1
|
Du Y, Zhang S, Jia X, Zhang X, Li X, Pan L, Li Z, Niu G, Liang T, Guo H. Radiomics Biomarkers to Predict Checkpoint Inhibitor Pneumonitis in Non-small Cell Lung Cancer. Acad Radiol 2024:S1076-6332(24)00703-7. [PMID: 39395887 DOI: 10.1016/j.acra.2024.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/14/2024]
Abstract
RATIONALE AND OBJECTIVES Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of non-small cell lung cancer (NSCLC). However, immune-related adverse events still occur, of which checkpoint inhibitor pneumonitis (CIP) is the most common. We aimed to construct and validate a contrast-enhanced computed tomography-based radiomic nomogram to predict the probability of CIP before ICIs treatment in NSCLC. MATERIALS AND METHODS We retrospectively analyzed 685 patients with NSCLC who were initially treated with ICIs. A total of 186 patients were included in our study, and an additional 52 patients from another hospital were considered for external validation. After radiomics feature extraction and selection, we applied a support vector machine classification model to distinguish CIP and used the probability as a radiomics signature. A radiomics-clinical logistic regression model was built using the filtered clinical parameters and a radiomic signature. Receiver operating characteristic, area under the curve (AUC), calibration curve, and decision curve analysis was used for inter-model comparison. RESULTS The combined radiomics-clinical model constructed using age, interstitial lung disease, emphysema at baseline, and radiomics signature showed an AUC of 0.935, 0.905, and 0.923 for the training, validation, and external validation cohorts, respectively. Compared with the clinical-only (AUC of 0.829, 0.826, and 0.809) and radiomics-only models (0.865, 0.847, and 0.841), the radiomics-clinical displayed better predictive power. CONCLUSION This combined radiomics-clinical model predicted the probability of CIP during ICIs treatment in patients with NSCLC with favorable accuracy and could therefore be used as an effective tool to guide clinical ICIs decisions.
Collapse
Affiliation(s)
- Yonghao Du
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Shuo Zhang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Xiaohui Jia
- Phase I Clinical Trial Ward, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi 710004, PR China (X.J., H.G.)
| | - Xi Zhang
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (X.Z.)
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (X.L.)
| | - Libo Pan
- Department of Radiology, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (L.P.)
| | - Zhihao Li
- Department of Pharmaceuticals Diagnostic, GE Healthcare, Xi'an, Shaanxi 710076, PR China (Z.L.)
| | - Gang Niu
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Ting Liang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Hui Guo
- Phase I Clinical Trial Ward, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi 710004, PR China (X.J., H.G.); Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi 710004, PR China (H.G.); Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China (H.G.); Key Laboratory of Surgical Critical Care and Life Support, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710061, PR China (H.G.).
| |
Collapse
|
2
|
Zhang Y, Cui Y, Liu H, Chang C, Yin Y, Wang R. Prognostic nomogram combining 18F-FDG PET/CT radiomics and clinical data for stage III NSCLC survival prediction. Sci Rep 2024; 14:20557. [PMID: 39231973 PMCID: PMC11374974 DOI: 10.1038/s41598-024-71003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
The aim of this study was to establish and validate the precision of a novel radiomics approach that integrates 18Fluorine-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)-computed tomography (CT) scan data with clinical information to improve the prognostication of survival rates in patients diagnosed with stage III Non-Small Cell Lung Cancer (NSCLC) who are not candidates for surgery. We evaluated pretreatment 18F-FDG PET-CT scans from 156 individuals diagnosed with stage III inoperable NSCLC at Shandong Cancer Hospital. These individuals were divided into two groups: a training set comprising 110 patients and an internal validation set consisting of 46 patients. By employing random forest classifier and cox proportional hazards model , we identified and utilized relevant features to create predictive models and a nomogram. The effectiveness of these models was assessed through the use of the receiver operating characteristics(ROC) curves, Kaplan-Meier (KM) curves, and the application of the nomogram. Our findings showed that the combined model, which integrates both clinical and radiomic data, outperformed those based solely on clinical or radiomic features in predicting 3-year overall survival(OS). Furthermore, calibration plots revealed a high level of agreement between predicted and actual survival times. The research successfully established a predictive radiomics model that integrates 18F-FDG PET/CT imaging with clinical indicators to enhance survival predictions for patients with stage III inoperable NSCLC.
Collapse
Affiliation(s)
- Yalin Zhang
- Department of Radiation Oncology, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Urumuqi, China
| | - Yongbin Cui
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Huiling Liu
- Department of Radiation Oncology, Binzhou People's Hospital, Binzhou, China
| | - Cheng Chang
- Department of Nuclear Medicine, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Urumuqi, China
| | - Yong Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Ruozheng Wang
- Department of Radiation Oncology, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Urumuqi, China.
- Xinjiang Key Laboratory of Oncology, Urumqi, China.
| |
Collapse
|
3
|
McGale JP, Chen DL, Trebeschi S, Farwell MD, Wu AM, Cutler CS, Schwartz LH, Dercle L. Artificial intelligence in immunotherapy PET/SPECT imaging. Eur Radiol 2024; 34:5829-5841. [PMID: 38355986 DOI: 10.1007/s00330-024-10637-3] [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: 09/27/2023] [Revised: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
OBJECTIVE Immunotherapy has dramatically altered the therapeutic landscape for oncology, but more research is needed to identify patients who are likely to achieve durable clinical benefit and those who may develop unacceptable side effects. We investigated the role of artificial intelligence in PET/SPECT-guided approaches for immunotherapy-treated patients. METHODS We performed a scoping review of MEDLINE, CENTRAL, and Embase databases using key terms related to immunotherapy, PET/SPECT imaging, and AI/radiomics through October 12, 2022. RESULTS Of the 217 studies identified in our literature search, 24 relevant articles were selected. The median (interquartile range) sample size of included patient cohorts was 63 (157). Primary tumors of interest were lung (n = 14/24, 58.3%), lymphoma (n = 4/24, 16.7%), or melanoma (n = 4/24, 16.7%). A total of 28 treatment regimens were employed, including anti-PD-(L)1 (n = 13/28, 46.4%) and anti-CTLA-4 (n = 4/28, 14.3%) monoclonal antibodies. Predictive models were built from imaging features using univariate radiomics (n = 7/24, 29.2%), radiomics (n = 12/24, 50.0%), or deep learning (n = 5/24, 20.8%) and were most often used to prognosticate (n = 6/24, 25.0%) or describe tumor phenotype (n = 5/24, 20.8%). Eighteen studies (75.0%) performed AI model validation. CONCLUSION Preliminary results suggest broad potential for the application of AI-guided immunotherapy management after further validation of models on large, prospective, multicenter cohorts. CLINICAL RELEVANCE STATEMENT This scoping review describes how artificial intelligence models are built to make predictions based on medical imaging and explores their application specifically in the PET and SPECT examination of immunotherapy-treated cancers. KEY POINTS • Immunotherapy has drastically altered the cancer treatment landscape but is known to precipitate response patterns that are not accurately accounted for by traditional imaging methods. • There is an unmet need for better tools to not only facilitate in-treatment evaluation but also to predict, a priori, which patients are likely to achieve a good response with a certain treatment as well as those who are likely to develop side effects. • Artificial intelligence applied to PET/SPECT imaging of immunotherapy-treated patients is mainly used to make predictions about prognosis or tumor phenotype and is built from baseline, pre-treatment images. Further testing is required before a true transition to clinical application can be realized.
Collapse
Affiliation(s)
- Jeremy P McGale
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Delphine L Chen
- Department of Molecular Imaging and Therapy, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna M Wu
- Department of Immunology and Theranostics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Cathy S Cutler
- Collider Accelerator Department, Brookhaven National Laboratory, Upton, NY, USA
| | - Lawrence H Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zandberg DP, Zenkin S, Ak M, Mamindla P, Peddagangireddy V, Hsieh R, Anderson JL, Delgoffe GM, Menk A, Skinner HD, Duvvuri U, Ferris RL, Colen RR. Evaluation of radiomics as a predictor of efficacy and the tumor immune microenvironment in anti-PD-1 mAb treated recurrent/metastatic squamous cell carcinoma of the head and neck patients. Head Neck 2024. [PMID: 39080968 DOI: 10.1002/hed.27878] [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: 08/23/2023] [Revised: 03/28/2024] [Accepted: 07/06/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND We retrospectively evaluated radiomics as a predictor of the tumor microenvironment (TME) and efficacy with anti-PD-1 mAb (IO) in R/M HNSCC. METHODS Radiomic feature extraction was performed on pre-treatment CT scans segmented using 3D slicer v4.10.2 and key features were selected using LASSO regularization method to build classification models with XGBoost algorithm by incorporating cross-validation techniques to calculate accuracy, sensitivity, and specificity. Outcome measures evaluated were disease control rate (DCR) by RECIST 1.1, PFS, and OS and hypoxia and CD8 T cells in the TME. RESULTS Radiomics features predicted DCR with accuracy, sensitivity, and specificity of 76%, 73%, and 83%, for OS 77%, 86%, 70%, PFS 82%, 75%, 89%, and in the TME, for high hypoxia 80%, 88%, and 72% and high CD8 T cells 91%, 83%, and 100%, respectively. CONCLUSION Radiomics accurately predicted the efficacy of IO and features of the TME in R/M HNSCC. Further study in a larger patient population is warranted.
Collapse
Affiliation(s)
- Dan P Zandberg
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Serafettin Zenkin
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Murat Ak
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Ronan Hsieh
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Ashely Menk
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | | | - Rivka R Colen
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Erasmus LT, Strange CD, Ahuja J, Agrawal R, Shroff GS, Marom EM, Truong MT. Imaging of Lung Cancer Staging: TNM 9 Updates. Semin Ultrasound CT MR 2024:S0887-2171(24)00045-3. [PMID: 39069273 DOI: 10.1053/j.sult.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Imaging plays a key role in clinical staging of lung cancer and guiding therapy. A thorough understanding of the staging system including the nomenclature and updates is necessary to tailor treatment plans and optimize patient care. The 9th edition of the Tumor, Node, Metastasis staging system for lung cancer has no changes for T classification and subdivides N2 and M1c categories. In nodal staging, N2 splits into N2a, ipsilateral mediastinal single station involvement and N2b, ipsilateral mediastinal multiple stations involvement. In the staging of multiple extrathoracic metastases, M1c splits into M1c1, multiple extrathoracic metastases in one organ system and M1c2, multiple extrathoracic metastases in multiple organ systems. Awareness of the proposed changes in TNM-9 staging classification is essential to provide methodical and accurate imaging interpretation.
Collapse
Affiliation(s)
- Lauren T Erasmus
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX
| | - Chad D Strange
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jitesh Ahuja
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rishi Agrawal
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Girish S Shroff
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Edith M Marom
- Department of Radiology, Chaim Sheba Medical Center, Tel Aviv, Israel
| | - Mylene T Truong
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
8
|
Lafon M, Cousin S, Alamé M, Nougaret S, Italiano A, Crombé A. Metastatic Lung Adenocarcinomas: Development and Evaluation of Radiomic-Based Methods to Measure Baseline Intra-Patient Inter-Tumor Lesion Heterogeneity. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01163-1. [PMID: 39020153 DOI: 10.1007/s10278-024-01163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/19/2024]
Abstract
Radiomics has traditionally focused on individual tumors, often neglecting the integration of metastatic disease, particularly in patients with non-small cell lung cancer. This study sought to examine intra-patient inter-tumor lesion heterogeneity indices using radiomics, exploring their relevance in metastatic lung adenocarcinoma. Consecutive adults newly diagnosed with metastatic lung adenocarcinoma underwent contrast-enhanced CT scans for lesion segmentation and radiomic feature extraction. Three methods were devised to measure distances between tumor lesion profiles within the same patient in radiomic space: centroid to lesion, lesion to lesion, and primitive to lesion, with subsequent calculation of mean, range, and standard deviation of these distances. Associations between HIs, disease control rate, objective response rate to first-line treatment, and overall survival were explored. The study included 167 patients (median age 62.3 years) between 2016 and 2019, divided randomly into experimental (N = 117,546 lesions) and validation (N = 50,232 tumor lesions) cohorts. Patients without disease control/objective response and with poorer survival consistently systematically exhibited values of all heterogeneity indices. Multivariable analyses revealed that the range of primitive-to-lesion distances was associated with disease control in both cohorts and with objective response in the validation cohort. This metrics showed univariable associations with overall survival in the experimental. In conclusion, we proposed original methods to estimate the intra-patient inter-tumor lesion heterogeneity using radiomics that demonstrated correlations with patient outcomes, shedding light on the clinical implications of inter-metastases heterogeneity. This underscores the potential of radiomics in understanding and potentially predicting treatment response and prognosis in metastatic lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Mathilde Lafon
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Mélissa Alamé
- Department of Biopathology, Institut Bergonié, Bordeaux, France
| | - Stéphanie Nougaret
- Medical Imaging Department, Montpellier Cancer Institute, Montpellier Cancer Research Institute (U1194), University of Montpellier, Montpellier, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
- SARCOTARGET Team, Bordeaux Research Institute in Oncology (BRIC) INSERM U1312 & University of Bordeaux, Bordeaux, France
| | - Amandine Crombé
- SARCOTARGET Team, Bordeaux Research Institute in Oncology (BRIC) INSERM U1312 & University of Bordeaux, Bordeaux, France.
