Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Bioinform ; 3: 1275402, 2023.
Article in English | MEDLINE | ID: mdl-37928169

ABSTRACT

Introduction: Tissue-based sampling and diagnosis are defined as the extraction of information from certain limited spaces and its diagnostic significance of a certain object. Pathologists deal with issues related to tumor heterogeneity since analyzing a single sample does not necessarily capture a representative depiction of cancer, and a tissue biopsy usually only presents a small fraction of the tumor. Many multiplex tissue imaging platforms (MTIs) make the assumption that tissue microarrays (TMAs) containing small core samples of 2-dimensional (2D) tissue sections are a good approximation of bulk tumors although tumors are not 2D. However, emerging whole slide imaging (WSI) or 3D tumor atlases that use MTIs like cyclic immunofluorescence (CyCIF) strongly challenge this assumption. In spite of the additional insight gathered by measuring the tumor microenvironment in WSI or 3D, it can be prohibitively expensive and time-consuming to process tens or hundreds of tissue sections with CyCIF. Even when resources are not limited, the criteria for region of interest (ROI) selection in tissues for downstream analysis remain largely qualitative and subjective as stratified sampling requires the knowledge of objects and evaluates their features. Despite the fact TMAs fail to adequately approximate whole tissue features, a theoretical subsampling of tissue exists that can best represent the tumor in the whole slide image. Methods: To address these challenges, we propose deep learning approaches to learn multi-modal image translation tasks from two aspects: 1) generative modeling approach to reconstruct 3D CyCIF representation and 2) co-embedding CyCIF image and Hematoxylin and Eosin (H&E) section to learn multi-modal mappings by a cross-domain translation for minimum representative ROI selection. Results and discussion: We demonstrate that generative modeling enables a 3D virtual CyCIF reconstruction of a colorectal cancer specimen given a small subset of the imaging data at training time. By co-embedding histology and MTI features, we propose a simple convex optimization for objective ROI selection. We demonstrate the potential application of ROI selection and the efficiency of its performance with respect to cellular heterogeneity.

3.
Cell Rep Med ; 3(2): 100525, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35243422

ABSTRACT

Mechanisms of therapeutic resistance and vulnerability evolve in metastatic cancers as tumor cells and extrinsic microenvironmental influences change during treatment. To support the development of methods for identifying these mechanisms in individual people, here we present an omic and multidimensional spatial (OMS) atlas generated from four serial biopsies of an individual with metastatic breast cancer during 3.5 years of therapy. This resource links detailed, longitudinal clinical metadata that includes treatment times and doses, anatomic imaging, and blood-based response measurements to clinical and exploratory analyses, which includes comprehensive DNA, RNA, and protein profiles; images of multiplexed immunostaining; and 2- and 3-dimensional scanning electron micrographs. These data report aspects of heterogeneity and evolution of the cancer genome, signaling pathways, immune microenvironment, cellular composition and organization, and ultrastructure. We present illustrative examples of how integrative analyses of these data reveal potential mechanisms of response and resistance and suggest novel therapeutic vulnerabilities.


Subject(s)
Breast Neoplasms , Biopsy , Breast Neoplasms/genetics , Female , Humans , Tumor Microenvironment/genetics
4.
Comput Med Imaging Graph ; 95: 102013, 2022 01.
Article in English | MEDLINE | ID: mdl-34864359

ABSTRACT

Emerging multiplexed imaging platforms provide an unprecedented view of an increasing number of molecular markers at subcellular resolution and the dynamic evolution of tumor cellular composition. As such, they are capable of elucidating cell-to-cell interactions within the tumor microenvironment that impact clinical outcome and therapeutic response. However, the rapid development of these platforms has far outpaced the computational methods for processing and analyzing the data they generate. While being technologically disparate, all imaging assays share many computational requirements for post-collection data processing. As such, our Image Analysis Working Group (IAWG), composed of researchers in the Cancer Systems Biology Consortium (CSBC) and the Physical Sciences - Oncology Network (PS-ON), convened a workshop on "Computational Challenges Shared by Diverse Imaging Platforms" to characterize these common issues and a follow-up hackathon to implement solutions for a selected subset of them. Here, we delineate these areas that reflect major axes of research within the field, including image registration, segmentation of cells and subcellular structures, and identification of cell types from their morphology. We further describe the logistical organization of these events, believing our lessons learned can aid others in uniting the imaging community around self-identified topics of mutual interest, in designing and implementing operational procedures to address those topics and in mitigating issues inherent in image analysis (e.g., sharing exemplar images of large datasets and disseminating baseline solutions to hackathon challenges through open-source code repositories).


Subject(s)
Image Processing, Computer-Assisted , Neoplasms , Diagnostic Imaging , Humans , Image Processing, Computer-Assisted/methods , Neoplasms/diagnostic imaging , Software , Tumor Microenvironment
5.
Cell Rep Methods ; 1(4)2021 08 23.
Article in English | MEDLINE | ID: mdl-34485971

ABSTRACT

The emergence of megascale single-cell multiplex tissue imaging (MTI) datasets necessitates reproducible, scalable, and robust tools for cell phenotyping and spatial analysis. We developed open-source, graphics processing unit (GPU)-accelerated tools for intensity normalization, phenotyping, and microenvironment characterization. We deploy the toolkit on a human breast cancer (BC) tissue microarray stained by cyclic immunofluorescence and present the first cross-validation of breast cancer cell phenotypes derived by using two different MTI platforms. Finally, we demonstrate an integrative phenotypic and spatial analysis revealing BC subtype-specific features.


Subject(s)
Breast Neoplasms , Diagnostic Imaging , Humans , Female , Breast Neoplasms/diagnostic imaging , Phenotype , Microarray Analysis , Fluorescent Antibody Technique , Tumor Microenvironment
6.
Sci Rep ; 11(1): 13630, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34211050

ABSTRACT

Metastatic progression defines the final stages of tumor evolution and underlies the majority of cancer-related deaths. The heterogeneity in disseminated tumor cell populations capable of seeding and growing in distant organ sites contributes to the development of treatment resistant disease. We recently reported the identification of a novel tumor-derived cell population, circulating hybrid cells (CHCs), harboring attributes from both macrophages and neoplastic cells, including functional characteristics important to metastatic spread. These disseminated hybrids outnumber conventionally defined circulating tumor cells (CTCs) in cancer patients. It is unknown if CHCs represent a generalized cancer mechanism for cell dissemination, or if this population is relevant to the metastatic cascade. Herein, we detect CHCs in the peripheral blood of patients with cancer in myriad disease sites encompassing epithelial and non-epithelial malignancies. Further, we demonstrate that in vivo-derived hybrid cells harbor tumor-initiating capacity in murine cancer models and that CHCs from human breast cancer patients express stem cell antigens, features consistent with the potential to seed and grow at metastatic sites. Finally, we reveal heterogeneity of CHC phenotypes reflect key tumor features, including oncogenic mutations and functional protein expression. Importantly, this novel population of disseminated neoplastic cells opens a new area in cancer biology and renewed opportunity for battling metastatic disease.


Subject(s)
Hybrid Cells/pathology , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cells, Cultured , Child , Child, Preschool , Female , Humans , Mice , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasms/blood
7.
Sci Rep ; 10(1): 17507, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060677

ABSTRACT

Spatially-resolved molecular profiling by immunostaining tissue sections is a key feature in cancer diagnosis, subtyping, and treatment, where it complements routine histopathological evaluation by clarifying tumor phenotypes. In this work, we present a deep learning-based method called speedy histological-to-immunofluorescent translation (SHIFT) which takes histologic images of hematoxylin and eosin (H&E)-stained tissue as input, then in near-real time returns inferred virtual immunofluorescence (IF) images that estimate the underlying distribution of the tumor cell marker pan-cytokeratin (panCK). To build a dataset suitable for learning this task, we developed a serial staining protocol which allows IF and H&E images from the same tissue to be spatially registered. We show that deep learning-extracted morphological feature representations of histological images can guide representative sample selection, which improved SHIFT generalizability in a small but heterogenous set of human pancreatic cancer samples. With validation in larger cohorts, SHIFT could serve as an efficient preliminary, auxiliary, or substitute for panCK IF by delivering virtual panCK IF images for a fraction of the cost and in a fraction of the time required by traditional IF.


Subject(s)
Coloring Agents/chemistry , Deep Learning , Image Processing, Computer-Assisted/methods , Pancreatic Neoplasms/diagnostic imaging , Actins/analysis , Aged , Algorithms , Biomarkers, Tumor/analysis , Female , Humans , Keratins/analysis , Microscopy, Fluorescence , Middle Aged , Pancreatic Neoplasms/pathology , Phenotype , Staining and Labeling
8.
Commun Biol ; 3(1): 111, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152447

ABSTRACT

Recent advances in multiplexed imaging technologies promise to improve the understanding of the functional states of individual cells and the interactions between the cells in tissues. This often requires compilation of results from multiple samples. However, quantitative integration of information between samples is complicated by variations in staining intensity and background fluorescence that obscure biological variations. Failure to remove these unwanted artifacts will complicate downstream analysis and diminish the value of multiplexed imaging for clinical applications. Here, to compensate for unwanted variations, we automatically identify negative control cells for each marker within the same tissue and use their expression levels to infer background signal level. The intensity profile is normalized by the inferred level of the negative control cells to remove between-sample variation. Using a tissue microarray data and a pair of longitudinal biopsy samples, we demonstrated that the proposed approach can remove unwanted variations effectively and shows robust performance.


Subject(s)
Algorithms , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Image Interpretation, Computer-Assisted , Immunohistochemistry , Microscopy , Artifacts , Automation, Laboratory , Biopsy , Breast Neoplasms/pathology , Cluster Analysis , Feasibility Studies , Tissue Array Analysis
9.
J Med Imaging (Bellingham) ; 7(1): 012706, 2020 Jan.
Article in English | MEDLINE | ID: mdl-34541020

ABSTRACT

Purpose: Pathologists rely on relevant clinical information, visual inspection of stained tissue slide morphology, and sophisticated molecular diagnostics to accurately infer the biological origin of secondary metastatic cancer. While highly effective, this process is expensive in terms of time and clinical resources. We seek to develop and evaluate a computer vision system designed to reasonably infer metastatic origin of secondary liver cancer directly from digitized histopathological whole slide images of liver biopsy. Approach: We illustrate a two-stage deep learning approach to accomplish this task. We first train a model to identify spatially localized regions of cancerous tumor within digitized hematoxylin and eosin (H&E)-stained tissue sections of secondary liver cancer based on a pathologist's annotation of several whole slide images. Then, a second model is trained to generate predictions of the cancers' metastatic origin belonging to one of three distinct clinically relevant classes as confirmed by immunohistochemistry. Results: Our approach achieves a classification accuracy of 90.2% in determining metastatic origin of whole slide images from a held-out test set, which compares favorably to an established clinical benchmark by three board-certified pathologists whose accuracies ranged from 90.2% to 94.1% on the same prediction task. Conclusions: We illustrate the potential impact of deep learning systems to leverage morphological and structural features of H&E-stained tissue sections to guide pathological and clinical determination of the metastatic origin of secondary liver cancers.

10.
Article in English | MEDLINE | ID: mdl-30283195

ABSTRACT

Multiplexed imaging such as multicolor immunofluorescence staining, multiplexed immunohistochemistry (mIHC) or cyclic immunofluorescence (cycIF) enables deep assessment of cellular complexity in situ and, in conjunction with standard histology stains like hematoxylin and eosin (H&E), can help to unravel the complex molecular relationships and spatial interdependencies that undergird disease states. However, these multiplexed imaging methods are costly and can degrade both tissue quality and antigenicity with each successive cycle of staining. In addition, computationally intensive image processing such as image registration across multiple channels is required. We have developed a novel method, speedy histopathological-to-immunofluorescent translation (SHIFT) of whole slide images (WSIs) using conditional generative adversarial networks (cGANs). This approach is rooted in the assumption that specific patterns captured in IF images by stains like DAPI, pan-cytokeratin (panCK), or α-smooth muscle actin ( α-SMA) are encoded in H&E images, such that a SHIFT model can learn useful feature representations or architectural patterns in the H&E stain that help generate relevant IF stain patterns. We demonstrate that the proposed method is capable of generating realistic tumor marker IF WSIs conditioned on corresponding H&E-stained WSIs with up to 94.5% accuracy in a matter of seconds. Thus, this method has the potential to not only improve our understanding of the mapping of histological and morphological profiles into protein expression profiles, but also greatly increase the e ciency of diagnostic and prognostic decision-making.

SELECTION OF CITATIONS
SEARCH DETAIL
...