Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Neoplasia ; 50: 100977, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38354688

RESUMO

BACKGROUND: The inconformity (IC) between pathological and imaging remissions after neoadjuvant immunotherapy in patients with NSCLC can affect the evaluation of curative effect of neoadjuvant therapy and the decision regarding the chance of surgery. MATERIALS AND METHODS: Patients who achieved disease control(CR/PR/SD) after neoadjuvant chemoimmunotherapy from a clinical trial (NCT04326153) and after neoadjuvant chemotherapy during the same period were enrolled in this study. All patients underwent radical resection and systematic mediastinal lymphadenectomy after neoadjuvant treatments. The pathological remission, immunohistochemistry (CD4, CD8, CD20, CD56, FoxP3, CD68, CD163, CD11b tumor-infiltrating lymphocytes, or macrophages), and single-source dual-energy computed tomography (ssDECT) scans were assessed. The IC between imaging remission by CT and pathological remission was investigated. The underlying cause of IC, the correlation between IC and DFS, and prognostic biomarkers were explored. RESULTS: After neoadjuvant immunotherapy, enhanced immune killing and reduced immunosuppressive performance were observed. 70 % of neoadjuvant chemoimmunotherapy patients were in high/medium IC level. Massive necrosis and repair around and inside the cancer nest were the main pathological changes observed 30-45 days post-treatment with PD1/PD-L1 antibody and were the main causes of IC between the pathology and imaging responses after neoadjuvant immunotherapy. High IC and preoperative CD8 expression (H score ≥ 3) indicate a high pathological response rate and prolonged DFS. Iodine material density ssDECT images showed that the iodine content in the lesion causes hyperattenuation in post-neoadjuvant lesion in PCR patient. CONCLUSIONS: Compared to chemotherapy and targeted therapy, the efficacy of neoadjuvant immunotherapy was underestimated based on the RECIST criteria due to the unique antitumor therapeutic mechanism. Preoperative CD8+ expression and ssDECT predict this IC and evaluate the residual tumor cells. This is of great significance for screening immune beneficiaries and making more accurate judgments about the timing of surgery.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Iodo , Neoplasias Pulmonares , Humanos , Terapia Neoadjuvante , Microambiente Tumoral , Carcinoma Pulmonar de Células não Pequenas/patologia , Tomografia Computadorizada por Raios X , Imunoterapia , Neoplasias Pulmonares/patologia , Iodo/farmacologia , Iodo/uso terapêutico
2.
Phys Med Biol ; 69(7)2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38224617

RESUMO

Objective.In the realm of utilizing artificial intelligence (AI) for medical image analysis, the paradigm of 'signal-image-knowledge' has remained unchanged. However, the process of 'signal to image' inevitably introduces information distortion, ultimately leading to irrecoverable biases in the 'image to knowledge' process. Our goal is to skip reconstruction and build a diagnostic model directly from the raw data (signal).Approach. This study focuses on computed tomography (CT) and its raw data (sinogram) as the research subjects. We simulate the real-world process of 'human-signal-image' using the workflow 'CT-simulated data- reconstructed CT,' and we develop a novel AI predictive model directly targeting raw data (RCTM). This model comprises orientation, spatial, and global analysis modules, embodying the fusion of local to global information extraction from raw data. We selected 1994 patients with retrospective cases of solid lung nodules and modeled different types of data.Main results. We employed predefined radiomic features to assess the diagnostic feature differences caused by reconstruction. The results indicated that approximately 14% of the features had Spearman correlation coefficients below 0.8. These findings suggest that despite the increasing maturity of CT reconstruction algorithms, they still introduce perturbations to diagnostic features. Moreover, our proposed RCTM achieved an area under the curve (AUC) of 0.863 in the diagnosis task, showcasing a comprehensive superiority over models constructed from secondary reconstructed CTs (0.840, 0.822, and 0.825). Additionally, the performance of RCTM closely resembled that of models constructed from original CT scans (0.868, 0.878, and 0.866).Significance. The diagnostic and therapeutic approach directly based on CT raw data can enhance the precision of AI models and the concept of 'signal-to-image' can be extended to other types of imaging. AI diagnostic models tailored to raw data offer the potential to disrupt the traditional paradigm of 'signal-image-knowledge', opening up new avenues for more accurate medical diagnostics.


Assuntos
Inteligência Artificial , Radiologia , Humanos , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/métodos , Algoritmos , Processamento de Imagem Assistida por Computador/métodos
3.
Med Phys ; 51(1): 179-191, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37929807

RESUMO

BACKGROUND: Lymphovascular invasion (LVI) status plays an important role in treatment decision-making in rectal cancer (RC). Intravoxel incoherent motion (IVIM) imaging has been shown to detect LVI; however, making better use of IVIM data remains an important issue that needs to be discussed. PURPOSE: We proposed to explore the best way to use IVIM quantitative parameters and images to construct radiomics models for the noninvasive detection of LVI in RC. METHODS: A total of 83 patients (LVI negative (LVI-): LVI positive (LVI+) = 51:32) with postoperative pathology-confirmed LVI status in RC were divided into a training group (n = 58) and a validation group (n = 25). Images were acquired from a 3.0 Tesla machine, including oblique axial T2 weighted imaging (T2WI) and IVIM with 11 b values. The ADC, D, D* and f values were measured on IVIM maps. The ROIs of tumors were delineated on T2WI, DWI, ADCmap , and Dmap images, and three mapping methods were used: ROIs_mapping from DWI, ROIs_mapping from ADCmap , and ROIs_mapping from Dmap . Three-dimensional radiomics features were extracted from the delineated ROIs. Multivariate logistic regression was used for radiomics feature selection. Radiomics models based on different mapping methods were developed. Receiver operating characteristic (ROC) curves, calibration, and decision curve analyses (DCA) were used to evaluate the performance of the models. RESULTS: Model B, which was constructed with radiomics features from ADCmap , Dmap and fmap by "ROIs_mapping from DWI" and T2WI (AUC 0.894), performed better than other models based on single sequence (AUC 0.600-0.806) and even better than Model A, which was based on "ROIs_mapping from ADC" and T2WI (AUC 0.838). Furthermore, an integrated model was constructed with Model B and the IVIM parameter (f value) with an AUC of 0.920 (95% CI: 0.820-1.000), which was higher than that of Model B, in the validation group. CONCLUSIONS: The integrated model incorporating the radiomics features and IVIM parameters accurately detected LVI of RC. The "ROIs_mapping from DWI" method provided the best results.


Assuntos
Radiômica , Neoplasias Retais , Humanos , Imageamento por Ressonância Magnética/métodos , Neoplasias Retais/diagnóstico por imagem , Neoplasias Retais/cirurgia , Neoplasias Retais/patologia , Curva ROC , Movimento (Física) , Imagem de Difusão por Ressonância Magnética/métodos , Estudos Retrospectivos
4.
Gut ; 72(11): 2149-2163, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37549980

RESUMO

OBJECTIVE: Selecting interventions for patients with solitary hepatocellular carcinoma (HCC) remains a challenge. Despite gross classification being proposed as a potential prognostic predictor, its widespread use has been restricted due to inadequate studies with sufficient patient numbers and the lack of established mechanisms. We sought to investigate the prognostic impacts on patients with HCC of different gross subtypes and assess their corresponding molecular landscapes. DESIGN: A prospective cohort of 400 patients who underwent hepatic resection for solitary HCC was reviewed and analysed and gross classification was assessed. Multiomics analyses were performed on tumours and non-tumour tissues from 49 patients to investigate the mechanisms underlying gross classification. Inverse probability of treatment weight (IPTW) was used to control for confounding factors. RESULTS: Overall 3-year survival rates varied significantly among the four gross subtypes (type I: 91%, type II: 80%, type III: 74.6%, type IV: 38.8%). Type IV was found to be independently associated with poor prognosis in both the entire cohort and the IPTW cohort. The four gross subtypes exhibited three distinct transcriptional modules. Particularly, type IV tumours exhibited increased angiogenesis and immune score as well as decreased metabolic pathways, together with highest frequency of TP53 mutations. Patients with type IV HCC may benefit from adjuvant intra-arterial therapy other than the other three subtypes. Accordingly, a modified trichotomous margin morphological gross classification was established. CONCLUSION: Different gross types of HCC showed significantly different prognosis and molecular characteristics. Gross classification may aid in development of precise individualised diagnosis and treatment strategies for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Estudos Prospectivos , Multiômica , Prognóstico
5.
Eur Radiol ; 33(12): 8936-8947, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37368104

RESUMO

OBJECTIVES: To evaluate the performance of a radiomics nomogram developed based on gadolinium-ethoxybenzyl-diethylenetriamine penta-acetic acid (Gd-EOB-DTPA) MRI for preoperative prediction of microvascular invasion (MVI) of hepatocellular carcinoma (HCC), and to identify patients who may benefit from the postoperative adjuvant transarterial chemoembolization (PA-TACE). METHODS: A total of 260 eligible patients were retrospectively enrolled from three hospitals (140, 65, and 55 in training, standardized external, and non-standardized external validation cohort). Radiomics features and image characteristics were extracted from Gd-EOB-DTPA MRI image before hepatectomy for each lesion. In the training cohort, a radiomics nomogram which incorporated the radiomics signature and radiological predictors was developed. The performance of the radiomics nomogram was assessed with respect to discrimination calibration, and clinical usefulness with external validation. A score (m-score) was constructed to stratify the patients and explored whether it could accurately predict patient who benefit from PA-TACE. RESULTS: A radiomics nomogram integrated with the radiomics signature, max-D(iameter) > 5.1 cm, peritumoral low intensity (PTLI), incomplete capsule, and irregular morphology had favorable discrimination in the training cohort (AUC = 0.982), the standardized external validation cohort (AUC = 0.969), and the non-standardized external validation cohort (AUC = 0.981). Decision curve analysis confirmed the clinical usefulness of the novel radiomics nomogram. The log-rank test revealed that PA-TACE significantly decreased the early recurrence in the high-risk group (p = 0.006) with no significant effect in the low-risk group (p = 0.270). CONCLUSIONS: The novel radiomics nomogram combining the radiomics signature and clinical radiological features achieved preoperative non-invasive MVI risk prediction and patient benefit assessment after PA-TACE, which may help clinicians implement more appropriate interventions. CLINICAL RELEVANCE STATEMENT: Our radiomics nomogram could represent a novel biomarker to identify patients who may benefit from the postoperative adjuvant transarterial chemoembolization, which may help clinicians to implement more appropriate interventions and perform individualized precision therapies. KEY POINTS: • The novel radiomics nomogram developed based on Gd-EOB-DTPA MRI achieved preoperative non-invasive MVI risk prediction. • An m-score based on the radiomics nomogram could stratify HCC patients and further identify individuals who may benefit from the PA-TACE. • The radiomics nomogram could help clinicians to implement more appropriate interventions and perform individualized precision therapies.


Assuntos
Carcinoma Hepatocelular , Quimioembolização Terapêutica , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/irrigação sanguínea , Nomogramas , Estudos Retrospectivos
6.
Mikrochim Acta ; 190(5): 181, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37046118

RESUMO

A simple fluorescence resonance energy transfer (FRET) sensing platform (termed as USP), comprised of upconversion nanoparticles (UCNPs) as the energy donor and Cy5 as the energy acceptor, has been synthesized for cathepsin B (CTSB) activity detection in vitro and in vivo. When Cy5-modified peptide substrate (peptide-Cy5) of CTSB is covalently linked on the surface of UCNPs, the FRET between the UCNPs (excitation: 980 nm; emission: 541 nm/655 nm) and Cy5 (excitation: 645 nm) leads to a reduction in the red upconversion luminescence (UCL) signal intensity of UCNPs. Cy5 can be liberated from UCNPs in the presence of CTSB through the cleavage of peptide-Cy5 by CTSB, leading to the recovery of the red UCL signal of UCNPs. Because the green UCL signal of UCNPs remains constant during the CTSB digestion, it can be considered as an internal reference. The findings demonstrate the ability of USP to detect CTSB with the linear detection ranges of 1 to 100 ng·mL-1 in buffer and 2 × 103 to 1 × 105 cells in 0.2 mL cell lysates. The limits of detection (LODs) are 0.30 ng·mL-1 in buffer and 887 cells in 0.2 mL of cell lysates (S/N = 3). The viability of USP to detect CTSB activity in tumor-bearing mice is has further been investigated using in vivo fluorescent imaging.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Nanopartículas , Animais , Camundongos , Catepsina B , Transferência Ressonante de Energia de Fluorescência/métodos , Peptídeos
7.
J Magn Reson Imaging ; 57(1): 45-56, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35993550

RESUMO

Rectal cancer (RC) accounts for approximately one-third of colorectal cancer (CRC), with death rates increasing in patients younger than 50 years old. Magnetic resonance imaging (MRI) is routinely performed for tumor evaluation. However, the semantic features from images alone remain insufficient to guide treatment decisions. Functional MRIs are useful for revealing microstructural and functional abnormalities and nevertheless have low or modest repeatability and reproducibility. Therefore, during the preoperative evaluation and follow-up treatment of patients with RC, novel noninvasive imaging markers are needed to describe tumor characteristics to guide treatment strategies and achieve individualized diagnosis and treatment. In recent years, the development of artificial intelligence (AI) has created new tools for RC evaluation based on MRI. In this review, we summarize the research progress of AI in the evaluation of staging, prediction of high-risk factors, genotyping, response to therapy, recurrence, metastasis, prognosis, and segmentation with RC. We further discuss the challenges of clinical application, including improvement in imaging, model performance, and the biological meaning of features, which may also be major development directions in the future. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 2.


Assuntos
Inteligência Artificial , Neoplasias Retais , Humanos , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Neoplasias Retais/patologia , Imageamento por Ressonância Magnética/métodos , Prognóstico
8.
ACS Omega ; 7(34): 30405-30411, 2022 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-36061664

RESUMO

The efficient and specific capture of circulating tumor cells (CTCs) from patients' peripheral blood is of significant value in precise cancer diagnosis and cancer therapy. As fine targeting molecules, lectins can recognize cancer cells specifically due to the abnormal glycosylation of molecules on the cancer cell membrane and the specific binding of lectin with glycoconjugates. Herein, a Ulex europaeus agglutinin-I (UEA-I)-based magnetic isolation strategy was developed to efficiently and specifically capture α-1,2-fucose overexpression CTCs from colorectal cancer (CRC) patients' peripheral blood. Using UEA-I-modified Fe3O4 magnetic beads (termed MB-UEA-I), up to 94 and 89% of target cells (i.e., SW480 CRC cells) were captured from the cell spiking complete cell culture medium and whole blood, respectively. More than 90% of captured cells show good viability and proliferation ability without detaching from MB-UEA-I. In combination with three-color immunocytochemistry (ICC) identification, MB-UEA-I has been successfully used to capture CTCs from CRC patients' peripheral blood. The experimental results indicate a correlation between CTC characterization and tumor metastasis. Specifically, MB-UEA-I can be applied to screen early CRC by capturing CTCs when served as a liquid biopsy. The presented work offers a new insight into developing cost-effective lectin-functionalized methods for biomedical applications.

9.
Hepatol Int ; 16(5): 1035-1051, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35829866

RESUMO

OBJECTIVE: To investigate the clinical, laboratory and genetic features of NAFLD patients based on MRI-PDFF in China. DESIGN: Patients with high ALT and with a diagnosis of fatty liver were included in this cross-sectional study. Fasting blood was collected to test biomarkers and SNPs. A total of 266 patients underwent MRI-PDFF and FibroScan examinations, and 38 underwent liver biopsy. Diagnostic models (decision tree, LASSO, and elastic net) were developed based on the diagnosis from MRI-PDFF reports. RESULTS: Approximately, 1/3 of the patients were found to have NASH and fibrosis. After quantifying liver steatosis by MRI-PDFF (healthy: n = 47; mild NAFLD: n = 136; moderate/severe NAFLD: n = 83; liver fat content (LFC): 3.6% vs. 8.7% vs. 19.0%), most biomarkers showed significant differences among the three groups, and patients without obesity were found to have a similar LFC as those with obesity (11.1% vs. 12.3%). Models including biomarkers showed strong diagnostic ability (accuracy: 0.80-0.91). Variant alleles of PNPLA3, HSD17B13 and MBOAT7 were identified as genetic risk factors causing higher LFC (8.7% vs. 12.3%; 11.0% vs. 14.5%; 8.5% vs. 10.2%, p < 0.05); those with the UQCC1 rs878639 variant allele showed lower LFC (10.4% vs. 8.4%; OR = 0.58, p < 0.05). Patients with more risk alleles had higher LFCs (8.1% vs. 10.7% vs. 11.6% vs. 14.5%). CONCLUSIONS: Based on MRI-PDFF, a combination of several specific biomarkers can accurately predict disease status. When the effects of genes on liver steatosis were first quantified by MRI-PDFF, the UQCC1 rs878639 G allele was identified as a protective factor, and the MBOAT7 T allele was identified as a risk only among nonobese individuals.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Biomarcadores , Estudos Transversais , Humanos , Fígado/diagnóstico por imagem , Fígado/patologia , Imageamento por Ressonância Magnética , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade
10.
Quant Imaging Med Surg ; 12(3): 1775-1786, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35284270

RESUMO

Background: Magnetic resonance (MR) images generated by different scanners generally have inconsistent contrast properties, making it difficult to perform a combined quantitative analysis of images from a range of scanners. In this study, we aimed to develop an automatic brain image segmentation model to provide a more reliable analysis of MR images taken with different scanners. Methods: The spatially localized atlas network tiles-27 (SLANT-27) deep learning model was used to train the automatic segmentation module, based on a multi-center dataset of 1,917 three-dimensional (3D) T1-weighted MR images. Subsequently, a framework called Qbrain, consisting of a new generative adversarial network (GAN) image transfer module and the SLANT-27 segmentation module, was developed. Another 3D T1-weighted MRI interscan dataset of 48 participants who were scanned in 3 MRI scanners (1.5T Siemens Avanto, 3T Siemens Trio Tim, and 3T Philips Ingenia) on the same day was used to train and test the Qbrain model. Volumetric T1-weighted images were processed with Qbrain, SLANT-27, and FreeSurfer (FS). The automatic segmentation reliability across the scanners was assessed using test-retest variability (TRV). Results: The reproducibility of different segmentation methods across scanners showed a consistent trend in the greater reliability and robustness of QBrain compared to SLANT-27 which, in turn, showed greater reliability and robustness compared to FS. Furthermore, when the GAN image transfer module was added, the mean segmentation error of the TRV of the 3T Siemens vs. 1.5T Siemens, the 3T Philips vs. 1.5T Siemens, and the 3T Siemens vs. 3T Philips scanners was reduced by 1.57%, 2.01%, and 0.56%, respectively. In addition, the segmentation model improved intra-scanner variability (0.9-1.67%) compared with that of FS (2.47-4.32%). Conclusions: The newly developed QBrain method combined with GAN image transfer module and a SLANT-27 segmentation module was shown to improve the reliability of whole-brain automatic structural segmentation results across multiple scanners, thus representing a suitable alternative quantitative method of comparative brain tissue analysis for individual patients.

12.
Front Oncol ; 11: 669437, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34336661

RESUMO

OBJECTIVE: Liver cancer is one of the most commonly diagnosed cancer, and energy-based tumor ablation is a widely accepted treatment. Automatic and robust segmentation of liver tumors and ablation zones would facilitate the evaluation of treatment success. The purpose of this study was to develop and evaluate an automatic deep learning based method for (1) segmentation of liver and liver tumors in both arterial and portal venous phase for pre-treatment CT, and (2) segmentation of liver and ablation zones in both arterial and portal venous phase for after ablation treatment. MATERIALS AND METHODS: 252 CT images from 63 patients undergoing liver tumor ablation at a large University Hospital were retrospectively included; each patient had pre-treatment and post-treatment multi-phase CT images. 3D voxel-wise manual segmentation of the liver, tumors and ablation region by the radiologist provided reference standard. Deep learning models for liver and lesion segmentation were initially trained on the public Liver Tumor Segmentation Challenge (LiTS) dataset to obtain base models. Then, transfer learning was applied to adapt the base models on the clinical training-set, to obtain tumor and ablation segmentation models both for arterial and portal venous phase images. For modeling, 2D residual-attention Unet (RA-Unet) was employed for liver segmentation and a multi-scale patch-based 3D RA-Unet for tumor and ablation segmentation. RESULTS: On the independent test-set, the proposed method achieved a dice similarity coefficient (DSC) of 0.96 and 0.95 for liver segmentation on arterial and portal venous phase, respectively. For liver tumors, the model on arterial phase achieved detection sensitivity of 71%, DSC of 0.64, and on portal venous phase sensitivity of 82%, DSC of 0.73. For liver tumors >0.5cm3 performance improved to sensitivity 79%, DSC 0.65 on arterial phase and, sensitivity 86%, DSC 0.72 on portal venous phase. For ablation zone, the model on arterial phase achieved detection sensitivity of 90%, DSC of 0.83, and on portal venous phase sensitivity of 90%, DSC of 0.89. CONCLUSION: The proposed deep learning approach can provide automated segmentation of liver tumors and ablation zones on multi-phase (arterial and portal venous) and multi-time-point (before and after treatment) CT enabling quantitative evaluation of treatment success.

13.
Sci Rep ; 11(1): 9429, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33941817

RESUMO

Perineural invasion (PNI) as a grossly underreported independent risk predictor in rectal cancer is hard to identify preoperatively. We aim to predict PNI status in rectal cancer using multi-modality radiomics. In total, 396 radiomics features were extracted from T2-weighted images (T2WIs), diffusion-weighted images (DWIs), and portal venous phase of contrast-enhanced CT (CE-CT) respectively of 94 consecutive patients with histologically confirmed rectal cancer. T2WI score, DWI score, and CT score were calculated via the radiomics features selection and optimization. Discrimination, calibration, and clinical benefit ability were used to evaluate the performance of the radiomics scores in both training and testing datasets. CT score and T2WI score were independent risk predictors [CT score, OR (95% CI) = 4.218 (1.070-16.620); T2WI score, OR (95% CI) = 105.721 (3.091-3615.790)]. The concise score which combined CT score and T2WI score, showed the best performance [training dataset, AUC (95% CI) = 0.906 (0.833-0.979); testing dataset, AUC (95% CI) = 0.884 (0.761-1.000)] and good calibration (P > 0.05 in the Hosmer-Lemeshow test for the training and testing datasets). Decision curve analysis showed that the multi-modality radiomics nomogram had a higher clinical net benefit. The multi-modality radiomics score could be used to preoperatively assess PNI status in rectal cancer.


Assuntos
Nervos Periféricos/patologia , Neoplasias Retais/diagnóstico , Neoplasias Retais/patologia , Região Sacrococcígea/inervação , Região Sacrococcígea/patologia , Idoso , Biologia Computacional , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Nomogramas , Radiometria/métodos , Neoplasias Retais/terapia , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
14.
Eur Radiol ; 31(8): 6030-6038, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33560457

RESUMO

OBJECTIVES: To develop and validate a PET/CT nomogram for preoperative estimation of lymph node (LN) staging in patients with non-small cell lung cancer (NSCLC). METHODS: A total of 263 pathologically confirmed LNs from 124 patients with NCSLC were retrospectively analyzed. Positron-emission tomography/computed tomography (PET/CT) examination was performed before treatment according to the clinical schedule. In the training cohort (N = 185), malignancy-related features, such as SUVmax, short-axis diameter (SAD), and CT radiomics features, were extracted from the regions of LN based on the PET/CT scan. The Minimum-Redundancy Maximum-Relevance (mRMR) algorithm and the Least Absolute Shrinkage and Selection Operator (LASSO) regression model were used for feature selection and radiomics score building. The radiomics score (Rad-Score) and SUVmax were incorporated in a PET/CT nomogram using the multivariable logistic regression analysis. The performance of the proposed model was evaluated with discrimination, calibration, and clinical application in an independent testing cohort (N = 78). RESULTS: The radiomics scores consisting of 14 selected features were significantly associated with LN status for both training cohort with AUC of 0.849 (95% confidence interval (CI), 0.796-0.903) and testing cohort with AUC of 0.828 (95% CI, 0.782-0.919). The PET/CT nomogram incorporating radiomics score and SUVmax showed moderate improvement of the efficiency with AUC of 0.881 (95% CI, 0.834-0.928) in the training cohort and AUC of 0.872 (95% CI, 0.797-0.946) in the testing cohort. The decision curve analysis indicated that the PET/CT nomogram was clinically useful. CONCLUSION: The PET/CT nomogram, which incorporates Rad-Score and SUVmax, can improve the diagnostic performance of LN metastasis. KEY POINTS: • The PET/CT nomogram (Int-Score) based on lymph node (LN) PET/CT images can reliably predict LN status in NSCLC. • Int-Score is a relatively objective diagnostic method, which can play an auxiliary role in the process of clinicians making treatment decisions.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Nomogramas , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
15.
RSC Adv ; 11(4): 2213-2220, 2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-35424166

RESUMO

Herein, a fluorescence turn-on nanosensor (MnIO@pep-FITC) has been proposed for detecting trypsin activity in vitro and in vivo through covalently immobilizing an FITC modified peptide substrate of trypsin (pep-FITC) on manganese-doped iron oxide nanoparticle (MnIO NP) surfaces via a polyethylene glycol (PEG) crosslinker. The conjugation of pep-FITC with MnIO NPs results in the quenching of FITC fluorescence. After trypsin cleavage, the FITC moiety is released from the MnIO NP surface, leading to a remarkable recovery of FITC fluorescence signal. Under the optimum experimental conditions, the recovery ratio of FITC fluorescence intensity is linearly dependent on the trypsin concentration in the range of 2 to 100 ng mL-1 in buffer and intracellular trypsin in the lysate of 5 × 102 to 1 × 104 HCT116 cells per mL, respectively. The detection limit of trypsin is 0.6 ng mL-1 in buffer or 359 cells per mL HCT116 cell lysate. The MnIO@pep-FITC is successfully employed to noninvasively monitor trypsin activity in the ultrasmall (ca. 4.9 mm3 in volume) BALB/c nude mouse-bearing HCT116 tumor by in vivo fluorescence imaging with external magnetic field assistance, demonstrating that it has excellent practicability.

16.
Transl Oncol ; 14(1): 100936, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33221688

RESUMO

In this study, we aimed to establish a radiomics nomogram that noninvasively evaluates the invasiveness of pulmonary adenocarcinomas manifesting as ground-glass nodules (GGNs). Computed tomography (CT) images of 509 patients manifesting as GGNs were collected: 70% of cases were included in the training cohort and 30% in the validation cohort. The Max-Relevance and Min-Redundancy (mRMR) and the least absolute shrinkage and selection operator (LASSO) algorithm were used to select the radiomics features and construct a radiomics signature. Univariate and multivariate logistic regression were used to select the invasiveness-related clinical and CT morphological predictors. Age, smoking history, long diameter, and average CT value were retained as independent predictors of GGN invasiveness. A radiomics nomogram was established by integrating clinical and CT morphological features with the radiomics signature. The radiomics nomogram showed good predictive ability in the training set (area under the curve [AUC], 0.940; 95% confidence interval [CI], 0.916-0.964) and validation set (AUC, 0.946; 95% CI, 0.907-0.986). This radiomics nomogram may serve as a noninvasive and accurate predictive tool to determine the invasiveness of GGNs prior to surgery and assist clinicians in creating personalized treatment strategies.

17.
BMC Med Imaging ; 20(1): 59, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32487083

RESUMO

BACKGROUND: The detection of Kirsten rat sarcoma viral oncogene homolog (KRAS) gene mutations in colorectal cancer (CRC) is key to the optimal design of individualized therapeutic strategies. The noninvasive prediction of the KRAS status in CRC is challenging. Deep learning (DL) in medical imaging has shown its high performance in diagnosis, classification, and prediction in recent years. In this paper, we investigated predictive performance by using a DL method with a residual neural network (ResNet) to estimate the KRAS mutation status in CRC patients based on pre-treatment contrast-enhanced CT imaging. METHODS: We have collected a dataset consisting of 157 patients with pathology-confirmed CRC who were divided into a training cohort (n = 117) and a testing cohort (n = 40). We developed an ResNet model that used portal venous phase CT images to estimate KRAS mutations in the axial, coronal, and sagittal directions of the training cohort and evaluated the model in the testing cohort. Several groups of expended region of interest (ROI) patches were generated for the ResNet model, to explore whether tissues around the tumor can contribute to cancer assessment. We also explored a radiomics model with the random forest classifier (RFC) to predict KRAS mutations and compared it with the DL model. RESULTS: The ResNet model in the axial direction achieved the higher area under the curve (AUC) value (0.90) in the testing cohort and peaked at 0.93 with an input of 'ROI and 20-pixel' surrounding area. AUC of radiomics model in testing cohorts were 0.818. In comparison, the ResNet model showed better predictive ability. CONCLUSIONS: Our experiments reveal that the computerized assessment of the pre-treatment CT images of CRC patients using a DL model has the potential to precisely predict KRAS mutations. This new model has the potential to assist in noninvasive KRAS mutation estimation.


Assuntos
Neoplasias Colorretais/diagnóstico por imagem , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Análise de Sequência de DNA/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/genética , Aprendizado Profundo , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Tomografia Computadorizada por Raios X
18.
Front Oncol ; 10: 457, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32328460

RESUMO

Objective: To explore a new predictive model of lymphatic vascular infiltration (LVI) in rectal cancer based on magnetic resonance (MR) and computed tomography (CT). Methods: A retrospective study was conducted on 94 patients with histologically confirmed rectal cancer, they were randomly divided into training cohort (n = 65) and validation cohort (n = 29). All patients underwent MR and CT examination within 2 weeks before treatment. On each slice of the tumor, we delineated the volume of interest on T2-weighted imaging, diffusion weighted imaging, and enhanced CT images, respectively. A total of 1,188 radiological features were extracted from each patient. Then, we used the student t-test or Mann-Whitney U-test, Spearman's rank correlation and least absolute shrinkage and selection operator (LASSO) algorithm to select the strongest features to establish a single and multimodal logic model for predicting LVI. Receiver operating characteristic (ROC) curves and calibration curves were plotted to determine how well they explored LVI prediction performance in the training and validation cohorts. Results: An optimal multi-mode radiology nomogram for LVI estimation was established, which had significant predictive power in training (AUC, 0.884; 95% CI, 0.803-0.964) and validation (AUC, 0.876; 95% CI, 0.721-1.000). Calibration curve and decision curve analysis showed that the multimodal radiomics model provides greater clinical benefits. Conclusion: Multimodal (MR/CT) radiomics models can serve as an effective visual prognostic tool for predicting LVI in rectal cancer. It demonstrated great potential of preoperative prediction to improve treatment decisions.

19.
Quant Imaging Med Surg ; 10(2): 397-414, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32190566

RESUMO

BACKGROUND: This article aims to develop and assess the radiomics paradigm for predicting colorectal cancer liver metastasis (CRLM) from the primary tumor. METHODS: This retrospective study included 100 patients from the First Hospital of Jilin University from June 2017 to December 2017. The 100 patients comprised 50 patients with and 50 without CRLM. The maximum-level enhanced computed tomography (CT) image of primary cancer in the portal venous phase of each patient was selected as the original image data. To automatically implement radiomics-related paradigms, we developed a toolkit called Radiomics Intelligent Analysis Toolkit (RIAT). RESULTS: With RIAT, the model based on logistic regression (LR) using both the radiomics and clinical information signatures showed the maximum net benefit. The area under the curve (AUC) value was 0.90±0.02 (sensitivity =0.85±0.02, specificity =0.79±0.04) for the training set, 0.86±0.11 (sensitivity =0.85±0.09, specificity =0.75±0.19) for the verification set, 0.906 (95% CI, 0.840-0.971; sensitivity =0.81, specificity =0.84) for the cross-validation set, and 0.899 (95% CI, 0.761-1.000; sensitivity =0.78, specificity =0.91) for the test set. CONCLUSIONS: The radiomics nomogram-based LR with clinical risk and radiomics features allows for a more accurate classification of CRLM using CT images with RIAT.

20.
Phys Med Biol ; 65(5): 055012, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31978901

RESUMO

To predict the epidermal growth factor receptor (EGFR) mutation status in patients with lung adenocarcinoma using quantitative radiomic biomarkers and semantic features. We analyzed the computed tomography (CT) images and medical record data of 104 patients with lung adenocarcinoma who underwent surgical excision and EGFR mutation detection from 2016 to 2018 at our center. CT radiomic and semantic features that reflect the tumors' heterogeneity and phenotype were extracted from preoperative non-enhanced CT scans. The least absolute shrinkage and selection operator method was applied to select the most distinguishable features. Three logistic regression models were built to predict the EGFR mutation status by combining the CT semantic with clinicopathological characteristics, using the radiomic features alone, and by combining the radiomic and clinicopathological features. Receiver operating characteristic (ROC) curve analysis was performed using five-fold cross-validation and the mean area under the curve (AUC) values were calculated and compared between the models to obtain the optimal model for predicting EGFR mutation. Furthermore, radiomic nomograms were constructed to demonstrate the performance of the model. In total, 1025 radiomic features were extracted and reduced to 13 features as the most important predictors to build the radiomic signature. The combined radiomic and clinicopathological features model was developed based on the radiomic signature, sex, smoking, vascular infiltration, and pathohistological type. The AUC was 0.90 ± 0.02 for the training, 0.88 ± 0.11 for the verification, and 0.894 for the test dataset. This model was superior to the other prediction models that used the combined CT semantic and clinicopathological features (AUC for the test dataset: 0.768) and radiomic features alone (AUC for the test dataset: 0.837). The prediction model built by radiomic biomarkers and clinicopathological features, including the radiomic signature, sex, smoking, vascular infiltration, and pathological type, outperformed the other two models and could effectively predict the EGFR mutation status in patients with peripheral lung adenocarcinoma. The radiomic nomogram of this model is expected to become an effective biomarker for patients with lung adenocarcinoma requiring adjuvant targeted treatment.


Assuntos
Adenocarcinoma de Pulmão/genética , Carcinoma Papilar/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Mutação , Nomogramas , Tomografia Computadorizada por Raios X/métodos , Adenocarcinoma de Pulmão/diagnóstico por imagem , Adenocarcinoma de Pulmão/patologia , Área Sob a Curva , Carcinoma Papilar/diagnóstico por imagem , Carcinoma Papilar/patologia , Feminino , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Curva ROC , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA