537 research outputs found

    Virtual liver biopsy: image processing and 3D visualization

    Get PDF

    Hepatic Vessel Segmentation for 3D Planning of Liver Surgery

    Get PDF
    none8Rationale and Objectives: The aim of this study was to identify the optimal parameter configuration of a new algorithm for fully automatic segmentation of hepatic vessels, evaluating its accuracy in view of its use in a computer system for three-dimensional (3D) planning of liver surgery. Materials and Methods: A phantom reproduction of a human liver with vessels up to the fourth subsegment order, corresponding to a minimum diameter of 0.2 mm, was realized through stereolithography, exploiting a 3D model derived from a real human computed tomographic data set. Algorithm parameter configuration was experimentally optimized, and the maximum achievable segmentation accuracy was quantified for both single two-dimensional slices and 3D reconstruction of the vessel network, through an analytic comparison of the automatic segmentation performed on contrast-enhanced computed tomographic phantom images with actual model features. Results: The optimal algorithm configuration resulted in a vessel detection sensitivity of 100% for vessels > 1 mm in diameter, 50% in the range 0.5 to 1 mm, and 14% in the range 0.2 to 0.5 mm. An average area overlap of 94.9% was obtained between automatically and manually segmented vessel sections, with an average difference of 0.06 mm2. The average values of corresponding false-positive and false-negative ratios were 7.7% and 2.3%, respectively. Conclusions: A robust and accurate algorithm for automatic extraction of the hepatic vessel tree from contrast-enhanced computed tomographic volume images was proposed and experimentally assessed on a liver model, showing unprecedented sensitivity in vessel delineation. This automatic segmentation algorithm is promising for supporting liver surgery planning and for guiding intraoperative resections.Francesco Conversano;Roberto Franchini;Christian Demitri;Laurent Massoptier;Francesco Montagna;Alfonso Maffezzoli;Antonio Malvasi;Sergio CasciaroFrancesco, Conversano; Roberto, Franchini; Demitri, Christian; Laurent, Massoptier; Francesco, Montagna; Maffezzoli, Alfonso; Antonio, Malvasi; Sergio, Casciar

    Crepuscular Rays for Tumor Accessibility Planning

    Get PDF

    A fully automatic method for vascular tortuosity feature extraction in the supra-aortic region: unraveling possibilities in stroke treatment planning

    Get PDF
    Vascular tortuosity of supra-aortic vessels is widely considered one of the main reasons for failure and delays in endovascular treatment of large vessel occlusion in patients with acute ischemic stroke. Characterization of tortuosity is a challenging task due to the lack of objective, robust and effective analysis tools. We present a fully automatic method for arterial segmentation, vessel labelling and tortuosity feature extraction applied to the supra-aortic region. A sample of 566 computed tomography angiography scans from acute ischemic stroke patients (aged 74.8 ยฑ 12.9, 51.0% females) were used for training, validation and testing of a segmentation module based on a U-Net architecture (162 cases) and a vessel labelling module powered by a graph U-Net (566 cases). Successively, 30 cases were processed for testing of a tortuosity feature extraction module. Measurements obtained through automatic processing were compared to manual annotations from two observers for a thorough validation of the method. The proposed feature extraction method presented similar performance to the inter-rater variability observed in the measurement of 33 geometrical and morphological features of the arterial anatomy in the supra-aortic region. This system will contribute to the development of more complex models to advance the treatment of stroke by adding immediate automation, objectivity, repeatability and robustness to the vascular tortuosity characterization of patients

    Liver Segmentation and its Application to Hepatic Interventions

    Get PDF
    The thesis addresses the development of an intuitive and accurate liver segmentation approach, its integration into software prototypes for the planning of liver interventions, and research on liver regeneration. The developed liver segmentation approach is based on a combination of the live wire paradigm and shape-based interpolation. Extended with two correction modes and integrated into a user-friendly workflow, the method has been applied to more than 5000 data sets. The combination of the liver segmentation with image analysis of hepatic vessels and tumors allows for the computation of anatomical and functional remnant liver volumes. In several projects with clinical partners world-wide, the benefit of the computer-assisted planning was shown. New insights about the postoperative liver function and regeneration could be gained, and most recent investigations into the analysis of MRI data provide the option to further improve hepatic intervention planning

    Segmentation and Deformable Modelling Techniques for a Virtual Reality Surgical Simulator in Hepatic Oncology

    No full text
    Liver surgical resection is one of the most frequently used curative therapies. However, resectability is problematic. There is a need for a computer-assisted surgical planning and simulation system which can accurately and efficiently simulate the liver, vessels and tumours in actual patients. The present project describes the development of these core segmentation and deformable modelling techniques. For precise detection of irregularly shaped areas with indistinct boundaries, the segmentation incorporated active contours - gradient vector flow (GVF) snakes and level sets. To improve efficiency, a chessboard distance transform was used to replace part of the GVF effort. To automatically initialize the liver volume detection process, a rotating template was introduced to locate the starting slice. For shape maintenance during the segmentation process, a simplified object shape learning step was introduced to avoid occasional significant errors. Skeletonization with fuzzy connectedness was used for vessel segmentation. To achieve real-time interactivity, the deformation regime of this system was based on a single-organ mass-spring system (MSS), which introduced an on-the-fly local mesh refinement to raise the deformation accuracy and the mesh control quality. This method was now extended to a multiple soft-tissue constraint system, by supplementing it with an adaptive constraint mesh generation. A mesh quality measure was tailored based on a wide comparison of classic measures. Adjustable feature and parameter settings were thus provided, to make tissues of interest distinct from adjacent structures, keeping the mesh suitable for on-line topological transformation and deformation. More than 20 actual patient CT and 2 magnetic resonance imaging (MRI) liver datasets were tested to evaluate the performance of the segmentation method. Instrument manipulations of probing, grasping, and simple cutting were successfully simulated on deformable constraint liver tissue models. This project was implemented in conjunction with the Division of Surgery, Hammersmith Hospital, London; the preliminary reality effect was judged satisfactory by the consultant hepatic surgeon

    RFA Guardian: Comprehensive Simulation of Radiofrequency Ablation Treatment of Liver Tumors

    Get PDF
    The RFA Guardian is a comprehensive application for high-performance patient-specific simulation of radiofrequency ablation of liver tumors. We address a wide range of usage scenarios. These include pre-interventional planning, sampling of the parameter space for uncertainty estimation, treatment evaluation and, in the worst case, failure analysis. The RFA Guardian is the first of its kind that exhibits sufficient performance for simulating treatment outcomes during the intervention. We achieve this by combining a large number of high-performance image processing, biomechanical simulation and visualization techniques into a generalized technical workflow. Further, we wrap the feature set into a single, integrated application, which exploits all available resources of standard consumer hardware, including massively parallel computing on graphics processing units. This allows us to predict or reproduce treatment outcomes on a single personal computer with high computational performance and high accuracy. The resulting low demand for infrastructure enables easy and cost-efficient integration into the clinical routine. We present a number of evaluation cases from the clinical practice where users performed the whole technical workflow from patient-specific modeling to final validation and highlight the opportunities arising from our fast, accurate prediction techniques

