Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 193(8): 1072-1080, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37236505

RESUMEN

The incidence of nonalcoholic fatty liver disease is a continuously growing health problem worldwide, along with obesity. Therefore, novel methods to both efficiently study the manifestation of nonalcoholic fatty liver disease and to analyze drug efficacy in preclinical models are needed. The present study developed a deep neural network-based model to quantify microvesicular and macrovesicular steatosis in the liver on hematoxylin-eosin-stained whole slide images, using the cloud-based platform, Aiforia Create. The training data included a total of 101 whole slide images from dietary interventions of wild-type mice and from two genetically modified mouse models with steatosis. The algorithm was trained for the following: to detect liver parenchyma, to exclude the blood vessels and any artefacts generated during tissue processing and image acquisition, to recognize and differentiate the areas of microvesicular and macrovesicular steatosis, and to quantify the recognized tissue area. The results of the image analysis replicated well the evaluation by expert pathologists and correlated well with the liver fat content measured by EchoMRI ex vivo, and the correlation with total liver triglycerides was notable. In conclusion, the developed deep learning-based model is a novel tool for studying liver steatosis in mouse models on paraffin sections and, thus, can facilitate reliable quantification of the amount of steatosis in large preclinical study cohorts.


Asunto(s)
Aprendizaje Profundo , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/diagnóstico por imagen , Hígado , Redes Neurales de la Computación , Algoritmos , Modelos Animales de Enfermedad
2.
Cancer Cell Int ; 24(1): 29, 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38218884

RESUMEN

PURPOSE: Platinum-based drugs are cytotoxic drugs commonly used in cancer treatment. They cause DNA damage, effects of which on chromatin and cellular responses are relatively well described. Yet, the nuclear stress responses related to RNA processing are incompletely known and may be relevant for the heterogeneity with which cancer cells respond to these drugs. Here, we determine the type and extent of nuclear stress responses of prostate cancer cells to clinically relevant platinum drugs. METHODS: We study nucleolar and Cajal body (CB) responses to cisplatin, carboplatin, and oxaliplatin with immunofluorescence-based methods in prostate cancer cells. We utilize organelle-specific markers NPM, Fibrillarin, Coilin, and SMN1, and study CB-regulatory proteins FUS and TDP-43 using siRNA-mediated downregulation. RESULTS: Different types of prostate cancer cells have different sensitivities to platinum drugs. With equally cytotoxic doses, cisplatin, and oxaliplatin induce prominent nucleolar and CB stress responses while the nuclear stress phenotypes to carboplatin are milder. We find that Coilin is a stress-specific marker for platinum drug response heterogeneity. We also find that CB-associated, stress-responsive RNA binding proteins FUS and TDP-43 control Coilin and CB biology in prostate cancer cells and, further, that TDP-43 is associated with stress-responsive CBs in prostate cancer cells. CONCLUSION: Our findings provide insight into the heterologous responses of prostate cancer cells to different platinum drug treatments and indicate Coilin and TDP-43 as stress mediators in the varied outcomes. These results help understand cancer drug responses at a cellular level and have implications in tackling heterogeneity in cancer treatment outcomes.

