1
|
Vincenzi MM, Mori M, Passoni P, Tummineri R, Slim N, Midulla M, Palazzo G, Belardo A, Spezi E, Picchio M, Reni M, Chiti A, del Vecchio A, Fiorino C, Di Muzio NG. Temporal Validation of an FDG-PET-Radiomic Model for Distant-Relapse-Free-Survival After Radio-Chemotherapy for Pancreatic Adenocarcinoma. Cancers (Basel) 2025; 17:1036. [PMID: 40149369 PMCID: PMC11941493 DOI: 10.3390/cancers17061036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Pancreatic cancer is a very aggressive disease with a poor prognosis, even when diagnosed at an early stage. This study aimed to validate and refine a radiomic-based [18F]FDG-PET model to predict distant relapse-free survival (DRFS) in patients with unresectable locally advanced pancreatic cancer (LAPC). Methods: A Cox regression model incorporating two radiomic features (RFs) and cancer stage (III vs. IV) was temporally validated using a larger cohort (215 patients treated between 2005-2022). Patients received concurrent chemoradiotherapy with capecitabine and hypo-fractionated Intensity Modulated Radiotherapy (IMRT). Data were split into training (145 patients, 2005-2017) and validation (70 patients, 2017-2022) groups. Seventy-eight RFs were extracted, harmonized, and analyzed using machine learning to develop refined models. Results: The model incorporating Statistical-Percentile10, Morphological-ComShift, and stage demonstrated moderate predictive accuracy (training: C-index = 0.632; validation: C-index = 0.590). When simplified to include only Statistical-Percentile10, performance improved slightly in the validation group (C-index = 0.601). Adding GLSZM3D-grayLevelVariance to Statistical-Percentile10, while excluding Morphological-ComShift, further enhanced accuracy (training: C-index = 0.654; validation: C-index = 0.623). Despite these refinements, all versions showed similar moderate ability to stratify patients into risk classes. Conclusions: [18F]FDG-PET radiomic features are robust predictors of DRFS after chemoradiotherapy in LAPC. Despite moderate performance, these models hold promise for patient risk stratification. Further validation with external cohorts is ongoing.
Collapse
Affiliation(s)
- Monica Maria Vincenzi
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Martina Mori
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Paolo Passoni
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Roberta Tummineri
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Najla Slim
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Martina Midulla
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gabriele Palazzo
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Alfonso Belardo
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Emiliano Spezi
- School of Engineering, Cardiff University, Cardiff CF24 4HQ, UK
| | - Maria Picchio
- Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Medical Oncology, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
- Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Arturo Chiti
- Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Medical Oncology, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
| | - Antonella del Vecchio
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Claudio Fiorino
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Nadia Gisella Di Muzio
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Imaging Diagnostics, Neuroradiology, and Radiotherapy, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
| |
Collapse
|
2
|
Guenther M, Surendran SA, Steinke LM, Liou I, Palm MA, Heinemann V, Haas M, Boeck S, Ormanns S. The Prognostic, Predictive and Clinicopathological Implications of KRT81/HNF1A- and GATA6-Based Transcriptional Subtyping in Pancreatic Cancer. Biomolecules 2025; 15:426. [PMID: 40149962 PMCID: PMC11940166 DOI: 10.3390/biom15030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Transcriptional subtypes of pancreatic ductal adenocarcinoma (PDAC) based on the expression of hallmark genes may have prognostic implications and potential predictive functions. The two most employed subtyping markers assess the combined expression of KRT81 and HNF1A or of GATA6 alone, which can be detected by immunohistochemistry (IHC). This study aimed to determine the prognostic or predictive impact of both subtyping marker panels in two large cohorts of advanced and resected pancreatic ductal adenocarcinoma (PDAC) patients. METHODS Transcriptional subtypes were determined by combining the expression of KRT81/HNF1A or assessing GATA6 expression alone by IHC in samples of two independent PDAC patient cohorts (advanced PDAC n = 139 and resected PDAC n = 411) as well as in 57 matched primary tumors and their corresponding metastases. RNAseq-based expression data of 316 resected PDAC patients was analyzed for validation. RESULTS Transcriptional subtypes widely overlapped in both marker panels (χ2p < 0.001) but switched during disease progression in up to 31.6% of patients. They had a modest impact on the patients' prognosis in both cohorts, with longer overall survival (OS) for patients with KRT81-/HNF1A+ or GATA6+ tumors but better progression-free survival (PFS) and disease-free survival (DFS) in patients with KRT81+/GATA6- tumors treated with palliative or adjuvant gemcitabine-based chemotherapy. RNAseq expression data confirmed the findings. CONCLUSIONS Transcriptional subtypes have differential responses to palliative and adjuvant gemcitabine-based chemotherapy and may change during disease progression. Both employed subtyping marker panels are equivalent and may be used to inform clinical therapy decisions.
Collapse
Affiliation(s)
- Michael Guenther
- Innpath Institute of Pathology, Tirol Kliniken, 6020 Innsbruck, Austria;
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Sai Agash Surendran
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Lea Margareta Steinke
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Iduna Liou
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Melanie Alexandra Palm
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Volker Heinemann
- Department of Hematology and Oncology, München Klinik Neuperlach, 81737 Munich, Germany; (V.H.); (M.H.); (S.B.)
| | - Michael Haas
- Department of Hematology and Oncology, München Klinik Neuperlach, 81737 Munich, Germany; (V.H.); (M.H.); (S.B.)
- Department of Internal Medicine III, Grosshadern University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Stefan Boeck
- Department of Hematology and Oncology, München Klinik Neuperlach, 81737 Munich, Germany; (V.H.); (M.H.); (S.B.)
- Department of Internal Medicine III, Grosshadern University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Steffen Ormanns
- Innpath Institute of Pathology, Tirol Kliniken, 6020 Innsbruck, Austria;
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
3
|
Qi L, Li X, Ni J, Du Y, Gu Q, Liu B, He J, Du J. Construction of feature selection and efficacy prediction model for transformation therapy of locally advanced pancreatic cancer based on CT, 18F-FDG PET/CT, DNA mutation, and CA199. Cancer Cell Int 2025; 25:19. [PMID: 39828699 PMCID: PMC11743000 DOI: 10.1186/s12935-025-03639-8] [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: 07/22/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Immunotherapy and radiotherapy play crucial roles in the transformation therapy of locally advanced pancreatic cancer; however, the exploration of effective predictive biomarkers has been unsatisfactory. With the rapid development of radiomics, next-generation sequencing, and machine learning, there is hope to identify biomarkers that can predict the efficacy of transformative treatment for locally advanced pancreatic cancer through simple and non-invasive clinical methods. Our study focuses on using computed tomography (CT), positron emission tomography/computed tomography (PET/CT), gene mutations, and baseline carbohydrate antigen 199 (CA199) to identify biomarkers for predicting the efficacy of transformative treatment. METHODS We retrospectively collected data from 70 patients with locally advanced pancreatic cancer who had undergone a biopsy for pathological diagnosis. These patients had complete baseline enhanced CT images and baseline CA199 results. Among them, 65 patients had efficacy evaluation results after 4 treatment cycles, 54 patients had complete baseline PET/CT images, 51 patients had complete DNA mutation detection results, and 34 patients had both complete PET/CT images and DNA mutation detection results. Additionally, 47 patients had complete available CT images at baseline, after 2 treatment cycles, and after 4 treatment cycles. We extracted radiomic features from the original lesion-enhanced CT images (including baseline and subsequent follow-up CT scans), radiomic features from baseline 18F-fluoro-2-deoxy-2-D-glucose (18F-FDG) PET, and patient-specific features related to abdominal and visceral fat. We used short-term and long-term treatment efficacy as the prediction outcomes and performed statistical and machine learning-based feature selection and COX regression analysis to identify potentially predictive features. Subsequently, we separately or in combination modeled the CT features, PET features, baseline CA199, and gene mutation data to construct efficacy prediction models. Finally, we investigated the mixed effects model of the dynamic changes in CT features at baseline, after 2 treatment cycles, and after 4 treatment cycles on the prediction of short-term treatment efficacy. RESULTS We found that a combination of CT radiomic features, including F1_ gray level co-occurrence matrix (GLCM), F2_gray level run length matrix (GLRLM), F5_neighboring gray tone difference matrix (NGTDM), and F6_Shape, PET radiomic features such as visceral adipose tissue (VAT), tumor-to-liver ratio (T/L), standardized uptake value mean (SUVmean), and GLCM, as well as baseline CA199, can be used to predict short-term treatment efficacy. Baseline CA199, GLCM, IntensityDirect, Shape, and PET/CT features are independent factors for long-term treatment efficacy. In constructing the short-term treatment efficacy prediction model, ensemble learning methods such as adaptive boosting (AdaBoost), extreme gradient boosting (XGBoost), and RandomForest performed the best. However, in terms of model interpretability, decision tree methods provide the most intuitive display of the predictive details of the model. For the time series data of patients' baseline CT, CT after 2 treatment cycles, and CT after 4 treatment cycles, long short-term memory (LSTM) modeling yielded better predictive models. CONCLUSION A multimodal combination of radiomics, DNA mutations, and baseline CA199 can predict the efficacy of transformative treatment in locally advanced pancreatic cancer. Various feature selection methods and multimodal fusion approaches contribute to guiding personalized and precise treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Liang Qi
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xiang Li
- Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiayao Ni
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yali Du
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, China
| | - Qing Gu
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China.
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Juan Du
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China.
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
4
|
Karalis VD. The Integration of Artificial Intelligence into Clinical Practice. APPLIED BIOSCIENCES 2024; 3:14-44. [DOI: 10.3390/applbiosci3010002] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The purpose of this literature review is to provide a fundamental synopsis of current research pertaining to artificial intelligence (AI) within the domain of clinical practice. Artificial intelligence has revolutionized the field of medicine and healthcare by providing innovative solutions to complex problems. One of the most important benefits of AI in clinical practice is its ability to investigate extensive volumes of data with efficiency and precision. This has led to the development of various applications that have improved patient outcomes and reduced the workload of healthcare professionals. AI can support doctors in making more accurate diagnoses and developing personalized treatment plans. Successful examples of AI applications are outlined for a series of medical specialties like cardiology, surgery, gastroenterology, pneumology, nephrology, urology, dermatology, orthopedics, neurology, gynecology, ophthalmology, pediatrics, hematology, and critically ill patients, as well as diagnostic methods. Special reference is made to legal and ethical considerations like accuracy, informed consent, privacy issues, data security, regulatory framework, product liability, explainability, and transparency. Finally, this review closes by critically appraising AI use in clinical practice and its future perspectives. However, it is also important to approach its development and implementation cautiously to ensure ethical considerations are met.
Collapse
Affiliation(s)
- Vangelis D. Karalis
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| |
Collapse
|
5
|
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
|
6
|
Yin H, Zhang F, Yang X, Meng X, Miao Y, Noor Hussain MS, Yang L, Li Z. Research trends of artificial intelligence in pancreatic cancer: a bibliometric analysis. Front Oncol 2022; 12:973999. [PMID: 35982967 PMCID: PMC9380440 DOI: 10.3389/fonc.2022.973999] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 01/03/2023] Open
Abstract
Purpose We evaluated the related research on artificial intelligence (AI) in pancreatic cancer (PC) through bibliometrics analysis and explored the research hotspots and current status from 1997 to 2021. Methods Publications related to AI in PC were retrieved from the Web of Science Core Collection (WoSCC) during 1997-2021. Bibliometrix package of R software 4.0.3 and VOSviewer were used to bibliometrics analysis. Results A total of 587 publications in this field were retrieved from WoSCC database. After 2018, the number of publications grew rapidly. The United States and Johns Hopkins University were the most influential country and institution, respectively. A total of 2805 keywords were investigated, 81 of which appeared more than 10 times. Co-occurrence analysis categorized these keywords into five types of clusters: (1) AI in biology of PC, (2) AI in pathology and radiology of PC, (3) AI in the therapy of PC, (4) AI in risk assessment of PC and (5) AI in endoscopic ultrasonography (EUS) of PC. Trend topics and thematic maps show that keywords " diagnosis ", “survival”, “classification”, and “management” are the research hotspots in this field. Conclusion The research related to AI in pancreatic cancer is still in the initial stage. Currently, AI is widely studied in biology, diagnosis, treatment, risk assessment, and EUS of pancreatic cancer. This bibliometrics study provided an insight into AI in PC research and helped researchers identify new research orientations.
Collapse
Affiliation(s)
- Hua Yin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Feixiong Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaoli Yang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiangkun Meng
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yu Miao
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
| | | | - Li Yang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
- *Correspondence: Zhaoshen Li, ; Li Yang,
| | - Zhaoshen Li
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- *Correspondence: Zhaoshen Li, ; Li Yang,
| |
Collapse
|
7
|
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
|
8
|
Xu ZH, Wang WQ, Liu L, Lou WH. A special subtype: Revealing the potential intervention and great value of KRAS wildtype pancreatic cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188751. [PMID: 35732240 DOI: 10.1016/j.bbcan.2022.188751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and has devastating consequences on affected families and society. Its dismal prognosis is attributed to poor specificity of symptoms during early stages. It is widely believed that PDAC patients with the wildtype (WT) KRAS gene benefit more from currently available treatments than those with KRAS mutations. The oncogenic genetic changes alternations generally found in KRAS wildtype PDAC are related to either the KRAS pathway or microsatellite instability/mismatch repair deficiency (MSI/dMMR), which enable the application of tailored treatments based on each patient's genetic characteristics. This review focuses on targeted therapies against alternative tumour mechanisms in KRAS WT PDAC.
Collapse
Affiliation(s)
- Zhi-Hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wen-Hui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Schlanger D, Graur F, Popa C, Moiș E, Al Hajjar N. The role of artificial intelligence in pancreatic surgery: a systematic review. Updates Surg 2022; 74:417-429. [PMID: 35237939 DOI: 10.1007/s13304-022-01255-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022]
Abstract
Artificial intelligence (AI), including machine learning (ML), is being slowly incorporated in medical practice, to provide a more precise and personalized approach. Pancreatic surgery is an evolving field, which offers the only curative option for patients with pancreatic cancer. Increasing amounts of data are available in medicine: AI and ML can help incorporate large amounts of information in clinical practice. We conducted a systematic review, based on PRISMA criteria, of studies that explored the use of AI or ML algorithms in pancreatic surgery. To our knowledge, this is the first systematic review on this topic. Twenty-five eligible studies were included in this review; 12 studies with implications in the preoperative diagnosis, while 13 studies had implications in patient evolution. Preoperative diagnosis, such as predicting the malignancy of IPMNs, differential diagnosis between pancreatic cystic lesions, classification of different pancreatic tumours, and establishment of the correct management for each of these lesions, can be facilitated through different AI or ML algorithms. Postoperative evolution can also be predicted, and some studies reported prediction models for complications, including postoperative pancreatic fistula, while other studies have analysed the implications for prognosis evaluation (from predicting a textbook outcome, the risk of metastasis or relapse, or the mortality rate and survival). One study discussed the possibility of predicting an intraoperative complication-massive intraoperative bleeding. Artificial intelligence and machine learning models have promising applications in pancreatic surgery, in the preoperative period (high-accuracy diagnosis) and postoperative setting (prognosis evaluation and complication prediction), and the intraoperative applications have been less explored.
Collapse
Affiliation(s)
- D Schlanger
- "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania street Emil Isac no 13, 400023, Cluj-Napoca, Romania.,Surgery Department, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. O. Fodor", Cluj-Napoca, Romania. Street Croitorilor no 19-21, 400162, Cluj-Napoca, Romania
| | - F Graur
- "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania street Emil Isac no 13, 400023, Cluj-Napoca, Romania. .,Surgery Department, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. O. Fodor", Cluj-Napoca, Romania. Street Croitorilor no 19-21, 400162, Cluj-Napoca, Romania.
| | - C Popa
- "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania street Emil Isac no 13, 400023, Cluj-Napoca, Romania.,Surgery Department, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. O. Fodor", Cluj-Napoca, Romania. Street Croitorilor no 19-21, 400162, Cluj-Napoca, Romania
| | - E Moiș
- "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania street Emil Isac no 13, 400023, Cluj-Napoca, Romania.,Surgery Department, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. O. Fodor", Cluj-Napoca, Romania. Street Croitorilor no 19-21, 400162, Cluj-Napoca, Romania
| | - N Al Hajjar
- "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania street Emil Isac no 13, 400023, Cluj-Napoca, Romania.,Surgery Department, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. O. Fodor", Cluj-Napoca, Romania. Street Croitorilor no 19-21, 400162, Cluj-Napoca, Romania
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Chaddad A, Sargos P, Desrosiers C. Modeling Texture in Deep 3D CNN for Survival Analysis. IEEE J Biomed Health Inform 2021; 25:2454-2462. [PMID: 32960772 DOI: 10.1109/jbhi.2020.3025901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Radiomics has shown remarkable potential for predicting the survival outcome for various types of cancers such as pancreatic ductal adenocarcinoma (PDAC). However, to date, there has been limited research using convolutional neural networks (CNN) with radiomic methods for this task, due to their requirement for large training sets. To overcome this issue, we propose a new type of radiomic descriptor modeling the distribution of learned features with a Gaussian mixture model (GMM). These parametric features (GMM-CNN) are computed from gross tumor volumes of PDAC patients defined semi-automatically in pre-operative computed tomography (CT) scans. We use the proposed GMM-CNN features as input to a robust classifier based on random forests (RF) to predict the survival outcome of patients with PDAC. Our experiments assess the advantage of GMM-CNN compared to employing the same 3D CNN model directly, standard radiomic (i.e., histogram, texture and shape), conditional entropy (CENT) based on 3DCNN, and clinical features (i.e., serum carbohydrate antigen 19-9 and chemotherapy neoadjuvant). Using the RF model (100 samples for training; 59 samples for validation), GMM-CNN features provided the highest area under the ROC curve (AUC) of 72.0% (p = 6.4×10-5) compared to 64.0% (p = 0.01) for the 3D CNN model output, 66.8% (p = 0.01) for standard radiomic features, 64.2% (p = 0.003) for CENT, and 57.6% (p = 0.3) for clinical variables. Our results suggest that the proposed GMM-CNN features used with a RF classifier can significantly improve the capacity to prognosticate PDAC patients prior to surgery via routinely-acquired imaging data.
Collapse
|
13
|
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
|