Next Article in Journal
Research on the Adhesive Performance of a Biomimetic Goat Hoof Track Shoe Pattern
Next Article in Special Issue
Explainable AI: A Neurally-Inspired Decision Stack Framework
Previous Article in Journal
Exploring the Binding Capacity of Mycelium and Wood-Based Composites for Use in Construction
Previous Article in Special Issue
Biomimetic Incremental Domain Generalization with a Graph Network for Surgical Scene Understanding
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Application of Artificial Intelligence in the Management of Pancreatic Cystic Lesions

1
Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
2
College of Medicine, The Ohio State University, Columbus, OH 43210, USA
3
Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
4
Department of Computer Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
5
Department of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Biomimetics 2022, 7(2), 79; https://doi.org/10.3390/biomimetics7020079
Submission received: 17 May 2022 / Revised: 7 June 2022 / Accepted: 7 June 2022 / Published: 14 June 2022
(This article belongs to the Special Issue Artificial Intelligence (AI))

Abstract

:
The rate of incidentally detected pancreatic cystic lesions (PCLs) has increased over the past decade and was recently reported at 8%. These lesions pose a unique challenge, as each subtype of PCL carries a different risk of malignant transformation, ranging from 0% (pancreatic pseudocyst) to 34–68% (main duct intraductal papillary mucinous neoplasm). It is imperative to correctly risk-stratify the malignant potential of these lesions in order to provide the correct care course for the patient, ranging from monitoring to surgical intervention. Even with the multiplicity of guidelines (i.e., the American Gastroenterology Association guidelines and Fukuoka/International Consensus guidelines) and multitude of diagnostic information, risk stratification of PCLs falls short. Studies have reported that 25–64% of patients undergoing PCL resection have pancreatic cysts with no malignant potential, and up to 78% of mucin-producing cysts resected harbor no malignant potential on pathological evaluation. Clinicians are now incorporating artificial intelligence technology to aid in the management of these difficult lesions. This review article focuses on advancements in artificial intelligence within digital pathomics, radiomics, and genomics as they apply to the diagnosis and risk stratification of PCLs.

1. Introduction

With the reduced cost and increased utilization of diagnostic testing for abdominal pathologies and an overall aging population, the rate of incidentally detected pancreatic cystic lesions (PCLs) has increased compared with previous decades and was recently reported at 8% (95% CI: 4–14%) [1]. PCLs present a unique challenge, as some of these lesions are pre-cancerous, such as mucinous PCLs, cystic neuroendocrine tumors, and solid pseudopapillary tumors. A specific subtype of PCLs, branch duct intraductal papillary mucinous neoplasm (BD-IPMN), is the most common precancerous PCL. BD-IPMNs are increasingly prevalent with increasing age at around 10% [1,2]. At the other end of the spectrum, some of the PCLs—such as serous cystadenomas (SCAs) and pseudocysts—represent benign lesions. Each of these PCLs carries a different risk of malignant transformation, which is outlined in Table 1 [2,3,4,5,6,7,8,9,10,11,12,13,14]. Therefore, it is imperative to correctly identify these cystic lesions and their characteristics to stratify their malignant potential. Classification systems such as the American Gastroenterology Association guidelines (AGA), American College of Gastroenterology guidelines, United European Gastroenterology guidelines (UEG), and Fukuoka/International Consensus guidelines (Fukuoka-ICG) rely on demographics, clinical features, fluid cytology, and cyst morphology based on CT, MRI, and endoscopic ultrasound (EUS) for PCL risk stratification [3,4,15,16].
Patients with PCLs will undergo multiple diagnostic testing modalities, including standard of care (SOC) imaging (CT/MRI) and EUS-guided fine needle aspiration (FNA). Some center may offer novel diagnostics that include cyst fluid next generation sequencing (NGS) analysis, EUS-guided through-the-needle biopsy (EUS-TTNB), and EUS-guided needle-based confocal laser endomicroscopy (EUS-nCLE), thus providing abundant information to clinicians [17]. As there are a multitude of PCLs with varied morphologies ranging from serous to mucinous, clinicians have had difficulty with accurately diagnosing and risk stratifying these lesions. Even with the multiplicity of guidelines and multitude of diagnostic information, risk stratification of PCLs falls short. Studies have reported that 25–64% of patients undergoing PCL resection have pancreatic cysts with no malignant potential, and up to 78% of mucin-producing cysts (IPMNs, mucinous cystic neoplasms (MCNs)) resected harbor no malignant potential on pathological evaluation [17,18,19]. This dissonance between risk stratification using SOC and pathological diagnosis is particularly troublesome in pancreatic surgery, as pancreaticoduodenectomies are associated with a morbidity of 30% and mortality rates of 2.1–5% [20,21]. While the goal of surgery in BD-IPMNs is to resect lesions with advanced neoplasia (high-grade dysplasia or carcinoma), multiple surgical series and a recent international multicenter study showed that 63% of resected BD-IPMNs had low-grade dysplasia (LGD), often representing overtreatment [1,22,23,24,25]. Given the difficulty with accurate current risk stratification and the risks of pancreatic surgery, researchers have utilized novel diagnostic models including computer-aided diagnostics with machine learning (ML) and artificial intelligence (AI) in order to risk-stratify PCLs.

1.1. Artificial Intelligence:

Artificial intelligence has applications across multiple practices and has recently been used in medicine to detect and diagnose disease. Given the plethora of diagnostic imaging, genomic information, and endoscopic information in patients being worked up for gastroenterological disease, practitioners have begun to use AI adjunctively to aid in detection and diagnosis of precancerous and cancerous lesions. Since 2010, AI has had multiple applications in gastroenterology, including endoscopic evaluation of lesions, analysis of inflammatory lesions on imaging, and assessment of liver fibrosis [20]. For example, in 2021, the FDA approved an AI assistance tool to help endoscopists identify lesions of concern during colonoscopies. Descriptions of AI terminology can be found in Table 2. AI lies on a spectrum of complexity ranging from machine learning (ML) to deep learning (DL). ML uses mathematical algorithms to analyze input values and predict output values without being explicitly programmed to do so [26]. There are numerous examples of ML, including linear discriminants, Bayesian networks, random forest, and support vector machines (SVMs) [26,27,28]. Each ML method of analysis requires training with data sets that have input data and outcome data [29].
Artificial neural networks (ANNs) are AI computing algorithms that mimic human neurons. Each ANN has an input layer that receives a signal (in the form of data) and an output layer that categorizes the input signal. Just as the neurons weigh action potentials to determine propagation of a signal, hidden layers between the input and output layers of ANNs weigh data characteristics and input from other neurons to determine the output [20,26,29]. DL is comprised of multiple ANNs, allowing for complex processing and automatic detection of the relevant features of input data [30]. DL can be supervised, semi-supervised, or unsupervised. DL algorithms have been applied to numerous specialties within medicine, and convoluted neural networks (CNNs) have recently outperformed both dermatologists and pathologists in identifying pathologies in photographs [31,32]. Advancements in AI have been applied to detection, classification, and diagnosis of PCLs in recent years. Artificial intelligence provides clinicians with modalities to process deeper layers of data on PCLs that are not discernable to humans. The intersection of AI and diagnostic modalities for PCLs, including digital pathomics, radiomics, and genomics, will allow improved risk stratification of these complex lesions. The remainder of this paper focuses on recent updates of AI utilization in PCL evaluation.

1.2. The Application of AI to Manage Pancreatic Cystic Lesions: EUS

The increased availability of EUS has allowed for enhanced pancreatic pathology surveillance. Due to the proximity of the transducer to the pancreas, EUS offers more granularity and improved spatial resolution in pancreatic imaging when compared with computed tomography (CT) and magnetic resonance imaging (MRI) and can be used to evaluate lesions as small as 1 cm [33]. The accuracy of CT and MRI in detecting pancreatic lesions < 30 mm is 53% and 67%, respectively, while EUS is 93% accurate [34,35]. Further, novel techniques such as contrast-enhanced EUS, EUS elastography, and EUS-nCLE have improved the diagnostic power of EUS when evaluating PCLs [35]. Given the amount of information in EUS images that is not perceptible to the human eye, clinicians are developing unique ML and DL algorithms to aid in PCL diagnosis and risk stratification.
AI has been utilized in EUS evaluation of pancreatic lesions since 2001, when it was first applied to differentiate focal pancreatitis from malignancy. In this study, a computer algorithm differentiated pancreatitis from malignancy with 73% specificity and 83% accuracy, compared with a blinded endosonographer with 89% and 85%, respectively (n = 35) [36]. Early on, AI models mainly employed ML techniques such as linear discrimination, Bayesian theory, and SVM when evaluating EUS images; however, with advancements in AI, DL concepts such as CNNs are now being applied to assist in the diagnosis and stratification of pancreatic lesions in EUS images. Multiple studies show that CNNs and DL models can differentiate between autoimmune pancreatitis, chronic pancreatitis, pancreatic ductal adenocarcinoma, and normal pancreas [37,38,39,40]. Given their varied morphologies and echogenicity, PCLs have been difficult to assess with AI until recently.

2. AI and EUS in PCL Risk Stratification

AI serves to help clinicians risk stratify PCLs such as IPMNs and differentiate between types of PCLs by identifying deeper characteristics in EUS images. A 2019 retrospective study (n = 50, IPMNs with malignancy = 23) used a DL algorithm to analyze 3790 EUS IPMN images and to evaluate malignant potential in patients having undergone resection with a pathological diagnosis. This study found the sensitivity, specificity, and accuracy of the DL program’s malignant probability to be 95.7%, 96.2%, and 94.0%, respectively. In comparison, the diagnostic accuracy of human interpretation was 56%, and that of the presence of an intracystic mural nodule was 68% [41]. AI has also been able to differentiate between types of PCLs: CNNs have been used to differentiate between EUS morphologies of MCNs (n = 60) and SCAs (n = 49) with 82.75% accuracy and a 0.88 (95% CI: 0.817–0.930) area under curve score [42].

3. AI and EUS-Guided Advanced Diagnostics

EUS-nCLE provides additional information that allows for real-time, high-resolution microscopic imaging of tissue, which facilitates in-vivo histopathology. EUS-nCLE allows practitioners to differentiate between IPMNs and other PCLs [43,44,45]. An image of an IPMN papilla can be seen in Figure 1, highlighting the vascular core and measurements of the epithelial width and density. Certain characteristics of EUS-nCLE allow for the prediction of advanced neoplasia in IPMNs. These characteristics include increased papillary epithelial width and darkness, which reflect cellular stratification and loss of polarity, respectively. These EUS-nCLE imaging characteristics can be seen with corresponding histopathology in Figure 2, which shows low- and high-grade PCL dysplasia. The EUS-nCLE images can be fed into AI algorithms that allow for pixel image processing to stratify based on epithelial dysplasia. In a 2021 study, researchers developed a CNN algorithm that correctly differentiated IPMNs with high-grade dysplasia from those without [46]. In this study, 15,027 EUS-nCLE video frames from 35 consecutive patients with histopathologically proven IPMN (18 with high-grade dysplasia) were used as inputs, with the model yielding higher sensitivity, accuracy, and comparable specificity in diagnosing high-grade dysplasia when compared with international guidelines: sensitivity (model 83.3%, AGA 55.6%, Fukuoka-ICG 55.6%), accuracy (model 85.7%, AGA 68.6%, Fukuoka-ICG 74.3%), and specificity (model 88.2%, AGA 82.4%, Fukuoka-ICG 94.1%) [46]. One of EUS-nCLE’s limitations is lack of widespread adaption. This is likely due to equipment costs (upfront investment), lack of training and exposure during advanced endoscopy fellowships, subtle variations in the advanced endosonography techniques of an nCLE procedure, and the need to learn novel in vivo histopathological imaging.

4. The Future of AI and EUS

The future of AI application to digital pathomics and EUS lies in real-time analytics. The aforementioned studies required trained experts to analyze EUS and EUS-nCLE images to identify regions of interest (ROI), in order to parse out high-utility PCL images from EUS image series to feed into DL models. This prevents on-the-fly real-time evaluations of PCLs. However, recent studies have made advancements in this regard. A 2021 study was able to correctly identify and segment PCL ROIs using DL [34]. Further, a recent study using a CNN-based model correctly identified areas of high-grade dysplasia in a series of IPMN EUS-nCLE images, showing concordance with expert validation with an area under the curve (AUC) sensitivity, and specificity of 0.84, 69%, and 89%, respectively [47].

4.1. Radiomics

Even with the advent of less-invasive options for the assessment of PCLs such as EUS-FNA and EUS through-the-needle biopsies (EUS-TTNB), there is still a significant morbidity with these procedures of 2.66–10% and 5–10%, respectively [48,49,50,51]. Radiomics, also referred to as quantitative imaging, is an emerging field within radiology that utilizes feature extraction and subsequent machine-based analysis of pixels or voxels from cross-sectional imaging to help create “radiological phenotypes” that can aid physicians in the diagnostic workup [50,52,53,54]. Non-invasive diagnostic options such as radiomics offer the potential to discern between PCLs without concurrent morbidity or mortality risk exposure.
Previous studies utilizing quantitative imaging with data obtained through computed tomography (CT) to discern between subtypes of PCLs have been promising, with reported accuracies of up to 84% [55,56]. When combining these machine-based learning models with the expertise of clinicians, appropriate identification of PCL subtypes reached near perfect [57,58]. However, there are limitations to the capabilities of CT. One example is the identification of isoattenuating lesions, which, for example, can represent up to 10% of pancreatic ductal adenocarcinomas (PDACs) [59]. This has led researchers to explore the capabilities of MRI-based radiomics for the identification and prognostication of PCLs. Udare et al. recently published a comparative analysis between CT- and MRI-based quantitative imaging to distinguish between benign and malignant PCLs that revealed comparable sensitivities and specificities [60].

4.1.1. Radiomics and PDAC

Discerning between PDACs and mass-forming chronic pancreatitis (MFCP) is challenging because of their similar clinical presentations and radiological features; however, these pathologies have drastically different outcomes, thus making an accurate diagnosis important. It has been reported that up to 11% of patients who have undergone a pancreaticoduodenectomy because of concern for PDAC were ultimately diagnosed with benign lesions of the pancreas [61]. Recently, Deng et al. published models discerning between PDAC and MFCP using arterial, portal, T1-weighted imaging (T1WI), and T2-weighted imaging (T2WI) phases of dynamic contrast-enhanced MRI [62]. Notably, these models performed better than a traditional clinical model as well as radiologists’ evaluation of the lesion on MRI. This has since been built upon by Liu et al., who developed a multiparametric MRI radiomics signature model that, when combined with independent clinical risk factors such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA 19-9) levels, demonstrated an AUC of 0.973 (95% CI: 0.904–0.997) [63]. MRI-based radiomic models which focus on tumor morphology have also been used to prognosticate and determine adjuvant therapy response in PDAC [64,65].

4.1.2. Radiomics and PCLs

High-risk PCLs such as IPMNs have also been evaluated with MRI-based radiomic models, which have proven to be superior in predicting malignant potential compared with CT models (AUC = 0.940 vs. 0.864, respectively) and yield improved radiometric feature reproducibility (89.4% for MRI vs. 60.5% and 66.8% in the CT arterial and venous phases, respectively) [66]. IPMNs with a high probability of malignant transformation demonstrated high intratumor entropy, and with optimal threshold entropic values set at >5.27, an accuracy of 83.3% was demonstrated. Similar to PDAC models, the incorporation of clinical factors such as pancreatic duct dilation, CEA, and CA 19-9 levels improved radiomic models in predicting high risk IPMNs (AUC = 0.836 vs. 0.903, respectively) [67,68]. Solid pseudopapillary neoplasms (SPNs) are another high-risk subtype of PCLs that confer a high rate of misdiagnosis, which affects timely treatment strategies and patient outcomes. Radiologically, these lesions appear very similar to other high-risk PCL subtypes such as cystic neuroendocrine tumors, as well as SCAs. To discern between these lesions, Gu et al. created an MRI-based radiomic model utilizing T1WI, T2WI, diffusion-weighted imaging (DWI), and contrast-enhanced (CE) sequencing that was able to achieve an accuracy of 84.1% when combined, which improved with the addition of clinical factors such as CEA and CA 19-9 levels [69].
There are some limitations to MRI-based radiomic models that must be considered. First, most studies have only been single-center analyses, and thus external validity and reproducibility between hospital systems is needed. Second, there is less standardization with MRI as compared with CT, and there is a broader range of scanner properties, image settings and sequences, and feature analyses that exist [70]. However, similar limitations also exist for CT-based radiomic models. To expand this field and improve patient outcomes, the development of standardized protocols and multi-center analyses is paramount.

4.2. Genomics

4.2.1. Current Use of Genomics in PCL Management

Genomic analysis is a sparingly used diagnostic technique in PCL management. Currently, the two major applications of this technique are diagnosis of cyst type and malignant risk-stratification. Positive cyst fluid cytology is a surgical indication amongst all current guidelines because it is highly specific for lesions harboring advanced neoplasia, although it is of limited sensitivity [71,72] Genomic analysis via NGS of cyst fluid is a well-studied method that lends to improved sensitivity while maintaining high specificity for the diagnosis of mucinous neoplasm and advanced neoplasia. Specifically, the presence of KRAS and GNAS mutations in cyst fluid has enhanced sensitivity and specificity for classifying mucinous lesions when compared with CEA and cytology [73,74]. Additional mutations (CDKN2A, SMAD4, PTEN, PIK3CA, and TP53) have been identified that are able to predict which lesions are at high risk for progression to malignancy with a sensitivity and specificity of 89% and 100%, respectively [74]. The use of genetics in PCL management has been clearly shown to be of potential benefit when compared with current diagnostic methods. The advancement of artificial intelligence brings the opportunity for improved lesion characterization via AI-based genetic analysis.

4.2.2. AI Genomics in Early Detection of Pancreatic Ductal Adenocarcinoma

Currently, the most utilized biomarker for early detection of PDAC is CA 19-9, but the sensitivity and specificity are suboptimal [75]. One application of AI is the identification of predictive biomarkers for PDAC. For example, one study used an SVM learning algorithm to identify a nine-gene signature that could identify early-stage PDAC with an accuracy, sensitivity, and specificity of 97.53%, 97.96%, and 93.22%, respectively [76]. Furthermore, the PancRISK trial demonstrated AI’s ability to analyze data and create risk-scoring systems. In this trial, three urine biomarkers (LYVE1, REG1B, and TFF1) were used to train and validate five ML algorithms (logistic regression, neural network, random forest, SVM, and neuro-fuzzy technology) in stratifying patients as “Normal” or “Abnormal” risk for PDAC. All algorithms performed similarly (Table 3), but combining PancRISK and CA 19-9 gave a specificity of 0.96 and sensitivity of 0.96 for detecting PDAC, which is better than the current standard of practice [77].
Furthermore, recent studies have utilized mRNA expression, DNA methylation, and miRNA not only as PDAC diagnostic markers but also to appropriately differentiate tumors into less and more aggressive subtypes [78,79,80]. Similarly to PDAC, researchers are applying AI to identify diagnostic and risk-stratification genetic markers for PCLs.

5. Risk Stratification Using AI + Genomics in PCL

It is important to accurately stratify low-risk from high-risk PCLs to shield patients with low-risk lesions from the morbidity of pancreatic surgery and conversely appropriately manage and monitor those with high-risk lesions. Through the input of integrated genomic analysis, clinical characteristics, and imaging features, AI has demonstrated the ability to risk-stratify lesions as either low-grade dysplasia or advanced neoplasia. A large multicenter study performed in 2019 by Springer et al. applied a machine learning technique to develop CompCyst. CompCyst utilizes cyst fluid genetic analysis, along with parameters discussed above, to classify patients into one of three groups: surgery, routine monitoring, or suitable for discharge. CompCyst significantly outperformed current clinical management in identifying patients with low risk for malignancy that could be safely discharged (60% vs. 19%, p = 1.3 × 10−4) [17]. In the subset of patients without advanced neoplasia (n = 140), CompCyst accurately recommended surveillance 68% of the time, whereas the standard of care only recommended monitoring 34% of the time (p = 0.02). Overall, use of the AI-based CompCyst test could have prevented unnecessary surgery in 60% of patients in this study. In addition, both standard management and CompCyst similarly identified lesions that required surgery with high accuracy (89% and 91%) [17].
Machine-learning algorithms have been used indirectly to identify markers related to the grade of cyst dysplasia to develop a risk stratifying biosignature. Maker et al. used an SVM-training algorithm to analyze the cyst fluid gene expression of mRNA, miRNA, KRAS, and GNAS. Through the application of AI, they were able to select appropriate genetic targets for the creation of a PCR-based assay that could characterize lesions as low-grade dysplasia or advanced neoplasia. Of all markers that underwent analysis, their AI algorithm selected the mRNA of IL1β, MUC4, and prostaglandin E synthase 2 to be the most accurate for discrimination of high-risk vs. low-risk cysts (AUC 0.86, p = 0.002) [81] There is yet to be significant research into AI-based genomics as the sole diagnostic and risk stratification tool. Patient outcomes could be further improved through computer-aided genomic algorithms and optimized biomarker panels.

6. The Future: Integrative Computational Models

At present, risk stratification of PCLs based on SOC variables relies on expert consensus-based guidelines, such as Fukuoka-ICG criteria [3,4,15,82]. Multiple studies have addressed the validation of the Fukuoka-ICG criteria and shown a wide range of sensitivity (50–80%) and specificity (55–85%) for the detection of high-grade dysplasia in BD-IPMNs [83,84,85]. While these expert-led consensus guidelines are crucial for the management of PCLs, the non-computational risk assessment of the complex multitude of data may oversimplify underlying relationships in the input risk predictors. Currently in medical practice, clinicians combine available data, apply guidelines such as Fukuoka-ICG, and often confer in multidisciplinary team meetings in order to estimate the risk of malignancy of PCLs. While each of the SOC modalities and advanced diagnostics (EUS-nCLE and cyst fluid NGS) can risk stratify these lesions, an ML/DL-powered integrative computational model can potentially optimize the combined diagnostic accuracy of the data available to clinicians in PCL evaluation (Figure 3). New evaluative modalities that use the tools of AI, such as CompCyst, may allow clinicians to more accurately risk-stratify PCLs and thereby more effectively and efficiently manage these complex lesions.