    ๋ณต๋ถ€ CT์—์„œ ๊ฐ„๊ณผ ํ˜ˆ๊ด€ ๋ถ„ํ•  ๊ธฐ๋ฒ•

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ(๋ฐ•์‚ฌ)--์„œ์šธ๋Œ€ํ•™๊ต ๋Œ€ํ•™์› :๊ณต๊ณผ๋Œ€ํ•™ ์ปดํ“จํ„ฐ๊ณตํ•™๋ถ€,2020. 2. ์‹ ์˜๊ธธ.๋ณต๋ถ€ ์ „์‚ฐํ™” ๋‹จ์ธต ์ดฌ์˜ (CT) ์˜์ƒ์—์„œ ์ •ํ™•ํ•œ ๊ฐ„ ๋ฐ ํ˜ˆ๊ด€ ๋ถ„ํ• ์€ ์ฒด์  ์ธก์ •, ์น˜๋ฃŒ ๊ณ„ํš ์ˆ˜๋ฆฝ ๋ฐ ์ถ”๊ฐ€์ ์ธ ์ฆ๊ฐ• ํ˜„์‹ค ๊ธฐ๋ฐ˜ ์ˆ˜์ˆ  ๊ฐ€์ด๋“œ์™€ ๊ฐ™์€ ์ปดํ“จํ„ฐ ์ง„๋‹จ ๋ณด์กฐ ์‹œ์Šคํ…œ์„ ๊ตฌ์ถ•ํ•˜๋Š”๋ฐ ํ•„์ˆ˜์ ์ธ ์š”์†Œ์ด๋‹ค. ์ตœ๊ทผ ๋“ค์–ด ์ปจ๋ณผ๋ฃจ์…”๋„ ์ธ๊ณต ์‹ ๊ฒฝ๋ง (CNN) ํ˜•ํƒœ์˜ ๋”ฅ ๋Ÿฌ๋‹์ด ๋งŽ์ด ์ ์šฉ๋˜๋ฉด์„œ ์˜๋ฃŒ ์˜์ƒ ๋ถ„ํ• ์˜ ์„ฑ๋Šฅ์ด ํ–ฅ์ƒ๋˜๊ณ  ์žˆ์ง€๋งŒ, ์‹ค์ œ ์ž„์ƒ์— ์ ์šฉํ•  ์ˆ˜ ์žˆ๋Š” ๋†’์€ ์ผ๋ฐ˜ํ™” ์„ฑ๋Šฅ์„ ์ œ๊ณตํ•˜๊ธฐ๋Š” ์—ฌ์ „ํžˆ ์–ด๋ ต๋‹ค. ๋˜ํ•œ ๋ฌผ์ฒด์˜ ๊ฒฝ๊ณ„๋Š” ์ „ํ†ต์ ์œผ๋กœ ์˜์ƒ ๋ถ„ํ• ์—์„œ ๋งค์šฐ ์ค‘์š”ํ•œ ์š”์†Œ๋กœ ์ด์šฉ๋˜์—ˆ์ง€๋งŒ, CT ์˜์ƒ์—์„œ ๊ฐ„์˜ ๋ถˆ๋ถ„๋ช…ํ•œ ๊ฒฝ๊ณ„๋ฅผ ์ถ”์ถœํ•˜๊ธฐ๊ฐ€ ์–ด๋ ต๊ธฐ ๋•Œ๋ฌธ์— ํ˜„๋Œ€ CNN์—์„œ๋Š” ์ด๋ฅผ ์‚ฌ์šฉํ•˜์ง€ ์•Š๊ณ  ์žˆ๋‹ค. ๊ฐ„ ํ˜ˆ๊ด€ ๋ถ„ํ•  ์ž‘์—…์˜ ๊ฒฝ์šฐ, ๋ณต์žกํ•œ ํ˜ˆ๊ด€ ์˜์ƒ์œผ๋กœ๋ถ€ํ„ฐ ํ•™์Šต ๋ฐ์ดํ„ฐ๋ฅผ ๋งŒ๋“ค๊ธฐ ์–ด๋ ต๊ธฐ ๋•Œ๋ฌธ์— ๋”ฅ ๋Ÿฌ๋‹์„ ์ ์šฉํ•˜๊ธฐ๊ฐ€ ์–ด๋ ต๋‹ค. ๋˜ํ•œ ์–‡์€ ํ˜ˆ๊ด€ ๋ถ€๋ถ„์˜ ์˜์ƒ ๋ฐ๊ธฐ ๋Œ€๋น„๊ฐ€ ์•ฝํ•˜์—ฌ ์›๋ณธ ์˜์ƒ์—์„œ ์‹๋ณ„ํ•˜๊ธฐ๊ฐ€ ๋งค์šฐ ์–ด๋ ต๋‹ค. ๋ณธ ๋…ผ๋ฌธ์—์„œ๋Š” ์œ„ ์–ธ๊ธ‰ํ•œ ๋ฌธ์ œ๋“ค์„ ํ•ด๊ฒฐํ•˜๊ธฐ ์œ„ํ•ด ์ผ๋ฐ˜ํ™” ์„ฑ๋Šฅ์ด ํ–ฅ์ƒ๋œ CNN๊ณผ ์–‡์€ ํ˜ˆ๊ด€์„ ํฌํ•จํ•˜๋Š” ๋ณต์žกํ•œ ๊ฐ„ ํ˜ˆ๊ด€์„ ์ •ํ™•ํ•˜๊ฒŒ ๋ถ„ํ• ํ•˜๋Š” ์•Œ๊ณ ๋ฆฌ์ฆ˜์„ ์ œ์•ˆํ•œ๋‹ค. ๊ฐ„ ๋ถ„ํ•  ์ž‘์—…์—์„œ ์šฐ์ˆ˜ํ•œ ์ผ๋ฐ˜ํ™” ์„ฑ๋Šฅ์„ ๊ฐ–๋Š” CNN์„ ๊ตฌ์ถ•ํ•˜๊ธฐ ์œ„ํ•ด, ๋‚ด๋ถ€์ ์œผ๋กœ ๊ฐ„ ๋ชจ์–‘์„ ์ถ”์ •ํ•˜๋Š” ๋ถ€๋ถ„์ด ํฌํ•จ๋œ ์ž๋™ ์ปจํ…์ŠคํŠธ ์•Œ๊ณ ๋ฆฌ์ฆ˜์„ ์ œ์•ˆํ•œ๋‹ค. ๋˜ํ•œ, CNN์„ ์‚ฌ์šฉํ•œ ํ•™์Šต์— ๊ฒฝ๊ณ„์„ ์˜ ๊ฐœ๋…์ด ์ƒˆ๋กญ๊ฒŒ ์ œ์•ˆ๋œ๋‹ค. ๋ชจํ˜ธํ•œ ๊ฒฝ๊ณ„๋ถ€๊ฐ€ ํฌํ•จ๋˜์–ด ์žˆ์–ด ์ „์ฒด ๊ฒฝ๊ณ„ ์˜์—ญ์„ CNN์— ํ›ˆ๋ จํ•˜๋Š” ๊ฒƒ์€ ๋งค์šฐ ์–ด๋ ต๊ธฐ ๋•Œ๋ฌธ์— ๋ฐ˜๋ณต๋˜๋Š” ํ•™์Šต ๊ณผ์ •์—์„œ ์ธ๊ณต ์‹ ๊ฒฝ๋ง์ด ์Šค์Šค๋กœ ์˜ˆ์ธกํ•œ ํ™•๋ฅ ์—์„œ ๋ถ€์ •ํ™•ํ•˜๊ฒŒ ์ถ”์ •๋œ ๋ถ€๋ถ„์  ๊ฒฝ๊ณ„๋งŒ์„ ์‚ฌ์šฉํ•˜์—ฌ ์ธ๊ณต ์‹ ๊ฒฝ๋ง์„ ํ•™์Šตํ•œ๋‹ค. ์‹คํ—˜์  ๊ฒฐ๊ณผ๋ฅผ ํ†ตํ•ด ์ œ์•ˆ๋œ CNN์ด ๋‹ค๋ฅธ ์ตœ์‹  ๊ธฐ๋ฒ•๋“ค๋ณด๋‹ค ์ •ํ™•๋„๊ฐ€ ์šฐ์ˆ˜ํ•˜๋‹ค๋Š” ๊ฒƒ์„ ๋ณด์ธ๋‹ค. ๋˜ํ•œ, ์ œ์•ˆ๋œ CNN์˜ ์ผ๋ฐ˜ํ™” ์„ฑ๋Šฅ์„ ๊ฒ€์ฆํ•˜๊ธฐ ์œ„ํ•ด ๋‹ค์–‘ํ•œ ์‹คํ—˜์„ ์ˆ˜ํ–‰ํ•œ๋‹ค. ๊ฐ„ ํ˜ˆ๊ด€ ๋ถ„ํ• ์—์„œ๋Š” ๊ฐ„ ๋‚ด๋ถ€์˜ ๊ด€์‹ฌ ์˜์—ญ์„ ์ง€์ •ํ•˜๊ธฐ ์œ„ํ•ด ์•ž์„œ ํš๋“ํ•œ ๊ฐ„ ์˜์—ญ์„ ํ™œ์šฉํ•œ๋‹ค. ์ •ํ™•ํ•œ ๊ฐ„ ํ˜ˆ๊ด€ ๋ถ„ํ• ์„ ์œ„ํ•ด ํ˜ˆ๊ด€ ํ›„๋ณด ์ ๋“ค์„ ์ถ”์ถœํ•˜์—ฌ ์‚ฌ์šฉํ•˜๋Š” ์•Œ๊ณ ๋ฆฌ์ฆ˜์„ ์ œ์•ˆํ•œ๋‹ค. ํ™•์‹คํ•œ ํ›„๋ณด ์ ๋“ค์„ ์–ป๊ธฐ ์œ„ํ•ด, ์‚ผ์ฐจ์› ์˜์ƒ์˜ ์ฐจ์›์„ ๋จผ์ € ์ตœ๋Œ€ ๊ฐ•๋„ ํˆฌ์˜ ๊ธฐ๋ฒ•์„ ํ†ตํ•ด ์ด์ฐจ์›์œผ๋กœ ๋‚ฎ์ถ˜๋‹ค. ์ด์ฐจ์› ์˜์ƒ์—์„œ๋Š” ๋ณต์žกํ•œ ํ˜ˆ๊ด€์˜ ๊ตฌ์กฐ๊ฐ€ ๋ณด๋‹ค ๋‹จ์ˆœํ™”๋  ์ˆ˜ ์žˆ๋‹ค. ์ด์–ด์„œ, ์ด์ฐจ์› ์˜์ƒ์—์„œ ํ˜ˆ๊ด€ ๋ถ„ํ• ์„ ์ˆ˜ํ–‰ํ•˜๊ณ  ํ˜ˆ๊ด€ ํ”ฝ์…€๋“ค์€ ์›๋ž˜์˜ ์‚ผ์ฐจ์› ๊ณต๊ฐ„์ƒ์œผ๋กœ ์—ญ ํˆฌ์˜๋œ๋‹ค. ๋งˆ์ง€๋ง‰์œผ๋กœ, ์ „์ฒด ํ˜ˆ๊ด€์˜ ๋ถ„ํ• ์„ ์œ„ํ•ด ์›๋ณธ ์˜์ƒ๊ณผ ํ˜ˆ๊ด€ ํ›„๋ณด ์ ๋“ค์„ ๋ชจ๋‘ ์‚ฌ์šฉํ•˜๋Š” ์ƒˆ๋กœ์šด ๋ ˆ๋ฒจ ์…‹ ๊ธฐ๋ฐ˜ ์•Œ๊ณ ๋ฆฌ์ฆ˜์„ ์ œ์•ˆํ•œ๋‹ค. ์ œ์•ˆ๋œ ์•Œ๊ณ ๋ฆฌ์ฆ˜์€ ๋ณต์žกํ•œ ๊ตฌ์กฐ๊ฐ€ ๋‹จ์ˆœํ™”๋˜๊ณ  ์–‡์€ ํ˜ˆ๊ด€์ด ๋” ์ž˜ ๋ณด์ด๋Š” ์ด์ฐจ์› ์˜์ƒ์—์„œ ์–ป์€ ํ›„๋ณด ์ ๋“ค์„ ์‚ฌ์šฉํ•˜๊ธฐ ๋•Œ๋ฌธ์— ์–‡์€ ํ˜ˆ๊ด€ ๋ถ„ํ• ์—์„œ ๋†’์€ ์ •ํ™•๋„๋ฅผ ๋ณด์ธ๋‹ค. ์‹คํ—˜์  ๊ฒฐ๊ณผ์— ์˜ํ•˜๋ฉด ์ œ์•ˆ๋œ ์•Œ๊ณ ๋ฆฌ์ฆ˜์€ ์ž˜๋ชป๋œ ์˜์—ญ์˜ ์ถ”์ถœ ์—†์ด ๋‹ค๋ฅธ ๋ ˆ๋ฒจ ์…‹ ๊ธฐ๋ฐ˜ ์•Œ๊ณ ๋ฆฌ์ฆ˜๋“ค๋ณด๋‹ค ์šฐ์ˆ˜ํ•œ ์„ฑ๋Šฅ์„ ๋ณด์ธ๋‹ค. ์ œ์•ˆ๋œ ์•Œ๊ณ ๋ฆฌ์ฆ˜์€ ๊ฐ„๊ณผ ํ˜ˆ๊ด€์„ ๋ถ„ํ• ํ•˜๋Š” ์ƒˆ๋กœ์šด ๋ฐฉ๋ฒ•์„ ์ œ์‹œํ•œ๋‹ค. ์ œ์•ˆ๋œ ์ž๋™ ์ปจํ…์ŠคํŠธ ๊ตฌ์กฐ๋Š” ์‚ฌ๋žŒ์ด ๋””์ž์ธํ•œ ํ•™์Šต ๊ณผ์ •์ด ์ผ๋ฐ˜ํ™” ์„ฑ๋Šฅ์„ ํฌ๊ฒŒ ํ–ฅ์ƒํ•  ์ˆ˜ ์žˆ๋‹ค๋Š” ๊ฒƒ์„ ๋ณด์ธ๋‹ค. ๊ทธ๋ฆฌ๊ณ  ์ œ์•ˆ๋œ ๊ฒฝ๊ณ„์„  ํ•™์Šต ๊ธฐ๋ฒ•์œผ๋กœ CNN์„ ์‚ฌ์šฉํ•œ ์˜์ƒ ๋ถ„ํ• ์˜ ์„ฑ๋Šฅ์„ ํ–ฅ์ƒํ•  ์ˆ˜ ์žˆ์Œ์„ ๋‚ดํฌํ•œ๋‹ค. ๊ฐ„ ํ˜ˆ๊ด€์˜ ๋ถ„ํ• ์€ ์ด์ฐจ์› ์ตœ๋Œ€ ๊ฐ•๋„ ํˆฌ์˜ ๊ธฐ๋ฐ˜ ์ด๋ฏธ์ง€๋กœ๋ถ€ํ„ฐ ํš๋“๋œ ํ˜ˆ๊ด€ ํ›„๋ณด ์ ๋“ค์„ ํ†ตํ•ด ์–‡์€ ํ˜ˆ๊ด€๋“ค์ด ์„ฑ๊ณต์ ์œผ๋กœ ๋ถ„ํ• ๋  ์ˆ˜ ์žˆ์Œ์„ ๋ณด์ธ๋‹ค. ๋ณธ ๋…ผ๋ฌธ์—์„œ ์ œ์•ˆ๋œ ์•Œ๊ณ ๋ฆฌ์ฆ˜์€ ๊ฐ„์˜ ํ•ด๋ถ€ํ•™์  ๋ถ„์„๊ณผ ์ž๋™ํ™”๋œ ์ปดํ“จํ„ฐ ์ง„๋‹จ ๋ณด์กฐ ์‹œ์Šคํ…œ์„ ๊ตฌ์ถ•ํ•˜๋Š” ๋ฐ ๋งค์šฐ ์ค‘์š”ํ•œ ๊ธฐ์ˆ ์ด๋‹ค.Accurate liver and its vessel segmentation on abdominal computed tomography (CT) images is one of the most important prerequisites for computer-aided diagnosis (CAD) systems such as volumetric measurement, treatment planning, and further augmented reality-based surgical guide. In recent years, the application of deep learning in the form of convolutional neural network (CNN) has improved the performance of medical image segmentation, but it is difficult to provide high generalization performance for the actual clinical practice. Furthermore, although the contour features are an important factor in the image segmentation problem, they are hard to be employed on CNN due to many unclear boundaries on the image. In case of a liver vessel segmentation, a deep learning approach is impractical because it is difficult to obtain training data from complex vessel images. Furthermore, thin vessels are hard to be identified in the original image due to weak intensity contrasts and noise. In this dissertation, a CNN with high generalization performance and a contour learning scheme is first proposed for liver segmentation. Secondly, a liver vessel segmentation algorithm is presented that accurately segments even thin vessels. To build a CNN with high generalization performance, the auto-context algorithm is employed. The auto-context algorithm goes through two pipelines: the first predicts the overall area of a liver and the second predicts the final liver using the first prediction as a prior. This process improves generalization performance because the network internally estimates shape-prior. In addition to the auto-context, a contour learning method is proposed that uses only sparse contours rather than the entire contour. Sparse contours are obtained and trained by using only the mispredicted part of the network's final prediction. Experimental studies show that the proposed network is superior in accuracy to other modern networks. Multiple N-fold tests are also performed to verify the generalization performance. An algorithm for accurate liver vessel segmentation is also proposed by introducing vessel candidate points. To obtain confident vessel candidates, the 3D image is first reduced to 2D through maximum intensity projection. Subsequently, vessel segmentation is performed from the 2D images and the segmented pixels are back-projected into the original 3D space. Finally, a new level set function is proposed that utilizes both the original image and vessel candidate points. The proposed algorithm can segment thin vessels with high accuracy by mainly using vessel candidate points. The reliability of the points can be higher through robust segmentation in the projected 2D images where complex structures are simplified and thin vessels are more visible. Experimental results show that the proposed algorithm is superior to other active contour models. The proposed algorithms present a new method of segmenting the liver and its vessels. The auto-context algorithm shows that a human-designed curriculum (i.e., shape-prior learning) can improve generalization performance. The proposed contour learning technique can increase the accuracy of a CNN for image segmentation by focusing on its failures, represented by sparse contours. The vessel segmentation shows that minor vessel branches can be successfully segmented through vessel candidate points obtained by reducing the image dimension. The algorithms presented in this dissertation can be employed for later analysis of liver anatomy that requires accurate segmentation techniques.Chapter 1 Introduction 1 1.1 Background and motivation 1 1.2 Problem statement 3 1.3 Main contributions 6 1.4 Contents and organization 9 Chapter 2 Related Works 10 2.1 Overview 10 2.2 Convolutional neural networks 11 2.2.1 Architectures of convolutional neural networks 11 2.2.2 Convolutional neural networks in medical image segmentation 21 2.3 Liver and vessel segmentation 37 2.3.1 Classical methods for liver segmentation 37 2.3.2 Vascular image segmentation 40 2.3.3 Active contour models 46 2.3.4 Vessel topology-based active contour model 54 2.4 Motivation 60 Chapter 3 Liver Segmentation via Auto-Context Neural Network with Self-Supervised Contour Attention 62 3.1 Overview 62 3.2 Single-pass auto-context neural network 65 3.2.1 Skip-attention module 66 3.2.2 V-transition module 69 3.2.3 Liver-prior inference and auto-context 70 3.2.4 Understanding the network 74 3.3 Self-supervising contour attention 75 3.4 Learning the network 81 3.4.1 Overall loss function 81 3.4.2 Data augmentation 81 3.5 Experimental Results 83 3.5.1 Overview 83 3.5.2 Data configurations and target of comparison 84 3.5.3 Evaluation metric 85 3.5.4 Accuracy evaluation 87 3.5.5 Ablation study 93 3.5.6 Performance of generalization 110 3.5.7 Results from ground-truth variations 114 3.6 Discussion 116 Chapter 4 Liver Vessel Segmentation via Active Contour Model with Dense Vessel Candidates 119 4.1 Overview 119 4.2 Dense vessel candidates 124 4.2.1 Maximum intensity slab images 125 4.2.2 Segmentation of 2D vessel candidates and back-projection 130 4.3 Clustering of dense vessel candidates 135 4.3.1 Virtual gradient-assisted regional ACM 136 4.3.2 Localized regional ACM 142 4.4 Experimental results 145 4.4.1 Overview 145 4.4.2 Data configurations and environment 146 4.4.3 2D segmentation 146 4.4.4 ACM comparisons 149 4.4.5 Evaluation of bifurcation points 154 4.4.6 Computational performance 159 4.4.7 Ablation study 160 4.4.8 Parameter study 162 4.5 Application to portal vein analysis 164 4.6 Discussion 168 Chapter 5 Conclusion and Future Works 170 Bibliography 172 ์ดˆ๋ก 197Docto
    • โ€ฆ
    corecore