1
|
Xie D, Yu J, He C, Jiang H, Qiu Y, Fu L, Kong L, Xu H. Predicting the immune therapy response of advanced non-small cell lung cancer based on primary tumor and lymph node radiomics features. Front Med (Lausanne) 2025; 12:1541376. [PMID: 40248083 PMCID: PMC12003267 DOI: 10.3389/fmed.2025.1541376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Objective To identify imaging biomarkers of primary tumors and lymph nodes in patients with stage III-IV non-small cell lung cancer (NSCLC) and assess their predictive ability for treatment response (response vs. non-response) to immune checkpoint inhibitors (ICIs) after 6 months. Methods Retrospective analysis of 83 NSCLC patients treated with ICIs. Quantitative imaging features of the maximum primary lung tumors and lymph nodes on contrast-enhanced CT imaging were extracted at baseline (time point 0, TP0) and after 2-3 cycles of immunotherapy (time point 1, TP1). Delta-radiomics features (delta-RFs) were defined as the net changes in radiomics features (RFs) between TP0 and TP1. Interobserver interclass coefficient (ICC) and Pearson correlation analyses were applied for feature selection, and logistic regression (LR) was used to build a model for predicting treatment response. Results Four and five important delta-RFs were selected to construct the nodal and tumor models, respectively. Δ Tumor diameter was used for constructing the clinical prediction model. The predictive efficacy of the nodal model for the treatment response status was higher than that of the tumor and clinical models. In the training set, the AUC values for the three models were 0.96 (95% CI = 0.90-1.00), 0.86 (95% CI = 0.76-0.95), and 0.82 (95% CI = 0.71-0.93), respectively. In the validation set, the AUC values were 0.94 (95% CI = 0.85-1.00), 0.77 (95% CI = 0.56-0.98), and 0.74 (95% CI = 0.48-1.00), respectively. Conclusion The nodal model based on delta-RFs performed well in distinguishing responders from non-responders and could identify patients more likely to benefit from immunotherapy. Finally, the nodal model exhibited a higher classification performance than the tumor model.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Jinna Yu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Han Jiang
- Department of Medical Oncology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Yonggang Qiu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Linfeng Fu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Lingting Kong
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Hongwei Xu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| |
Collapse
|
2
|
Mai H, Li L, Xin X, Jiang Z, Tang Y, Huang J, Lei Y, Chen L, Dong T, Zhong X. Prediction of immunotherapy response in nasopharyngeal carcinoma: a comparative study using MRI-based radiomics signature and programmed cell death ligand 1 expression score. Eur Radiol 2025:10.1007/s00330-025-11350-5. [PMID: 39853331 DOI: 10.1007/s00330-025-11350-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/29/2024] [Accepted: 12/10/2024] [Indexed: 01/26/2025]
Abstract
OBJECTIVES To compare an MRI-based radiomics signature with the programmed cell death ligand 1 (PD-L1) expression score for predicting immunotherapy response in nasopharyngeal carcinoma (NPC). METHODS Consecutive patients with NPC who received immunotherapy between January 2019 and June 2022 were divided into training (n = 111) and validation (n = 66) sets. Tumor radiomics features were extracted from pretreatment MR images. PD-L1 combined positive score (CPS) was calculated using immunohistochemistry. The least absolute shrinkage and selection operator (LASSO) algorithm was used for feature selection and radiomics signature construction. Receiver operating characteristic (ROC) curve analysis was performed to assess prediction performance. RESULTS A total of eleven radiomics features with the greatest discrimination capability were identified by the LASSO algorithm to construct the radiomics signature. In predicting patients with objective response to immunotherapy, radiomics score (Rd-score) yielded a significantly higher area under the ROC curve than that of CPS in both the training (0.790 vs. 0.645, p = 0.025) and the validation (0.735 vs. 0.608, p = 0.038) sets. Multivariate analysis identified the Rd-score as an independent influencing factor in predicting immunotherapy response (odds ratio = 19.963, p < 0.001). Kaplan-Meier analysis indicated that patients with Rd-score ≥ 0.5 showed longer progression-free survival than patients with Rd-score < 0.5 (log-rank p < 0.01). CONCLUSION An MRI-based radiomics signature demonstrated greater efficacy than the PD-L1 expression score in predicting immunotherapy response in patients with NPC. KEY POINTS Question How does an MRI-based radiomics signature compare with the programmed cell death ligand 1 expression score for predicting immunotherapy response in nasopharyngeal carcinoma? Findings The MRI-based radiomics signature demonstrated superior predictive value compared with programmed cell death ligand 1 expression score in identifying immunotherapy responders. Clinical relevance MRI-based radiomics are a promising novel noninvasive tool for predicting immunotherapy outcomes in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Hui Mai
- Department of Radiology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Li Li
- Department of Otolaryngology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xin Xin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhike Jiang
- Department of Radiology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongfang Tang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jie Huang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanxing Lei
- Department of Radiology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lianzhi Chen
- Department of Radiology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tianfa Dong
- Department of Radiology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xi Zhong
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Magnin CY, Lauer D, Ammeter M, Gote-Schniering J. From images to clinical insights: an educational review on radiomics in lung diseases. Breathe (Sheff) 2025; 21:230225. [PMID: 40104259 PMCID: PMC11915127 DOI: 10.1183/20734735.0225-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/20/2025] Open
Abstract
Radiological imaging is a cornerstone in the clinical workup of lung diseases. Radiomics represents a significant advancement in clinical lung imaging, offering a powerful tool to complement traditional qualitative image analysis. Radiomic features are quantitative and computationally describe shape, intensity, texture and wavelet characteristics from medical images that can uncover detailed and often subtle information that goes beyond the visual capabilities of radiological examiners. By extracting this quantitative information, radiomics can provide deep insights into the pathophysiology of lung diseases and support clinical decision-making as well as personalised medicine approaches. In this educational review, we provide a step-by-step guide to radiomics-based medical image analysis, discussing the technical challenges and pitfalls, and outline the potential clinical applications of radiomics in diagnosing, prognosticating and evaluating treatment responses in respiratory medicine.
Collapse
Affiliation(s)
- Cheryl Y Magnin
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Both authors contributed equally
| | - David Lauer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Both authors contributed equally
| | - Michael Ammeter
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Janine Gote-Schniering
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Lee G, Moon SH, Kim JH, Jeong DY, Choi J, Choi JY, Lee HY. Multimodal Imaging Approach for Tumor Treatment Response Evaluation in the Era of Immunotherapy. Invest Radiol 2025; 60:11-26. [PMID: 39018248 DOI: 10.1097/rli.0000000000001096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
ABSTRACT Immunotherapy is likely the most remarkable advancement in lung cancer treatment during the past decade. Although immunotherapy provides substantial benefits, their therapeutic responses differ from those of conventional chemotherapy and targeted therapy, and some patients present unique immunotherapy response patterns that cannot be judged under the current measurement standards. Therefore, the response monitoring of immunotherapy can be challenging, such as the differentiation between real response and pseudo-response. This review outlines the various tumor response patterns to immunotherapy and discusses methods for quantifying computed tomography (CT) and 18 F-fluorodeoxyglucose positron emission tomography (PET) in the field of lung cancer. Emerging technologies in magnetic resonance imaging (MRI) and non-FDG PET tracers are also explored. With immunotherapy responses, the role for imaging is essential in both anatomical radiological responses (CT/MRI) and molecular changes (PET imaging). Multiple aspects must be considered when assessing treatment responses using CT and PET. Finally, we introduce multimodal approaches that integrate imaging and nonimaging data, and we discuss future directions for the assessment and prediction of lung cancer responses to immunotherapy.
Collapse
Affiliation(s)
- Geewon Lee
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (G.L., D.Y.J., J.C., H.Y.L.); Department of Radiology and Medical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, South Korea (G.L.); Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (S.H.M., J.Y.C.); Industrial Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea (J.H.K.); Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, South Korea (J.C.); and Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea (H.Y.L.)
| | | | | | | | | | | | | |
Collapse
|
5
|
Balbi M, Mazzaschi G, Leo L, Moron Dalla Tor L, Milanese G, Marrocchio C, Silva M, Mura R, Favia P, Bocchialini G, Trentini F, Minari R, Ampollini L, Quaini F, Roti G, Tiseo M, Sverzellati N. Longitudinal Changes of CT-radiomic and Systemic Inflammatory Features Predict Survival in Advanced Non-Small Cell Lung Cancer Patients Treated With Immune Checkpoint Inhibitors. J Thorac Imaging 2025; 40:00005382-990000000-00147. [PMID: 39188157 PMCID: PMC11654449 DOI: 10.1097/rti.0000000000000801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
PURPOSE This study aims to determine whether longitudinal changes in CT radiomic features (RFs) and systemic inflammatory indices outperform single-time-point assessment in predicting survival in advanced non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs). MATERIALS AND METHODS We retrospectively acquired pretreatment (T0) and first disease assessment (T1) RFs and systemic inflammatory indices from a single-center cohort of stage IV NSCLC patients and computed their delta (Δ) variation as [(T1-T0)/T0]. RFs from the primary tumor were selected for building baseline-radiomic (RAD) and Δ-RAD scores using the linear combination of standardized predictors detected by LASSO Cox regression models. Cox models were generated using clinical features alone or combined with baseline and Δ blood parameters and integrated with baseline-RAD and Δ-RAD. All models were 3-fold cross-validated. A prognostic index (PI) of each model was tested to stratify overall survival (OS) through Kaplan-Meier analysis. RESULTS We included 90 ICI-treated NSCLC patients (median age 70 y [IQR=42 to 85], 63 males). Δ-RAD outperformed baseline-RAD for predicting OS [c-index: 0.632 (95%CI: 0.628 to 0.636) vs. 0.605 (95%CI: 0.601 to 0.608) in the test splits]. Integrating longitudinal changes of systemic inflammatory indices and Δ-RAD with clinical data led to the best model performance [Integrated-Δ model, c-index: 0.750 (95% CI: 0.749 to 0.751) in training and 0.718 (95% CI: 0.715 to 0.721) in testing splits]. PI enabled significant OS stratification within all the models ( P -value <0.01), reaching the greatest discriminative ability in Δ models (high-risk group HR up to 7.37, 95% CI: 3.9 to 13.94, P <0.01). CONCLUSION Δ-RAD improved OS prediction compared with single-time-point radiomic in advanced ICI-treated NSCLC. Integrating Δ-RAD with a longitudinal assessment of clinical and laboratory data further improved the prognostic performance.
Collapse
Affiliation(s)
- Maurizio Balbi
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giulia Mazzaschi
- Medical Oncology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ludovica Leo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Gianluca Milanese
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Cristina Marrocchio
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mario Silva
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Rebecca Mura
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Pasquale Favia
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Francesca Trentini
- Medical Oncology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Luca Ampollini
- Thoracic Surgery Unit, University Hospital of Parma
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giovanni Roti
- Translational Hematology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicola Sverzellati
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
6
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
7
|
Yang J, Yang C, Feng J, Zhu F, Zhao Z. Predicting Microwave Ablation Early Efficacy in Pulmonary Malignancies via Δ Radiomics Models. J Comput Assist Tomogr 2024; 48:794-802. [PMID: 38657155 DOI: 10.1097/rct.0000000000001611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
OBJECTIVE This study aimed to explore the value of preoperative and postoperative computed tomography (CT)-based radiomic signatures and Δ radiomic signatures for evaluating the early efficacy of microwave ablation (MWA) for pulmonary malignancies. METHODS In total, 115 patients with pulmonary malignancies who underwent MWA treatment were categorized into response and nonresponse groups according to relevant guidelines and consensus. Quantitative image features of the largest pulmonary malignancies were extracted from CT noncontrast scan images preoperatively (time point 0, TP0) and immediately postoperatively (time point 1, TP1). Critical features were selected from TP0 and TP1 and as Δ radiomics signatures for building radiomics models. In addition, a combined radiomics model (C-RO) was developed by integrating radiomics parameters with clinical risk factors. Prediction performance was assessed using the area under the receiver operating characteristic curve (AUC) and decision curve analysis (DCA). RESULTS The radiomics model using Δ features outperformed the radiomics model using TP0 and TP1 features, with training and validation AUCs of 0.892, 0.808, and 0.787, and 0.705, 0.825, and 0.778, respectively. By combining the TP0, TP1, and Δ features, the logistic regression model exhibited the best performance, with training and validation AUCs of 0.945 and 0.744, respectively. The DCA confirmed the clinical utility of the Δ radiomics model. CONCLUSIONS A combined prediction model, including TP0, TP1, and Δ radiometric features, can be used to evaluate the early efficacy of MWA in pulmonary malignancies.
Collapse
Affiliation(s)
- Jing Yang
- From the School of Medicine, Shaoxing University
| | - Chen Yang
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing
| | - Jianju Feng
- Department of Radiology, Zhuji People's Hospital, Zhuji, Zhejiang, China
| | - Fandong Zhu
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing
| |
Collapse
|
8
|
Zheng J, Xu S, Wang G, Shi Y. Applications of CT-based radiomics for the prediction of immune checkpoint markers and immunotherapeutic outcomes in non-small cell lung cancer. Front Immunol 2024; 15:1434171. [PMID: 39238640 PMCID: PMC11374640 DOI: 10.3389/fimmu.2024.1434171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
In recent years, there has been significant research interest in the field of immunotherapy for non-small cell lung cancer (NSCLC) within the academic community. Given the observed variations in individual responses, despite similarities in histopathologic type, immunohistochemical index, TNM stage, or mutation status, the identification of a reliable biomarker for early prediction of therapeutic responses is of utmost importance. Conventional medical imaging techniques primarily focus on macroscopic tumor monitoring, which may no longer adequately fulfill the requirements of clinical diagnosis and treatment. CT (computerized tomography) or PEF/CT-based radiomics has the potential to investigate the molecular-level biological attributes of tumors, such as PD-1/PD-L1 expression and tumor mutation burden, which offers a novel approach to assess the effectiveness of immunotherapy and forecast patient prognosis. The utilization of cutting-edge radiological imaging techniques, including radiomics, PET/CT, machine learning, and artificial intelligence, demonstrates significant potential in predicting diagnosis, treatment response, immunosuppressive characteristics, and immune-related adverse events. The current review highlights that CT scan-based radiomics is a reliable and feasible way to predict the benefits of immunotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Radiology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang, China
| | - Shuang Xu
- Department of Radiology, Redcliffe Hospital, The University of Queensland, Redcliffe, QLD, Australia
| | - Guoyu Wang
- Department of Radiology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang, China
| | - Yiming Shi
- Department of Radiology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang, China
| |
Collapse
|
9
|
HOU Y, ZHANG T, WANG H. [Advancements in Radiomics for Immunotherapy of Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:637-644. [PMID: 39318257 PMCID: PMC11425675 DOI: 10.3779/j.issn.1009-3419.2024.102.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Indexed: 09/26/2024]
Abstract
Lung cancer is the main cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) being the predominant subtype. At present, immunotherapy represented by immune checkpoint inhibitors (ICIs) of programmed cell death receptor 1 or its ligand has been widely used in the clinical diagnosis and treatment of patients with NSCLC. However, only a few patients can benefit from it, and reliable predictive markers for immunotherapy are lacking. Radiomics is a tool that uses computer software and algorithms to extract a large amount of quantitative information from biomedical images. A large number of studies have confirmed that the radiomic model that predicts the immune efficacy of NSCLC can be used as a new type of immune efficacy predictive marker, which is expected to guide the individualized diagnosis and treatment decisions for patients with lung cancer and has a bright application prospect. This article reviews the research progress of radiomics in predicting the immune therapy response of NSCLC, identifying pseudo-progression and hyperprogression, ICIs-related pneumonia, cachexia risk, and combining with other genomics.
.
Collapse
|
10
|
Chen L, Zhu W, Zhang W, Chen E, Zhou W. Magnetic resonance imaging radiomics-based prediction of severe inflammatory response in locally advanced rectal cancer patients after neoadjuvant radiochemotherapy. Langenbecks Arch Surg 2024; 409:218. [PMID: 39017754 PMCID: PMC11255083 DOI: 10.1007/s00423-024-03416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE To predict severe inflammatory response after neoadjuvant radiochemotherapy in locally advanced rectal cancer (RC) patients using magnetic resonance imaging (MRI) radiomics models. METHODS This retrospective study included patients who underwent radical surgery for RC cancer after neoadjuvant radiochemotherapy between July 2017 and December 2019 at XXX Hospital. MRI radiomics features were extracted from T2WI images before (pre-nRCT-RF) and after (post-nRCT-RF) neoadjuvant radiochemotherapy, and the variation of radiomics features before and after neoadjuvant radiochemotherapy (delta-RF) were calculated. Eight, eight, and five most relevant features were identified for pre-nRCT-RF, post-nRCT-RF, and delta-RF, respectively. RESULTS Eighty-six patients were included and randomized 3:1 to the training and test set (n = 65 and n = 21, respectively). The prediction model based on delta-RF had areas under the curve (AUCs) of 0.80 and 0.85 in the training and test set, respectively. A higher rate of difficult operations was observed in patients with severe inflammation (65.5% vs. 42.9%, P = 0.045). CONCLUSION The prediction model based on MRI delta-RF may be a useful tool for predicting severe inflammatory response after neoadjuvant radiochemotherapy in locally advanced RC patients.
Collapse
Affiliation(s)
- Li Chen
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.
| | - Wenchao Zhu
- Department of Radiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Engeng Chen
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wei Zhou
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Holder AM, Dedeilia A, Sierra-Davidson K, Cohen S, Liu D, Parikh A, Boland GM. Defining clinically useful biomarkers of immune checkpoint inhibitors in solid tumours. Nat Rev Cancer 2024; 24:498-512. [PMID: 38867074 DOI: 10.1038/s41568-024-00705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
Although more than a decade has passed since the approval of immune checkpoint inhibitors (ICIs) for the treatment of melanoma and non-small-cell lung, breast and gastrointestinal cancers, many patients still show limited response. US Food and Drug Administration (FDA)-approved biomarkers include programmed cell death 1 ligand 1 (PDL1) expression, microsatellite status (that is, microsatellite instability-high (MSI-H)) and tumour mutational burden (TMB), but these have limited utility and/or lack standardized testing approaches for pan-cancer applications. Tissue-based analytes (such as tumour gene signatures, tumour antigen presentation or tumour microenvironment profiles) show a correlation with immune response, but equally, these demonstrate limited efficacy, as they represent a single time point and a single spatial assessment. Patient heterogeneity as well as inter- and intra-tumoural differences across different tissue sites and time points represent substantial challenges for static biomarkers. However, dynamic biomarkers such as longitudinal biopsies or novel, less-invasive markers such as blood-based biomarkers, radiomics and the gut microbiome show increasing potential for the dynamic identification of ICI response, and patient-tailored predictors identified through neoadjuvant trials or novel ex vivo tumour models can help to personalize treatment. In this Perspective, we critically assess the multiple new static, dynamic and patient-specific biomarkers, highlight the newest consortia and trial efforts, and provide recommendations for future clinical trials to make meaningful steps forwards in the field.
Collapse
Affiliation(s)
- Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sonia Cohen
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - David Liu
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Aparna Parikh
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Nemoto H, Saito M, Satoh Y, Komiyama T, Marino K, Aoki S, Suzuki H, Sano N, Nonaka H, Watanabe H, Funayama S, Onishi H. Evaluation of the performance of both machine learning models using PET and CT radiomics for predicting recurrence following lung stereotactic body radiation therapy: A single-institutional study. J Appl Clin Med Phys 2024; 25:e14322. [PMID: 38436611 PMCID: PMC11244675 DOI: 10.1002/acm2.14322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/14/2024] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
PURPOSE Predicting recurrence following stereotactic body radiotherapy (SBRT) for non-small cell lung cancer provides important information for the feasibility of the individualized radiotherapy and allows to select the appropriate treatment strategy based on the risk of recurrence. In this study, we evaluated the performance of both machine learning models using positron emission tomography (PET) and computed tomography (CT) radiomic features for predicting recurrence after SBRT. METHODS Planning CT and PET images of 82 non-small cell lung cancer patients who performed SBRT at our hospital were used. First, tumors were delineated on each CT and PET of each patient, and 111 unique radiomic features were extracted, respectively. Next, the 10 features were selected using three different feature selection algorithms, respectively. Recurrence prediction models based on the selected features and four different machine learning algorithms were developed, respectively. Finally, we compared the predictive performance of each model for each recurrence pattern using the mean area under the curve (AUC) calculated following the 0.632+ bootstrap method. RESULTS The highest performance for local recurrence, regional lymph node metastasis, and distant metastasis were observed in models using Support vector machine with PET features (mean AUC = 0.646), Naive Bayes with PET features (mean AUC = 0.611), and Support vector machine with CT features (mean AUC = 0.645), respectively. CONCLUSIONS We comprehensively evaluated the performance of prediction model developed for recurrence following SBRT. The model in this study would provide information to predict the recurrence pattern and assist in making treatment strategies.
Collapse
Affiliation(s)
- Hikaru Nemoto
- Department of Advanced Biomedical ImagingUniversity of YamanashiChuoYamanashiJapan
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Masahide Saito
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Yoko Satoh
- Imaging CenterFujita Medical Innovation Center TokyoTokyoJapan
| | | | - Kan Marino
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Shinichi Aoki
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Hidekazu Suzuki
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Naoki Sano
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Hotaka Nonaka
- Department of RadiologyFuji City General HospitalFujiShizuokaJapan
| | - Hiroaki Watanabe
- Department of RadiologyYamanashi Central HospitalKofuYamanashiJapan
| | - Satoshi Funayama
- Department of RadiologyHamamatsu University school of medicineHamamatsuShizuokaJapan
| | - Hiroshi Onishi
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| |
Collapse
|
13
|
Ligero M, Gielen B, Navarro V, Cresta Morgado P, Prior O, Dienstmann R, Nuciforo P, Trebeschi S, Beets-Tan R, Sala E, Garralda E, Perez-Lopez R. A whirl of radiomics-based biomarkers in cancer immunotherapy, why is large scale validation still lacking? NPJ Precis Oncol 2024; 8:42. [PMID: 38383736 PMCID: PMC10881558 DOI: 10.1038/s41698-024-00534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
The search for understanding immunotherapy response has sparked interest in diverse areas of oncology, with artificial intelligence (AI) and radiomics emerging as promising tools, capable of gathering large amounts of information to identify suitable patients for treatment. The application of AI in radiology has grown, driven by the hypothesis that radiology images capture tumor phenotypes and thus could provide valuable insights into immunotherapy response likelihood. However, despite the rapid growth of studies, no algorithms in the field have reached clinical implementation, mainly due to the lack of standardized methods, hampering study comparisons and reproducibility across different datasets. In this review, we performed a comprehensive assessment of published data to identify sources of variability in radiomics study design that hinder the comparison of the different model performance and, therefore, clinical implementation. Subsequently, we conducted a use-case meta-analysis using homogenous studies to assess the overall performance of radiomics in estimating programmed death-ligand 1 (PD-L1) expression. Our findings indicate that, despite numerous attempts to predict immunotherapy response, only a limited number of studies share comparable methodologies and report sufficient data about cohorts and methods to be suitable for meta-analysis. Nevertheless, although only a few studies meet these criteria, their promising results underscore the importance of ongoing standardization and benchmarking efforts. This review highlights the importance of uniformity in study design and reporting. Such standardization is crucial to enable meaningful comparisons and demonstrate the validity of biomarkers across diverse populations, facilitating their implementation into the immunotherapy patient selection process.
Collapse
Affiliation(s)
- Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Bente Gielen
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Victor Navarro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pablo Cresta Morgado
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
- Prostate Cancer Translational Research Group, Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Olivia Prior
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Regina Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Evis Sala
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
14
|
Chiu HY, Wang TW, Hsu MS, Chao HS, Liao CY, Lu CF, Wu YT, Chen YM. Progress in Serial Imaging for Prognostic Stratification of Lung Cancer Patients Receiving Immunotherapy: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:615. [PMID: 38339369 PMCID: PMC10854498 DOI: 10.3390/cancers16030615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Immunotherapy, particularly with checkpoint inhibitors, has revolutionized non-small cell lung cancer treatment. Enhancing the selection of potential responders is crucial, and researchers are exploring predictive biomarkers. Delta radiomics, a derivative of radiomics, holds promise in this regard. For this study, a meta-analysis was conducted that adhered to PRISMA guidelines, searching PubMed, Embase, Web of Science, and the Cochrane Library for studies on the use of delta radiomics in stratifying lung cancer patients receiving immunotherapy. Out of 223 initially collected studies, 10 were included for qualitative synthesis. Stratifying patients using radiomic models, the pooled analysis reveals a predictive power with an area under the curve of 0.81 (95% CI 0.76-0.86, p < 0.001) for 6-month response, a pooled hazard ratio of 4.77 (95% CI 2.70-8.43, p < 0.001) for progression-free survival, and 2.15 (95% CI 1.73-2.66, p < 0.001) for overall survival at 6 months. Radiomics emerges as a potential prognostic predictor for lung cancer, but further research is needed to compare traditional radiomics and deep-learning radiomics.
Collapse
Affiliation(s)
- Hwa-Yen Chiu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-Y.C.); (T.-W.W.); (M.-S.H.); (H.-S.C.)
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Internal Medicine, Taipei Veterans General Hospital, Hsinchu Branch, Chutong 310, Taiwan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Ting-Wei Wang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-Y.C.); (T.-W.W.); (M.-S.H.); (H.-S.C.)
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ming-Sheng Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-Y.C.); (T.-W.W.); (M.-S.H.); (H.-S.C.)
| | - Heng-Shen Chao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (H.-Y.C.); (T.-W.W.); (M.-S.H.); (H.-S.C.)
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chien-Yi Liao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (C.-Y.L.); (C.-F.L.)
| | - Chia-Feng Lu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (C.-Y.L.); (C.-F.L.)
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yuh-Ming Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| |
Collapse
|
15
|
Schroeder KE, Acharya L, Mani H, Furqan M, Sieren JC. Radiomic biomarkers from chest computed tomography are assistive in immunotherapy response prediction for non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:1023-1033. [PMID: 37323179 PMCID: PMC10261870 DOI: 10.21037/tlcr-22-763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/12/2023] [Indexed: 06/17/2023]
Abstract
Background Immunotherapies, such as programmed death 1/programmed death ligand 1 (PD-1/PD-L1) antibodies have been shown to improve overall and progression-free survival (PFS) in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC). However, not all patients derive a meaningful clinical benefit. Additionally, patients receiving anti-PD-1/PD-L1 therapy can experience immune-related adverse events (irAEs). Clinically significant irAEs may require temporary pause or discontinuation of treatment. Having a tool to identify patients who may not benefit and/or are at risk for developing severe irAEs from immunotherapy will aid in an informed decision-making process for the patients and their physicians. Methods Computed tomography (CT) scans and clinical data were retrospectively collected for this study to develop three prediction models using (I) radiomic features, (II) clinical features, and (III) radiomic and clinical features combined. Each subject had 6 clinical features and 849 radiomic features extracted. Selected features were run through an artificial neural network (NN) trained on 70% of the cohort, maintaining the case and control ratio. The NN was assessed by calculating the area-under-the-receiver-operating-characteristic curve (AUC-ROC), area-under-the-precision-recall curve (AUC-PR), sensitivity, and specificity. Results A cohort of 132 subjects, of which 43 (33%) had a PFS ≤90 days and 89 (67%) of which had a PFS >90 days was used to develop the prediction models. The radiomic model was able to predict progression-free survival with a training AUC-ROC of 87% and testing AUC-ROC, sensitivity, and specificity of 83%, 75%, and 81%, respectively. In this cohort, the clinical and radiomic combined features did add a slight increase in the specificity (85%) but with a decrease in sensitivity (75%) and AUC-ROC (81%). Conclusions Whole lung segmentation and feature extraction can identify those that would see a benefit from anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
| | - Luna Acharya
- Department of Internal Medicine, Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa, Iowa City, IA, USA
| | - Hariharasudan Mani
- Department of Internal Medicine, Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa, Iowa City, IA, USA
| | - Muhammad Furqan
- Department of Internal Medicine, Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Jessica C. Sieren
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
16
|
Li K, Li Y, Wang Z, Huang C, Sun S, Liu X, Fan W, Zhang G, Li X. Delta-radiomics based on CT predicts pathologic complete response in ESCC treated with neoadjuvant immunochemotherapy and surgery. Front Oncol 2023; 13:1131883. [PMID: 37251937 PMCID: PMC10213404 DOI: 10.3389/fonc.2023.1131883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Background and purpose Unnecessary surgery can be avoided, and more appropriate treatment plans can be developed for patients if the efficacy of neoadjuvant immunochemotherapy for esophageal cancer (EC) can be predicted before surgery. The purpose of this study was to evaluate the ability of machine learning models based on delta features of immunochemotherapy CT images to predict the efficacy of neoadjuvant immunochemotherapy in patients with esophageal squamous cell carcinoma (ESCC) compared with machine learning models based solely on postimmunochemotherapy CT images. Materials and methods A total of 95 patients were enrolled in our study and randomly divided into a training group (n = 66) and test group (n = 29). We extracted preimmunochemotherapy radiomics features from preimmunochemotherapy enhanced CT images in the preimmunochemotherapy group (pregroup) and postimmunochemotherapy radiomics features from postimmunochemotherapy enhanced CT images in the postimmunochemotherapy group (postgroup). We then subtracted the preimmunochemotherapy features from the postimmunochemotherapy features and obtained a series of new radiomics features that were included in the delta group. The reduction and screening of radiomics features were carried out by using the Mann-Whitney U test and LASSO regression. Five pairwise machine learning models were established, the performance of which was evaluated by receiver operating characteristic (ROC) curve and decision curve analyses. Results The radiomics signature of the postgroup was composed of 6 radiomics features; that of the delta-group was composed of 8 radiomics features. The area under the ROC curve (AUC) of the machine learning model with the best efficacy was 0.824 (0.706-0.917) in the postgroup and 0.848 (0.765-0.917) in the delta group. The decision curve showed that our machine learning models had good predictive performance. The delta group performed better than the postgroup for each corresponding machine learning model. Conclusion We established machine learning models that have good predictive efficacy and can provide certain reference values for clinical treatment decision-making. Our machine learning models based on delta imaging features performed better than those based on single time-stage postimmunochemotherapy imaging features.
Collapse
Affiliation(s)
- Kaiyuan Li
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuetong Li
- Clinical Medical College, Henan University, Henan, Kaifeng, China
| | - Zhulin Wang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunyao Huang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shaowu Sun
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xu Liu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenbo Fan
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guoqing Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangnan Li
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
17
|
Pabst L, Lopes S, Bertrand B, Creusot Q, Kotovskaya M, Pencreach E, Beau-Faller M, Mascaux C. Prognostic and Predictive Biomarkers in the Era of Immunotherapy for Lung Cancer. Int J Mol Sci 2023; 24:ijms24087577. [PMID: 37108738 PMCID: PMC10145126 DOI: 10.3390/ijms24087577] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The therapeutic algorithm of lung cancer has recently been revolutionized by the emergence of immune checkpoint inhibitors. However, an objective and durable response rate remains low with those recent therapies and some patients even experience severe adverse events. Prognostic and predictive biomarkers are therefore needed in order to select patients who will respond. Nowadays, the only validated biomarker is the PD-L1 expression, but its predictive value remains imperfect, and it does not offer any certainty of a sustained response to treatment. With recent progresses in molecular biology, genome sequencing techniques, and the understanding of the immune microenvironment of the tumor and its host, new molecular features have been highlighted. There are evidence in favor of the positive predictive value of the tumor mutational burden, as an example. From the expression of molecular interactions within tumor cells to biomarkers circulating in peripheral blood, many markers have been identified as associated with the response to immunotherapy. In this review, we would like to summarize the latest knowledge about predictive and prognostic biomarkers of immune checkpoint inhibitors efficacy in order to go further in the field of precision immuno-oncology.
Collapse
Affiliation(s)
- Lucile Pabst
- Pulmonology Department, University Hospital of Strasbourg, 67000 Strasbourg, France
| | - Sébastien Lopes
- Pharmacy Department, University Hospital of Strasbourg, 67000 Strasbourg, France
| | - Basil Bertrand
- Pulmonology Department, University Hospital of Strasbourg, 67000 Strasbourg, France
- Laboratory Streinth (STress REsponse and INnovative THerapy against Cancer), Inserm UMR_S 1113, IRFAC, Université de Strasbourg, ITI InnoVec, 67000 Strasbourg, France
| | - Quentin Creusot
- Pulmonology Department, University Hospital of Strasbourg, 67000 Strasbourg, France
- Laboratory Streinth (STress REsponse and INnovative THerapy against Cancer), Inserm UMR_S 1113, IRFAC, Université de Strasbourg, ITI InnoVec, 67000 Strasbourg, France
| | - Maria Kotovskaya
- Pulmonology Department, University Hospital of Strasbourg, 67000 Strasbourg, France
- Laboratory Streinth (STress REsponse and INnovative THerapy against Cancer), Inserm UMR_S 1113, IRFAC, Université de Strasbourg, ITI InnoVec, 67000 Strasbourg, France
| | - Erwan Pencreach
- Laboratory Streinth (STress REsponse and INnovative THerapy against Cancer), Inserm UMR_S 1113, IRFAC, Université de Strasbourg, ITI InnoVec, 67000 Strasbourg, France
- Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 67000 Strasbourg, France
| | - Michèle Beau-Faller
- Laboratory Streinth (STress REsponse and INnovative THerapy against Cancer), Inserm UMR_S 1113, IRFAC, Université de Strasbourg, ITI InnoVec, 67000 Strasbourg, France
- Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 67000 Strasbourg, France
| | - Céline Mascaux
- Pulmonology Department, University Hospital of Strasbourg, 67000 Strasbourg, France
- Laboratory Streinth (STress REsponse and INnovative THerapy against Cancer), Inserm UMR_S 1113, IRFAC, Université de Strasbourg, ITI InnoVec, 67000 Strasbourg, France
| |
Collapse
|
18
|
Cousin F, Louis T, Dheur S, Aboubakar F, Ghaye B, Occhipinti M, Vos W, Bottari F, Paulus A, Sibille A, Vaillant F, Duysinx B, Guiot J, Hustinx R. Radiomics and Delta-Radiomics Signatures to Predict Response and Survival in Patients with Non-Small-Cell Lung Cancer Treated with Immune Checkpoint Inhibitors. Cancers (Basel) 2023; 15:cancers15071968. [PMID: 37046629 PMCID: PMC10093736 DOI: 10.3390/cancers15071968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The aim of our study was to determine the potential role of CT-based radiomics in predicting treatment response and survival in patients with advanced NSCLC treated with immune checkpoint inhibitors. We retrospectively included 188 patients with NSCLC treated with PD-1/PD-L1 inhibitors from two independent centers. Radiomics analysis was performed on pre-treatment contrast-enhanced CT. A delta-radiomics analysis was also conducted on a subset of 160 patients who underwent a follow-up contrast-enhanced CT after 2 to 4 treatment cycles. Linear and random forest (RF) models were tested to predict response at 6 months and overall survival. Models based on clinical parameters only and combined clinical and radiomics models were also tested and compared to the radiomics and delta-radiomics models. The RF delta-radiomics model showed the best performance for response prediction with an AUC of 0.8 (95% CI: 0.65−0.95) on the external test dataset. The Cox regression delta-radiomics model was the most accurate at predicting survival with a concordance index of 0.68 (95% CI: 0.56−0.80) (p = 0.02). The baseline CT radiomics signatures did not show any significant results for treatment response prediction or survival. In conclusion, our results demonstrated the ability of a CT-based delta-radiomics signature to identify early on patients with NSCLC who were more likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- François Cousin
- Department of Nuclear Medicine and Oncological Imaging, University Hospital (CHU) of Liège, 4000 Liège, Belgium
- Correspondence: ; Tel.: +32-475972109
| | - Thomas Louis
- Radiomics (Oncoradiomics SA), 4000 Liège, Belgium
| | - Sophie Dheur
- Department of Radiology, University Hospital (CHU) of Liège, 4000 Liège, Belgium
| | - Frank Aboubakar
- Department of Pulmonology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Bruxelles, Belgium
- Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Bruxelles, Belgium
| | - Benoit Ghaye
- Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Bruxelles, Belgium
- Department of Radiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Bruxelles, Belgium
| | | | - Wim Vos
- Radiomics (Oncoradiomics SA), 4000 Liège, Belgium
| | | | - Astrid Paulus
- Department of Respiratory Medicine, University Hospital (CHU) of Liège, 4000 Liège, Belgium
| | - Anne Sibille
- Department of Respiratory Medicine, University Hospital (CHU) of Liège, 4000 Liège, Belgium
| | - Frédérique Vaillant
- Department of Respiratory Medicine, University Hospital (CHU) of Liège, 4000 Liège, Belgium
| | - Bernard Duysinx
- Department of Respiratory Medicine, University Hospital (CHU) of Liège, 4000 Liège, Belgium
| | - Julien Guiot
- Department of Respiratory Medicine, University Hospital (CHU) of Liège, 4000 Liège, Belgium
| | - Roland Hustinx
- Department of Nuclear Medicine and Oncological Imaging, University Hospital (CHU) of Liège, 4000 Liège, Belgium
- GIGA-CRC In Vivo Imaging, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
19
|
Farina B, Guerra ADR, Bermejo-Peláez D, Miras CP, Peral AA, Madueño GG, Jaime JC, Vilalta-Lacarra A, Pérez JR, Muñoz-Barrutia A, Peces-Barba GR, Maceiras LS, Gil-Bazo I, Gómez MD, Ledesma-Carbayo MJ. Integration of longitudinal deep-radiomics and clinical data improves the prediction of durable benefits to anti-PD-1/PD-L1 immunotherapy in advanced NSCLC patients. J Transl Med 2023; 21:174. [PMID: 36872371 PMCID: PMC9985838 DOI: 10.1186/s12967-023-04004-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/16/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Identifying predictive non-invasive biomarkers of immunotherapy response is crucial to avoid premature treatment interruptions or ineffective prolongation. Our aim was to develop a non-invasive biomarker for predicting immunotherapy clinical durable benefit, based on the integration of radiomics and clinical data monitored through early anti-PD-1/PD-L1 monoclonal antibodies treatment in patients with advanced non-small cell lung cancer (NSCLC). METHODS In this study, 264 patients with pathologically confirmed stage IV NSCLC treated with immunotherapy were retrospectively collected from two institutions. The cohort was randomly divided into a training (n = 221) and an independent test set (n = 43), ensuring the balanced availability of baseline and follow-up data for each patient. Clinical data corresponding to the start of treatment was retrieved from electronic patient records, and blood test variables after the first and third cycles of immunotherapy were also collected. Additionally, traditional radiomics and deep-radiomics features were extracted from the primary tumors of the computed tomography (CT) scans before treatment and during patient follow-up. Random Forest was used to implementing baseline and longitudinal models using clinical and radiomics data separately, and then an ensemble model was built integrating both sources of information. RESULTS The integration of longitudinal clinical and deep-radiomics data significantly improved clinical durable benefit prediction at 6 and 9 months after treatment in the independent test set, achieving an area under the receiver operating characteristic curve of 0.824 (95% CI: [0.658,0.953]) and 0.753 (95% CI: [0.549,0.931]). The Kaplan-Meier survival analysis showed that, for both endpoints, the signatures significantly stratified high- and low-risk patients (p-value< 0.05) and were significantly correlated with progression-free survival (PFS6 model: C-index 0.723, p-value = 0.004; PFS9 model: C-index 0.685, p-value = 0.030) and overall survival (PFS6 models: C-index 0.768, p-value = 0.002; PFS9 model: C-index 0.736, p-value = 0.023). CONCLUSIONS Integrating multidimensional and longitudinal data improved clinical durable benefit prediction to immunotherapy treatment of advanced non-small cell lung cancer patients. The selection of effective treatment and the appropriate evaluation of clinical benefit are important for better managing cancer patients with prolonged survival and preserving quality of life.
Collapse
Affiliation(s)
- Benito Farina
- Biomedical Image Technologies, ETSI Telecomunicación, Universidad Politécnica de Madrid, 28040, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| | - Ana Delia Ramos Guerra
- Biomedical Image Technologies, ETSI Telecomunicación, Universidad Politécnica de Madrid, 28040, Madrid, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - David Bermejo-Peláez
- Biomedical Image Technologies, ETSI Telecomunicación, Universidad Politécnica de Madrid, 28040, Madrid, Spain
| | | | | | | | | | | | | | - Arrate Muñoz-Barrutia
- Bioengineering Department, Universidad Carlos III de Madrid, 28911, Leganés, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, 28007, Madrid, Spain
| | - German R Peces-Barba
- Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Pamplona, Spain
| | - Luis Seijo Maceiras
- Clínica Universidad de Navarra, 28027, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Pamplona, Spain
| | - Ignacio Gil-Bazo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008, Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, 31008, Pamplona, Spain.,Program in Solid Tumors, Center for Applied Medical Research (CIMA), 31008, Pamplona, Spain.,Navarra Institute for Health Research, IdiSNA, 31008, Pamplona, Spain.,Department of Oncology, Fundación Instituto Valenciano de Oncología (FIVO), 46009, Valencia, Spain
| | | | - María J Ledesma-Carbayo
- Biomedical Image Technologies, ETSI Telecomunicación, Universidad Politécnica de Madrid, 28040, Madrid, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
20
|
Staal FC, Taghavi M, Hong EK, Tissier R, van Treijen M, Heeres BC, van der Zee D, Tesselaar ME, Beets-Tan RG, Maas M. CT-based radiomics to distinguish progressive from stable neuroendocrine liver metastases treated with somatostatin analogues: an explorative study. Acta Radiol 2023; 64:1062-1070. [PMID: 35702011 DOI: 10.1177/02841851221106598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Accurate response evaluation in patients with neuroendocrine liver metastases (NELM) remains a challenge. Radiomics has shown promising results regarding response assessment. PURPOSE To differentiate progressive (PD) from stable disease (SD) with radiomics in patients with NELM undergoing somatostatin analogue (SSA) treatment. MATERIAL AND METHODS A total of 46 patients with histologically confirmed gastroenteropancreatic neuroendocrine tumors (GEP-NET) with ≥1 NELM and ≥2 computed tomography (CT) scans were included. Response was assessed with Response Evaluation Criteria in Solid Tumors (RECIST1.1). Hepatic target lesions were manually delineated and analyzed with radiomics. Radiomics features were extracted from each NELM on both arterial-phase (AP) and portal-venous-phase (PVP) CT. Multiple instance learning with regularized logistic regression via LASSO penalization (with threefold cross-validation) was used to classify response. Three models were computed: (i) AP model; (ii) PVP model; and (iii) AP + PVP model for a lesion-based and patient-based outcome. Next, clinical features were added to each model. RESULTS In total, 19 (40%) patients had PD. Median follow-up was 13 months (range 1-50 months). Radiomics models could not accurately classify response (area under the curve 0.44-0.60). Adding clinical variables to the radiomics models did not significantly improve the performance of any model. CONCLUSION Radiomics features were not able to accurately classify response of NELM on surveillance CT scans during SSA treatment.
Collapse
Affiliation(s)
- Femke Cr Staal
- Department of Radiology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, 5211Maastricht University Medical Centre, Maastricht, The Netherlands
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, 1228Netherlands Cancer Institute Amsterdam/University Medical Center Utrecht, Utrecht, The Netherlands
| | - M Taghavi
- Department of Radiology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, 5211Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Eun K Hong
- Department of Radiology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, 5211Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Radiology, 26725Seoul National University Hospital, Seoul, Republic of Korea
| | - Renaud Tissier
- Biostatistics Center, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark van Treijen
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, 1228Netherlands Cancer Institute Amsterdam/University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Endocrine Oncology, 8124University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Birthe C Heeres
- Department of Radiology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Margot Et Tesselaar
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, 1228Netherlands Cancer Institute Amsterdam/University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Oncology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Regina Gh Beets-Tan
- Department of Radiology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, 5211Maastricht University Medical Centre, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Monique Maas
- Department of Radiology, 1228The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Association of Multi-Phasic MR-Based Radiomic and Dosimetric Features with Treatment Response in Unresectable Hepatocellular Carcinoma Patients following Novel Sequential TACE-SBRT-Immunotherapy. Cancers (Basel) 2023; 15:cancers15041105. [PMID: 36831445 PMCID: PMC9954441 DOI: 10.3390/cancers15041105] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
This study aims to investigate the association of pre-treatment multi-phasic MR-based radiomics and dosimetric features with treatment response to a novel sequential trans-arterial chemoembolization (TACE) plus stereotactic body radiotherapy (SBRT) plus immunotherapy regimen in unresectable Hepatocellular Carcinoma (HCC) sub-population. Twenty-six patients with unresectable HCC were retrospectively analyzed. Radiomic features were extracted from 42 lesions on arterial phase (AP) and portal-venous phase (PVP) MR images. Delta-phase (DeltaP) radiomic features were calculated as AP-to-PVP ratio. Dosimetric data of the tumor was extracted from dose-volume-histograms. A two-sided independent Mann-Whitney U test was used to assess the clinical association of each feature, and the classification performance of each significant independent feature was assessed using logistic regression. For the 3-month timepoint, four DeltaP-derived radiomics that characterize the temporal change in intratumoral randomness and uniformity were the only contributors to the treatment response association (p-value = 0.038-0.063, AUC = 0.690-0.766). For the 6-month timepoint, DeltaP-derived radiomic features (n = 4) maintained strong clinical associations with the treatment response (p-value = 0.047-0.070, AUC = 0.699-0.788), additional AP-derived radiomic features (n = 4) that reflect baseline tumoral arterial-enhanced signal pattern and tumor morphology (n = 1) that denotes initial tumor burden were shown to have strong associations with treatment response (p-value = 0.028-0.074, AUC = 0.719-0.773). This pilot study successfully demonstrated associations of pre-treatment multi-phasic MR-based radiomics with tumor response to the novel treatment regimen.
Collapse
|
22
|
Ge G, Zhang J. Feature selection methods and predictive models in CT lung cancer radiomics. J Appl Clin Med Phys 2023; 24:e13869. [PMID: 36527376 PMCID: PMC9860004 DOI: 10.1002/acm2.13869] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/31/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Radiomics is a technique that extracts quantitative features from medical images using data-characterization algorithms. Radiomic features can be used to identify tissue characteristics and radiologic phenotyping that is not observable by clinicians. A typical workflow for a radiomics study includes cohort selection, radiomic feature extraction, feature and predictive model selection, and model training and validation. While there has been increasing attention given to radiomic feature extraction, standardization, and reproducibility, currently, there is a lack of rigorous evaluation of feature selection methods and predictive models. Herein, we review the published radiomics investigations in CT lung cancer and provide an overview of the commonly used radiomic feature selection methods and predictive models. We also compare limitations of various methods in clinical applications and present sources of uncertainty associated with those methods. This review is expected to help raise awareness of the impact of radiomic feature and model selection methods on the integrity of radiomics studies.
Collapse
Affiliation(s)
- Gary Ge
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Jie Zhang
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
23
|
Zhu Z, Chen M, Hu G, Pan Z, Han W, Tan W, Zhou Z, Wang M, Mao L, Li X, Sui X, Song L, Xu Y, Song W, Yu Y, Jin Z. A pre-treatment CT-based weighted radiomic approach combined with clinical characteristics to predict durable clinical benefits of immunotherapy in advanced lung cancer. Eur Radiol 2022; 33:3918-3930. [PMID: 36515714 PMCID: PMC9748402 DOI: 10.1007/s00330-022-09337-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/08/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To develop a pre-treatment CT-based predictive model to anticipate inoperable lung cancer patients' progression-free survival (PFS) to immunotherapy. METHODS This single-center retrospective study developed and cross-validated a radiomic model in 185 patients and tested it in 48 patients. The binary endpoint is the durable clinical benefit (DCB, PFS ≥ 6 months) and non-DCB (NDCB, PFS < 6 months). Radiomic features were extracted from multiple intrapulmonary lesions and weighted by an attention-based multiple-instance learning model. Aggregated features were then selected through L2-regularized ridge regression. Five machine-learning classifiers were conducted to build predictive models using radiomic and clinical features alone and then together. Lastly, the predictive value of the model with the best performance was validated by Kaplan-Meier survival analysis. RESULTS The predictive models based on the weighted radiomic approach showed superior performance across all classifiers (AUCs: 0.75-0.82) compared with the largest lesion approach (AUCs: 0.70-0.78) and the average sum approach (AUCs: 0.64-0.80). Among them, the logistic regression model yielded the most balanced performance (AUC = 0.87 [95%CI 0.84-0.89], 0.75 [0.68-0.82], 0.80 [0.68-0.92] in the training, validation, and test cohort respectively). The addition of five clinical characteristics significantly enhanced the performance of radiomic-only model (train: AUC 0.91 [0.89-0.93], p = .042; validation: AUC 0.86 [0.80-0.91], p = .011; test: AUC 0.86 [0.76-0.96], p = .026). Kaplan-Meier analysis of the radiomic-based predictive models showed a clear stratification between classifier-predicted DCB versus NDCB for PFS (HR = 2.40-2.95, p < 0.05). CONCLUSIONS The adoption of weighted radiomic features from multiple intrapulmonary lesions has the potential to predict long-term PFS benefits for patients who are candidates for PD-1/PD-L1 immunotherapies. KEY POINTS • Weighted radiomic-based model derived from multiple intrapulmonary lesions on pre-treatment CT images has the potential to predict durable clinical benefits of immunotherapy in lung cancer. • Early line immunotherapy is associated with longer progression-free survival in advanced lung cancer.
Collapse
Affiliation(s)
- Zhenchen Zhu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Ge Hu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Zhengsong Pan
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Institute of Basic Medicine Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005 China
| | - Weixiong Tan
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Zhen Zhou
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Li Mao
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Xiuli Li
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Xin Sui
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yizhou Yu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| |
Collapse
|
24
|
Kim N, Lee ES, Won SE, Yang M, Lee AJ, Shin Y, Ko Y, Pyo J, Park HJ, Kim KW. Evolution of Radiological Treatment Response Assessments for Cancer Immunotherapy: From iRECIST to Radiomics and Artificial Intelligence. Korean J Radiol 2022; 23:1089-1101. [PMID: 36098343 PMCID: PMC9614294 DOI: 10.3348/kjr.2022.0225] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has revolutionized and opened a new paradigm for cancer treatment. In the era of immunotherapy and molecular targeted therapy, precision medicine has gained emphasis, and an early response assessment is a key element of this approach. Treatment response assessment for immunotherapy is challenging for radiologists because of the rapid development of immunotherapeutic agents, from immune checkpoint inhibitors to chimeric antigen receptor-T cells, with which many radiologists may not be familiar, and the atypical responses to therapy, such as pseudoprogression and hyperprogression. Therefore, new response assessment methods such as immune response assessment, functional/molecular imaging biomarkers, and artificial intelligence (including radiomics and machine learning approaches) have been developed and investigated. Radiologists should be aware of recent trends in immunotherapy development and new response assessment methods.
Collapse
Affiliation(s)
- Nari Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Eun Sung Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Eun Won
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Mihyun Yang
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Amy Junghyun Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Youngbin Shin
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yousun Ko
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Junhee Pyo
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung Won Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
25
|
Sun R, Lerousseau M, Briend-Diop J, Routier E, Roy S, Henry T, Ka K, Jiang R, Temar N, Carré A, Laville A, Hamaoui A, Laurent PA, Rouyar A, Robert C, Robert C, Deutsch E. Radiomics to evaluate interlesion heterogeneity and to predict lesion response and patient outcomes using a validated signature of CD8 cells in advanced melanoma patients treated with anti-PD1 immunotherapy. J Immunother Cancer 2022; 10:jitc-2022-004867. [PMID: 36307149 PMCID: PMC9621183 DOI: 10.1136/jitc-2022-004867] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
PURPOSE While there is still a significant need to identify potential biomarkers that can predict which patients are most likely to respond to immunotherapy treatments, radiomic approaches have shown promising results. The objectives of this study were to evaluate whether a previously validated radiomics signature of CD8 T-cells could predict progressions at a lesion level and whether the spatial heterogeneity of this radiomics score could be used at a patient level to assess the clinical response and survival of melanoma patients. METHODS Clinical data from patients with advanced melanoma treated in our center with immunotherapy were retrieved. Radiomic features were extracted and the CD8 radiomics signature was applied. A progressive lesion was defined by an increase in lesion size of 20% or more. Dispersion metrics of the radiomics signature were estimated to evaluate the impact of interlesion heterogeneity on patient's response. Fine-tuned cut-offs for predicting overall survival were evaluated after splitting data into training and test sets. RESULTS A total of 136 patients were included in this study, with 1120 segmented lesions at baseline, and 1052 lesions at first evaluation. A low CD8 radiomics score at baseline was associated with a significantly higher risk of lesion progression (AUC=0.55, p=0.0091), especially for lesions larger than >1 mL (AUC=0.59 overall, p=0.0035, with AUC=0.75, p=0.002 for subcutaneous lesions, AUC=0.68, p=0.01, for liver lesions and AUC=0.62, p=0.03 for nodes). The least infiltrated lesion according to the radiomics score of CD8 T-cells was positively associated with overall survival (training set HR=0.31, p=0.00062, test set HR=0.28, p=0.016), which remained significant in a multivariate analysis including clinical and biological variables. CONCLUSIONS These results confirm the predictive value at a lesion level of the biologically inspired CD8 radiomics score in melanoma patients treated with anti-PD1-based immunotherapy and may be interesting to assess the disease spatial heterogeneity to evaluate the patient prognosis with potential clinical implication such as tumor selection for focal ablative therapies.
Collapse
Affiliation(s)
- Roger Sun
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Marvin Lerousseau
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Jade Briend-Diop
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Emilie Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Gustave Roussy, Villejuif, France
| | - Severine Roy
- Dermatology Unit, Department of Medicine, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Gustave Roussy, Villejuif, France
| | - Théophraste Henry
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France,Imaging Department, Gustave Roussy, Villejuif, France
| | - Kanta Ka
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - Rui Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Nawal Temar
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - Alexandre Carré
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Adrien Laville
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Anthony Hamaoui
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Pierre-Antoine Laurent
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Angela Rouyar
- Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Charlotte Robert
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| | - Caroline Robert
- Dermatology Unit, Department of Medicine, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Gustave Roussy, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France,Université Paris Saclay, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Gustave Roussy, Villejuif, France
| |
Collapse
|
26
|
Xie D, Xu F, Zhu W, Pu C, Huang S, Lou K, Wu Y, Huang D, He C, Hu H. Delta radiomics model for the prediction of progression-free survival time in advanced non-small-cell lung cancer patients after immunotherapy. Front Oncol 2022; 12:990608. [PMID: 36276082 PMCID: PMC9583844 DOI: 10.3389/fonc.2022.990608] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
Objective To assess the validity of pre- and posttreatment computed tomography (CT)-based radiomics signatures and delta radiomics signatures for predicting progression-free survival (PFS) in stage III-IV non-small-cell lung cancer (NSCLC) patients after immune checkpoint inhibitor (ICI) therapy. Methods Quantitative image features of the largest primary lung tumours were extracted on CT-enhanced imaging at baseline (time point 0, TP0) and after the 2nd-3rd immunotherapy cycles (time point 1, TP1). The critical features were selected to construct TP0, TP1 and delta radiomics signatures for the risk stratification of patient survival after ICI treatment. In addition, a prediction model integrating the clinicopathologic risk characteristics and phenotypic signature was developed for the prediction of PFS. Results The C-index of TP0, TP1 and delta radiomics models in the training and validation cohort were 0.64, 0.75, 0.80, and 0.61, 0.68, 0.78, respectively. The delta radiomics score exhibited good accuracy for distinguishing patients with slow and rapid progression to ICI treatment. The predictive accuracy of the combined prediction model was higher than that of the clinical prediction model in both training and validation sets (P<0.05), with a C-index of 0.83 and 0.70, respectively. Additionally, the delta radiomics model (C-index of 0.86) had a higher predictive accuracy compared to PD-L1 expression (C-index of 0.50) (P<0.0001). Conclusions The combined prediction model including clinicopathologic characteristics (tumour anatomical classification and brain metastasis) and the delta radiomics signature could achieve the individualized prediction of PFS in ICIs-treated NSCLC patients.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, China
| | - Fangyi Xu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenchao Zhu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cailing Pu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shaoyu Huang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Ningbo Medical Center LiHuili Hospital, Ningbo, China
| | - Kaihua Lou
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingpin Huang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Hongjie Hu,
| |
Collapse
|
27
|
Ter Maat LS, van Duin IAJ, Elias SG, van Diest PJ, Pluim JPW, Verhoeff JJC, de Jong PA, Leiner T, Veta M, Suijkerbuijk KPM. Imaging to predict checkpoint inhibitor outcomes in cancer. A systematic review. Eur J Cancer 2022; 175:60-76. [PMID: 36096039 DOI: 10.1016/j.ejca.2022.07.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Checkpoint inhibition has radically improved the perspective for patients with metastatic cancer, but predicting who will not respond with high certainty remains difficult. Imaging-derived biomarkers may be able to provide additional insights into the heterogeneity in tumour response between patients. In this systematic review, we aimed to summarise and qualitatively assess the current evidence on imaging biomarkers that predict response and survival in patients treated with checkpoint inhibitors in all cancer types. METHODS PubMed and Embase were searched from database inception to 29th November 2021. Articles eligible for inclusion described baseline imaging predictive factors, radiomics and/or imaging machine learning models for predicting response and survival in patients with any kind of malignancy treated with checkpoint inhibitors. Risk of bias was assessed using the QUIPS and PROBAST tools and data was extracted. RESULTS In total, 119 studies including 15,580 patients were selected. Of these studies, 73 investigated simple imaging factors. 45 studies investigated radiomic features or deep learning models. Predictors of worse survival were (i) higher tumour burden, (ii) presence of liver metastases, (iii) less subcutaneous adipose tissue, (iv) less dense muscle and (v) presence of symptomatic brain metastases. Hazard rate ratios did not exceed 2.00 for any predictor in the larger and higher quality studies. The added value of baseline fluorodeoxyglucose positron emission tomography parameters in predicting response to treatment was limited. Pilot studies of radioactive drug tracer imaging showed promising results. Reports on radiomics were almost unanimously positive, but numerous methodological concerns exist. CONCLUSIONS There is well-supported evidence for several imaging biomarkers that can be used in clinical decision making. Further research, however, is needed into biomarkers that can more accurately identify which patients who will not benefit from checkpoint inhibition. Radiomics and radioactive drug labelling appear to be promising approaches for this purpose.
Collapse
Affiliation(s)
- Laurens S Ter Maat
- Image Science Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Isabella A J van Duin
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Josien P W Pluim
- Image Science Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Medical Image Analysis, Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Joost J C Verhoeff
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Pim A de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Tim Leiner
- Utrecht University, Utrecht, the Netherlands; Department of Radiology, Mayo Clinical, Rochester, MN, USA
| | - Mitko Veta
- Medical Image Analysis, Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
28
|
Kothari G. Role of radiomics in predicting immunotherapy response. J Med Imaging Radiat Oncol 2022; 66:575-591. [PMID: 35581928 PMCID: PMC9323544 DOI: 10.1111/1754-9485.13426] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022]
Abstract
Immunotherapies have revolutionised cancer management. Despite their success, durable responses are limited to a subset of patients. Prediction of immunotherapy response in patients has proven to be difficult due to a lack of robust biomarkers. Routinely collected imaging may offer an additional information source to personalise patient treatment, with advantages over tissue-based biomarkers. Quantitative image analysis or radiomics, which involves the high-throughput extraction of imaging features, has the potential to non-invasively predict cancer histology, outcomes and prognosis. This review evaluates the value of radiomics in patients undergoing immunotherapy, with a summary provided of the performance of radiomics models in predicting immunotherapy response and toxicity, as well as immune correlates. Much of the literature focussed on clinical endpoints and correlates to tissue biomarkers, particularly in lung cancer, while few studies investigated association with immune-related adverse events. Strengths of the studies included more frequent use of clinical trial datasets, homogenous patient cohorts and high-quality diagnostic scans. Limitations of the studies include heterogeneity in study methodology, lack of well-defined homogenous imaging datasets, limited open publishing of imaging datasets, coding and parameters used for radiomics signature development and limited use of external validation datasets. Future research should address the above limitations, as well as further explore the relationship between radiomics and immune-related adverse effects and less well-studied biological correlates such tumour mutational burden, and incorporate known clinical prognostic scores into radiomics models.
Collapse
Affiliation(s)
- Gargi Kothari
- Department of Radiation OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of Oncology, University of MelbournePeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| |
Collapse
|
29
|
Multi-Omics Approaches for the Prediction of Clinical Endpoints after Immunotherapy in Non-Small Cell Lung Cancer: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061237. [PMID: 35740259 PMCID: PMC9219996 DOI: 10.3390/biomedicines10061237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the management of locally advanced and advanced non-small lung cancer (NSCLC). With an improvement in the overall survival (OS) as both first- and second-line treatments, ICIs, and especially programmed-death 1 (PD-1) and programmed-death ligands 1 (PD-L1), changed the landscape of thoracic oncology. The PD-L1 level of expression is commonly accepted as the most used biomarker, with both prognostic and predictive values. However, even in a low expression level of PD-L1, response rates remain significant while a significant number of patients will experience hyperprogression or adverse events. The dentification of such subtypes is thus of paramount importance. While several studies focused mainly on the prediction of the PD-L1 expression status, others aimed directly at the development of prediction/prognostic models. The response to ICIs depends on a complex physiopathological cascade, intricating multiple mechanisms from the molecular to the macroscopic level. With the high-throughput extraction of features, omics approaches aim for the most comprehensive assessment of each patient. In this article, we will review the place of the different biomarkers (clinical, biological, genomics, transcriptomics, proteomics and radiomics), their clinical implementation and discuss the most recent trends projecting on the future steps in prediction modeling in NSCLC patients treated with ICI.
Collapse
|
30
|
Wang Z, Yang C, Han W, Sui X, Zheng F, Xue F, Xu X, Wu P, Chen Y, Gu W, Song W, Jiang J. Quantifying lung cancer heterogeneity using novel CT features: a cross-institute study. Insights Imaging 2022; 13:82. [PMID: 35482262 PMCID: PMC9050978 DOI: 10.1186/s13244-022-01204-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/04/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Radiomics-based image metrics are not used in the clinic despite the rapidly growing literature. We selected eight promising radiomic features and validated their value in decoding lung cancer heterogeneity. METHODS CT images of 236 lung cancer patients were obtained from three different institutes, whereupon radiomic features were extracted according to a standardized procedure. The predictive value for patient long-term prognosis and association with routinely used semantic, genetic (e.g., epidermal growth factor receptor (EGFR)), and histopathological cancer profiles were validated. Feature measurement reproducibility was assessed. RESULTS All eight selected features were robust across repeat scans (intraclass coefficient range: 0.81-0.99), and were associated with at least one of the cancer profiles: prognostic, semantic, genetic, and histopathological. For instance, "kurtosis" had a high predictive value of early death (AUC at first year: 0.70-0.75 in two independent cohorts), negative association with histopathological grade (Spearman's r: - 0.30), and altered expression levels regarding EGFR mutation and semantic characteristics (solid intensity, spiculated shape, juxtapleural location, and pleura tag; all p < 0.05). Combined as a radiomic score, the features had a higher area under curve for predicting 5-year survival (train: 0.855, test: 0.780, external validation: 0.760) than routine characteristics (0.733, 0.622, 0.613, respectively), and a better capability in patient death risk stratification (hazard ratio: 5.828, 95% confidence interval: 2.915-11.561) than histopathological staging and grading. CONCLUSIONS We highlighted the clinical value of radiomic features. Following confirmation, these features may change the way in which we approach CT imaging and improve the individualized care of lung cancer patients.
Collapse
Affiliation(s)
- Zixing Wang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Cuihong Yang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xin Sui
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fuling Zheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fang Xue
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoli Xu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Peng Wu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yali Chen
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wentao Gu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| | - Jingmei Jiang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences / School of Basic Medicine, Peking Union Medical College, Beijing, China.
| |
Collapse
|
31
|
The Role of Radiomics in the Era of Immune Checkpoint Inhibitors: A New Protagonist in the Jungle of Response Criteria. J Clin Med 2022; 11:jcm11061740. [PMID: 35330068 PMCID: PMC8948743 DOI: 10.3390/jcm11061740] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The introduction of immune checkpoint inhibitors has represented a milestone in cancer treatment. Despite PD-L1 expression being the standard biomarker used before the start of therapy, there is still a strict need to identify complementary non-invasive biomarkers in order to better select patients. In this context, radiomics is an emerging approach for examining medical images and clinical data by capturing multiple features hidden from human eye and is potentially able to predict response assessment and survival in the course of immunotherapy. We reviewed the available studies investigating the role of radiomics in cancer patients, focusing on non-small cell lung cancer treated with immune checkpoint inhibitors. Although preliminary research shows encouraging results, different issues need to be solved before radiomics can enter into clinical practice. Abstract Immune checkpoint inhibitors (ICI) have demonstrated encouraging results in terms of durable clinical benefit and survival in several malignancies. Nevertheless, the search to identify an “ideal” biomarker for predicting response to ICI is still far from over. Radiomics is a new translational field of study aiming to extract, by dedicated software, several features from a given medical image, ranging from intensity distribution and spatial heterogeneity to higher-order statistical parameters. Based on these premises, our review aims to summarize the current status of radiomics as a potential predictor of clinical response following immunotherapy treatment. A comprehensive search of PubMed results was conducted. All studies published in English up to and including December 2021 were selected, comprising those that explored computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) for radiomic analyses in the setting of ICI. Several studies have demonstrated the potential applicability of radiomic features in the monitoring of the therapeutic response beyond the traditional morphologic and metabolic criteria, as well as in the prediction of survival or non-invasive assessment of the tumor microenvironment. Nevertheless, important limitations emerge from our review in terms of standardization in feature selection, data sharing, and methods, as well as in external validation. Additionally, there is still need for prospective clinical trials to confirm the potential significant role of radiomics during immunotherapy.
Collapse
|
32
|
Wu M, Zhang Y, Zhang J, Zhang Y, Wang Y, Chen F, Luo Y, He S, Liu Y, Yang Q, Li Y, Wei H, Zhang H, Lu N, Wang S, Guo Y, Ye Z, Liu Y. A Combined-Radiomics Approach of CT Images to Predict Response to Anti-PD-1 Immunotherapy in NSCLC: A Retrospective Multicenter Study. Front Oncol 2022; 11:688679. [PMID: 35083133 PMCID: PMC8784873 DOI: 10.3389/fonc.2021.688679] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Based on non-contrast-enhanced (NCE)/contrast-enhanced (CE) computed tomography (CT) images, we try to identify a combined-radiomics model and evaluate its predictive capacity regarding response to anti-PD1 immunotherapy of patients with non-small-cell lung cancer (NSCLC). METHODS 131 patients with NSCLC undergoing anti-PD1 immunotherapy were retrospectively enrolled from 7 institutions. Using largest lesion (LL) and target lesions (TL) approaches, we performed a radiomics analysis based on pretreatment NCE-CT (NCE-radiomics) and CE-CT images (CE-radiomics), respectively. Meanwhile, a combined-radiomics model based on NCE-CT and CE-CT images was constructed. Finally, we developed their corresponding nomograms incorporating clinical factors. ROC was used to evaluate models' predictive performance in the training and testing set, and a DeLong test was employed to compare the differences between different models. RESULTS For TL approach, both NCE-radiomics and CE-radiomics performed poorly in predicting response to immunotherapy. For LL approach, NCE-radiomics nomograms and CE-radiomics nomograms incorporating with clinical factor of distant metastasis all showed satisfactory results, reflected by the AUCs in the training (AUC=0.84, 95% CI: 0.75-0.92; AUC=0.77, 95% CI: 0.67-0.87) and test sets (AUC=0.78, 95% CI: 0.64-0.92, AUC=0.73, 95% CI: 0.57-0.88), respectively. Compared with the NCE-radiomics nomograms, the combined-radiomics nomogram showed incremental predictive capacity in the training set (AUC=0.85, 95% CI: 0.77-0.92) and test set (AUC=0.81, 95% CI: 0.67-0.94), respectively, but no statistical difference (P=0.86, P=0.79). CONCLUSION Compared with radiomics based on single NCE or CE-CT images, the combined-radiomics model has potential advantages to identify patients with NSCLC most likely to benefit from immunotherapy, and may effectively improve more precise and individualized decision support.
Collapse
Affiliation(s)
- Minghao Wu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanyan Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jianing Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yuwei Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yina Wang
- Department of Medical Oncology, 1st Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Chen
- Department of Radiology, 1st Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yahong Luo
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Shuai He
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yulin Liu
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Yang
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanying Li
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhang
- Department of Radiology, Tianjin Chest Hospital, Tianjin, China
| | - Nian Lu
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Guangzhou, China
| | - Sicong Wang
- Prognostic Diagnosis, GE Healthcare China, Beijing, China
| | - Yan Guo
- Prognostic Diagnosis, GE Healthcare China, Beijing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ying Liu
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
33
|
Exploring Response to Immunotherapy in Non-Small Cell Lung Cancer Using Delta-Radiomics. Cancers (Basel) 2022; 14:cancers14020350. [PMID: 35053513 PMCID: PMC8773717 DOI: 10.3390/cancers14020350] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 01/16/2023] Open
Abstract
Delta-radiomics is a branch of radiomics in which features are confronted after time or after introducing an external factor (such as treatment with chemotherapy or radiotherapy) to extrapolate prognostic data or to monitor a certain condition. Immune checkpoint inhibitors (ICIs) are currently revolutionizing the treatment of non-small cell lung cancer (NSCLC); however, there are still many issues in defining the response to therapy. Contrast-enhanced CT scans of 33 NSCLC patients treated with ICIs were analyzed; altogether, 43 lung lesions were considered. The radiomic features of the lung lesions were extracted from CT scans at baseline and at first reassessment, and their variation (delta, Δ) was calculated by means of the absolute difference and relative reduction. This variation was related to the final response of each lesion to evaluate the predictive ability of the variation itself. Twenty-seven delta features have been identified that are able to discriminate radiologic response to ICIs with statistically significant accuracy. Furthermore, the variation of nine features significantly correlates with pseudo-progression.
Collapse
|
34
|
Kang CY, Duarte SE, Kim HS, Kim E, Park J, Lee AD, Kim Y, Kim L, Cho S, Oh Y, Gim G, Park I, Lee D, Abazeed M, Velichko YS, Chae YK. OUP accepted manuscript. Oncologist 2022; 27:e471-e483. [PMID: 35348765 PMCID: PMC9177100 DOI: 10.1093/oncolo/oyac036] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/14/2022] [Indexed: 11/17/2022] Open
Abstract
The recent, rapid advances in immuno-oncology have revolutionized cancer treatment and spurred further research into tumor biology. Yet, cancer patients respond variably to immunotherapy despite mounting evidence to support its efficacy. Current methods for predicting immunotherapy response are unreliable, as these tests cannot fully account for tumor heterogeneity and microenvironment. An improved method for predicting response to immunotherapy is needed. Recent studies have proposed radiomics—the process of converting medical images into quantitative data (features) that can be processed using machine learning algorithms to identify complex patterns and trends—for predicting response to immunotherapy. Because patients undergo numerous imaging procedures throughout the course of the disease, there exists a wealth of radiological imaging data available for training radiomics models. And because radiomic features reflect cancer biology, such as tumor heterogeneity and microenvironment, these models have enormous potential to predict immunotherapy response more accurately than current methods. Models trained on preexisting biomarkers and/or clinical outcomes have demonstrated potential to improve patient stratification and treatment outcomes. In this review, we discuss current applications of radiomics in oncology, followed by a discussion on recent studies that use radiomics to predict immunotherapy response and toxicity.
Collapse
Affiliation(s)
| | | | - Hye Sung Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eugene Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Alice Daeun Lee
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yeseul Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leeseul Kim
- Department of Internal Medicine, AMITA Health Saint Francis Hospital, Evanston, IL, USA
| | - Sukjoo Cho
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yoojin Oh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gahyun Gim
- Department of Hematology and Oncology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Inae Park
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dongyup Lee
- Department of Physical Medicine and Rehabilitation, Geisinger Health System, Danville, PA, USA
| | - Mohamed Abazeed
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yury S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Young Kwang Chae
- Corresponding author: Young Kwang Chae, Department of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
35
|
Nardone V, Reginelli A, Grassi R, Boldrini L, Vacca G, D'Ippolito E, Annunziata S, Farchione A, Belfiore MP, Desideri I, Cappabianca S. Delta radiomics: a systematic review. Radiol Med 2021; 126:1571-1583. [PMID: 34865190 DOI: 10.1007/s11547-021-01436-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/18/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Radiomics can provide quantitative features from medical imaging that can be correlated with various biological features and clinical endpoints. Delta radiomics, on the other hand, consists in the analysis of feature variation at different acquisition time points, usually before and after therapy. The aim of this study was to provide a systematic review of the different delta radiomics approaches. METHODS Eligible articles were searched in Embase, PubMed, and ScienceDirect using a search string that included free text and/or Medical Subject Headings (MeSH) with three key search terms: "radiomics", "texture", and "delta". Studies were analysed using QUADAS-2 and the RQS tool. RESULTS Forty-eight studies were finally included. The studies were divided into preclinical/methodological (five studies, 10.4%); rectal cancer (six studies, 12.5%); lung cancer (twelve studies, 25%); sarcoma (five studies, 10.4%); prostate cancer (three studies, 6.3%), head and neck cancer (six studies, 12.5%); gastrointestinal malignancies excluding rectum (seven studies, 14.6%), and other disease sites (four studies, 8.3%). The median RQS of all studies was 25% (mean 21% ± 12%), with 13 studies (30.2%) achieving a quality score < 10% and 22 studies (51.2%) < 25%. CONCLUSIONS Delta radiomics shows potential benefit for several clinical endpoints in oncology (differential diagnosis, prognosis and prediction of treatment response, and evaluation of side effects). Nevertheless, the studies included in this systematic review suffer from the bias of overall low quality, so that the conclusions are currently heterogeneous, not robust, and not replicable. Further research with prospective and multicentre studies is needed for the clinical validation of delta radiomics approaches.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy.
| | - Roberta Grassi
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Luca Boldrini
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giovanna Vacca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Emma D'Ippolito
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Salvatore Annunziata
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Alessandra Farchione
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Maria Paola Belfiore
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
36
|
Chen Q, Zhang L, Mo X, You J, Chen L, Fang J, Wang F, Jin Z, Zhang B, Zhang S. Current status and quality of radiomic studies for predicting immunotherapy response and outcome in patients with non-small cell lung cancer: a systematic review and meta-analysis. Eur J Nucl Med Mol Imaging 2021; 49:345-360. [PMID: 34402924 DOI: 10.1007/s00259-021-05509-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE Prediction of immunotherapy response and outcome in patients with non-small cell lung cancer (NSCLC) is challenging due to intratumoral heterogeneity and lack of robust biomarkers. The aim of this study was to systematically evaluate the methodological quality of radiomic studies for predicting immunotherapy response or outcome in patients with NSCLC. METHODS We systematically searched for eligible studies in the PubMed and Web of Science datasets up to April 1, 2021. The methodological quality of included studies was evaluated using the phase classification criteria for image mining studies and the radiomics quality scoring (RQS) tool. A meta-analysis of studies regarding the prediction of immunotherapy response and outcome in patients with NSCLC was performed. RESULTS Fifteen studies were identified with sample sizes ranging from 30 to 228. Seven studies were classified as phase II, and the remaining as discovery science (n = 2), phase 0 (n = 4), phase I (n = 1), and phase III (n = 1). The mean RQS score of all studies was 29.6%, varying from 0 to 68.1%. The pooled diagnostic odds ratio for predicting immunotherapy response in NSCLC using radiomics was 14.99 (95% confidence interval [CI] 8.66-25.95). In addition, radiomics could divide patients into high- and low-risk group with significantly different overall survival (pooled hazard ratio [HR]: 1.96, 95%CI 1.61-2.40, p < 0.001) and progression-free survival (pooled HR: 2.39, 95%CI 1.69-3.38, p < 0.001). CONCLUSIONS Radiomics has potential to noninvasively predict immunotherapy response and outcome in patients with NSCLC. However, it has not yet been implemented as a clinical decision-making tool. Further external validation and evaluation within clinical pathway can facilitate personalized treatment for patients with NSCLC.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Lu Zhang
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaokai Mo
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
| | - Jingjing You
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Luyan Chen
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Jin Fang
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
| | - Zhe Jin
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China.
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China.
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital, Jinan University, No. 613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, People's Republic of China.
- Graduate College, Jinan University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|