Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Med Imaging ; 24(1): 141, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862884

RESUMEN

OBJECTIVE: To evaluate the consistency between doctors and artificial intelligence (AI) software in analysing and diagnosing pulmonary nodules, and assess whether the characteristics of pulmonary nodules derived from the two methods are consistent for the interpretation of carcinomatous nodules. MATERIALS AND METHODS: This retrospective study analysed participants aged 40-74 in the local area from 2011 to 2013. Pulmonary nodules were examined radiologically using a low-dose chest CT scan, evaluated by an expert panel of doctors in radiology, oncology, and thoracic departments, as well as a computer-aided diagnostic(CAD) system based on the three-dimensional(3D) convolutional neural network (CNN) with DenseNet architecture(InferRead CT Lung, IRCL). Consistency tests were employed to assess the uniformity of the radiological characteristics of the pulmonary nodules. The receiver operating characteristic (ROC) curve was used to evaluate the diagnostic accuracy. Logistic regression analysis is utilized to determine whether the two methods yield the same predictive factors for cancerous nodules. RESULTS: A total of 570 subjects were included in this retrospective study. The AI software demonstrated high consistency with the panel's evaluation in determining the position and diameter of the pulmonary nodules (kappa = 0.883, concordance correlation coefficient (CCC) = 0.809, p = 0.000). The comparison of the solid nodules' attenuation characteristics also showed acceptable consistency (kappa = 0.503). In patients diagnosed with lung cancer, the area under the curve (AUC) for the panel and AI were 0.873 (95%CI: 0.829-0.909) and 0.921 (95%CI: 0.884-0.949), respectively. However, there was no significant difference (p = 0.0950). The maximum diameter, solid nodules, subsolid nodules were the crucial factors for interpreting carcinomatous nodules in the analysis of expert panel and IRCL pulmonary nodule characteristics. CONCLUSION: AI software can assist doctors in diagnosing nodules and is consistent with doctors' evaluations and diagnosis of pulmonary nodules.


Asunto(s)
Inteligencia Artificial , Diagnóstico por Computador , Neoplasias Pulmonares , Tomografía Computarizada por Rayos X , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Estudios Retrospectivos , Persona de Mediana Edad , Masculino , Anciano , Femenino , Adulto , Diagnóstico por Computador/métodos , Tomografía Computarizada por Rayos X/métodos , Detección Precoz del Cáncer/métodos , Curva ROC , Redes Neurales de la Computación , Interpretación de Imagen Radiográfica Asistida por Computador/métodos , Programas Informáticos
2.
Eur J Radiol Open ; 12: 100548, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38298532

RESUMEN

Background: Kirsten rat sarcoma virus (KRAS) has evolved from a genotype with predictive value to a therapeutic target recently. The study aimed to establish non-invasive radiomics models based on MRI to discriminate KRAS from epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) mutations in lung cancer patients with brain metastases (BM), then further explore the optimal sequence for prediction. Methods: This retrospective study involved 317 patients (218 patients in training cohort and 99 patients in testing cohort) who had confirmed of KRAS, EGFR or ALK mutations. Radiomics features were separately extracted from T2WI, T2 fluid-attenuated inversion recovery (T2-FLAIR), diffusion weighted imaging (DWI) and contrast-enhanced T1-weighted imaging (T1-CE) sequences. The maximal information coefficient and recursive feature elimination method were used to select informative features. Then we built four radiomics models for differentiating KRAS from EGFR or ALK using random forest classifier. ROC curves were used to validate the capability of the models. Results: The four radiomics models for discriminating KRAS from EGFR all worked well, especially DWI and T2WI models (AUCs: 0.942, 0.942 in training cohort, 0.949, 0.954 in testing cohort). When KRAS compared to ALK, DWI and T2-FLAIR models showed excellent performance in two cohorts (AUCs: 0.947, 0.917 in training cohort, 0.850, 0.824 in testing cohort). Conclusions: Radiomics classifiers integrating MRI have potential to discriminate KRAS from EGFR or ALK, which are helpful to guide treatment and facilitate the discovery of new approaches capable of achieving this long-sought goal of cure in lung cancer patients with KRAS.

3.
Eur Radiol Exp ; 8(1): 2, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38169047

RESUMEN

BACKGROUND: To establish a predictive model based on multisequence magnetic resonance imaging (MRI) using deep learning to identify wild-type (WT) epidermal growth factor receptor (EGFR), EGFR exon 19 deletion (19Del), and EGFR exon 21-point mutation (21L858R) simultaneously. METHODS: A total of 399 patients with proven brain metastases of non-small cell lung cancer (NSCLC) were retrospectively enrolled and divided into training (n = 306) and testing (n = 93) cohorts separately based on two timepoints. All patients underwent 3.0-T brain MRI including T2-weighted, T2-weighted fluid-attenuated inversion recovery, diffusion-weighted imaging, and contrast-enhanced T1-weighted sequences. Radiomics features were extracted from each lesion based on four sequences. An algorithm combining radiomics approach with graph convolutional networks architecture (Radio-GCN) was designed for the prediction of EGFR mutation status and subtype. The area under the curve (AUC) at receiver operating characteristic analysis was used to evaluate the predication capabilities of each model. RESULTS: We extracted 1,290 radiomics features from each MRI sequence. The AUCs of the Radio-GCN model for identifying EGFR 19Del, 21L858R, and WT for the lesion-wise analysis were 0.996 ± 0.004, 0.971 ± 0.013, and 1.000 ± 0.000 on the independent testing cohort separately. It also yielded AUCs of 1.000 ± 0.000, 0.991 ± 0.009, and 1.000 ± 0.000 for predicting EGFR mutations respectively for the patient-wise analysis. The κ coefficients were 0.735 and 0.812, respectively. CONCLUSIONS: The constructed Radio-GCN model is a new potential tool to predict the EGFR mutation status and subtype in NSCLC patients with brain metastases. RELEVANCE STATEMENT: The study demonstrated that a deep learning approach based on multisequence MRI can help to predict the EGFR mutation status in NSCLC patients with brain metastases, which is beneficial to guide a personalized treatment. KEY POINTS: • This is the first study to predict the EGFR mutation subtype simultaneously. • The Radio-GCN model holds the potential to be used as a diagnostic tool. • This study provides an imaging surrogate for identifying the EGFR mutation subtype.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Aprendizaje Profundo , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/genética , Estudios Retrospectivos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/genética , Imagen por Resonancia Magnética , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Receptores ErbB/genética , Mutación
4.
Quant Imaging Med Surg ; 13(12): 8599-8610, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38106277

RESUMEN

Background: Predicting whether T790M emerges early is crucial to the adjustment of targeted drugs for non-small cell lung cancer (NSCLC) patients. This study aimed to evaluate the risk of T790M resistance in progressive new brain metastases (BMs) based on multisequence magnetic resonance imaging (MRI) radiomics. Methods: This retrospective study included 405 consecutive patients (training cohort: 294 patients; testing cohort: 111 patients) with proven NSCLC with disease progression of new BM. The radiomics features were separately extracted from T2-weighted imaging (T2WI), T2 fluid-attenuated inversion recovery (T2-FLAIR), diffusion-weighted imaging (DWI), and contrast-enhanced T1-weighted imaging (T1-CE) sequence of baseline MRI. Then, we calculated radiomics scores (rad-score) of the 4 sequences respectively and established predictive models (lesion- or patient-level) to evaluate T790M resistance within up to 14 months using random forest classifier. Receiver operating characteristic (ROC) curves and F1 scores were used to validate the performance of two models in both the training and testing cohort. Results: There were significant differences in rad-scores of the four sequences between T790M-positive and negative groups whether in the training or testing cohort (P<0.05). The lesion-level model consisting of rad-scores showed excellent discrimination, with an area under the curve (AUC) and F1-score of 0.879 and 0.798 in the training cohort, and 0.834 and 0.742 in the testing cohort, respectively. The patient-level model also showed a favorable discriminatory ability with an AUC and F1 score of 0.851 and 0.837, which was confirmed with an AUC and F1 score of 0.734 and 0.716 in the testing cohort. Conclusions: The MRI-based radiomics signatures may be new markers to identify patients at high risk of developing resistance in the early period.

5.
Infect Drug Resist ; 16: 6893-6904, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37920476

RESUMEN

Background: Sputum culture result at the sixth month is essential for predicting therapeutic response to longer multidrug-resistant tuberculosis (MDR-TB) regimens. This study aimed to construct a predictive model using cavity-based radiomics to predict sputum status at the sixth month for MDR-TB patients treated with longer regimens. Methods: This retrospective study recruited 315 MDR-TB patients treated with longer regimens from two centers (250 patients from center 1 and 65 patients from center 2), who were divided into persistently positive and conversion to negative sputum culture groups according to sputum results. Radiomics features were extracted based on the cavity, and a radiomics model was selected and established using a random forest classifier. The clinical characteristics and primary CT signs with significant differences were integrated to build a clinical model. A combined model was generated using the radiomics and clinical model. ROC curves, F1-score and DCA curves were used to assess the predictive performance of the models. Results: Twenty-eight radiomics features were selected to build a radiomics model for predicting the sputum status. The radiomics model achieved good performance, with AUCs of 0.892 and 0.839 in the training and testing cohort, respectively, which was similar to the performance of the combined model (0.913 and 0.815) and much higher than that of the clinical model (0.688 and 0.525) in the two cohorts. Conclusion: The cavity-based radiomics model has the potential to predict sputum culture status for MDR-TB patients receiving longer regimens, which could guide follow-up treatment effectively.

6.
Eur Radiol Exp ; 7(1): 64, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37914925

RESUMEN

BACKGROUND: To evaluate the value of computed tomography (CT) radiomics in predicting the risk of developing epidermal growth factor receptor (EGFR) T790M resistance mutation for metastatic non-small lung cancer (NSCLC) patients before first-line EGFR-tyrosine kinase inhibitors (EGFR-TKIs) therapy. METHODS: A total of 162 metastatic NSCLC patients were recruited and split into training and testing cohort. Radiomics features were extracted from tumor lesions on nonenhanced CT (NECT) and contrast-enhanced CT (CECT). Radiomics score (rad-score) of two CT scans was calculated respectively. A nomogram combining two CT scans was developed to evaluate T790M resistance within up to 14 months. Patients were followed up to calculate the time of T790M occurrence. Models were evaluated by area under the curve at receiver operating characteristic analysis (ROC-AUC), calibration curve, and decision curve analysis (DCA). The association of the nomogram with the time of T790M occurrence was evaluated by Kaplan-Meier survival analysis. RESULTS: The nomogram constructed with the rad-score of NECT and CECT for predicting T790M resistance within 14 months achieved the highest ROC-AUCs of 0.828 and 0.853 in training and testing cohorts, respectively. The DCA showed that the nomogram was clinically useful. The Kaplan-Meier analysis showed that the occurrence time of T790M difference between the high- and low-risk groups distinguished by the rad-score was significant (p < 0.001). CONCLUSIONS: The CT-based radiomics signature may provide prognostic information and improve pretreatment risk stratification in EGFR NSCLC patients before EGFR-TKIs therapy. The multimodal radiomics nomogram further improved the capability. RELEVANCE STATEMENT: Radiomics based on NECT and CECT images can effectively identify and stratify the risk of T790M resistance before the first-line TKIs treatment in metastatic non-small cell lung cancer patients. KEY POINTS: • Early identification of the risk of T790M resistance before TKIs treatment is clinically relevant. • Multimodel radiomics nomogram holds potential to be a diagnostic tool. • It provided an imaging surrogate for identifying the pretreatment risk of T790M.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Nomogramas , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/tratamiento farmacológico , Receptores ErbB/genética , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Tomografía Computarizada por Rayos X/métodos , Medición de Riesgo
7.
Eur J Radiol Open ; 11: 100521, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37692549

RESUMEN

Background: Osimertinib resistance is a major problem in the course of targeted therapy for non-small cell lung cancer (NSCLC) patients. To develop and validate a multisequence MRI-based radiomics nomogram for early prediction of osimertinib resistance in NSCLC with brain metastases (BM). Methods: Pretreatment brain MRI of 251 NSCLC patients proven with BM were retrospectively enrolled from two centers (training cohort: 196 patients; testing cohort: 55 patients). According to the gene test result of osimertinib resistance, patients were labeled as resistance and non-resistance groups (training cohort: 65 versus 131 patients; testing cohort: 25 versus 30 patients). Radiomics features were extracted from T2WI, T2 fluid-attenuated inversion recovery (T2-FLAIR), diffusion weighted imaging (DWI) and contrast-enhanced T1-weighted imaging (T1-CE) sequences separately and radiomics score (rad-score) were built from the four sequences. Then a multisequence MRI-based nomogram was developed and the predictive ability was evaluated by ROC curves and calibration curves. Results: The rad-scores of the four sequences has significant differences between resistance and non-resistance groups in both training and testing cohorts. The nomogram achieved the highest predictive ability with area under the curve (AUC) of 0.989 (95 % confidence interval, 0.976-1.000) and 0.923 (95 % confidence interval, 0.851-0.995) in the training and testing cohort respectively. The calibration curves showed excellent concordance between the predicted and actual probability of osimertinib resistance using the radiomics nomogram. Conclusions: The multisequence MRI-based radiomics nomogram can be used as a noninvasive auxiliary tool to identify candidates who were resistant to osimertinib, which could guide clinical therapy for NSCLC patients with BM.

8.
Eur Radiol ; 33(9): 6308-6317, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37004571

RESUMEN

OBJECTIVES: Multidrug-resistant TB (MDR-TB) is a severe burden and public health threat worldwide. This study aimed to develop a radiomics model based on the tree-in-bud (TIB) sign and nodules and validate its predictive performance for MDR-TB. METHODS: We retrospectively recruited 454 patients with proven active TB from two hospitals and classified them into three training and testing cohorts: TIB (n = 295, 102), nodules (n = 302, 97), and their combination (n = 261, 81). Radiomics features relating to TIB and nodules were separately extracted. The maximal information coefficient and recursive feature elimination were used to select informative features per the two signs. Two radiomics models were constructed to predict MDR-TB using a random forest classifier. Then, a combined model was built incorporating radiomics features based on these two signs. The capability of the models in the combined training and testing cohorts was validated with ROC curves. RESULTS: Sixteen features were extracted from TIB and 15 from nodules. The AUCs of the combined model were slightly higher than those of the TIB model in the combined training cohort (0.911 versus 0.877, p > 0.05) and testing cohort (0.820 versus 0.786, p < 0.05) and similar to the performance of the nodules model in the combined training cohort (0.911 versus 0.933, p > 0.05) and testing cohort (0.820 versus 0.855, p > 0.05). CONCLUSIONS: The CT-based radiomics models hold promise for use as a non-invasive tool in the prediction of MDR-TB. CLINICAL RELEVANCE STATEMENT: Our study revealed that complementary information regarding MDR-TB can be provided by radiomics based on the TIB sign and nodules. The proposed radiomics models may be new markers to predict MDR in active TB patients. KEY POINTS: • This is the first study to build, validate, and apply radiomics based on tree-in-bud sign and nodules for the prediction of MDR-TB. • The radiomics model showed a favorable performance for the identification of MDR-TB. • The combined model holds potential to be used as a diagnostic tool in routine clinical practice.


Asunto(s)
Tomografía Computarizada por Rayos X , Tuberculosis Resistente a Múltiples Medicamentos , Humanos , Estudios Retrospectivos , Tuberculosis Resistente a Múltiples Medicamentos/diagnóstico por imagen , Pulmón , Resistencia a Múltiples Medicamentos
9.
Acad Radiol ; 30(9): 1887-1895, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36586758

RESUMEN

RATIONALE AND OBJECTIVES: Timely identifying T790M mutation for non-small cell lung cancer (NSCLC) patients with brain metastases (BM) is essential to adjust targeted treatment strategies. To develop and validate radiomics models based on multisequence MRI for differentiating patients with T790M resistance from no T790M mutation in BM and explore the optimal sequence for prediction. MATERIALS AND METHODS: This retrospective study enrolled 233 patients with proven of BM in NSCLC which included 95 with T790M and 138 without T790M from two hospitals as the training cohort and testing cohort separately. Radiomics features extracted from T2WI, T2 fluid-attenuated inversion recovery (T2-FLAIR), diffusion weighted imaging (DWI) and contrast-enhanced T1-weighted imaging (T1-CE) sequence respectively. The most predictable features were selected based on the maximal information coefficient and Boruta method. Then four radiomics models were built to characterize T790M mutation by random forest classifier. ROC curves, F1 score and DCA curves were constructed to validate the capability and verify the performance of four models. RESULTS: The DWI model showed best performance with AUC and F1 score of 0.886 and 0.789 in the training cohort, 0.850 and 0.743 in the testing cohort. DCA curves also showed higher overall net benefit from the DWI model than from the remaining three models in the testing cohort. Other three models also had some classification power whether in the training or testing cohort, especially T2-FLAIR model. CONCLUSION: Multisequence MRI-based radiomics has potential to predict the emergence of EGFR T790M resistance mutations especially the radiomics signature based on DWI sequence.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Estudios Retrospectivos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/genética , Imagen por Resonancia Magnética/métodos , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Mutación , Receptores ErbB/genética
10.
Eur J Radiol ; 155: 110499, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36049410

RESUMEN

PURPOSE: More and more small brain metastases (BMs) in asymptomatic patients can be detected even prior to their primary lung cancer with the development of MRI. The aim of this study was to develop a predictive radiomics model to identify epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutation status in BM and explore the optimal MR sequence for predication. METHODS: This retrospective study included 186 patients with proven BM of lung cancer (training cohort: 70 patients with EGFR mutations and 65 patients with ALK rearrangements; testing cohort: 26 patients with EGFR mutations and 25 patients with ALK rearrangements). Radiomics features were separately extracted from contrast-enhanced T1-weighted imaging (T1-CE), T2 fluid-attenuated inversion recovery (T2-FLAIR) and T2WI sequences. The model for three MR sequences were constructed using a random forest classifier. ROC curves were used to validate the capability of the models in the training and testing cohorts. RESULTS: The AUCs of the T2-FLAIR model were significantly higher than those of the T1-CE model in training cohort (0.991 versus 0.954) and testing cohort (0.950 versus 0.867) and much higher than those of the T2WI model in training cohort (0.991 versus 0.880) and testing cohort (0.950 versus 0.731). Besides, the F1 scores of the T1-CE model were slightly higher than the T2-FLAIR model and much higher than the T2WI model in two cohorts. CONCLUSION: T2-FLAIR and T1-CE radiomics models that can be used as noninvasive tools for identifying EGFR and ALK mutation status are helpful to guide therapeutic strategies.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Pulmonares , Quinasa de Linfoma Anaplásico/genética , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/genética , Imagen por Resonancia Magnética/métodos , Mutación , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...