Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
J Clin Pathol ; 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39168612

RESUMEN

AIMS: The International Association for the Study of Lung Cancer (IASLC) has proposed a new histological grading system for invasive lung adenocarcinoma (LUAD). However, the efficacy of this grading system in predicting distant metastases in patients with LUAD remains unexplored. This study aims to assess the potential of the IASLC grading system in predicting the occurrence of brain and bone metastases in patients with resectable LUAD, thereby identifying individuals at high risk of post-surgery distant metastasis. METHODS: We retrospectively analysed clinical data and pathological reports of 174 patients with early-stage LUAD who underwent surgical resection between 2008 and 2015 at our cancer center. Patients were monitored for 5 years, and their bone and brain metastasis-free survival rates were determined. RESULTS: 28 out of 174 patients developed distant metastases in 5 years with a median overall survival of 60 months for metastasis-free patients and 38.3 months for patients with distant metastasis. Tumour grading of all samples was evaluated by both IASLC grading and predominant pattern-based grading systems. Receiver operating characteristic (ROC) curves were used to evaluate the predictive capabilities of the IASLC grading system and tumour stage for distant metastasis. Compared with the predominant pattern-based grading system, the IASLC grading system showed a better correlation with the incidence of distant metastasis and lymphovascular invasion. ROC analyses revealed that the IASLC grading system outperformed tumour stage in predicting distant metastasis. CONCLUSIONS: Our study indicates that the IASLC grading system is capable of predicting the incidence of distant metastasis among patients with early-stage invasive LUAD.

2.
EBioMedicine ; 107: 105287, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39154539

RESUMEN

BACKGROUND: Multiplexed immunofluorescence (mIF) staining, such as CODEX and MIBI, holds significant clinical value for various fields, such as disease diagnosis, biological research, and drug development. However, these techniques are often hindered by high time and cost requirements. METHODS: Here we present a Multimodal-Attention-based virtual mIF Staining (MAS) system that utilises a deep learning model to extract potential antibody-related features from dual-modal non-antibody-stained fluorescence imaging, specifically autofluorescence (AF) and DAPI imaging. The MAS system simultaneously generates predictions of mIF with multiple survival-associated biomarkers in gastric cancer using self- and multi-attention learning mechanisms. FINDINGS: Experimental results with 180 pathological slides from 94 patients with gastric cancer demonstrate the efficiency and consistent performance of the MAS system in both cancer and noncancer gastric tissues. Furthermore, we showcase the prognostic accuracy of the virtual mIF images of seven gastric cancer related biomarkers, including CD3, CD20, FOXP3, PD1, CD8, CD163, and PD-L1, which is comparable to those obtained from the standard mIF staining. INTERPRETATION: The MAS system rapidly generates reliable multiplexed staining, greatly reducing the cost of mIF and improving clinical workflow. FUNDING: Stanford 2022 HAI Seed Grant; National Institutes of Health 1R01CA256890.


Asunto(s)
Biomarcadores de Tumor , Técnica del Anticuerpo Fluorescente , Imagen Óptica , Neoplasias Gástricas , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/diagnóstico por imagen , Humanos , Pronóstico , Biomarcadores de Tumor/metabolismo , Imagen Óptica/métodos , Técnica del Anticuerpo Fluorescente/métodos , Coloración y Etiquetado/métodos , Procesamiento de Imagen Asistido por Computador/métodos
3.
ACS Meas Sci Au ; 4(4): 338-417, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39193565

RESUMEN

Proteomics is the large scale study of protein structure and function from biological systems through protein identification and quantification. "Shotgun proteomics" or "bottom-up proteomics" is the prevailing strategy, in which proteins are hydrolyzed into peptides that are analyzed by mass spectrometry. Proteomics studies can be applied to diverse studies ranging from simple protein identification to studies of proteoforms, protein-protein interactions, protein structural alterations, absolute and relative protein quantification, post-translational modifications, and protein stability. To enable this range of different experiments, there are diverse strategies for proteome analysis. The nuances of how proteomic workflows differ may be challenging to understand for new practitioners. Here, we provide a comprehensive overview of different proteomics methods. We cover from biochemistry basics and protein extraction to biological interpretation and orthogonal validation. We expect this Review will serve as a handbook for researchers who are new to the field of bottom-up proteomics.

4.
J Proteome Res ; 23(8): 3649-3658, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39007500

RESUMEN

Noninvasive detection of protein biomarkers in plasma is crucial for clinical purposes. Liquid chromatography-mass spectrometry (LC-MS) is the gold standard technique for plasma proteome analysis, but despite recent advances, it remains limited by throughput, cost, and coverage. Here, we introduce a new hybrid method that integrates direct infusion shotgun proteome analysis (DISPA) with nanoparticle (NP) protein corona enrichment for high-throughput and efficient plasma proteomic profiling. We realized over 280 protein identifications in 1.4 min collection time, which enables a potential throughput of approximately 1000 samples daily. The identified proteins are involved in valuable pathways, and 44 of the proteins are FDA-approved biomarkers. The robustness and quantitative accuracy of this method were evaluated across multiple NPs and concentrations with a mean coefficient of variation of 17%. Moreover, different protein corona profiles were observed among various NPs based on their distinct surface modifications, and all NP protein profiles exhibited deeper coverage and better quantification than neat plasma. Our streamlined workflow merges coverage and throughput with precise quantification, leveraging both DISPA and NP protein corona enrichment. This underscores the significant potential of DISPA when paired with NP sample preparation techniques for plasma proteome studies.


Asunto(s)
Proteínas Sanguíneas , Nanopartículas , Corona de Proteínas , Proteoma , Proteómica , Humanos , Proteínas Sanguíneas/análisis , Proteínas Sanguíneas/química , Nanopartículas/química , Corona de Proteínas/química , Corona de Proteínas/análisis , Proteoma/análisis , Proteómica/métodos , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Biomarcadores/sangre
5.
Int J Surg ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884256

RESUMEN

BACKGROUND: Tertiary lymphoid structures (TLSs) are associated with favorable prognosis and enhanced response to anti-cancer therapy. A digital assessment of TLSs could provide an objective alternative that mitigates variability inherent in manual evaluation. This study aimed to develop and validate a digital gene panel based on biological prior knowledge for assessment of TLSs, and further investigate its associations with survival and multiple anti-cancer therapies. MATERIALS AND METHODS: The present study involved 1,704 patients with gastric cancer from seven cancer centers. TLSs were identified morphologically through hematoxylin-and-eosin staining. We further developed a digital score based on targeted gene expression profiling to assess TLSs status, recorded as gene signature of tertiary lymphoid structures (gsTLS). For enhanced interpretability, we employed the SHapley Additive exPlanation (SHAP) analysis to elucidate its contribution to the prediction. We next evaluated the signature's associations with prognosis, and investigated its predictive accuracy for multiple anti-cancer therapies, including adjuvant chemotherapy and immunotherapy. RESULTS: The gsTLS panel with nine gene features achieved high accuracies in predicting TLSs status in the training, internal and external validation cohorts (area under the curve, range: 0.729-0.791). In multivariable analysis, gsTLS remained an independent predictor of disease-free and overall survival (hazard ratio, range: 0.346-0.743, all P < 0.05) after adjusting for other clinicopathological variables. SHAP analysis highlighted gsTLS as the strongest predictor of TLSs status compared with clinical features. Importantly, patients with high gsTLS (but not those with low gsTLS) exhibited substantial benefits from adjuvant chemotherapy (P < 0.05). Furthermore, we found that the objective response rate to anti-programmed cell death protein 1 (anti-PD-1) immunotherapy was significantly higher in the high-gsTLS group (40.7%) versus the low-gsTLS group (5.6%, P = 0.036), and the diagnosis was independent from Epstein-Barr virus (EBV), tumor mutation burden (TMB), and programmed cell death-ligand 1 (PD-L1) expression. CONCLUSION: The gsTLS digital panel enables accurate assessment of TLSs status, and provides information regarding prognosis and responses to multiple therapies for gastric cancer.

6.
J Immunother Cancer ; 12(5)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38749538

RESUMEN

BACKGROUND: Only a subset of patients with gastric cancer experience long-term benefits from immune checkpoint inhibitors (ICIs). Currently, there is a deficiency in precise predictive biomarkers for ICI efficacy. The aim of this study was to develop and validate a pathomics-driven ensemble model for predicting the response to ICIs in gastric cancer, using H&E-stained whole slide images (WSI). METHODS: This multicenter study retrospectively collected and analyzed H&E-stained WSIs and clinical data from 584 patients with gastric cancer. An ensemble model, integrating four classifiers: least absolute shrinkage and selection operator, k-nearest neighbors, decision trees, and random forests, was developed and validated using pathomics features, with the objective of predicting the therapeutic efficacy of immune checkpoint inhibition. Model performance was evaluated using metrics including the area under the curve (AUC), sensitivity, and specificity. Additionally, SHAP (SHapley Additive exPlanations) analysis was used to explain the model's predicted values as the sum of the attribution values for each input feature. Pathogenomics analysis was employed to explain the molecular mechanisms underlying the model's predictions. RESULTS: Our pathomics-driven ensemble model effectively stratified the response to ICIs in training cohort (AUC 0.985 (95% CI 0.971 to 0.999)), which was further validated in internal validation cohort (AUC 0.921 (95% CI 0.839 to 0.999)), as well as in external validation cohort 1 (AUC 0.914 (95% CI 0.837 to 0.990)), and external validation cohort 2 (0.927 (95% CI 0.802 to 0.999)). The univariate Cox regression analysis revealed that the prediction signature of pathomics-driven ensemble model was a prognostic factor for progression-free survival in patients with gastric cancer who underwent immunotherapy (p<0.001, HR 0.35 (95% CI 0.24 to 0.50)), and remained an independent predictor after multivariable Cox regression adjusted for clinicopathological variables, (including sex, age, carcinoembryonic antigen, carbohydrate antigen 19-9, therapy regime, line of therapy, differentiation, location and programmed death ligand 1 (PD-L1) expression in all patients (p<0.001, HR 0.34 (95% CI 0.24 to 0.50)). Pathogenomics analysis suggested that the ensemble model is driven by molecular-level immune, cancer, metabolism-related pathways, and was correlated with the immune-related characteristics, including immune score, Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data score, and tumor purity. CONCLUSIONS: Our pathomics-driven ensemble model exhibited high accuracy and robustness in predicting the response to ICIs using WSIs. Therefore, it could serve as a novel and valuable tool to facilitate precision immunotherapy.


Asunto(s)
Inmunoterapia , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Masculino , Femenino , Inmunoterapia/métodos , Estudios Retrospectivos , Persona de Mediana Edad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Anciano
7.
J Proteome Res ; 23(6): 1871-1882, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38713528

RESUMEN

The coevolution of liquid chromatography (LC) with mass spectrometry (MS) has shaped contemporary proteomics. LC hyphenated to MS now enables quantification of more than 10,000 proteins in a single injection, a number that likely represents most proteins in specific human cells or tissues. Separations by ion mobility spectrometry (IMS) have recently emerged to complement LC and further improve the depth of proteomics. Given the theoretical advantages in speed and robustness of IMS in comparison to LC, we envision that ongoing improvements to IMS paired with MS may eventually make LC obsolete, especially when combined with targeted or simplified analyses, such as rapid clinical proteomics analysis of defined biomarker panels. In this perspective, we describe the need for faster analysis that might drive this transition, the current state of direct infusion proteomics, and discuss some technical challenges that must be overcome to fully complete the transition to entirely gas phase proteomics.


Asunto(s)
Espectrometría de Movilidad Iónica , Proteómica , Proteómica/métodos , Espectrometría de Movilidad Iónica/métodos , Humanos , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos
8.
bioRxiv ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38370692

RESUMEN

Non-invasive detection of protein biomarkers in plasma is crucial for clinical purposes. Liquid chromatography mass spectrometry (LC-MS) is the gold standard technique for plasma proteome analysis, but despite recent advances, it remains limited by throughput, cost, and coverage. Here, we introduce a new hybrid method, which integrates direct infusion shotgun proteome analysis (DISPA) with nanoparticle (NP) protein coronas enrichment for high throughput and efficient plasma proteomic profiling. We realized over 280 protein identifications in 1.4 minutes collection time, which enables a potential throughput of approximately 1,000 samples daily. The identified proteins are involved in valuable pathways and 44 of the proteins are FDA approved biomarkers. The robustness and quantitative accuracy of this method were evaluated across multiple NPs and concentrations with a mean coefficient of variation at 17%. Moreover, different protein corona profiles were observed among various nanoparticles based on their distinct surface modifications, and all NP protein profiles exhibited deeper coverage and better quantification than neat plasma. Our streamlined workflow merges coverage and throughput with precise quantification, leveraging both DISPA and NP protein corona enrichments. This underscores the significant potential of DISPA when paired with NP sample preparation techniques for plasma proteome studies.

9.
Comput Med Imaging Graph ; 112: 102336, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38244280

RESUMEN

Rigid pre-registration involving local-global matching or other large deformation scenarios is crucial. Current popular methods rely on unsupervised learning based on grayscale similarity, but under circumstances where different poses lead to varying tissue structures, or where image quality is poor, these methods tend to exhibit instability and inaccuracies. In this study, we propose a novel method for medical image registration based on arbitrary voxel point of interest matching, called query point quizzer (QUIZ). QUIZ focuses on the correspondence between local-global matching points, specifically employing CNN for feature extraction and utilizing the Transformer architecture for global point matching queries, followed by applying average displacement for local image rigid transformation.We have validated this approach on a large deformation dataset of cervical cancer patients, with results indicating substantially smaller deviations compared to state-of-the-art methods. Remarkably, even for cross-modality subjects, it achieves results surpassing the current state-of-the-art.


Asunto(s)
Algoritmos , Neoplasias del Cuello Uterino , Femenino , Humanos , Neoplasias del Cuello Uterino/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos
10.
J Clin Invest ; 134(6)2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38271117

RESUMEN

BACKGROUNDThe tumor immune microenvironment can provide prognostic and therapeutic information. We aimed to develop noninvasive imaging biomarkers from computed tomography (CT) for comprehensive evaluation of immune context and investigate their associations with prognosis and immunotherapy response in gastric cancer (GC).METHODSThis study involved 2,600 patients with GC from 9 independent cohorts. We developed and validated 2 CT imaging biomarkers (lymphoid radiomics score [LRS] and myeloid radiomics score [MRS]) for evaluating the IHC-derived lymphoid and myeloid immune context respectively, and integrated them into a combined imaging biomarker [LRS/MRS: low(-) or high(+)] with 4 radiomics immune subtypes: 1 (-/-), 2 (+/-), 3 (-/+), and 4 (+/+). We further evaluated the imaging biomarkers' predictive values on prognosis and immunotherapy response.RESULTSThe developed imaging biomarkers (LRS and MRS) had a high accuracy in predicting lymphoid (AUC range: 0.765-0.773) and myeloid (AUC range: 0.736-0.750) immune context. Further, similar to the IHC-derived immune context, 2 imaging biomarkers (HR range: 0.240-0.761 for LRS; 1.301-4.012 for MRS) and the combined biomarker were independent predictors for disease-free and overall survival in the training and all validation cohorts (all P < 0.05). Additionally, patients with high LRS or low MRS may benefit more from immunotherapy (P < 0.001). Further, a highly heterogeneous outcome on objective response ​rate was observed in 4 imaging subtypes: 1 (-/-) with 27.3%, 2 (+/-) with 53.3%, 3 (-/+) with 10.2%, and 4 (+/+) with 30.0% (P < 0.0001).CONCLUSIONThe noninvasive imaging biomarkers could accurately evaluate the immune context and provide information regarding prognosis and immunotherapy for GC.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/terapia , Radiómica , Inmunoterapia , Tomografía Computarizada por Rayos X , Microambiente Tumoral , Biomarcadores , Pronóstico
11.
J Natl Cancer Inst ; 116(4): 555-564, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-37982756

RESUMEN

BACKGROUND: Intratumor heterogeneity drives disease progression and treatment resistance, which can lead to poor patient outcomes. Here, we present a computational approach for quantification of cancer cell diversity in routine hematoxylin-eosin-stained histopathology images. METHODS: We analyzed publicly available digitized whole-slide hematoxylin-eosin images for 2000 patients. Four tumor types were included: lung, head and neck, colon, and rectal cancers, representing major histology subtypes (adenocarcinomas and squamous cell carcinomas). We performed single-cell analysis on hematoxylin-eosin images and trained a deep convolutional autoencoder to automatically learn feature representations of individual cancer nuclei. We then computed features of intranuclear variability and internuclear diversity to quantify tumor heterogeneity. Finally, we used these features to build a machine-learning model to predict patient prognosis. RESULTS: A total of 68 million cancer cells were segmented and analyzed for nuclear image features. We discovered multiple morphological subtypes of cancer cells (range = 15-20) that co-exist within the same tumor, each with distinct phenotypic characteristics. Moreover, we showed that a higher morphological diversity is associated with chromosome instability and genomic aneuploidy. A machine-learning model based on morphological diversity demonstrated independent prognostic values across tumor types (hazard ratio range = 1.62-3.23, P < .035) in validation cohorts and further improved prognostication when combined with clinical risk factors. CONCLUSIONS: Our study provides a practical approach for quantifying intratumor heterogeneity based on routine histopathology images. The cancer cell diversity score can be used to refine risk stratification and inform personalized treatment strategies.


Asunto(s)
Carcinoma de Células Escamosas , Humanos , Hematoxilina , Eosina Amarillenta-(YS) , Pronóstico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad
12.
IEEE Trans Pattern Anal Mach Intell ; 46(5): 3692-3706, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38147423

RESUMEN

Facial editing is to manipulate the facial attributes of a given face image. Nowadays, with the development of generative models, users can easily generate 2D and 3D facial images with high fidelity and 3D-aware consistency. However, existing works are incapable of delivering a continuous and fine-grained editing mode (e.g., editing a slightly smiling face to a big laughing one) with natural interactions with users. In this work, we propose Talk-to-Edit, an interactive facial editing framework that performs fine-grained attribute manipulation through dialog between the user and the system. Our key insight is to model a continual "semantic field" in the GAN latent space. 1) Unlike previous works that regard the editing as traversing straight lines in the latent space, here the fine-grained editing is formulated as finding a curving trajectory that respects fine-grained attribute landscape on the semantic field. 2) The curvature at each step is location-specific and determined by the input image as well as the users' language requests. 3) To engage the users in a meaningful dialog, our system generates language feedback by considering both the user request and the current state of the semantic field. We demonstrate the effectiveness of our proposed framework on both 2D and 3D-aware generative models. We term the semantic field for the 3D-aware models as "tri-plane" flow, as it corresponds to the changes not only in the color space but also in the density space. We also contribute CelebA-Dialog, a visual-language facial editing dataset to facilitate large-scale study. Specifically, each image has manually annotated fine-grained attribute annotations as well as template-based textual descriptions in natural language. Extensive quantitative and qualitative experiments demonstrate the superiority of our framework in terms of 1) the smoothness of fine-grained editing, 2) the identity/attribute preservation, and 3) the visual photorealism and dialog fluency. Notably, the user study validates that our overall system is consistently favored by around 80% of the participants.

13.
Med Image Anal ; 91: 102984, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37837690

RESUMEN

The accurate delineation of organs-at-risk (OARs) is a crucial step in treatment planning during radiotherapy, as it minimizes the potential adverse effects of radiation on surrounding healthy organs. However, manual contouring of OARs in computed tomography (CT) images is labor-intensive and susceptible to errors, particularly for low-contrast soft tissue. Deep learning-based artificial intelligence algorithms surpass traditional methods but require large datasets. Obtaining annotated medical images is both time-consuming and expensive, hindering the collection of extensive training sets. To enhance the performance of medical image segmentation, augmentation strategies such as rotation and Gaussian smoothing are employed during preprocessing. However, these conventional data augmentation techniques cannot generate more realistic deformations, limiting improvements in accuracy. To address this issue, this study introduces a statistical deformation model-based data augmentation method for volumetric medical image segmentation. By applying diverse and realistic data augmentation to CT images from a limited patient cohort, our method significantly improves the fully automated segmentation of OARs across various body parts. We evaluate our framework on three datasets containing tumor OARs from the head, neck, chest, and abdomen. Test results demonstrate that the proposed method achieves state-of-the-art performance in numerous OARs segmentation challenges. This innovative approach holds considerable potential as a powerful tool for various medical imaging-related sub-fields, effectively addressing the challenge of limited data access.


Asunto(s)
Inteligencia Artificial , Neoplasias , Humanos , Algoritmos , Cuello , Tomografía Computarizada por Rayos X/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Planificación de la Radioterapia Asistida por Computador/métodos
14.
Med Image Anal ; 91: 102998, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37857066

RESUMEN

Radiotherapy serves as a pivotal treatment modality for malignant tumors. However, the accuracy of radiotherapy is significantly compromised due to respiratory-induced fluctuations in the size, shape, and position of the tumor. To address this challenge, we introduce a deep learning-anchored, volumetric tumor tracking methodology that employs single-angle X-ray projection images. This process involves aligning the intraoperative two-dimensional (2D) X-ray images with the pre-treatment three-dimensional (3D) planning Computed Tomography (CT) scans, enabling the extraction of the 3D tumor position and segmentation. Prior to therapy, a bespoke patient-specific tumor tracking model is formulated, leveraging a hybrid data augmentation, style correction, and registration network to create a mapping from single-angle 2D X-ray images to the corresponding 3D tumors. During the treatment phase, real-time X-ray images are fed into the trained model, producing the respective 3D tumor positioning. Rigorous validation conducted on actual patient lung data and lung phantoms attests to the high localization precision of our method at lowered radiation doses, thus heralding promising strides towards enhancing the precision of radiotherapy.


Asunto(s)
Aprendizaje Profundo , Neoplasias , Humanos , Imagenología Tridimensional/métodos , Rayos X , Tomografía Computarizada por Rayos X/métodos , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia , Tomografía Computarizada de Haz Cónico/métodos
16.
Sensors (Basel) ; 23(21)2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37960379

RESUMEN

Batch process monitoring datasets usually contain missing data, which decreases the performance of data-driven modeling for fault identification and optimal control. Many methods have been proposed to impute missing data; however, they do not fulfill the need for data quality, especially in sensor datasets with different types of missing data. We propose a hybrid missing data imputation method for batch process monitoring datasets with multi-type missing data. In this method, the missing data is first classified into five categories based on the continuous missing duration and the number of variables missing simultaneously. Then, different categories of missing data are step-by-step imputed considering their unique characteristics. A combination of three single-dimensional interpolation models is employed to impute transient isolated missing values. An iterative imputation based on a multivariate regression model is designed for imputing long-term missing variables, and a combination model based on single-dimensional interpolation and multivariate regression is proposed for imputing short-term missing variables. The Long Short-Term Memory (LSTM) model is utilized to impute both short-term and long-term missing samples. Finally, a series of experiments for different categories of missing data were conducted based on a real-world batch process monitoring dataset. The results demonstrate that the proposed method achieves higher imputation accuracy than other comparative methods.

17.
ArXiv ; 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38013887

RESUMEN

Proteomics is the large scale study of protein structure and function from biological systems through protein identification and quantification. "Shotgun proteomics" or "bottom-up proteomics" is the prevailing strategy, in which proteins are hydrolyzed into peptides that are analyzed by mass spectrometry. Proteomics studies can be applied to diverse studies ranging from simple protein identification to studies of proteoforms, protein-protein interactions, protein structural alterations, absolute and relative protein quantification, post-translational modifications, and protein stability. To enable this range of different experiments, there are diverse strategies for proteome analysis. The nuances of how proteomic workflows differ may be challenging to understand for new practitioners. Here, we provide a comprehensive overview of different proteomics methods to aid the novice and experienced researcher. We cover from biochemistry basics and protein extraction to biological interpretation and orthogonal validation. We expect this work to serve as a basic resource for new practitioners in the field of shotgun or bottom-up proteomics.

18.
Cell Rep Med ; 4(8): 101146, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37557177

RESUMEN

The tumor microenvironment (TME) plays a critical role in disease progression and is a key determinant of therapeutic response in cancer patients. Here, we propose a noninvasive approach to predict the TME status from radiological images by combining radiomics and deep learning analyses. Using multi-institution cohorts of 2,686 patients with gastric cancer, we show that the radiological model accurately predicted the TME status and is an independent prognostic factor beyond clinicopathologic variables. The model further predicts the benefit from adjuvant chemotherapy for patients with localized disease. In patients treated with checkpoint blockade immunotherapy, the model predicts clinical response and further improves predictive accuracy when combined with existing biomarkers. Our approach enables noninvasive assessment of the TME, which opens the door for longitudinal monitoring and tracking response to cancer therapy. Given the routine use of radiologic imaging in oncology, our approach can be extended to many other solid tumor types.


Asunto(s)
Aprendizaje Profundo , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/terapia , Microambiente Tumoral , Inmunoterapia , Quimioterapia Adyuvante
19.
Nat Commun ; 14(1): 5135, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612313

RESUMEN

Substantial progress has been made in using deep learning for cancer detection and diagnosis in medical images. Yet, there is limited success on prediction of treatment response and outcomes, which has important implications for personalized treatment strategies. A significant hurdle for clinical translation of current data-driven deep learning models is lack of interpretability, often attributable to a disconnect from the underlying pathobiology. Here, we present a biology-guided deep learning approach that enables simultaneous prediction of the tumor immune and stromal microenvironment status as well as treatment outcomes from medical images. We validate the model for predicting prognosis of gastric cancer and the benefit from adjuvant chemotherapy in a multi-center international study. Further, the model predicts response to immune checkpoint inhibitors and complements clinically approved biomarkers. Importantly, our model identifies a subset of mismatch repair-deficient tumors that are non-responsive to immunotherapy and may inform the selection of patients for combination treatments.


Asunto(s)
Neoplasias Encefálicas , Aprendizaje Profundo , Humanos , Inmunoterapia , Quimioterapia Adyuvante , Biología , Microambiente Tumoral
20.
J Am Soc Mass Spectrom ; 34(9): 1858-1867, 2023 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-37463334

RESUMEN

Skeletal muscle is a major regulatory tissue of whole-body metabolism and is composed of a diverse mixture of cell (fiber) types. Aging and several diseases differentially affect the various fiber types, and therefore, investigating the changes in the proteome in a fiber-type specific manner is essential. Recent breakthroughs in isolated single muscle fiber proteomics have started to reveal heterogeneity among fibers. However, existing procedures are slow and laborious, requiring 2 h of mass spectrometry time per single muscle fiber; 50 fibers would take approximately 4 days to analyze. Thus, to capture the high variability in fibers both within and between individuals requires advancements in high throughput single muscle fiber proteomics. Here we use a single cell proteomics method to enable quantification of single muscle fiber proteomes in 15 min total instrument time. As proof of concept, we present data from 53 isolated skeletal muscle fibers obtained from two healthy individuals analyzed in 13.25 h. Adapting single cell data analysis techniques to integrate the data, we can reliably separate type 1 and 2A fibers. Ninety-four proteins were statistically different between clusters indicating alteration of proteins involved in fatty acid oxidation, oxidative phosphorylation, and muscle structure and contractile function. Our results indicate that this method is significantly faster than prior single fiber methods in both data collection and sample preparation while maintaining sufficient proteome depth. We anticipate this assay will enable future studies of single muscle fibers across hundreds of individuals, which has not been possible previously due to limitations in throughput.


Asunto(s)
Proteoma , Proteómica , Humanos , Proteoma/metabolismo , Proteómica/métodos , Flujo de Trabajo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...