299 research outputs found

    Co-Segmentation Methods for Improving Tumor Target Delineation in PET-CT Images

    Get PDF
    Positron emission tomography (PET)-Computed tomography (CT) plays an important role in cancer management. As a multi-modal imaging technique it provides both functional and anatomical information of tumor spread. Such information improves cancer treatment in many ways. One important usage of PET-CT in cancer treatment is to facilitate radiotherapy planning, for the information it provides helps radiation oncologists to better target the tumor region. However, currently most tumor delineations in radiotherapy planning are performed by manual segmentation, which consumes a lot of time and work. Most computer-aided algorithms need a knowledgeable user to locate roughly the tumor area as a starting point. This is because, in PET-CT imaging, some tissues like heart and kidney may also exhibit a high level of activity similar to that of a tumor region. In order to address this issue, a novel co-segmentation method is proposed in this work to enhance the accuracy of tumor segmentation using PET-CT, and a localization algorithm is developed to differentiate and segment tumor regions from normal regions. On a combined dataset containing 29 patients with lung tumor, the combined method shows good segmentation results as well as good tumor recognition rate

    Classification of small renal masses based on CT images and machine learning algorithms

    Get PDF
    Kidney tumor is among the leading causes of tumors and deaths worldwide. In all kidney tumor cases, an increasing number of small renal masses (SRMs) with a size smaller than 4 cm have been detected and they are becoming a typical problem for radiologists and surgeons. Most SRMs are either of renal angiomyolipoma (AML) or renal cell carcinoma (RCC), the former being benign and the latter being malignant. The malignant ones can be further classified into three types, clear cell renal cell carcinoma (ccRCC), papillary renal cell carcinoma (pRCC), and chromophobe renal cell carcinoma (chRCC). Different kind of renal tumor requires varied treatment and management. In recent years, four-phase computer tomography (CT) has become the standard approach for kidney tumor examination. In most circumstances, classic AMLs and RCCs can be classified by a radiologist reading the CT images. While fat poor angiomyolipomas (fp-AML) set barriers to this classification method due to the loss of typical diagnosis characteristics. Radiologists are also incapable of differentiating malignant tumors. For now, SRM classification is mainly performed by pathological examination, which is time and resource consuming. Machine learning and one of its branch, deep learning, has been extended to medical image processing field. In this paper, support vector machine (SVM) and convolutional neural network (CNN) were respectively used to build models with the input of one of the last three phases of CT images and the combination of them. For the establishment of each model, at least 20% of overall patient cases were picked out randomly as independent testing subset and the rest undertook 10-fold cross validation for an objective and reliable evaluation of the models. It turned out that SVM algorithm using a linear kernel with phase 2 (corticomedullary) images as input acquired an accuracy of 0.93 and a sensitivity of 0.97 on patient’s tumor type prediction of fp-AML/RCC classification. CNN algorithm, consisting of 12 layers including 4 convolutional layers each followed by a max-pooling layer, one flatten layer, and three densely connected layers, with the help of activation functions, dropout strategy, and stochastic gradient descent (SGD) optimization method, achieved an accuracy of 0.85 on pRCC/chRCC/ccRCC categorization with phase 2 images as input. Images of corticomedullary stage were proved to be eligible for classifiers. This can be seen as a breakthrough since it is the first successful application of deep learning networks in renal tumor classification. Meanwhile, these two models were both balanced over different classes and they together provide a comprehensive solution to SRM classification. Given these findings, the two models can be a preliminary step for machine learning and especially deep learning algorithms to assist, improve, and finally revolutionize the conventional clinical decision making process to guide appropriate management and treatment

    Studies on Category Prediction of Ovarian Cancers Based on Magnetic Resonance Images

    Get PDF
    Ovarian cancer is the gynecological malignant tumor with low early diagnosis rate and high mortality. Ovarian epithelial cancer (OEC) is the most common subtype of ovarian cancer. Pathologically, OEC is divided into two subtypes: Type I and Type II. These two subtypes of OEC have different biological characteristics and treatment response. Therefore, it is important to accurately categorize these two groups of patients and provide the reference for clinicians in designing treatment plans. In the current magnetic resonance (MR) examination, the diagnoses given by the radiologists are largely based on individual judgment and not sufficiently accurate. Because of the low accuracy of the results and the risk of suffering Type II OEC, most patients will undertake the fine-needle aspiration, which may cause harm to patients’ bodies. Therefore, there is need for the method for OEC subtype classification based on MR images. This thesis proposes the automatic diagnosis system of ovarian cancer based on the combination of deep learning and radiomics. The method utilizes four common useful sequences for ovarian cancer diagnosis: sagittal fat-suppressed T2WI (Sag-fs-T2WI), coronal T2WI (Cor-T2WI), axial T1WI (Axi-T1WI), and apparent diffusion coefficient map (ADC) to establish a multi-sequence diagnostic model. The system starts with the segmentation of the ovarian tumors, and then obtains the radiomic features from lesion parts together with the network features. Selected Features are used to build model to predict the malignancy of ovarian cancers, the subtype of OEC and the survival condition. Bi-atten-ResUnet is proposed in this thesis as the segmentation model. The network is established on the basis of U-Net with adopting Residual block and non-local attention module. It preserves the classic encoder/decoder architecture in the U-Net network. The encoder part is reconstructed by the pretrained ResNet to make use of transfer learning knowledge, and bi-non-local attention modules are added to the decoder part on each level. The application of these techniques enhances the network’s performance in segmentation tasks. The model achieves 0.918, 0.905, 0.831, and 0.820 Dice coefficient respectively in segmenting on four MR sequences. After the segmentation work, the thesis proposes a diagnostic model with three steps: quantitative description feature extraction, feature selection, and establishment of prediction models. First, radiomic features and network features are obtained. Then iterative sparse representation (ISR) method is adopted as the feature selection to reduce the redundancy and correlation. The selected features are used to establish a predictive model, and support vector machine (SVM) is used as the classifier. The model achieves an AUC of 0.967 in distinguishing between benign and malignant ovarian tumors. For discriminating Type I and Type II OEC, the model yields an AUC of 0.823. In the survival prediction, patients categorized in high risk group are more likely to have poor prognosis with hazard ratio 4.169

    Quantitative analysis with machine learning models for multi-parametric brain imaging data

    Get PDF
    Gliomas are considered to be the most common primary adult malignant brain tumor. With the dramatic increases in computational power and improvements in image analysis algorithms, computer-aided medical image analysis has been introduced into clinical applications. Precision tumor grading and genotyping play an indispensable role in clinical diagnosis, treatment and prognosis. Gliomas diagnostic procedures include histopathological imaging tests, molecular imaging scans and tumor grading. Pathologic review of tumor morphology in histologic sections is the traditional method for cancer classification and grading, yet human study has limitations that can result in low reproducibility and inter-observer agreement. Compared with histopathological images, Magnetic resonance (MR) imaging present the different structure and functional features, which might serve as noninvasive surrogates for tumor genotypes. Therefore, computer-aided image analysis has been adopted in clinical application, which might partially overcome these shortcomings due to its capacity to quantitatively and reproducibly measure multilevel features on multi-parametric medical information. Imaging features obtained from a single modal image do not fully represent the disease, so quantitative imaging features, including morphological, structural, cellular and molecular level features, derived from multi-modality medical images should be integrated into computer-aided medical image analysis. The image quality differentiation between multi-modality images is a challenge in the field of computer-aided medical image analysis. In this thesis, we aim to integrate the quantitative imaging data obtained from multiple modalities into mathematical models of tumor prediction response to achieve additional insights into practical predictive value. Our major contributions in this thesis are: 1. Firstly, to resolve the imaging quality difference and observer-dependent in histological image diagnosis, we proposed an automated machine-learning brain tumor-grading platform to investigate contributions of multi-parameters from multimodal data including imaging parameters or features from Whole Slide Images (WSI) and the proliferation marker KI-67. For each WSI, we extract both visual parameters such as morphology parameters and sub-visual parameters including first-order and second-order features. A quantitative interpretable machine learning approach (Local Interpretable Model-Agnostic Explanations) was followed to measure the contribution of features for single case. Most grading systems based on machine learning models are considered β€œblack boxes,” whereas with this system the clinically trusted reasoning could be revealed. The quantitative analysis and explanation may assist clinicians to better understand the disease and accordingly to choose optimal treatments for improving clinical outcomes. 2. Based on the automated brain tumor-grading platform we propose, multimodal Magnetic Resonance Images (MRIs) have been introduced in our research. A new imaging–tissue correlation based approach called RA-PA-Thomics was proposed to predict the IDH genotype. Inspired by the concept of image fusion, we integrate multimodal MRIs and the scans of histopathological images for indirect, fast, and cost saving IDH genotyping. The proposed model has been verified by multiple evaluation criteria for the integrated data set and compared to the results in the prior art. The experimental data set includes public data sets and image information from two hospitals. Experimental results indicate that the model provided improves the accuracy of glioma grading and genotyping

    Role of artificial intelligence in risk prediction, prognostication, and therapy response assessment in colorectal cancer: current state and future directions

    Get PDF
    Artificial Intelligence (AI) is a branch of computer science that utilizes optimization, probabilistic and statistical approaches to analyze and make predictions based on a vast amount of data. In recent years, AI has revolutionized the field of oncology and spearheaded novel approaches in the management of various cancers, including colorectal cancer (CRC). Notably, the applications of AI to diagnose, prognosticate, and predict response to therapy in CRC, is gaining traction and proving to be promising. There have also been several advancements in AI technologies to help predict metastases in CRC and in Computer-Aided Detection (CAD) Systems to improve miss rates for colorectal neoplasia. This article provides a comprehensive review of the role of AI in predicting risk, prognosis, and response to therapies among patients with CRC

    Artificial intelligence - based ultrasound elastography for disease evaluation -Β a narrative review

    Get PDF
    Ultrasound elastography (USE) provides complementary information of tissue stiffness and elasticity to conventional ultrasound imaging. It is noninvasive and free of radiation, and has become a valuable tool to improve diagnostic performance with conventional ultrasound imaging. However, the diagnostic accuracy will be reduced due to high operator-dependence and intra- and inter-observer variability in visual observations of radiologists. Artificial intelligence (AI) has great potential to perform automatic medical image analysis tasks to provide a more objective, accurate and intelligent diagnosis. More recently, the enhanced diagnostic performance of AI applied to USE have been demonstrated for various disease evaluations. This review provides an overview of the basic concepts of USE and AI techniques for clinical radiologists and then introduces the applications of AI in USE imaging that focus on the following anatomical sites: liver, breast, thyroid and other organs for lesion detection and segmentation, machine learning (ML) - assisted classification and prognosis prediction. In addition, the existing challenges and future trends of AI in USE are also discussed

    A Medical Analysis for Colorectal Lymphomas using 3D MRI Images and Deep Residual Boltzmann CNN Mechanism

    Get PDF
    In this technological world the healthcare is very crucial and difficult to spend time for the wellbeing. The lifestyle disease can transform in to the life threating disease and lead to critical stages. Colorectal lymphomas are the 3rd most malignancy death in the entire world. The estimation of the volume of lymphomas is often used by Magnetic Resonance Imaging during medical diagnosis, particularly in advanced stages. The research study can be classified in multiple stages. In the initial stages, an automated method is used to calculated the volume of the colorectal lymphomas using 3D MRI images. The process begins with feature extraction using Iterative Multilinear Component Analysis and Multiscale Phase level set segmentation based on CNN model. Then, a logical frustum model is utilized for 3D simulation of colon lymphoma for rendering the medical data. The next stages is focused on tackling the matter of segmentation and classification of abnormality and normality of lymph nodes. A semi supervised fuzzy logic algorithm for clustering is used for segmentation, whereas bee herd optimization algorithm with scale down for employed to intensify corresponding classifier rate of detection. Finally, classification is performed using Deep residual Boltzmann CNN. Our proposed methodology gives a better results and diagnosis prediction for lymphomas for an accuracy 97.7%, sensitivity 95.7% and specify as 95.8% which is superior than the traditional approach

    Artificial intelligence in gastroenterology: a state-of-the-art review

    Get PDF
    The development of artificial intelligence (AI) has increased dramatically in the last 20 years, with clinical applications progressively being explored for most of the medical specialties. The field of gastroenterology and hepatology, substantially reliant on vast amounts of imaging studies, is not an exception. The clinical applications of AI systems in this field include the identification of premalignant or malignant lesions (e.g., identification of dysplasia or esophageal adenocarcinoma in Barrett's esophagus, pancreatic malignancies), detection of lesions (e.g., polyp identification and classification, small-bowel bleeding lesion on capsule endoscopy, pancreatic cystic lesions), development of objective scoring systems for risk stratification, predicting disease prognosis or treatment response [e.g., determining survival in patients post-resection of hepatocellular carcinoma), determining which patients with inflammatory bowel disease (IBD) will benefit from biologic therapy], or evaluation of metrics such as bowel preparation score or quality of endoscopic examination. The objective of this comprehensive review is to analyze the available AI-related studies pertaining to the entirety of the gastrointestinal tract, including the upper, middle and lower tracts; IBD; the hepatobiliary system; and the pancreas, discussing the findings and clinical applications, as well as outlining the current limitations and future directions in this field.Cellular mechanisms in basic and clinical gastroenterology and hepatolog

    Π˜ΡΠΊΡƒΡΡΡ‚Π²Π΅Π½Π½Ρ‹ΠΉ ΠΈΠ½Ρ‚Π΅Π»Π»Π΅ΠΊΡ‚ ΠΏΡ€ΠΈ ΠΊΠΎΠ»ΠΎΡ€Π΅ΠΊΡ‚Π°Π»ΡŒΠ½ΠΎΠΌ Ρ€Π°ΠΊΠ΅: ΠΎΠ±Π·ΠΎΡ€

    Get PDF
    The study objective: the study objective is to examine the use of artificial intelligence (AI) in the diagnosis, treatment, and prognosis of Colorectal Cancer (CRC) and discuss the future potential of AI in CRC. Material and Methods. The Web of Science, Scopus, PubMed, Medline, and eLIBRARY databases were used to search for the publications. A study on the application of Artificial Intelligence (AI) to the diagnosis, treatment, and prognosis of Colorectal Cancer (CRC) was discovered in more than 100 sources. In the review, data from 83 articles were incorporated. Results. The review article explores the use of artificial intelligence (AI) in medicine, specifically focusing on its applications in colorectal cancer (CRC). It discusses the stages of AI development for CRC, including molecular understanding, image-based diagnosis, drug design, and individualized treatment. The benefits of AI in medical image analysis are highlighted, improving diagnosis accuracy and inspection quality. Challenges in AI development are addressed, such as data standardization and the interpretability of machine learning algorithms. The potential of AI in treatment decision support, precision medicine, and prognosis prediction is discussed, emphasizing the role of AI in selecting optimal treatments and improving surgical precision. Ethical and regulatory considerations in integrating AI are mentioned, including patient trust, data security, and liability in AI-assisted surgeries. The review emphasizes the importance of an AI standard system, dataset standardization, and integrating clinical knowledge into AI algorithms. Overall, the article provides an overview of the current research on AI in CRC diagnosis, treatment, and prognosis, discussing its benefits, challenges, and future prospects in improving medical outcomes.ЦСль исслСдования - ΠΎΡ†Π΅Π½ΠΊΠ° возмоТностСй использования искусствСнного ΠΈΠ½Ρ‚Π΅Π»Π»Π΅ΠΊΡ‚Π° (ИИ) Π² диагностикС, Π»Π΅Ρ‡Π΅Π½ΠΈΠΈ ΠΈ ΠΏΡ€ΠΎΠ³Π½ΠΎΠ·ΠΈΡ€ΠΎΠ²Π°Π½ΠΈΠΈ ΠΊΠΎΠ»ΠΎΡ€Π΅ΠΊΡ‚Π°Π»ΡŒΠ½ΠΎΠ³ΠΎ Ρ€Π°ΠΊΠ° (КРР), Π° Ρ‚Π°ΠΊΠΆΠ΅ обсуТдСниС ΠΏΠΎΡ‚Π΅Π½Ρ†ΠΈΠ°Π»Π° ИИ Π² Π»Π΅Ρ‡Π΅Π½ΠΈΠΈ КРР. ΠœΠ°Ρ‚Π΅Ρ€ΠΈΠ°Π» ΠΈ ΠΌΠ΅Ρ‚ΠΎΠ΄Ρ‹. ΠŸΡ€ΠΎΠ²Π΅Π΄Π΅Π½ поиск Π½Π°ΡƒΡ‡Π½Ρ‹Ρ… ΠΏΡƒΠ±Π»ΠΈΠΊΠ°Ρ†ΠΈΠΉ Π² поисковых систСмах Web of Science, Scopus, PubMed, Medline ΠΈ eLIBRARY. Π‘Ρ‹Π»ΠΎ просмотрСно Π±ΠΎΠ»Π΅Π΅ 100 источников ΠΏΠΎ ΠΏΡ€ΠΈΠΌΠ΅Π½Π΅Π½ΠΈΡŽ ИИ для диагностики, лСчСния ΠΈ прогнозирования КРР. Π’ ΠΎΠ±Π·ΠΎΡ€ Π²ΠΊΠ»ΡŽΡ‡Π΅Π½Ρ‹ Π΄Π°Π½Π½Ρ‹Π΅ ΠΈΠ· 83 статСй. Π Π΅Π·ΡƒΠ»ΡŒΡ‚Π°Ρ‚Ρ‹. ΠŸΡ€ΠΎΠ²Π΅Π΄Π΅Π½ Π°Π½Π°Π»ΠΈΠ· Π»ΠΈΡ‚Π΅Ρ€Π°Ρ‚ΡƒΡ€Ρ‹, посвящСнной ΠΏΡ€ΠΈΠΌΠ΅Π½Π΅Π½ΠΈΡŽ искусствСнного ΠΈΠ½Ρ‚Π΅Π»Π»Π΅ΠΊΡ‚Π° Π² ΠΌΠ΅Π΄ΠΈΡ†ΠΈΠ½Π΅, особоС Π²Π½ΠΈΠΌΠ°Π½ΠΈΠ΅ ΡƒΠ΄Π΅Π»Π΅Π½ΠΎ Π΅Π³ΠΎ использованию ΠΏΡ€ΠΈ ΠΊΠΎΠ»ΠΎΡ€Π΅ΠΊΡ‚Π°Π»ΡŒΠ½ΠΎΠΌ Ρ€Π°ΠΊΠ΅. ΠžΠ±ΡΡƒΠΆΠ΄Π°ΡŽΡ‚ΡΡ этапы развития ИИ ΠΏΡ€ΠΈ КРР, Π²ΠΊΠ»ΡŽΡ‡Π°Ρ ΠΌΠΎΠ»Π΅ΠΊΡƒΠ»ΡΡ€Π½ΡƒΡŽ Π²Π΅Ρ€ΠΈΡ„ΠΈΠΊΠ°Ρ†ΠΈΡŽ, Π»ΡƒΡ‡Π΅Π²ΡƒΡŽ диагностику, Ρ€Π°Π·Ρ€Π°Π±ΠΎΡ‚ΠΊΡƒ лСкарств ΠΈ ΠΈΠ½Π΄ΠΈΠ²ΠΈΠ΄ΡƒΠ°Π»ΡŒΠ½ΠΎΠ΅ Π»Π΅Ρ‡Π΅Π½ΠΈΠ΅. ΠŸΠΎΠ΄Ρ‡Π΅Ρ€ΠΊΠ½ΡƒΡ‚Ρ‹ прСимущСства ИИ Π² Π°Π½Π°Π»ΠΈΠ·Π΅ мСдицинских ΠΈΠ·ΠΎΠ±Ρ€Π°ΠΆΠ΅Π½ΠΈΠΉ, Ρ‚Π°ΠΊΠΈΡ… ΠΊΠ°ΠΊ КВ, МРВ ΠΈ ПЭВ, Ρ‡Ρ‚ΠΎ ΠΏΠΎΠ²Ρ‹ΡˆΠ°Π΅Ρ‚ Ρ‚ΠΎΡ‡Π½ΠΎΡΡ‚ΡŒ диагностики. Π Π°ΡΡΠΌΠ°Ρ‚Ρ€ΠΈΠ²Π°ΡŽΡ‚ΡΡ Ρ‚Π°ΠΊΠΈΠ΅ ΠΏΡ€ΠΎΠ±Π»Π΅ΠΌΡ‹ развития ИИ, ΠΊΠ°ΠΊ стандартизация Π΄Π°Π½Π½Ρ‹Ρ… ΠΈ ΠΈΠ½Ρ‚Π΅Ρ€ΠΏΡ€Π΅Ρ‚ΠΈΡ€ΡƒΠ΅ΠΌΠΎΡΡ‚ΡŒ Π°Π»Π³ΠΎΡ€ΠΈΡ‚ΠΌΠΎΠ² машинного обучСния. ΠŸΠΎΠ΄Ρ‡Π΅Ρ€ΠΊΠΈΠ²Π°Π΅Ρ‚ΡΡ Ρ€ΠΎΠ»ΡŒ ИИ Π² Π²Ρ‹Π±ΠΎΡ€Π΅ ΠΎΠΏΡ‚ΠΈΠΌΠ°Π»ΡŒΠ½ΠΎΠΉ Ρ‚Π°ΠΊΡ‚ΠΈΠΊΠΈ лСчСния ΠΈ ΠΏΠΎΠ²Ρ‹ΡˆΠ΅Π½ΠΈΠΈ эффСктивности хирургичСского Π²ΠΌΠ΅ΡˆΠ°Ρ‚Π΅Π»ΡŒΡΡ‚Π²Π°. Π£Ρ‡ΠΈΡ‚Ρ‹Π²Π°ΡŽΡ‚ΡΡ этичСскиС ΠΈ Π½ΠΎΡ€ΠΌΠ°Ρ‚ΠΈΠ²Π½Ρ‹Π΅ аспСкты ИИ, Π²ΠΊΠ»ΡŽΡ‡Π°Ρ Π΄ΠΎΠ²Π΅Ρ€ΠΈΠ΅ ΠΏΠ°Ρ†ΠΈΠ΅Π½Ρ‚ΠΎΠ², Π±Π΅Π·ΠΎΠΏΠ°ΡΠ½ΠΎΡΡ‚ΡŒ Π΄Π°Π½Π½Ρ‹Ρ… ΠΈ ΠΎΡ‚Π²Π΅Ρ‚ΡΡ‚Π²Π΅Π½Π½ΠΎΡΡ‚ΡŒ Π² ΠΏΡ€ΠΎΠ²Π΅Π΄Π΅Π½ΠΈΠΈ ΠΎΠΏΠ΅Ρ€Π°Ρ†ΠΈΠΉ с использованиСм ИИ. ΠžΠ±ΡΡƒΠΆΠ΄Π°ΡŽΡ‚ΡΡ прСимущСства ИИ Π² диагностикС, Π»Π΅Ρ‡Π΅Π½ΠΈΠΈ ΠΈ ΠΏΡ€ΠΎΠ³Π½ΠΎΠ·ΠΈΡ€ΠΎΠ²Π°Π½ΠΈΠΈ ΠΊΠΎΠ»ΠΎΡ€Π΅ΠΊΡ‚Π°Π»ΡŒΠ½ΠΎΠ³ΠΎ Ρ€Π°ΠΊΠ°, ΠΏΡ€ΠΎΠ±Π»Π΅ΠΌΡ‹ ΠΈ пСрспСктивы ΡƒΠ»ΡƒΡ‡ΡˆΠ΅Π½ΠΈΡ Ρ€Π΅Π·ΡƒΠ»ΡŒΡ‚Π°Ρ‚ΠΎΠ² лСчСния

    Artificial Intelligence Predictive Models of Response to Cytotoxic Chemotherapy Alone or Combined to Targeted Therapy for Metastatic Colorectal Cancer Patients: A Systematic Review and Meta-Analysis

    Get PDF
    Simple Summary Metastatic colorectal cancer (mCRC) has high incidence and mortality. Nevertheless, innovative biomarkers have been developed for predicting the response to therapy. We have examined the ability of learning methods to build prognostic and predictive models to predict response to chemotherapy, alone or combined with targeted therapy in mCRC patients, by targeting specific narrative publications. After a literature search, 26 original articles met inclusion and exclusion criteria and were included in the study. We showed that all investigations conducted in this field provided generally promising results in predicting the response to therapy or toxic side-effects, using a meta-analytic approach. We found that radiomics and molecular biomarker signatures were able to discriminate response vs. non-response by correctly identifying up to 99% of mCRC patients who were responders and up to 100% of patients who were non-responders. Our study supports the use of computer science for developing personalized treatment decision processes for mCRC patients. Tailored treatments for metastatic colorectal cancer (mCRC) have not yet completely evolved due to the variety in response to drugs. Therefore, artificial intelligence has been recently used to develop prognostic and predictive models of treatment response (either activity/efficacy or toxicity) to aid in clinical decision making. In this systematic review, we have examined the ability of learning methods to predict response to chemotherapy alone or combined with targeted therapy in mCRC patients by targeting specific narrative publications in Medline up to April 2022 to identify appropriate original scientific articles. After the literature search, 26 original articles met inclusion and exclusion criteria and were included in the study. Our results show that all investigations conducted on this field have provided generally promising results in predicting the response to therapy or toxic side-effects. By a meta-analytic approach we found that the overall weighted means of the area under the receiver operating characteristic (ROC) curve (AUC) were 0.90, 95% C.I. 0.80-0.95 and 0.83, 95% C.I. 0.74-0.89 in training and validation sets, respectively, indicating a good classification performance in discriminating response vs. non-response. The calculation of overall HR indicates that learning models have strong ability to predict improved survival. Lastly, the delta-radiomics and the 74 gene signatures were able to discriminate response vs. non-response by correctly identifying up to 99% of mCRC patients who were responders and up to 100% of patients who were non-responders. Specifically, when we evaluated the predictive models with tests reaching 80% sensitivity (SE) and 90% specificity (SP), the delta radiomics showed an SE of 99% and an SP of 94% in the training set and an SE of 85% and SP of 92 in the test set, whereas for the 74 gene signatures the SE was 97.6% and the SP 100% in the training set
    • …
    corecore