31 research outputs found

    Differently stained whole slide image registration technique with landmark validation

    Get PDF
    Abstract. One of the most significant features in digital pathology is to compare and fuse successive differently stained tissue sections, also called slides, visually. Doing so, aligning different images to a common frame, ground truth, is required. Current sample scanning tools enable to create images full of informative layers of digitalized tissues, stored with a high resolution into whole slide images. However, there are a limited amount of automatic alignment tools handling large images precisely in acceptable processing time. The idea of this study is to propose a deep learning solution for histopathology image registration. The main focus is on the understanding of landmark validation and the impact of stain augmentation on differently stained histopathology images. Also, the developed registration method is compared with the state-of-the-art algorithms which utilize whole slide images in the field of digital pathology. There are previous studies about histopathology, digital pathology, whole slide imaging and image registration, color staining, data augmentation, and deep learning that are referenced in this study. The goal is to develop a learning-based registration framework specifically for high-resolution histopathology image registration. Different whole slide tissue sample images are used with a resolution of up to 40x magnification. The images are organized into sets of consecutive, differently dyed sections, and the aim is to register the images based on only the visible tissue and ignore the background. Significant structures in the tissue are marked with landmarks. The quality measurements include, for example, the relative target registration error, structural similarity index metric, visual evaluation, landmark-based evaluation, matching points, and image details. These results are comparable and can be used also in the future research and in development of new tools. Moreover, the results are expected to show how the theory and practice are combined in whole slide image registration challenges. DeepHistReg algorithm will be studied to better understand the development of stain color feature augmentation-based image registration tool of this study. Matlab and Aperio ImageScope are the tools to annotate and validate the image, and Python is used to develop the algorithm of this new registration tool. As cancer is globally a serious disease regardless of age or lifestyle, it is important to find ways to develop the systems experts can use while working with patients’ data. There is still a lot to improve in the field of digital pathology and this study is one step toward it.Eri menetelmin värjättyjen virtuaalinäytelasien rekisteröintitekniikka kiintopisteiden validointia hyödyntäen. Tiivistelmä. Yksi tärkeimmistä digitaalipatologian ominaisuuksista on verrata ja fuusioida peräkkäisiä eri menetelmin värjättyjä kudosleikkeitä toisiinsa visuaalisesti. Tällöin keskenään lähes identtiset kuvat kohdistetaan samaan yhteiseen kehykseen, niin sanottuun pohjatotuuteen. Nykyiset näytteiden skannaustyökalut mahdollistavat sellaisten kuvien luonnin, jotka ovat täynnä kerroksittaista tietoa digitalisoiduista näytteistä, tallennettuna erittäin korkean resoluution virtuaalisiin näytelaseihin. Tällä hetkellä on olemassa kuitenkin vain kourallinen automaattisia työkaluja, jotka kykenevät käsittelemään näin valtavia kuvatiedostoja tarkasti hyväksytyin aikarajoin. Tämän työn tarkoituksena on syväoppimista hyväksikäyttäen löytää ratkaisu histopatologisten kuvien rekisteröintiin. Tärkeimpänä osa-alueena on ymmärtää kiintopisteiden validoinnin periaatteet sekä eri väriaineiden augmentoinnin vaikutus. Lisäksi tässä työssä kehitettyä rekisteröintialgoritmia tullaan vertailemaan muihin kirjallisuudessa esitettyihin algoritmeihin, jotka myös hyödyntävät virtuaalinäytelaseja digitaalipatologian saralla. Kirjallisessa osiossa tullaan siteeraamaan aiempia tutkimuksia muun muassa seuraavista aihealueista: histopatologia, digitaalipatologia, virtuaalinäytelasi, kuvantaminen ja rekisteröinti, näytteen värjäys, data-augmentointi sekä syväoppiminen. Tavoitteena on kehittää oppimispohjainen rekisteröintikehys erityisesti korkearesoluutioisille digitalisoiduille histopatologisille kuville. Erilaisissa näytekuvissa tullaan käyttämään jopa 40-kertaista suurennosta. Kuvat kudoksista on järjestetty eri menetelmin värjättyihin peräkkäisiin kuvasarjoihin ja tämän työn päämääränä on rekisteröidä kuvat pohjautuen ainoastaan kudosten näkyviin osuuksiin, jättäen kuvien tausta huomioimatta. Kudosten merkittävimmät rakenteet on merkattu niin sanotuin kiintopistein. Työn laatumittauksina käytetään arvoja, kuten kohteen suhteellinen rekisteröintivirhe (rTRE), rakenteellisen samankaltaisuuindeksin mittari (SSIM), sekä visuaalista arviointia, kiintopisteisiin pohjautuvaa arviointia, yhteensopivuuskohtia, ja kuvatiedoston yksityiskohtia. Nämä arvot ovat verrattavissa myös tulevissa tutkimuksissa ja samaisia arvoja voidaan käyttää uusia työkaluja kehiteltäessä. DeepHistReg metodi toimii pohjana tässä työssä kehitettävälle näytteen värjäyksen parantamiseen pohjautuvalle rekisteröintityökalulle. Matlab ja Aperio ImageScope ovat ohjelmistoja, joita tullaan hyödyntämään tässä työssä kuvien merkitsemiseen ja validointiin. Ohjelmointikielenä käytetään Pythonia. Syöpä on maailmanlaajuisesti vakava sairaus, joka ei katso ikää eikä elämäntyyliä. Siksi on tärkeää löytää uusia keinoja kehittää työkaluja, joita asiantuntijat voivat hyödyntää jokapäiväisessä työssään potilastietojen käsittelyssä. Digitaalipatologian osa-alueella on vielä paljon innovoitavaa ja tämä työ on yksi askel eteenpäin taistelussa syöpäsairauksia vastaan

    Translating AI to digital pathology workflow: Dealing with scarce data and high variation by minimising complexities in data and models

    Get PDF
    The recent conversion to digital pathology using Whole Slide Images (WSIs) from conventional pathology opened the doors for Artificial Intelligence (AI) in pathology workflow. The recent interests in machine learning and deep learning have gained a high interest in medical image processing. However, WSIs differ from generic medical images. WSIs are complex images which can reveal various information to support different diagnosis varying from cancer to unknown underlying conditions which were not discovered in other medical investigations. These investigations require expert knowledge spending a long time for investigations, applying different stains to the WSIs, and comparing the WSIs. Differences in WSI differentiate general machine learning methods that are applied for medical image processing. Co-analysing multistained WSIs, high variation of the WSIs from different sites, and lack of labelled data are the main key interest areas that directly influence in developing machine learning models that support pathologists in their investigations. However, most of the state-ofthe- art machine learning approaches cannot be applied in the general clinical workflow without using high compute power, expert knowledge, and time. Therefore, this thesis explores avenues to translate the highly computational and time intensive model to a clinical workflow. Co-analysing multi-stained WSIs require registering differently stained WSI together. In order to get a high precision in the registration exploring nonrigid and rigid transformation is required. The non-rigid transformation requires complex deep learning approaches. Using super-convergence on a small Convolutional Neural Network model it is possible to achieve high precision compared to larger auto-encoders and other state-of-the-art models. High variation of the WSIs from different sites heavily effect machine learning models in their predictions. The thesis presents an approach of using a pre-trained model by using only a small number of samples from the new site. Therefore, re-training larger deep learning models are not required which saves expert time for re-labelling and computational power. Finally, lack of labelled data is one of the main issues in training any supervised machine learning or deep learning model. Using a Generative Adversarial Networks (GAN) is an approach which can be easily implemented to avoid this issue. However, GANs are time and computationally expensive. These are not applicable in a general clinical workflow. Therefore, this thesis presents an approach using a simpler GANthat can generate accurate sample labelled data. The synthetic data are used to train classifier and the thesis demonstrates that the predictive model can generate higher accuracy in the test environment. This thesis demonstrates that machine learning and deep learning models can be applied to a clinical workflow, without exploiting expert time and high computing power

    Registration of serial sections: An evaluation method based on distortions of the ground truths

    Get PDF
    Registration of histological serial sections is a challenging task. Serial sections exhibit distortions and damage from sectioning. Missing information on how the tissue looked before cutting makes a realistic validation of 2D registrations extremely difficult. This work proposes methods for ground-truth-based evaluation of registrations. Firstly, we present a methodology to generate test data for registrations. We distort an innately registered image stack in the manner similar to the cutting distortion of serial sections. Test cases are generated from existing 3D data sets, thus the ground truth is known. Secondly, our test case generation premises evaluation of the registrations with known ground truths. Our methodology for such an evaluation technique distinguishes this work from other approaches. Both under- and over-registration become evident in our evaluations. We also survey existing validation efforts. We present a full-series evaluation across six different registration methods applied to our distorted 3D data sets of animal lungs. Our distorted and ground truth data sets are made publicly available.Comment: Supplemental data available under https://zenodo.org/record/428244

    Patch-based nonlinear image registration for gigapixel whole slide images

    Get PDF
    Producción CientíficaImage registration of whole slide histology images allows the fusion of fine-grained information-like different immunohistochemical stains-from neighboring tissue slides. Traditionally, pathologists fuse this information by looking subsequently at one slide at a time. If the slides are digitized and accurately aligned at cell level, automatic analysis can be used to ease the pathologist's work. However, the size of those images exceeds the memory capacity of regular computers. Methods: We address the challenge to combine a global motion model that takes the physical cutting process of the tissue into account with image data that is not simultaneously globally available. Typical approaches either reduce the amount of data to be processed or partition the data into smaller chunks to be processed separately. Our novel method first registers the complete images on a low resolution with a nonlinear deformation model and later refines this result on patches by using a second nonlinear registration on each patch. Finally, the deformations computed on all patches are combined by interpolation to form one globally smooth nonlinear deformation. The NGF distance measure is used to handle multistain images. Results: The method is applied to ten whole slide image pairs of human lung cancer data. The alignment of 85 corresponding structures is measured by comparing manual segmentations from neighboring slides. Their offset improves significantly, by at least 15%, compared to the low-resolution nonlinear registration. Conclusion/Significance: The proposed method significantly improves the accuracy of multistain registration which allows us to compare different antibodies at cell level

    DEVELOPING NOVEL COMPUTER-AIDED DETECTION AND DIAGNOSIS SYSTEMS OF MEDICAL IMAGES

    Get PDF
    Reading medical images to detect and diagnose diseases is often difficult and has large inter-reader variability. To address this issue, developing computer-aided detection and diagnosis (CAD) schemes or systems of medical images has attracted broad research interest in the last several decades. Despite great effort and significant progress in previous studies, only limited CAD schemes have been used in clinical practice. Thus, developing new CAD schemes is still a hot research topic in medical imaging informatics field. In this dissertation, I investigate the feasibility of developing several new innovative CAD schemes for different application purposes. First, to predict breast tumor response to neoadjuvant chemotherapy and reduce unnecessary aggressive surgery, I developed two CAD schemes of breast magnetic resonance imaging (MRI) to generate quantitative image markers based on quantitative analysis of global kinetic features. Using the image marker computed from breast MRI acquired pre-chemotherapy, CAD scheme enables to predict radiographic complete response (CR) of breast tumors to neoadjuvant chemotherapy, while using the imaging marker based on the fusion of kinetic and texture features extracted from breast MRI performed after neoadjuvant chemotherapy, CAD scheme can better predict the pathologic complete response (pCR) of the patients. Second, to more accurately predict prognosis of stroke patients, quantifying brain hemorrhage and ventricular cerebrospinal fluid depicting on brain CT images can play an important role. For this purpose, I developed a new interactive CAD tool to segment hemorrhage regions and extract radiological imaging marker to quantitatively determine the severity of aneurysmal subarachnoid hemorrhage at presentation and correlate the estimation with various homeostatic/metabolic derangements and predict clinical outcome. Third, to improve the efficiency of primary antibody screening processes in new cancer drug development, I developed a CAD scheme to automatically identify the non-negative tissue slides, which indicate reactive antibodies in digital pathology images. Last, to improve operation efficiency and reliability of storing digital pathology image data, I developed a CAD scheme using optical character recognition algorithm to automatically extract metadata from tissue slide label images and reduce manual entry for slide tracking and archiving in the tissue pathology laboratories. In summary, in these studies, we developed and tested several innovative approaches to identify quantitative imaging markers with high discriminatory power. In all CAD schemes, the graphic user interface-based visual aid tools were also developed and implemented. Study results demonstrated feasibility of applying CAD technology to several new application fields, which has potential to assist radiologists, oncologists and pathologists improving accuracy and consistency in disease diagnosis and prognosis assessment of using medical image

    Registration of histology and magnetic resonance imaging of the brain

    Get PDF
    Combining histology and non-invasive imaging has been attracting the attention of the medical imaging community for a long time, due to its potential to correlate macroscopic information with the underlying microscopic properties of tissues. Histology is an invasive procedure that disrupts the spatial arrangement of the tissue components but enables visualisation and characterisation at a cellular level. In contrast, macroscopic imaging allows non-invasive acquisition of volumetric information but does not provide any microscopic details. Through the establishment of spatial correspondences obtained via image registration, it is possible to compare micro- and macroscopic information and to recover the original histological arrangement in three dimensions. In this thesis, I present: (i) a survey of the literature relative to methods for histology reconstruction with and without the help of 3D medical imaging; (ii) a graph-theoretic method for histology volume reconstruction from sets of 2D sections, without external information; (iii) a method for multimodal 2D linear registration between histology and MRI based on partial matching of shape-informative boundaries

    A survey on artificial intelligence in histopathology image analysis

    Get PDF
    The increasing adoption of the whole slide image (WSI) technology in histopathology has dramatically transformed pathologists' workflow and allowed the use of computer systems in histopathology analysis. Extensive research in Artificial Intelligence (AI) with a huge progress has been conducted resulting in efficient, effective, and robust algorithms for several applications including cancer diagnosis, prognosis, and treatment. These algorithms offer highly accurate predictions but lack transparency, understandability, and actionability. Thus, explainable artificial intelligence (XAI) techniques are needed not only to understand the mechanism behind the decisions made by AI methods and increase user trust but also to broaden the use of AI algorithms in the clinical setting. From the survey of over 150 papers, we explore different AI algorithms that have been applied and contributed to the histopathology image analysis workflow. We first address the workflow of the histopathological process. We present an overview of various learning-based, XAI, and actionable techniques relevant to deep learning methods in histopathological imaging. We also address the evaluation of XAI methods and the need to ensure their reliability on the field

    A model-based cortical parcellation scheme for high-resolution 7 Tesla MRI data

    No full text

    Towards automated three-dimensional tracking of nephrons through stacked histological image sets

    Get PDF
    A dissertation submitted to the Faculty of Engineering and the Built Environment, University of Witwatersrand for the degree of Master of Science in Engineering. August, 2015The three-dimensional microarchitecture of the mammalian kidney is of keen interest in the fields of cell biology and biomedical engineering as it plays a crucial role in renal function. This study presents a novel approach to the automatic tracking of individual nephrons through three-dimensional histological image sets of mouse and rat kidneys. The image database forms part of a previous study carried out at the University of Aarhus, Denmark. The previous study involved manually tracking a few hundred nephrons through the image sets in order to explore the renal microarchitecture, the results of which forms the gold standard for this study. The purpose of the current research is to develop methods which contribute towards creating an automated, intelligent system as a standard tool for such image sets. This would reduce the excessive time and human effort previously required for the tracking task, enabling a larger sample of nephrons to be tracked. It would also be desirable, in future, to explore the renal microstructure of various species and diseased specimens. The developed algorithm is robust, able to isolate closely packed nephrons and track their convoluted paths despite a number of non-ideal conditions such as local image distortions, artefacts and connective tissue interference. The system consists of initial image pre-processing steps such as background removal, adaptive histogram equalisation and image segmentation. A feature extraction stage achieves data abstraction and information concentration by extracting shape iii descriptors, radial shape profiles and key coordinates for each nephron crosssection. A custom graph-based tracking algorithm is implemented to track the nephrons using the extracted coordinates. A rule-base and machine learning algorithms including an Artificial Neural Network and Support Vector Machine are used to evaluate the shape features and other information to validate the algorithm’s results through each of its iterations. The validation steps prove to be highly effective in rejecting incorrect tracking moves, with the rule-base having greater than 90% accuracy and the Artificial Neural Network and Support Vector Machine both producing 93% classification accuracies. Comparison of a selection of automatically and manually tracked nephrons yielded results of 95% accuracy and 98% tracking extent for the proximal convoluted tubule, proximal straight tubule and ascending thick limb of the loop of Henle. The ascending and descending thin limbs of the loop of Henle pose a challenge, having low accuracy and low tracking extent due to the low resolution, narrow diameter and high density of cross-sections in the inner medulla. Limited manual intervention is proposed as a solution to these limitations, enabling full nephron paths to be obtained with an average of 17 manual corrections per mouse nephron and 58 manual corrections per rat nephron. The developed semi-automatic system saves a considerable amount of time and effort in comparison with the manual task. Furthermore, the developed methodology forms a foundation for future development towards a fully automated tracking system for nephrons
    corecore