48 research outputs found

    First Observation Of The Spin Rotational Structure Of The Hydroxymethyl Radical (h2coh) In The Ch2 Asymmetric Mode

    Get PDF
    Rotationally-resolved direct infrared absorption spectra of hydroxymethyl radical (H2_{2}COH) in the CH2_{2} asymmetric mode (\nub{2}) were observed for the first time using the Boulder difference frequency generation infrared spectrometer. Hydroxymethyl radical was formed with chemical selectivity via the reaction of Cl radical with CH3_{3}OH in a discharge slit-jet supersonic expansion. As a result of sub-Doppler linewidth and low rotational temperature, the \textit{b}-type rotational structure and spin-rotation splitting were fully resolved. In particular, tunneling splitting was observed due to the large-amplitude COH torsional mode. Because of the feasible permutation of hydrogens in the methylenic group, nuclear spin intensity alternation was given as: 3:1 for \textit{Ka_{a}}=even, odd in the 0+^{+} level, and 1:3 for \textit{Ka_{a}}=even, odd in the 0^{-} level of the ground vibrational state. The assignments were confirmed rigorously by four-line ground state combination differences, which agreed within the experimental frequency uncertainty (10 MHz). The identified transitions were fit with a Watson \textit{A}-reduction Hamiltonian including the spin rotational interaction, leading to unambiguous determination of asymmetric top spectroscopic constants, as well as spin rotational constants (ϵ\epsilonaa_{aa}, ϵ\epsilonbb_{bb}, ϵ\epsiloncc_{cc}) for the first time

    Deep Learning for the detection of microsatellite instability from histology images in colorectal cancer: a systematic literature review

    Get PDF
    Microsatellite instability (MSI) or deficient mismatch repair (dMMR) is a clinically important genetic feature affecting 10-15% of colorectal cancer (CRC) patients. Patients with metastatic MSI/dMMR CRC are eligible for therapy with immune checkpoint inhibitors, making MSI/dMMR the most important immuno-oncological biomarker in CRC. Gold standard tests for detection of MSI/dMMR in CRC are based on wet laboratory tests such as immunohistochemistry (IHC) or DNA extraction with subsequent polymerase chain reaction (PCR). However, since 2019, advances in Deep Learning (DL), an Artificial Intelligence (AI) technology, have enabled the prediction of MSI/dMMR directly from digitized routine haematoxylin and eosin (H&E) histopathology slides with high accuracy. In addition to the initial proof-of-concept publication in 2019, twelve subsequent studies have refined, improved, and further validated this approach. At this moment, MSI/dMMR prediction using Deep Learning has become a widely used benchmark task for academic studies in the field of computational pathology. Beyond academic use, this assay has attracted commercial interest from companies with the possibility of approval as a diagnostic device in the near future. In this review, we summarize and quantitatively compare the existing evidence on Deep-Learning-based detection of MSI/dMMR in CRC and discuss the need for further improvement and potential for integration into routine pathological workflows. Ultimately, this DL-based method could facilitate the identification of patients eligible for treatment with immune checkpoint inhibitors by pre-screening or replacement of current methods

    Artificial intelligence for detection of microsatellite instability in colorectal cancer-a multicentric analysis of a pre-screening tool for clinical application.

    Get PDF
    BACKGROUND Microsatellite instability (MSI)/mismatch repair deficiency (dMMR) is a key genetic feature which should be tested in every patient with colorectal cancer (CRC) according to medical guidelines. Artificial intelligence (AI) methods can detect MSI/dMMR directly in routine pathology slides, but the test performance has not been systematically investigated with predefined test thresholds. METHOD We trained and validated AI-based MSI/dMMR detectors and evaluated predefined performance metrics using nine patient cohorts of 8343 patients across different countries and ethnicities. RESULTS Classifiers achieved clinical-grade performance, yielding an area under the receiver operating curve (AUROC) of up to 0.96 without using any manual annotations. Subsequently, we show that the AI system can be applied as a rule-out test: by using cohort-specific thresholds, on average 52.73% of tumors in each surgical cohort [total number of MSI/dMMR = 1020, microsatellite stable (MSS)/ proficient mismatch repair (pMMR) = 7323 patients] could be identified as MSS/pMMR with a fixed sensitivity at 95%. In an additional cohort of N = 1530 (MSI/dMMR = 211, MSS/pMMR = 1319) endoscopy biopsy samples, the system achieved an AUROC of 0.89, and the cohort-specific threshold ruled out 44.12% of tumors with a fixed sensitivity at 95%. As a more robust alternative to cohort-specific thresholds, we showed that with a fixed threshold of 0.25 for all the cohorts, we can rule-out 25.51% in surgical specimens and 6.10% in biopsies. INTERPRETATION When applied in a clinical setting, this means that the AI system can rule out MSI/dMMR in a quarter (with global thresholds) or half of all CRC patients (with local fine-tuning), thereby reducing cost and turnaround time for molecular profiling

    Pan-cancer image-based detection of clinically actionable genetic alterations

    Get PDF
    Molecular alterations in cancer can cause phenotypic changes in tumor cells and their microenvironment. Routine histopathology tissue slides, which are ubiquitously available, can reflect such morphological changes. Here, we show that deep learning can consistently infer a wide range of genetic mutations, molecular tumor subtypes, gene expression signatures and standard pathology biomarkers directly from routine histology. We developed, optimized, validated and publicly released a one-stop-shop workflow and applied it to tissue slides of more than 5,000 patients across multiple solid tumors. Our findings show that a single deep learning algorithm can be trained to predict a wide range of molecular alterations from routine, paraffin-embedded histology slides stained with hematoxylin and eosin. These predictions generalize to other populations and are spatially resolved. Our method can be implemented on mobile hardware, potentially enabling point-of-care diagnostics for personalized cancer treatment. More generally, this approach could elucidate and quantify genotype–phenotype links in cancer

    Clinical-grade Detection of Microsatellite Instability in Colorectal Tumors by Deep Learning

    Get PDF
    Background and Aims: Microsatellite instability (MSI) and mismatch-repair deficiency (dMMR) in colorectal tumors are used to select treatment for patients. Deep learning can detect MSI and dMMR in tumor samples on routine histology slides faster and cheaper than molecular assays. But clinical application of this technology requires high performance and multisite validation, which have not yet been performed. Methods: We collected hematoxylin and eosin-stained slides, and findings from molecular analyses for MSI and dMMR, from 8836 colorectal tumors (of all stages) included in the MSIDETECT consortium study, from Germany, the Netherlands, the United Kingdom, and the United States. Specimens with dMMR were identified by immunohistochemistry analyses of tissue microarrays for loss of MLH1, MSH2, MSH6, and/or PMS2. Specimens with MSI were identified by genetic analyses. We trained a deep-learning detector to identify samples with MSI from these slides; performance was assessed by cross-validation (n=6406 specimens) and validated in an external cohort (n=771 specimens). Prespecified endpoints were area under the receiver operating characteristic (AUROC) curve and area under the precision-recall curve (AUPRC). Results: The deep-learning detector identified specimens with dMMR or MSI with a mean AUROC curve of 0.92 (lower bound 0.91, upper bound 0.93) and an AUPRC of 0.63 (range, 0.59–0.65), or 67% specificity and 95% sensitivity, in the cross-validation development cohort. In the validation cohort, the classifier identified samples with dMMR with an AUROC curve of 0.95 (range, 0.92–0.96) without image-preprocessing and an AUROC curve of 0.96 (range, 0.93–0.98) after color normalization. Conclusions: We developed a deep-learning system that detects colorectal cancer specimens with dMMR or MSI using hematoxylin and eosin-stained slides; it detected tissues with dMMR with an AUROC of 0.96 in a large, international validation cohort. This system might be used for high-throughput, low-cost evaluation of colorectal tissue specimens

    Bentonite der Insel Milos/Griechenland

    No full text
    corecore