1
|
Gou X, Feng A, Feng C, Cheng J, Hong N. Imaging genomics of cancer: a bibliometric analysis and review. Cancer Imaging 2025; 25:24. [PMID: 40038813 DOI: 10.1186/s40644-025-00841-9] [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: 12/09/2024] [Accepted: 02/13/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Imaging genomics is a burgeoning field that seeks to connections between medical imaging and genomic features. It has been widely applied to explore heterogeneity and predict responsiveness and disease progression in cancer. This review aims to assess current applications and advancements of imaging genomics in cancer. METHODS Literature on imaging genomics in cancer was retrieved and selected from PubMed, Web of Science, and Embase before July 2024. Detail information of articles, such as systems and imaging features, were extracted and analyzed. Citation information was extracted from Web of Science and Scopus. Additionally, a bibliometric analysis of the included studies was conducted using the Bibliometrix R package and VOSviewer. RESULTS A total of 370 articles were included in the study. The annual growth rate of articles on imaging genomics in cancer is 24.88%. China (133) and the USA (107) were the most productive countries. The top 2 keywords plus were "survival" and "classification". The current research mainly focuses on the central nervous system (121) and the genitourinary system (110, including 44 breast cancer articles). Despite different systems utilizing different imaging modalities, more than half of the studies in each system employed radiomics features. CONCLUSIONS Publication databases provide data support for imaging genomics research. The development of artificial intelligence algorithms, especially in feature extraction and model construction, has significantly advanced this field. It is conducive to enhancing the related-models' interpretability. Nonetheless, challenges such as the sample size and the standardization of feature extraction and model construction must overcome. And the research trends revealed in this study will guide the development of imaging genomics in the future and contribute to more accurate cancer diagnosis and treatment in the clinic.
Collapse
Affiliation(s)
- Xinyi Gou
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Aobo Feng
- College of Computer and Information, Inner Mongolia Medical University, Inner Mongolia, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Beijing, China.
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
2
|
Bitar R, Salem R, Finn R, Greten TF, Goldberg SN, Chapiro J, Atzen S. Interventional Oncology Meets Immuno-oncology: Combination Therapies for Hepatocellular Carcinoma. Radiology 2024; 313:e232875. [PMID: 39560477 PMCID: PMC11605110 DOI: 10.1148/radiol.232875] [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: 10/28/2023] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 11/20/2024]
Abstract
The management of hepatocellular carcinoma (HCC) is undergoing transformational changes due to the emergence of various novel immunotherapies and their combination with image-guided locoregional therapies. In this setting, immunotherapy is expected to become one of the standards of care in both neoadjuvant and adjuvant settings across all disease stages of HCC. Currently, more than 50 ongoing prospective clinical trials are investigating various end points for the combination of immunotherapy with both percutaneous and catheter-directed therapies. This review will outline essential tumor microenvironment mechanisms responsible for disease evolution and therapy resistance, discuss the rationale for combining locoregional therapy with immunotherapy, summarize ongoing clinical trials, and report on developing imaging end points and novel biomarkers that are relevant to both diagnostic and interventional radiologists participating in the management of HCC.
Collapse
Affiliation(s)
- Ryan Bitar
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Riad Salem
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Richard Finn
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Tim F. Greten
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - S. Nahum Goldberg
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Julius Chapiro
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Sarah Atzen
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| |
Collapse
|
3
|
Casillas Meléndez C. Ways of analysing extracellular gadolinium enhancement. RADIOLOGIA 2024; 66 Suppl 2:S65-S74. [PMID: 39603742 DOI: 10.1016/j.rxeng.2024.11.001] [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: 02/26/2024] [Accepted: 04/03/2024] [Indexed: 11/29/2024]
Abstract
The use of extracellular gadolinium-based contrast agents provides valuable information in magnetic resonance studies, thus increasing diagnostic confidence. These contrast agents make it easier to detect and define injuries, and narrow down the differential diagnosis. They are indicated for several different reasons, both for diagnostic purposes and for evaluating the response to treatment. Morphological analysis can assess the type of uptake, the qualitative and semiquantitative study of the signal intensity vs time curves in multiphase sequences, and the quantitative analysis of the uptake with T1 or T2* perfusion studies associated with pharmacokinetic models. Multiphase dynamic studies with 3D sequences contain valuable information that is not exploited by a simple visual analysis of 2D images. To take advantage of this information and the imaging biomarkers provided, computational analysis should be used. To this end, the future role of artificial intelligence is increasingly evident.
Collapse
Affiliation(s)
- C Casillas Meléndez
- Servicio de Diagnóstico por Imagen, Consorcio Hospitalario Provincial de Castellón, Castellón, Spain.
| |
Collapse
|
4
|
Tang S, Fan T, Wang X, Yu C, Zhang C, Zhou Y. Cancer Immunotherapy and Medical Imaging Research Trends from 2003 to 2023: A Bibliometric Analysis. J Multidiscip Healthc 2024; 17:2105-2120. [PMID: 38736544 PMCID: PMC11086400 DOI: 10.2147/jmdh.s457367] [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: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose With the rapid development of immunotherapy, cancer treatment has entered a new phase. Medical imaging, as a primary diagnostic method, is closely related to cancer immunotherapy. However, until now, there has been no systematic bibliometric analysis of the state of this field. Therefore, the main purpose of this article is to clarify the past research trajectory, summarize current research hotspots, reveal dynamic scientific developments, and explore future research directions. Patients and Methods A comprehensive search was conducted on the Web of Science Core Collection (WoSCC) database to identify publications related to immunotherapy specifically for the medical imaging of carcinoma. The search spanned the period from the year 2003 to 2023. Several analytical tools were employed. These included CiteSpace (6.2.4), and the Microsoft Office Excel (2016). Results By searching the database, a total of 704 English articles published between 2003 and 2023 were obtained. We have observed a rapid increase in the number of publications since 2018. The two most active countries are the United States (n=265) and China (n=170). Pittock, Sean J and Abu-sbeih, Hamzah are very concerned about the relationship between cancer immunotherapy and medical images and have published more academic papers (n = 5; n = 4). Among the top 10 co-cited authors, Topalian Sl (n=43) cited ranked first, followed by Graus F (n=40) cited. According to clustering, timeline, and burst word analysis, the results show that the current research focus is on "MRI", "deep learning", "tumor microenvironment" and so on. Conclusion Medical imaging and cancer immunotherapy are hot topics. The United States is the country with the most publications and the greatest influence in this field, followed by China. "MRI", "PET/PET-CT", "deep learning", "immune-related adverse events" and "tumor microenvironment" are currently hot research topics and potential targets.
Collapse
Affiliation(s)
- Shuli Tang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Tiantian Fan
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Xinxin Wang
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Can Yu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Chunhui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Yang Zhou
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| |
Collapse
|
5
|
Foster JB, Alonso MM, Sayour E, Davidson TB, Persson ML, Dun MD, Kline C, Mueller S, Vitanza NA, van der Lugt J. Translational considerations for immunotherapy clinical trials in pediatric neuro-oncology. Neoplasia 2023; 42:100909. [PMID: 37244226 DOI: 10.1016/j.neo.2023.100909] [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: 10/13/2022] [Revised: 04/20/2023] [Accepted: 05/11/2023] [Indexed: 05/29/2023]
Abstract
While immunotherapy for pediatric cancer has made great strides in recent decades, including the FDA approval of agents such as dinutuximab and tisgenlecleucel, these successes have rarely impacted children with pediatric central nervous system (CNS) tumors. As our understanding of the biological underpinnings of these tumors evolves, new immunotherapeutics are undergoing rapid clinical translation specifically designed for children with CNS tumors. Most recently, there have been notable clinical successes with oncolytic viruses, vaccines, adoptive cellular therapy, and immune checkpoint inhibition. In this article, the immunotherapy working group of the Pacific Pediatric Neuro-Oncology Consortium (PNOC) reviews the current and future state of immunotherapeutic CNS clinical trials with a focus on clinical trial development. Based on recent therapeutic trials, we discuss unique immunotherapy clinical trial challenges, including toxicity considerations, disease assessment, and correlative studies. Combinatorial strategies and future directions will be addressed. Through internationally collaborative efforts and consortia, we aim to direct this promising field of immuno-oncology to the next frontier of successful application against pediatric CNS tumors.
Collapse
Affiliation(s)
- Jessica B Foster
- Division of Oncology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA.
| | - Marta M Alonso
- Department of Pediatrics, Program of Solid Tumors, University Clinic of Navarra, Center for the Applied Medical Research (CIMA), Pamplona, Spain
| | - Elias Sayour
- Lillian S. Wells Department of Neurosurgery, Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL USA
| | - Tom B Davidson
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States
| | - Mika L Persson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Mark Hughes Foundation Centre for Brain Cancer Research, Paediatric Program, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Cassie Kline
- Division of Oncology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Sabine Mueller
- Department of Neurology, Department of Neurosurgery and Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Nicholas A Vitanza
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
6
|
Lin G, Wang X, Ye H, Cao W. Radiomic Models Predict Tumor Microenvironment Using Artificial Intelligence-the Novel Biomarkers in Breast Cancer Immune Microenvironment. Technol Cancer Res Treat 2023; 22:15330338231218227. [PMID: 38111330 PMCID: PMC10734346 DOI: 10.1177/15330338231218227] [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: 09/12/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Breast cancer is the most common malignancy in women, and some subtypes are associated with a poor prognosis with a lack of efficacious therapy. Moreover, immunotherapy and the use of other novel antibody‒drug conjugates have been rapidly incorporated into the standard management of advanced breast cancer. To extract more benefit from these therapies, clarifying and monitoring the tumor microenvironment (TME) status is critical, but this is difficult to accomplish based on conventional approaches. Radiomics is a method wherein radiological image features are comprehensively collected and assessed to build connections with disease diagnosis, prognosis, therapy efficacy, the TME, etc In recent years, studies focused on predicting the TME using radiomics have increasingly emerged, most of which demonstrate meaningful results and show better capability than conventional methods in some aspects. Beyond predicting tumor-infiltrating lymphocytes, immunophenotypes, cytokines, infiltrating inflammatory factors, and other stromal components, radiomic models have the potential to provide a completely new approach to deciphering the TME and facilitating tumor management by physicians.
Collapse
Affiliation(s)
- Guang Lin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hunan Ye
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wenming Cao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Zhu Z, Chen M, Hu G, Pan Z, Han W, Tan W, Zhou Z, Wang M, Mao L, Li X, Sui X, Song L, Xu Y, Song W, Yu Y, Jin Z. A pre-treatment CT-based weighted radiomic approach combined with clinical characteristics to predict durable clinical benefits of immunotherapy in advanced lung cancer. Eur Radiol 2022; 33:3918-3930. [PMID: 36515714 PMCID: PMC9748402 DOI: 10.1007/s00330-022-09337-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/08/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To develop a pre-treatment CT-based predictive model to anticipate inoperable lung cancer patients' progression-free survival (PFS) to immunotherapy. METHODS This single-center retrospective study developed and cross-validated a radiomic model in 185 patients and tested it in 48 patients. The binary endpoint is the durable clinical benefit (DCB, PFS ≥ 6 months) and non-DCB (NDCB, PFS < 6 months). Radiomic features were extracted from multiple intrapulmonary lesions and weighted by an attention-based multiple-instance learning model. Aggregated features were then selected through L2-regularized ridge regression. Five machine-learning classifiers were conducted to build predictive models using radiomic and clinical features alone and then together. Lastly, the predictive value of the model with the best performance was validated by Kaplan-Meier survival analysis. RESULTS The predictive models based on the weighted radiomic approach showed superior performance across all classifiers (AUCs: 0.75-0.82) compared with the largest lesion approach (AUCs: 0.70-0.78) and the average sum approach (AUCs: 0.64-0.80). Among them, the logistic regression model yielded the most balanced performance (AUC = 0.87 [95%CI 0.84-0.89], 0.75 [0.68-0.82], 0.80 [0.68-0.92] in the training, validation, and test cohort respectively). The addition of five clinical characteristics significantly enhanced the performance of radiomic-only model (train: AUC 0.91 [0.89-0.93], p = .042; validation: AUC 0.86 [0.80-0.91], p = .011; test: AUC 0.86 [0.76-0.96], p = .026). Kaplan-Meier analysis of the radiomic-based predictive models showed a clear stratification between classifier-predicted DCB versus NDCB for PFS (HR = 2.40-2.95, p < 0.05). CONCLUSIONS The adoption of weighted radiomic features from multiple intrapulmonary lesions has the potential to predict long-term PFS benefits for patients who are candidates for PD-1/PD-L1 immunotherapies. KEY POINTS • Weighted radiomic-based model derived from multiple intrapulmonary lesions on pre-treatment CT images has the potential to predict durable clinical benefits of immunotherapy in lung cancer. • Early line immunotherapy is associated with longer progression-free survival in advanced lung cancer.
Collapse
Affiliation(s)
- Zhenchen Zhu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Ge Hu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Zhengsong Pan
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Institute of Basic Medicine Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005 China
| | - Weixiong Tan
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Zhen Zhou
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Li Mao
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Xiuli Li
- Deepwise Artificial Intelligence (AI) Lab, Beijing Deepwise & League of PhD technology Co. Ltd, Beijing, 100080 China
| | - Xin Sui
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yizhou Yu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| |
Collapse
|
8
|
Kim N, Lee ES, Won SE, Yang M, Lee AJ, Shin Y, Ko Y, Pyo J, Park HJ, Kim KW. Evolution of Radiological Treatment Response Assessments for Cancer Immunotherapy: From iRECIST to Radiomics and Artificial Intelligence. Korean J Radiol 2022; 23:1089-1101. [PMID: 36098343 PMCID: PMC9614294 DOI: 10.3348/kjr.2022.0225] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has revolutionized and opened a new paradigm for cancer treatment. In the era of immunotherapy and molecular targeted therapy, precision medicine has gained emphasis, and an early response assessment is a key element of this approach. Treatment response assessment for immunotherapy is challenging for radiologists because of the rapid development of immunotherapeutic agents, from immune checkpoint inhibitors to chimeric antigen receptor-T cells, with which many radiologists may not be familiar, and the atypical responses to therapy, such as pseudoprogression and hyperprogression. Therefore, new response assessment methods such as immune response assessment, functional/molecular imaging biomarkers, and artificial intelligence (including radiomics and machine learning approaches) have been developed and investigated. Radiologists should be aware of recent trends in immunotherapy development and new response assessment methods.
Collapse
Affiliation(s)
- Nari Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Eun Sung Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Eun Won
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Mihyun Yang
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Amy Junghyun Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Youngbin Shin
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yousun Ko
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Junhee Pyo
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung Won Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
9
|
Li W, Le NN, Onishi N, Newitt DC, Wilmes LJ, Gibbs JE, Carmona-Bozo J, Liang J, Partridge SC, Price ER, Joe BN, Kornak J, Magbanua MJM, Nanda R, LeStage B, Esserman LJ, I-Spy Imaging Working Group, I-Spy Investigator Network, Van't Veer LJ, Hylton NM. Diffusion-Weighted MRI for Predicting Pathologic Complete Response in Neoadjuvant Immunotherapy. Cancers (Basel) 2022; 14:4436. [PMID: 36139594 PMCID: PMC9497087 DOI: 10.3390/cancers14184436] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022] Open
Abstract
This study tested the hypothesis that a change in the apparent diffusion coefficient (ADC) measured in diffusion-weighted MRI (DWI) is an independent imaging marker, and ADC performs better than functional tumor volume (FTV) for assessing treatment response in patients with locally advanced breast cancer receiving neoadjuvant immunotherapy. A total of 249 patients were randomized to standard neoadjuvant chemotherapy with pembrolizumab (pembro) or without pembrolizumab (control). DCE-MRI and DWI, performed prior to and 3 weeks after the start of treatment, were analyzed. Percent changes of tumor ADC metrics (mean, 5th to 95th percentiles of ADC histogram) and FTV were evaluated for the prediction of pathologic complete response (pCR) using a logistic regression model. The area under the ROC curve (AUC) estimated for the percent change in mean ADC was higher in the pembro cohort (0.73, 95% confidence interval [CI]: 0.52 to 0.93) than in the control cohort (0.63, 95% CI: 0.43 to 0.83). In the control cohort, the percent change of the 95th percentile ADC achieved the highest AUC, 0.69 (95% CI: 0.52 to 0.85). In the pembro cohort, the percent change of the 25th percentile ADC achieved the highest AUC, 0.75 (95% CI: 0.55 to 0.95). AUCs estimated for percent change of FTV were 0.61 (95% CI: 0.39 to 0.83) and 0.66 (95% CI: 0.47 to 0.85) for the pembro and control cohorts, respectively. Tumor ADC may perform better than FTV to predict pCR at an early treatment time-point during neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Wen Li
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Nu N Le
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Natsuko Onishi
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - David C Newitt
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Lisa J Wilmes
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Jessica E Gibbs
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Julia Carmona-Bozo
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Jiachao Liang
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Savannah C Partridge
- Department of Radiology, University of Washington, 1100 Fairview Ave N, Seattle, Washington, DC 98109, USA
| | - Elissa R Price
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Bonnie N Joe
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Mark Jesus M Magbanua
- Department of Laboratory Medicine, University of California, 2340 Sutter Street, San Francisco, CA 94115, USA
| | - Rita Nanda
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Barbara LeStage
- I-SPY 2 Advocacy Group, 499 Illinois Street, San Francisco, CA 94158, USA
| | - Laura J Esserman
- Department of Surgery, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | | | - I-Spy Investigator Network
- Department of Radiology, University of Washington, 1100 Fairview Ave N, Seattle, Washington, DC 98109, USA
| | - Laura J Van't Veer
- Department of Laboratory Medicine, University of California, 2340 Sutter Street, San Francisco, CA 94115, USA
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| |
Collapse
|
10
|
Sheikhbahaei S, Marcus CV, Sadaghiani MS, Rowe SP, Pomper MG, Solnes LB. Imaging of Cancer Immunotherapy: Response Assessment Methods, Atypical Response Patterns, and Immune-Related Adverse Events, From the AJR Special Series on Imaging of Inflammation. AJR Am J Roentgenol 2022; 218:940-952. [PMID: 34612682 DOI: 10.2214/ajr.21.26538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The introduction of immunotherapy with immune-checkpoint inhibitors (ICIs) has revolutionized cancer treatment paradigms. Since FDA approval of the first ICI in 2011, multiple additional ICIs have been approved and granted marketing authorization, and many promising agents are in early clinical adoption. Due to the distinctive biologic mechanisms of ICIs, the patterns of tumor response and progression seen with immunotherapy differ from those observed with cytotoxic chemothera-pies. With increasing clinical adoption of immunotherapy, it is critical for radiologists to recognize different response patterns and common pitfalls to avoid misinterpretation of imaging studies or prompt premature cessation of potentially effective treatment. This review provides an overview of ICIs and their mechanisms of action and discusses anatomic and metabolic immune-related response assessment methods, typical and atypical patterns of immunotherapy response (including pseudoprogression, hyperprogression, dissociated response, and durable response), and common imaging features of immune-related adverse events. Future multicenter trials are needed to validate the proposed immune-related response criteria and identify the functional imaging markers of early treatment response and survival.
Collapse
Affiliation(s)
- Sara Sheikhbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N Caroline St, Johns Hopkins Outpatient Center, JHOC #3009, Baltimore, MD 21287
| | | | - Mohammad S Sadaghiani
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N Caroline St, Johns Hopkins Outpatient Center, JHOC #3009, Baltimore, MD 21287
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N Caroline St, Johns Hopkins Outpatient Center, JHOC #3009, Baltimore, MD 21287
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N Caroline St, Johns Hopkins Outpatient Center, JHOC #3009, Baltimore, MD 21287
| | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N Caroline St, Johns Hopkins Outpatient Center, JHOC #3009, Baltimore, MD 21287
| |
Collapse
|
11
|
Keenan KE, Delfino JG, Jordanova KV, Poorman ME, Chirra P, Chaudhari AS, Baessler B, Winfield J, Viswanath SE, deSouza NM. Challenges in ensuring the generalizability of image quantitation methods for MRI. Med Phys 2022; 49:2820-2835. [PMID: 34455593 PMCID: PMC8882689 DOI: 10.1002/mp.15195] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 01/31/2023] Open
Abstract
Image quantitation methods including quantitative MRI, multiparametric MRI, and radiomics offer great promise for clinical use. However, many of these methods have limited clinical adoption, in part due to issues of generalizability, that is, the ability to translate methods and models across institutions. Researchers can assess generalizability through measurement of repeatability and reproducibility, thus quantifying different aspects of measurement variance. In this article, we review the challenges to ensuring repeatability and reproducibility of image quantitation methods as well as present strategies to minimize their variance to enable wider clinical implementation. We present possible solutions for achieving clinically acceptable performance of image quantitation methods and briefly discuss the impact of minimizing variance and achieving generalizability towards clinical implementation and adoption.
Collapse
Affiliation(s)
- Kathryn E. Keenan
- Physical Measurement Laboratory, National Institute of Standards and Technology, 325 Broadway, Boulder, CO 80305, USA
| | - Jana G. Delfino
- Center for Devices and Radiological Health, US Food and Drug Administration, 10993 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Kalina V. Jordanova
- Physical Measurement Laboratory, National Institute of Standards and Technology, 325 Broadway, Boulder, CO 80305, USA
| | - Megan E. Poorman
- Physical Measurement Laboratory, National Institute of Standards and Technology, 325 Broadway, Boulder, CO 80305, USA
| | - Prathyush Chirra
- Dept of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Akshay S. Chaudhari
- Department of Radiology, Stanford University, 450 Serra Mall, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University, 450 Serra Mall, Stanford, CA 94305, USA
| | - Bettina Baessler
- University Hospital of Zurich and University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Jessica Winfield
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
- MRI Unit, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Satish E. Viswanath
- Dept of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Nandita M. deSouza
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
- MRI Unit, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
| |
Collapse
|
12
|
Xu Y, Su GH, Ma D, Xiao Y, Shao ZM, Jiang YZ. Technological advances in cancer immunity: from immunogenomics to single-cell analysis and artificial intelligence. Signal Transduct Target Ther 2021; 6:312. [PMID: 34417437 PMCID: PMC8377461 DOI: 10.1038/s41392-021-00729-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/06/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapies play critical roles in cancer treatment. However, given that only a few patients respond to immune checkpoint blockades and other immunotherapeutic strategies, more novel technologies are needed to decipher the complicated interplay between tumor cells and the components of the tumor immune microenvironment (TIME). Tumor immunomics refers to the integrated study of the TIME using immunogenomics, immunoproteomics, immune-bioinformatics, and other multi-omics data reflecting the immune states of tumors, which has relied on the rapid development of next-generation sequencing. High-throughput genomic and transcriptomic data may be utilized for calculating the abundance of immune cells and predicting tumor antigens, referring to immunogenomics. However, as bulk sequencing represents the average characteristics of a heterogeneous cell population, it fails to distinguish distinct cell subtypes. Single-cell-based technologies enable better dissection of the TIME through precise immune cell subpopulation and spatial architecture investigations. In addition, radiomics and digital pathology-based deep learning models largely contribute to research on cancer immunity. These artificial intelligence technologies have performed well in predicting response to immunotherapy, with profound significance in cancer therapy. In this review, we briefly summarize conventional and state-of-the-art technologies in the field of immunogenomics, single-cell and artificial intelligence, and present prospects for future research.
Collapse
Affiliation(s)
- Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Response Prediction and Evaluation Using PET in Patients with Solid Tumors Treated with Immunotherapy. Cancers (Basel) 2021; 13:cancers13123083. [PMID: 34205572 PMCID: PMC8234914 DOI: 10.3390/cancers13123083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary In cancer treatment, immunotherapy is increasingly becoming important as a component of first-line treatment and has improved the prognosis of patients since its introduction. A large group of patients, however, do not respond to immunotherapy, and predicting a treatment response remains challenging. Furthermore, evaluating a response using conventional computed tomography (CT) scans is not straightforward due to the different mechanism of action of immunotherapy compared to chemotherapy. This review provides an overview of positron emission tomography (PET) in predicting and evaluating treatment response to immunotherapy. Abstract In multiple malignancies, checkpoint inhibitor therapy has an established role in the first-line treatment setting. However, only a subset of patients benefit from checkpoint inhibition, and as a result, the field of biomarker research is active. Molecular imaging with the use of positron emission tomography (PET) is one of the biomarkers that is being studied. PET tracers such as conventional 18F-FDG but also PD-(L)1 directed tracers are being evaluated for their predictive power. Furthermore, the use of artificial intelligence is under evaluation for the purpose of response prediction. Response evaluation during checkpoint inhibitor therapy can be challenging due to the different response patterns that can be observed compared to traditional chemotherapy. The additional information provided by PET can potentially be of value to evaluate a response early after the start of treatment and provide the clinician with important information about the efficacy of immunotherapy. Furthermore, the use of PET to stratify between patients with a complete response and those with a residual disease can potentially guide clinicians to identify patients for which immunotherapy can be discontinued and patients for whom the treatment needs to be escalated. This review provides an overview of the use of positron emission tomography (PET) to predict and evaluate treatment response to immunotherapy.
Collapse
|
14
|
Challenges and advances for the treatment of renal cancer patients with brain metastases: From immunological background to upcoming clinical evidence on immune-checkpoint inhibitors. Crit Rev Oncol Hematol 2021; 163:103390. [PMID: 34090998 DOI: 10.1016/j.critrevonc.2021.103390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/27/2021] [Accepted: 05/25/2021] [Indexed: 01/12/2023] Open
Abstract
The introduction of checkpoint inhibitors (ICIs) in renal cell carcinoma (RCC) treatment landscape, resulted in improvements in overall survival (OS) in metastatic patients. Brain metastases (BMs) are a specific metastatic site of interest representing a predictive factor of poor prognosis. Patients with BMs were usually excluded from prospective clinical trials in the past. Despite recent evidence suggest the efficacy and safety of ICIs, the BMs treatment remains a challenge; the immunotherapy responsiveness seems to be multifactorial and dependent on several factors, such as the genetic intratumor heterogeneity and the immunosuppressive role of the brain tumor microenvironment. This review, starting from the immunological background in RCC BMs, provide an overview of the upcoming evidence from clinical trials, address the issues related to the neuroradiological immunotherapy response evaluation and, with a look to the future, describes how the epigenetic modulation of immune evasion could represent a background for new therapeutic strategies.
Collapse
|
15
|
Schäfer JF, Herrmann J, Kammer B, Koerber F, Tsiflikas I, von Kalle T, Mentzel HJ. Fortschrittliche radiologische Diagnostik bei soliden Tumoren im Kindes- und Jugendalter. DER ONKOLOGE 2021; 27:410-426. [DOI: 10.1007/s00761-021-00910-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 01/04/2025]
|
16
|
Shen L, Fu H, Tao G, Liu X, Yuan Z, Ye X. Pre-Immunotherapy Contrast-Enhanced CT Texture-Based Classification: A Useful Approach to Non-Small Cell Lung Cancer Immunotherapy Efficacy Prediction. Front Oncol 2021; 11:591106. [PMID: 33968716 PMCID: PMC8103028 DOI: 10.3389/fonc.2021.591106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022] Open
Abstract
Objective: To investigate the utility of the pre-immunotherapy contrast-enhanced CT-based texture classification in predicting response to non-small cell lung cancer (NSCLC) immunotherapy treatment. Methods: Sixty-three patients with 72 lesions who received immunotherapy were enrolled in this study. We extracted textures including histogram, absolute gradient, run-length matrix, gray-level co-occurrence matrix, autoregressive model, and wavelet transform from pre-immunotherapy contrast-enhanced CT by using Mazda software. Three different methods, namely, Fisher coefficient, mutual information measure (MI), and minimization of classification error probability combined average correlation coefficients (POE + ACC), were performed to select 10 optimal texture feature sets, respectively. The patients were divided into non-progressive disease (non-PD) and progressive disease (PD) groups. t-test or Mann–Whitney U-test was performed to test the differences in each texture feature set between the above two groups. Each texture feature set was analyzed by principal component analysis (PCA), linear discriminant analysis (LDA), and non-linear discriminant analysis (NDA). The area under the curve (AUC) was used to quantify the predictive accuracy of the above three analysis models for each texture feature set, and the sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV) were also calculated, respectively. Results: Among the three texture feature sets, the texture parameter differences of kurtosis (2.12 ± 3.92 vs. 0.78 ± 1.10, p = 0.047), “S(2,2)SumEntrp” (1.14 ± 0.31 vs. 1.24 ± 0.12, p = 0.036), and “S(1,0)SumEntrp” (1.18 ± 0.27 vs. 1.28 ± 0.11, p = 0.046) between the non-PD and PD group were statistically significant (all p < 0.05). The classification result of texture feature set selected by POE + ACC and analyzed by NDA was identified as the best model (AUC = 0.812, 95% CI: 0.706–0.919) with a sensitivity, specificity, accuracy, PPV, and NPV of 88.2, 76.3, 81.9, 76.9, and 87.9%, respectively. Conclusion: Pre-immunotherapy contrast-enhanced CT-based texture provides a new method for clinical evaluation of the NSCLC immunotherapy efficacy prediction.
Collapse
Affiliation(s)
- Leilei Shen
- Department of Radiology, Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Hongchao Fu
- Department of Radiology, Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Guangyu Tao
- Department of Radiology, Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Xuemei Liu
- Department of Radiology, Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Zheng Yuan
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Xiaodan Ye
- Department of Radiology, Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|