- Department of Radiology, Institut Bergonié, Bordeaux, France.
- Department of Radiology, Pellegrin University Hospital, Bordeaux, France.
| |
Collapse
|
9
|
Chen L, Yin G, Wang Z, Liu Z, Sui C, Chen K, Song T, Xu W, Qi L, Li X. A predictive radiotranscriptomics model based on DCE-MRI for tumor immune landscape and immunotherapy in cholangiocarcinoma. Biosci Trends 2024; 18:263-276. [PMID: 38853000 DOI: 10.5582/bst.2024.01121] [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: 06/11/2024]
Abstract
This study aims to determine the predictive role of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) derived radiomic model in tumor immune profiling and immunotherapy for cholangiocarcinoma. To perform radiomic analysis, immune related subgroup clustering was first performed by single sample gene set enrichment analysis (ssGSEA). Second, a total of 806 radiomic features for each phase of DCE-MRI were extracted by utilizing the Python package Pyradiomics. Then, a predictive radiomic signature model was constructed after a three-step features reduction and selection, and receiver operating characteristic (ROC) curve was employed to evaluate the performance of this model. In the end, an independent testing cohort involving cholangiocarcinoma patients with anti-PD-1 Sintilimab treatment after surgery was used to verify the potential application of the established radiomic model in immunotherapy for cholangiocarcinoma. Two distinct immune related subgroups were classified using ssGSEA based on transcriptome sequencing. For radiomic analysis, a total of 10 predictive radiomic features were finally identified to establish a radiomic signature model for immune landscape classification. Regarding to the predictive performance, the mean AUC of ROC curves was 0.80 in the training/validation cohort. For the independent testing cohort, the individual predictive probability by radiomic model and the corresponding immune score derived from ssGSEA was significantly correlated. In conclusion, radiomic signature model based on DCE-MRI was capable of predicting the immune landscape of chalangiocarcinoma. Consequently, a potentially clinical application of this developed radiomic model to guide immunotherapy for cholangiocarcinoma was suggested.
Collapse
Affiliation(s)
- Lu Chen
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guotao Yin
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zifan Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Kun Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
10
|
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
|
11
|
Shao H, Zhu J, Shi L, Yao J, Wang Y, Ma C, Swierniak A, Ni B. Value of computed tomography radiomics combined with inflammation indices in predicting the efficacy of immunotherapy in patients with locally advanced and metastatic non-small cell lung cancer. J Thorac Dis 2024; 16:3213-3227. [PMID: 38883654 PMCID: PMC11170425 DOI: 10.21037/jtd-24-526] [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: 03/29/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024]
Abstract
Background Although immunotherapy has revolutionized the treatment landscape of lung cancer and improved the prognosis of this malignancy, many patients with lung cancer still are not able to benefit from it because of many different reasons. The expression of programmed death ligand-1 (PD-L1) in tumor cells has been approved for the prediction of immunotherapy efficacy; however, its clinical application has been limited by the invasiveness of PD-L1 determination and the heterogeneity of tumor cells. As a promising technology, radiomics has made significant progress in the diagnosis and treatment of lung cancer. Thus, we constructed a noninvasive predictive model which based on radiomics to predict the immunotherapy efficacy of lung caner patients. Methods Data of 82 patients with stage IIIa/IVb NSCLC who received immunotherapy at the First Affiliated Hospital of Soochow University from December 2019 to January 2023 were retrospectively collected. These patients were followed up for durable clinical benefit (DCB), as defined by whether progression-free survival (PFS) reached 12 months. The least absolute shrinkage and selection operator (LASSO) algorithm was used to screen for the radiomic features in the training set, and a radiomics score (Rad-score) was calculated. The clinical baseline data were analyzed, and the peripheral blood inflammation indices were calculated. Univariate and multivariate analyses were performed to identify the applicable indices, which were combined with the Rad-score to create a comprehensive forecasting model (CFM) and nomograms. Internal validation was performed in the validation set. Results Up to the last follow-up time, 48 of 82 patients had a PFS of more than 12 months. The area under the receiver operating characteristic (ROC) curve (AUC) of the Rad-score was 0.858 and 0.812, respectively, in the training set and validation set. A systemic immune-inflammation index (SII) score of <500.88 after two cycles of immunotherapy was a protective factor for PFS >12 months [odds ratio (OR) 0.054; P=0.003]. The CFM had an AUC of 0.930 and 0.922, respectively, in the training and validation sets. The calibration curves and decision curve analysis (DCA) demonstrated the reliability and clinical applicability of the model, respectively. Conclusions The radiomics model performed well in predicting whether patients with locally advanced or metastatic NSCLC can achieve DCB after receiving immunotherapy. The CFM had good predictive performance and reliability.
Collapse
Affiliation(s)
- Hancheng Shao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Shi
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Yao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxuan Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chonggang Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Andrzej Swierniak
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Bin Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Lococo F, Ghaly G, Chiappetta M, Flamini S, Evangelista J, Bria E, Stefani A, Vita E, Martino A, Boldrini L, Sassorossi C, Campanella A, Margaritora S, Mohammed A. Implementation of Artificial Intelligence in Personalized Prognostic Assessment of Lung Cancer: A Narrative Review. Cancers (Basel) 2024; 16:1832. [PMID: 38791910 PMCID: PMC11119930 DOI: 10.3390/cancers16101832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Artificial Intelligence (AI) has revolutionized the management of non-small-cell lung cancer (NSCLC) by enhancing different aspects, including staging, prognosis assessment, treatment prediction, response evaluation, recurrence/prognosis prediction, and personalized prognostic assessment. AI algorithms may accurately classify NSCLC stages using machine learning techniques and deep imaging data analysis. This could potentially improve precision and efficiency in staging, facilitating personalized treatment decisions. Furthermore, there are data suggesting the potential application of AI-based models in predicting prognosis in terms of survival rates and disease progression by integrating clinical, imaging and molecular data. In the present narrative review, we will analyze the preliminary studies reporting on how AI algorithms could predict responses to various treatment modalities, such as surgery, radiotherapy, chemotherapy, targeted therapy, and immunotherapy. There is robust evidence suggesting that AI also plays a crucial role in predicting the likelihood of tumor recurrence after surgery and the pattern of failure, which has significant implications for tailoring adjuvant treatments. The successful implementation of AI in personalized prognostic assessment requires the integration of different data sources, including clinical, molecular, and imaging data. Machine learning (ML) and deep learning (DL) techniques enable AI models to analyze these data and generate personalized prognostic predictions, allowing for a precise and individualized approach to patient care. However, challenges relating to data quality, interpretability, and the ability of AI models to generalize need to be addressed. Collaboration among clinicians, data scientists, and regulators is critical for the responsible implementation of AI and for maximizing its benefits in providing a more personalized prognostic assessment. Continued research, validation, and collaboration are essential to fully exploit the potential of AI in NSCLC management and improve patient outcomes. Herein, we have summarized the state of the art of applications of AI in lung cancer for predicting staging, prognosis, and pattern of recurrence after treatment in order to provide to the readers a large comprehensive overview of this challenging issue.
Collapse
Affiliation(s)
- Filippo Lococo
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Galal Ghaly
- Faculty of Medicine and Surgery, Thoracic Surgery Unit, Cairo University, Giza 12613, Egypt; (G.G.); (A.M.)
| | - Marco Chiappetta
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Sara Flamini
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Jessica Evangelista
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Emilio Bria
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Medical Oncology, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy
| | - Alessio Stefani
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Medical Oncology, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy
| | - Emanuele Vita
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Medical Oncology, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy
| | - Antonella Martino
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Radiotherapy Unit, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy
| | - Luca Boldrini
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Radiotherapy Unit, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy
| | - Carolina Sassorossi
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Annalisa Campanella
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Stefano Margaritora
- Faculty of Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (J.E.); (E.B.); (A.S.); (E.V.); (A.M.); (L.B.); (C.S.); (S.M.)
- Thoracic Surgery, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy; (S.F.); (A.C.)
| | - Abdelrahman Mohammed
- Faculty of Medicine and Surgery, Thoracic Surgery Unit, Cairo University, Giza 12613, Egypt; (G.G.); (A.M.)
| |
Collapse
|
13
|
Dixon D, Sattar H, Moros N, Kesireddy SR, Ahsan H, Lakkimsetti M, Fatima M, Doshi D, Sadhu K, Junaid Hassan M. Unveiling the Influence of AI Predictive Analytics on Patient Outcomes: A Comprehensive Narrative Review. Cureus 2024; 16:e59954. [PMID: 38854327 PMCID: PMC11161909 DOI: 10.7759/cureus.59954] [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] [Accepted: 05/08/2024] [Indexed: 06/11/2024] Open
Abstract
This comprehensive literature review explores the transformative impact of artificial intelligence (AI) predictive analytics on healthcare, particularly in improving patient outcomes regarding disease progression, treatment response, and recovery rates. AI, encompassing capabilities such as learning, problem-solving, and decision-making, is leveraged to predict disease progression, optimize treatment plans, and enhance recovery rates through the analysis of vast datasets, including electronic health records (EHRs), imaging, and genetic data. The utilization of machine learning (ML) and deep learning (DL) techniques in predictive analytics enables personalized medicine by facilitating the early detection of conditions, precision in drug discovery, and the tailoring of treatment to individual patient profiles. Ethical considerations, including data privacy, bias, and accountability, emerge as vital in the responsible implementation of AI in healthcare. The findings underscore the potential of AI predictive analytics in revolutionizing clinical decision-making and healthcare delivery, emphasizing the necessity of ethical guidelines and continuous model validation to ensure its safe and effective use in augmenting human judgment in medical practice.
Collapse
Affiliation(s)
- Diny Dixon
- Medicine, Jubilee Mission Medical College and Research Institute, Thrissur, IND
| | - Hina Sattar
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Natalia Moros
- Medicine, Pontifical Javeriana University Medical School, Bogotá, COL
| | | | - Huma Ahsan
- Medicine, Jinnah Postgraduate Medical Centre, Karachi, PAK
| | | | - Madiha Fatima
- Medicine, Fatima Jinnah Medical University, Lahore, PAK
| | - Dhruvi Doshi
- Medicine, Gujarat Cancer Society Medical College, Hospital & Research Centre, Ahmedabad, IND
| | | | | |
Collapse
|
14
|
Ni S, Liang Q, Jiang X, Ge Y, Jiang Y, Liu L. Prognostic models for immunotherapy in non-small cell lung cancer: A comprehensive review. Heliyon 2024; 10:e29840. [PMID: 38681577 PMCID: PMC11053285 DOI: 10.1016/j.heliyon.2024.e29840] [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: 09/26/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024] Open
Abstract
The introduction of immune checkpoint inhibitors (ICIs) has revolutionized the treatment of lung cancer. Given the limited clinical benefits of immunotherapy in patients with non-small cell lung cancer (NSCLC), various predictors have been shown to significantly influence prognosis. However, no single predictor is adequate to forecast patients' survival benefit. Therefore, it's imperative to develop a prognostic model that integrates multiple predictors. This model would be instrumental in identifying patients who might benefit from ICIs. Retrospective analysis and small case series have demonstrated the potential role of these models in prognostic prediction, though further prospective investigation is required to evaluate more rigorously their application in these contexts. This article presents and summarizes the latest research advancements on immunotherapy prognostic models for NSCLC from multiple omics perspectives and discuss emerging strategies being developed to enhance the domain.
Collapse
Affiliation(s)
- Siqi Ni
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qi Liang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xingyu Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yinping Ge
- The Friendship Hospital of Ili Kazakh Autonomous Prefecture Ili & Jiangsu Joint Institute of Health, Yining 835000, Xinjiang Uygur Autonomous Regio, China
| | - Yali Jiang
- The Friendship Hospital of Ili Kazakh Autonomous Prefecture Ili & Jiangsu Joint Institute of Health, Yining 835000, Xinjiang Uygur Autonomous Regio, China
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
15
|
Xu J, Wang P, Li Y, Shi X, Yin T, Yu J, Teng F. Development and validation of an MRI-Based nomogram to predict the effectiveness of immunotherapy for brain metastasis in patients with non-small cell lung cancer. Front Immunol 2024; 15:1373330. [PMID: 38686383 PMCID: PMC11057328 DOI: 10.3389/fimmu.2024.1373330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction The variability and unpredictability of immune checkpoint inhibitors (ICIs) in treating brain metastases (BMs) in patients with advanced non-small cell lung cancer (NSCLC) is the main concern. We assessed the utility of novel imaging biomarkers (radiomics) for discerning patients with NSCLC and BMs who would derive advantages from ICIs treatment. Methods Data clinical outcomes and pretreatment magnetic resonance images (MRI) were collected on patients with NSCLC with BMs treated with ICIs between June 2019 and June 2022 and divided into training and test sets. Metastatic brain lesions were contoured using ITK-SNAP software, and 3748 radiomic features capturing both intra- and peritumoral texture patterns were extracted. A clinical radiomic nomogram (CRN) was built to evaluate intracranial progression-free survival, progression-free survival, and overall survival. The prognostic value of the CRN was assessed by Kaplan-Meier survival analysis and log-rank tests. Results In the study, a total of 174 patients were included, and 122 and 52 were allocated to the training and validation sets correspondingly. The intratumoral radiomic signature, peritumoral radiomic signature, clinical signature, and CRN predicted intracranial objective response rate. Kaplan-Meier analyses showed a significantly longer intracranial progression-free survival in the low-CRN group than in the high-CRN group (p < 0.001). The CRN was also significantly associated with progression-free survival (p < 0.001) but not overall survival. Discussion Radiomics biomarkers from pretreatment MRI images were predictive of intracranial response. Pretreatment radiomics may allow the early prediction of benefits.
Collapse
Affiliation(s)
- Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Peiliang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaonan Shi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tianwen Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
16
|
Nakajima EC, Simpson A, Bogaerts J, de Vries EGE, Do R, Garalda E, Goldmacher G, Kinahan PE, Lambin P, LeStage B, Li Q, Lin F, Litière S, Perez-Lopez R, Petrick N, Schwartz L, Seymour L, Shankar L, Laurie SA. Tumor Size Is Not Everything: Advancing Radiomics as a Precision Medicine Biomarker in Oncology Drug Development and Clinical Care. A Report of a Multidisciplinary Workshop Coordinated by the RECIST Working Group. JCO Precis Oncol 2024; 8:e2300687. [PMID: 38635935 DOI: 10.1200/po.23.00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
Radiomics, the science of extracting quantifiable data from routine medical images, is a powerful tool that has many potential applications in oncology. The Response Evaluation Criteria in Solid Tumors Working Group (RWG) held a workshop in May 2022, which brought together various stakeholders to discuss the potential role of radiomics in oncology drug development and clinical trials, particularly with respect to response assessment. This article summarizes the results of that workshop, reviewing radiomics for the practicing oncologist and highlighting the work that needs to be done to move forward the incorporation of radiomics into clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Richard Do
- Memorial Sloan-Kettering Cancer Center, NY, NY
| | - Elena Garalda
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | | | | | - Frank Lin
- University of Sydney, Sydney, Australia
| | | | | | | | | | - Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Lalitha Shankar
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Scott A Laurie
- The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
17
|
Qu W, Chen C, Cai C, Gong M, Luo Q, Song Y, Yang M, Shi M. Non-invasive prediction for pathologic complete response to neoadjuvant chemoimmunotherapy in lung cancer using CT-based deep learning: a multicenter study. Front Immunol 2024; 15:1327779. [PMID: 38596674 PMCID: PMC11003263 DOI: 10.3389/fimmu.2024.1327779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Neoadjuvant chemoimmunotherapy has revolutionized the therapeutic strategy for non-small cell lung cancer (NSCLC), and identifying candidates likely responding to this advanced treatment is of important clinical significance. The current multi-institutional study aims to develop a deep learning model to predict pathologic complete response (pCR) to neoadjuvant immunotherapy in NSCLC based on computed tomography (CT) imaging and further prob the biologic foundation of the proposed deep learning signature. A total of 248 participants administrated with neoadjuvant immunotherapy followed by surgery for NSCLC at Ruijin Hospital, Ningbo Hwamei Hospital, and Affiliated Hospital of Zunyi Medical University from January 2019 to September 2023 were enrolled. The imaging data within 2 weeks prior to neoadjuvant chemoimmunotherapy were retrospectively extracted. Patients from Ruijin Hospital were grouped as the training set (n = 104) and the validation set (n = 69) at the 6:4 ratio, and other participants from Ningbo Hwamei Hospital and Affiliated Hospital of Zunyi Medical University served as an external cohort (n = 75). For the entire population, pCR was obtained in 29.4% (n = 73) of cases. The areas under the curve (AUCs) of our deep learning signature for pCR prediction were 0.775 (95% confidence interval [CI]: 0.649 - 0.901) and 0.743 (95% CI: 0.618 - 0.869) in the validation set and the external cohort, significantly superior than 0.579 (95% CI: 0.468 - 0.689) and 0.569 (95% CI: 0.454 - 0.683) of the clinical model. Furthermore, higher deep learning scores correlated to the upregulation for pathways of cell metabolism and more antitumor immune infiltration in microenvironment. Our developed deep learning model is capable of predicting pCR to neoadjuvant chemoimmunotherapy in patients with NSCLC.
Collapse
Affiliation(s)
- Wendong Qu
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Cheng Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chuang Cai
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ming Gong
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Luo
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongxiang Song
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo Hwamei Hospital, Chinese Academy of Sciences, Zhejiang, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Hospital of Civil Aviation Administration of China, Shanghai, China
- Department of Oncology, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Wuxi, China
| |
Collapse
|
18
|
de Miguel-Perez D, Ak M, Mamindla P, Russo A, Zenkin S, Ak N, Peddagangireddy V, Lara-Mejia L, Gunasekaran M, Cardona AF, Naing A, Hirsch FR, Arrieta O, Colen RR, Rolfo C. Validation of a multiomic model of plasma extracellular vesicle PD-L1 and radiomics for prediction of response to immunotherapy in NSCLC. J Exp Clin Cancer Res 2024; 43:81. [PMID: 38486328 PMCID: PMC10941547 DOI: 10.1186/s13046-024-02997-x] [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: 01/31/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICIs) have showed unprecedent efficacy in the treatment of patients with advanced non-small cell lung cancer (NSCLC). However, not all patients manifest clinical benefit due to the lack of reliable predictive biomarkers. We showed preliminary data on the predictive role of the combination of radiomics and plasma extracellular vesicle (EV) PD-L1 to predict durable response to ICIs. MAIN BODY Here, we validated this model in a prospective cohort of patients receiving ICIs plus chemotherapy and compared it with patients undergoing chemotherapy alone. This multiparametric model showed high sensitivity and specificity at identifying non-responders to ICIs and outperformed tissue PD-L1, being directly correlated with tumor change. SHORT CONCLUSION These findings indicate that the combination of radiomics and EV PD-L1 dynamics is a minimally invasive and promising biomarker for the stratification of patients to receive ICIs.
Collapse
Affiliation(s)
- Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai, 1470 Madison Ave, New York, NY, 10029, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Murat Ak
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | | | - Nursima Ak
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Vishal Peddagangireddy
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Luis Lara-Mejia
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Muthukumar Gunasekaran
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Departments of Surgery and Pediatrics, Feinberg School of Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL, USA
| | - Andres F Cardona
- Molecular Oncology and Biology Systems Research Group (Fox G), Universidad El Bosque, Bogota, Colombia
| | - Aung Naing
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai, 1470 Madison Ave, New York, NY, 10029, USA
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Rivka R Colen
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai, 1470 Madison Ave, New York, NY, 10029, USA.
| |
Collapse
|
19
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [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: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
20
|
Fiste O, Gkiozos I, Charpidou A, Syrigos NK. Artificial Intelligence-Based Treatment Decisions: A New Era for NSCLC. Cancers (Basel) 2024; 16:831. [PMID: 38398222 PMCID: PMC10887017 DOI: 10.3390/cancers16040831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality among women and men, in developed countries, despite the public health interventions including tobacco-free campaigns, screening and early detection methods, recent therapeutic advances, and ongoing intense research on novel antineoplastic modalities. Targeting oncogenic driver mutations and immune checkpoint inhibition has indeed revolutionized NSCLC treatment, yet there still remains the unmet need for robust and standardized predictive biomarkers to accurately inform clinical decisions. Artificial intelligence (AI) represents the computer-based science concerned with large datasets for complex problem-solving. Its concept has brought a paradigm shift in oncology considering its immense potential for improved diagnosis, treatment guidance, and prognosis. In this review, we present the current state of AI-driven applications on NSCLC management, with a particular focus on radiomics and pathomics, and critically discuss both the existing limitations and future directions in this field. The thoracic oncology community should not be discouraged by the likely long road of AI implementation into daily clinical practice, as its transformative impact on personalized treatment approaches is undeniable.
Collapse
Affiliation(s)
- Oraianthi Fiste
- Oncology Unit, Third Department of Internal Medicine and Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (A.C.); (N.K.S.)
| | | | | | | |
Collapse
|
21
|
McGale JP, Howell HJ, Beddok A, Tordjman M, Sun R, Chen D, Wu AM, Assi T, Ammari S, Dercle L. Integrating Artificial Intelligence and PET Imaging for Drug Discovery: A Paradigm Shift in Immunotherapy. Pharmaceuticals (Basel) 2024; 17:210. [PMID: 38399425 PMCID: PMC10892847 DOI: 10.3390/ph17020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
The integration of artificial intelligence (AI) and positron emission tomography (PET) imaging has the potential to become a powerful tool in drug discovery. This review aims to provide an overview of the current state of research and highlight the potential for this alliance to advance pharmaceutical innovation by accelerating the development and deployment of novel therapeutics. We previously performed a scoping review of three databases (Embase, MEDLINE, and CENTRAL), identifying 87 studies published between 2018 and 2022 relevant to medical imaging (e.g., CT, PET, MRI), immunotherapy, artificial intelligence, and radiomics. Herein, we reexamine the previously identified studies, performing a subgroup analysis on articles specifically utilizing AI and PET imaging for drug discovery purposes in immunotherapy-treated oncology patients. Of the 87 original studies identified, 15 met our updated search criteria. In these studies, radiomics features were primarily extracted from PET/CT images in combination (n = 9, 60.0%) rather than PET imaging alone (n = 6, 40.0%), and patient cohorts were mostly recruited retrospectively and from single institutions (n = 10, 66.7%). AI models were used primarily for prognostication (n = 6, 40.0%) or for assisting in tumor phenotyping (n = 4, 26.7%). About half of the studies stress-tested their models using validation sets (n = 4, 26.7%) or both validation sets and test sets (n = 4, 26.7%), while the remaining six studies (40.0%) either performed no validation at all or used less stringent methods such as cross-validation on the training set. Overall, the integration of AI and PET imaging represents a paradigm shift in drug discovery, offering new avenues for more efficient development of therapeutics. By leveraging AI algorithms and PET imaging analysis, researchers could gain deeper insights into disease mechanisms, identify new drug targets, or optimize treatment regimens. However, further research is needed to validate these findings and address challenges such as data standardization and algorithm robustness.
Collapse
Affiliation(s)
- Jeremy P. McGale
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA (H.J.H.)
| | - Harrison J. Howell
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA (H.J.H.)
| | - Arnaud Beddok
- Department of Radiation Oncology, Institut Godinot, 51100 Reims, France
| | - Mickael Tordjman
- Department of Radiology, Hôtel Dieu Hospital, APHP, 75014 Paris, France
| | - Roger Sun
- Department of Radiation Oncology, Gustave Roussy, 94800 Villejuif, France
| | - Delphine Chen
- Department of Molecular Imaging and Therapy, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Anna M. Wu
- Department of Immunology and Theranostics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
| | - Tarek Assi
- International Department, Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Samy Ammari
- Department of Medical Imaging, BIOMAPS, UMR1281 INSERM, CEA, CNRS, Gustave Roussy, Université Paris-Saclay, 94800 Villejuif, France
- ELSAN Department of Radiology, Institut de Cancérologie Paris Nord, 95200 Sarcelles, France
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA (H.J.H.)
| |
Collapse
|
22
|
Zhong Y, Cai C, Chen T, Gui H, Chen C, Deng J, Yang M, Yu B, Song Y, Wang T, Chen Y, Shi H, Xie D, Chen C, She Y. PET/CT-based deep learning grading signature to optimize surgical decisions for clinical stage I invasive lung adenocarcinoma and biologic basis under its prediction: a multicenter study. Eur J Nucl Med Mol Imaging 2024; 51:521-534. [PMID: 37725128 DOI: 10.1007/s00259-023-06434-7] [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: 04/26/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE No consensus on a grading system for invasive lung adenocarcinoma had been built over a long period of time. Until October 2020, a novel grading system was proposed to quantify the whole landscape of histologic subtypes and proportions of pulmonary adenocarcinomas. This study aims to develop a deep learning grading signature (DLGS) based on positron emission tomography/computed tomography (PET/CT) to personalize surgical treatments for clinical stage I invasive lung adenocarcinoma and explore the biologic basis under its prediction. METHODS A total of 2638 patients with clinical stage I invasive lung adenocarcinoma from 4 medical centers were retrospectively included to construct and validate the DLGS. The predictive performance of the DLGS was evaluated by the area under the receiver operating characteristic curve (AUC), its potential to optimize surgical treatments was investigated via survival analyses in risk groups defined by the DLGS, and its biological basis was explored by comparing histologic patterns, genotypic alternations, genetic pathways, and infiltration of immune cells in microenvironments between risk groups. RESULTS The DLGS to predict grade 3 achieved AUCs of 0.862, 0.844, and 0.851 in the validation set (n = 497), external cohort (n = 382), and prospective cohort (n = 600), respectively, which were significantly better than 0.814, 0.810, and 0.806 of the PET model, 0.813, 0.795, and 0.824 of the CT model, and 0.762, 0.734, and 0.751 of the clinical model. Additionally, for DLGS-defined high-risk population, lobectomy yielded an improved prognosis compared to sublobectomy p = 0.085 for overall survival [OS] and p = 0.038 for recurrence-free survival [RFS]) and systematic nodal dissection conferred a superior prognosis to limited nodal dissection (p = 0.001 for OS and p = 0.041 for RFS). CONCLUSION The DLGS harbors the potential to predict the histologic grade and personalize the surgical treatments for clinical stage I invasive lung adenocarcinoma. Its applicability to other territories should be further validated by a larger international study.
Collapse
Affiliation(s)
- Yifan Zhong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chuang Cai
- School of Computer Science and Communication Engineering , Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Gui
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Cheng Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi Medical College, Guizhou, China
| | - Jiajun Deng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo HwaMei Hospital, Chinese Academy of Sciences, Zhejiang, China
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yongxiang Song
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi Medical College, Guizhou, China
| | - Tingting Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangchun Chen
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huazheng Shi
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
23
|
Nakajo M, Jinguji M, Ito S, Tani A, Hirahara M, Yoshiura T. Clinical application of 18F-fluorodeoxyglucose positron emission tomography/computed tomography radiomics-based machine learning analyses in the field of oncology. Jpn J Radiol 2024; 42:28-55. [PMID: 37526865 PMCID: PMC10764437 DOI: 10.1007/s11604-023-01476-1] [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: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023]
Abstract
Machine learning (ML) analyses using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) radiomics features have been applied in the field of oncology. The current review aimed to summarize the current clinical articles about 18F-FDG PET/CT radiomics-based ML analyses to solve issues in classifying or constructing prediction models for several types of tumors. In these studies, lung and mediastinal tumors were the most commonly evaluated lesions, followed by lymphatic, abdominal, head and neck, breast, gynecological, and other types of tumors. Previous studies have commonly shown that 18F-FDG PET radiomics-based ML analysis has good performance in differentiating benign from malignant tumors, predicting tumor characteristics and stage, therapeutic response, and prognosis by examining significant differences in the area under the receiver operating characteristic curves, accuracies, or concordance indices (> 0.70). However, these studies have reported several ML algorithms. Moreover, different ML models have been applied for the same purpose. Thus, various procedures were used in 18F-FDG PET/CT radiomics-based ML analysis in oncology, and 18F-FDG PET/CT radiomics-based ML models, which are easy and universally applied in clinical practice, would be expected to be established.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Megumi Jinguji
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Soichiro Ito
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atushi Tani
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Mitsuho Hirahara
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
24
|
Fang Y, Chen X, Cao C. Cancer immunotherapy efficacy and machine learning. Expert Rev Anticancer Ther 2024; 24:21-28. [PMID: 38288663 DOI: 10.1080/14737140.2024.2311684] [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: 07/25/2023] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
INTRODUCTION Immunotherapy is one of the major breakthroughs in the treatment of cancer, and it has become a powerful clinical strategy, however, not all patients respond to immune checkpoint blockade and other immunotherapy strategies. Applying machine learning (ML) techniques to predict the efficacy of cancer immunotherapy is useful for clinical decision-making. AREAS COVERED Applying ML including deep learning (DL) in radiomics, pathomics, tumor microenvironment (TME) and immune-related genes analysis to predict immunotherapy efficacy. The studies in this review were searched from PubMed and ClinicalTrials.gov (January 2023). EXPERT OPINION An increasing number of studies indicate that ML has been applied to various aspects of oncology research, with the potential to provide more effective individualized immunotherapy strategies and enhance treatment decisions. With advances in ML technology, more efficient methods of predicting the efficacy of immunotherapy may become available in the future.
Collapse
Affiliation(s)
- Yuting Fang
- Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Xiaozhong Chen
- Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Caineng Cao
- Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
25
|
Roisman LC, Kian W, Anoze A, Fuchs V, Spector M, Steiner R, Kassel L, Rechnitzer G, Fried I, Peled N, Bogot NR. Radiological artificial intelligence - predicting personalized immunotherapy outcomes in lung cancer. NPJ Precis Oncol 2023; 7:125. [PMID: 37990050 PMCID: PMC10663598 DOI: 10.1038/s41698-023-00473-x] [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: 07/17/2023] [Accepted: 10/24/2023] [Indexed: 11/23/2023] Open
Abstract
Personalized medicine has revolutionized approaches to treatment in the field of lung cancer by enabling therapies to be specific to each patient. However, physicians encounter an immense number of challenges in providing the optimal treatment regimen for the individual given the sheer complexity of clinical aspects such as tumor molecular profile, tumor microenvironment, expected adverse events, acquired or inherent resistance mechanisms, the development of brain metastases, the limited availability of biomarkers and the choice of combination therapy. The integration of innovative next-generation technologies such as deep learning-a subset of machine learning-and radiomics has the potential to transform the field by supporting clinical decision making in cancer treatment and the delivery of precision therapies while integrating numerous clinical considerations. In this review, we present a brief explanation of the available technologies, the benefits of using these technologies in predicting immunotherapy response in lung cancer, and the expected future challenges in the context of precision medicine.
Collapse
Affiliation(s)
- Laila C Roisman
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel.
- Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Waleed Kian
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
- The Institute of Oncology, Assuta Ashdod, Ashdod, Israel
| | - Alaa Anoze
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Vered Fuchs
- Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Maria Spector
- The Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Roee Steiner
- The Institute for Nuclear Medicine, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Levi Kassel
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Gilad Rechnitzer
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Iris Fried
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nir Peled
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Naama R Bogot
- The Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
26
|
Erasmus LT, Strange TA, Agrawal R, Strange CD, Ahuja J, Shroff GS, Truong MT. Lung Cancer Staging: Imaging and Potential Pitfalls. Diagnostics (Basel) 2023; 13:3359. [PMID: 37958255 PMCID: PMC10649001 DOI: 10.3390/diagnostics13213359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths in men and women in the United States. Accurate staging is needed to determine prognosis and devise effective treatment plans. The International Association for the Study of Lung Cancer (IASLC) has made multiple revisions to the tumor, node, metastasis (TNM) staging system used by the Union for International Cancer Control and the American Joint Committee on Cancer to stage lung cancer. The eighth edition of this staging system includes modifications to the T classification with cut points of 1 cm increments in tumor size, grouping of lung cancers associated with partial or complete lung atelectasis or pneumonitis, grouping of tumors with involvement of a main bronchus regardless of distance from the carina, and upstaging of diaphragmatic invasion to T4. The N classification describes the spread to regional lymph nodes and no changes were proposed for TNM-8. In the M classification, metastatic disease is divided into intra- versus extrathoracic metastasis, and single versus multiple metastases. In order to optimize patient outcomes, it is important to understand the nuances of the TNM staging system, the strengths and weaknesses of various imaging modalities used in lung cancer staging, and potential pitfalls in image interpretation.
Collapse
Affiliation(s)
- Lauren T. Erasmus
- Department of Anatomy and Cell Biology, Faculty of Sciences, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Taylor A. Strange
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Rishi Agrawal
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Chad D. Strange
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Jitesh Ahuja
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Girish S. Shroff
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Mylene T. Truong
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| |
Collapse
|
27
|
Pan F, Feng L, Liu B, Hu Y, Wang Q. Application of radiomics in diagnosis and treatment of lung cancer. Front Pharmacol 2023; 14:1295511. [PMID: 38027000 PMCID: PMC10646419 DOI: 10.3389/fphar.2023.1295511] [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: 09/16/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Radiomics has become a research field that involves the process of converting standard nursing images into quantitative image data, which can be combined with other data sources and subsequently analyzed using traditional biostatistics or artificial intelligence (Al) methods. Due to the capture of biological and pathophysiological information by radiomics features, these quantitative radiomics features have been proven to provide fast and accurate non-invasive biomarkers for lung cancer risk prediction, diagnosis, prognosis, treatment response monitoring, and tumor biology. In this review, radiomics has been emphasized and discussed in lung cancer research, including advantages, challenges, and drawbacks.
Collapse
Affiliation(s)
- Feng Pan
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of CT, Jilin Province FAW General Hospital, Changchun, China
| | - Li Feng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baocai Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qian Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Wu S, Zhan W, Liu L, Xie D, Yao L, Yao H, Liao G, Huang L, Zhou Y, You P, Huang Z, Li Q, Xu B, Wang S, Wang G, Zhang DK, Qiao G, Chan LWC, Lanuti M, Zhou H. Pretreatment radiomic biomarker for immunotherapy responder prediction in stage IB-IV NSCLC (LCDigital-IO Study): a multicenter retrospective study. J Immunother Cancer 2023; 11:e007369. [PMID: 37865396 PMCID: PMC10603353 DOI: 10.1136/jitc-2023-007369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND The predictive efficacy of current biomarker of immune checkpoint inhibitors (ICIs) is not sufficient. This study investigated the causality between radiomic biomarkers and immunotherapy response status in patients with stage IB-IV non-small cell lung cancer (NSCLC), including its biological context for ICIs treatment response prediction. METHODS CT images from 319 patients with pretreatment NSCLC receiving immunotherapy between January 2015 and November 2021 were retrospectively collected and composed a discovery (n=214), independent validation (n=54), and external test cohort (n=51). A set of 851 features was extracted from tumorous and peritumoral volumes of interest (VOIs). The reference standard is the durable clinical benefit (DCB, sustained disease control for more than 6 months assessed via radiological evaluation). The predictive value of combined radiomic signature (CRS) for pathological response was subsequently assessed in another cohort of 98 patients with resectable NSCLC receiving ICIs preoperatively. The association between radiomic features and tumor immune landscape on the online data set (n=60) was also examined. A model combining clinical predictor and radiomic signatures was constructed to improve performance further. RESULTS CRS discriminated DCB and non-DCB patients well in the training and validation cohorts with an area under the curve (AUC) of 0.82, 95% CI: 0.75 to 0.88, and 0.75, 95% CI: 0.64 to 0.87, respectively. In this study, the predictive value of CRS was better than programmed cell death ligand-1 (PD-L1) expression (AUC of PD-L1 subset: 0.59, 95% CI: 0.50 to 0.69) or clinical model (AUC: 0.66, 95% CI: 0.51 to 0.81). After combining the clinical signature with CRS, the predictive performance improved further with an AUC of 0.837, 0.790 and 0.781 in training, validation and D2 cohorts, respectively. When predicting pathological response, CRS divided patients into a major pathological response (MPR) and non-MPR group (AUC: 0.76, 95% CI: 0.67 to 0.81). Moreover, CRS showed a promising stratification ability on overall survival (HR: 0.49, 95% CI: 0.27 to 0.89; p=0.020) and progression-free survival (HR: 0.43, 95% CI: 0.26 to 0.74; p=0.002). CONCLUSION By analyzing both tumorous and peritumoral regions of CT images in a radiomic strategy, we developed a non-invasive biomarker for distinguishing responders of ICIs therapy and stratifying their survival outcome efficiently, which may support the clinical decisions on the use of ICIs in advanced as well as patients with resectable NSCLC.
Collapse
Affiliation(s)
- Shaowei Wu
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Weijie Zhan
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Daipeng Xie
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Lintong Yao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Shantou University Medical College, Shantou, China
| | - Henian Yao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangdong Medical University, Zhanjiang, China
| | - Guoqing Liao
- Department of Thoracic Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Luyu Huang
- Department of Surgery, Competence Center of Thoracic Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yubo Zhou
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Peimeng You
- Department of Radiology, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zekai Huang
- Guangdong Medical University, Zhanjiang, China
| | - Qiaxuan Li
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Shantou University Medical College, Shantou, China
| | - Bin Xu
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Siyun Wang
- Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Guangyi Wang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Dong-Kun Zhang
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Lawrence Wing-Chi Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Michael Lanuti
- Department of Thoracic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Haiyu Zhou
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
29
|
Cheng Y, Chen ZY, Huang JJ, Shao D. Efficacy evaluation of neoadjuvant immunotherapy plus chemotherapy for non-small-cell lung cancer: comparison of PET/CT with postoperative pathology. Eur Radiol 2023; 33:6625-6635. [PMID: 37515634 DOI: 10.1007/s00330-023-09922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/16/2023] [Accepted: 05/29/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVES To assess the value of positron emission tomography/computed tomography (PET/CT) in the efficacy evaluation of patients undergoing neoadjuvant immunotherapy plus chemotherapy, and to analyze its correlation with postoperative pathology. METHODS The PET/CT metabolic parameters and CT size were retrospectively analyzed before and after neoadjuvant immunotherapy plus chemotherapy in 67 patients with resectable stage II/IIIA non-small-cell lung cancer (NSCLC). CT assessment based on immune response evaluation criteria in solid tumor criteria ((i)RECIST) was compared with PET/CT assessment based on the response criteria in solid tumors (PERCIST). The correlations between PET/CT metabolic parameters and postoperative pathology were analyzed. The value of PET/CT in the efficacy evaluation was assessed. RESULTS The PET/CT assessment showed high consistency with postoperative pathological evaluation, yet the CT assessment showed low consistency with postoperative pathological evaluation. The (i)RECIST and PERCIST criteria showed statistically significant differences (p < 0.001). The postoperative pathological response was negatively associated with ΔSUVmax (%) (r = - 0.812, p < 0.001), ΔSUVmean (%) (r = - 0.805, p < 0.001), and ΔSUVpeak (%) (r = - 0.800, p < 0.001). The cut-off values of 75.8 for ΔSUVmax (%), 67.8 for ΔSUVmean (%), and 74.6 for ΔSUVpeak (%) had the highest sensitivity and specificity. CONCLUSION The PERCIST criteria are more sensitive and accurate than (i)RECIST criteria to identify more responders when evaluating the response of neoadjuvant immunotherapy plus chemotherapy for NSCLC. PET/CT shows high accuracy in predicting postoperative pathological response. Our study shows the important role PET/CT plays in the efficacy evaluation of NSCLC patients undergoing neoadjuvant immunotherapy plus chemotherapy, as well as in predicting the prognosis and guiding postoperative treatment. CLINICAL RELEVANCE STATEMENT Neoadjuvant immunotherapy plus chemotherapy is highly effective in the treatment of non-small-cell lung cancer. And PET/CT played an important role in the efficacy evaluation following neoadjuvant immunotherapy plus chemotherapy for non-small-cell lung cancer. KEY POINTS • Neoadjuvant immunotherapy plus chemotherapy is highly effective in the treatment of NSCLC. • The PERCIST criteria are more sensitive and accurate than (i)RECIST criteria to identify more responders when evaluating the response of neoadjuvant immunotherapy plus chemotherapy for NSCLC. • PET/CT played an important role in the efficacy evaluation; ΔSUVmax (%), ΔSUVmean (%), and ΔSUVpeak (%) following neoadjuvant immunotherapy plus chemotherapy for NSCLC had high consistency and strong correlations with postoperative pathology.
Collapse
Affiliation(s)
- You Cheng
- Department of PET Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Room 526, 5/F, Weilun Building, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Zhi-Yong Chen
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jian-Jiang Huang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Dan Shao
- Department of PET Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Room 526, 5/F, Weilun Building, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
30
|
Shu Y, Xu W, Su R, Ran P, Liu L, Zhang Z, Zhao J, Chao Z, Fu G. Clinical applications of radiomics in non-small cell lung cancer patients with immune checkpoint inhibitor-related pneumonitis. Front Immunol 2023; 14:1251645. [PMID: 37799725 PMCID: PMC10547882 DOI: 10.3389/fimmu.2023.1251645] [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: 07/02/2023] [Accepted: 08/24/2023] [Indexed: 10/07/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) modulate the body's immune function to treat tumors but may also induce pneumonitis. Immune checkpoint inhibitor-related pneumonitis (ICIP) is a serious immune-related adverse event (irAE). Immunotherapy is currently approved as a first-line treatment for non-small cell lung cancer (NSCLC), and the incidence of ICIP in NSCLC patients can be as high as 5%-19% in clinical practice. ICIP can be severe enough to lead to the death of NSCLC patients, but there is a lack of a gold standard for the diagnosis of ICIP. Radiomics is a method that uses computational techniques to analyze medical images (e.g., CT, MRI, PET) and extract important features from them, which can be used to solve classification and regression problems in the clinic. Radiomics has been applied to predict and identify ICIP in NSCLC patients in the hope of transforming clinical qualitative problems into quantitative ones, thus improving the diagnosis and treatment of ICIP. In this review, we summarize the pathogenesis of ICIP and the process of radiomics feature extraction, review the clinical application of radiomics in ICIP of NSCLC patients, and discuss its future application prospects.
Collapse
Affiliation(s)
- Yang Shu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wei Xu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Rui Su
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Pancen Ran
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Liu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhizhao Zhang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jing Zhao
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhen Chao
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Guobin Fu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Oncology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Oncology, The Third Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
31
|
Liang S, Wang H, Tian H, Xu Z, Wu M, Hua D, Li C. The prognostic biological markers of immunotherapy for non-small cell lung cancer: current landscape and future perspective. Front Immunol 2023; 14:1249980. [PMID: 37753089 PMCID: PMC10518408 DOI: 10.3389/fimmu.2023.1249980] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
The emergence of immunotherapy, particularly programmed cell death 1 (PD-1) and programmed cell death ligand-1 (PD-L1) produced profound transformations for treating non-small cell lung cancer (NSCLC). Nevertheless, not all NSCLC patients can benefit from immunotherapy in clinical practice. In addition to limited response rates, exorbitant treatment costs, and the substantial threats involved with immune-related adverse events, the intricate interplay between long-term survival outcomes and early disease progression, including early immune hyperprogression, remains unclear. Consequently, there is an urgent imperative to identify robust predictive and prognostic biological markers, which not only possess the potential to accurately forecast the therapeutic efficacy of immunotherapy in NSCLC but also facilitate the identification of patient subgroups amenable to personalized treatment approaches. Furthermore, this advancement in patient stratification based on certain biological markers can also provide invaluable support for the management of immunotherapy in NSCLC patients. Hence, in this review, we comprehensively examine the current landscape of individual biological markers, including PD-L1 expression, tumor mutational burden, hematological biological markers, and gene mutations, while also exploring the potential of combined biological markers encompassing radiological and radiomic markers, as well as prediction models that have the potential to better predict responders to immunotherapy in NSCLC with an emphasis on some directions that warrant further investigation which can also deepen the understanding of clinicians and provide a reference for clinical practice.
Collapse
Affiliation(s)
- Shuai Liang
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Hanyu Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Haixia Tian
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Zhicheng Xu
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Min Wu
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Dong Hua
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chengming Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
32
|
Shao J, Feng J, Li J, Liang S, Li W, Wang C. Novel tools for early diagnosis and precision treatment based on artificial intelligence. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:148-160. [PMID: 39171128 PMCID: PMC11332840 DOI: 10.1016/j.pccm.2023.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Indexed: 08/23/2024]
Abstract
Lung cancer has the highest mortality rate among all cancers in the world. Hence, early diagnosis and personalized treatment plans are crucial to improving its 5-year survival rate. Chest computed tomography (CT) serves as an essential tool for lung cancer screening, and pathology images are the gold standard for lung cancer diagnosis. However, medical image evaluation relies on manual labor and suffers from missed diagnosis or misdiagnosis, and physician heterogeneity. The rapid development of artificial intelligence (AI) has brought a whole novel opportunity for medical task processing, demonstrating the potential for clinical application in lung cancer diagnosis and treatment. AI technologies, including machine learning and deep learning, have been deployed extensively for lung nodule detection, benign and malignant classification, and subtype identification based on CT images. Furthermore, AI plays a role in the non-invasive prediction of genetic mutations and molecular status to provide the optimal treatment regimen, and applies to the assessment of therapeutic efficacy and prognosis of lung cancer patients, enabling precision medicine to become a reality. Meanwhile, histology-based AI models assist pathologists in typing, molecular characterization, and prognosis prediction to enhance the efficiency of diagnosis and treatment. However, the leap to extensive clinical application still faces various challenges, such as data sharing, standardized label acquisition, clinical application regulation, and multimodal integration. Nevertheless, AI holds promising potential in the field of lung cancer to improve cancer care.
Collapse
Affiliation(s)
- Jun Shao
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaming Feng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shufan Liang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
33
|
Li X, Younis MH, Wei W, Cai W. PD-L1 - targeted magnetic fluorescent hybrid nanoparticles: Illuminating the path of image-guided cancer immunotherapy. Eur J Nucl Med Mol Imaging 2023; 50:2240-2243. [PMID: 36943430 PMCID: PMC10272096 DOI: 10.1007/s00259-023-06202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Affiliation(s)
- Xiaoyan Li
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Muhsin H Younis
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA.
| |
Collapse
|
34
|
Evangelista L, Fiz F, Laudicella R, Bianconi F, Castello A, Guglielmo P, Liberini V, Manco L, Frantellizzi V, Giordano A, Urso L, Panareo S, Palumbo B, Filippi L. PET Radiomics and Response to Immunotherapy in Lung Cancer: A Systematic Review of the Literature. Cancers (Basel) 2023; 15:3258. [PMID: 37370869 PMCID: PMC10296704 DOI: 10.3390/cancers15123258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
The aim of this review is to provide a comprehensive overview of the existing literature concerning the applications of positron emission tomography (PET) radiomics in lung cancer patient candidates or those undergoing immunotherapy. MATERIALS AND METHODS A systematic review was conducted on databases and web sources. English-language original articles were considered. The title and abstract were independently reviewed to evaluate study inclusion. Duplicate, out-of-topic, and review papers, or editorials, articles, and letters to editors were excluded. For each study, the radiomics analysis was assessed based on the radiomics quality score (RQS 2.0). The review was registered on the PROSPERO database with the number CRD42023402302. RESULTS Fifteen papers were included, thirteen were qualified as using conventional radiomics approaches, and two used deep learning radiomics. The content of each study was different; indeed, seven papers investigated the potential ability of radiomics to predict PD-L1 expression and tumor microenvironment before starting immunotherapy. Moreover, two evaluated the prediction of response, and four investigated the utility of radiomics to predict the response to immunotherapy. Finally, two papers investigated the prediction of adverse events due to immunotherapy. CONCLUSIONS Radiomics is promising for the evaluation of TME and for the prediction of response to immunotherapy, but some limitations should be overcome.
Collapse
Affiliation(s)
- Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Francesco Fiz
- Nuclear Medicine Department, E.O. “Ospedali Galliera”, 16128 Genoa, Italy;
- Nuclear Medicine Department and Clinical Molecular Imaging, University Hospital, 72076 Tübingen, Germany
| | - Riccardo Laudicella
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90100 Palermo, Italy;
| | - Francesco Bianconi
- Department of Engineering, Università degli Studi di Perugia, Via Goffredo Duranti, 06125 Perugia, Italy;
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Priscilla Guglielmo
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy;
| | - Virginia Liberini
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy;
| | - Luigi Manco
- Medical Physics Unit, Azienda USL of Ferrara, 45100 Ferrara, Italy;
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy;
| | - Alessia Giordano
- Nuclear Medicine Unit, IRCCS CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Luca Urso
- Department of Nuclear Medicine PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, 41124 Modena, Italy;
| | - Barbara Palumbo
- Section of Nuclear Medicine and Health Physics, Department of Medicine and Surgery, Università degli Studi di Perugia, 06125 Perugia, Italy;
| | - Luca Filippi
- Nuclear Medicine Section, Santa Maria Goretti Hospital, 04100 Latina, Italy;
| |
Collapse
|
35
|
Song R, Liu F, Ping Y, Zhang Y, Wang L. Potential non-invasive biomarkers in tumor immune checkpoint inhibitor therapy: response and prognosis prediction. Biomark Res 2023; 11:57. [PMID: 37268978 DOI: 10.1186/s40364-023-00498-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/07/2023] [Indexed: 06/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically enhanced the treatment outcomes for diverse malignancies. Yet, only 15-60% of patients respond significantly. Therefore, accurate responder identification and timely ICI administration are critical issues in tumor ICI therapy. Recent rapid developments at the intersection of oncology, immunology, biology, and computer science have provided an abundance of predictive biomarkers for ICI efficacy. These biomarkers can be invasive or non-invasive, depending on the specific sample collection method. Compared with invasive markers, a host of non-invasive markers have been confirmed to have superior availability and accuracy in ICI efficacy prediction. Considering the outstanding advantages of dynamic monitoring of the immunotherapy response and the potential for widespread clinical application, we review the recent research in this field with the aim of contributing to the identification of patients who may derive the greatest benefit from ICI therapy.
Collapse
Affiliation(s)
- Ruixia Song
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou University, Zhengzhou, Henan, China
| | - Fengsen Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Ping
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou University, Zhengzhou, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China.
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
36
|
Gao Q, Yang L, Lu M, Jin R, Ye H, Ma T. The artificial intelligence and machine learning in lung cancer immunotherapy. J Hematol Oncol 2023; 16:55. [PMID: 37226190 DOI: 10.1186/s13045-023-01456-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Since the past decades, more lung cancer patients have been experiencing lasting benefits from immunotherapy. It is imperative to accurately and intelligently select appropriate patients for immunotherapy or predict the immunotherapy efficacy. In recent years, machine learning (ML)-based artificial intelligence (AI) was developed in the area of medical-industrial convergence. AI can help model and predict medical information. A growing number of studies have combined radiology, pathology, genomics, proteomics data in order to predict the expression levels of programmed death-ligand 1 (PD-L1), tumor mutation burden (TMB) and tumor microenvironment (TME) in cancer patients or predict the likelihood of immunotherapy benefits and side effects. Finally, with the advancement of AI and ML, it is believed that "digital biopsy" can replace the traditional single assessment method to benefit more cancer patients and help clinical decision-making in the future. In this review, the applications of AI in PD-L1/TMB prediction, TME prediction and lung cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Luyu Yang
- Department of Respiratory and Critical Care Medicine, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Renjing Jin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Huan Ye
- Department of Respiratory and Critical Care Medicine, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
37
|
Zhao J, Sun Z, Yu Y, Yuan Z, Lin Y, Tan Y, Duan X, Yao H, Wang Y, Liu J. Radiomic and clinical data integration using machine learning predict the efficacy of anti-PD-1 antibodies-based combinational treatment in advanced breast cancer: a multicentered study. J Immunother Cancer 2023; 11:jitc-2022-006514. [PMID: 37217246 DOI: 10.1136/jitc-2022-006514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs)-based therapy, is regarded as one of the major breakthroughs in cancer treatment. However, it is challenging to accurately identify patients who may benefit from ICIs. Current biomarkers for predicting the efficacy of ICIs require pathological slides, and their accuracy is limited. Here we aim to develop a radiomics model that could accurately predict response of ICIs for patients with advanced breast cancer (ABC). METHODS Pretreatment contrast-enhanced CT (CECT) image and clinicopathological features of 240 patients with ABC who underwent ICIs-based treatment in three academic hospitals from February 2018 to January 2022 were assigned into a training cohort and an independent validation cohort. For radiomic features extraction, CECT images of patients 1 month prior to ICIs-based therapies were first delineated with regions of interest. Data dimension reduction, feature selection and radiomics model construction were carried out with multilayer perceptron. Combined the radiomics signatures with independent clinicopathological characteristics, the model was integrated by multivariable logistic regression analysis. RESULTS Among the 240 patients, 171 from Sun Yat-sen Memorial Hospital and Sun Yat-sen University Cancer Center were evaluated as a training cohort, while other 69 from Sun Yat-sen University Cancer Center and the First Affiliated Hospital of Sun Yat-sen University were the validation cohort. The area under the curve (AUC) of radiomics model was 0.994 (95% CI: 0.988 to 1.000) in the training and 0.920 (95% CI: 0.824 to 1.000) in the validation set, respectively, which were significantly better than the performance of clinical model (0.672 for training and 0.634 for validation set). The integrated clinical-radiomics model showed increased but not statistical different predictive ability in both the training (AUC=0.997, 95% CI: 0.993 to 1.000) and validation set (AUC=0.961, 95% CI: 0.885 to 1.000) compared with the radiomics model. Furthermore, the radiomics model could divide patients under ICIs-therapies into high-risk and low-risk group with significantly different progression-free survival both in training (HR=2.705, 95% CI: 1.888 to 3.876, p<0.001) and validation set (HR=2.625, 95% CI: 1.506 to 4.574, p=0.001), respectively. Subgroup analyses showed that the radiomics model was not influenced by programmed death-ligand 1 status, tumor metastatic burden or molecular subtype. CONCLUSIONS This radiomics model provided an innovative and accurate way that could stratify patients with ABC who may benefit more from ICIs-based therapies.
Collapse
Affiliation(s)
- Jianli Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhixian Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunfang Yu
- Department of Medical Oncology, Yat-sen Supercomputer Intelligent Medical Joint Research Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Lin
- Breast Disease Center, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Yujie Tan
- Department of Medical Oncology, Yat-sen Supercomputer Intelligent Medical Joint Research Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieqiong Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Erasmus JJ, Vlahos I. Editorial Commentary: Baseline Radiomic Signature to Estimate Overall Survival in Patients With NSCLC. J Thorac Oncol 2023; 18:556-558. [PMID: 37087116 DOI: 10.1016/j.jtho.2023.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/24/2023]
Affiliation(s)
- Jeremy J Erasmus
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ioannis Vlahos
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
39
|
Bianchi A, De Rimini ML, Sciuto R, Annovazzi A, Di Traglia S, Bauckneht M, Lanfranchi F, Morbelli S, Nappi AG, Ferrari C, Rubini G, Panareo S, Urso L, Bartolomei M, D'Arienzo D, Valente T, Rossetti V, Caroli P, Matteucci F, Aricò D, Bombaci M, Caponnetto D, Bertagna F, Albano D, Dondi F, Gusella S, Spimpolo A, Carriere C, Balma M, Buschiazzo A, Gallicchio R, Storto G, Ruffini L, Scarlattei M, Baldari G, Cervino AR, Cuppari L, Burei M, Trifirò G, Brugola E, Zanini CA, Alessi A, Fuoco V, Seregni E, Deandreis D, Liberini V, Moreci AM, Ialuna S, Pulizzi S, Evangelista L. Can Baseline [18F]FDG PET/CT Predict Response to Immunotherapy After 6 Months and Overall Survival in Patients with Lung Cancer or Malignant Melanoma? A Multicenter Retrospective Study. Cancer Biother Radiopharm 2023; 38:256-267. [PMID: 37098169 DOI: 10.1089/cbr.2022.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023] Open
Abstract
Aim: To assess the role of baseline 18F-fluorodeoxyglucose ([18F]FDG)-positron emission tomography/computed tomography (PET/CT) in predicting response to immunotherapy after 6 months and overall survival (OS) in patients with lung cancer (LC) or malignant melanoma (MM). Methods: Data from a multicenter, retrospective study conducted between March and November 2021 were analyzed. Patients >18 years old with a confirmed diagnosis of LC or MM, who underwent a baseline [18F]FDG-PET/CT within 1-2 months before starting immunotherapy and had a follow-up of at least 12 months were included. PET scans were examined visually and semiquantitatively by physicians at peripheral centers. The metabolic tumor burden (number of lesions with [18F]FDG-uptake) and other parameters were recorded. Clinical response was assessed at 3 and 6 months after starting immunotherapy, and OS was calculated as the time elapsing between the PET scan and death or latest follow-up. Results: The study concerned 177 patients with LC and 101 with MM. Baseline PET/CT was positive in primary or local recurrent lesions in 78.5% and 9.9% of cases, in local/distant lymph nodes in 71.8% and 36.6%, in distant metastases in 58.8% and 84%, respectively, in LC and in MM patients. Among patients with LC, [18F]FDG-uptake in primary/recurrent lung lesions was more often associated with no clinical response to immunotherapy after 6 months than in cases without any tracer uptake. After a mean 21 months, 46.5% of patients with LC and 37.1% with MM had died. A significant correlation emerged between the site/number of [18F]FDG foci and death among patients with LC, but not among those with MM. Conclusions: In patients with LC who are candidates for immunotherapy, baseline [18F]FDG-PET/CT can help to predict response to this therapy after 6 months, and to identify those with a poor prognosis based on their metabolic parameters. For patients with MM, there was only a weak correlation between baseline PET/CT parameters, response to therapy, and survival.
Collapse
Affiliation(s)
- Andrea Bianchi
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
| | - Maria Luisa De Rimini
- Nuclear Medicine Unit, Department of Health Service, AORN Ospedali dei Colli, Naples, Italy
| | - Rosa Sciuto
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessio Annovazzi
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Di Traglia
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Matteo Bauckneht
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Lanfranchi
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Morbelli
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Anna Giulia Nappi
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Cristina Ferrari
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Giuseppe Rubini
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, Modena, Italy
| | - Luca Urso
- Nuclear Medicine Unit, Oncology and Specialistic Department, University Hospital of Ferrara, Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncology and Specialistic Department, University Hospital of Ferrara, Ferrara, Italy
| | - Davide D'Arienzo
- Nuclear Medicine Unit, Department of Health Service, AORN Ospedali dei Colli, Naples, Italy
| | - Tullio Valente
- U.O.C. Radiologia, Department of Servizi, AORN Ospedali dei Colli, Napoli, Italy
| | - Virginia Rossetti
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Paola Caroli
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Federica Matteucci
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Demetrio Aricò
- Servizio di Medicina Nucleare, Humanitas Istituto Clinico Catanese, Misterbianco, Italy
| | - Michelangelo Bombaci
- Servizio di Medicina Nucleare, Humanitas Istituto Clinico Catanese, Misterbianco, Italy
| | - Domenica Caponnetto
- Servizio di Medicina Nucleare, Humanitas Istituto Clinico Catanese, Misterbianco, Italy
| | - Francesco Bertagna
- Nuclear Medicine Unit, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Domenico Albano
- Nuclear Medicine Unit, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Francesco Dondi
- Nuclear Medicine Unit, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Sara Gusella
- Nuclear Medicine Department, Central Hospital Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Alessandro Spimpolo
- Nuclear Medicine Department, Central Hospital Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Cinzia Carriere
- Dermatology Department, Central Hospital Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Michele Balma
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
| | - Rosj Gallicchio
- Nuclear Medicine Unit, IRCCS CROB Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Giovanni Storto
- Nuclear Medicine Unit, IRCCS CROB Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Livia Ruffini
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Maura Scarlattei
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Giorgio Baldari
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Anna Rita Cervino
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Lea Cuppari
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Marta Burei
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Giuseppe Trifirò
- Servizio di Medicina Nucleare ICS MAUGERI SPA SB-IRCCS, Pavia, Italy
| | | | - Carolina Arianna Zanini
- Department of Nuclear Medicine, Università Degli Studi di Milano, Milano Statale, Milan, Italy
| | - Alessandra Alessi
- Nuclear Medicine Division, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Fuoco
- Nuclear Medicine Division, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ettore Seregni
- Nuclear Medicine Division, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Désirée Deandreis
- Nuclear Medicine Division, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Virginia Liberini
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
- Nuclear Medicine Division, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Antonino Maria Moreci
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, Palermo, Italy
| | - Salvatore Ialuna
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, Palermo, Italy
| | - Sabina Pulizzi
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, Palermo, Italy
| | - Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Padua, Italy
| |
Collapse
|
40
|
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: 10] [Impact Index Per Article: 10.0] [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
|
41
|
Malet J, Ancel J, Moubtakir A, Papathanassiou D, Deslée G, Dewolf M. Assessment of the Association between Entropy in PET/CT and Response to Anti-PD-1/PD-L1 Monotherapy in Stage III or IV NSCLC. Life (Basel) 2023; 13:life13041051. [PMID: 37109580 PMCID: PMC10142835 DOI: 10.3390/life13041051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Anti-PD-1/PD-L1 therapy indications are broadened in non-small cell lung cancer (NSCLC) although immune checkpoint inhibitors (ICI) do not provide benefits for the entire population. Texture features based on positron emission tomography/computed tomography (PET/CT), especially entropy (based on a gray-level co-occurrence matrix (GLCM)), could be interesting as predictors in NSCLC. The aim of our retrospective study was to evaluate the association between GLCM-entropy and response to anti-PD-1/PD-L1 monotherapy at the first evaluation in stage III or IV NSCLC, comparing patients with progressive disease (PD) and non-progressive disease (non-PD). In total, 47 patients were included. Response Evaluation Criteria in Solid Tumors (RECIST 1.1) were used to evaluate the response to ICI treatment (nivolumab, pembrolizumab, or atezolizumab). At the first evaluation, 25 patients were PD and 22 were non-PD. GLCM-entropy was not predictive of response at the first evaluation. Furthermore, GLCM-entropy was not associated with progression-free survival (PFS) (p = 0.393) or overall survival (OS) (p = 0.220). Finally, GLCM-entropy measured in PET/CT performed before ICI initiation in stage III or IV NSCLC was not predictive of response at the first evaluation. However, this study demonstrates the feasibility of using texture parameters in routine clinical practice. The interest of measuring PET/CT texture parameters in NSCLC remains to be evaluated in larger prospective studies.
Collapse
Affiliation(s)
- Julie Malet
- Department of Respiratory Diseases, Reims University Hospital, 45, Rue Cognacq-Jay, 51100 Reims, France
| | - Julien Ancel
- Department of Respiratory Diseases, Reims University Hospital, 45, Rue Cognacq-Jay, 51100 Reims, France
- INSERM UMRS 1250, University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Abdenasser Moubtakir
- Department of Nuclear Medicine, Institut Godinot, 1, Rue du Général Koenig, 51100 Reims, France
| | - Dimitri Papathanassiou
- Department of Nuclear Medicine, Institut Godinot, 1, Rue du Général Koenig, 51100 Reims, France
- UFR de Médecine, Reims-Champagne Ardenne University, 1, Rue Cognacq-Jay, CEDEX 51095 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l'Information et de la Communication), EA 3804, University of Reims Champagne-Ardenne, Moulin de la Housse, BP 1039, CEDEX 51687 Reims, France
| | - Gaëtan Deslée
- Department of Respiratory Diseases, Reims University Hospital, 45, Rue Cognacq-Jay, 51100 Reims, France
- INSERM UMRS 1250, University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Maxime Dewolf
- Department of Respiratory Diseases, Reims University Hospital, 45, Rue Cognacq-Jay, 51100 Reims, France
| |
Collapse
|
42
|
Ventura D, Schindler P, Masthoff M, Görlich D, Dittmann M, Heindel W, Schäfers M, Lenz G, Wardelmann E, Mohr M, Kies P, Bleckmann A, Roll W, Evers G. Radiomics of Tumor Heterogeneity in 18F-FDG-PET-CT for Predicting Response to Immune Checkpoint Inhibition in Therapy-Naïve Patients with Advanced Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:cancers15082297. [PMID: 37190228 DOI: 10.3390/cancers15082297] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
We aimed to evaluate the predictive and prognostic value of baseline 18F-FDG-PET-CT (PET-CT) radiomic features (RFs) for immune checkpoint-inhibitor (CKI)-based first-line therapy in advanced non-small-cell lung cancer (NSCLC) patients. In this retrospective study 44 patients were included. Patients were treated with either CKI-monotherapy or combined CKI-based immunotherapy-chemotherapy as first-line treatment. Treatment response was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST). After a median follow-up of 6.4 months patients were stratified into "responder" (n = 33) and "non-responder" (n = 11). RFs were extracted from baseline PET and CT data after segmenting PET-positive tumor volume of all lesions. A Radiomics-based model was developed based on a Radiomics signature consisting of reliable RFs that allow classification of response and overall progression using multivariate logistic regression. These RF were additionally tested for their prognostic value in all patients by applying a model-derived threshold. Two independent PET-based RFs differentiated well between responders and non-responders. For predicting response, the area under the curve (AUC) was 0.69 for "PET-Skewness" and 0.75 predicting overall progression for "PET-Median". In terms of progression-free survival analysis, patients with a lower value of PET-Skewness (threshold < 0.2014; hazard ratio (HR) 0.17, 95% CI 0.06-0.46; p < 0.001) and higher value of PET-Median (threshold > 0.5233; HR 0.23, 95% CI 0.11-0.49; p < 0.001) had a significantly lower probability of disease progression or death. Our Radiomics-based model might be able to predict response in advanced NSCLC patients treated with CKI-based first-line therapy.
Collapse
Affiliation(s)
- David Ventura
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center (WTZ), 48149 Muenster, Germany
| | - Philipp Schindler
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Clinic for Radiology, University and University Hospital Muenster, 48149 Muenster, Germany
| | - Max Masthoff
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Clinic for Radiology, University and University Hospital Muenster, 48149 Muenster, Germany
| | - Dennis Görlich
- Institute of Biostatistics and Clinical Research, University of Muenster, 48149 Muenster, Germany
| | - Matthias Dittmann
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center (WTZ), 48149 Muenster, Germany
| | - Walter Heindel
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Clinic for Radiology, University and University Hospital Muenster, 48149 Muenster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center (WTZ), 48149 Muenster, Germany
| | - Georg Lenz
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Department of Medicine A-Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
| | - Eva Wardelmann
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, 48149 Muenster, Germany
| | - Michael Mohr
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Department of Medicine A-Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
| | - Peter Kies
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center (WTZ), 48149 Muenster, Germany
| | - Annalen Bleckmann
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Department of Medicine A-Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany
- West German Cancer Center (WTZ), 48149 Muenster, Germany
| | - Georg Evers
- West German Cancer Center (WTZ), 48149 Muenster, Germany
- Department of Medicine A-Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Muenster, 48149 Muenster, Germany
| |
Collapse
|
43
|
Yan W, Quan C, Waleed M, Yuan J, Shi Z, Yang J, Lu Q, Zhang J. Application of radiomics in lung immuno‐oncology. PRECISION RADIATION ONCOLOGY 2023. [DOI: 10.1002/pro6.1191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Affiliation(s)
- Weisi Yan
- Baptist Health System Lexington Kentucky USA
| | - Chen Quan
- City of Hope Comprehensive Cancer Center Duarte California USA
| | - Mourad Waleed
- Department of Radiation Medicine University of Kentucky Lexington Kentucky USA
| | - Jianda Yuan
- Translational Oncology at Merck & Co Kenilworth New Jersey USA
| | | | - Jun Yang
- Foshan Chancheng Hospital Foshan Guangdong China
| | - Qiuxia Lu
- Foshan Chancheng Hospital Foshan Guangdong China
| | - Jie Zhang
- Department of Radiology University of Kentucky Lexington Kentucky USA
| |
Collapse
|
44
|
Xie J, Luo X, Deng X, Tang Y, Tian W, Cheng H, Zhang J, Zou Y, Guo Z, Xie X. Advances in artificial intelligence to predict cancer immunotherapy efficacy. Front Immunol 2023; 13:1076883. [PMID: 36685496 PMCID: PMC9845588 DOI: 10.3389/fimmu.2022.1076883] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
Tumor immunotherapy, particularly the use of immune checkpoint inhibitors, has yielded impressive clinical benefits. Therefore, it is critical to accurately screen individuals for immunotherapy sensitivity and forecast its efficacy. With the application of artificial intelligence (AI) in the medical field in recent years, an increasing number of studies have indicated that the efficacy of immunotherapy can be better anticipated with the help of AI technology to reach precision medicine. This article focuses on the current prediction models based on information from histopathological slides, imaging-omics, genomics, and proteomics, and reviews their research progress and applications. Furthermore, we also discuss the existing challenges encountered by AI in the field of immunotherapy, as well as the future directions that need to be improved, to provide a point of reference for the early implementation of AI-assisted diagnosis and treatment systems in the future.
Collapse
Affiliation(s)
- Jindong Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiyuan Luo
- School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinpei Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuhui Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenwen Tian
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hui Cheng
- School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Junsheng Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yutian Zou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China,*Correspondence: Xiaoming Xie, ; Zhixing Guo, ; Yutian Zou,
| | - Zhixing Guo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China,*Correspondence: Xiaoming Xie, ; Zhixing Guo, ; Yutian Zou,
| | - Xiaoming Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China,*Correspondence: Xiaoming Xie, ; Zhixing Guo, ; Yutian Zou,
| |
Collapse
|
45
|
Zhou H, Luo Q, Wu W, Li N, Yang C, Zou L. Radiomics-guided checkpoint inhibitor immunotherapy for precision medicine in cancer: A review for clinicians. Front Immunol 2023; 14:1088874. [PMID: 36936913 PMCID: PMC10014595 DOI: 10.3389/fimmu.2023.1088874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) is a breakthrough in oncology development and has been applied to multiple solid tumors. However, unlike traditional cancer treatment approaches, immune checkpoint inhibitors (ICIs) initiate indirect cytotoxicity by generating inflammation, which causes enlargement of the lesion in some cases. Therefore, rather than declaring progressive disease (PD) immediately, confirmation upon follow-up radiological evaluation after four-eight weeks is suggested according to immune-related Response Evaluation Criteria in Solid Tumors (ir-RECIST). Given the difficulty for clinicians to immediately distinguish pseudoprogression from true disease progression, we need novel tools to assist in this field. Radiomics, an innovative data analysis technique that quantifies tumor characteristics through high-throughput extraction of quantitative features from images, can enable the detection of additional information from early imaging. This review will summarize the recent advances in radiomics concerning immunotherapy. Notably, we will discuss the potential of applying radiomics to differentiate pseudoprogression from PD to avoid condition exacerbation during confirmatory periods. We also review the applications of radiomics in hyperprogression, immune-related biomarkers, efficacy, and immune-related adverse events (irAEs). We found that radiomics has shown promising results in precision cancer immunotherapy with early detection in noninvasive ways.
Collapse
Affiliation(s)
- Huijie Zhou
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Qian Luo
- Department of Hematology, the Second Affiliated Hospital Zhejiang University School of Medicine, Zhejiang, China
| | - Wanchun Wu
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Na Li
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Chunli Yang
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Liqun Zou
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
- *Correspondence: Liqun Zou,
| |
Collapse
|
46
|
Hu Q, Li K, Yang C, Wang Y, Huang R, Gu M, Xiao Y, Huang Y, Chen L. The role of artificial intelligence based on PET/CT radiomics in NSCLC: Disease management, opportunities, and challenges. Front Oncol 2023; 13:1133164. [PMID: 36959810 PMCID: PMC10028142 DOI: 10.3389/fonc.2023.1133164] [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: 12/28/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Objectives Lung cancer has been widely characterized through radiomics and artificial intelligence (AI). This review aims to summarize the published studies of AI based on positron emission tomography/computed tomography (PET/CT) radiomics in non-small-cell lung cancer (NSCLC). Materials and methods A comprehensive search of literature published between 2012 and 2022 was conducted on the PubMed database. There were no language or publication status restrictions on the search. About 127 articles in the search results were screened and gradually excluded according to the exclusion criteria. Finally, this review included 39 articles for analysis. Results Classification is conducted according to purposes and several studies were identified at each stage of disease:1) Cancer detection (n=8), 2) histology and stage of cancer (n=11), 3) metastases (n=6), 4) genotype (n=6), 5) treatment outcome and survival (n=8). There is a wide range of heterogeneity among studies due to differences in patient sources, evaluation criteria and workflow of radiomics. On the whole, most models show diagnostic performance comparable to or even better than experts, and the common problems are repeatability and clinical transformability. Conclusion AI-based PET/CT Radiomics play potential roles in NSCLC clinical management. However, there is still a long way to go before being translated into clinical application. Large-scale, multi-center, prospective research is the direction of future efforts, while we need to face the risk of repeatability of radiomics features and the limitation of access to large databases.
Collapse
Affiliation(s)
- Qiuyuan Hu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Conghui Yang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yue Wang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Rong Huang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Mingqiu Gu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yuqiang Xiao
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| | - Long Chen
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| |
Collapse
|
47
|
Peng B, Wang K, Xu R, Guo C, Lu T, Li Y, Wang Y, Wang C, Chang X, Shen Z, Shi J, Xu C, Zhang L. Preoperative computed tomography-based tumoral radiomic features prediction for overall survival in resectable non-small cell lung cancer. Front Oncol 2023; 13:1131816. [PMID: 37207163 PMCID: PMC10189057 DOI: 10.3389/fonc.2023.1131816] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
Objectives The purpose of this study was to evaluate whether preoperative radiomics features could meliorate risk stratification for the overall survival (OS) of non-small cell lung cancer (NSCLC) patients. Methods After rigorous screening, the 208 NSCLC patients without any pre-operative adjuvant therapy were eventually enrolled. We segmented the 3D volume of interest (VOI) based on malignant lesion of computed tomography (CT) imaging and extracted 1542 radiomics features. Interclass correlation coefficients (ICC) and LASSO Cox regression analysis were utilized to perform feature selection and radiomics model building. In the model evaluation phase, we carried out stratified analysis, receiver operating characteristic (ROC) curve, concordance index (C-index), and decision curve analysis (DCA). In addition, integrating the clinicopathological trait and radiomics score, we developed a nomogram to predict the OS at 1 year, 2 years, and 3 years, respectively. Results Six radiomics features, including gradient_glcm_InverseVariance, logarithm_firstorder_Median, logarithm_firstorder_RobustMeanAbsoluteDeviation, square_gldm_LargeDependenceEmphasis, wavelet_HLL_firstorder_Kurtosis, and wavelet_LLL_firstorder_Maximum, were selected to construct the radiomics signature, whose areas under the curve (AUCs) for 3-year prediction reached 0.857 in the training set (n=146) and 0.871 in the testing set (n=62). The results of multivariate analysis revealed that the radiomics score, radiological sign, and N stage were independent prognostic factors in NSCLC. Moreover, compared with clinical factors and the separate radiomics model, the established nomogram exhibited a better performance in predicting 3-year OS. Conclusions Our radiomics model may provide a promising non-invasive approach for preoperative risk stratification and personalized postoperative surveillance for resectable NSCLC patients.
Collapse
Affiliation(s)
- Bo Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kaiyu Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ran Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Congying Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tong Lu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongchao Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiqiao Wang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenghao Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Chang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiping Shen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxin Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengyu Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Linyou Zhang,
| |
Collapse
|
48
|
Artificial intelligence for prediction of response to cancer immunotherapy. Semin Cancer Biol 2022; 87:137-147. [PMID: 36372326 DOI: 10.1016/j.semcancer.2022.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Artificial intelligence (AI) indicates the application of machines to imitate intelligent behaviors for solving complex tasks with minimal human intervention, including machine learning and deep learning. The use of AI in medicine improves health-care systems in multiple areas such as diagnostic confirmation, risk stratification, analysis, prognosis prediction, treatment surveillance, and virtual health support, which has considerable potential to revolutionize and reshape medicine. In terms of immunotherapy, AI has been applied to unlock underlying immune signatures to associate with responses to immunotherapy indirectly as well as predict responses to immunotherapy responses directly. The AI-based analysis of high-throughput sequences and medical images can provide useful information for management of cancer immunotherapy considering the excellent abilities in selecting appropriate subjects, improving therapeutic regimens, and predicting individualized prognosis. In present review, we aim to evaluate a broad framework about AI-based computational approaches for prediction of response to cancer immunotherapy on both indirect and direct manners. Furthermore, we summarize our perspectives about challenges and opportunities of further AI applications on cancer immunotherapy relating to clinical practicability.
Collapse
|
49
|
She Y, He B, Wang F, Zhong Y, Wang T, Liu Z, Yang M, Yu B, Deng J, Sun X, Wu C, Hou L, Zhu Y, Yang Y, Hu H, Dong D, Chen C, Tian J. Deep learning for predicting major pathological response to neoadjuvant chemoimmunotherapy in non-small cell lung cancer: A multicentre study. EBioMedicine 2022; 86:104364. [PMID: 36395737 PMCID: PMC9672965 DOI: 10.1016/j.ebiom.2022.104364] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
Background This study, based on multicentre cohorts, aims to utilize computed tomography (CT) images to construct a deep learning model for predicting major pathological response (MPR) to neoadjuvant chemoimmunotherapy in non-small cell lung cancer (NSCLC) and further explore the biological basis under its prediction. Methods 274 patients undergoing curative surgery after neoadjuvant chemoimmunotherapy for NSCLC at 4 centres from January 2019 to December 2021 were included and divided into a training cohort, an internal validation cohort, and an external validation cohort. ShuffleNetV2x05-based features of the primary tumour on the CT scans within the 2 weeks preceding neoadjuvant administration were employed to develop a deep learning score for distinguishing MPR and non-MPR. To reveal the underlying biological basis of the deep learning score, a genetic analysis was conducted based on 25 patients with RNA-sequencing data. Findings MPR was achieved in 54.0% (n = 148) patients. The area under the curve (AUC) of the deep learning score to predict MPR was 0.73 (95% confidence interval [CI]: 0.58–0.86) and 0.72 (95% CI: 0.58–0.85) in the internal validation and external validation cohorts, respectively. After integrating the clinical characteristic into the deep learning score, the combined model achieved satisfactory performance in the internal validation (AUC: 0.77, 95% CI: 0.64–0.89) and external validation cohorts (AUC: 0.75, 95% CI: 0.62–0.87). In the biological basis exploration for the deep learning score, a high deep learning score was associated with the downregulation of pathways mediating tumour proliferation and the promotion of antitumour immune cell infiltration in the microenvironment. Interpretation The proposed deep learning model could effectively predict MPR in NSCLC patients treated with neoadjuvant chemoimmunotherapy. Funding This study was supported by National Key Research and Development Program of China, China (2017YFA0205200); National Natural Science Foundation of China, China (91959126, 82022036, 91959130, 81971776, 81771924, 6202790004, 81930053, 9195910169, 62176013, 8210071009); Beijing Natural Science Foundation, China (L182061); Strategic Priority Research Program of Chinese Academy of Sciences, China (XDB38040200); Chinese Academy of Sciences, China (GJJSTD20170004, QYZDJ-SSW-JSC005); Shanghai Hospital Development Center, China (SHDC2020CR3047B); and Science and Technology Commission of Shanghai Municipality, China (21YF1438200).
Collapse
|
50
|
Tankyevych O, Trousset F, Latappy C, Berraho M, Dutilh J, Tasu JP, Lamour C, Cheze Le Rest C. Development of Radiomic-Based Model to Predict Clinical Outcomes in Non-Small Cell Lung Cancer Patients Treated with Immunotherapy. Cancers (Basel) 2022; 14:cancers14235931. [PMID: 36497415 PMCID: PMC9739232 DOI: 10.3390/cancers14235931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/07/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose: We aimed to assess the ability of radiomics features extracted from baseline (PET/CT0) and follow-up PET/CT scans, as well as their evolution (delta-radiomics), to predict clinical outcome (durable clinical benefit (DCB), progression, response to therapy, OS and PFS) in non-small cell lung cancer (NSCLC) patients treated with immunotherapy. Methods: 83 NSCLC patients treated with immunotherapy who underwent a baseline PET/CT were retrospectively included. Response was assessed at 6−8 weeks (PET/CT1) using PERCIST criteria and at 3 months with iPERCIST (PET/CT2) or RECIST 1.1 criteria using CT. The predictive performance of clinical parameters (CP), standard PET metrics (SUV, Metabolic Tumor volume, Total Lesion Glycolysis), delta-radiomics and PET and CT radiomics features extracted at baseline and during follow-up were studied. Seven multivariate models with different combinations of CP and radiomics were trained on a subset of patients (75%) using least absolute shrinkage, selection operator (LASSO) and random forest classification with 10-fold cross-validation to predict outcome. Model validation was performed on the remaining patients (25%). Overall and progression-free survival was also performed by Kaplan−Meier survival analysis. Results: Numerous radiomics and delta-radiomics parameters had a high individual predictive value of patient outcome with areas under receiver operating characteristics curves (AUCs) >0.80. Their performance was superior to that of CP and standard PET metrics. Several multivariate models were also promising, especially for the prediction of progression (AUCs of 1 and 0.96 for the training and testing subsets with the PET-CT model (PET/CT0)) or DCB (AUCs of 0.85 and 0.83 with the PET-CT-CP model (PET/CT0)). Conclusions: Delta-radiomics and radiomics features extracted from baseline and follow-up PET/CT images could predict outcome in NSCLC patients treated with immunotherapy and identify patients who would benefit from this new standard. These data reinforce the rationale for the use of advanced image analysis of PET/CT scans to further improve personalized treatment management in advanced NSCLC.
Collapse
Affiliation(s)
- Olena Tankyevych
- Nuclear Medicine Department, Poitiers University Hospital, 86000 Poitiers, France
- LaTIM, INSERM, UMR 1101, 29200 Brest, France
| | - Flora Trousset
- Nuclear Medicine Department, Poitiers University Hospital, 86000 Poitiers, France
| | - Claire Latappy
- Nuclear Medicine Department, Poitiers University Hospital, 86000 Poitiers, France
| | - Moran Berraho
- Nuclear Medicine Department, Poitiers University Hospital, 86000 Poitiers, France
| | - Julien Dutilh
- Oncology Department, Poitiers University Hospital, 86000 Poitiers, France
| | - Jean Pierre Tasu
- LaTIM, INSERM, UMR 1101, 29200 Brest, France
- Medical School, University of Poitiers, 86000 Poitiers, France
- Radiology Department, Poitiers University Hospital, 86000 Poitiers, France
| | - Corinne Lamour
- Oncology Department, Poitiers University Hospital, 86000 Poitiers, France
| | - Catherine Cheze Le Rest
- Nuclear Medicine Department, Poitiers University Hospital, 86000 Poitiers, France
- LaTIM, INSERM, UMR 1101, 29200 Brest, France
- Medical School, University of Poitiers, 86000 Poitiers, France
- Correspondence:
| |
Collapse
|