1
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
2
|
Jiang J, Yan Y, Yang C, Cai H. Immunogenic Cell Death and Metabolic Reprogramming in Cancer: Mechanisms, Synergies, and Innovative Therapeutic Strategies. Biomedicines 2025; 13:950. [PMID: 40299564 PMCID: PMC12024911 DOI: 10.3390/biomedicines13040950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
Immunogenic cell death (ICD) is a promising cancer therapy where dying tumor cells release damage-associated molecular patterns (DAMPs) to activate immune responses. Recent research highlights the critical role of metabolic reprogramming in tumor cells, including the Warburg effect, oxidative stress, and lipid metabolism, in modulating ICD and shaping the immune microenvironment. These metabolic changes enhance immune activation, making tumors more susceptible to immune surveillance. This review explores the molecular mechanisms linking ICD and metabolism, including mitochondrial oxidative stress, endoplasmic reticulum (ER) stress, and ferroptosis. It also discusses innovative therapeutic strategies, such as personalized combination therapies, metabolic inhibitors, and targeted delivery systems, to improve ICD efficacy. The future of cancer immunotherapy lies in integrating metabolic reprogramming and immune activation to overcome tumor immune evasion, with multi-omics approaches and microbiome modulation offering new avenues for enhanced treatment outcomes.
Collapse
Affiliation(s)
| | | | - Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China; (J.J.); (Y.Y.)
| | - Hong Cai
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China; (J.J.); (Y.Y.)
| |
Collapse
|
3
|
Chen L, Yin G, Wang Z, Liu Z, Sui C, Chen K, Song T, Xu W, Qi L, Li X. A predictive radiotranscriptomics model based on DCE-MRI for tumor immune landscape and immunotherapy in cholangiocarcinoma. Biosci Trends 2024; 18:263-276. [PMID: 38853000 DOI: 10.5582/bst.2024.01121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
This study aims to determine the predictive role of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) derived radiomic model in tumor immune profiling and immunotherapy for cholangiocarcinoma. To perform radiomic analysis, immune related subgroup clustering was first performed by single sample gene set enrichment analysis (ssGSEA). Second, a total of 806 radiomic features for each phase of DCE-MRI were extracted by utilizing the Python package Pyradiomics. Then, a predictive radiomic signature model was constructed after a three-step features reduction and selection, and receiver operating characteristic (ROC) curve was employed to evaluate the performance of this model. In the end, an independent testing cohort involving cholangiocarcinoma patients with anti-PD-1 Sintilimab treatment after surgery was used to verify the potential application of the established radiomic model in immunotherapy for cholangiocarcinoma. Two distinct immune related subgroups were classified using ssGSEA based on transcriptome sequencing. For radiomic analysis, a total of 10 predictive radiomic features were finally identified to establish a radiomic signature model for immune landscape classification. Regarding to the predictive performance, the mean AUC of ROC curves was 0.80 in the training/validation cohort. For the independent testing cohort, the individual predictive probability by radiomic model and the corresponding immune score derived from ssGSEA was significantly correlated. In conclusion, radiomic signature model based on DCE-MRI was capable of predicting the immune landscape of chalangiocarcinoma. Consequently, a potentially clinical application of this developed radiomic model to guide immunotherapy for cholangiocarcinoma was suggested.
Collapse
Affiliation(s)
- Lu Chen
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guotao Yin
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zifan Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Kun Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
4
|
Zhang Z, Du Y, Shi X, Wang K, Qu Q, Liang Q, Ma X, He K, Chi C, Tang J, Liu B, Ji J, Wang J, Dong J, Hu Z, Tian J. NIR-II light in clinical oncology: opportunities and challenges. Nat Rev Clin Oncol 2024; 21:449-467. [PMID: 38693335 DOI: 10.1038/s41571-024-00892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Novel strategies utilizing light in the second near-infrared region (NIR-II; 900-1,880 nm wavelengths) offer the potential to visualize and treat solid tumours with enhanced precision. Over the past few decades, numerous techniques leveraging NIR-II light have been developed with the aim of precisely eliminating tumours while maximally preserving organ function. During cancer surgery, NIR-II optical imaging enables the visualization of clinically occult lesions and surrounding vital structures with increased sensitivity and resolution, thereby enhancing surgical quality and improving patient prognosis. Furthermore, the use of NIR-II light promises to improve cancer phototherapy by enabling the selective delivery of increased therapeutic energy to tissues at greater depths. Initial clinical studies of NIR-II-based imaging and phototherapy have indicated impressive potential to decrease cancer recurrence, reduce complications and prolong survival. Despite the encouraging results achieved, clinical translation of innovative NIR-II techniques remains challenging and inefficient; multidisciplinary cooperation is necessary to bridge the gap between preclinical research and clinical practice, and thus accelerate the translation of technical advances into clinical benefits. In this Review, we summarize the available clinical data on NIR-II-based imaging and phototherapy, demonstrating the feasibility and utility of integrating these technologies into the treatment of cancer. We also introduce emerging NIR-II-based approaches with substantial potential to further enhance patient outcomes, while also highlighting the challenges associated with imminent clinical studies of these modalities.
Collapse
Affiliation(s)
- Zeyu Zhang
- Key Laboratory of Big Data-Based Precision Medicine of Ministry of Industry and Information Technology, School of Engineering Medicine, Beihang University, Beijing, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Xiaojing Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Qiaojun Qu
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Qian Liang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Xiaopeng Ma
- School of Control Science and Engineering, Shandong University, Jinan, China
| | - Kunshan He
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Chongwei Chi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Jianqiang Tang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Liu
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiafu Ji
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Jun Wang
- Thoracic Oncology Institute/Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China.
| | - Jiahong Dong
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China.
| | - Jie Tian
- Key Laboratory of Big Data-Based Precision Medicine of Ministry of Industry and Information Technology, School of Engineering Medicine, Beihang University, Beijing, China.
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China.
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China.
| |
Collapse
|
5
|
Shi G, Liu X, Du Y, Tian J. RGD targeted magnetic ferrite nanoparticles enhance antitumor immunotherapeutic efficacy by activating STING signaling pathway. iScience 2024; 27:109062. [PMID: 38660408 PMCID: PMC11039334 DOI: 10.1016/j.isci.2024.109062] [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: 09/19/2023] [Revised: 12/06/2023] [Accepted: 01/25/2024] [Indexed: 04/26/2024] Open
Abstract
Manganese has been used in tumor imaging for their ability to provide T1-weighted MRI signal. Recent research find Mn2+ can induce activation of the stimulator of interferon gene (STING) pathway to create an active and favorable tumor immune microenvironment. However, the direct injection of Mn2+ often results in toxicity. In this study, we developed an RGD targeted magnetic ferrite nanoparticle (RGD-MnFe2O4) to facilitate tumor targeted imaging and improve tumor immunotherapy. Magnetic resonance imaging and fluorescence molecular imaging were performed to monitor its in vivo biodistribution. We found that RGD-MnFe2O4 showed active tumor targeting and longer accumulation at tumor sites. Moreover, RGD-MnFe2O4 can activate STING pathway with low toxicity to enhance the PD-L1 expression. Furthermore, combining RGD-MnFe2O4 and anti-PD-L1 antibody (aPD-L1) can treat several types of immunogenic tumors through promoting the accumulation of tumor-infiltrating cytotoxic T cells. In general, our study provides a promising new strategy for the targeted and multifunctional theranostic nanoparticle for the improvement of tumor immunotherapy.
Collapse
Affiliation(s)
- Guangyuan Shi
- University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoli Liu
- Northwest University, Xi’an 710127, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100080, China
| | - Jie Tian
- Science and Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
6
|
Liu N, Yang X, Gao C, Wang J, Zeng Y, Zhang L, Yin Q, Zhang T, Zhou H, Li K, Du J, Zhou S, Zhao X, Zhu H, Yang Z, Liu Z. Noninvasively Deciphering the Immunosuppressive Tumor Microenvironment Using Galectin-1 PET to Inform Immunotherapy Responses. J Nucl Med 2024; 65:728-734. [PMID: 38514084 DOI: 10.2967/jnumed.123.266888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Immune checkpoint blockade (ICB) has achieved groundbreaking results in clinical cancer therapy; however, only a subset of patients experience durable benefits. The aim of this study was to explore strategies for predicting tumor responses to optimize the intervention approach using ICB therapy. Methods: We used a bilateral mouse model for proteomics analysis to identify new imaging biomarkers for tumor responses to ICB therapy. A PET radiotracer was synthesized by radiolabeling the identified biomarker-targeting antibody with 124I. The radiotracer was then tested for PET prediction of tumor responses to ICB therapy. Results: We identified galectin-1 (Gal-1), a member of the carbohydrate-binding lectin family, as a potential negative biomarker for ICB efficacy. We established that Gal-1 inhibition promotes a sensitive immune phenotype within the tumor microenvironment (TME) for ICB therapy. To assess the pre-ICB treatment status of the TME, a Gal-1-targeted PET radiotracer, 124I-αGal-1, was developed. PET imaging with 124I-αGal-1 showed the pretreatment immunosuppressive status of the TME before the initiation of therapy, thus enabling the prediction of ICB resistance in advance. Moreover, the use of hydrogel scaffolds loaded with a Gal-1 inhibitor, thiodigalactoside, demonstrated that a single dose of thiodigalactoside-hydrogel significantly potentiated ICB and adoptive cell transfer immunotherapies by remodeling the immunosuppressive TME. Conclusion: Our study underscores the potential of Gal-1-targeted PET imaging as a valuable strategy for early-stage monitoring of tumor responses to ICB therapy. Additionally, Gal-1 inhibition effectively counteracts the immunosuppressive TME, resulting in enhanced immunotherapy efficacy.
Collapse
Affiliation(s)
- Ning Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiujie Yang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chao Gao
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jianze Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yuwen Zeng
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Linyu Zhang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Qi Yin
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ting Zhang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Haoyi Zhou
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kui Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jinhong Du
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Shixin Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China;
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
- Department of Nuclear Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; and
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing, China
| |
Collapse
|
7
|
Zhou M, Xiang S, Zhao Y, Tang Y, Yang J, Yin X, Tian J, Hu S, Du Y. [ 68Ga]Ga-AUNP-12 PET imaging to assess the PD-L1 status in preclinical and first-in-human study. Eur J Nucl Med Mol Imaging 2024; 51:369-379. [PMID: 37759096 DOI: 10.1007/s00259-023-06447-2] [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: 07/03/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE PD-L1 PET imaging, as a non-invasive procedure, can perform a real-time, dynamic and quantitative analysis of PD-L1 expression at tumor sites. In this study, we developed a novel peptide-based PET tracer, [68 Ga]Ga-AUNP-12, for preclinical and first-of-its-kind imaging of PD-L1 expression in patients. METHODS Radiosynthesis of [68 Ga]Ga-AUNP-12 was conducted. Assays for cellular uptake and binding were conducted on the PANC02, CT26, and B16F10 cell lines. Preclinical models were used to investigate its biodistribution, imaging capacity, and pharmacokinetics. Furthermore, interferon-γ (IFN-γ) was used for development of an animal model with high PD-L1 expression for targeted PET imaging and efficacy evaluation of PD-L1 blocking therapy. In healthy volunteers and cancer patients, the PD-L1 imaging, radiation dosimetry, safety, and biodistribution were further evaluated. RESULTS In vitro and in vivo animal studies showed that [68 Ga]Ga-AUNP-12 PET imaging displayed a high specificity in evaluating PD-L1 expression. The radiochemical yield of [68 Ga]Ga-AUNP-12 was 71.7 ± 8.2%. Additionally, its molar activity and radiochemical purity were satisfactory. The B16F10 tumor was visualized with the tumor uptake of 6.86 ± 0.71% ID/g and tumor-to-muscle ratio of 6.83 ± 0.36 at 60 min after [68 Ga]Ga-AUNP-12 injection. Furthermore, [68 Ga]Ga-AUNP-12 PET imaging could sensitively detect the PD-L1 dynamic changes in CT26 tumor xenograft models regulated by IFN-γ treatment, and correspondingly can effectively guide immunotherapy. Regarding radiation dosimetry, [68 Ga]Ga-AUNP-12 is safe for human use. The first human study found that [68 Ga]Ga-AUNP-12 can be rapidly cleared from blood and other nonspecific organs through the kidney excretion, leading to form a clear imaging contrast in the clinical framework. The specificity of [68 Ga]Ga-AUNP-12 was validated and tumor uptake strongly correlated with the high PD-L1 expression in patients with lung adenocarcinoma and oesophageal squamous cell carcinoma (OSCC). CONCLUSION [68 Ga]Ga-AUNP-12 was successfully developed as a PD-L1-specific PET imaging tracer in preclinical and first-in-human studies.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shijun Xiang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yajie Zhao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jinhui Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, No. 95 Zhongguancun East Road, Beijing, 100190, China.
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, China.
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, People's Republic of China.
| |
Collapse
|
8
|
An Y, Wang H, Li J, Li G, Ma X, Du Y, Tian J. Reconstruction based on adaptive group least angle regression for fluorescence molecular tomography. BIOMEDICAL OPTICS EXPRESS 2023; 14:2225-2239. [PMID: 37206151 PMCID: PMC10191665 DOI: 10.1364/boe.486451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 05/21/2023]
Abstract
Fluorescence molecular tomography can combine two-dimensional fluorescence imaging with anatomical information to reconstruct three-dimensional images of tumors. Reconstruction based on traditional regularization with tumor sparsity priors does not take into account that tumor cells form clusters, so it performs poorly when multiple light sources are used. Here we describe reconstruction based on an "adaptive group least angle regression elastic net" (AGLEN) method, in which local spatial structure correlation and group sparsity are integrated with elastic net regularization, followed by least angle regression. The AGLEN method works iteratively using the residual vector and a median smoothing strategy in order to adaptively obtain a robust local optimum. The method was verified using numerical simulations as well as imaging of mice bearing liver or melanoma tumors. AGLEN reconstruction performed better than state-of-the-art methods with different sizes of light sources at different distances from the sample and in the presence of Gaussian noise at 5-25%. In addition, AGLEN-based reconstruction accurately imaged tumor expression of cell death ligand-1, which can guide immunotherapy.
