1
|
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
|
2
|
Zhang M, Li X, Zhou P, Zhang P, Wang G, Lin X. Prediction value study of breast cancer tumor infiltrating lymphocyte levels based on ultrasound imaging radiomics. Front Oncol 2024; 14:1411261. [PMID: 38903726 PMCID: PMC11187250 DOI: 10.3389/fonc.2024.1411261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Objective Construct models based on grayscale ultrasound and radiomics and compare the efficacy of different models in preoperatively predicting the level of tumor-infiltrating lymphocytes in breast cancer. Materials and methods This study retrospectively collected clinical data and preoperative ultrasound images from 185 breast cancer patients confirmed by surgical pathology. Patients were randomly divided into a training set (n=111) and a testing set (n=74) using a 6:4 ratio. Based on a 10% threshold for tumor-infiltrating lymphocytes (TIL) levels, patients were classified into low-level and high-level groups. Radiomic features were extracted and selected using the training set. The evaluation included assessing the relationship between TIL levels and both radiomic features and grayscale ultrasound features. Subsequently, grayscale ultrasound models, radiomic models, and nomograms combining radiomics score (Rad-score) and grayscale ultrasound features were established. The predictive performance of different models was evaluated through receiver operating characteristic (ROC) analysis. Calibration curves assessed the fit of the nomograms, and decision curve analysis (DCA) evaluated the clinical effectiveness of the models. Results Univariate analyses and multivariate logistic regression analyses revealed that indistinct margin (P<0.001, Odds Ratio [OR]=0.214, 95% Confidence Interval [CI]: 0.103-1.026), posterior acoustic enhancement (P=0.027, OR=2.585, 95% CI: 1.116-5.987), and ipsilateral axillary lymph node enlargement (P=0.001, OR=4.214, 95% CI: 1.798-9.875) were independent predictive factors for high levels of TIL in breast cancer. In comparison to grayscale ultrasound model (Training set: Area under curve [AUC] 0.795; Testing set: AUC 0.720) and radiomics model (Training set: AUC 0.803; Testing set: AUC 0.759), the nomogram demonstrated superior discriminative ability on both the training (AUC 0.884) and testing (AUC 0.820) datasets. Calibration curves indicated high consistency between the nomogram model's predicted probability of breast cancer TIL levels and the actual occurrence probability. DCA revealed that the radiomics model and the nomogram model achieved higher clinical net benefits compared to the grayscale ultrasound model. Conclusion The nomogram based on preoperative ultrasound radiomics features exhibits robust predictive capacity for the non-invasive evaluation of breast cancer TIL levels, potentially providing a significant basis for individualized treatment decisions in breast cancer.
Collapse
Affiliation(s)
- Min Zhang
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Xuanyu Li
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Pin Zhou
- Department of Pathology, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China
| | - Panpan Zhang
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China
| | - Gang Wang
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China
| | - Xianfang Lin
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China
| |
Collapse
|
3
|
Hu L, Jin P, Xu W, Wang C, Huang P. Clinical and radiomics integrated nomogram for preoperative prediction of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer. Front Oncol 2024; 14:1370466. [PMID: 38567151 PMCID: PMC10985173 DOI: 10.3389/fonc.2024.1370466] [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: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Objectives The present study aimed to develop a radiomics nomogram based on conventional ultrasound (CUS) to preoperatively distinguish high tumor-infiltrating lymphocytes (TILs) and low TILs in triple-negative breast cancer (TNBC) patients. Methods In the present study, 145 TNBC patients were retrospectively included. Pathological evaluation of TILs in the hematoxylin and eosin sections was set as the gold standard. The patients were randomly allocated into training dataset and validation dataset with a ratio of 7:3. Clinical features (age and CUS features) and radiomics features were collected. Then, the Rad-score model was constructed after the radiomics feature selection. The clinical features model and clinical features plus Rad-score (Clin+RS) model were built using logistic regression analysis. Furthermore, the performance of the models was evaluated by analyzing the receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). Results Univariate analysis and LASSO regression were employed to identify a subset of 25 radiomics features from a pool of 837 radiomics features, followed by the calculation of Rad-score. The Clin+RS integrated model, which combined posterior echo and Rad-score, demonstrated better predictive performance compared to both the Rad-score model and clinical model, achieving AUC values of 0.848 in the training dataset and 0.847 in the validation dataset. Conclusion The Clin+RS integrated model, incorporating posterior echo and Rad-score, demonstrated an acceptable preoperative evaluation of the TIL level. The Clin+RS integrated nomogram holds tremendous potential for preoperative individualized prediction of the TIL level in TNBC.
Collapse
Affiliation(s)
- Ling Hu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Ultrasound in Medicine, Hangzhou Women’s Hospital, Hangzhou, Zhejiang, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wen Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao Wang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Landén AH, Chin K, Kovács A, Holmberg E, Molnar E, Stenmark Tullberg A, Wärnberg F, Karlsson P. Evaluation of tumor-infiltrating lymphocytes and mammographic density as predictors of response to neoadjuvant systemic therapy in breast cancer. Acta Oncol 2023; 62:1862-1872. [PMID: 37934084 DOI: 10.1080/0284186x.2023.2274483] [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: 06/16/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Response rates vary among breast cancer patients treated with neoadjuvant systemic therapy (NAST). Thus, there is a need for reliable treatment predictors. Evidence suggests tumor-infiltrating lymphocytes (TILs) predict NAST response. Still, TILs are seldom used clinically as a treatment determinant. Mammographic density (MD) is another potential marker for NAST benefit and its relationship with TILs is unknown. Our aims were to investigate TILs and MD as predictors of NAST response and to study the unexplored relationship between TILs and MD. MATERIAL AND METHODS We studied 315 invasive breast carcinomas treated with NAST between 2013 and 2020. Clinicopathological data were retrieved from medical records. The endpoint was defined as pathological complete response (pCR) in the breast. TILs were evaluated in pre-treatment core biopsies and categorized as high (≥10%) or low (<10%). MD was scored (a-d) according to the breast imaging reporting and data system (BI-RADS) fifth edition. Binary logistic regression and Spearman's test of correlation were performed using SPSS. RESULTS Out of 315 carcinomas, 136 achieved pCR. 94 carcinomas had high TILs and 215 had low TILs. Six carcinomas had no available TIL data. The number of carcinomas in each BI-RADS category were 37, 122, 112, and 44 for a, b, c, and d, respectively. High TILs were independently associated with pCR (OR: 2.95; 95% CI: 1.59-5.46) compared to low TILs. In the univariable analysis, MD (BI-RADS d vs. a) showed a tendency of higher likelihood for pCR (OR: 2.43; 95% CI: 0.99-5.98). However, the association was non-significant, which is consistent with the result of the multivariable analysis (OR: 2.51; 95% CI: 0.78-8.04). We found no correlation between TILs and MD (0.02; p = .80). CONCLUSION TILs significantly predicted NAST response. We could not define MD as a significant predictor of NAST response. These findings should be further replicated.
Collapse
Affiliation(s)
- Amalia H Landén
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kian Chin
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Erik Holmberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eva Molnar
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Axel Stenmark Tullberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Wärnberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Ansari G, Mirza-Aghazadeh-Attari M, Mohseni A, Madani SP, Shahbazian H, Pawlik TM, Kamel IR. Response Assessment of Primary Liver Tumors to Novel Therapies: an Imaging Perspective. J Gastrointest Surg 2023; 27:2245-2259. [PMID: 37464140 DOI: 10.1007/s11605-023-05762-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/11/2023] [Indexed: 07/20/2023]
Abstract
The latest developments in cancer immunotherapy, namely the introduction of immune checkpoint inhibitors, have led to a fundamental change in advanced cancer treatments. Imaging is crucial to identify tumor response accurately and delineate prognosis in immunotherapy-treated patients. Simultaneously, advances in image acquisition techniques, notably functional and molecular imaging, have facilitated more accurate pretreatment evaluation, assessment of response to therapy, and monitoring for tumor recurrence. Traditional approaches to assessing tumor progression, such as RECIST, rely on changes in tumor size, while new strategies for evaluating tumor response to therapy, such as the mRECIST and the EASL, rely on tumor enhancement. Moreover, the assessment of tumor volume, enhancement, cellularity, and perfusion are some novel techniques that have been investigated. Validation of these novel approaches should rely on comparing their results with those of standard evaluation methods (EASL, mRECIST) while considering the ultimate outcome, which is patient survival. More recently, immunotherapy has been used in the management of primary liver tumors. However, little is known about its efficacy. This article reviews imaging modalities and techniques for assessing tumor response and survival in immunotherapy-treated patients with primary hepatic malignancies.
Collapse
Affiliation(s)
- Golnoosh Ansari
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Mohammad Mirza-Aghazadeh-Attari
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Timothy M Pawlik
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, James Comprehensive Cancer Center, Columbus, OH, USA
| | - Ihab R Kamel
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA.
| |
Collapse
|
6
|
Kang W, Qiu X, Luo Y, Luo J, Liu Y, Xi J, Li X, Yang Z. Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosis. J Transl Med 2023; 21:598. [PMID: 37674169 PMCID: PMC10481579 DOI: 10.1186/s12967-023-04437-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
The advent of immunotherapy, a groundbreaking advancement in cancer treatment, has given rise to the prominence of the tumor microenvironment (TME) as a critical area of research. The clinical implications of an improved understanding of the TME are significant and far-reaching. Radiomics has been increasingly utilized in the comprehensive assessment of the TME and cancer prognosis. Similarly, the advancement of pathomics, which is based on pathological images, can offer additional insights into the panoramic view and microscopic information of tumors. The combination of pathomics and radiomics has revolutionized the concept of a "digital biopsy". As genomics and transcriptomics continue to evolve, integrating radiomics with genomic and transcriptomic datasets can offer further insights into tumor and microenvironment heterogeneity and establish correlations with biological significance. Therefore, the synergistic analysis of digital image features (radiomics, pathomics) and genetic phenotypes (genomics) can comprehensively decode and characterize the heterogeneity of the TME as well as predict cancer prognosis. This review presents a comprehensive summary of the research on important radiomics biomarkers for predicting the TME, emphasizing the interplay between radiomics, genomics, transcriptomics, and pathomics, as well as the application of multiomics in decoding the TME and predicting cancer prognosis. Finally, we discuss the challenges and opportunities in multiomics research. In conclusion, this review highlights the crucial role of radiomics and multiomics associations in the assessment of the TME and cancer prognosis. The combined analysis of radiomics, pathomics, genomics, and transcriptomics is a promising research direction with substantial research significance and value for comprehensive TME evaluation and cancer prognosis assessment.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiang Qiu
- Obstetrics and Gynecology Hospital of, Fudan University, Shanghai, 200011, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Jianwei Luo
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yang Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junqing Xi
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
7
|
Ma Q, Shen C, Gao Y, Duan Y, Li W, Lu G, Qin X, Zhang C, Wang J. Radiomics Analysis of Breast Lesions in Combination with Coronal Plane of ABVS and Strain Elastography. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:381-390. [PMID: 37260586 PMCID: PMC10228588 DOI: 10.2147/bctt.s410356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Background Breast cancer is the most common tumor globally. Automated Breast Volume Scanner (ABVS) and strain elastography (SE) can provide more useful breast information. The use of radiomics combined with ABVS and SE images to predict breast cancer has become a new focus. Therefore, this study developed and validated a radiomics analysis of breast lesions in combination with coronal plane of ABVS and SE to improve the differential diagnosis of benign and malignant breast diseases. Patients and Methods 620 pathologically confirmed breast lesions from January 2017 to August 2021 were retrospectively analyzed and randomly divided into a training set (n=434) and a validation set (n=186). Radiomic features of the lesions were extracted from ABVS, B-ultrasound, and strain elastography (SE) images, respectively. These were then filtered by Gradient Boosted Decision Tree (GBDT) and multiple logistic regression. The ABVS model is based on coronal plane features for the breast, B+SE model is based on features of B-ultrasound and SE, and the multimodal model is based on features of three examinations. The evaluation of the predicted performance of the three models used the receiver operating characteristic (ROC) and decision curve analysis (DCA). Results The area under the curve, accuracy, specificity, and sensitivity of the multimodal model in the training set are 0.975 (95% CI:0.959-0.991),93.78%, 92.02%, and 96.49%, respectively, and 0.946 (95% CI:0.913 -0.978), 87.63%, 83.93%, and 93.24% in the validation set, respectively. The multimodal model outperformed the ABVS model and B+SE model in both the training (P < 0.001, P = 0.002, respectively) and validation sets (P < 0.001, P = 0.034, respectively). Conclusion Radiomics from the coronal plane of the breast lesion provide valuable information for identification. A multimodal model combination with radiomics from ABVS, B-ultrasound, and SE could improve the diagnostic efficacy of breast masses.
Collapse
Affiliation(s)
- Qianqing Ma
- Department of Ultrasound, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Chunyun Shen
- Department of Ultrasound, Wuhu No. 2 People’s Hospital, Wuhu, People’s Republic of China
| | - Yankun Gao
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Yayang Duan
- Department of Ultrasound, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Wanyan Li
- Department of Ultrasound, Linquan Country People’s Hospital, Fuyang, People’s Republic of China
| | - Gensheng Lu
- Department of Pathology, Wuhu No. 2 People’s Hospital, Wuhu, People’s Republic of China
| | - Xiachuan Qin
- Department of Ultrasound, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Chaoxue Zhang
- Department of Ultrasound, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Junli Wang
- Department of Ultrasound, Wuhu No. 2 People’s Hospital, Wuhu, People’s Republic of China
| |
Collapse
|
8
|
Frankowska K, Zarobkiewicz M, Dąbrowska I, Bojarska-Junak A. Tumor infiltrating lymphocytes and radiological picture of the tumor. Med Oncol 2023; 40:176. [PMID: 37178270 PMCID: PMC10182948 DOI: 10.1007/s12032-023-02036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
Tumor microenvironment (TME) is a complex entity that includes besides the tumor cells also a whole range of immune cells. Among various populations of immune cells infiltrating the tumor, tumor infiltrating lymphocytes (TILs) are a population of lymphocytes characterized by high reactivity against the tumor component. As, TILs play a key role in mediating responses to several types of therapy and significantly improve patient outcomes in some cancer types including for instance breast cancer and lung cancer, their assessment has become a good predictive tool in the evaluation of potential treatment efficacy. Currently, the evaluation of the density of TILs infiltration is performed by histopathological. However, recent studies have shed light on potential utility of several imaging methods, including ultrasonography, magnetic resonance imaging (MRI), positron emission tomography-computed tomography (PET-CT), and radiomics, in the assessment of TILs levels. The greatest attention concerning the utility of radiology methods is directed to breast and lung cancers, nevertheless imaging methods of TILs are constantly being developed also for other malignancies. Here, we focus on reviewing the radiological methods used to assess the level of TILs in different cancer types and on the extraction of the most favorable radiological features assessed by each method.
Collapse
Affiliation(s)
- Karolina Frankowska
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | - Michał Zarobkiewicz
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland.
| | - Izabela Dąbrowska
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
9
|
Jeon SH, Kim SW, Na K, Seo M, Sohn YM, Lim YJ. Radiomic models based on magnetic resonance imaging predict the spatial distribution of CD8 + tumor-infiltrating lymphocytes in breast cancer. Front Immunol 2022; 13:1080048. [PMID: 36601118 PMCID: PMC9806253 DOI: 10.3389/fimmu.2022.1080048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Infiltration of CD8+ T cells and their spatial contexture, represented by immunophenotype, predict the prognosis and therapeutic response in breast cancer. However, a non-surgical method using radiomics to evaluate breast cancer immunophenotype has not been explored. Here, we assessed the CD8+ T cell-based immunophenotype in patients with breast cancer undergoing upfront surgery (n = 182). We extracted radiomic features from the four phases of dynamic contrast-enhanced magnetic resonance imaging, and randomly divided the patients into training (n = 137) and validation (n = 45) cohorts. For predicting the immunophenotypes, radiomic models (RMs) that combined the four phases demonstrated superior performance to those derived from a single phase. For discriminating the inflamed tumor from the non-inflamed tumor, the feature-based combination model from the whole tumor (RM-wholeFC) showed high performance in both training (area under the receiver operating characteristic curve [AUC] = 0.973) and validation cohorts (AUC = 0.985). Similarly, the feature-based combination model from the peripheral tumor (RM-periFC) discriminated between immune-desert and excluded tumors with high performance in both training (AUC = 0.993) and validation cohorts (AUC = 0.984). Both RM-wholeFC and RM-periFC demonstrated good to excellent performance for every molecular subtype. Furthermore, in patients who underwent neoadjuvant chemotherapy (n = 64), pre-treatment images showed that tumors exhibiting complete response to neoadjuvant chemotherapy had significantly higher scores from RM-wholeFC and lower scores from RM-periFC. Our RMs predicted the immunophenotype of breast cancer based on the spatial distribution of CD8+ T cells with high accuracy. This approach can be used to stratify patients non-invasively based on the status of the tumor-immune microenvironment.
Collapse
Affiliation(s)
- Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - So-Woon Kim
- Department of Pathology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Mirinae Seo
- Department of Radiology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Yu-Mee Sohn
- Department of Radiology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Yu Jin Lim
- Department of Radiation Oncology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea,*Correspondence: Yu Jin Lim,
| |
Collapse
|
10
|
Su GH, Xiao Y, Jiang L, Zheng RC, Wang H, Chen Y, Gu YJ, You C, Shao ZM. Radiomics features for assessing tumor-infiltrating lymphocytes correlate with molecular traits of triple-negative breast cancer. Lab Invest 2022; 20:471. [PMID: 36243806 PMCID: PMC9571493 DOI: 10.1186/s12967-022-03688-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) have become a promising biomarker for assessing tumor immune microenvironment and predicting immunotherapy response. However, the assessment of TILs relies on invasive pathological slides. METHODS We retrospectively extracted radiomics features from magnetic resonance imaging (MRI) to develop a radiomic cohort of triple-negative breast cancer (TNBC) (n = 139), among which 116 patients underwent transcriptomic sequencing. This radiomic cohort was randomly divided into the training cohort (n = 98) and validation cohort (n = 41) to develop radiomic signatures to predict the level of TILs through a non-invasive method. Pathologically evaluated TILs in the H&E sections were set as the gold standard. Elastic net and logistic regression were utilized to perform radiomics feature selection and model training, respectively. Transcriptomics was utilized to infer the detailed composition of the tumor microenvironment and to validate the radiomic signatures. RESULTS We selected three radiomics features to develop a TILs-predicting radiomics model, which performed well in the validation cohort (AUC 0.790, 95% confidence interval (CI) 0.638-0.943). Further investigation with transcriptomics verified that tumors with high TILs predicted by radiomics (Rad-TILs) presented activated immune-related pathways, such as antigen processing and presentation, and immune checkpoints pathways. In addition, a hot immune microenvironment, including upregulated T cell infiltration gene signatures, cytokines, costimulators and major histocompatibility complexes (MHCs), as well as more CD8+ T cells, follicular helper T cells and memory B cells, was found in high Rad-TILs tumors. CONCLUSIONS Our study demonstrated the feasibility of radiomics model in predicting TILs status and provided a method to make the features interpretable, which will pave the way toward precision medicine for TNBC.
Collapse
Affiliation(s)
- Guan-Hua Su
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi Xiao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lin Jiang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ren-Cheng Zheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 201203, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 201203, China
| | - Yan Chen
- Division of Cancer and Stem Cell, School of Medicine at University of Nottingham, Nottingham, UK
| | - Ya-Jia Gu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
| | - Chao You
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
11
|
Jiang X, Song J, Duan S, Cheng W, Chen T, Liu X. MRI radiomics combined with clinicopathologic features to predict disease-free survival in patients with early-stage cervical cancer. Br J Radiol 2022; 95:20211229. [PMID: 35604668 PMCID: PMC10162065 DOI: 10.1259/bjr.20211229] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/21/2022] [Accepted: 05/06/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To establish a comprehensive model including MRI radiomics and clinicopathological features to predict post-operative disease-free survival (DFS) in early-stage (pre-operative FIGO Stage IB-IIA) cervical cancer. METHODS A total of 183 patients with early-stage cervical cancer admitted to our Jiangsu Province Hospital underwent radical hysterectomy were enrolled in this retrospective study from January 2013 to June 2018 and their clinicopathology and MRI information were collected. They were then divided into training cohort (n = 129) and internal validation cohort (n = 54). The radiomic features were extracted from the pre-operative T1 contrast-enhanced (T1CE) and T2 weighted image of each patient. Least absolute shrinkage and selection operator regression and multivariate Cox proportional hazard model were used for feature selection, and the rad-score (RS) of each patient were evaluated individually. The clinicopathology model, T1CE_RS model, T1CE + T2_RS model, and clinicopathology combined with T1CE_RS model were established and compared. Patients were divided into high- and low-risk groups according to the optimum cut-off values of four models. RESULTS T1CE_RS model showed better performance on DFS prediction of early-stage cervical cancer than clinicopathological model (C-index: 0.724 vs 0.659). T1CE+T2_RS model did not improve predictive performance (C-index: 0.671). The combination of T1CE_RS and clinicopathology features showed more accurate predictive ability (C-index=0.773). CONCLUSION The combination of T1CE_RS and clinicopathology features showed more accurate predictive performance for DFS of patients with early-stage (pre-operative IB-IIA) cervical cancer which can aid in the design of individualised treatment strategies and regular follow-up. ADVANCES IN KNOWLEDGE A radiomics signature composed of T1CE radiomic features combined with clinicopathology features allowed differentiating patients at high or low risk of recurrence.
Collapse
Affiliation(s)
- Xiaoting Jiang
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiacheng Song
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shaofeng Duan
- GE Healthcare, Precision Health Institution, Shanghai, China
| | - Wenjun Cheng
- Department of Gynaecology and Obstetrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Chen
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xisheng Liu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Kang CY, Duarte SE, Kim HS, Kim E, Park J, Lee AD, Kim Y, Kim L, Cho S, Oh Y, Gim G, Park I, Lee D, Abazeed M, Velichko YS, Chae YK. OUP accepted manuscript. Oncologist 2022; 27:e471-e483. [PMID: 35348765 PMCID: PMC9177100 DOI: 10.1093/oncolo/oyac036] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/14/2022] [Indexed: 11/17/2022] Open
Abstract
The recent, rapid advances in immuno-oncology have revolutionized cancer treatment and spurred further research into tumor biology. Yet, cancer patients respond variably to immunotherapy despite mounting evidence to support its efficacy. Current methods for predicting immunotherapy response are unreliable, as these tests cannot fully account for tumor heterogeneity and microenvironment. An improved method for predicting response to immunotherapy is needed. Recent studies have proposed radiomics—the process of converting medical images into quantitative data (features) that can be processed using machine learning algorithms to identify complex patterns and trends—for predicting response to immunotherapy. Because patients undergo numerous imaging procedures throughout the course of the disease, there exists a wealth of radiological imaging data available for training radiomics models. And because radiomic features reflect cancer biology, such as tumor heterogeneity and microenvironment, these models have enormous potential to predict immunotherapy response more accurately than current methods. Models trained on preexisting biomarkers and/or clinical outcomes have demonstrated potential to improve patient stratification and treatment outcomes. In this review, we discuss current applications of radiomics in oncology, followed by a discussion on recent studies that use radiomics to predict immunotherapy response and toxicity.
Collapse
Affiliation(s)
| | | | - Hye Sung Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eugene Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Alice Daeun Lee
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yeseul Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leeseul Kim
- Department of Internal Medicine, AMITA Health Saint Francis Hospital, Evanston, IL, USA
| | - Sukjoo Cho
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yoojin Oh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gahyun Gim
- Department of Hematology and Oncology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Inae Park
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dongyup Lee
- Department of Physical Medicine and Rehabilitation, Geisinger Health System, Danville, PA, USA
| | - Mohamed Abazeed
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yury S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Young Kwang Chae
- Corresponding author: Young Kwang Chae, Department of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
13
|
Huang Z, Lyu M, Ai Z, Chen Y, Liang Y, Xiang Z. Pre-operative Prediction of Ki-67 Expression in Various Histological Subtypes of Lung Adenocarcinoma Based on CT Radiomic Features. Front Surg 2021; 8:736737. [PMID: 34733879 PMCID: PMC8558627 DOI: 10.3389/fsurg.2021.736737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/09/2021] [Indexed: 12/26/2022] Open
Abstract
Purpose: The aims of this study were to combine CT images with Ki-67 expression to distinguish various subtypes of lung adenocarcinoma and to pre-operatively predict the Ki-67 expression level based on CT radiomic features. Methods: Data from 215 patients with 237 pathologically proven lung adenocarcinoma lesions who underwent CT and immunohistochemical Ki-67 from January 2019 to April 2021 were retrospectively analyzed. The receiver operating curve (ROC) identified the Ki-67 cut-off value for differentiating subtypes of lung adenocarcinoma. A chi-square test or t-test analyzed the differences in the CT images between the negative expression group (n = 132) and the positive expression group (n = 105), and then the risk factors affecting the expression level of Ki-67 were evaluated. Patients were randomly divided into a training dataset (n = 165) and a validation dataset (n = 72) in a ratio of 7:3. A total of 1,316 quantitative radiomic features were extracted from the Analysis Kinetics (A.K.) software. Radiomic feature selection and radiomic classifier were generated through a least absolute shrinkage and selection operator (LASSO) regression and logistic regression analysis model. The predictive capacity of the radiomic classifiers for the Ki-67 levels was investigated through the ROC curves in the training and testing groups. Results: The cut-off value of the Ki-67 to distinguish subtypes of lung adenocarcinoma was 5%. A comparison of clinical data and imaging features between the two groups showed that histopathological subtypes and air bronchograms could be used as risk factors to evaluate the expression of Ki-67 in lung adenocarcinoma (p = 0.005, p = 0.045, respectively). Through radiomic feature selection, eight top-class features constructed the radiomic model to pre-operatively predict the expression of Ki-67, and the area under the ROC curves of the training group and the testing group were 0.871 and 0.8, respectively. Conclusion: Ki-67 expression level with a cut-off value of 5% could be used to differentiate non-invasive lung adenocarcinomas from invasive lung adenocarcinomas. It is feasible and reliable to pre-operatively predict the expression level of Ki-67 in lung adenocarcinomas based on CT radiomic features, as a non-invasive biomarker to predict the degree of malignant invasion of lung adenocarcinoma, and to evaluate the prognosis of the tumor.
Collapse
Affiliation(s)
- Zhiwei Huang
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Mo Lyu
- Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, China.,School of Life Sciences, South China Normal University, Guangzhou, China
| | - Zhu Ai
- Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Yirong Chen
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Yuying Liang
- Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Zhiming Xiang
- Department of Radiology, Guangzhou Panyu Central Hospital, Guangzhou, China
| |
Collapse
|