Author Contributions

Conceptualization: S.R., D.R.A. and S.G.K.; Investigation: S.R., D.R.A., B.R., J.M., R.T., S.C., W.-L.C. and S.G.K.; Resources: S.R., D.R.A., B.R., J.M., R.T., S.C., W.-L.C. and S.G.K.; Writing—original draft preparation: S.R., D.R.A., B.R., J.M., R.T. and S.G.K.; Writing—review and editing: S.R., D.R.A., B.R., J.M., R.T., S.C., W.-L.C. and S.G.K.; Visualization: S.R., D.R.A. and S.G.K.; Supervision: S.C., W.-L.C. and S.G.K.; Project administration: S.G.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Informed consent was not necessary as this was a review article and did not deal with human subjects.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zerboni, G.; Signoretti, M.; Crippa, S.; Falconi, M.; Arcidiacono, P.G.; Capurso, G. Systematic review and meta-analysis: Prevalence of incidentally detected pancreatic cystic lesions in asymptomatic individuals. Pancreatology 2019, 19, 2–9. [Google Scholar] [CrossRef] [PubMed]
  2. Stark, A.; Donahue, T.R.; Reber, H.A.; Hines, O.J. Pancreatic Cyst Disease. JAMA 2016, 315, 1882. [Google Scholar] [CrossRef] [PubMed]
  3. Tanaka, M.; Fernández-del Castillo, C.; Kamisawa, T.; Jang, J.Y.; Levy, P.; Ohtsuka, T.; Salvia, R.; Shimizu, Y.; Tada, M.; Wolfgang, C.L. Revisions of international consensus Fukuoka guidelines for the management of IPMN of the pancreas. Pancreatology 2017, 17, 738–753. [Google Scholar] [CrossRef]
  4. Elta, G.H.; Enestvedt, B.K.; Sauer, B.G.; Lennon, A.M. ACG Clinical Guideline: Diagnosis and Management of Pancreatic Cysts. Am. J. Gastroenterol. 2018, 113, 464–479. [Google Scholar] [CrossRef] [PubMed]
  5. Babiker, H.M.; Hoilat, G.J.; Recio-Boiles, A. Mucinous Cystic Pancreatic Neoplasms; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  6. Park, J.W.; Jang, J.Y.; Kang, M.J.; Kwon, W.; Chang, Y.R.; Kim, S.W. Mucinous cystic neoplasm of the pancreas: Is surgical resection recommended for all surgically fit patients? Pancreatology 2014, 14, 131–136. [Google Scholar] [CrossRef]
  7. Dababneh, Y.; Mousa, O.Y. Pancreatic Serous Cystadenoma; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  8. Law, J.K.; Ahmed, A.; Singh, V.K.; Akshintala, V.S.; Olson, M.T.; Raman, S.P.; Ali, S.Z.; Fishman, E.; Kamel, I.; Canto, M.I.; et al. A Systematic Review of Solid-Pseudopapillary Neoplasms. Pancreas 2014, 43, 331–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Jais, B.; Rebours, V.; Malleo, G.; Salvia, R.; Fontana, M.; Maggino, L.; Bassi, C.; Manfredi, R.; Moran, R.; Lennon, A.M.; et al. Serous cystic neoplasm of the pancreas: A multinational study of 2622 patients under the auspices of the International Association of Pancreatology and European Pancreatic Club (European Study Group on Cystic Tumors of the Pancreas). Gut 2016, 65, 305–312. [Google Scholar] [CrossRef] [Green Version]
  10. Morales-Oyarvide, V.; Fong, Z.V.; Fernández-del Castillo, C.; Warshaw, A.L. Intraductal Papillary Mucinous Neoplasms of the Pancreas: Strategic Considerations. Visc. Med. 2017, 33, 466–476. [Google Scholar] [CrossRef]
  11. Oyama, H.; Tada, M.; Takagi, K.; Tateishi, K.; Hamada, T.; Nakai, Y.; Hakuta, R.; Ijichi, H.; Ishigaki, K.; Kanai, S.; et al. Long-term Risk of Malignancy in Branch-Duct Intraductal Papillary Mucinous Neoplasms. Gastroenterology 2020, 158, 226–237.e5. [Google Scholar] [CrossRef] [Green Version]
  12. Shuja, A.; Alkimawi, K.A. Solid pseudopapillary tumor: A rare neoplasm of the pancreas. Gastroenterol. Rep. 2014, 2, 145–149. [Google Scholar] [CrossRef]
  13. Kawamoto, S.; Johnson, P.T.; Shi, C.; Singhi, A.D.; Hruban, R.H.; Wolfgang, C.L.; Edil, B.H.; Fishman, E.K. Pancreatic Neuroendocrine Tumor with Cystlike Changes: Evaluation with MDCT. Am. J. Roentgenol. 2013, 200, W283–W290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ro, C.; Chai, W.; Yu, V.E.; Yu, R. Pancreatic neuroendocrine tumors: Biology, diagnosis, and treatment. Chin. J. Cancer 2013, 32, 312–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Vege, S.S.; Ziring, B.; Jain, R.; Moayyedi, P.; Adams, M.A.; Dorn, S.D.; Dudley-Brown, S.L.; Flamm, S.L.; Gellad, Z.F.; Gruss, C.B.; et al. American Gastroenterological Association Institute Guideline on the Diagnosis and Management of Asymptomatic Neoplastic Pancreatic Cysts. Gastroenterology 2015, 148, 819–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Tanaka, M.; Fernández-del Castillo, C.; Adsay, V.; Chari, S.; Falconi, M.; Jang, J.-Y.; Kimura, W.; Levy, P.; Pitman, M.B.; Schmidt, C.M.; et al. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology 2012, 12, 183–197. [Google Scholar] [CrossRef]
  17. Springer, S.; Masica, D.L.; Dal Molin, M.; Douville, C.; Thoburn, C.J.; Afsari, B.; Li, L.; Cohen, J.D.; Thompson, E.; Allen, P.J.; et al. A multimodality test to guide the management of patients with a pancreatic cyst. Sci. Transl. Med. 2019, 11, eaav4772. [Google Scholar] [CrossRef]
  18. Perri, G.; Marchegiani, G.; Frigerio, I.; Dervenis, C.G.; Conlon, K.C.; Bassi, C.; Salvia, R. Management of Pancreatic Cystic Lesions. Dig. Surg. 2020, 37, 1–9. [Google Scholar] [CrossRef]
  19. Litchinko, A.; Kobayashi, K.; Halkic, N. A retrospective study of histological outcome for IPMN after surgery in Lausanne, Switzerland: A case series. Ann. Med. Surg. 2020, 60, 110–114. [Google Scholar] [CrossRef]
  20. Le Berre, C.; Sandborn, W.J.; Aridhi, S.; Devignes, M.-D.; Fournier, L.; Smaïl-Tabbone, M.; Danese, S.; Peyrin-Biroulet, L. Application of Artificial Intelligence to Gastroenterology and Hepatology. Gastroenterology 2020, 158, 76–94.e2. [Google Scholar] [CrossRef] [Green Version]
  21. Sahora, K.; Ferrone, C.R.; Brugge, W.R.; Morales-Oyarvide, V.; Warshaw, A.L.; Lillemoe, K.D.; Castillo, C.F.-D. Effects of Comorbidities on Outcomes of Patients with Intraductal Papillary Mucinous Neoplasms. Clin. Gastroenterol. Hepatol. 2015, 13, 1816–1823. [Google Scholar] [CrossRef]
  22. Gaujoux, S.; Brennan, M.F.; Gonen, M.; D’Angelica, M.I.; DeMatteo, R.; Fong, Y.; Schattner, M.; DiMaio, C.; Janakos, M.; Jarnagin, W.R.; et al. Cystic Lesions of the Pancreas: Changes in the Presentation and Management of 1424 Patients at a Single Institution over a 15-Year Time Period. J. Am. Coll. Surg. 2011, 212, 590–600. [Google Scholar] [CrossRef] [Green Version]
  23. Valsangkar, N.P.; Morales-Oyarvide, V.; Thayer, S.P.; Ferrone, C.R.; Wargo, J.A.; Warshaw, A.L.; Fernandez-del Castillo, C. 851 resected cystic tumors of the pancreas: A 33-year experience at the Massachusetts General Hospital. Surgery 2012, 152, S4–S12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Marchegiani, G.; Pollini, T.; Andrianello, S.; Tomasoni, G.; Biancotto, M.; Javed, A.A.; Kinny-Köster, B.; Amini, N.; Han, Y.; Kim, H.; et al. Progression vs Cyst Stability of Branch-Duct Intraductal Papillary Mucinous Neoplasms After Observation and Surgery. JAMA Surg. 2021, 156, 654. [Google Scholar] [CrossRef] [PubMed]
  25. Sahora, K.; Mino-Kenudson, M.; Brugge, W.; Thayer, S.P.; Ferrone, C.R.; Sahani, D.; Pitman, M.B.; Warshaw, A.L.; Lillemoe, K.D.; Fernandez-del Castillo, C.F. Branch Duct Intraductal Papillary Mucinous Neoplasms. Ann. Surg. 2013, 258, 466–475. [Google Scholar] [CrossRef] [PubMed]
  26. Yang, Y.J.; Bang, C.S. Application of artificial intelligence in gastroenterology. World J. Gastroenterol. 2019, 25, 1666–1683. [Google Scholar] [CrossRef] [PubMed]
  27. Jiang, F.; Jiang, Y.; Zhi, H.; Dong, Y.; Li, H.; Ma, S.; Wang, Y.; Dong, Q.; Shen, H.; Wang, Y. Artificial intelligence in healthcare: Past, present and future. Stroke Vasc. Neurol. 2017, 2, 230–243. [Google Scholar] [CrossRef] [PubMed]
  28. Dalal, V.; Carmicheal, J.; Dhaliwal, A.; Jain, M.; Kaur, S.; Batra, S.K. Radiomics in stratification of pancreatic cystic lesions: Machine learning in action. Cancer Lett. 2020, 469, 228–237. [Google Scholar] [CrossRef]
  29. Rajkomar, A.; Dean, J.; Kohane, I. Machine Learning in Medicine. N. Engl. J. Med. 2019, 380, 1347–1358. [Google Scholar] [CrossRef]
  30. Min, J.K.; Kwak, M.S.; Cha, J.M. Overview of Deep Learning in Gastrointestinal Endoscopy. Gut Liver 2019, 13, 388–393. [Google Scholar] [CrossRef] [Green Version]
  31. Hekler, A.; Utikal, J.S.; Enk, A.H.; Solass, W.; Schmitt, M.; Klode, J.; Schadendorf, D.; Sondermann, W.; Franklin, C.; Bestvater, F.; et al. Deep learning outperformed 11 pathologists in the classification of histopathological melanoma images. Eur. J. Cancer 2019, 118, 91–96. [Google Scholar] [CrossRef] [Green Version]
  32. Brinker, T.J.; Hekler, A.; Enk, A.H.; Klode, J.; Hauschild, A.; Berking, C.; Schilling, B.; Haferkamp, S.; Schadendorf, D.; Holland-Letz, T.; et al. Deep learning outperformed 136 of 157 dermatologists in a head-to-head dermoscopic melanoma image classification task. Eur. J. Cancer 2019, 113, 47–54. [Google Scholar] [CrossRef] [Green Version]
  33. Yamaguchi, K.; Okusaka, T.; Shimizu, K.; Furuse, J.; Ito, Y.; Hanada, K.; Shimosegawa, T.; Okazaki, K. Clinical Practice Guidelines for Pancreatic Cancer 2016 from the Japan Pancreas Society. Pancreas 2017, 46, 595–604. [Google Scholar] [CrossRef] [PubMed]
  34. Oh, S.; Kim, Y.J.; Park, Y.T.; Kim, K.G. Automatic Pancreatic Cyst Lesion Segmentation on EUS Images Using a Deep-Learning Approach. Sensors 2021, 22, 245. [Google Scholar] [CrossRef] [PubMed]
  35. Kitano, M.; Yoshida, T.; Itonaga, M.; Tamura, T.; Hatamaru, K.; Yamashita, Y. Impact of endoscopic ultrasonography on diagnosis of pancreatic cancer. J. Gastroenterol. 2019, 54, 19–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Norton, I.D.; Zheng, Y.; Wiersema, M.S.; Greenleaf, J.; Clain, J.E.; DiMagno, E.P. Neural network analysis of EUS images to differentiate between pancreatic malignancy and pancreatitis. Gastrointest. Endosc. 2001, 54, 625–629. [Google Scholar] [CrossRef]
  37. Marya, N.B.; Powers, P.D.; Chari, S.T.; Gleeson, F.C.; Leggett, C.L.; Abu Dayyeh, B.K.; Chandrasekhara, V.; Iyer, P.G.; Majumder, S.; Pearson, R.K.; et al. Utilisation of artificial intelligence for the development of an EUS-convolutional neural network model trained to enhance the diagnosis of autoimmune pancreatitis. Gut 2021, 70, 1335–1344. [Google Scholar] [CrossRef] [PubMed]
  38. Zhu, J.; Wang, L.; Chu, Y.; Hou, X.; Xing, L.; Kong, F.; Zhou, Y.; Wang, Y.; Jin, Z.; Li, Z. A new descriptor for computer-aided diagnosis of EUS imaging to distinguish autoimmune pancreatitis from chronic pancreatitis. Gastrointest. Endosc. 2015, 82, 831–836.e1. [Google Scholar] [CrossRef]
  39. Săftoiu, A.; Vilmann, P.; Gorunescu, F.; Janssen, J.; Hocke, M.; Larsen, M.; Iglesias–Garcia, J.; Arcidiacono, P.G.; Will, U.; Giovannini, M.; et al. Efficacy of an Artificial Neural Network–Based Approach to Endoscopic Ultrasound Elastography in Diagnosis of Focal Pancreatic Masses. Clin. Gastroenterol. Hepatol. 2012, 10, 84–90. [Google Scholar] [CrossRef]
  40. Tonozuka, R.; Itoi, T.; Nagata, N.; Kojima, H.; Sofuni, A.; Tsuchiya, T.; Ishii, K.; Tanaka, R.; Nagakawa, Y.; Mukai, S. Deep learning analysis for the detection of pancreatic cancer on endosonographic images: A pilot study. J. Hepato-Biliary-Pancreat. Sci. 2021, 28, 95–104. [Google Scholar] [CrossRef]
  41. Kuwahara, T.; Hara, K.; Mizuno, N.; Okuno, N.; Matsumoto, S.; Obata, M.; Kurita, Y.; Koda, H.; Toriyama, K.; Onishi, S.; et al. Usefulness of Deep Learning Analysis for the Diagnosis of Malignancy in Intraductal Papillary Mucinous Neoplasms of the Pancreas. Clin. Transl. Gastroenterol. 2019, 10, e00045. [Google Scholar] [CrossRef]
  42. Nguon, L.; Seo, K.; Lim, J.-H.; Song, T.-J.; Cho, S.-H.; Park, J.-S.; Park, S. Deep Learning-Based Differentiation between Mucinous Cystic Neoplasm and Serous Cystic Neoplasm in the Pancreas Using Endoscopic Ultrasonography. Diagnostics 2021, 11, 1052. [Google Scholar] [CrossRef]
  43. Nakai, Y.; Iwashita, T.; Park, D.H.; Samarasena, J.B.; Lee, J.G.; Chang, K.J. Diagnosis of pancreatic cysts: EUS-guided, through-the-needle confocal laser-induced endomicroscopy and cystoscopy trial: DETECT study. Gastrointest. Endosc. 2015, 81, 1204–1214. [Google Scholar] [CrossRef] [PubMed]
  44. Konda, V.J.A.; Meining, A.; Jamil, L.H.; Giovannini, M.; Hwang, J.H.; Wallace, M.B.; Chang, K.J.; Siddiqui, U.D.; Hart, J.; Lo, S.K.; et al. A pilot study of in vivo identification of pancreatic cystic neoplasms with needle-based confocal laser endomicroscopy under endosonographic guidance. Endoscopy 2013, 45, 1006–1013. [Google Scholar] [CrossRef] [PubMed]
  45. Krishna, S.G.; Hart, P.A.; Malli, A.; Kruger, A.J.; McCarthy, S.T.; Eldika, S.; Walker, J.P.; Dillhoff, M.E.; Manilchuk, A.; Schmidt, C.R.; et al. Endoscopic Ultrasound-Guided Confocal Laser Endomicroscopy Increases Accuracy of Differentiation of Pancreatic Cystic Lesions. Clin. Gastroenterol. Hepatol. 2020, 18, 432–440. [Google Scholar] [CrossRef] [PubMed]
  46. Machicado, J.D.; Chao, W.-L.; Carlyn, D.E.; Pan, T.-Y.; Poland, S.; Alexander, V.L.; Maloof, T.G.; Dubay, K.; Ueltschi, O.; Middendorf, D.M.; et al. High performance in risk stratification of intraductal papillary mucinous neoplasms by confocal laser endomicroscopy image analysis with convolutional neural networks (with video). Gastrointest. Endosc. 2021, 94, 78–87.e2. [Google Scholar] [CrossRef] [PubMed]
  47. Krishna, S.G.; Chao, W.-L.; Strobel, S.G.; Stanich, P.P.; Patel, A.; Luthra, A.; Chan, M.Q.; Blaszczak, A.; Lee, D.; Porter, K.; et al. Mo2052—Application of Machine Learning and Artificial Intelligence in the Detection of Dyplasia in Intraductal Papillary Mucinous Neoplasms Using Eus-Guided Needle-Based Confocal Laser Endomicroscopy. Gastroenterology 2019, 156, S-938. [Google Scholar] [CrossRef]
  48. Zhu, H.; Jiang, F.; Zhu, J.; Du, Y.; Jin, Z.; Li, Z. Assessment of morbidity and mortality associated with endoscopic ultrasound-guided fine-needle aspiration for pancreatic cystic lesions: A systematic review and meta-analysis. Dig. Endosc. 2017, 29, 667–675. [Google Scholar] [CrossRef]
  49. Bertani, H.; Pezzilli, R.; Pigò, F.; Bruno, M.; De Angelis, C.; Manfredi, G.; Delconte, G.; Conigliaro, R.; Buscarini, E. Needle-based confocal endomicroscopy in the discrimination of mucinous from non-mucinous pancreatic cystic lesions. World J. Gastrointest. Endosc. 2021, 13, 555–564. [Google Scholar] [CrossRef]
  50. Kovacevic, B.; Antonelli, G.; Klausen, P.; Hassan, C.; Larghi, A.; Vilmann, P.; Karstensen, J. EUS-guided biopsy versus confocal laser endomicroscopy in patients with pancreatic cystic lesions: A systematic review and meta-analysis. Endosc. Ultrasound 2021, 10, 270. [Google Scholar] [CrossRef]
  51. Guzmán-Calderón, E.; Martinez-Moreno, B.; Casellas, J.A.; de Madaria, E.; Aparicio, J.R. Endoscopic ultrasound-guided, through-the-needle forceps biopsy for diagnosis of pancreatic cystic lesions: A systematic review. Endosc. Int. Open 2020, 8, E1123–E1133. [Google Scholar] [CrossRef]
  52. Mayerhoefer, M.E.; Materka, A.; Langs, G.; Häggström, I.; Szczypiński, P.; Gibbs, P.; Cook, G. Introduction to Radiomics. J. Nucl. Med. 2020, 61, 488–495. [Google Scholar] [CrossRef]
  53. Varghese, B.A.; Cen, S.Y.; Hwang, D.H.; Duddalwar, V.A. Texture Analysis of Imaging: What Radiologists Need to Know. Am. J. Roentgenol. 2019, 212, 520–528. [Google Scholar] [CrossRef] [PubMed]
  54. Rogers, W.; Seetha, S.T.; Refaee, T.A.G.; Lieverse, R.I.Y.; Granzier, R.W.Y.; Ibrahim, A.; Keek, S.A.; Sanduleanu, S.; Primakov, S.P.; Beuque, M.P.L.; et al. Radiomics: From qualitative to quantitative imaging. Br. J. Radiol. 2020, 93, 20190948. [Google Scholar] [CrossRef] [PubMed]
  55. Dmitriev, K.; Kaufman, A.E.; Javed, A.A.; Hruban, R.H.; Fishman, E.K.; Lennon, A.M.; Saltz, J.H. Classification of Pancreatic Cysts in Computed Tomography Images Using a Random Forest and Convolutional Neural Network Ensemble. Lect. Notes Comput. Sci. 2017, 10435, 150–158. [Google Scholar] [CrossRef]
  56. Shen, X.; Yang, F.; Yang, P.; Yang, M.; Xu, L.; Zhuo, J.; Wang, J.; Lu, D.; Liu, Z.; Zheng, S.-S.; et al. A Contrast-Enhanced Computed Tomography Based Radiomics Approach for Preoperative Differentiation of Pancreatic Cystic Neoplasm Subtypes: A Feasibility Study. Front. Oncol. 2020, 10, 248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Xie, H.; Ma, S.; Guo, X.; Zhang, X.; Wang, X. Preoperative differentiation of pancreatic mucinous cystic neoplasm from macrocystic serous cystic adenoma using radiomics: Preliminary findings and comparison with radiological model. Eur. J. Radiol. 2020, 122, 108747. [Google Scholar] [CrossRef] [Green Version]
  58. Wei, R.; Lin, K.; Yan, W.; Guo, Y.; Wang, Y.; Li, J.; Zhu, J. Computer-Aided Diagnosis of Pancreas Serous Cystic Neoplasms: A Radiomics Method on Preoperative MDCT Images. Technol. Cancer Res. Treat. 2019, 18, 1–9. [Google Scholar] [CrossRef] [Green Version]
  59. Granata, V.; Grassi, R.; Fusco, R.; Galdiero, R.; Setola, S.V.; Palaia, R.; Belli, A.; Silvestro, L.; Cozzi, D.; Brunese, L.; et al. Pancreatic cancer detection and characterization: State of the art and radiomics. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 3684–3699. [Google Scholar] [CrossRef]
  60. Udare, A.; Agarwal, M.; Alabousi, M.; McInnes, M.; Rubino, J.G.; Marcaccio, M.; van der Pol, C.B. Diagnostic Accuracy of MRI for Differentiation of Benign and Malignant Pancreatic Cystic Lesions Compared to CT and Endoscopic Ultrasound: Systematic Review and Meta-analysis. J. Magn. Reson. Imaging 2021, 54, 1126–1137. [Google Scholar] [CrossRef]
  61. Kennedy, T.; Preczewski, L.; Stocker, S.J.; Rao, S.M.; Parsons, W.G.; Wayne, J.D.; Bell, R.H.; Talamonti, M.S. Incidence of benign inflammatory disease in patients undergoing Whipple procedure for clinically suspected carcinoma: A single-institution experience. Am. J. Surg. 2006, 191, 437–441. [Google Scholar] [CrossRef]
  62. Deng, Y.; Ming, B.; Zhou, T.; Wu, J.-L.; Chen, Y.; Liu, P.; Zhang, J.; Zhang, S.-Y.; Chen, T.-W.; Zhang, X.-M. Radiomics Model Based on MR Images to Discriminate Pancreatic Ductal Adenocarcinoma and Mass-Forming Chronic Pancreatitis Lesions. Front. Oncol. 2021, 11, 811. [Google Scholar] [CrossRef]
  63. Liu, J.; Hu, L.; Zhou, B.; Wu, C.; Cheng, Y. Development and validation of a novel model incorporating MRI-based radiomics signature with clinical biomarkers for distinguishing pancreatic carcinoma from mass-forming chronic pancreatitis. Transl. Oncol. 2022, 18, 101357. [Google Scholar] [CrossRef] [PubMed]
  64. Jeon, S.K.; Jang, J.-Y.; Kwon, W.; Kim, H.; Han, Y.; Kim, D.; Park, D.; Kim, J.H. Diffusion-weighted MR imaging in pancreatic ductal adenocarcinoma: Prediction of next-generation sequencing-based tumor cellularity and prognosis after surgical resection. Abdom. Radiol. 2021, 46, 4787–4799. [Google Scholar] [CrossRef]
  65. Liang, L.; Ding, Y.; Yu, Y.; Liu, K.; Rao, S.; Ge, Y.; Zeng, M. Whole-tumour evaluation with MRI and radiomics features to predict the efficacy of S-1 for adjuvant chemotherapy in postoperative pancreatic cancer patients: A pilot study. BMC Med. Imaging 2021, 21, 75. [Google Scholar] [CrossRef] [PubMed]
  66. Cheng, S.; Shi, H.; Lu, M.; Wang, C.; Duan, S.; Xu, Q.; Shi, H. Radiomics Analysis for Predicting Malignant Potential of Intraductal Papillary Mucinous Neoplasms of the Pancreas: Comparison of CT and MRI. Acad. Radiol. 2022, 29, 367–375. [Google Scholar] [CrossRef] [PubMed]
  67. Hoffman, D.H.; Ream, J.M.; Hajdu, C.H.; Rosenkrantz, A.B. Utility of whole-lesion ADC histogram metrics for assessing the malignant potential of pancreatic intraductal papillary mucinous neoplasms (IPMNs). Abdom. Radiol. 2017, 42, 1222–1228. [Google Scholar] [CrossRef]
  68. Cui, S.; Tang, T.; Su, Q.; Wang, Y.; Shu, Z.; Yang, W.; Gong, X. Radiomic nomogram based on MRI to predict grade of branching type intraductal papillary mucinous neoplasms of the pancreas: A multicenter study. Cancer Imaging 2021, 21, 26. [Google Scholar] [CrossRef]
  69. Gu, H.; Liang, H.; Zhong, J.; Wei, Y.; Ma, Y. How does the pancreatic solid pseudopapillary neoplasm confuse us: Analyzing from the point view of MRI-based radiomics? Magn. Reson. Imaging 2022, 85, 38–43. [Google Scholar] [CrossRef]
  70. Tobaly, D.; Santinha, J.; Sartoris, R.; Burgio, M.D.; Matos, C.; Cros, J.; Couvelard, A.; Rebours, V.; Sauvanet, A.; Ronot, M.; et al. CT-Based Radiomics Analysis to Predict Malignancy in Patients with Intraductal Papillary Mucinous Neoplasm (IPMN) of the Pancreas. Cancers 2020, 12, 3089. [Google Scholar] [CrossRef]
  71. Buerlein, R.C.D.; Shami, V.M. Management of pancreatic cysts and guidelines: What the gastroenterologist needs to know. Ther. Adv. Gastrointest. Endosc. 2021, 14, 1–21. [Google Scholar] [CrossRef]
  72. Ardeshna, D.R.; Cao, T.; Rodgers, B.; Onongaya, C.; Jones, D.; Chen, W.; Koay, E.J.; Krishna, S.G. Recent advances in the diagnostic evaluation of pancreatic cystic lesions. World J. Gastroenterol. 2022, 28, 624–634. [Google Scholar] [CrossRef]
  73. Ren, R.; Krishna, S.G.; Chen, W.; Frankel, W.L.; Shen, R.; Zhao, W.; Avenarius, M.R.; Garee, J.; Caruthers, S.; Jones, D. Activation of the RAS pathway through uncommon BRAF mutations in mucinous pancreatic cysts without KRAS mutation. Mod. Pathol. 2021, 34, 438–444. [Google Scholar] [CrossRef] [PubMed]
  74. Singhi, A.D.; McGrath, K.; Brand, R.E.; Khalid, A.; Zeh, H.J.; Chennat, J.S.; Fasanella, K.E.; Papachristou, G.I.; Slivka, A.; Bartlett, D.L.; et al. Preoperative next-generation sequencing of pancreatic cyst fluid is highly accurate in cyst classification and detection of advanced neoplasia. Gut 2018, 67, 2131–2141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Goonetilleke, K.S.; Siriwardena, A.K. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur. J. Surg. Oncol. 2007, 33, 266–270. [Google Scholar] [CrossRef]
  76. Zhang, Z.M.; Wang, J.S.; Zulfiqar, H.; Lv, H.; Dao, F.Y.; Lin, H. Early Diagnosis of Pancreatic Ductal Adenocarcinoma by Combining Relative Expression Orderings with Machine-Learning Method. Front. Cell Dev. Biol. 2020, 8, 582864. [Google Scholar] [CrossRef]
  77. Blyuss, O.; Zaikin, A.; Cherepanova, V.; Munblit, D.; Kiseleva, E.M.; Prytomanova, O.M.; Duffy, S.W.; Crnogorac-Jurcevic, T. Development of PancRISK, a urine biomarker-based risk score for stratified screening of pancreatic cancer patients. Br. J. Cancer 2020, 122, 692–696. [Google Scholar] [CrossRef] [PubMed]
  78. Sinkala, M.; Mulder, N.; Martin, D. Machine Learning and Network Analyses Reveal Disease Subtypes of Pancreatic Cancer and their Molecular Characteristics. Sci. Rep. 2020, 10, 1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Savareh, B.A.; Aghdaie, H.A.; Behmanesh, A.; Bashiri, A.; Sadeghi, A.; Zali, M.; Shams, R. A machine learning approach identified a diagnostic model for pancreatic cancer through using circulating microRNA signatures. Pancreatology 2020, 20, 1195–1204. [Google Scholar] [CrossRef] [PubMed]
  80. Baek, B.; Lee, H. Prediction of survival and recurrence in patients with pancreatic cancer by integrating multi-omics data. Sci. Rep. 2020, 10, 18951. [Google Scholar] [CrossRef]
  81. Maker, A.V.; Hu, V.; Kadkol, S.S.; Hong, L.; Brugge, W.; Winter, J.; Yeo, C.J.; Hackert, T.; Büchler, M.; Lawlor, R.T.; et al. Cyst Fluid Biosignature to Predict Intraductal Papillary Mucinous Neoplasms of the Pancreas with High Malignant Potential. J. Am. Coll. Surg. 2019, 228, 721–729. [Google Scholar] [CrossRef]
  82. Scheiman, J.M.; Hwang, J.H.; Moayyedi, P. American Gastroenterological Association Technical Review on the Diagnosis and Management of Asymptomatic Neoplastic Pancreatic Cysts. Gastroenterology 2015, 148, 824–848.e22. [Google Scholar] [CrossRef] [Green Version]
  83. Dbouk, M.; Gutierrez, O.I.B.; Lennon, A.M.; Chuidian, M.; Shin, E.J.; Kamel, I.R.; Fishman, E.K.; He, J.; Burkhart, R.A.; Wolfgang, C.L.; et al. Guidelines on management of pancreatic cysts detected in high-risk individuals: An evaluation of the 2017 Fukuoka guidelines and the 2020 International Cancer of the Pancreas Screening (CAPS) consortium statements. Pancreatology 2021, 21, 613–621. [Google Scholar] [CrossRef] [PubMed]
  84. Kaimakliotis, P.; Riff, B.; Pourmand, K.; Chandrasekhara, V.; Furth, E.E.; Siegelman, E.S.; Drebin, J.; Vollmer, C.M.; Kochman, M.L.; Ginsberg, G.G.; et al. Sendai and Fukuoka Consensus Guidelines Identify Advanced Neoplasia in Patients with Suspected Mucinous Cystic Neoplasms of the Pancreas. Clin. Gastroenterol. Hepatol. 2015, 13, 1808–1815. [Google Scholar] [CrossRef] [PubMed]
  85. Heckler, M.; Michalski, C.W.; Schaefle, S.; Kaiser, J.; Büchler, M.W.; Hackert, T. The Sendai and Fukuoka consensus criteria for the management of branch duct IPMN—A meta-analysis on their accuracy. Pancreatology 2017, 17, 255–262. [Google Scholar] [CrossRef] [PubMed]
Figure 1. EUS-nCLE image of IPMN. A: Left panel: IPMN epithelium and vascular core. Each linear marking corresponds to a different epithelial thickness, as shown in the adjacent measurement. B: Right panel: IPMN epithelium and vascular core, with measurements of epithelial density as proxied by pixel intensity in image, with corresponding histogram of mean pixel intensity.
Figure 1. EUS-nCLE image of IPMN. A: Left panel: IPMN epithelium and vascular core. Each linear marking corresponds to a different epithelial thickness, as shown in the adjacent measurement. B: Right panel: IPMN epithelium and vascular core, with measurements of epithelial density as proxied by pixel intensity in image, with corresponding histogram of mean pixel intensity.
Biomimetics 07 00079 g001
Figure 2. A comparison of EUS-nCLE images. Top panel: IPMNs with low grade dysplasia. The thin and translucent epithelium is noted by red arrows on EUS-nCLE images. Bottom panel: IPMN with high grade dysplasia. The thicker and darker epithelium is noted by yellow arrows.
Figure 2. A comparison of EUS-nCLE images. Top panel: IPMNs with low grade dysplasia. The thin and translucent epithelium is noted by red arrows on EUS-nCLE images. Bottom panel: IPMN with high grade dysplasia. The thicker and darker epithelium is noted by yellow arrows.
Biomimetics 07 00079 g002
Figure 3. Integration of diagnostics for the prediction of HGD-Ca: Standard of care (SOC) variables: Demographics and patient characteristics; age, gender, onset of diabetes, family history symptoms, pancreatitis history, serum CA 19-9, and cyst fluid analysis (glucose, CEA, cytology). Cyst and pancreas morphology: CT/MRI/EUS: size, wall, thickness, mural nodules, growth rate, and pancreatic duct diameter. nCLE: needle-based confocal laser endomicroscopy. NGS: Next generation sequencing.
Figure 3. Integration of diagnostics for the prediction of HGD-Ca: Standard of care (SOC) variables: Demographics and patient characteristics; age, gender, onset of diabetes, family history symptoms, pancreatitis history, serum CA 19-9, and cyst fluid analysis (glucose, CEA, cytology). Cyst and pancreas morphology: CT/MRI/EUS: size, wall, thickness, mural nodules, growth rate, and pancreatic duct diameter. nCLE: needle-based confocal laser endomicroscopy. NGS: Next generation sequencing.
Biomimetics 07 00079 g003
Table 1. Type, characteristics, and malignant potential of pancreatic cystic lesions.
Table 1. Type, characteristics, and malignant potential of pancreatic cystic lesions.
Cyst TypeCharacteristicsRate of Malignancy (%)
Main duct IPMNMucinous cyst with variable malignant potential, characterized by main pancreatic duct dilation > 5 mm in the absence of other causes of obstruction [3]38–68% [2]
Branch duct IPMNMucinous cyst with variable malignant potential, characterized as a cyst > 5 mm in diameter that is in communication with the main pancreatic duct. Most common IPMN type [3].15–17% [3,11]
Mixed IPMNDisplays features of both MD-IPMN and BD-IPMN [3]28-31% [10]
Mucinous cystic neoplasmFound almost exclusively in middle-aged women. Mucinous cyst most commonly found in the body or tail of the pancreas. Usually no communication with the pancreatic duct. Columnar epithelium with ovarian stroma differentiates from IPMN [5].10% [6]
Cystic pancreatic neuroendocrine tumorCan be solid, cystic, or mixed composition. Can mimic other cyst types on imaging. Can be associated with Multiple Endocrine Neoplasia type 1 (MEN1) [13].6–31% [14]
Serous cystadenomaMore common in women. Benign, usually found in the tail of the pancreas. Imaging shows microcystic or macrocystic appearance. Central stellate scar is characteristic but not always present. Associated with von Hippel-Lindau disease [7].0.01% [9]
Solid pseudopapillary neoplasmMore common in younger women, commonly third decade of life. Can occur anywhere in the pancreas. Small tumors are usually solid. Large tumors usually have mixed solid and cystic components. Generally well encapsulated and carry a good prognosis [12].10% [8]
PseudocystBenign cyst in patients with history of pancreatitis. Typically high lipase and amylase in cyst fluid.0% [4]
Table 2. Artificial intelligence (AI) terms.
Table 2. Artificial intelligence (AI) terms.
TermDefinitionSubset of AI
Machine learning (ML)Models that use historical data (inputs) to categorize and predict outcomes (outputs). Requires human intervention via algorithm training.ML
Deep learning (DL)A subfield of ML that uses layered neural networks to automatically record and categorize data outputs without human intervention.DL
Linear discriminantsA method used to create a linear combination of characteristics that separates/characterizes data into two subsetsML
Bayesian networksA probabilistic model that relies on independent/dependent input variables to identify causal probabilities of scenariosML
Random forestA model made up of a large number of decision trees, each producing their own prediction. Predictions are combined to formulate a more accurate prediction of an event occurrence.ML
Support vector machines (SVM)Supervised ML algorithm that is capable of performing regression, classification, and outlier predictionML
Artificial neural networks (ANN)Computing algorithms that mimic the human neuron. Each ANN has an input layer and an output layer. Between these layers are hidden layers in which variables are weighted, similar to action potentials.ML/DL
Convoluted neural networks (CNN)Type of ANN that allows for unsupervised evaluation of input data, usually in the form of image, speech, or textDL
Table 3. PDAC detection performance of five machine learning algorithms in PancRisk trial.
Table 3. PDAC detection performance of five machine learning algorithms in PancRisk trial.
MethodLogistic RegressionNeural NetworkRandom ForestSupport Vector MachinesNeuro-Fuzzy Technology
Sensitivity 0.810.810.860.820.87
Specificity 0.90.90.820.890.9
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rangwani, S.; Ardeshna, D.R.; Rodgers, B.; Melnychuk, J.; Turner, R.; Culp, S.; Chao, W.-L.; Krishna, S.G. Application of Artificial Intelligence in the Management of Pancreatic Cystic Lesions. Biomimetics 2022, 7, 79. https://doi.org/10.3390/biomimetics7020079

AMA Style

Rangwani S, Ardeshna DR, Rodgers B, Melnychuk J, Turner R, Culp S, Chao W-L, Krishna SG. Application of Artificial Intelligence in the Management of Pancreatic Cystic Lesions. Biomimetics. 2022; 7(2):79. https://doi.org/10.3390/biomimetics7020079

Chicago/Turabian Style

Rangwani, Shiva, Devarshi R. Ardeshna, Brandon Rodgers, Jared Melnychuk, Ronald Turner, Stacey Culp, Wei-Lun Chao, and Somashekar G. Krishna. 2022. "Application of Artificial Intelligence in the Management of Pancreatic Cystic Lesions" Biomimetics 7, no. 2: 79. https://doi.org/10.3390/biomimetics7020079

Article Metrics

Back to TopTop