3.
Lab Invest ; 103(5): 100070, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36801642

RESUMEN

Tissue structures, phenotypes, and pathology are routinely investigated based on histology. This includes chemically staining the transparent tissue sections to make them visible to the human eye. Although chemical staining is fast and routine, it permanently alters the tissue and often consumes hazardous reagents. On the other hand, on using adjacent tissue sections for combined measurements, the cell-wise resolution is lost owing to sections representing different parts of the tissue. Hence, techniques providing visual information of the basic tissue structure enabling additional measurements from the exact same tissue section are required. Here we tested unstained tissue imaging for the development of computational hematoxylin and eosin (HE) staining. We used unsupervised deep learning (CycleGAN) and whole slide images of prostate tissue sections to compare the performance of imaging tissue in paraffin, as deparaffinized in air, and as deparaffinized in mounting medium with section thicknesses varying between 3 and 20 µm. We showed that although thicker sections increase the information content of tissue structures in the images, thinner sections generally perform better in providing information that can be reproduced in virtual staining. According to our results, tissue imaged in paraffin and as deparaffinized provides a good overall representation of the tissue for virtually HE-stained images. Further, using a pix2pix model, we showed that the reproduction of overall tissue histology can be clearly improved with image-to-image translation using supervised learning and pixel-wise ground truth. We also showed that virtual HE staining can be used for various tissues and used with both 20× and 40× imaging magnifications. Although the performance and methods of virtual staining need further development, our study provides evidence of the feasibility of whole slide unstained microscopy as a fast, cheap, and feasible approach to producing virtual staining of tissue histology while sparing the exact same tissue section ready for subsequent utilization with follow-up methods at single-cell resolution.


Asunto(s)
Microscopía , Parafina , Masculino , Humanos , Hematoxilina , Eosina Amarillenta-(YS) , Microscopía/métodos , Coloración y Etiquetado
4.
Histopathology ; 82(6): 837-845, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36645163

RESUMEN

AIMS: There is strong evidence that cribriform morphology indicates a worse prognosis of prostatic adenocarcinoma. Our aim was to investigate its interobserver reproducibility in prostate needle biopsies. METHODS AND RESULTS: A panel of nine prostate pathology experts from five continents independently reviewed 304 digitised biopsies for cribriform cancer according to recent International Society of Urological Pathology criteria. The biopsies were collected from a series of 702 biopsies that were reviewed by one of the panellists for enrichment of high-grade cancer and potentially cribriform structures. A 2/3 consensus diagnosis of cribriform and noncribriform cancer was reached in 90% (272/304) of the biopsies with a mean kappa value of 0.56 (95% confidence interval 0.52-0.61). The prevalence of consensus cribriform cancers was estimated to 4%, 12%, 21%, and 20% of Gleason scores 7 (3 + 4), 7 (4 + 3), 8, and 9-10, respectively. More than two cribriform structures per level or a largest cribriform mass with ≥9 lumina or a diameter of ≥0.5 mm predicted a consensus diagnosis of cribriform cancer in 88% (70/80), 84% (87/103), and 90% (56/62), respectively, and noncribriform cancer in 3% (2/80), 5% (5/103), and 2% (1/62), respectively (all P < 0.01). CONCLUSION: Cribriform prostate cancer was seen in a minority of needle biopsies with high-grade cancer. Stringent diagnostic criteria enabled the identification of cribriform patterns and the generation of a large set of consensus cases for standardisation.


Asunto(s)
Adenocarcinoma , Neoplasias de la Próstata , Masculino , Humanos , Próstata/patología , Reproducibilidad de los Resultados , Biopsia con Aguja , Adenocarcinoma/diagnóstico , Adenocarcinoma/patología , Biopsia , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/patología , Clasificación del Tumor
5.
Bioinformatics ; 37(21): 3995-3997, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34358287

RESUMEN

SUMMARY: Digital pathology enables applying computational methods, such as deep learning, in pathology for improved diagnostics and prognostics, but lack of interoperability between whole slide image formats of different scanner vendors is a challenge for algorithm developers. We present OpenPhi-Open PatHology Interface, an Application Programming Interface for seamless access to the iSyntax format used by the Philips Ultra Fast Scanner, the first digital pathology scanner approved by the United States Food and Drug Administration. OpenPhi is extensible and easily interfaced with existing vendor-neutral applications. AVAILABILITY AND IMPLEMENTATION: OpenPhi is implemented in Python and is available as open-source under the MIT license at: https://gitlab.com/BioimageInformaticsGroup/openphi. The Philips Software Development Kit is required and available at: https://www.openpathology.philips.com. OpenPhi version 1.1.1 is additionally provided as Supplementary Data. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Programas Informáticos , Estados Unidos
6.
BMC Cancer ; 21(1): 1133, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686173

