Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
BMC Cancer ; 24(1): 441, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38594604

RESUMO

BACKGROUND: We recently found that epiplakin 1 (EPPK1) alterations were present in 12% of lung adenocarcinoma (LUAD) cases and were associated with a poor prognosis in early-stage LUAD when combined with other molecular alterations. This study aimed to identify a probable crucial role for EPPK1 in cancer development. METHODS: EPPK1 mRNA and protein expression was analyzed with clinical variables. Normal bronchial epithelial cell lines were exposed to cigarette smoke for 16 weeks to determine whether EPPK1 protein expression was altered after exposure. Further, we used CRISPR-Cas9 to knock out (KO) EPPK1 in LUAD cell lines and observed how the cancer cells were altered functionally and genetically. RESULTS: EPPK1 protein expression was associated with smoking and poor prognosis in early-stage LUAD. Moreover, a consequential mesenchymal-to-epithelial transition was observed, subsequently resulting in diminished cell proliferation and invasion after EPPK1 KO. RNA sequencing revealed that EPPK1 KO induced downregulation of 11 oncogenes, 75 anti-apoptosis, and 22 angiogenesis genes while upregulating 8 tumor suppressors and 12 anti-cell growth genes. We also observed the downregulation of MYC and upregulation of p53 expression at both protein and RNA levels following EPPK1 KO. Gene ontology enrichment analysis of molecular functions highlighted the correlation of EPPK1 with the regulation of mesenchymal cell proliferation, mesenchymal differentiation, angiogenesis, and cell growth after EPPK1 KO. CONCLUSIONS: Our data suggest that EPPK1 is linked to smoking, epithelial to mesenchymal transition, and the regulation of cancer progression, indicating its potential as a therapeutic target for LUAD.


Assuntos
Adenocarcinoma de Pulmão , Adenocarcinoma , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/patologia , Transição Epitelial-Mesenquimal/genética , Prognóstico , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma/patologia , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral
2.
BMC Cancer ; 23(1): 783, 2023 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-37612638

RESUMO

BACKGROUND: There is a need for biomarkers that improve accuracy compared with current demographic risk indices to detect individuals at the highest lung cancer risk. Improved risk determination will enable more effective lung cancer screening and better stratification of lung nodules into high or low-risk category. We previously reported discovery of a biomarker for lung cancer risk characterized by increased prevalence of TP53 somatic mutations in airway epithelial cells (AEC). Here we present results from a validation study in an independent retrospective case-control cohort. METHODS: Targeted next generation sequencing was used to identify mutations within three TP53 exons spanning 193 base pairs in AEC genomic DNA. RESULTS: TP53 mutation prevalence was associated with cancer status (P < 0.001). The lung cancer detection receiver operator characteristic (ROC) area under the curve (AUC) for the TP53 biomarker was 0.845 (95% confidence limits 0.749-0.942). In contrast, TP53 mutation prevalence was not significantly associated with age or smoking pack-years. The combination of TP53 mutation prevalence with PLCOM2012 risk score had an ROC AUC of 0.916 (0.846-0.986) and this was significantly higher than that for either factor alone (P < 0.03). CONCLUSIONS: These results support the validity of the TP53 mutation prevalence biomarker and justify taking additional steps to assess this biomarker in AEC specimens from a prospective cohort and in matched nasal brushing specimens as a potential non-invasive surrogate specimen.


Assuntos
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/epidemiologia , Neoplasias Pulmonares/genética , Detecção Precoce de Câncer , Estudos Prospectivos , Estudos Retrospectivos , Epitélio , Biomarcadores , Pulmão , Proteína Supressora de Tumor p53/genética
3.
Am J Respir Crit Care Med ; 204(11): 1306-1316, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34464235

RESUMO

Rationale: Patients with indeterminate pulmonary nodules (IPNs) at risk of cancer undergo high rates of invasive, costly, and morbid procedures. Objectives: To train and externally validate a risk prediction model that combined clinical, blood, and imaging biomarkers to improve the noninvasive management of IPNs. Methods: In this prospectively collected, retrospective blinded evaluation study, probability of cancer was calculated for 456 patient nodules using the Mayo Clinic model, and patients were categorized into low-, intermediate-, and high-risk groups. A combined biomarker model (CBM) including clinical variables, serum high sensitivity CYFRA 21-1 level, and a radiomic signature was trained in cohort 1 (n = 170) and validated in cohorts 2-4 (total n = 286). All patients were pooled to recalibrate the model for clinical implementation. The clinical utility of the CBM compared with current clinical care was evaluated in 2 cohorts. Measurements and Main Results: The CBM provided improved diagnostic accuracy over the Mayo Clinic model with an improvement in area under the curve of 0.124 (95% bootstrap confidence interval, 0.091-0.156; P < 2 × 10-16). Applying 10% and 70% risk thresholds resulted in a bias-corrected clinical reclassification index for cases and control subjects of 0.15 and 0.12, respectively. A clinical utility analysis of patient medical records estimated that a CBM-guided strategy would have reduced invasive procedures from 62.9% to 50.6% in the intermediate-risk benign population and shortened the median time to diagnosis of cancer from 60 to 21 days in intermediate-risk cancers. Conclusions: Integration of clinical, blood, and image biomarkers improves noninvasive diagnosis of patients with IPNs, potentially reducing the rate of unnecessary invasive procedures while shortening the time to diagnosis.


Assuntos
Carcinoma/diagnóstico por imagem , Carcinoma/metabolismo , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/metabolismo , Nódulos Pulmonares Múltiplos/diagnóstico por imagem , Nódulos Pulmonares Múltiplos/metabolismo , Idoso , Biomarcadores/metabolismo , Carcinoma/patologia , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Nódulos Pulmonares Múltiplos/patologia , Valor Preditivo dos Testes , Curva ROC , Fatores de Risco , Tomografia Computadorizada por Raios X
4.
Eur Respir J ; 57(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33303552

RESUMO

INTRODUCTION: Implementation of low-dose chest computed tomography (CT) lung cancer screening and the ever-increasing use of cross-sectional imaging are resulting in the identification of many screen- and incidentally detected indeterminate pulmonary nodules. While the management of nodules with low or high pre-test probability of malignancy is relatively straightforward, those with intermediate pre-test probability commonly require advanced imaging or biopsy. Noninvasive risk stratification tools are highly desirable. METHODS: We previously developed the BRODERS classifier (Benign versus aggRessive nODule Evaluation using Radiomic Stratification), a conventional predictive radiomic model based on eight imaging features capturing nodule location, shape, size, texture and surface characteristics. Herein we report its external validation using a dataset of incidentally identified lung nodules (Vanderbilt University Lung Nodule Registry) in comparison to the Brock model. Area under the curve (AUC), as well as sensitivity, specificity, negative and positive predictive values were calculated. RESULTS: For the entire Vanderbilt validation set (n=170, 54% malignant), the AUC was 0.87 (95% CI 0.81-0.92) for the Brock model and 0.90 (95% CI 0.85-0.94) for the BRODERS model. Using the optimal cut-off determined by Youden's index, the sensitivity was 92.3%, the specificity was 62.0%, the positive (PPV) and negative predictive values (NPV) were 73.7% and 87.5%, respectively. For nodules with intermediate pre-test probability of malignancy, Brock score of 5-65% (n=97), the sensitivity and specificity were 94% and 46%, respectively, the PPV was 78.4% and the NPV was 79.2%. CONCLUSIONS: The BRODERS radiomic predictive model performs well on an independent dataset and may facilitate the management of indeterminate pulmonary nodules.


Assuntos
Neoplasias Pulmonares , Nódulos Pulmonares Múltiplos , Nódulo Pulmonar Solitário , Área Sob a Curva , Detecção Precoce de Câncer , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Nódulos Pulmonares Múltiplos/diagnóstico por imagem , Nódulo Pulmonar Solitário/diagnóstico por imagem , Tomografia Computadorizada por Raios X
5.
Am J Respir Crit Care Med ; 202(2): 241-249, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326730

RESUMO

Rationale: The management of indeterminate pulmonary nodules (IPNs) remains challenging, resulting in invasive procedures and delays in diagnosis and treatment. Strategies to decrease the rate of unnecessary invasive procedures and optimize surveillance regimens are needed.Objectives: To develop and validate a deep learning method to improve the management of IPNs.Methods: A Lung Cancer Prediction Convolutional Neural Network model was trained using computed tomography images of IPNs from the National Lung Screening Trial, internally validated, and externally tested on cohorts from two academic institutions.Measurements and Main Results: The areas under the receiver operating characteristic curve in the external validation cohorts were 83.5% (95% confidence interval [CI], 75.4-90.7%) and 91.9% (95% CI, 88.7-94.7%), compared with 78.1% (95% CI, 68.7-86.4%) and 81.9 (95% CI, 76.1-87.1%), respectively, for a commonly used clinical risk model for incidental nodules. Using 5% and 65% malignancy thresholds defining low- and high-risk categories, the overall net reclassifications in the validation cohorts for cancers and benign nodules compared with the Mayo model were 0.34 (Vanderbilt) and 0.30 (Oxford) as a rule-in test, and 0.33 (Vanderbilt) and 0.58 (Oxford) as a rule-out test. Compared with traditional risk prediction models, the Lung Cancer Prediction Convolutional Neural Network was associated with improved accuracy in predicting the likelihood of disease at each threshold of management and in our external validation cohorts.Conclusions: This study demonstrates that this deep learning algorithm can correctly reclassify IPNs into low- or high-risk categories in more than a third of cancers and benign nodules when compared with conventional risk models, potentially reducing the number of unnecessary invasive procedures and delays in diagnosis.


Assuntos
Aprendizado Profundo , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/fisiopatologia , Nódulos Pulmonares Múltiplos/diagnóstico por imagem , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Algoritmos , Humanos , Neoplasias Pulmonares/epidemiologia , Redes Neurais de Computação , Estados Unidos/epidemiologia
6.
Neurocomputing (Amst) ; 397: 48-59, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32863584

RESUMO

With the rapid development of image acquisition and storage, multiple images per class are commonly available for computer vision tasks (e.g., face recognition, object detection, medical imaging, etc.). Recently, the recurrent neural network (RNN) has been widely integrated with convolutional neural networks (CNN) to perform image classification on ordered (sequential) data. In this paper, by permutating multiple images as multiple dummy orders, we generalize the ordered "RNN+CNN" design (longitudinal) to a novel unordered fashion, called Multi-path x-D Recurrent Neural Network (MxDRNN) for image classification. To the best of our knowledge, few (if any) existing studies have deployed the RNN framework to unordered intra-class images to leverage classification performance. Specifically, multiple learning paths are introduced in the MxDRNN to extract discriminative features by permutating input dummy orders. Eight datasets from five different fields (MNIST, 3D-MNIST, CIFAR, VGGFace2, and lung screening computed tomography) are included to evaluate the performance of our method. The proposed MxDRNN improves the baseline performance by a large margin across the different application fields (e.g., accuracy from 46.40% to 76.54% in VGGFace2 test pose set, AUC from 0.7418 to 0.8162 in NLST lung dataset). Additionally, empirical experiments show the MxDRNN is more robust to category-irrelevant attributes (e.g., expression, pose in face images), which may introduce difficulties for image classification and algorithm generalizability. The code is publicly available.

7.
Cancer Biomark ; 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38517780

RESUMO

BACKGROUND: Large community cohorts are useful for lung cancer research, allowing for the analysis of risk factors and development of predictive models. OBJECTIVE: A robust methodology for (1) identifying lung cancer and pulmonary nodules diagnoses as well as (2) associating multimodal longitudinal data with these events from electronic health record (EHRs) is needed to optimally curate cohorts at scale. METHODS: In this study, we leveraged (1) SNOMED concepts to develop ICD-based decision rules for building a cohort that captured lung cancer and pulmonary nodules and (2) clinical knowledge to define time windows for collecting longitudinal imaging and clinical concepts. We curated three cohorts with clinical data and repeated imaging for subjects with pulmonary nodules from our Vanderbilt University Medical Center. RESULTS: Our approach achieved an estimated sensitivity 0.930 (95% CI: [0.879, 0.969]), specificity of 0.996 (95% CI: [0.989, 1.00]), positive predictive value of 0.979 (95% CI: [0.959, 1.000]), and negative predictive value of 0.987 (95% CI: [0.976, 0.994]) for distinguishing lung cancer from subjects with SPNs. CONCLUSION: This work represents a general strategy for high-throughput curation of multi-modal longitudinal cohorts at risk for lung cancer from routinely collected EHRs.

8.
Artigo em Inglês | MEDLINE | ID: mdl-37465098

RESUMO

In lung cancer screening, estimation of future lung cancer risk is usually guided by demographics and smoking status. The role of constitutional profiles of human body, a.k.a. body habitus, is increasingly understood to be important, but has not been integrated into risk models. Chest low dose computed tomography (LDCT) is the standard imaging study in lung cancer screening, with the capability to discriminate differences in body composition and organ arrangement in the thorax. We hypothesize that the primary phenotypes identified using lung screening chest LDCT can form a representation of body habitus and add predictive power for lung cancer risk stratification. In this pilot study, we evaluated the feasibility of body habitus image-based phenotyping on a large lung screening LDCT dataset. A thoracic imaging manifold was estimated based on an intensity-based pairwise (dis)similarity metric for pairs of spatial normalized chest LDCT images. We applied the hierarchical clustering method on this manifold to identify the primary phenotypes. Body habitus features of each identified phenotype were evaluated and associated with future lung cancer risk using time-to-event analysis. We evaluated the method on the baseline LDCT scans of 1,200 male subjects sampled from National Lung Screening Trial. Five primary phenotypes were identified, which were associated with highly distinguishable clinical and body habitus features. Time-to-event analysis against future lung cancer incidences showed two of the five identified phenotypes were associated with elevated future lung cancer risks (HR=1.61, 95% CI = [1.08, 2.38], p=0.019; HR=1.67, 95% CI = [0.98, 2.86], p=0.057). These results indicated that it is feasible to capture the body habitus by image-base phenotyping using lung screening LDCT and the learned body habitus representation can potentially add value for future lung cancer risk stratification.

9.
Med Image Anal ; 88: 102852, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37276799

RESUMO

Field-of-view (FOV) tissue truncation beyond the lungs is common in routine lung screening computed tomography (CT). This poses limitations for opportunistic CT-based body composition (BC) assessment as key anatomical structures are missing. Traditionally, extending the FOV of CT is considered as a CT reconstruction problem using limited data. However, this approach relies on the projection domain data which might not be available in application. In this work, we formulate the problem from the semantic image extension perspective which only requires image data as inputs. The proposed two-stage method identifies a new FOV border based on the estimated extent of the complete body and imputes missing tissues in the truncated region. The training samples are simulated using CT slices with complete body in FOV, making the model development self-supervised. We evaluate the validity of the proposed method in automatic BC assessment using lung screening CT with limited FOV. The proposed method effectively restores the missing tissues and reduces BC assessment error introduced by FOV tissue truncation. In the BC assessment for large-scale lung screening CT datasets, this correction improves both the intra-subject consistency and the correlation with anthropometric approximations. The developed method is available at https://github.com/MASILab/S-EFOV.


Assuntos
Processamento de Imagem Assistida por Computador , Semântica , Humanos , Processamento de Imagem Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Tórax , Composição Corporal , Imagens de Fantasmas , Algoritmos
10.
Sci Rep ; 13(1): 17604, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37848457

RESUMO

Lung adenocarcinoma (LUAD) is the predominant type of lung cancer in the U.S. and exhibits a broad variety of behaviors ranging from indolent to aggressive. Identification of the biological determinants of LUAD behavior at early stages can improve existing diagnostic and treatment strategies. Extracellular matrix (ECM) remodeling and cancer-associated fibroblasts play a crucial role in the regulation of cancer aggressiveness and there is a growing need to investigate their role in the determination of LUAD behavior at early stages. We analyzed tissue samples isolated from patients with LUAD at early stages and used imaging-based biomarkers to predict LUAD behavior. Single-cell RNA sequencing and histological assessment showed that aggressive LUADs are characterized by a decreased number of ADH1B+ CAFs in comparison to indolent tumors. ADH1B+ CAF enrichment is associated with distinct ECM and immune cell signatures in early-stage LUADs. Also, we found a positive correlation between the gene expression of ADH1B+ CAF markers in early-stage LUADs and better survival. We performed TCGA dataset analysis to validate our findings. Identified associations can be used for the development of the predictive model of LUAD aggressiveness and novel therapeutic approaches.


Assuntos
Adenocarcinoma de Pulmão , Fibroblastos Associados a Câncer , Síndrome de DiGeorge , Neoplasias Pulmonares , Humanos , Adenocarcinoma de Pulmão/genética , Agressão , Neoplasias Pulmonares/genética , Prognóstico , Biomarcadores Tumorais/genética
11.
Med Image Comput Comput Assist Interv ; 14221: 649-659, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38779102

RESUMO

The accuracy of predictive models for solitary pulmonary nodule (SPN) diagnosis can be greatly increased by incorporating repeat imaging and medical context, such as electronic health records (EHRs). However, clinically routine modalities such as imaging and diagnostic codes can be asynchronous and irregularly sampled over different time scales which are obstacles to longitudinal multimodal learning. In this work, we propose a transformer-based multimodal strategy to integrate repeat imaging with longitudinal clinical signatures from routinely collected EHRs for SPN classification. We perform unsupervised disentanglement of latent clinical signatures and leverage time-distance scaled self-attention to jointly learn from clinical signatures expressions and chest computed tomography (CT) scans. Our classifier is pretrained on 2,668 scans from a public dataset and 1,149 subjects with longitudinal chest CTs, billing codes, medications, and laboratory tests from EHRs of our home institution. Evaluation on 227 subjects with challenging SPNs revealed a significant AUC improvement over a longitudinal multimodal baseline (0.824 vs 0.752 AUC), as well as improvements over a single cross-section multimodal scenario (0.809 AUC) and a longitudinal imaging-only scenario (0.741 AUC). This work demonstrates significant advantages with a novel approach for co-learning longitudinal imaging and non-imaging phenotypes with transformers. Code available at https://github.com/MASILab/lmsignatures.

12.
JTO Clin Res Rep ; 4(9): 100504, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37674811

RESUMO

Introduction: Lung cancer is the deadliest cancer in the United States and worldwide, and lung adenocarcinoma (LUAD) is the most prevalent histologic subtype in the United States. LUAD exhibits a wide range of aggressiveness and risk of recurrence, but the biological underpinnings of this behavior are poorly understood. Past studies have focused on the biological characteristics of the tumor itself, but the ability of the immune response to contain tumor growth represents an alternative or complementary hypothesis. Emerging technologies enable us to investigate the spatial distribution of specific cell types within the tumor nest and characterize this immune response. This study aimed to investigate the association between immune cell density within the primary tumor and recurrence-free survival (RFS) in stage I and II LUAD. Methods: This study is a prospective collection with retrospective evaluation. A total of 100 patients with surgically resected LUAD and at least 5-year follow-ups, including 69 stage I and 31 stages II tumors, were enrolled. Multiplexed immunohistochemistry panels for immune markers were used for measurement. Results: Cox regression models adjusted for sex and EGFR mutation status revealed that the risk of recurrence was reduced by 50% for the unit of one interquartile range (IQR) change in the tumoral T-cell (adjusted hazard ratio per IQR increase = 0.50, 95% confidence interval: 0.27-0.93) and decreased by 64% in mast cell density (adjusted hazard ratio per IQR increase = 0.36, confidence interval: 0.15-0.84). The analyses were reported without the type I error correction for the multiple types of immune cell testing. Conclusions: Analysis of the density of immune cells within the tumor and surrounding stroma reveals an association between the density of T-cells and RFS and between mast cells and RFS in early-stage LUAD. This preliminary result is a limited study with a small sample size and a lack of an independent validation set.

13.
Sci Rep ; 13(1): 6157, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37061539

RESUMO

A deep learning model (LCP CNN) for the stratification of indeterminate pulmonary nodules (IPNs) demonstrated better discrimination than commonly used clinical prediction models. However, the LCP CNN score is based on a single timepoint that ignores longitudinal information when prior imaging studies are available. Clinically, IPNs are often followed over time and temporal trends in nodule size or morphology inform management. In this study we investigated whether the change in LCP CNN scores over time was different between benign and malignant nodules. This study used a prospective-specimen collection, retrospective-blinded-evaluation (PRoBE) design. Subjects with incidentally or screening detected IPNs 6-30 mm in diameter with at least 3 consecutive CT scans prior to diagnosis (slice thickness ≤ 1.5 mm) with the same nodule present were included. Disease outcome was adjudicated by biopsy-proven malignancy, biopsy-proven benign disease and absence of growth on at least 2-year imaging follow-up. Lung nodules were analyzed using the Optellum LCP CNN model. Investigators performing image analysis were blinded to all clinical data. The LCP CNN score was determined for 48 benign and 32 malignant nodules. There was no significant difference in the initial LCP CNN score between benign and malignant nodules. Overall, the LCP CNN scores of benign nodules remained relatively stable over time while that of malignant nodules continued to increase over time. The difference in these two trends was statistically significant. We also developed a joint model that incorporates longitudinal LCP CNN scores to predict future probability of cancer. Malignant and benign nodules appear to have distinctive trends in LCP CNN score over time. This suggests that longitudinal modeling may improve radiomic prediction of lung cancer over current models. Additional studies are needed to validate these early findings.


Assuntos
Neoplasias Pulmonares , Nódulos Pulmonares Múltiplos , Nódulo Pulmonar Solitário , Humanos , Estudos Retrospectivos , Estudos Prospectivos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Redes Neurais de Computação , Nódulos Pulmonares Múltiplos/diagnóstico por imagem , Nódulo Pulmonar Solitário/diagnóstico por imagem , Pulmão/patologia
14.
Cancer Biomark ; 33(4): 449-465, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35491773

RESUMO

The Early Detection Research Network's (EDRN) purpose is to discover, develop and validate biomarkers and imaging methods to detect early-stage cancers or at-risk individuals. The EDRN is composed of sites that fall into four categories: Biomarker Developmental Laboratories (BDL), Biomarker Reference Laboratories (BRL), Clinical Validation Centers (CVC) and Data Management and Coordinating Centers. Each component has a crucial role to play within the mission of the EDRN. The primary role of the CVCs is to support biomarker developers through validation trials on promising biomarkers discovered by both EDRN and non-EDRN investigators. The second round of funding for the EDRN Lung CVC at Vanderbilt University Medical Center (VUMC) was funded in October 2016 and we intended to accomplish the three missions of the CVCs: To conduct innovative research on the validation of candidate biomarkers for early cancer detection and risk assessment of lung cancer in an observational study; to compare biomarker performance; and to serve as a resource center for collaborative research within the Network and partner with established EDRN BDLs and BRLs, new laboratories and industry partners. This report outlines the impact of the VUMC EDRN Lung CVC and describes the role in promoting and validating biological and imaging biomarkers.


Assuntos
Biomarcadores Tumorais , Detecção Precoce de Câncer , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/diagnóstico , Estudos de Validação como Assunto
15.
PLoS One ; 17(3): e0265427, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35294486

RESUMO

BACKGROUND: 18F-fluorodeoxyglucose (FDG) PET/CT is recommended for evaluation of intermediate-risk indeterminate pulmonary nodules (IPNs). While highly sensitive, the specificity of FDG remains suboptimal for differentiating malignant from benign nodules, particularly in areas where fungal lung diseases are prevalent. Thus, a cancer-specific imaging probe is greatly needed. In this study, we tested the hypothesis that a PET radiotracer (S)-4-(3-[18F]-fluoropropyl)-L-glutamic acid (FSPG) improves the diagnostic accuracy of IPNs compared to 18F-FDG PET/CT. METHODS: This study was conducted at a major academic medical center and an affiliated VA medical center. Twenty-six patients with newly discovered IPNs 7-30mm diameter or newly diagnosed lung cancer completed serial PET/CT scans utilizing 18F-FDG and 18F-FSPG, without intervening treatment of the lesion. The scans were independently reviewed by two dual-trained diagnostic radiology and nuclear medicine physicians. Characteristics evaluated included quantitative SUVmax values of the pulmonary nodules and metastases. RESULTS: A total of 17 out of 26 patients had cancer and 9 had benign lesions. 18F-FSPG was negative in 6 of 9 benign lesions compared to 7 of 9 with 18F-FDG. 18F-FSPG and 18F-FDG were positive in 14 of 17 and 12 of 17 malignant lesions, respectively. 18F-FSPG detected brain and intracardiac metastases missed by 18F-FDG PET in one case, while 18F-FDG detected a metastasis to the kidney missed by 18F-FSPG. CONCLUSION: In this pilot study, there was no significant difference in overall diagnostic accuracy between 18F-FSPG and 18F-FDG for the evaluation of IPNs and staging of lung cancer. Additional studies will be needed to determine the clinical utility of this tracer in the management of IPNs and lung cancer.


Assuntos
Neoplasias Pulmonares , Nódulos Pulmonares Múltiplos , Fluordesoxiglucose F18 , Ácido Glutâmico , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Nódulos Pulmonares Múltiplos/diagnóstico por imagem , Projetos Piloto , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Sensibilidade e Especificidade
16.
Comput Biol Med ; 150: 106113, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36198225

RESUMO

OBJECTIVE: Patients with indeterminate pulmonary nodules (IPN) with an intermediate to a high probability of lung cancer generally undergo invasive diagnostic procedures. Chest computed tomography image and clinical data have been in estimating the pretest probability of lung cancer. In this study, we apply a deep learning network to integrate multi-modal data from CT images and clinical data (including blood-based biomarkers) to improve lung cancer diagnosis. Our goal is to reduce uncertainty and to avoid morbidity, mortality, over- and undertreatment of patients with IPNs. METHOD: We use a retrospective study design with cross-validation and external-validation from four different sites. We introduce a deep learning framework with a two-path structure to learn from CT images and clinical data. The proposed model can learn and predict with single modality if the multi-modal data is not complete. We use 1284 patients in the learning cohort for model development. Three external sites (with 155, 136 and 96 patients, respectively) provided patient data for external validation. We compare our model to widely applied clinical prediction models (Mayo and Brock models) and image-only methods (e.g., Liao et al. model). RESULTS: Our co-learning model improves upon the performance of clinical-factor-only (Mayo and Brock models) and image-only (Liao et al.) models in both cross-validation of learning cohort (e.g. , AUC: 0.787 (ours) vs. 0.707-0.719 (baselines), results reported in validation fold and external-validation using three datasets from University of Pittsburgh Medical Center (e.g., 0.918 (ours) vs. 0.828-0.886 (baselines)), Detection of Early Cancer Among Military Personnel (e.g., 0.712 (ours) vs. 0.576-0.709 (baselines)), and University of Colorado Denver (e.g., 0.847 (ours) vs. 0.679-0.746 (baselines)). In addition, our model achieves better re-classification performance (cNRI 0.04 to 0.20) in all cross- and external-validation sets compared to the Mayo model. CONCLUSIONS: Lung cancer risk estimation in patients with IPNs can benefit from the co-learning of CT image and clinical data. Learning from more subjects, even though those only have a single modality, can improve the prediction accuracy. An integrated deep learning model can achieve reasonable discrimination and re-classification performance.


Assuntos
Aprendizado Profundo , Neoplasias Pulmonares , Nódulos Pulmonares Múltiplos , Humanos , Estudos Retrospectivos , Incerteza , Nódulos Pulmonares Múltiplos/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem
17.
Artigo em Inglês | MEDLINE | ID: mdl-36303578

RESUMO

Certain body composition phenotypes, like sarcopenia, are well established as predictive markers for post-surgery complications and overall survival of lung cancer patients. However, their association with incidental lung cancer risk in the screening population is still unclear. We study the feasibility of body composition analysis using chest low dose computed tomography (LDCT). A two-stage fully automatic pipeline is developed to assess the cross-sectional area of body composition components including subcutaneous adipose tissue (SAT), muscle, visceral adipose tissue (VAT), and bone on T5, T8 and T10 vertebral levels. The pipeline is developed using 61 cases of the VerSe'20 dataset, 40 annotated cases of NLST, and 851 inhouse screening cases. On a test cohort consisting of 30 cases from the inhouse screening cohort (age 55 - 73, 50% female) and 42 cases of NLST (age 55 - 75, 59.5% female), the pipeline achieves a root mean square error (RMSE) of 7.25 mm (95% CI: [6.61, 7.85]) for the vertebral level identification and mean Dice similarity score (DSC) 0.99 ± 0.02, 0.96 ± 0.03, and 0.95 ± 0.04 for SAT, muscle, and VAT, respectively for body composition segmentation. The pipeline is generalized to the CT arm of the NLST dataset (25,205 subjects, 40.8% female, 1,056 lung cancer incidences). Time-to-event analysis for lung cancer incidence indicates inverse association between measured muscle cross-sectional area and incidental lung cancer risks (p < 0.001 female, p < 0.001 male). In conclusion, automatic body composition analysis using routine lung screening LDCT is feasible.

18.
Cancer Res ; 82(16): 2838-2847, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35748739

RESUMO

Genomic profiling of bronchoalveolar lavage (BAL) samples may be useful for tumor profiling and diagnosis in the clinic. Here, we compared tumor-derived mutations detected in BAL samples from subjects with non-small cell lung cancer (NSCLC) to those detected in matched plasma samples. Cancer Personalized Profiling by Deep Sequencing (CAPP-Seq) was used to genotype DNA purified from BAL, plasma, and tumor samples from patients with NSCLC. The characteristics of cell-free DNA (cfDNA) isolated from BAL fluid were first characterized to optimize the technical approach. Somatic mutations identified in tumor were then compared with those identified in BAL and plasma, and the potential of BAL cfDNA analysis to distinguish lung cancer patients from risk-matched controls was explored. In total, 200 biofluid and tumor samples from 38 cases and 21 controls undergoing BAL for lung cancer evaluation were profiled. More tumor variants were identified in BAL cfDNA than plasma cfDNA in all stages (P < 0.001) and in stage I to II disease only. Four of 21 controls harbored low levels of cancer-associated driver mutations in BAL cfDNA [mean variant allele frequency (VAF) = 0.5%], suggesting the presence of somatic mutations in nonmalignant airway cells. Finally, using a Random Forest model with leave-one-out cross-validation, an exploratory BAL genomic classifier identified lung cancer with 69% sensitivity and 100% specificity in this cohort and detected more cancers than BAL cytology. Detecting tumor-derived mutations by targeted sequencing of BAL cfDNA is technically feasible and appears to be more sensitive than plasma profiling. Further studies are required to define optimal diagnostic applications and clinical utility. SIGNIFICANCE: Hybrid-capture, targeted deep sequencing of lung cancer mutational burden in cell-free BAL fluid identifies more tumor-derived mutations with increased allele frequencies compared with plasma cell-free DNA. See related commentary by Rolfo et al., p. 2826.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Ácidos Nucleicos Livres , Neoplasias Pulmonares , Biomarcadores Tumorais/genética , Líquido da Lavagem Broncoalveolar , DNA de Neoplasias/genética , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias Pulmonares/patologia , Mutação
19.
Chest ; 159(1): e53-e56, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33422242

RESUMO

CASE PRESENTATION: A 56-year-old man presented to the lung nodule clinic with abnormal chest imaging prompted by a chronic cough and hemoptysis. Approximately 2.5 years earlier, while kneeling beside his car fixing a flat tire, he fell backwards while holding the tire cap in his mouth, causing him to inhale sharply and aspirate the cap. He immediately developed an intractable cough productive of flecks of blood. He presented to an emergency room but left before being seen because of a long wait time and his lack of health-care insurance. He self-medicated for severe cough and chest discomfort with codeine, eventually developing a dependency. Approximately 3 weeks after aspirating the tire cap, his cough became productive, and he developed fever and chills. His symptoms improved transiently with antibiotics and additional narcotics. Ultimately, his chronic cough with intermittent hemoptysis affected his ability to work, and 30 months later he sought medical attention and was diagnosed with pneumonia and reactive airway disease. He was prescribed doxycycline, steroids, inhaled albuterol, and dextromethorphan, with initial improvement, but his symptoms recurred multiple times despite quitting smoking, leading to repeated medication courses.


Assuntos
Brônquios , Tosse/etiologia , Corpos Estranhos/complicações , Corpos Estranhos/diagnóstico por imagem , Hemoptise/etiologia , Broncoscopia , Tosse/diagnóstico por imagem , Corpos Estranhos/cirurgia , Hemoptise/diagnóstico por imagem , Humanos , Masculino , Pessoa de Meia-Idade , Tomografia Computadorizada por Raios X
20.
Artigo em Inglês | MEDLINE | ID: mdl-34650321

RESUMO

Clinical data elements (CDEs) (e.g., age, smoking history), blood markers and chest computed tomography (CT) structural features have been regarded as effective means for assessing lung cancer risk. These independent variables can provide complementary information and we hypothesize that combining them will improve the prediction accuracy. In practice, not all patients have all these variables available. In this paper, we propose a new network design, termed as multi-path multi-modal missing network (M3Net), to integrate the multi-modal data (i.e., CDEs, biomarker and CT image) considering missing modality with multiple paths neural network. Each path learns discriminative features of one modality, and different modalities are fused in a second stage for an integrated prediction. The network can be trained end-to-end with both medical image features and CDEs/biomarkers, or make a prediction with single modality. We evaluate M3Net with datasets including three sites from the Consortium for Molecular and Cellular Characterization of Screen-Detected Lesions (MCL) project. Our method is cross validated within a cohort of 1291 subjects (383 subjects with complete CDEs/biomarkers and CT images), and externally validated with a cohort of 99 subjects (99 with complete CDEs/biomarkers and CT images). Both cross-validation and external-validation results show that combining multiple modality significantly improves the predicting performance of single modality. The results suggest that integrating subjects with missing either CDEs/biomarker or CT imaging features can contribute to the discriminatory power of our model (p < 0.05, bootstrap two-tailed test). In summary, the proposed M3Net framework provides an effective way to integrate image and non-image data in the context of missing information.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA