Do you want to publish a course? Click here

High-resolution Image Registration of Consecutive and Re-stained Sections in Histopathology

123   0   0.0 ( 0 )
 Added by Johannes Lotz
 Publication date 2021
and research's language is English




Ask ChatGPT about the research

We compare variational image registration in consectutive and re-stained sections from histopathology. We present a fully-automatic algorithm for non-parametric (nonlinear) image registration and apply it to a previously existing dataset from the ANHIR challenge (230 slide pairs, consecutive sections) and a new dataset (hybrid re-stained and consecutive, 81 slide pairs, ca. 3000 landmarks) which is made publicly available. Registration hyperparameters are obtained in the ANHIR dataset and applied to the new dataset without modification. In the new dataset, landmark errors after registration range from 13.2 micrometers for consecutive sections to 1 micrometer for re-stained sections. We observe that non-parametric registration leads to lower landmark errors in both cases, even though the effect is smaller in re-stained sections. The nucleus-level alignment after non-parametric registration of re-stained sections provides a valuable tool to generate automatic ground-truth for machine learning applications in histopathology.

rate research

Read More

135 - Zhe Xu , Jie Luo , Jiangpeng Yan 2020
Deformable image registration (DIR) is essential for many image-guided therapies. Recently, deep learning approaches have gained substantial popularity and success in DIR. Most deep learning approaches use the so-called mono-stream high-to-low, low-to-high network structure, and can achieve satisfactory overall registration results. However, accurate alignments for some severely deformed local regions, which are crucial for pinpointing surgical targets, are often overlooked. Consequently, these approaches are not sensitive to some hard-to-align regions, e.g., intra-patient registration of deformed liver lobes. In this paper, we propose a novel unsupervised registration network, namely the Full-Resolution Residual Registration Network (F3RNet), for deformable registration of severely deformed organs. The proposed method combines two parallel processing streams in a residual learning fashion. One stream takes advantage of the full-resolution information that facilitates accurate voxel-level registration. The other stream learns the deep multi-scale residual representations to obtain robust recognition. We also factorize the 3D convolution to reduce the training parameters and enhance network efficiency. We validate the proposed method on a clinically acquired intra-patient abdominal CT-MRI dataset and a public inspiratory and expiratory thorax CT dataset. Experiments on both multimodal and unimodal registration demonstrate promising results compared to state-of-the-art approaches.
The distribution and appearance of nuclei are essential markers for the diagnosis and study of cancer. Despite the importance of nuclear morphology, there is a lack of large scale, accurate, publicly accessible nucleus segmentation data. To address this, we developed an analysis pipeline that segments nuclei in whole slide tissue images from multiple cancer types with a quality control process. We have generated nucleus segmentation results in 5,060 Whole Slide Tissue images from 10 cancer types in The Cancer Genome Atlas. One key component of our work is that we carried out a multi-level quality control process (WSI-level and image patch-level), to evaluate the quality of our segmentation results. The image patch-level quality control used manual segmentation ground truth data from 1,356 sampled image patches. The datasets we publish in this work consist of roughly 5 billion quality controlled nuclei from more than 5,060 TCGA WSIs from 10 different TCGA cancer types and 1,356 manually segmented TCGA image patches from the same 10 cancer types plus additional 4 cancer types. Data is available at https://doi.org/10.7937/tcia.2019.4a4dkp9u
Joint analysis of multiple biomarker images and tissue morphology is important for disease diagnosis, treatment planning and drug development. It requires cross-staining comparison among Whole Slide Images (WSIs) of immuno-histochemical and hematoxylin and eosin (H&E) microscopic slides. However, automatic, and fast cross-staining alignment of enormous gigapixel WSIs at single-cell precision is challenging. In addition to morphological deformations introduced during slide preparation, there are large variations in cell appearance and tissue morphology across different staining. In this paper, we propose a two-step automatic feature-based cross-staining WSI alignment to assist localization of even tiny metastatic foci in the assessment of lymph node. Image pairs were aligned allowing for translation, rotation, and scaling. The registration was performed automatically by first detecting landmarks in both images, using the scale-invariant image transform (SIFT), followed by the fast sample consensus (FSC) protocol for finding point correspondences and finally aligned the images. The Registration results were evaluated using both visual and quantitative criteria using the Jaccard index. The average Jaccard similarity index of the results produced by the proposed system is 0.942 when compared with the manual registration.
Histopathological images provide rich information for disease diagnosis. Large numbers of histopathological images have been digitized into high resolution whole slide images, opening opportunities in developing computational image analysis tools to reduce pathologists workload and potentially improve inter- and intra- observer agreement. Most previous work on whole slide image analysis has focused on classification or segmentation of small pre-selected regions-of-interest, which requires fine-grained annotation and is non-trivial to extend for large-scale whole slide analysis. In this paper, we proposed a multi-resolution multiple instance learning model that leverages saliency maps to detect suspicious regions for fine-grained grade prediction. Instead of relying on expensive region- or pixel-level annotations, our model can be trained end-to-end with only slide-level labels. The model is developed on a large-scale prostate biopsy dataset containing 20,229 slides from 830 patients. The model achieved 92.7% accuracy, 81.8% Cohens Kappa for benign, low grade (i.e. Grade group 1) and high grade (i.e. Grade group >= 2) prediction, an area under the receiver operating characteristic curve (AUROC) of 98.2% and an average precision (AP) of 97.4% for differentiating malignant and benign slides. The model obtained an AUROC of 99.4% and an AP of 99.8% for cancer detection on an external dataset.
Classical pairwise image registration methods search for a spatial transformation that optimises a numerical measure that indicates how well a pair of moving and fixed images are aligned. Current learning-based registration methods have adopted the same paradigm and typically predict, for any new input image pair, dense correspondences in the form of a dense displacement field or parameters of a spatial transformation model. However, in many applications of registration, the spatial transformation itself is only required to propagate points or regions of interest (ROIs). In such cases, detailed pixel- or voxel-level correspondence within or outside of these ROIs often have little clinical value. In this paper, we propose an alternative paradigm in which the location of corresponding image-specific ROIs, defined in one image, within another image is learnt. This results in replacing image registration by a conditional segmentation algorithm, which can build on typical image segmentation networks and their widely-adopted training strategies. Using the registration of 3D MRI and ultrasound images of the prostate as an example to demonstrate this new approach, we report a median target registration error (TRE) of 2.1 mm between the ground-truth ROIs defined on intraoperative ultrasound images and those propagated from the preoperative MR images. Significantly lower (>34%) TREs were obtained using the proposed conditional segmentation compared with those obtained from a previously-proposed spatial-transformation-predicting registration network trained with the same multiple ROI labels for individual image pairs. We conclude this work by using a quantitative bias-variance analysis to provide one explanation of the observed improvement in registration accuracy.
comments
Fetching comments Fetching comments
Sign in to be able to follow your search criteria
mircosoft-partner

هل ترغب بارسال اشعارات عن اخر التحديثات في شمرا-اكاديميا