RESUMEN

BACKGROUND: Virtual reality (VR) enables data visualization in an immersive and engaging manner, and it can be used for creating ways to explore scientific data. Here, we use VR for visualization of 3D histology data, creating a novel interface for digital pathology to aid cancer research. METHODS: Our contribution includes 3D modeling of a whole organ and embedded objects of interest, fusing the models with associated quantitative features and full resolution serial section patches, and implementing the virtual reality application. Our VR application is multi-scale in nature, covering two object levels representing different ranges of detail, namely organ level and sub-organ level. In addition, the application includes several data layers, including the measured histology image layer and multiple representations of quantitative features computed from the histology. RESULTS: In our interactive VR application, the user can set visualization properties, select different samples and features, and interact with various objects, which is not possible in the traditional 2D-image view used in digital pathology. In this work, we used whole mouse prostates (organ level) with prostate cancer tumors (sub-organ objects of interest) as example cases, and included quantitative histological features relevant for tumor biology in the VR model. CONCLUSIONS: Our application enables a novel way for exploration of high-resolution, multidimensional data for biomedical research purposes, and can also be used in teaching and researcher training. Due to automated processing of the histology data, our application can be easily adopted to visualize other organs and pathologies from various origins.


Asunto(s)
Imagenología Tridimensional/métodos , Preservación de Órganos/métodos , Realidad Virtual , Animales , Humanos , Ratones
7.
Lancet Oncol ; 21(2): 222-232, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31926806

RESUMEN

BACKGROUND: An increasing volume of prostate biopsies and a worldwide shortage of urological pathologists puts a strain on pathology departments. Additionally, the high intra-observer and inter-observer variability in grading can result in overtreatment and undertreatment of prostate cancer. To alleviate these problems, we aimed to develop an artificial intelligence (AI) system with clinically acceptable accuracy for prostate cancer detection, localisation, and Gleason grading. METHODS: We digitised 6682 slides from needle core biopsies from 976 randomly selected participants aged 50-69 in the Swedish prospective and population-based STHLM3 diagnostic study done between May 28, 2012, and Dec 30, 2014 (ISRCTN84445406), and another 271 from 93 men from outside the study. The resulting images were used to train deep neural networks for assessment of prostate biopsies. The networks were evaluated by predicting the presence, extent, and Gleason grade of malignant tissue for an independent test dataset comprising 1631 biopsies from 246 men from STHLM3 and an external validation dataset of 330 biopsies from 73 men. We also evaluated grading performance on 87 biopsies individually graded by 23 experienced urological pathologists from the International Society of Urological Pathology. We assessed discriminatory performance by receiver operating characteristics and tumour extent predictions by correlating predicted cancer length against measurements by the reporting pathologist. We quantified the concordance between grades assigned by the AI system and the expert urological pathologists using Cohen's kappa. FINDINGS: The AI achieved an area under the receiver operating characteristics curve of 0·997 (95% CI 0·994-0·999) for distinguishing between benign (n=910) and malignant (n=721) biopsy cores on the independent test dataset and 0·986 (0·972-0·996) on the external validation dataset (benign n=108, malignant n=222). The correlation between cancer length predicted by the AI and assigned by the reporting pathologist was 0·96 (95% CI 0·95-0·97) for the independent test dataset and 0·87 (0·84-0·90) for the external validation dataset. For assigning Gleason grades, the AI achieved a mean pairwise kappa of 0·62, which was within the range of the corresponding values for the expert pathologists (0·60-0·73). INTERPRETATION: An AI system can be trained to detect and grade cancer in prostate needle biopsy samples at a ranking comparable to that of international experts in prostate pathology. Clinical application could reduce pathology workload by reducing the assessment of benign biopsies and by automating the task of measuring cancer length in positive biopsy cores. An AI system with expert-level grading performance might contribute a second opinion, aid in standardising grading, and provide pathology expertise in parts of the world where it does not exist. FUNDING: Swedish Research Council, Swedish Cancer Society, Swedish eScience Research Center, EIT Health.


