang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Christiane Bruns
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yue Zhao
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
- Key laboratory of whole-period monitoring and precise intervention of digestive cancer, Shanghai Municipal Health Commission (SMHC), Shanghai, China.
| |
Collapse
|
29
|
Arendt CT, Leithner D, Mayerhoefer ME, Gibbs P, Czerny C, Arnoldner C, Burck I, Leinung M, Tanyildizi Y, Lenga L, Martin SS, Vogl TJ, Schernthaner RE. Radiomics of high-resolution computed tomography for the differentiation between cholesteatoma and middle ear inflammation: effects of post-reconstruction methods in a dual-center study. Eur Radiol 2021; 31:4071-4078. [PMID: 33277670 PMCID: PMC8128805 DOI: 10.1007/s00330-020-07564-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/07/2020] [Accepted: 11/25/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To evaluate the performance of radiomic features extracted from high-resolution computed tomography (HRCT) for the differentiation between cholesteatoma and middle ear inflammation (MEI), and to investigate the impact of post-reconstruction harmonization and data resampling. METHODS One hundred patients were included in this retrospective dual-center study: 48 with histology-proven cholesteatoma (center A: 23; center B: 25) and 52 with MEI (A: 27; B: 25). Radiomic features (co-occurrence and run-length matrix, absolute gradient, autoregressive model, Haar wavelet transform) were extracted from manually defined 2D-ROIs. The ten best features for lesion differentiation were selected using probability of error and average correlation coefficients. A multi-layer perceptron feed-forward artificial neural network (MLP-ANN) was used for radiomics-based classification, with histopathology serving as the reference standard (70% of cases for training, 30% for validation). The analysis was performed five times each on (a) unmodified data and on data that were (b) resampled to the same matrix size, and (c) corrected for acquisition protocol differences using ComBat harmonization. RESULTS Using unmodified data, the MLP-ANN classification yielded an overall median area under the receiver operating characteristic curve (AUC) of 0.78 (0.72-0.84). Using original data from center A and resampled data from center B, an overall median AUC of 0.88 (0.82-0.99) was yielded, while using ComBat harmonized data, an overall median AUC of 0.89 (0.79-0.92) was revealed. CONCLUSION Radiomic features extracted from HRCT differentiate between cholesteatoma and MEI. When using multi-centric data obtained with differences in CT acquisition parameters, data resampling and ComBat post-reconstruction harmonization clearly improve radiomics-based lesion classification. KEY POINTS • Unenhanced high-resolution CT coupled with radiomics analysis may be useful for the differentiation between cholesteatoma and middle ear inflammation. • Pooling of data extracted from inhomogeneous CT datasets does not appear meaningful without further post-processing. • When using multi-centric CT data obtained with differences in acquisition parameters, post-reconstruction harmonization and data resampling clearly improve radiomics-based soft-tissue differentiation.
Collapse
Affiliation(s)
- Christophe T Arendt
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Doris Leithner
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Biomedical Imaging and Image-guided Therapy, Division of General and Pediatric Radiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christian Czerny
- Department of Biomedical Imaging and Image-guided Therapy, Division of General and Pediatric Radiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christoph Arnoldner
- Department of Otorhinolaryngology, Head Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Iris Burck
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Martin Leinung
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Yasemin Tanyildizi
- Department of Neuroradiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Lukas Lenga
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Simon S Martin
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ruediger E Schernthaner
- Department of Biomedical Imaging and Image-guided Therapy, Division of General and Pediatric Radiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| |
Collapse
|
30
|
Xie XJ, Liu SY, Chen JY, Zhao Y, Jiang J, Wu L, Zhang XW, Wu Y, Duan H, He B, Luo H, Han D. Development of unenhanced CT-based imaging signature for BAP1 mutation status prediction in malignant pleural mesothelioma: Consideration of 2D and 3D segmentation. Lung Cancer 2021; 157:30-39. [PMID: 34052706 DOI: 10.1016/j.lungcan.2021.04.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 01/07/2023]
Abstract
OBJECTIVES We aimed to explore the feasibility of 2D and 3D radiomics signature based on the unenhanced computed tomography (CT) images to predict BRCA1-associated protein 1 (BAP1) gene mutation status for malignant pleural mesothelioma (MPM) patients. MATERIALS AND METHODS 74 patients with MPM were retrospectively enrolled (22 mutant BAP1, 52 wild-type BAP1 demonstrated by Sanger sequencing). The radiomic features were extracted respectively from the 2D and 3D segmentation of unenhanced pre-treatment CT images, and the dataset was randomly divided into training (n = 51) and test (n = 23) sets for radiomics model development and internal validation. The synthetic minority over-sampling technique (SMOTE) was used for data balancing in the training set. 2D or 3D features were sequentially selected by ICC > 0.8, correlation analysis (cut-value 0.7), univariate analysis or univariate logistic regression (LR), which were involved into multivariate LR for LR model construction. Following the comparison of the 2D and 3D models by the ROC analysis and Delong test for AUC, the calibration and clinical utility of 2D and 3D models were evaluated. RESULTS 3D radiomic features showed better ICCs compared with 2D in both intra- (P < 0.001) and inter-observer (P < 0.001) analysis. 3D radiomic model based on selected features developed from a balanced training dataset presented a favorable predictive performance with AUC of 0.786 and 0.768 in the training and test sets, respectively. The predictive performance of 3D model was superior to 2D model (1 feature) both in the training (AUC 0.786 vs. 0.683, P = 0.036) and the test (AUC 0.768 vs.0.652, P = 0.441) set. The calibration curve and decision curves also indicate a better BAP1 prediction performance and clinical benefit for 3D model than that of 2D model. CONCLUSION The developed unenhanced CT-based 3D radiomics signature is potential as a noninvasive marker for predicting BAP1 mutation status.
Collapse
Affiliation(s)
- Xiao-Jie Xie
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Si-Yun Liu
- Precision Health Institution, GE Healthcare (China), Beijing, 100176, China
| | - Jian-You Chen
- Department of Radiology, Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650106, China
| | - Yi Zhao
- Department of Pathology, the People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, 675099, China
| | - Jie Jiang
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Li Wu
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Xing-Wen Zhang
- Department of Radiology, the People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, 675099, China
| | - Yi Wu
- Department of Radiology, the People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, 675099, China
| | - Hui Duan
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Bing He
- Department of Pathology, the People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, 675099, China
| | - Heng Luo
- Office of the Vice President, the People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, 675099, China.
| | - Dan Han
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| |
Collapse
|
31
|
Laoveeravat P, Abhyankar PR, Brenner AR, Gabr MM, Habr FG, Atsawarungruangkit A. Artificial intelligence for pancreatic cancer detection: Recent development and future direction. Artif Intell Gastroenterol 2021; 2:56-68. [DOI: 10.35712/aig.v2.i2.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Artificial intelligence (AI) has been increasingly utilized in medical applications, especially in the field of gastroenterology. AI can assist gastroenterologists in imaging-based testing and prediction of clinical diagnosis, for examples, detecting polyps during colonoscopy, identifying small bowel lesions using capsule endoscopy images, and predicting liver diseases based on clinical parameters. With its high mortality rate, pancreatic cancer can highly benefit from AI since the early detection of small lesion is difficult with conventional imaging techniques and current biomarkers. Endoscopic ultrasound (EUS) is a main diagnostic tool with high sensitivity for pancreatic adenocarcinoma and pancreatic cystic lesion. The standard tumor markers have not been effective for diagnosis. There have been recent research studies in AI application in EUS and novel biomarkers to early detect and differentiate malignant pancreatic lesions. The findings are impressive compared to the available traditional methods. Herein, we aim to explore the utility of AI in EUS and novel serum and cyst fluid biomarkers for pancreatic cancer detection.
Collapse
Affiliation(s)
- Passisd Laoveeravat
- Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Priya R Abhyankar
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Aaron R Brenner
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Moamen M Gabr
- Division of Digestive Diseases and Nutrition, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Fadlallah G Habr
- Division of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Amporn Atsawarungruangkit
- Division of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| |
Collapse
|
32
|
Prediction of Tumor Cellularity in Resectable PDAC from Preoperative Computed Tomography Imaging. Cancers (Basel) 2021; 13:cancers13092069. [PMID: 33922981 PMCID: PMC8123300 DOI: 10.3390/cancers13092069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/17/2021] [Accepted: 04/21/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) remains a devastating disease. However, variations in tumor biology influence individual patient outcomes greatly. We previously showed a strong association between magnetic resonance imaging-based tumor cell estimates and patient survival. In this study we aimed to transfer this finding to more broadly applied computed tomography (CT) imaging for non-invasive risk stratification. We correlated in vivo CT imaging with histopathological analyses and could show a strong association between regional Hounsfield Units (HU) and tumor cellularity. In conclusion, our study suggests CT-based tumor cell estimates as a widely applicable way of non-invasive tumor cellularity characterization in PDAC. Abstract Background: PDAC remains a tumor entity with poor prognosis and a 5-year survival rate below 10%. Recent research has revealed invasive biomarkers, such as distinct molecular subtypes, predictive for therapy response and patient survival. Non-invasive prediction of individual patient outcome however remains an unresolved task. Methods: Discrete cellularity regions of PDAC resection specimen (n = 43) were analyzed by routine histopathological work up. Regional tumor cellularity and CT-derived Hounsfield Units (HU, n = 66) as well as iodine concentrations were regionally matched. One-way ANOVA and pairwise t-tests were performed to assess the relationship between different cellularity level in conventional, virtual monoenergetic 40 keV (monoE 40 keV) and iodine map reconstructions. Results: A statistically significant negative correlation between regional tumor cellularity in histopathology and CT-derived HU from corresponding image regions was identified. Radiological differentiation was best possible in monoE 40 keV CT images. However, HU values differed significantly in conventional reconstructions as well, indicating the possibility of a broad clinical application of this finding. Conclusion: In this study we establish a novel method for CT-based prediction of tumor cellularity for in-vivo tumor characterization in PDAC patients.
Collapse
|
33
|
Gorris M, Hoogenboom SA, Wallace MB, van Hooft JE. Artificial intelligence for the management of pancreatic diseases. Dig Endosc 2021; 33:231-241. [PMID: 33065754 PMCID: PMC7898901 DOI: 10.1111/den.13875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 12/16/2022]
Abstract
Novel artificial intelligence techniques are emerging in all fields of healthcare, including gastroenterology. The aim of this review is to give an overview of artificial intelligence applications in the management of pancreatic diseases. We performed a systematic literature search in PubMed and Medline up to May 2020 to identify relevant articles. Our results showed that the development of machine-learning based applications is rapidly evolving in the management of pancreatic diseases, guiding precision medicine in clinical, endoscopic and radiologic settings. Before implementation into clinical practice, further research should focus on the external validation of novel techniques, clarifying the accuracy and robustness of these models.
Collapse
Affiliation(s)
- Myrte Gorris
- Department of Gastroenterology and HepatologyAmsterdam Gastroenterology Endocrinology MetabolismAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sanne A. Hoogenboom
- Department of Gastroenterology and HepatologyAmsterdam Gastroenterology Endocrinology MetabolismAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Michael B. Wallace
- Department of Gastroenterology and HepatologyMayo Clinic JacksonvilleJacksonvilleUSA
| | - Jeanin E. van Hooft
- Department of Gastroenterology and HepatologyAmsterdam Gastroenterology Endocrinology MetabolismAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
34
|
Zha HL, Zong M, Liu XP, Pan JZ, Wang H, Gong HY, Xia TS, Liu XA, Li CY. Preoperative ultrasound-based radiomics score can improve the accuracy of the Memorial Sloan Kettering Cancer Center nomogram for predicting sentinel lymph node metastasis in breast cancer. Eur J Radiol 2020; 135:109512. [PMID: 33429302 DOI: 10.1016/j.ejrad.2020.109512] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/25/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE To develop a combined nomogram by incorporating the Memorial Sloan Kettering Cancer Center (MSKCC) nomogram and ultrasound (US)-based radiomics score (Radscore) for predicting sentinel lymph node (SLN) metastasis in invasive breast cancer. MATERIALS AND METHODS This retrospective study was approved by the ethics committee of our institution, and written informed consent was waived. A total of 452 patients with invasive breast cancer who received SLN Biopsy in a single center were included between January 2016 and December 2019. The patients were divided into a training set (n = 318) and a validation set (n = 134). A total of 1216 features were extracted from the regions of interest (ROIs) of the tumors on conventional ultrasound. The maximum relevance minimum redundancy (mRMR) and the least absolute shrinkage and selection operator (LASSO) algorithm were used to build the Radscore. Afterward, the diagnostic performance was assessed and validated. Comparison of receiver operating characteristic (ROC) curves and decision curve analysis (DCA) were performed to evaluate the incremental value of the combined model. RESULTS Obtained from 18 features, the Radscore indicated a favorable discriminatory capability in the training set with an area under the curve (AUC) of 0.834, whereas a value of 0.770 was observed in the validation set. The AUC of the combined model was 0.901 (95 % confidence interval (95 % CI): 0.865-0.938) in the training set and 0.833 (95 % CI: 0.788-0.878) in the validation set. Both of them were superior to MSKCC or imaging Radscore alone (P < 0.05). DCA demonstrated that the combined model was superior to the others in terms of clinical practicability. CONCLUSION Preoperative US-based Radscore can improve the accuracy of clinical MSKCC nomogram for SLN metastasis prediction in breast cancer.
Collapse
Affiliation(s)
- Hai-Ling Zha
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Min Zong
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Xin-Pei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Jia-Zhen Pan
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Hui Wang
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Hai-Yan Gong
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Tian-Song Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Xiao-An Liu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Cui-Ying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
35
|
Ziegelmayer S, Kaissis G, Harder F, Jungmann F, Müller T, Makowski M, Braren R. Deep Convolutional Neural Network-Assisted Feature Extraction for Diagnostic Discrimination and Feature Visualization in Pancreatic Ductal Adenocarcinoma (PDAC) versus Autoimmune Pancreatitis (AIP). J Clin Med 2020; 9:jcm9124013. [PMID: 33322559 PMCID: PMC7764649 DOI: 10.3390/jcm9124013] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
The differentiation of autoimmune pancreatitis (AIP) and pancreatic ductal adenocarcinoma (PDAC) poses a relevant diagnostic challenge and can lead to misdiagnosis and consequently poor patient outcome. Recent studies have shown that radiomics-based models can achieve high sensitivity and specificity in predicting both entities. However, radiomic features can only capture low level representations of the input image. In contrast, convolutional neural networks (CNNs) can learn and extract more complex representations which have been used for image classification to great success. In our retrospective observational study, we performed a deep learning-based feature extraction using CT-scans of both entities and compared the predictive value against traditional radiomic features. In total, 86 patients, 44 with AIP and 42 with PDACs, were analyzed. Whole pancreas segmentation was automatically performed on CT-scans during the portal venous phase. The segmentation masks were manually checked and corrected if necessary. In total, 1411 radiomic features were extracted using PyRadiomics and 256 features (deep features) were extracted using an intermediate layer of a convolutional neural network (CNN). After feature selection and normalization, an extremely randomized trees algorithm was trained and tested using a two-fold shuffle-split cross-validation with a test sample of 20% (n = 18) to discriminate between AIP or PDAC. Feature maps were plotted and visual difference was noted. The machine learning (ML) model achieved a sensitivity, specificity, and ROC-AUC of 0.89 ± 0.11, 0.83 ± 0.06, and 0.90 ± 0.02 for the deep features and 0.72 ± 0.11, 0.78 ± 0.06, and 0.80 ± 0.01 for the radiomic features. Visualization of feature maps indicated different activation patterns for AIP and PDAC. We successfully trained a machine learning model using deep feature extraction from CT-images to differentiate between AIP and PDAC. In comparison to traditional radiomic features, deep features achieved a higher sensitivity, specificity, and ROC-AUC. Visualization of deep features could further improve the diagnostic accuracy of non-invasive differentiation of AIP and PDAC.
Collapse
Affiliation(s)
- Sebastian Ziegelmayer
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
| | - Georgios Kaissis
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
- Department of Computing, Faculty of Engineering, Technology and Medicine, Imperial College of Science, London SW7 2BU, UK
| | - Felix Harder
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
| | - Friederike Jungmann
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
| | - Tamara Müller
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
| | - Marcus Makowski
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
| | - Rickmer Braren
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (G.K.); (F.H.); (F.J.); (T.M.); (M.M.)
- German Cancer Consortium, Partner Site Technical University of Munich, D-69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-89-4140-5627
| |
Collapse
|
36
|
Abunahel BM, Pontre B, Kumar H, Petrov MS. Pancreas image mining: a systematic review of radiomics. Eur Radiol 2020; 31:3447-3467. [PMID: 33151391 DOI: 10.1007/s00330-020-07376-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
37
|
Collisson EA. Distinctions with a Difference: RNA Subtyping and Clinical Outcome in Pancreatic Cancer. Clin Cancer Res 2020; 26:4715-4716. [PMID: 32661068 DOI: 10.1158/1078-0432.ccr-20-1062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/12/2020] [Accepted: 07/08/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic cancer patients are in desperate need of effective therapy virtually from the moment of their diagnosis. As we acquire more therapies, how best to deploy them, in what order and to which patients is emerging as an important clinical question. Pancreatic cancer subtypes, identifiable with common lab diagnostics in diagnostic biopsy samples, may be helpful in guiding therapy selection.See related article by O'Kane et al., p. 4901.
Collapse
Affiliation(s)
- Eric A Collisson
- University of California, San Francisco, San Francisco, California.
| |
Collapse
|
38
|
Deng Y, Zhou T, Wu JL, Chen Y, Shen CY, Zeng M, Chen T, Zhang XM. The impact of molecular classification based on the transcriptome of pancreatic cancer: from bench to bedside. CHINESE JOURNAL OF ACADEMIC RADIOLOGY 2020; 3:67-75. [DOI: 10.1007/s42058-020-00037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 07/25/2024]
|
39
|
Kaissis GA, Jungmann F, Ziegelmayer S, Lohöfer FK, Harder FN, Schlitter AM, Muckenhuber A, Steiger K, Schirren R, Friess H, Schmid R, Weichert W, Makowski MR, Braren RF. Multiparametric Modelling of Survival in Pancreatic Ductal Adenocarcinoma Using Clinical, Histomorphological, Genetic and Image-Derived Parameters. J Clin Med 2020; 9:jcm9051250. [PMID: 32344944 PMCID: PMC7287805 DOI: 10.3390/jcm9051250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: Pancreatic ductal adenocarcinoma (PDAC) remains a tumor entity of exceptionally poor prognosis, and several biomarkers are under current investigation for the prediction of patient prognosis. Many studies focus on promoting newly developed imaging biomarkers without a rigorous comparison to other established parameters. To assess the true value and leverage the potential of all efforts in this field, a multi-parametric evaluation of the available biomarkers for PDAC survival prediction is warranted. Here we present a multiparametric analysis to assess the predictive value of established parameters and the added contribution of newly developed imaging features such as biomarkers for overall PDAC patient survival. Methods: 103 patients with resectable PDAC were retrospectively enrolled. Clinical and histopathological data (age, sex, chemotherapy regimens, tumor size, lymph node status, grading and resection status), morpho-molecular and genetic data (tumor morphology, molecular subtype, tp53, kras, smad4 and p16 genetics), image-derived features and the combination of all parameters were tested for their prognostic strength based on the concordance index (CI) of multivariate Cox proportional hazards survival modelling after unsupervised machine learning preprocessing. Results: The average CIs of the out-of-sample data were: 0.63 for the clinical and histopathological features, 0.53 for the morpho-molecular and genetic features, 0.65 for the imaging features and 0.65 for the combined model including all parameters. Conclusions: Imaging-derived features represent an independent survival predictor in PDAC and enable the multiparametric, machine learning-assisted modelling of postoperative overall survival with a high performance compared to clinical and morpho-molecular/genetic parameters. We propose that future studies systematically include imaging-derived features to benchmark their additive value when evaluating biomarker-based model performance.
Collapse
Affiliation(s)
- Georgios A. Kaissis
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Department of Computing, Faculty of Engineering, Imperial College of Science, Technology and Medicine, London SW7 2BU, UK
| | - Friederike Jungmann
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Sebastian Ziegelmayer
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Fabian K. Lohöfer
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Felix N. Harder
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Anna Melissa Schlitter
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- German Cancer Consortium, Partner Site Technical University of Munich, D-69120 Heidelberg, Germany
| | - Alexander Muckenhuber
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rebekka Schirren
- School of Medicine, Surgical Clinic and Policlinic, Technical University of Munich, 81675 Munich, Germany
| | - Helmut Friess
- School of Medicine, Surgical Clinic and Policlinic, Technical University of Munich, 81675 Munich, Germany
| | - Roland Schmid
- Department of Internal Medicine II, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Wilko Weichert
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Marcus R. Makowski
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rickmer F. Braren
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-5627
| |
Collapse
|