Collapse
Affiliation(s)
- Yu An
- the Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Hanfan Wang
- the CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiaqian Li
- the Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Guanghui Li
- the Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Xiaopeng Ma
- School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Yang Du
- the CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jie Tian
- the Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, School of Engineering Medicine, Beihang University, Beijing, 100191, China
- the CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
9
|
CEACAM1 Is a Prognostic Biomarker and Correlated with Immune Cell Infiltration in Clear Cell Renal Cell Carcinoma. DISEASE MARKERS 2023; 2023:3606362. [PMID: 36712923 PMCID: PMC9876685 DOI: 10.1155/2023/3606362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/24/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
Background CEACAM1 has been shown to be aberrantly expressed in a variety of tumors, and modulation of CEACAM1-related signaling pathways has been suggested as a novel approach for cancer immunotherapy in recent years. However, its role in clear cell renal cell carcinoma (ccRCC) is unclear. Methods The relationship between CEACAM1 and ccRCC was demonstrated based on data from TCGA, GEO, and HPA databases. And the relationship between clinicopathological features and CEACAM1 expression was also assessed. Survival curve analysis was performed to analyze the prognostic relationship between CEACAM1 expression and ccRCC. Protein interaction network analysis was used to analyze the relationship between CEACAM1 and microenvironment-related proteins. In addition, the immunomodulatory role of CEACAM1 in ccRCC was assessed by analyzing CEACAM1 and immune cell infiltration. Results The expression of CEACAM1 was lower in ccRCC tissues than in adjacent normal tissues, and its expression level was negatively correlated with tumor size status (P < 0.001), metastasis status (P = 0.009), pathological stage (P = 0.002), gender (P < 0.001), histological grade (P < 0.001), and primary therapy outcome (P = 0.045) of ccRCC. Survival curve analysis showed that ccRCC patients with lower CEACAM1 expression exhibited shorter overall survival (P < 0.001), and CEACAM1 interacted with microenvironmental molecules such as fibronectin and integrins. Furthermore, immune infiltration analysis showed that CEACAM1 expression correlated with CD8+ and CD4+ T cells, macrophage, neutrophil, and dendritic cell infiltration in ccRCC. Conclusions CEACAM1 expression correlates with progression, prognosis, and immune cell infiltration in ccRCC patients, and it may be a promising prognostic biomarker and therapeutic target for ccRCC.
Collapse
|
10
|
Li C, Zhu P, Xiang H, Jin Y, Lu B, Shen Y, Wang W, Huang B, Chen Y. 3D-CEUS tracking of injectable chemo-sonodynamic therapy-enabled mop-up of residual renal cell carcinoma after thermal ablation. Mater Today Bio 2022; 18:100513. [PMID: 36569591 PMCID: PMC9771734 DOI: 10.1016/j.mtbio.2022.100513] [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: 10/13/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Thermal ablation (TA), as a minimally invasive therapeutic technique, has been extensively used to the treatment of solid tumors, such as renal cell carcinoma (RCC), which, unfortunately, still fails to overcome the high risk of local recurrence and distant metastasis since the incomplete ablation cannot be ignored due to various factors such as the indistinguishable tumor margins and limited ablation zone. Herein, we report the injectable thermosensitive hydrogel by confining curcumin (Cur)-loaded hollow mesoporous organosilica nanoparticles (Cur@HMON@gel) which can locate in tumor site more than half a month and mop up the residual RCC under ultrasound (US) irradiation after transforming from colloidal sol status to elastic gel matrix at physiological temperature. Based on the US-triggered accelerated diffusion of the model chemotherapy drug with multi-pharmacologic functions, the sustained and controlled release of Cur has been demonstrated in vitro. Significantly, US is employed as an external energy to trigger Cur, as a sonosensitizer also, to generate reactive oxygen species (ROS) for sonodynamic tumor therapy (SDT) in parallel. Tracking by the three-dimensional contrast-enhanced ultrasound (3D-CEUS) imaging, the typical decreased blood perfusions have been observed since the residual xenograft tumor after incomplete TA were effectively suppressed during the chemo-sonodynamic therapy process. The high in vivo biocompatibility and biodegradability of the multifunctional nanoplatform confined by thermogel provide the potential of their further clinical translation for the solid tumor eradication under the guidance and monitoring of 3D-CEUS.
Collapse
Affiliation(s)
- Cuixian Li
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Piao Zhu
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, PR China,Corresponding author.
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Yunjie Jin
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Beilei Lu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Yujia Shen
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China,Corresponding author. Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| | - Beijian Huang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China,Corresponding author. Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China,Corresponding author.
| |
Collapse
|
11
|
Ma X, Zhang MJ, Wang J, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Emerging Biomaterials Imaging Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204034. [PMID: 35728795 DOI: 10.1002/adma.202204034] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/19/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy is one of the most promising clinical modalities for the treatment of malignant tumors and has shown excellent therapeutic outcomes in clinical settings. However, it continues to face several challenges, including long treatment cycles, high costs, immune-related adverse events, and low response rates. Thus, it is critical to predict the response rate to immunotherapy by using imaging technology in the preoperative and intraoperative. Here, the latest advances in nanosystem-based biomaterials used for predicting responses to immunotherapy via the imaging of immune cells and signaling molecules in the immune microenvironment are comprehensively summarized. Several imaging methods, such as fluorescence imaging, magnetic resonance imaging, positron emission tomography imaging, ultrasound imaging, and photoacoustic imaging, used in immune predictive imaging, are discussed to show the potential of nanosystems for distinguishing immunotherapy responders from nonresponders. Nanosystem-based biomaterials aided by various imaging technologies are expected to enable the effective prediction and diagnosis in cases of tumors, inflammation, and other public diseases.
Collapse
Affiliation(s)
- Xianbin Ma
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jingting Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Tian Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
12
|
Glucose–Thymidine Ratio as a Metabolism Index Using 18F-FDG and 18F-FLT PET Uptake as a Potential Imaging Biomarker for Evaluating Immune Checkpoint Inhibitor Therapy. Int J Mol Sci 2022; 23:ijms23169273. [PMID: 36012530 PMCID: PMC9409370 DOI: 10.3390/ijms23169273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in cancer immunotherapy, requiring effective methods for response monitoring. This study evaluated changes in 18F-2-fluoro-2-deoxy-D-glucose (FDG) and 18F-fluorothymidine (FLT) uptake by tumors following ICI treatment as potential imaging biomarkers in mice. Tumor uptakes of 18F-FDG and 18F-FLT were measured and compared between the ICI treatment and control groups. A combined imaging index of glucose–thymidine uptake ratio (GTR) was defined and compared between groups. In the ICI treatment group, tumor growth was effectively inhibited, and higher proportions of immune cells were observed. In the early phase, 18F-FDG uptake was higher in the treatment group, whereas 18F-FLT uptake was not different. There was no difference in 18F-FDG uptake between the two groups in the late phase. However, 18F-FLT uptake of the control group was markedly increased compared with the ICI treatment group. GTR was consistently higher in the ICI treatment group in the early and late phases. After ICI treatment, changes in tumor cell proliferation were observed with 18F-FLT, whereas 18F-FDG showed altered metabolism in both tumor and immune cells. A combination of 18F-FLT and 18F-FDG PET, such as GTR, is expected to serve as a potentially effective imaging biomarker for monitoring ICI treatment.
Collapse
|
13
|
Lin JX, Lin JP, Weng Y, Lv CB, Chen JH, Zhan CY, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Zhou WX, Zhang XJ, Zheng CH, Cai LS, Ma YB, Huang CM. Radiographical Evaluation of Tumor Immunosuppressive Microenvironment and Treatment Outcomes in Gastric Cancer: A Retrospective, Multicohort Study. Ann Surg Oncol 2022; 29:5022-5033. [PMID: 35532827 DOI: 10.1245/s10434-022-11499-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
|
14
|
An Y, Bian C, Yan D, Wang H, Wang Y, Du Y, Tian J. A Fast and Automated FMT/XCT Reconstruction Strategy Based on Standardized Imaging Space. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:657-666. [PMID: 34648436 DOI: 10.1109/tmi.2021.3120011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The traditional finite element method-based fluorescence molecular tomography (FMT)/ X-ray computed tomography (XCT) imaging reconstruction suffers from complicated mesh generation and dual-modality image data fusion, which limits the application of in vivo imaging. To solve this problem, a novel standardized imaging space reconstruction (SISR) method for the quantitative determination of fluorescent probe distributions inside small animals was developed. In conjunction with a standardized dual-modality image data fusion technology, and novel reconstruction strategy based on Laplace regularization and L1-fused Lasso method, the in vivo distribution can be calculated rapidly and accurately, which enables standardized and algorithm-driven data process. We demonstrated the method's feasibility through numerical simulations and quantitatively monitored in vivo programmed death ligand 1 (PD-L1) expression in mouse tumor xenografts, and the results demonstrate that our proposed SISR can increase data throughput and reproducibility, which helps to realize the dynamically and accurately in vivo imaging.
Collapse
|
15
|
Zhu Y, Yao W, Xu BC, Lei YY, Guo QK, Liu LZ, Li HJ, Xu M, Yan J, Chang DD, Feng ST, Zhu ZH. Predicting response to immunotherapy plus chemotherapy in patients with esophageal squamous cell carcinoma using non-invasive Radiomic biomarkers. BMC Cancer 2021; 21:1167. [PMID: 34717582 PMCID: PMC8557514 DOI: 10.1186/s12885-021-08899-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/11/2021] [Indexed: 12/22/2022] Open
Abstract
Objectives To develop and validate a radiomics model for evaluating treatment response to immune-checkpoint inhibitor plus chemotherapy (ICI + CT) in patients with advanced esophageal squamous cell carcinoma (ESCC). Methods A total of 64 patients with advance ESCC receiving first-line ICI + CT at two centers between January 2019 and June 2020 were enrolled in this study. Both 2D ROIs and 3D ROIs were segmented. ComBat correction was applied to minimize the potential bias on the results due to different scan protocols. A total of 788 features were extracted and radiomics models were built on corrected/uncorrected 2D and 3D features by using 5-fold cross-validation. The performance of the radiomics models was assessed by its discrimination, calibration and clinical usefulness with independent validation. Results Five features and support vector machine algorithm were selected to build the 2D uncorrected, 2D corrected, 3D uncorrected and 3D corrected radiomics models. The 2D radiomics models significantly outperformed the 3D radiomics models in both primary and validation cohorts. When ComBat correction was used, the performance of 2D models was better (p = 0.0059) in the training cohort, and significantly better (p < 0.0001) in the validation cohort. The 2D corrected radiomics model yielded the optimal performance and was used to build the nomogram. The calibration curve of the radiomics model demonstrated good agreement between prediction and observation and the decision curve analysis confirmed the clinical utility. Conclusions The easy-to-use 2D corrected radiomics model could facilitate noninvasive preselection of ESCC patients who would benefit from ICI + CT. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08899-x.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510080, Province Guangdong, People's Republic of China.,Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Wang Yao
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Bing-Chen Xu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Yi-Yan Lei
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Qi-Kun Guo
- Department of Radiological Interventional, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Li-Zhi Liu
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Hao-Jiang Li
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Min Xu
- Scientific Collaboration, CT-MR Division, Canon Medical System (China), Jiuxianqiao North Road, Chaoyang District, 100015, Beijing, People's Republic of China
| | - Jing Yan
- Scientific Collaboration, CT-MR Division, Canon Medical System (China), Jiuxianqiao North Road, Chaoyang District, 100015, Beijing, People's Republic of China
| | - Dan-Dan Chang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Province Guangdong, People's Republic of China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Province Guangdong, People's Republic of China.
| | - Zhi-Hua Zhu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510080, Province Guangdong, People's Republic of China.
| |
Collapse
|
16
|
Xu L, Wang Y, Ma Y, Huan S, Song G. Monitoring Immunotherapy With Optical Molecular Imaging. ChemMedChem 2021; 16:2547-2557. [PMID: 33949786 DOI: 10.1002/cmdc.202100260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Indexed: 01/17/2023]
Abstract
Immunotherapy is an effective way to mobilize the body's own immune system to confront tumor cells. However, the efficacy of immunotherapy is affected by tumor heterogeneity, and the low therapeutic response to immunotherapy may lead to negative outcomes, which reinforces the urgency for early benefit predictors. Evaluating the infiltration of immune cells in solid tumors and metabolism changes of tumors provide potential response targets for monitoring immune response. Non-invasive imaging identifying prognostic biomarkers can select the beneficiaries of targeted immunotherapy from non-responses. Quantitative biomarkers may eventually improve the cancer management, help customize individual treatment plans and predict the treatment outcomes. In this review, we summarize the non-invasive optical molecular imaging methods for monitoring immunotherapy. With the combination of imaging and immunotherapy, the prediction of immunotherapy response may promote the development of precision medicine.
Collapse
Affiliation(s)
- Li Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Youjuan Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Yuan Ma
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Shuangyan Huan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| |
Collapse
|
17
|
Bian C, Wang Y, Lu Z, An Y, Wang H, Kong L, Du Y, Tian J. ImmunoAIzer: A Deep Learning-Based Computational Framework to Characterize Cell Distribution and Gene Mutation in Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13071659. [PMID: 33916145 PMCID: PMC8036970 DOI: 10.3390/cancers13071659] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Spatial distribution of tumor infiltrating lymphocytes (TILs) and cancer cells in the tumor microenvironment (TME) along with tumor gene mutation status are of vital importance to the guidance of cancer immunotherapy and prognoses. In this work, we developed a deep learning-based computational framework, termed ImmunoAIzer, which involves: (1) the implementation of a semi-supervised strategy to train a cellular biomarker distribution prediction network (CBDPN) to make predictions of spatial distributions of CD3, CD20, PanCK, and DAPI biomarkers in the tumor microenvironment with an accuracy of 90.4%; (2) using CBDPN to select tumor areas on hematoxylin and eosin (H&E) staining tissue slides and training a multilabel tumor gene mutation detection network (TGMDN), which can detect APC, KRAS, and TP53 mutations with area-under-the-curve (AUC) values of 0.76, 0.77, and 0.79. These findings suggest that ImmunoAIzer could provide comprehensive information of cell distribution and tumor gene mutation status of colon cancer patients efficiently and less costly; hence, it could serve as an effective auxiliary tool for the guidance of immunotherapy and prognoses. The method is also generalizable and has the potential to be extended for application to other types of cancers other than colon cancer.
Collapse
Affiliation(s)
- Chang Bian
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Wang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China;
| | - Yu An
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine Science and Engineering, Beihang University, Beijing 100191, China
| | - Hanfan Wang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- School of Life Science and Technology, Xidian University, Xi’an 710071, China
| | - Lingxin Kong
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (Y.D.); (J.T.)
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; (C.B.); (Y.W.); (Y.A.); (H.W.); (L.K.)
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine Science and Engineering, Beihang University, Beijing 100191, China
- School of Life Science and Technology, Xidian University, Xi’an 710071, China
- Correspondence: (Y.D.); (J.T.)
| |
Collapse
|
18
|
Hricak H, Abdel-Wahab M, Atun R, Lette MM, Paez D, Brink JA, Donoso-Bach L, Frija G, Hierath M, Holmberg O, Khong PL, Lewis JS, McGinty G, Oyen WJG, Shulman LN, Ward ZJ, Scott AM. Medical imaging and nuclear medicine: a Lancet Oncology Commission. Lancet Oncol 2021; 22:e136-e172. [PMID: 33676609 PMCID: PMC8444235 DOI: 10.1016/s1470-2045(20)30751-8] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
The diagnosis and treatment of patients with cancer requires access to imaging to ensure accurate management decisions and optimal outcomes. Our global assessment of imaging and nuclear medicine resources identified substantial shortages in equipment and workforce, particularly in low-income and middle-income countries (LMICs). A microsimulation model of 11 cancers showed that the scale-up of imaging would avert 3·2% (2·46 million) of all 76·0 million deaths caused by the modelled cancers worldwide between 2020 and 2030, saving 54·92 million life-years. A comprehensive scale-up of imaging, treatment, and care quality would avert 9·55 million (12·5%) of all cancer deaths caused by the modelled cancers worldwide, saving 232·30 million life-years. Scale-up of imaging would cost US$6·84 billion in 2020-30 but yield lifetime productivity gains of $1·23 trillion worldwide, a net return of $179·19 per $1 invested. Combining the scale-up of imaging, treatment, and quality of care would provide a net benefit of $2·66 trillion and a net return of $12·43 per $1 invested. With the use of a conservative approach regarding human capital, the scale-up of imaging alone would provide a net benefit of $209·46 billion and net return of $31·61 per $1 invested. With comprehensive scale-up, the worldwide net benefit using the human capital approach is $340·42 billion and the return per dollar invested is $2·46. These improved health and economic outcomes hold true across all geographical regions. We propose actions and investments that would enhance access to imaging equipment, workforce capacity, digital technology, radiopharmaceuticals, and research and training programmes in LMICs, to produce massive health and economic benefits and reduce the burden of cancer globally.
Collapse
Affiliation(s)
- Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| | - May Abdel-Wahab
- International Atomic Energy Agency, Division of Human Health, Vienna, Austria; Radiation Oncology, National Cancer Institute, Cairo University, Cairo, Egypt; Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rifat Atun
- Department of Global Health and Population, Harvard TH Chan School of Public Health, Boston, MA, USA; Department of Global Health and Social Medicine, Harvard Medical School, Harvard University, Boston, MA, USA
| | | | - Diana Paez
- International Atomic Energy Agency, Division of Human Health, Vienna, Austria
| | - James A Brink
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Lluís Donoso-Bach
- Department of Medical Imaging, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Spain
| | | | | | - Ola Holmberg
- Radiation Protection of Patients Unit, International Atomic Energy Agency, Vienna, Austria
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jason S Lewis
- Department of Radiology and Molecular Pharmacology Programme, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Departments of Pharmacology and Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Geraldine McGinty
- Departments of Radiology and Population Science, Weill Cornell Medical College, New York, NY, USA; American College of Radiology, Reston, VA, USA
| | - Wim J G Oyen
- Department of Biomedical Sciences and Humanitas Clinical and Research Centre, Department of Nuclear Medicine, Humanitas University, Milan, Italy; Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, Netherlands; Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Lawrence N Shulman
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary J Ward
- Center for Health Decision Science, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Yan M, Wang W. A radiomics model of predicting tumor volume change of patients with stage III non-small cell lung cancer after radiotherapy. Sci Prog 2021; 104:36850421997295. [PMID: 33687294 PMCID: PMC10453712 DOI: 10.1177/0036850421997295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To predict the volume change of stage III NSCLC after radiotherapy with 60 Gy.This retrospective study included two independent cohorts, a train cohort of 192 patients, and a test cohort of 31 patients. We developed a radiomics model based on radiomics features and clinical variables. LIFEx package was used to extract radiomics texture features from CT images. The classification method was logistic regression analysis and feature selection was performed by correlation coefficients. Performance metrics of logistic regression include accuracy, precision, the receiver operating characteristic curves, and recall.The combination features of clinical variables and radiomics can predict the tumor volume change after radiotherapy with 88.7% accuracy (88.6% precision, 88.7% recall, and 88.7% ROC area).Radiomics features combined with medical knowledge have a great potential to predict accurately tumor volume change of stage III NSCLC after radiotherapy with 60 Gy.
Collapse
Affiliation(s)
- Mengmeng Yan
- Urban Vocational College of Sichuan, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Chengdu, China
- Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
20
|
García-Figueiras R, Baleato-González S, Luna A, Muñoz-Iglesias J, Oleaga L, Vallejo Casas JA, Martín-Noguerol T, Broncano J, Areses MC, Vilanova JC. Assessing Immunotherapy with Functional and Molecular Imaging and Radiomics. Radiographics 2020; 40:1987-2010. [PMID: 33035135 DOI: 10.1148/rg.2020200070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunotherapy is changing the treatment paradigm for cancer and has introduced new challenges in medical imaging. Because not all patients benefit from immunotherapy, pretreatment imaging should be performed to identify not only prognostic factors but also factors that allow prediction of response to immunotherapy. Follow-up studies must allow detection of nonresponders, without confusion of pseudoprogression with real progression to prevent premature discontinuation of treatment that can benefit the patient. Conventional imaging techniques and classic tumor response criteria are limited for the evaluation of the unusual patterns of response that arise from the specific mechanisms of action of immunotherapy, so advanced imaging methods must be developed to overcome these shortcomings. The authors present the fundamentals of the tumor immune microenvironment and immunotherapy and how they influence imaging findings. They also discuss advances in functional and molecular imaging techniques for the assessment of immunotherapy in clinical practice, including their use to characterize immune phenotypes, assess patient prognosis and response to therapy, and evaluate immune-related adverse events. Finally, the development of radiomics and radiogenomics in these therapies and the future role of imaging biomarkers for immunotherapy are discussed. Online supplemental material is available for this article. ©RSNA, 2020.
Collapse
Affiliation(s)
- Roberto García-Figueiras
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Sandra Baleato-González
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Antonio Luna
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - José Muñoz-Iglesias
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Laura Oleaga
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Juan Antonio Vallejo Casas
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Teodoro Martín-Noguerol
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Jordi Broncano
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - María Carmen Areses
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| | - Joan C Vilanova
- From the Department of Radiology, Oncologic Imaging, Hospital Clínico Universitario de Santiago de Compostela, Choupana s/n, 15706, Santiago de Compostela, Spain (R.G.F., S.B.G.); Department of Radiology, HT Medica, Jaén, Spain (A.L, J.B.); Department of Nuclear Medicine, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (J.M.I.); Department of Radiology, Hospital Clínic Barcelona, Barcelona, Spain (L.O.); Unidad de Gestión Clínica de Medicina Nuclear, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain (J.A.V.C.); MRI Unit, HT Medica, Jaén, Spain (T.M.N.); Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain (M.C.A.); and Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging, Girona, Spain (J.C.V.)
| |
Collapse
|
21
|
Porcu M, Solinas C, Mannelli L, Micheletti G, Lambertini M, Willard-Gallo K, Neri E, Flanders AE, Saba L. Radiomics and "radi-…omics" in cancer immunotherapy: a guide for clinicians. Crit Rev Oncol Hematol 2020; 154:103068. [PMID: 32805498 DOI: 10.1016/j.critrevonc.2020.103068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/13/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years the concept of precision medicine has become a popular topic particularly in medical oncology. Besides the identification of new molecular prognostic and predictive biomarkers and the development of new targeted and immunotherapeutic drugs, imaging has started to play a central role in this new era. Terms such as "radiomics", "radiogenomics" or "radi…-omics" are becoming increasingly common in the literature and soon they will represent an integral part of clinical practice. The use of artificial intelligence, imaging and "-omics" data can be used to develop models able to predict, for example, the features of the tumor immune microenvironment through imaging, and to monitor the therapeutic response beyond the standard radiological criteria. The aims of this narrative review are to provide a simplified guide for clinicians to these concepts, and to summarize the existing evidence on radiomics and "radi…-omics" in cancer immunotherapy.
Collapse
Affiliation(s)
- Michele Porcu
- Department of Radiology, AOU of Cagliari, University of Cagliari, Italy.
| | - Cinzia Solinas
- Medical Oncology, Azienda Tutela Salute Sardegna, Hospital Antonio Segni, Ozieri, SS, Italy
| | | | - Giulio Micheletti
- Department of Radiology, AOU of Cagliari, University of Cagliari, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | | | | | - Adam E Flanders
- Department of Radiology, Division of Neuroradiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Luca Saba
- Department of Radiology, AOU of Cagliari, University of Cagliari, Italy
| |
Collapse
|
22
|
Wang H. Precision oncology-featuring the special issue guest editor "Cancer Letters". Cancer Lett 2020; 482:72-73. [PMID: 32200040 DOI: 10.1016/j.canlet.2020.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Hongyang Wang
- National Center for Liver Cancer, Shanghai, China; International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai, China; Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, China; Shanghai Key Laboratory of Hepato-biliary Tumor Biology, China.
| |
Collapse
|