Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Br J Cancer ; 125(5): 641-657, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33958734

RESUMO

The natural history and treatment landscape of primary brain tumours are complicated by the varied tumour behaviour of primary or secondary gliomas (high-grade transformation of low-grade lesions), as well as the dilemmas with identification of radiation necrosis, tumour progression, and pseudoprogression on MRI. Radiomics and radiogenomics promise to offer precise diagnosis, predict prognosis, and assess tumour response to modern chemotherapy/immunotherapy and radiation therapy. This is achieved by a triumvirate of morphological, textural, and functional signatures, derived from a high-throughput extraction of quantitative voxel-level MR image metrics. However, the lack of standardisation of acquisition parameters and inconsistent methodology between working groups have made validations unreliable, hence multi-centre studies involving heterogenous study populations are warranted. We elucidate novel radiomic and radiogenomic workflow concepts and state-of-the-art descriptors in sub-visual MR image processing, with relevant literature on applications of such machine learning techniques in glioma management.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Genômica/métodos , Glioma/diagnóstico por imagem , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioma/genética , Glioma/patologia , Humanos , Aprendizado de Máquina , Imageamento por Ressonância Magnética , Gradação de Tumores , Prognóstico
2.
Radiology ; 290(3): 783-792, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30561278

RESUMO

Purpose To evaluate ability of radiomic (computer-extracted imaging) features to distinguish non-small cell lung cancer adenocarcinomas from granulomas at noncontrast CT. Materials and Methods For this retrospective study, screening or standard diagnostic noncontrast CT images were collected for 290 patients (mean age, 68 years; range, 18-92 years; 125 men [mean age, 67 years; range, 18-90 years] and 165 women [mean age, 68 years; range, 33-92 years]) from two institutions between 2007 and 2013. Histopathologic analysis was available for one nodule per patient. Corresponding nodule of interest was identified on axial CT images by a radiologist with manual annotation. Nodule shape, wavelet (Gabor), and texture-based (Haralick and Laws energy) features were extracted from intra- and perinodular regions. Features were pruned to train machine learning classifiers with 145 patients. In a test set of 145 patients, classifier results were compared against a convolutional neural network (CNN) and diagnostic readings of two radiologists. Results Support vector machine classifier with intranodular radiomic features achieved an area under the receiver operating characteristic curve (AUC) of 0.75 on the test set. Combining radiomics of intranodular with perinodular regions improved the AUC to 0.80. On the same test set, CNN resulted in an AUC of 0.76. Radiologist readers achieved AUCs of 0.61 and 0.60, respectively. Conclusion Radiomic features from intranodular and perinodular regions of nodules can distinguish non-small cell lung cancer adenocarcinomas from benign granulomas at noncontrast CT. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Nishino in this issue.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Granuloma/diagnóstico por imagem , Pneumopatias/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Nódulo Pulmonar Solitário/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Máquina de Vetores de Suporte
3.
ArXiv ; 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38855551

RESUMO

Background: Predictive biomarkers of treatment response are lacking for metastatic clearcell renal cell carcinoma (ccRCC), a tumor type that is treated with angiogenesis inhibitors, immune checkpoint inhibitors, mTOR inhibitors and a HIF2 inhibitor. The Angioscore, an RNA-based quantification of angiogenesis, is arguably the best candidate to predict anti-angiogenic (AA) response. However, the clinical adoption of transcriptomic assays faces several challenges including standardization, time delay, and high cost. Further, ccRCC tumors are highly heterogenous, and sampling multiple areas for sequencing is impractical. Approach: Here we present a novel deep learning (DL) approach to predict the Angioscore from ubiquitous histopathology slides. In order to overcome the lack of interpretability, one of the biggest limitations of typical DL models, our model produces a visual vascular network which is the basis of the model's prediction. To test its reliability, we applied this model to multiple cohorts including a clinical trial dataset. Results: Our model accurately predicts the RNA-based Angioscore on multiple independent cohorts (spearman correlations of 0.77 and 0.73). Further, the predictions help unravel meaningful biology such as association of angiogenesis with grade, stage, and driver mutation status. Finally, we find our model is able to predict response to AA therapy, in both a real-world cohort and the IMmotion150 clinical trial. The predictive power of our model vastly exceeds that of CD31, a marker of vasculature, and nearly rivals the performance (c-index 0.66 vs 0.67) of the ground truth RNA-based Angioscore at a fraction of the cost. Conclusion: By providing a robust yet interpretable prediction of the Angioscore from histopathology slides alone, our approach offers insights into angiogenesis biology and AA treatment response.

4.
IEEE Trans Med Imaging ; 41(7): 1764-1777, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35108202

RESUMO

The concept of tumor field effect implies that cancer is a systemic disease with its impact way beyond the visible tumor confines. For instance, in Glioblastoma (GBM), an aggressive brain tumor, the increase in intracranial pressure due to tumor burden often leads to brain herniation and poor outcomes. Our work is based on the rationale that highly aggressive tumors tend to grow uncontrollably, leading to pronounced biomechanical tissue deformations in the normal parenchyma, which when combined with local morphological differences in the tumor confines on MRI scans, will comprehensively capture tumor field effect. Specifically, we present an integrated MRI-based descriptor, radiomic-Deformation and Textural Heterogeneity (r-DepTH). This descriptor comprises measurements of the subtle perturbations in tissue deformations throughout the surrounding normal parenchyma due to mass effect. This involves non-rigidly aligning the patients' MRI scans to a healthy atlas via diffeomorphic registration. The resulting inverse mapping is used to obtain the deformation field magnitudes in the normal parenchyma. These measurements are then combined with a 3D texture descriptor, Co-occurrence of Local Anisotropic Gradient Orientations (COLLAGE), which captures the morphological heterogeneity and infiltration within the tumor confines, on MRI scans. In this work, we extensively evaluated r-DepTH for survival risk-stratification on a total of 207 GBM cases from 3 different cohorts (Cohort 1 ( n1 = 53 ), Cohort 2 ( n2 = 75 ), and Cohort 3 ( n3 = 79 )), where each of these three cohorts was used as a training set for our model separately, and the other two cohorts were used for testing, independently, for each training experiment. When employing Cohort 1 for training, r-DepTH yielded Concordance indices (C-indices) of 0.7 and 0.65, hazard ratios (HR) and Confidence Intervals (CI) of 10 (6 - 19) and 5 (3 - 8) on Cohorts 2 and 3, respectively. Similarly, training on Cohort 2 yielded C-indices of 0.6 and 0.7, HR and CI of 1 (0.7 - 2) and 3 (2 - 5) on Cohorts 1 and 3, respectively. Finally, training on Cohort 3 yielded C-indices of 0.75 and 0.63, HR and CI of 24 (10 - 57) and 12 (6 - 21) on Cohorts 1 and 2, respectively. Our results show that r-DepTH descriptor may serve as a comprehensive and a robust MRI-based prognostic marker of disease aggressiveness and survival in solid tumors.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Anisotropia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Estudos de Coortes , Glioblastoma/diagnóstico por imagem , Glioblastoma/patologia , Humanos , Imageamento por Ressonância Magnética/métodos , Prognóstico
5.
NPJ Precis Oncol ; 6(1): 33, 2022 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-35661148

RESUMO

Despite known histological, biological, and clinical differences between lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC), relatively little is known about the spatial differences in their corresponding immune contextures. Our study of over 1000 LUAD and LUSC tumors revealed that computationally derived patterns of tumor-infiltrating lymphocytes (TILs) on H&E images were different between LUAD (N = 421) and LUSC (N = 438), with TIL density being prognostic of overall survival in LUAD and spatial arrangement being more prognostically relevant in LUSC. In addition, the LUAD-specific TIL signature was associated with OS in an external validation set of 100 NSCLC treated with more than six different neoadjuvant chemotherapy regimens, and predictive of response to therapy in the clinical trial CA209-057 (n = 303). In LUAD, the prognostic TIL signature was primarily comprised of CD4+ T and CD8+ T cells, whereas in LUSC, the immune patterns were comprised of CD4+ T, CD8+ T, and CD20+ B cells. In both subtypes, prognostic TIL features were associated with transcriptomics-derived immune scores and biological pathways implicated in immune recognition, response, and evasion. Our results suggest the need for histologic subtype-specific TIL-based models for stratifying survival risk and predicting response to therapy. Our findings suggest that predictive models for response to therapy will need to account for the unique morphologic and molecular immune patterns as a function of histologic subtype of NSCLC.

6.
Neuro Oncol ; 23(2): 251-263, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33068415

RESUMO

BACKGROUND: Recent epidemiological studies have suggested that sexual dimorphism influences treatment response and prognostic outcome in glioblastoma (GBM). To this end, we sought to (i) identify distinct sex-specific radiomic phenotypes-from tumor subcompartments (peritumoral edema, enhancing tumor, and necrotic core) using pretreatment MRI scans-that are prognostic of overall survival (OS) in GBMs, and (ii) investigate radiogenomic associations of the MRI-based phenotypes with corresponding transcriptomic data, to identify the signaling pathways that drive sex-specific tumor biology and treatment response in GBM. METHODS: In a retrospective setting, 313 GBM patients (male = 196, female = 117) were curated from multiple institutions for radiomic analysis, where 130 were used for training and independently validated on a cohort of 183 patients. For the radiogenomic analysis, 147 GBM patients (male = 94, female = 53) were used, with 125 patients in training and 22 cases for independent validation. RESULTS: Cox regression models of radiomic features from gadolinium T1-weighted MRI allowed for developing more precise prognostic models, when trained separately on male and female cohorts. Our radiogenomic analysis revealed higher expression of Laws energy features that capture spots and ripple-like patterns (representative of increased heterogeneity) from the enhancing tumor region, as well as aggressive biological processes of cell adhesion and angiogenesis to be more enriched in the "high-risk" group of poor OS in the male population. In contrast, higher expressions of Laws energy features (which detect levels and edges) from the necrotic core with significant involvement of immune related signaling pathways was observed in the "low-risk" group of the female population. CONCLUSIONS: Sexually dimorphic radiogenomic models could help risk-stratify GBM patients for personalized treatment decisions.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Prognóstico , Estudos Retrospectivos
7.
Neurooncol Adv ; 2(Suppl 4): iv3-iv14, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33521636