Asunto(s)
Inteligencia Artificial , Diagnóstico por Computador , Interpretación de Imagen Asistida por Computador , Clasificación del Tumor , Neoplasias de la Próstata/patología , Anciano , Biopsia , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Reproducibilidad de los Resultados , Suecia
8.
BMC Bioinformatics ; 20(1): 80, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30767778

RESUMEN

BACKGROUND: Cell counting from cell cultures is required in multiple biological and biomedical research applications. Especially, accurate brightfield-based cell counting methods are needed for cell growth analysis. With deep learning, cells can be detected with high accuracy, but manually annotated training data is required. We propose a method for cell detection that requires annotated training data for one cell line only, and generalizes to other, unseen cell lines. RESULTS: Training a deep learning model with one cell line only can provide accurate detections for similar unseen cell lines (domains). However, if the new domain is very dissimilar from training domain, high precision but lower recall is achieved. Generalization capabilities of the model can be improved with training data transformations, but only to a certain degree. To further improve the detection accuracy of unseen domains, we propose iterative unsupervised domain adaptation method. Predictions of unseen cell lines with high precision enable automatic generation of training data, which is used to train the model together with parts of the previously used annotated training data. We used U-Net-based model, and three consecutive focal planes from brightfield image z-stacks. We trained the model initially with PC-3 cell line, and used LNCaP, BT-474 and 22Rv1 cell lines as target domains for domain adaptation. Highest improvement in accuracy was achieved for 22Rv1 cells. F1-score after supervised training was only 0.65, but after unsupervised domain adaptation we achieved a score of 0.84. Mean accuracy for target domains was 0.87, with mean improvement of 16 percent. CONCLUSIONS: With our method for generalized cell detection, we can train a model that accurately detects different cell lines from brightfield images. A new cell line can be introduced to the model without a single manual annotation, and after iterative domain adaptation the model is ready to detect these cells with high accuracy.


Asunto(s)
Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador/métodos , Neoplasias de la Próstata/patología , Humanos , Masculino , Células Tumorales Cultivadas
9.
Bioinformatics ; 34(17): 3013-3021, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29684099

RESUMEN

Motivation: Digital pathology enables new approaches that expand beyond storage, visualization or analysis of histological samples in digital format. One novel opportunity is 3D histology, where a three-dimensional reconstruction of the sample is formed computationally based on serial tissue sections. This allows examining tissue architecture in 3D, for example, for diagnostic purposes. Importantly, 3D histology enables joint mapping of cellular morphology with spatially resolved omics data in the true 3D context of the tissue at microscopic resolution. Several algorithms have been proposed for the reconstruction task, but a quantitative comparison of their accuracy is lacking. Results: We developed a benchmarking framework to evaluate the accuracy of several free and commercial 3D reconstruction methods using two whole slide image datasets. The results provide a solid basis for further development and application of 3D histology algorithms and indicate that methods capable of compensating for local tissue deformation are superior to simpler approaches. Availability and implementation: Code: https://github.com/BioimageInformaticsTampere/RegBenchmark. Whole slide image datasets: http://urn.fi/urn: nbn: fi: csc-kata20170705131652639702. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Técnicas Histológicas , Imagenología Tridimensional/métodos
10.
Cell Commun Signal ; 17(1): 148, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31730483

RESUMEN

BACKGROUND: Progression of prostate cancer from benign local tumors to metastatic carcinomas is a multistep process. Here we have investigated the signaling pathways that support migration and invasion of prostate cancer cells, focusing on the role of the NFATC1 transcription factor and its post-translational modifications. We have previously identified NFATC1 as a substrate for the PIM1 kinase and shown that PIM1-dependent phosphorylation increases NFATC1 activity without affecting its subcellular localization. Both PIM kinases and NFATC1 have been reported to promote cancer cell migration, invasion and angiogenesis, but it has remained unclear whether the effects of NFATC1 are phosphorylation-dependent and which downstream targets are involved. METHODS: We used mass spectrometry to identify PIM1 phosphorylation target sites in NFATC1, and analysed their functional roles in three prostate cancer cell lines by comparing phosphodeficient mutants to wild-type NFATC1. We used luciferase assays to determine effects of phosphorylation on NFAT-dependent transcriptional activity, and migration and invasion assays to evaluate effects on cell motility. We also performed a microarray analysis to identify novel PIM1/NFATC1 targets, and validated one of them with both cellular expression analyses and in silico in clinical prostate cancer data sets. RESULTS: Here we have identified ten PIM1 target sites in NFATC1 and found that prevention of their phosphorylation significantly decreases the transcriptional activity as well as the pro-migratory and pro-invasive effects of NFATC1 in prostate cancer cells. We observed that also PIM2 and PIM3 can phosphorylate NFATC1, and identified several novel putative PIM1/NFATC1 target genes. These include the ITGA5 integrin, which is differentially expressed in the presence of wild-type versus phosphorylation-deficient NFATC1, and which is coexpressed with PIM1 and NFATC1 in clinical prostate cancer specimens. CONCLUSIONS: Based on our data, phosphorylation of PIM1 target sites stimulates NFATC1 activity and enhances its ability to promote prostate cancer cell migration and invasion. Therefore, inhibition of the interplay between PIM kinases and NFATC1 may have therapeutic implications for patients with metastatic forms of cancer.


Asunto(s)
Movimiento Celular , Factores de Transcripción NFATC/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proliferación Celular , Humanos , Masculino , Espectrometría de Masas , Células PC-3 , Fosforilación , Neoplasias de la Próstata/patología , Transducción de Señal , Células Tumorales Cultivadas
11.
Am J Pathol ; 187(11): 2546-2557, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28827140

RESUMEN

miRNAs are important regulators of gene expression and are often deregulated in cancer. We have previously shown that miR-32 is an androgen receptor-regulated miRNA overexpressed in castration-resistant prostate cancer and that miR-32 can improve prostate cancer cell growth in vitro. To assess the effects of miR-32 in vivo, we developed transgenic mice overexpressing miR-32 in the prostate. The study indicated that transgenic miR-32 expression increases replicative activity in the prostate epithelium. We further observed an aging-associated increase in the incidence of goblet cell metaplasia in the prostate epithelium. Furthermore, aged miR-32 transgenic mice exhibited metaplasia-associated prostatic intraepithelial neoplasia at a low frequency. When crossbred with mice lacking the other allele of tumor-suppressor Pten (miR-32xPten+/- mice), miR-32 expression increased both the incidence and the replicative activity of prostatic intraepithelial neoplasia lesions in the dorsal prostate. The miR-32xPten+/- mice also demonstrated increased goblet cell metaplasia compared with Pten+/- mice. By performing a microarray analysis of mouse prostate tissue to screen downstream targets and effectors of miR-32, we identified RAC2 as a potential, and clinically relevant, target of miR-32. We also demonstrate down-regulation of several interesting, potentially prostate cancer-relevant genes (Spink1, Spink5, and Casp1) by miR-32 in the prostate tissue. The results demonstrate that miR-32 increases proliferation and promotes metaplastic transformation in mouse prostate epithelium, which may promote neoplastic alterations in the prostate.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Próstata/patología , Neoplasias de la Próstata/genética , Animales , Proliferación Celular/genética , Transformación Celular Neoplásica/patología , Epitelio/patología , Masculino , Ratones , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo
12.
Cell Microbiol ; 19(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27665309

RESUMEN

We have demonstrated previously that the human picornavirus Echovirus 1 (EV1) triggers an infectious internalization pathway that follows closely, but seems to stay separate, from the epidermal growth factor receptor (EGFR) pathway triggered by epidermal growth factor (EGF). Here, we confirmed by using live and confocal microscopy that EGFR and EV1 vesicles are following intimately each other but are distinct entities with different degradation kinetics. We show here that despite being sorted to different pathways and located in distinct endosomes, EV1 inhibits EGFR downregulation. Simultaneous treatment with EV1 and EGF led to an accumulation of EGFR in cytoplasmic endosomes, which was evident already 15 min p.i. and more pronounced after 2 hr p.i. EV1 treatment led to reduced downregulation, which was proven by increased total cellular amount of EGFR. Confocal microscopy studies revealed that EGFR accumulated in large endosomes, presumably macropinosomes, which were not positive for markers of the early, recycling, or late endosomes/lysosomes. Interestingly, EV1 did not have a similar blocking effect on bulk endosomal trafficking or transferrin recycling along the clathrin pathway suggesting that EV1 did not have a general effect on cellular trafficking pathways. Importantly, EGF treatment increased EV1 infection and increased cell viability during infection. Simultaneous EV1 and EGF treatment seemed to moderately enhance phosphorylation of protein kinase C α. Furthermore, similar phenotype of EGFR trafficking could be produced by phorbol 12-myristate 13-acetate treatment, further suggesting that activated protein kinase C α could be contributing to EGFR phenotype. These results altogether demonstrate that EV1 specifically affects EGFR trafficking, leading to EGFR downregulation, which is beneficial to EV1 infection.


Asunto(s)
Enterovirus Humano B/fisiología , Receptores ErbB/biosíntesis , Interacciones Huésped-Patógeno , Internalización del Virus , Línea Celular , Regulación hacia Abajo , Endosomas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Microscopía Confocal
13.
Cytometry A ; 91(6): 555-565, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28426134

RESUMEN

Digital pathology has led to a demand for automated detection of regions of interest, such as cancerous tissue, from scanned whole slide images. With accurate methods using image analysis and machine learning, significant speed-up, and savings in costs through increased throughput in histological assessment could be achieved. This article describes a machine learning approach for detection of cancerous tissue from scanned whole slide images. Our method is based on feature engineering and supervised learning with a random forest model. The features extracted from the whole slide images include several local descriptors related to image texture, spatial structure, and distribution of nuclei. The method was evaluated in breast cancer metastasis detection from lymph node samples. Our results show that the method detects metastatic areas with high accuracy (AUC = 0.97-0.98 for tumor detection within whole image area, AUC = 0.84-0.91 for tumor vs. normal tissue detection) and that the method generalizes well for images from more than one laboratory. Further, the method outputs an interpretable classification model, enabling the linking of individual features to differences between tissue types. © 2017 International Society for Advancement of Cytometry.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Histocitoquímica/estadística & datos numéricos , Interpretación de Imagen Asistida por Computador/métodos , Ganglios Linfáticos/diagnóstico por imagen , Aprendizaje Automático , Adulto , Área Bajo la Curva , Neoplasias de la Mama/patología , Núcleo Celular/patología , Núcleo Celular/ultraestructura , Eosina Amarillenta-(YS) , Femenino , Hematoxilina , Humanos , Ganglios Linfáticos/patología , Metástasis Linfática , Linfocitos/patología , Linfocitos/ultraestructura , Persona de Mediana Edad , Curva ROC , Programas Informáticos
14.
JAMA ; 318(22): 2199-2210, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29234806

RESUMEN

Importance: Application of deep learning algorithms to whole-slide pathology images can potentially improve diagnostic accuracy and efficiency. Objective: Assess the performance of automated deep learning algorithms at detecting metastases in hematoxylin and eosin-stained tissue sections of lymph nodes of women with breast cancer and compare it with pathologists' diagnoses in a diagnostic setting. Design, Setting, and Participants: Researcher challenge competition (CAMELYON16) to develop automated solutions for detecting lymph node metastases (November 2015-November 2016). A training data set of whole-slide images from 2 centers in the Netherlands with (n = 110) and without (n = 160) nodal metastases verified by immunohistochemical staining were provided to challenge participants to build algorithms. Algorithm performance was evaluated in an independent test set of 129 whole-slide images (49 with and 80 without metastases). The same test set of corresponding glass slides was also evaluated by a panel of 11 pathologists with time constraint (WTC) from the Netherlands to ascertain likelihood of nodal metastases for each slide in a flexible 2-hour session, simulating routine pathology workflow, and by 1 pathologist without time constraint (WOTC). Exposures: Deep learning algorithms submitted as part of a challenge competition or pathologist interpretation. Main Outcomes and Measures: The presence of specific metastatic foci and the absence vs presence of lymph node metastasis in a slide or image using receiver operating characteristic curve analysis. The 11 pathologists participating in the simulation exercise rated their diagnostic confidence as definitely normal, probably normal, equivocal, probably tumor, or definitely tumor. Results: The area under the receiver operating characteristic curve (AUC) for the algorithms ranged from 0.556 to 0.994. The top-performing algorithm achieved a lesion-level, true-positive fraction comparable with that of the pathologist WOTC (72.4% [95% CI, 64.3%-80.4%]) at a mean of 0.0125 false-positives per normal whole-slide image. For the whole-slide image classification task, the best algorithm (AUC, 0.994 [95% CI, 0.983-0.999]) performed significantly better than the pathologists WTC in a diagnostic simulation (mean AUC, 0.810 [range, 0.738-0.884]; P < .001). The top 5 algorithms had a mean AUC that was comparable with the pathologist interpreting the slides in the absence of time constraints (mean AUC, 0.960 [range, 0.923-0.994] for the top 5 algorithms vs 0.966 [95% CI, 0.927-0.998] for the pathologist WOTC). Conclusions and Relevance: In the setting of a challenge competition, some deep learning algorithms achieved better diagnostic performance than a panel of 11 pathologists participating in a simulation exercise designed to mimic routine pathology workflow; algorithm performance was comparable with an expert pathologist interpreting whole-slide images without time constraints. Whether this approach has clinical utility will require evaluation in a clinical setting.


Asunto(s)
Neoplasias de la Mama/patología , Metástasis Linfática/diagnóstico , Aprendizaje Automático , Patólogos , Algoritmos , Femenino , Humanos , Metástasis Linfática/patología , Patología Clínica , Curva ROC
15.
Cytometry A ; 89(12): 1057-1072, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27922735

RESUMEN

The simulations of cells and microscope images thereof have been used to facilitate the development, selection, and validation of image analysis algorithms employed in cytometry as well as for modeling and understanding cell structure and dynamics beyond what is visible in the eyepiece. The simulation approaches vary from simple parametric models of specific cell components-especially shapes of cells and cell nuclei-to learning-based synthesis and multi-stage simulation models for complex scenes that simultaneously visualize multiple object types and incorporate various properties of the imaged objects and laws of image formation. This review covers advances in artificial digital cell generation at scales ranging from particles up to tissue synthesis and microscope image simulation methods, provides examples of the use of simulated images for various purposes ranging from subcellular object detection to cell tracking, and discusses how such simulators have been validated. Finally, the future possibilities and limitations of simulation-based validation are considered. © 2016 International Society for Advancement of Cytometry.


Asunto(s)
Citometría de Imagen/métodos , Algoritmos , Inteligencia Artificial , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Reconocimiento de Normas Patrones Automatizadas/métodos
16.
Trends Biotechnol ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480025

RESUMEN

In pathology and biomedical research, histology is the cornerstone method for tissue analysis. Currently, the histological workflow consumes plenty of chemicals, water, and time for staining procedures. Deep learning is now enabling digital replacement of parts of the histological staining procedure. In virtual staining, histological stains are created by training neural networks to produce stained images from an unstained tissue image, or through transferring information from one stain to another. These technical innovations provide more sustainable, rapid, and cost-effective alternatives to traditional histological pipelines, but their development is in an early phase and requires rigorous validation. In this review we cover the basic concepts of virtual staining for histology and provide future insights into the utilization of artificial intelligence (AI)-enabled virtual histology.

17.
BMC Bioinformatics ; 14 Suppl 10: S6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24267488

RESUMEN

BACKGROUND: High-throughput genome-wide screening to study gene-specific functions, e.g. for drug discovery, demands fast automated image analysis methods to assist in unraveling the full potential of such studies. Image segmentation is typically at the forefront of such analysis as the performance of the subsequent steps, for example, cell classification, cell tracking etc., often relies on the results of segmentation. METHODS: We present a cell cytoplasm segmentation framework which first separates cell cytoplasm from image background using novel approach of image enhancement and coefficient of variation of multi-scale Gaussian scale-space representation. A novel outline-learning based classification method is developed using regularized logistic regression with embedded feature selection which classifies image pixels as outline/non-outline to give cytoplasm outlines. Refinement of the detected outlines to separate cells from each other is performed in a post-processing step where the nuclei segmentation is used as contextual information. RESULTS AND CONCLUSIONS: We evaluate the proposed segmentation methodology using two challenging test cases, presenting images with completely different characteristics, with cells of varying size, shape, texture and degrees of overlap. The feature selection and classification framework for outline detection produces very simple sparse models which use only a small subset of the large, generic feature set, that is, only 7 and 5 features for the two cases. Quantitative comparison of the results for the two test cases against state-of-the-art methods show that our methodology outperforms them with an increase of 4-9% in segmentation accuracy with maximum accuracy of 93%. Finally, the results obtained for diverse datasets demonstrate that our framework not only produces accurate segmentation but also generalizes well to different segmentation tasks.


Asunto(s)
División Celular/fisiología , Ensayos Analíticos de Alto Rendimiento , Algoritmos , Animales , Células Cultivadas , Citoplasma/fisiología , Drosophila melanogaster/citología , Células HeLa , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Aumento de la Imagen/métodos , Microscopía Fluorescente , Distribución Normal , Procesamiento Proteico-Postraduccional/fisiología , Distribución Aleatoria
18.
Cancer Cell ; 41(9): 1543-1545, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37652005

RESUMEN

Artificial intelligence (AI) is rapidly gaining interest in medicine, including pathological assessments for personalized medicine. In this issue of Cancer Cell, Wagner et al. demonstrate superior accuracy of transformer-based deep learning in predicting biomarker status in CRC. The work has implications for increased efficiency and accuracy in clinical diagnostics guiding treatment decisions in precision oncology.


Asunto(s)
Neoplasias Colorrectales , Aprendizaje Profundo , Humanos , Biomarcadores de Tumor , Inteligencia Artificial , Medicina de Precisión
19.
Patterns (N Y) ; 4(5): 100725, 2023 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-37223268

RESUMEN

Conventional histopathology has relied on chemical staining for over a century. The staining process makes tissue sections visible to the human eye through a tedious and labor-intensive procedure that alters the tissue irreversibly, preventing repeated use of the sample. Deep learning-based virtual staining can potentially alleviate these shortcomings. Here, we used standard brightfield microscopy on unstained tissue sections and studied the impact of increased network capacity on the resulting virtually stained H&E images. Using the generative adversarial neural network model pix2pix as a baseline, we observed that replacing simple convolutions with dense convolution units increased the structural similarity score, peak signal-to-noise ratio, and nuclei reproduction accuracy. We also demonstrated highly accurate reproduction of histology, especially with increased network capacity, and demonstrated applicability to several tissues. We show that network architecture optimization can improve the image translation accuracy of virtual H&E staining, highlighting the potential of virtual staining in streamlining histopathological analysis.

20.
Med Image Anal ; 90: 102940, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37666115

RESUMEN

Cross-modality image synthesis is an active research topic with multiple medical clinically relevant applications. Recently, methods allowing training with paired but misaligned data have started to emerge. However, no robust and well-performing methods applicable to a wide range of real world data sets exist. In this work, we propose a generic solution to the problem of cross-modality image synthesis with paired but non-aligned data by introducing new deformation equivariance encouraging loss functions. The method consists of joint training of an image synthesis network together with separate registration networks and allows adversarial training conditioned on the input even with misaligned data. The work lowers the bar for new clinical applications by allowing effortless training of cross-modality image synthesis networks for more difficult data sets.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA