Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 237
Filter
1.
Mod Pathol ; : 100625, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39332710

ABSTRACT

Tumors of the major and minor salivary gland histologically encompass a diverse and partly overlapping spectrum of frequently diagnostically challenging neoplasms. Despite recent advances in molecular testing and the identification of tumor-specific mutations or gene fusions, there is an unmet need to identify additional diagnostic biomarkers for entities lacking specific alterations. In this study, we collected a comprehensive cohort of 363 cases encompassing 20 different salivary gland tumor entities and explored the potential of DNA methylation to classify these tumors. We were able to show that most entities show specific epigenetic signatures and present a machine learning algorithm that achieved a mean balanced accuracy of 0.991. Of note, we showed that cribriform adenocarcinoma is epigenetically distinct from classical polymorphous adenocarcinoma, which could support risk stratification of these tumors. Myoepithelioma and pleomorphic adenoma form a uniform epigenetic class, supporting the theory of a single entity with a broad but continuous morphological spectrum. Furthermore, we identified a histomorphologically heterogeneous but epigenetically distinct class that could represent a novel tumor entity. In conclusion, our study provides a comprehensive resource of the DNA methylation landscape of salivary gland tumors. Our data provides novel insight into disputed entities and shows the potential of DNA methylation to identify new tumor classes. Furthermore, in future, our machine learning classifier could support the histopathological diagnosis of salivary gland tumors.

2.
Eur J Cancer ; 211: 114292, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39276594

ABSTRACT

INTRODUCTION: Molecular profiling of lung cancer is essential to identify genetic alterations that predict response to targeted therapy. While deep learning shows promise for predicting oncogenic mutations from whole tissue images, existing studies often face challenges such as limited sample sizes, a focus on earlier stage patients, and insufficient analysis of robustness and generalizability. METHODS: This retrospective study evaluates factors influencing mutation prediction accuracy using the large Heidelberg Lung Adenocarcinoma Cohort (HLCC), a cohort of 2356 late-stage FFPE samples. Validation is performed in the publicly available TCGA-LUAD cohort. RESULTS: Models trained on the larger HLCC cohort generalized well to the TCGA dataset for mutations in EGFR (AUC 0.76), STK11 (AUC 0.71) and TP53 (AUC 0.75), in line with the hypothesis that larger cohort sizes improve model robustness. Variation in performance due to pre-processing and modeling choices, such as mutation variant calling, affected EGFR prediction accuracy by up to 7 %. DISCUSSION: Model explanations suggest that acinar and papillary growth patterns are critical for the detection of EGFR mutations, whereas solid growth patterns and large nuclei are indicative of TP53 mutations. These findings highlight the importance of specific morphological features in mutation detection and the potential of deep learning models to improve mutation prediction accuracy. CONCLUSION: Although deep learning models trained on larger cohorts show improved robustness and generalizability in predicting oncogenic mutations, they cannot replace comprehensive molecular profiling. However, they may support patient pre-selection for clinical trials and deepen the insight in genotype-phenotype relationships.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Mutation , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Retrospective Studies , Female , Male , ErbB Receptors/genetics , Biomarkers, Tumor/genetics , Middle Aged , Aged , Tumor Suppressor Protein p53/genetics , Deep Learning
3.
Nat Commun ; 15(1): 6539, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107296

ABSTRACT

Recent years have seen vast progress in the development of machine learned force fields (MLFFs) based on ab-initio reference calculations. Despite achieving low test errors, the reliability of MLFFs in molecular dynamics (MD) simulations is facing growing scrutiny due to concerns about instability over extended simulation timescales. Our findings suggest a potential connection between robustness to cumulative inaccuracies and the use of equivariant representations in MLFFs, but the computational cost associated with these representations can limit this advantage in practice. To address this, we propose a transformer architecture called SO3KRATES that combines sparse equivariant representations (Euclidean variables) with a self-attention mechanism that separates invariant and equivariant information, eliminating the need for expensive tensor products. SO3KRATES achieves a unique combination of accuracy, stability, and speed that enables insightful analysis of quantum properties of matter on extended time and system size scales. To showcase this capability, we generate stable MD trajectories for flexible peptides and supra-molecular structures with hundreds of atoms. Furthermore, we investigate the PES topology for medium-sized chainlike molecules (e.g., small peptides) by exploring thousands of minima. Remarkably, SO3KRATES demonstrates the ability to strike a balance between the conflicting demands of stability and the emergence of new minimum-energy conformations beyond the training data, which is crucial for realistic exploration tasks in the field of biochemistry.

4.
IEEE Trans Pattern Anal Mach Intell ; 46(11): 7283-7299, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38607718

ABSTRACT

Explainable AI aims to overcome the black-box nature of complex ML models like neural networks by generating explanations for their predictions. Explanations often take the form of a heatmap identifying input features (e.g. pixels) that are relevant to the model's decision. These explanations, however, entangle the potentially multiple factors that enter into the overall complex decision strategy. We propose to disentangle explanations by extracting at some intermediate layer of a neural network, subspaces that capture the multiple and distinct activation patterns (e.g. visual concepts) that are relevant to the prediction. To automatically extract these subspaces, we propose two new analyses, extending principles found in PCA or ICA to explanations. These novel analyses, which we call principal relevant component analysis (PRCA) and disentangled relevant subspace analysis (DRSA), maximize relevance instead of e.g. variance or kurtosis. This allows for a much stronger focus of the analysis on what the ML model actually uses for predicting, ignoring activations or concepts to which the model is invariant. Our approach is general enough to work alongside common attribution techniques such as Shapley Value, Integrated Gradients, or LRP. Our proposed methods show to be practically useful and compare favorably to the state of the art as demonstrated on benchmarks and three use cases.

5.
6.
Pathologie (Heidelb) ; 45(2): 133-139, 2024 Mar.
Article in German | MEDLINE | ID: mdl-38315198

ABSTRACT

With the advancements in precision medicine, the demands on pathological diagnostics have increased, requiring standardized, quantitative, and integrated assessments of histomorphological and molecular pathological data. Great hopes are placed in artificial intelligence (AI) methods, which have demonstrated the ability to analyze complex clinical, histological, and molecular data for disease classification, biomarker quantification, and prognosis estimation. This paper provides an overview of the latest developments in pathology AI, discusses the limitations, particularly concerning the black box character of AI, and describes solutions to make decision processes more transparent using methods of so-called explainable AI (XAI).


Subject(s)
Artificial Intelligence , Pathology, Molecular , Hope , Precision Medicine
7.
IEEE Trans Pattern Anal Mach Intell ; 46(5): 3257-3274, 2024 May.
Article in English | MEDLINE | ID: mdl-38055368

ABSTRACT

Counterfactuals can explain classification decisions of neural networks in a human interpretable way. We propose a simple but effective method to generate such counterfactuals. More specifically, we perform a suitable diffeomorphic coordinate transformation and then perform gradient ascent in these coordinates to find counterfactuals which are classified with great confidence as a specified target class. We propose two methods to leverage generative models to construct such suitable coordinate systems that are either exactly or approximately diffeomorphic. We analyze the generation process theoretically using Riemannian differential geometry and validate the quality of the generated counterfactuals using various qualitative and quantitative measures.

8.
Annu Rev Pathol ; 19: 541-570, 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-37871132

ABSTRACT

The rapid development of precision medicine in recent years has started to challenge diagnostic pathology with respect to its ability to analyze histological images and increasingly large molecular profiling data in a quantitative, integrative, and standardized way. Artificial intelligence (AI) and, more precisely, deep learning technologies have recently demonstrated the potential to facilitate complex data analysis tasks, including clinical, histological, and molecular data for disease classification; tissue biomarker quantification; and clinical outcome prediction. This review provides a general introduction to AI and describes recent developments with a focus on applications in diagnostic pathology and beyond. We explain limitations including the black-box character of conventional AI and describe solutions to make machine learning decisions more transparent with so-called explainable AI. The purpose of the review is to foster a mutual understanding of both the biomedical and the AI side. To that end, in addition to providing an overview of the relevant foundations in pathology and machine learning, we present worked-through examples for a better practical understanding of what AI can achieve and how it should be done.


Subject(s)
Artificial Intelligence , Precision Medicine , Humans
9.
Neural Netw ; 167: 233-243, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37660672

ABSTRACT

Domain shifts in the training data are common in practical applications of machine learning; they occur for instance when the data is coming from different sources. Ideally, a ML model should work well independently of these shifts, for example, by learning a domain-invariant representation. However, common ML losses do not give strong guarantees on how consistently the ML model performs for different domains, in particular, whether the model performs well on a domain at the expense of its performance on another domain. In this paper, we build new theoretical foundations for this problem, by contributing a set of mathematical relations between classical losses for supervised ML and the Wasserstein distance in joint space (i.e. representation and output space). We show that classification or regression losses, when combined with a GAN-type discriminator between domains, form an upper-bound to the true Wasserstein distance between domains. This implies a more invariant representation and also more stable prediction performance across domains. Theoretical results are corroborated empirically on several image datasets. Our proposed approach systematically produces the highest minimum classification accuracy across domains, and the most invariant representation.


Subject(s)
Machine Learning
10.
Phys Chem Chem Phys ; 25(38): 26370-26379, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37750554

ABSTRACT

In recent years, the prediction of quantum mechanical observables with machine learning methods has become increasingly popular. Message-passing neural networks (MPNNs) solve this task by constructing atomic representations, from which the properties of interest are predicted. Here, we introduce a method to automatically identify chemical moieties (molecular building blocks) from such representations, enabling a variety of applications beyond property prediction, which otherwise rely on expert knowledge. The required representation can either be provided by a pretrained MPNN, or be learned from scratch using only structural information. Beyond the data-driven design of molecular fingerprints, the versatility of our approach is demonstrated by enabling the selection of representative entries in chemical databases, the automatic construction of coarse-grained force fields, as well as the identification of reaction coordinates.

11.
J Phys Chem Lett ; 14(31): 7092-7099, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37530451

ABSTRACT

Essential for understanding far-from-equilibrium processes, nonadiabatic (NA) molecular dynamics (MD) requires expensive calculations of the excitation energies and NA couplings. Machine learning (ML) can simplify computation; however, the NA Hamiltonian requires complex ML models due to its intricate relationship to atomic geometry. Working directly in the time domain, we employ bidirectional long short-term memory networks (Bi-LSTM) to interpolate the Hamiltonian. Applying this multiscale approach to three metal-halide perovskite systems, we achieve two orders of magnitude computational savings compared to direct ab initio calculation. Reasonable charge trapping and recombination times are obtained with NA Hamiltonian sampling every half a picosecond. The Bi-LSTM-NAMD method outperforms earlier models and captures both slow and fast time scales. In combination with ML force fields, the methodology extends NAMD simulation times from picoseconds to nanoseconds, comparable to charge carrier lifetimes in many materials. Nanosecond sampling is particularly important in systems containing defects, boundaries, interfaces, etc. that can undergo slow rearrangements.

12.
J Phys Chem C Nanomater Interfaces ; 127(28): 13817-13836, 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37492192

ABSTRACT

A bold vision in nanofabrication is the assembly of functional molecular structures using a scanning probe microscope (SPM). This approach requires continuous monitoring of the molecular configuration during manipulation. Until now, this has been impossible because the SPM tip cannot simultaneously act as an actuator and an imaging probe. Here, we implement configuration monitoring using experimental data other than images collected during the manipulation process. We model the manipulation as a partially observable Markov decision process (POMDP) and approximate the actual configuration in real time using a particle filter. To achieve this, the models underlying the POMDP are precomputed and organized in the form of a finite-state automaton, allowing the use of complex atomistic simulations. We exemplify the configuration monitoring process and reveal structural motifs behind measured force gradients. The proposed methodology marks an important step toward the piece-by-piece creation of supramolecular structures in a robotic and possibly automated manner.

13.
PLoS Comput Biol ; 19(5): e1011105, 2023 05.
Article in English | MEDLINE | ID: mdl-37228169

ABSTRACT

Single-pulse electrical stimulation in the nervous system, often called cortico-cortical evoked potential (CCEP) measurement, is an important technique to understand how brain regions interact with one another. Voltages are measured from implanted electrodes in one brain area while stimulating another with brief current impulses separated by several seconds. Historically, researchers have tried to understand the significance of evoked voltage polyphasic deflections by visual inspection, but no general-purpose tool has emerged to understand their shapes or describe them mathematically. We describe and illustrate a new technique to parameterize brain stimulation data, where voltage response traces are projected into one another using a semi-normalized dot product. The length of timepoints from stimulation included in the dot product is varied to obtain a temporal profile of structural significance, and the peak of the profile uniquely identifies the duration of the response. Using linear kernel PCA, a canonical response shape is obtained over this duration, and then single-trial traces are parameterized as a projection of this canonical shape with a residual term. Such parameterization allows for dissimilar trace shapes from different brain areas to be directly compared by quantifying cross-projection magnitudes, response duration, canonical shape projection amplitudes, signal-to-noise ratios, explained variance, and statistical significance. Artifactual trials are automatically identified by outliers in sub-distributions of cross-projection magnitude, and rejected. This technique, which we call "Canonical Response Parameterization" (CRP) dramatically simplifies the study of CCEP shapes, and may also be applied in a wide range of other settings involving event-triggered data.


Subject(s)
Brain , Evoked Potentials , Evoked Potentials/physiology , Brain Mapping/methods , Electrodes, Implanted , Electric Stimulation/methods
14.
J Chem Theory Comput ; 19(14): 4619-4630, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37156733

ABSTRACT

Kernel machines have sustained continuous progress in the field of quantum chemistry. In particular, they have proven to be successful in the low-data regime of force field reconstruction. This is because many equivariances and invariances due to physical symmetries can be incorporated into the kernel function to compensate for much larger data sets. So far, the scalability of kernel machines has however been hindered by its quadratic memory and cubical runtime complexity in the number of training points. While it is known that iterative Krylov subspace solvers can overcome these burdens, their convergence crucially relies on effective preconditioners, which are elusive in practice. Effective preconditioners need to partially presolve the learning problem in a computationally cheap and numerically robust manner. Here, we consider the broad class of Nyström-type methods to construct preconditioners based on successively more sophisticated low-rank approximations of the original kernel matrix, each of which provides a different set of computational trade-offs. All considered methods aim to identify a representative subset of inducing (kernel) columns to approximate the dominant kernel spectrum.

15.
Nucleic Acids Res ; 51(4): e20, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36629274

ABSTRACT

The molecular heterogeneity of cancer cells contributes to the often partial response to targeted therapies and relapse of disease due to the escape of resistant cell populations. While single-cell sequencing has started to improve our understanding of this heterogeneity, it offers a mostly descriptive view on cellular types and states. To obtain more functional insights, we propose scGeneRAI, an explainable deep learning approach that uses layer-wise relevance propagation (LRP) to infer gene regulatory networks from static single-cell RNA sequencing data for individual cells. We benchmark our method with synthetic data and apply it to single-cell RNA sequencing data of a cohort of human lung cancers. From the predicted single-cell networks our approach reveals characteristic network patterns for tumor cells and normal epithelial cells and identifies subnetworks that are observed only in (subgroups of) tumor cells of certain patients. While current state-of-the-art methods are limited by their ability to only predict average networks for cell populations, our approach facilitates the reconstruction of networks down to the level of single cells which can be utilized to characterize the heterogeneity of gene regulation within and across tumors.


Subject(s)
Deep Learning , Gene Regulatory Networks , Neoplasms , Single-Cell Gene Expression Analysis , Humans , Gene Expression Regulation , Neoplasms/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology
16.
Sci Adv ; 9(2): eadf0873, 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36630510

ABSTRACT

Global machine learning force fields, with the capacity to capture collective interactions in molecular systems, now scale up to a few dozen atoms due to considerable growth of model complexity with system size. For larger molecules, locality assumptions are introduced, with the consequence that nonlocal interactions are not described. Here, we develop an exact iterative approach to train global symmetric gradient domain machine learning (sGDML) force fields (FFs) for several hundred atoms, without resorting to any potentially uncontrolled approximations. All atomic degrees of freedom remain correlated in the global sGDML FF, allowing the accurate description of complex molecules and materials that present phenomena with far-reaching characteristic correlation lengths. We assess the accuracy and efficiency of sGDML on a newly developed MD22 benchmark dataset containing molecules from 42 to 370 atoms. The robustness of our approach is demonstrated in nanosecond path-integral molecular dynamics simulations for supramolecular complexes in the MD22 dataset.

17.
IEEE Trans Neural Netw Learn Syst ; 34(10): 7675-7688, 2023 Oct.
Article in English | MEDLINE | ID: mdl-35133968

ABSTRACT

Domain translation is the task of finding correspondence between two domains. Several deep neural network (DNN) models, e.g., CycleGAN and cross-lingual language models, have shown remarkable successes on this task under the unsupervised setting-the mappings between the domains are learned from two independent sets of training data in both domains (without paired samples). However, those methods typically do not perform well on a significant proportion of test samples. In this article, we hypothesize that many of such unsuccessful samples lie at the fringe-relatively low-density areas-of data distribution, where the DNN was not trained very well, and propose to perform the Langevin dynamics to bring such fringe samples toward high-density areas. We demonstrate qualitatively and quantitatively that our strategy, called Langevin cooling (L-Cool), enhances state-of-the-art methods in image translation and language translation tasks.

18.
Neuropathol Appl Neurobiol ; 49(1): e12866, 2023 02.
Article in English | MEDLINE | ID: mdl-36519297

ABSTRACT

AIM: Analysis of cerebrospinal fluid (CSF) is essential for diagnostic workup of patients with neurological diseases and includes differential cell typing. The current gold standard is based on microscopic examination by specialised technicians and neuropathologists, which is time-consuming, labour-intensive and subjective. METHODS: We, therefore, developed an image analysis approach based on expert annotations of 123,181 digitised CSF objects from 78 patients corresponding to 15 clinically relevant categories and trained a multiclass convolutional neural network (CNN). RESULTS: The CNN classified the 15 categories with high accuracy (mean AUC 97.3%). By using explainable artificial intelligence (XAI), we demonstrate that the CNN identified meaningful cellular substructures in CSF cells recapitulating human pattern recognition. Based on the evaluation of 511 cells selected from 12 different CSF samples, we validated the CNN by comparing it with seven board-certified neuropathologists blinded for clinical information. Inter-rater agreement between the CNN and the ground truth was non-inferior (Krippendorff's alpha 0.79) compared with the agreement of seven human raters and the ground truth (mean Krippendorff's alpha 0.72, range 0.56-0.81). The CNN assigned the correct diagnostic label (inflammatory, haemorrhagic or neoplastic) in 10 out of 11 clinical samples, compared with 7-11 out of 11 by human raters. CONCLUSIONS: Our approach provides the basis to overcome current limitations in automated cell classification for routine diagnostics and demonstrates how a visual explanation framework can connect machine decision-making with cell properties and thus provide a novel versatile and quantitative method for investigating CSF manifestations of various neurological diseases.


Subject(s)
Deep Learning , Humans , Artificial Intelligence , Neural Networks, Computer , Image Processing, Computer-Assisted/methods
19.
Sci Rep ; 12(1): 18991, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36347879

ABSTRACT

Histological sections of the lymphatic system are usually the basis of static (2D) morphological investigations. Here, we performed a dynamic (4D) analysis of human reactive lymphoid tissue using confocal fluorescent laser microscopy in combination with machine learning. Based on tracks for T-cells (CD3), B-cells (CD20), follicular T-helper cells (PD1) and optical flow of follicular dendritic cells (CD35), we put forward the first quantitative analysis of movement-related and morphological parameters within human lymphoid tissue. We identified correlations of follicular dendritic cell movement and the behavior of lymphocytes in the microenvironment. In addition, we investigated the value of movement and/or morphological parameters for a precise definition of cell types (CD clusters). CD-clusters could be determined based on movement and/or morphology. Differentiating between CD3- and CD20 positive cells is most challenging and long term-movement characteristics are indispensable. We propose morphological and movement-related prototypes of cell entities applying machine learning models. Finally, we define beyond CD clusters new subgroups within lymphocyte entities based on long term movement characteristics. In conclusion, we showed that the combination of 4D imaging and machine learning is able to define characteristics of lymphocytes not visible in 2D histology.


Subject(s)
Dendritic Cells, Follicular , Lymphoid Tissue , Humans , Lymphoid Tissue/pathology , Dendritic Cells, Follicular/metabolism , T-Lymphocytes, Helper-Inducer , Lymphocytes , Machine Learning
20.
IEEE Trans Med Imaging ; PP2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36423312

ABSTRACT

We consider the reconstruction of brain activity from electroencephalography (EEG). This inverse problem can be formulated as a linear regression with independent Gaussian scale mixture priors for both the source and noise components. Crucial factors influencing the accuracy of the source estimation are not only the noise level but also its correlation structure, but existing approaches have not addressed the estimation of noise covariance matrices with full structure. To address this shortcoming, we develop hierarchical Bayesian (type-II maximum likelihood) models for observations with latent variables for source and noise, which are estimated jointly from data. As an extension to classical sparse Bayesian learning (SBL), where across-sensor observations are assumed to be independent and identically distributed, we consider Gaussian noise with full covariance structure. Using the majorization-maximization framework and Riemannian geometry, we derive an efficient algorithm for updating the noise covariance along the manifold of positive definite matrices. We demonstrate that our algorithm has guaranteed and fast convergence and validate it in simulations and with real MEG data. Our results demonstrate that the novel framework significantly improves upon state-of-the-art techniques in the real-world scenario where the noise is indeed non-diagonal and fullstructured. Our method has applications in many domains beyond biomagnetic inverse problems.

SELECTION OF CITATIONS
SEARCH DETAIL