RESUMO

Neuro-oncology largely consists of malignancies of the brain and central nervous system including both primary as well as metastatic tumors. Currently, a significant clinical challenge in neuro-oncology is to tailor therapies for patients based on a priori knowledge of their survival outcome or treatment response to conventional or experimental therapies. Radiomics or the quantitative extraction of subvisual data from conventional radiographic imaging has recently emerged as a powerful data-driven approach to offer insights into clinically relevant questions related to diagnosis, prediction, prognosis, as well as assessing treatment response. Furthermore, radiogenomic approaches provide a mechanism to establish statistical correlations of radiomic features with point mutations and next-generation sequencing data to further leverage the potential of routine MRI scans to serve as "virtual biopsy" maps. In this review, we provide an introduction to radiomic and radiogenomic approaches in neuro-oncology, including a brief description of the workflow involving preprocessing, tumor segmentation, and extraction of "hand-crafted" features from the segmented region of interest, as well as identifying radiogenomic associations that could ultimately lead to the development of reliable prognostic and predictive models in neuro-oncology applications. Lastly, we discuss the promise of radiomics and radiogenomic approaches in personalizing treatment decisions in neuro-oncology, as well as the challenges with clinical adoption, which will rely heavily on their demonstrated resilience to nonstandardization in imaging protocols across sites and scanners, as well as in their ability to demonstrate reproducibility across large multi-institutional cohorts.

8.
Radiol Artif Intell ; 2(6): e190168, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33330847

RESUMO

PURPOSE: To identify radiomic features extracted from the tumor habitat on routine MR images that are prognostic for progression-free survival (PFS) and to assess their morphologic basis with corresponding histopathologic attributes in glioblastoma (GBM). MATERIALS AND METHODS: In this retrospective study, 156 pretreatment GBM MR images (gadolinium-enhanced T1-weighted, T2-weighted, and fluid-attenuated inversion recovery [FLAIR] images) were curated. Of these 156 images, 122 were used for training (90 from The Cancer Imaging Archive and 32 from the Cleveland Clinic, acquired between December 1, 2011, and May 1, 2018) and 34 were used for validation. The validation set was obtained from the Ivy Glioblastoma Atlas Project database, for which the percentage extent of 11 histologic attributes was available on corresponding histopathologic specimens of the resected tumor. Following expert annotations of the tumor habitat (necrotic core, enhancing tumor, and FLAIR-hyperintense subcompartments), 1008 radiomic descriptors (eg, Haralick texture features, Laws energy features, co-occurrence of local anisotropic gradient orientations [CoLIAGe]) were extracted from the three MRI sequences. The top radiomic features were obtained from each subcompartment in the training set on the basis of their ability to risk-stratify patients according to PFS. These features were then concatenated to create a radiomics risk score (RRS). The RRS was independently validated on a holdout set. In addition, correlations (P < .05) of RRS features were computed, with the percentage extent of the 11 histopathologic attributes, using Spearman correlation analysis. RESULTS: RRS yielded a concordance index of 0.80 on the validation set and constituted radiomic features, including Laws (capture edges, waves, ripple patterns) and CoLIAGe (capture disease heterogeneity) from enhancing tumor and FLAIR hyperintensity. These radiomic features were correlated with histopathologic attributes associated with disease aggressiveness in GBM, particularly tumor infiltration (P = .0044) and hyperplastic blood vessels (P = .0005). CONCLUSION: Preliminary findings demonstrated significant associations of prognostic radiomic features with disease-specific histologic attributes, with implications for risk-stratifying patients with GBM for personalized treatment decisions. Supplemental material is available for this article. © RSNA, 2020.

9.
Med Phys ; 47(12): 6029-6038, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33176026

RESUMO

PURPOSE: There is an increasing availability of large imaging cohorts [such as through The Cancer Imaging Archive (TCIA)] for computational model development and imaging research. To ensure development of generalizable computerized models, there is a need to quickly determine relative quality differences in these cohorts, especially when considering MRI datasets which can exhibit wide variations in image appearance. The purpose of this study is to present a quantitative quality control tool, MRQy, to help interrogate MR imaging datasets for: (a) site- or scanner-specific variations in image resolution or image contrast, and (b) imaging artifacts such as noise or inhomogeneity; which need correction prior to model development. METHODS: Unlike existing imaging quality control tools, MRQy has been generalized to work with images from any body region to efficiently extract a series of quality measures (e.g., noise ratios, variation metrics) and MR image metadata (e.g., voxel resolution and image dimensions). MRQy also offers a specialized HTML5-based front-end designed for real-time filtering and trend visualization of quality measures. RESULTS: MRQy was used to evaluate (a) n = 133 brain MRIs from TCIA (7 sites) and (b) n = 104 rectal MRIs (3 local sites). MRQy measures revealed significant site-specific variations in both cohorts, indicating potential batch effects. Before processing, MRQy measures could be used to identify each of the seven sites within the TCIA cohort with 87.5%, 86.4%, 90%, 93%, 90%, 60%, and 92.9% accuracy and the three sites within the rectal cohort with 91%, 82.8%, and 88.9% accuracy using unsupervised clustering. After processing, none of the sites could be distinctively clustered via MRQy measures in either cohort; suggesting that batch effects had been largely accounted for. Marked differences in specific MRQy measures were also able to identify outlier MRI datasets that needed to be corrected for common acquisition artifacts. CONCLUSIONS: MRQy is designed to be a standalone, unsupervised tool that can be efficiently run on a standard desktop computer. It has been made freely accessible and open-source at http://github.com/ccipd/MRQy for community use and feedback.


Assuntos
Artefatos , Imageamento por Ressonância Magnética , Estudos de Coortes , Humanos , Processamento de Imagem Assistida por Computador , Controle de Qualidade
10.
Lancet Digit Health ; 2(3): e116-e128, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32123864

RESUMO

Background: Development and validation of a quantitative radiomic risk score (QuRiS) and associated nomogram (QuRNom) for early-stage non-small cell lung cancer (ES-NSCLC) that is prognostic of disease-free survival (DFS) and predictive of the added benefit of adjuvant chemotherapy (ACT) following surgery. Methods: QuRiS was developed using radiomic texture features derived from within and outside the primary lung nodule on chest CT scans using a cohort D1 of 329 patients from the Cleveland Clinic. A LASSO-Cox regularization model was used for data dimension reduction, feature selection, and QuRiS construction. QuRiS was independently validated on D2(N=114; University of Pennsylvania) and D3(N=82; TCIA). QuRNom was constructed by integrating QuRiS with T-, N-Descriptors, and LVI. The added benefit of ACT using QuRiS and QuRNom was validated by comparing patients who received ACT against patients who underwent surgery alone in D1-D3. To explore the underlying morphologic basis of the QuRiS, we explored associations with corresponding whole-slide tissue scans (WSIs) and mRNA sequencing data using subsets of D1 and D3. Findings: QuRiS consisted three intra- and ten peri-tumoral CT-radiomic features and was found to be significantly associated with DFS (D1: HR=1.60 [1.10-2.20];p<·05; D2:HR=2.70 [1.40-5.10]; p<·01; D3:HR=2.70 [1.20-5.70];p<·01). Patients were partitioned into three risk groups (QH, QI, QL) based off their corresponding QuRiS score. High QuRiS group, QH, patients were observed to have significantly prolonged survival with ACT when compared to surgery alone (D1: HR=0·27[0.07-0.95],p<0.05; D2+D3: HR=0·08[0.01-0.42],p<0.01). For developed QuRNom, the actual efficacy of ACT was predictive of nomogram-estimated survival benefit (D1: HR= D1:0·25 [0·12-0·55], D3: HR=0·13 [0·004-0·99]). QuRiS features were found to be associated with the spatial arrangement of TILs and cancer nuclei on corresponding WSIs (D1: Rho=0·23,p<0·05, N=70). They were also observed to have an association with biological pathways implicated in chemotaxis (D3,p<0·05, N=86) and other immune specific biological pathways. Interpretation: QuRiS and QuRNom were validated as being prognostic of DFS and predictive of the added benefit of ACT.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Estadiamento de Neoplasias , Avaliação de Resultados em Cuidados de Saúde/métodos , Tomografia Computadorizada por Raios X , Idoso , Quimioterapia Adjuvante , Intervalo Livre de Doença , Feminino , Previsões , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
11.
Med Phys ; 47(12): 6039-6052, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33118182

RESUMO

PURPOSE: The availability of radiographic magnetic resonance imaging (MRI) scans for the Ivy Glioblastoma Atlas Project (Ivy GAP) has opened up opportunities for development of radiomic markers for prognostic/predictive applications in glioblastoma (GBM). In this work, we address two critical challenges with regard to developing robust radiomic approaches: (a) the lack of availability of reliable segmentation labels for glioblastoma tumor sub-compartments (i.e., enhancing tumor, non-enhancing tumor core, peritumoral edematous/infiltrated tissue) and (b) identifying "reproducible" radiomic features that are robust to segmentation variability across readers/sites. ACQUISITION AND VALIDATION METHODS: From TCIA's Ivy GAP cohort, we obtained a paired set (n = 31) of expert annotations approved by two board-certified neuroradiologists at the Hospital of the University of Pennsylvania (UPenn) and at Case Western Reserve University (CWRU). For these studies, we performed a reproducibility study that assessed the variability in (a) segmentation labels and (b) radiomic features, between these paired annotations. The radiomic variability was assessed on a comprehensive panel of 11 700 radiomic features including intensity, volumetric, morphologic, histogram-based, and textural parameters, extracted for each of the paired sets of annotations. Our results demonstrated (a) a high level of inter-rater agreement (median value of DICE ≥0.8 for all sub-compartments), and (b) ≈24% of the extracted radiomic features being highly correlated (based on Spearman's rank correlation coefficient) to annotation variations. These robust features largely belonged to morphology (describing shape characteristics), intensity (capturing intensity profile statistics), and COLLAGE (capturing heterogeneity in gradient orientations) feature families. DATA FORMAT AND USAGE NOTES: We make publicly available on TCIA's Analysis Results Directory (https://doi.org/10.7937/9j41-7d44), the complete set of (a) multi-institutional expert annotations for the tumor sub-compartments, (b) 11 700 radiomic features, and (c) the associated reproducibility meta-analysis. POTENTIAL APPLICATIONS: The annotations and the associated meta-data for Ivy GAP are released with the purpose of enabling researchers toward developing image-based biomarkers for prognostic/predictive applications in GBM.


Assuntos
Glioblastoma , Estudos de Coortes , Glioblastoma/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Reprodutibilidade dos Testes
12.
Lancet Digit Health ; 2(3): e116-e128, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-33334576

RESUMO

BACKGROUND: Use of adjuvant chemotherapy in patients with early-stage lung cancer is controversial because no definite biomarker exists to identify patients who would receive added benefit from it. We aimed to develop and validate a quantitative radiomic risk score (QuRiS) and associated nomogram (QuRNom) for early-stage non-small cell lung cancer (NSCLC) that is prognostic of disease-free survival and predictive of the added benefit of adjuvant chemotherapy following surgery. METHODS: We did a retrospective multicohort study of individuals with early-stage NSCLC (stage I and II) who either received surgery alone or surgery plus adjuvant chemotherapy. We selected patients for whom we had available pre-treatment diagnostic CT scans and corresponding survival information. We used radiomic texture features derived from within and outside the primary lung nodule on chest CT scans of patients from the Cleveland Clinic Foundation (Cleveland, OH, USA; cohort D1) to develop QuRiS. A least absolute shrinkage and selection operator-Cox regularisation model was used for data dimension reduction, feature selection, and QuRiS construction. QuRiS was independently validated on a cohort of patients from the University of Pennsylvania (Philadephia, PA, USA; cohort D2) and a cohort of patients whose CT scans were derived from The Cancer Imaging Archive (cohort D3). QuRNom was constructed by integrating QuRiS with tumour and node descriptors (according to the tumour, node, metastasis staging system) and lymphovascular invasion. The primary endpoint of the study was the assessment of the performance of QuRiS and QuRNom in predicting disease-free survival. The added benefit of adjuvant chemotherapy estimated using QuRiS and QuRNom was validated by comparing patients who received adjuvant chemotherapy versus patients who underwent surgery alone in cohorts D1-D3. FINDINGS: We included: 329 patients in cohort D1 (73 [22%] had surgery plus adjuvant chemotherapy and 256 (78%) had surgery alone); 114 patients in cohort D2 (33 [29%] had surgery plus adjuvant chemotherapy and 81 (71%) had surgery alone); and 82 patients in cohort D3 (24 [29%] had surgery plus adjuvant chemotherapy and 58 (71%) had surgery alone). QuRiS comprised three intratumoral and 10 peritumoral CT-radiomic features and was found to be significantly associated with disease-free survival (ie, prognostic validation of QuRiS; hazard ratio for predicted high-risk vs predicted low-risk groups 1·56, 95% CI 1·08-2·23, p=0·016 for cohort D1; 2·66, 1·24-5·68, p=0·011 for cohort D2; and 2·67, 1·39-5·11, p=0·0029 for cohort D3). To validate the predictive performance of QuRiS, patients were partitioned into three risk groups (high, intermediate, and low risk) on the basis of their corresponding QuRiS. Patients in the high-risk group were observed to have significantly longer survival with adjuvant chemotherapy than patients who underwent surgery alone (0·27, 0·08-0·95, p=0·042, for cohort D1; 0·08, 0·01-0·42, p=0·0029, for cohorts D2 and D3 combined). As concerns QuRNom, the nomogram-estimated survival benefit was predictive of the actual efficacy of adjuvant chemotherapy (0·25, 0·12-0·55, p<0·0001, for cohort D1; 0·13, <0·01-0·99, p=0·0019 for cohort D3). INTERPRETATION: QuRiS and QuRNom were validated as being prognostic of disease-free survival and predictive of the added benefit of adjuvant chemotherapy, especially in clinically defined low-risk groups. Since QuRiS is based on routine chest CT imaging, with additional multisite independent validation it could potentially be employed for decision management in non-invasive treatment of resectable lung cancer. FUNDING: National Cancer Institute of the US National Institutes of Health, National Center for Research Resources, US Department of Veterans Affairs Biomedical Laboratory Research and Development Service, Department of Defence, National Institute of Diabetes and Digestive and Kidney Diseases, Wallace H Coulter Foundation, Case Western Reserve University, and Dana Foundation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/tratamento farmacológico , Tomografia Computadorizada por Raios X , Idoso , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Quimioterapia Adjuvante , Estudos de Coortes , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Prognóstico , Estudos Retrospectivos
13.
Lancet Digit Health ; 2(11): e594-e606, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33163952

RESUMO

Background: Intratumoural heterogeneity has been previously shown to be related to clonal evolution and genetic instability and associated with tumour progression. Phenotypically, it is reflected in the diversity of appearance and morphology within cell populations. Computer-extracted features relating to tumour cellular diversity on routine tissue images might correlate with outcome. This study investigated the prognostic ability of computer-extracted features of tumour cellular diversity (CellDiv) from haematoxylin and eosin (H&E)-stained histology images of non-small cell lung carcinomas (NSCLCs). Methods: In this multicentre, retrospective study, we included 1057 patients with early-stage NSCLC with corresponding diagnostic histology slides and overall survival information from four different centres. CellDiv features quantifying local cellular morphological diversity from H&E-stained histology images were extracted from the tumour epithelium region. A Cox proportional hazards model based on CellDiv was used to construct risk scores for lung adenocarcinoma (LUAD; 270 patients) and lung squamous cell carcinoma (LUSC; 216 patients) separately using data from two of the cohorts, and was validated in the two remaining independent cohorts (comprising 236 patients with LUAD and 335 patients with LUSC). We used multivariable Cox regression analysis to examine the predictive ability of CellDiv features for 5-year overall survival, controlling for the effects of clinical and pathological parameters. We did a gene set enrichment and Gene Ontology analysis on 405 patients to identify associations with differentially expressed biological pathways implicated in lung cancer pathogenesis. Findings: For prognosis of patients with early-stage LUSC, the CellDiv LUSC model included 11 discriminative CellDiv features, whereas for patients with early-stage LUAD, the model included 23 features. In the independent validation cohorts, patients predicted to be at a higher risk by the univariable CellDiv model had significantly worse 5-year overall survival (hazard ratio 1·48 [95% CI 1·06-2·08]; p=0·022 for The Cancer Genome Atlas [TCGA] LUSC group, 2·24 [1·04-4·80]; p=0·039 for the University of Bern LUSC group, and 1·62 [1·15-2·30]; p=0·0058 for the TCGA LUAD group). The identified CellDiv features were also found to be strongly associated with apoptotic signalling and cell differentiation pathways. Interpretation: CellDiv features were strongly prognostic of 5-year overall survival in patients with early-stage NSCLC and also associated with apoptotic signalling and cell differentiation pathways. The CellDiv-based risk stratification model could potentially help to determine which patients with early-stage NSCLC might receive added benefit from adjuvant therapy. Funding: National Institue of Health and US Department of Defense.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Análise de Sobrevida , Idoso , Bases de Dados Factuais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos
14.
Clin Cancer Res ; 26(8): 1866-1876, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32079590

RESUMO

PURPOSE: To (i) create a survival risk score using radiomic features from the tumor habitat on routine MRI to predict progression-free survival (PFS) in glioblastoma and (ii) obtain a biological basis for these prognostic radiomic features, by studying their radiogenomic associations with molecular signaling pathways. EXPERIMENTAL DESIGN: Two hundred three patients with pretreatment Gd-T1w, T2w, T2w-FLAIR MRI were obtained from 3 cohorts: The Cancer Imaging Archive (TCIA; n = 130), Ivy GAP (n = 32), and Cleveland Clinic (n = 41). Gene-expression profiles of corresponding patients were obtained for TCIA cohort. For every study, following expert segmentation of tumor subcompartments (necrotic core, enhancing tumor, peritumoral edema), 936 3D radiomic features were extracted from each subcompartment across all MRI protocols. Using Cox regression model, radiomic risk score (RRS) was developed for every protocol to predict PFS on the training cohort (n = 130) and evaluated on the holdout cohort (n = 73). Further, Gene Ontology and single-sample gene set enrichment analysis were used to identify specific molecular signaling pathway networks associated with RRS features. RESULTS: Twenty-five radiomic features from the tumor habitat yielded the RRS. A combination of RRS with clinical (age and gender) and molecular features (MGMT and IDH status) resulted in a concordance index of 0.81 (P < 0.0001) on training and 0.84 (P = 0.03) on the test set. Radiogenomic analysis revealed associations of RRS features with signaling pathways for cell differentiation, cell adhesion, and angiogenesis, which contribute to chemoresistance in GBM. CONCLUSIONS: Our findings suggest that prognostic radiomic features from routine Gd-T1w MRI may also be significantly associated with key biological processes that affect response to chemotherapy in GBM.


Assuntos
Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/mortalidade , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Mutação , Medição de Risco/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Transdução de Sinais , Taxa de Sobrevida , Adulto Jovem
15.
Sci Rep ; 9(1): 1145, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718547

RESUMO

Subtle tissue deformations caused by mass-effect in Glioblastoma (GBM) are often not visually evident, and may cause neurological deficits, impacting survival. Radiomic features provide sub-visual quantitative measures to uncover disease characteristics. We present a new radiomic feature to capture mass effect-induced deformations in the brain on Gadolinium-contrast (Gd-C) T1w-MRI, and their impact on survival. Our rationale is that larger variations in deformation within functionally eloquent areas of the contralateral hemisphere are likely related to decreased survival. Displacements in the cortical and subcortical structures were measured by aligning the Gd-C T1w-MRI to a healthy atlas. The variance of deformation magnitudes was measured and defined as Mass Effect Deformation Heterogeneity (MEDH) within the brain structures. MEDH values were then correlated with overall-survival of 89 subjects on the discovery cohort, with tumors on the right (n = 41) and left (n = 48) cerebral hemispheres, and evaluated on a hold-out cohort (n = 49 subjects). On both cohorts, decreased survival time was found to be associated with increased MEDH in areas of language comprehension, social cognition, visual perception, emotion, somato-sensory, cognitive and motor-control functions, particularly in the memory areas in the left-hemisphere. Our results suggest that higher MEDH in functionally eloquent areas of the left-hemisphere due to GBM in the right-hemisphere may be associated with poor-survival.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Cérebro/diagnóstico por imagem , Cérebro/patologia , Glioblastoma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Estudos de Coortes , Estudos de Viabilidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sobrevida
17.
JAMA Netw Open ; 2(4): e192561, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-31002322

RESUMO

Importance: There has been significant recent interest in understanding the utility of quantitative imaging to delineate breast cancer intrinsic biological factors and therapeutic response. No clinically accepted biomarkers are as yet available for estimation of response to human epidermal growth factor receptor 2 (currently known as ERBB2, but referred to as HER2 in this study)-targeted therapy in breast cancer. Objective: To determine whether imaging signatures on clinical breast magnetic resonance imaging (MRI) could noninvasively characterize HER2-positive tumor biological factors and estimate response to HER2-targeted neoadjuvant therapy. Design, Setting, and Participants: In a retrospective diagnostic study encompassing 209 patients with breast cancer, textural imaging features extracted within the tumor and annular peritumoral tissue regions on MRI were examined as a means to identify increasingly granular breast cancer subgroups relevant to therapeutic approach and response. First, among a cohort of 117 patients who received an MRI prior to neoadjuvant chemotherapy (NAC) at a single institution from April 27, 2012, through September 4, 2015, imaging features that distinguished HER2+ tumors from other receptor subtypes were identified. Next, among a cohort of 42 patients with HER2+ breast cancers with available MRI and RNaseq data accumulated from a multicenter, preoperative clinical trial (BrUOG 211B), a signature of the response-associated HER2-enriched (HER2-E) molecular subtype within HER2+ tumors (n = 42) was identified. The association of this signature with pathologic complete response was explored in 2 patient cohorts from different institutions, where all patients received HER2-targeted NAC (n = 28, n = 50). Finally, the association between significant peritumoral features and lymphocyte distribution was explored in patients within the BrUOG 211B trial who had corresponding biopsy hematoxylin-eosin-stained slide images. Data analysis was conducted from January 15, 2017, to February 14, 2019. Main Outcomes and Measures: Evaluation of imaging signatures by the area under the receiver operating characteristic curve (AUC) in identifying HER2+ molecular subtypes and distinguishing pathologic complete response (ypT0/is) to NAC with HER2-targeting. Results: In the 209 patients included (mean [SD] age, 51.1 [11.7] years), features from the peritumoral regions better discriminated HER2-E tumors (maximum AUC, 0.85; 95% CI, 0.79-0.90; 9-12 mm from the tumor) compared with intratumoral features (AUC, 0.76; 95% CI, 0.69-0.84). A classifier combining peritumoral and intratumoral features identified the HER2-E subtype (AUC, 0.89; 95% CI, 0.84-0.93) and was significantly associated with response to HER2-targeted therapy in both validation cohorts (AUC, 0.80; 95% CI, 0.61-0.98 and AUC, 0.69; 95% CI, 0.53-0.84). Features from the 0- to 3-mm peritumoral region were significantly associated with the density of tumor-infiltrating lymphocytes (R2 = 0.57; 95% CI, 0.39-0.75; P = .002). Conclusions and Relevance: A combination of peritumoral and intratumoral characteristics appears to identify intrinsic molecular subtypes of HER2+ breast cancers from imaging, offering insights into immune response within the peritumoral environment and suggesting potential benefit for treatment guidance.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Imageamento por Ressonância Magnética/estatística & dados numéricos , Radiometria/estatística & dados numéricos , Receptor ErbB-2/metabolismo , Adulto , Biomarcadores Tumorais/análise , Neoplasias da Mama/metabolismo , Feminino , Humanos , Linfócitos do Interstício Tumoral/patologia , Pessoa de Meia-Idade , Terapia Neoadjuvante , Período Pré-Operatório , Estudos Retrospectivos , Resultado do Tratamento
18.
Lung Cancer ; 115: 34-41, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29290259

RESUMO

Lung cancer is responsible for a large proportion of cancer-related deaths across the globe, with delayed detection being perhaps the most significant factor for its high mortality rate. Though the National Lung Screening Trial argues for screening of certain at-risk populations, the practical implementation of these screening efforts has not yet been successful and remains in high demand. Radiomics refers to the computerized extraction of data from radiologic images, and provides unique potential for making lung cancer screening more rapid and accurate using machine learning algorithms. The quantitative features analyzed express subvisual characteristics of images which correlate with pathogenesis of diseases. These features are broadly classified into four categories: intensity, structure, texture/gradient, and wavelet, based on the types of image attributes they capture. Many studies have been done to show correlation between these features and the malignant potential of a nodule on a chest CT. In cancer patients, these nodules also have features that can be correlated with prognosis and mutation status. The major limitations of radiomics are the lack of standardization of acquisition parameters, inconsistent radiomic methods, and lack of reproducibility. Researchers are working on overcoming these limitations, which would make radiomics more acceptable in the medical community.


Assuntos
Detecção Precoce de Câncer/métodos , Processamento de Imagem Assistida por Computador , Neoplasias Pulmonares/diagnóstico por imagem , Pulmão/diagnóstico por imagem , Algoritmos , Animais , Diagnóstico por Imagem , Humanos , Neoplasias Pulmonares/genética , Prognóstico , Padrões de Referência , Software , Tomografia Computadorizada por Raios X
19.
Sci Rep ; 8(1): 15290, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30327507

RESUMO

Adenocarcinomas and active granulomas can both have a spiculated appearance on computed tomography (CT) and both are often fluorodeoxyglucose (FDG) avid on positron emission tomography (PET) scan, making them difficult to distinguish. Consequently, patients with benign granulomas are often subjected to invasive surgical biopsies or resections. In this study, quantitative vessel tortuosity (QVT), a novel CT imaging biomarker to distinguish between benign granulomas and adenocarcinomas on routine non-contrast lung CT scans is introduced. Our study comprised of CT scans of 290 patients from two different institutions, one cohort for training (N = 145) and the other (N = 145) for independent validation. In conjunction with a machine learning classifier, the top informative and stable QVT features yielded an area under receiver operating characteristic curve (ROC AUC) of 0.85 in the independent validation set. On the same cohort, the corresponding AUCs for two human experts including a radiologist and a pulmonologist were found to be 0.61 and 0.60, respectively. QVT features also outperformed well known shape and textural radiomic features which had a maximum AUC of 0.73 (p-value = 0.002), as well as features learned using a convolutional neural network AUC = 0.76 (p-value = 0.028). Our results suggest that QVT features could potentially serve as a non-invasive imaging biomarker to distinguish granulomas from adenocarcinomas on non-contrast CT scans.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Vasos Sanguíneos/patologia , Granuloma/diagnóstico por imagem , Pneumopatias Fúngicas/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Pulmão/irrigação sanguínea , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Idoso , Idoso de 80 Anos ou mais , Conjuntos de Dados como Assunto , Diagnóstico Diferencial , Feminino , Humanos , Aprendizado de Máquina , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
20.
Sci Rep ; 8(1): 7, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311558

RESUMO

Hypoxia, a characteristic trait of Glioblastoma (GBM), is known to cause resistance to chemo-radiation treatment and is linked with poor survival. There is hence an urgent need to non-invasively characterize tumor hypoxia to improve GBM management. We hypothesized that (a) radiomic texture descriptors can capture tumor heterogeneity manifested as a result of molecular variations in tumor hypoxia, on routine treatment naïve MRI, and (b) these imaging based texture surrogate markers of hypoxia can discriminate GBM patients as short-term (STS), mid-term (MTS), and long-term survivors (LTS). 115 studies (33 STS, 41 MTS, 41 LTS) with gadolinium-enhanced T1-weighted MRI (Gd-T1w) and T2-weighted (T2w) and FLAIR MRI protocols and the corresponding RNA sequences were obtained. After expert segmentation of necrotic, enhancing, and edematous/nonenhancing tumor regions for every study, 30 radiomic texture descriptors were extracted from every region across every MRI protocol. Using the expression profile of 21 hypoxia-associated genes, a hypoxia enrichment score (HES) was obtained for the training cohort of 85 cases. Mutual information score was used to identify a subset of radiomic features that were most informative of HES within 3-fold cross-validation to categorize studies as STS, MTS, and LTS. When validated on an additional cohort of 30 studies (11 STS, 9 MTS, 10 LTS), our results revealed that the most discriminative features of HES were also able to distinguish STS from LTS (p = 0.003).


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Hipóxia/genética , Hipóxia/metabolismo , Adulto , Idoso , Biomarcadores , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/mortalidade , Feminino , Perfilação da Expressão Gênica , Genômica/métodos , Glioblastoma/diagnóstico por imagem , Glioblastoma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA