1
|
Wu F, Cao G, Lu J, Ye S, Tang X. Correlation between 18 F-FDG PET/CT metabolic parameters and microvascular invasion before liver transplantation in patients with hepatocellular carcinoma. Nucl Med Commun 2024; 45:1033-1038. [PMID: 39267532 PMCID: PMC11537472 DOI: 10.1097/mnm.0000000000001897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Microvascular infiltration (MVI) before liver transplantation (LT) in patients with hepatocellular carcinoma (HCC) is associated with postoperative tumor recurrence and survival. MVI is mainly assessed by pathological analysis of tissue samples, which is invasive and heterogeneous. PET/computed tomography (PET/CT) with 18 F-labeled fluorodeoxyglucose ( 18 F-FDG) as a tracer has been widely used in the examination of malignant tumors. This study investigated the association between 18 F-FDG PET/CT metabolic parameters and MVI before LT in HCC patients. METHODS About 124 HCC patients who had 18 F-FDG PET/CT examination before LT were included. The patients' clinicopathological features and 18 F-FDG PET/CT metabolic parameters were recorded. Correlations between clinicopathological features, 18 F-FDG PET/CT metabolic parameters, and MVI were analyzed. ROC curve was used to determine the optimal diagnostic cutoff value, area under the curve (AUC), sensitivity, and specificity for predictors of MVI. RESULT In total 72 (58.06%) patients were detected with MVI among the 124 HCC patients. Univariate analysis showed that tumor size ( P = 0.001), T stage ( P < 0.001), maximum standardized uptake value (SUV max ) ( P < 0.001), minimum standardized uptake value (SUV min ) ( P = 0.031), mean standardized uptake value (SUV mean ) ( P = 0.001), peak standardized uptake value (SUV peak ) ( P = 0.001), tumor-to-liver ratio (SUV ratio ) ( P = 0.010), total lesion glycolysis (TLG) ( P = 0.006), metabolic tumor volume (MTV) ( P = 0.011) and MVI were significantly different. Multivariate logistic regression showed that tumor size ( P = 0.018), T stage ( P = 0.017), TLG ( P = 0.023), and MTV ( P = 0.015) were independent predictors of MVI. In the receiver operating characteristic curve, TLG predicted MVI with an AUC value of 0.645. MTV predicted MVI with an AUC value of 0.635. Patients with tumor size ≥5 cm, T3-4, TLG > 400.67, and MTV > 80.58 had a higher incidence of MVI. CONCLUSION 18 F-FDG PET/CT metabolic parameters correlate with MVI and may be used as a noninvasive technique to predict MVI before LT in HCC patients.
Collapse
Affiliation(s)
- Fan Wu
- Department of Nuclear Medicine and Radiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University
| | - Guohong Cao
- Department of Nuclear Medicine and Radiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University
| | - Jinlan Lu
- Department of Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital
| | - Shengli Ye
- Department of Nuclear Medicine and Radiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University
| | - Xin Tang
- Department of Radiology, Hangzhou Wuyunshan Hospital, Hangzhou Health Promotion Research Institute, Hangzhou, China
| |
Collapse
|
2
|
Jin N, An Y, Tian Y, Zhang Z, He K, Chi C, Mu W, Tian J, Du Y. Multispectral fluorescence imaging of EGFR and PD-L1 for precision detection of oral squamous cell carcinoma: a preclinical and clinical study. BMC Med 2024; 22:342. [PMID: 39183296 PMCID: PMC11346054 DOI: 10.1186/s12916-024-03559-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Early detection and treatment are effective methods for the management of oral squamous cell carcinoma (OSCC), which can be facilitated by the detection of tumor-specific OSCC biomarkers. The epidermal growth factor receptor (EGFR) and programmed death-ligand 1 (PD-L1) are important therapeutic targets for OSCC. Multispectral fluorescence molecular imaging (FMI) can facilitate the detection of tumor multitarget expression with high sensitivity and safety. Hence, we developed Nimotuzumab-ICG and Atezolizumab-Cy5.5 imaging probes, in combination with multispectral FMI, to sensitively and noninvasively identify EGFR and PD-L1 expression for the detection and comprehensive treatment of OSCC. METHODS The expression of EGFR and PD-L1 was analyzed using bioinformatics data sources and specimens. Nimotuzumab-ICG and Atezolizumab-Cy5.5 imaging probes were developed and tested on preclinical OSCC cell line and orthotopic OSCC mouse model, fresh OSCC patients' biopsied samples, and further clinical mouthwash trials were conducted in OSCC patients. RESULTS EGFR and PD-L1 were specifically expressed in human OSCC cell lines and tumor xenografts. Nimotuzumab-ICG and Atezolizumab-Cy5.5 imaging probes can specifically target to the tumor sites in an in situ human OSCC mouse model with good safety. The detection sensitivity and specificity of Nimotuzumab-ICG in patients were 96.4% and 100%, and 95.2% and 88.9% for Atezolizumab-Cy5.5. CONCLUSIONS EGFR and PD-L1 are highly expressed in OSCC, the combination of which is important for a precise prognosis of OSCC. EGFR and PD-L1 expression can be sensitively detected using the newly synthesized multispectral fluorescence imaging probes Nimotuzumab-ICG and Atezolizumab-Cy5.5, which can facilitate the sensitive and specific detection of OSCC and improve treatment outcomes. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR2100045738. Registered 23 April 2021, https://www.chictr.org.cn/bin/project/edit?pid=125220.
Collapse
Affiliation(s)
- Nenghao Jin
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yu An
- 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
| | - Yu Tian
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Stomatology, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, 100039, China
| | - Zeyu Zhang
- 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
| | - Kunshan He
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- State Key Laboratory of Computer Science and Beijing Key Lab of Human-Computer Interaction, Institute of Software, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chongwei Chi
- 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
| | - Wei Mu
- 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.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- 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.
| | - 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.
| |
Collapse
|
3
|
Zhang X, Wang P, Shi G, Tang C, Xue H. AUNP-12 Near-Infrared Fluorescence Probes across NIR-I to NIR-II Enable In Vivo Detection of PD-1/PD-L1 Axis in the Tumor Microenvironment. Bioconjug Chem 2024; 35:1064-1074. [PMID: 38980173 PMCID: PMC11261610 DOI: 10.1021/acs.bioconjchem.4c00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
The innovative PD-1/PD-L1 pathway strategy is gaining significant traction in cancer therapeutics. However, fluctuating response rates of 20-40% to PD-1/PD-L1 inhibitors, coupled with the risk of hyperprogression after immunotherapy, underscore the need for accurate patient selection and the identification of more beneficiaries. Molecular imaging, specifically near-infrared (NIR) fluorescence imaging, is a valuable alternative for real-time, noninvasive visualization of dynamic PD-L1 expression in vivo. This research introduces AUNP-12, a novel PD-L1-targeting peptide antagonist conjugated with Cy5.5 and CH1055 for first (NIR-I) and second near-infrared (NIR-II) imaging. These probes have proven to be effective in mapping PD-L1 expression across various mouse tumor models, offering insights into tumor-immune interactions. This study highlights the potential of AUNP-12-Cy5.5 and AUNP-12-CH1055 for guiding clinical immunotherapy through precise patient stratification and dynamic monitoring, supporting the shift toward molecular imaging for personalized cancer care.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department
of Radiology, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Peking
Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Ping Wang
- Department
of Cardiology, The Second Medical Center & National Clinical Research
Center for Geriatric Diseases, Chinese People’s
Liberation Army (PLA) General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Guangyuan Shi
- University
of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, China
| | - Chu Tang
- Engineering
Research Center of Molecular and Neuro Imaging of Ministry of Education,
School of Life Science and Technology, Xidian
University, Xi’an 710126, China
| | - Huadan Xue
- Department
of Radiology, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Peking
Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Astudillo-Ortiz E, Babo PS, Sunde PT, Galler KM, Gomez-Florit M, Gomes ME. Endodontic Tissue Regeneration: A Review for Tissue Engineers and Dentists. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:491-513. [PMID: 37051704 DOI: 10.1089/ten.teb.2022.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
The paradigm shift in the endodontic field from replacement toward regenerative therapies has witnessed the ever-growing research in tissue engineering and regenerative medicine targeting pulp-dentin complex in the past few years. Abundant literature on the subject that has been produced, however, is scattered over diverse areas of knowledge. Moreover, the terminology and concepts are not always consensual, reflecting the range of research fields addressing this subject, from endodontics to biology, genetics, and engineering, among others. This fact triggered some misinterpretations, mainly when the denominations of different approaches were used as synonyms. The evaluation of results is not precise, leading to biased conjectures. Therefore, this literature review aims to conceptualize the commonly used terminology, summarize the main research areas on pulp regeneration, identify future trends, and ultimately clarify whether we are really on the edge of a paradigm shift in contemporary endodontics toward pulp regeneration.
Collapse
Affiliation(s)
- Esteban Astudillo-Ortiz
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
- Department of Endodontics, School of Dentistry, University of Cuenca, Cuenca, Ecuador
| | - Pedro S Babo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Pia T Sunde
- Department of Endodontics, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Kerstin M Galler
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
7
|
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
|
8
|
Zhou M, Liang S, Liu D, Ma K, Peng Y, Wang Z. Engineered Nanoprobes for Immune Activation Monitoring. ACS NANO 2022; 16:19940-19958. [PMID: 36454191 DOI: 10.1021/acsnano.2c09743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The activation of the immune system is critical for cancer immunotherapy and treatments of inflammatory diseases. Non-invasive visualization of immunoactivation is designed to monitor the dynamic nature of the immune response and facilitate the assessment of therapeutic outcomes, which, however, remains challenging. Conventional imaging modalities, such as positron emission tomography, computed tomography, etc., were utilized for imaging immune-related biomarkers. To explore the dynamic immune monitoring, probes with signals correlated to biomarkers of immune activation or prognosis are urgently needed. These emerging molecular probes, which turn on the signal only in the presence of the intended biomarker, can improve the detection specificity. These probes with "turn on" signals enable non-invasive, dynamic, and real-time imaging with high sensitivity and efficiency, showing significance for multifunctionality/multimodality imaging. As a result, more and more innovative engineered nanoprobes combined with diverse imaging modalities were developed to assess the activation of the immune system. In this work, we comprehensively review the recent and emerging advances in engineered nanoprobes for monitoring immune activation in cancer or other immune-mediated inflammatory diseases and discuss the potential in predicting the efficacy following treatments. Research on real-time in vivo immunoimaging is still under exploration, and this review can provide guidance and facilitate the development and application of next-generation imaging technologies.
Collapse
Affiliation(s)
- Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
9
|
Nguyen A, Kumar S, Kulkarni AA. Nanotheranostic Strategies for Cancer Immunotherapy. SMALL METHODS 2022; 6:e2200718. [PMID: 36382571 PMCID: PMC11056828 DOI: 10.1002/smtd.202200718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Despite advancements in cancer immunotherapy, heterogeneity in tumor response impose barriers to successful treatments and accurate prognosis. Effective therapy and early outcome detection are critical as toxicity profiles following immunotherapies can severely affect patients' quality of life. Existing imaging techniques, including positron emission tomography, computed tomography, magnetic resonance imaging, or multiplexed imaging, are often used in clinics yet suffer from limitations in the early assessment of immune response. Conventional strategies to validate immune response mainly rely on the Response Evaluation Criteria in Solid Tumors (RECIST) and the modified iRECIST for immuno-oncology drug trials. However, accurate monitoring of immunotherapy efficacy is challenging since the response does not always follow conventional RECIST criteria due to delayed and variable kinetics in immunotherapy responses. Engineered nanomaterials for immunotherapy applications have significantly contributed to overcoming these challenges by improving drug delivery and dynamic imaging techniques. This review summarizes challenges in recent immune-modulation approaches and traditional imaging tools, followed by emerging developments in three-in-one nanoimmunotheranostic systems co-opting nanotechnology, immunotherapy, and imaging. In addition, a comprehensive overview of imaging modalities in recent cancer immunotherapy research and a brief outlook on how nanotheranostic platforms can potentially advance to clinical translations for the field of immuno-oncology is presented.
Collapse
Affiliation(s)
- Anh Nguyen
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Sahana Kumar
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Ashish A. Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
10
|
Gao Y, Wu C, Chen X, Ma L, Zhang X, Chen J, Liao X, Liu M. PET/CT molecular imaging in the era of immune-checkpoint inhibitors therapy. Front Immunol 2022; 13:1049043. [PMID: 36341331 PMCID: PMC9630646 DOI: 10.3389/fimmu.2022.1049043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/10/2022] [Indexed: 04/24/2024] Open
Abstract
Cancer immunotherapy, especially immune-checkpoint inhibitors (ICIs), has paved a new way for the treatment of many types of malignancies, particularly advanced-stage cancers. Accumulating evidence suggests that as a molecular imaging modality, positron emission tomography/computed tomography (PET/CT) can play a vital role in the management of ICIs therapy by using different molecular probes and metabolic parameters. In this review, we will provide a comprehensive overview of the clinical data to support the importance of 18F-fluorodeoxyglucose PET/CT (18F-FDG PET/CT) imaging in the treatment of ICIs, including the evaluation of the tumor microenvironment, discovery of immune-related adverse events, evaluation of therapeutic efficacy, and prediction of therapeutic prognosis. We also discuss perspectives on the development direction of 18F-FDG PET/CT imaging, with a particular emphasis on possible challenges in the future. In addition, we summarize the researches on novel PET molecular probes that are expected to potentially promote the precise application of ICIs.
Collapse
|
11
|
Liu WL, Zhang YQ, Li LT, Zhu YY, Ming ZH, Chen WL, Yang RQ, Li RH, Chen M, Zhang GJ. Application of molecular imaging in immune checkpoints therapy: From response assessment to prognosis prediction. Crit Rev Oncol Hematol 2022; 176:103746. [PMID: 35752425 DOI: 10.1016/j.critrevonc.2022.103746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/30/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Recently, immune checkpoint therapy (ICT) represented by programmed cell death1 (PD-1) and its major ligands, programmed death ligand 1 (PD-L1), has achieved significant success. Detection of PD-L1 by immunohistochemistry (IHC) is a classic method to guide the treatment of ICT patients. However, PD-L1 expression in the tumor microenvironment is highly complex. Thus, PD-L1 IHC is inadequate to fully understand the relevance of PD-L1 levels in the whole body and their dynamics to improve therapeutic outcomes. Intriguingly, numerous studies have revealed that molecular imaging technologies could potentially meet this need. Therefore, the purpose of this narrative review is to summarize the preclinical and clinical application of ICT guided by molecular imaging technology, and to explore the future opportunities and practical difficulties of these innovations.
Collapse
Affiliation(s)
- Wan-Ling Liu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Liang-Tao Li
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Yuan-Yuan Zhu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Zi-He Ming
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Wei-Ling Chen
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Rui-Qin Yang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Rong-Hui Li
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Department of Medical Oncology, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China
| | - Min Chen
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China.
| | - Guo-Jun Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China; Cancer Research Center, School of Medicine, Xiamen University, 4221 South Xiang'an Road, Xiamen, China.
| |
Collapse
|
12
|
Kramer CS, Dimitrakopoulou-Strauss A. Immuno-Imaging (PET/SPECT)-Quo Vadis? MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103354. [PMID: 35630835 PMCID: PMC9147562 DOI: 10.3390/molecules27103354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023]
Abstract
The use of immunotherapy has revolutionized the treatment regimen of certain cancer types, but response assessment has become a difficult task with conventional methods such as CT/MRT or FDG PET-CT and the classical response criteria such as RECIST or PERCIST which have been developed for chemotherapeutic treatment. Plenty of new tracers have been published to improve the assessment of treatment response and to stratify the patient population. We gathered the information on published tracers (in total, 106 individual SPECT/PET tracers were identified) and performed a descriptor-based analysis; in this way, we classify the tracers with regard to target choice, developability (probability to progress from preclinical stage into the clinic), translatability (probability to be widely applied in the 'real world'), and (assumed) diagnostic quality. In our analysis, we show that most tracers are targeting PD-L1, PD-1, CTLA-4, and CD8 receptors by using antibodies or their fragments. Another finding is that plenty of tracers possess only minor iterations regarding chelators and nuclides instead of approaching the problem in a new innovative way. Based on the data, we suggest an orthogonal approach by targeting intracellular targets with PET-activatable small molecules that are currently underrepresented.
Collapse
Affiliation(s)
- Carsten S. Kramer
- Curanosticum Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, D-65191 Wiesbaden, Germany
- Correspondence:
| | | |
Collapse
|
13
|
Cheng Z, Du Y, Yu L, Yuan Z, Tian J. Application of Noninvasive Imaging to Combined Immune Checkpoint Inhibitors for Breast Cancer: Facts and Future. Mol Imaging Biol 2022; 24:264-279. [PMID: 35102468 DOI: 10.1007/s11307-021-01688-9] [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] [Received: 07/19/2021] [Revised: 11/13/2021] [Accepted: 11/24/2021] [Indexed: 12/19/2022]
Abstract
With the application of mono-immunotherapy in cancer, particularly immune checkpoint inhibitors, improved outcomes have been achieved. However, there are several limitations to immunotherapy, such as a poor response to the drugs, immune resistance, and immune-related adverse events. In recent years, studies of preclinical animal models and clinical trials have demonstrated that immune checkpoint inhibitors for breast cancer can significantly prolong the overall survival and quality of patients' lives. Meanwhile, combined immune checkpoint inhibitor treatment has attracted researchers' attention and showed great potential in the comprehensive treatment of breast cancer patients. Additionally, noninvasive imaging enables physicians to predict response to combined immunotherapeutic drugs, achieve treatment efficacy, and lead to better clinical management. Herein, we review the background of combined immune checkpoint inhibitor therapy and summarize its targeted imaging as well as progress in noninvasive imaging aimed at evaluating therapeutic outcomes. Finally, we describe several factors that may influence the outcome of this combined immunotherapy, the future direction of medical imaging, and the potential application of artificial intelligence in breast cancer. With further development of noninvasive imaging for the guidance of combined immune checkpoint inhibitors, cures for this disease may be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex System, Institute of Automation, Chinese Academy of Sciences, BeijingBeijing, 100190, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex System, Institute of Automation, Chinese Academy of Sciences, BeijingBeijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, China.
| | - Leyi Yu
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex System, Institute of Automation, Chinese Academy of Sciences, BeijingBeijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, 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, Shaanxi, 710071, China.
| |
Collapse
|
14
|
Li X, Wang R, Zhang Y, Han S, Gan Y, Liang Q, Ma X, Rong P, Wang W, Li W. Molecular imaging of tumor-associated macrophages in cancer immunotherapy. Ther Adv Med Oncol 2022; 14:17588359221076194. [PMID: 35251314 PMCID: PMC8891912 DOI: 10.1177/17588359221076194] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/10/2022] [Indexed: 12/20/2022] Open
Abstract
Tumor-associated macrophages (TAMs), the most abundant inflammatory cell group in the tumor microenvironment, play an essential role in tumor immune regulation. The infiltration degree of TAMs in the tumor microenvironment is closely related to tumor growth and metastasis, and TAMs have become a promising target in tumor immunotherapy. Molecular imaging is a new interdisciplinary subject that combines medical imaging technology with molecular biology, nuclear medicine, radiation medicine, and computer science. The latest progress in molecular imaging allows the biological processes of cells to be visualized in vivo, which makes it possible to better understand the density and distribution of macrophages in the tumor microenvironment. This review mainly discusses the application of targeting TAM in tumor immunotherapy and the imaging characteristics and progress of targeting TAM molecular probes using various imaging techniques.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Ruike Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yangnan Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Qi Liang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Xiaoqian Ma
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
15
|
Lecocq Q, Debie P, Puttemans J, Awad RM, De Beck L, Ertveldt T, De Vlaeminck Y, Goyvaerts C, Raes G, Keyaerts M, Breckpot K, Devoogdt N. Evaluation of single domain antibodies as nuclear tracers for imaging of the immune checkpoint receptor human lymphocyte activation gene-3 in cancer. EJNMMI Res 2021; 11:115. [PMID: 34727262 PMCID: PMC8563901 DOI: 10.1186/s13550-021-00857-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022] Open
Abstract
Recent advancements in the field of immune-oncology have led to a significant increase in life expectancy of patients with diverse forms of cancer, such as hematologic malignancies, melanoma and lung cancer. Unfortunately, these encouraging results are not observed in the majority of patients, who remain unresponsive and/or encounter adverse events. Currently, researchers are collecting more insight into the cellular and molecular mechanisms that underlie these variable responses. As an example, the human lymphocyte activation gene-3 (huLAG-3), an inhibitory immune checkpoint receptor, is increasingly studied as a therapeutic target in immune-oncology. Noninvasive molecular imaging of the immune checkpoint programmed death protein-1 (PD-1) or its ligand PD-L1 has shown its value as a strategy to guide and monitor PD-1/PD-L1-targeted immune checkpoint therapy. Yet, radiotracers that allow dynamic, whole body imaging of huLAG-3 expression are not yet described. We here developed single-domain antibodies (sdAbs) that bind huLAG-3 and showed that these sdAbs can image huLAG-3 in tumors, therefore representing promising tools for further development into clinically applicable radiotracers.
Collapse
Affiliation(s)
- Q Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - P Debie
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium
| | - J Puttemans
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium
| | - R M Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - L De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - T Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Y De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - C Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - G Raes
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium.,Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - K Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| | - N Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium.
| |
Collapse
|
16
|
Lecocq Q, Awad RM, De Vlaeminck Y, de Mey W, Ertveldt T, Goyvaerts C, Raes G, Thielemans K, Keyaerts M, Devoogdt N, Breckpot K. Single-Domain Antibody Nuclear Imaging Allows Noninvasive Quantification of LAG-3 Expression by Tumor-Infiltrating Leukocytes and Predicts Response of Immune Checkpoint Blockade. J Nucl Med 2021; 62:1638-1644. [PMID: 33712537 PMCID: PMC8612328 DOI: 10.2967/jnumed.120.258871] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/03/2021] [Indexed: 11/29/2022] Open
Abstract
Recent advances in the field of immune-oncology led to the discovery of next-generation immune checkpoints (ICPs). Lymphocyte activation gene-3 (LAG-3), being the most widely studied among them, is being explored as a target for the treatment of cancer patients. Several antagonistic anti-LAG-3 antibodies are being developed and are prime candidates for clinical application. Furthermore, validated therapies targeting cytotoxic T-lymphocyte-associated protein-4, programmed cell-death protein-1, or programmed cell-death ligand-1 showed that only subsets of patients respond. This finding highlights the need for better tools for patient selection and monitoring. The potential of molecular imaging to detect ICPs noninvasively in cancer is supported by several preclinical and clinical studies. Here, we report on a single-domain antibody to evaluate whole-body LAG-3 expression in various syngeneic mouse cancer models using nuclear imaging. Methods: SPECT/CT scans of tumor-bearing mice were performed 1 h after injection with radiolabeled single-domain antibody. Organs and tumors of mice were isolated and evaluated for the presence of the radiolabeled tracer and LAG-3-expressing immune cells using a γ-counter and flow cytometry respectively. PD-1/LAG-3-blocking antibodies were injected in MC38-bearing mice. Results: The radiolabeled single-domain antibody detected LAG-3 expression on tumor-infiltrating lymphocytes (TILs) as soon as 1 h after injection in MC38, MO4, and TC-1 cancer models. The single-domain antibody tracer visualized a compensatory upregulation of LAG-3 on TILs in MC38 tumors of mice treated with PD-1-blocking antibodies. When PD-1 blockade was combined with LAG-3 blockade, a synergistic effect on tumor growth delay was observed. Conclusion: These findings consolidate LAG-3 as a next-generation ICP and support the use of single-domain antibodies as tools to noninvasively monitor the dynamic evolution of LAG-3 expression by TILs, which could be exploited to predict therapy outcome.
Collapse
Affiliation(s)
- Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wout de Mey
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Raes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium; and
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
17
|
Jiang C, Zhang L, Xu X, Qi M, Zhang J, He S, Tian Q, Song S. Engineering a Smart Agent for Enhanced Immunotherapy Effect by Simultaneously Blocking PD-L1 and CTLA-4. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102500. [PMID: 34473430 PMCID: PMC8529437 DOI: 10.1002/advs.202102500] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/04/2021] [Indexed: 05/06/2023]
Abstract
Combinations of immune checkpoint therapies show encouraging results in the treatment of many human cancers. However, the higher costs and greater side effects of such combinations compared with single-agent immunotherapies limit their further applications. In this work, a novel smart agent, KN046@19 F-ZIF-8, is developed to overcome these limitations. KN046 is a novel recombinant humanized PD-L1/CTLA-4 bispecific single-domain antibody-Fc fusion protein, which can bind to both PD-L1 and CTLA-4 effectively. ZIF-8 is a smart delivery system, which can safely and effectively deliver KN406 to a tumor. In vitro and in vivo results demonstrate that the smart agent KN046@19 F-ZIF-8 not only improves the immune response rate of the antibody drug in treatment of tumors but also reduces its toxic side effects, thereby achieving excellent antitumor efficacy. This study provides an engineering strategy for clinical applications of a more effective immunotherapy.
Collapse
Affiliation(s)
- Chunjuan Jiang
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Le Zhang
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
- Department of Research and DevelopmentShanghai Proton and Heavy Ion CenterShanghai201321China
| | - Xiaoping Xu
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Ming Qi
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Jianping Zhang
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Simin He
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular ImagingShanghai University of Medicine and Health SciencesShanghai201318China
| | - Shaoli Song
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterShanghai200032China
- Center for Biomedical ImagingFudan UniversityShanghai200032China
- Shanghai Engineering Research Center of Molecular Imaging ProbesShanghai200032China
- Department of Research and DevelopmentShanghai Proton and Heavy Ion CenterShanghai201321China
| |
Collapse
|
18
|
Leung D, Bonacorsi S, Smith RA, Weber W, Hayes W. Molecular Imaging and the PD-L1 Pathway: From Bench to Clinic. Front Oncol 2021; 11:698425. [PMID: 34497758 PMCID: PMC8420047 DOI: 10.3389/fonc.2021.698425] [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: 04/21/2021] [Accepted: 07/22/2021] [Indexed: 01/24/2023] Open
Abstract
Programmed death-1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors target the important molecular interplay between PD-1 and PD-L1, a key pathway contributing to immune evasion in the tumor microenvironment (TME). Long-term clinical benefit has been observed in patients receiving PD-(L)1 inhibitors, alone and in combination with other treatments, across multiple tumor types. PD-L1 expression has been associated with response to immune checkpoint inhibitors, and treatment strategies are often guided by immunohistochemistry-based diagnostic tests assessing expression of PD-L1. However, challenges related to the implementation, interpretation, and clinical utility of PD-L1 diagnostic tests have led to an increasing number of preclinical and clinical studies exploring interrogation of the TME by real-time imaging of PD-(L)1 expression by positron emission tomography (PET). PET imaging utilizes radiolabeled molecules to non-invasively assess PD-(L)1 expression spatially and temporally. Several PD-(L)1 PET tracers have been tested in preclinical and clinical studies, with clinical trials in progress to assess their use in a number of cancer types. This review will showcase the development of PD-(L)1 PET tracers from preclinical studies through to clinical use, and will explore the opportunities in drug development and possible future clinical implementation.
Collapse
Affiliation(s)
- David Leung
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Samuel Bonacorsi
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Ralph Adam Smith
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Wolfgang Weber
- Technische Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wendy Hayes
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
19
|
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
|
20
|
Wu J, Liu J, Lin B, Lv R, Yuan Y, Tao X. Met-Targeted Dual-Modal MRI/NIR II Imaging for Specific Recognition of Head and Neck Squamous Cell Carcinoma. ACS Biomater Sci Eng 2021; 7:1640-1650. [DOI: 10.1021/acsbiomaterials.0c01807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Jun Wu
- Department of Radiology, School of Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jun Liu
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shanxi 710071, China
| | - Bi Lin
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shanxi 710071, China
| | - Ruichan Lv
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shanxi 710071, China
| | - Ying Yuan
- Department of Radiology, School of Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Xiaofeng Tao
- Department of Radiology, School of Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| |
Collapse
|
21
|
Abousaway O, Rakhshandehroo T, Van den Abbeele AD, Kircher MF, Rashidian M. Noninvasive Imaging of Cancer Immunotherapy. Nanotheranostics 2021; 5:90-112. [PMID: 33391977 PMCID: PMC7738948 DOI: 10.7150/ntno.50860] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has revolutionized the treatment of several malignancies. Notwithstanding the encouraging results, many patients do not respond to treatments. Evaluation of the efficacy of treatments is challenging and robust methods to predict the response to treatment are not yet available. The outcome of immunotherapy results from changes that treatment evokes in the tumor immune landscape. Therefore, a better understanding of the dynamics of immune cells that infiltrate into the tumor microenvironment may fundamentally help in addressing this challenge and provide tools to assess or even predict the response. Noninvasive imaging approaches, such as PET and SPECT that provide whole-body images are currently seen as the most promising tools that can shed light on the events happening in tumors in response to treatment. Such tools can provide critical information that can be used to make informed clinical decisions. Here, we review recent developments in the field of noninvasive cancer imaging with a focus on immunotherapeutics and nuclear imaging technologies and will discuss how the field can move forward to address the challenges that remain unresolved.
Collapse
Affiliation(s)
- Omar Abousaway
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| | - Taha Rakhshandehroo
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| | - Annick D. Van den Abbeele
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, USA
| | - Moritz F. Kircher
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, USA
| | - Mohammad Rashidian
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
22
|
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
|
23
|
Radiomics-based prediction model for outcomes of PD-1/PD-L1 immunotherapy in metastatic urothelial carcinoma. Eur Radiol 2020; 30:5392-5403. [PMID: 32394281 DOI: 10.1007/s00330-020-06847-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/27/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To evaluate the usefulness of a radiomics-based prediction model for predicting response and survival outcomes of patients with metastatic urothelial carcinoma treated with immunotherapy targeting programmed cell death 1 (PD-1) and its ligand (PD-L1). METHODS Sixty-two patients who underwent immunotherapy were divided into training (n = 41) and validation sets (n = 21). A total of 224 measurable lesions were identified on contrast-enhanced CT. A radiomics signature was constructed with features selected using a least absolute shrinkage and selection operator algorithm in the training set. A radiomics-based model was built based on a radiomics signature consisting of five reliable RFs and the presence of visceral organ involvement using multivariate logistic regression. According to a cutoff determined on the training set, patients in the validation set were assigned to either high- or low-risk groups. Kaplan-Meier analysis was performed to compare progression-free and overall survival between high- and low-risk groups. RESULTS For predicting objective response and disease control, the areas under the receiver operating characteristic curves of the radiomics-based model were 0.87 (95% CI, 0.65-0.97) and 0.88 (95% CI, 0.67-0.98) for the validation set, providing larger net benefit determined by decision curve analysis than without radiomics-based model. The high-risk group in the validation set showed shorter progression-free and overall survival than the low-risk group (log-rank p = 0.044 and p = 0.035). CONCLUSIONS The radiomics-based model may predict the response and survival outcome in patients treated with PD-1/PD-L1 immunotherapy for metastatic urothelial carcinoma. This approach may provide important and decision tool for planning immunotherapy. KEY POINTS • A radiomics-based model was built based on radiomics features and the presence of visceral organ involvement for prediction of outcomes in metastatic urothelial carcinoma treated with immunotherapy. • This prediction model demonstrated good prediction of treatment response and higher net benefit than no model in the independent validation set. • This radiomics-based model demonstrated significant associations with progression-free and overall survival between low-risk and high-risk groups.
Collapse
|
24
|
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, Wang Z. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1628. [PMID: 32314552 PMCID: PMC7507140 DOI: 10.1002/wnan.1628] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/06/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - María J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Applied Physics Graduate Program, Rice University, Houston, Texas, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anh-Dung Le
- Nanoscience and Microsystems Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Shreya Goel
- Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
25
|
Mouse Models for Immunotherapy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11111800. [PMID: 31731753 PMCID: PMC6896030 DOI: 10.3390/cancers11111800] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
Liver cancer is one of the dominant causes of cancer-related mortality, and the survival rate of liver cancer is among the lowest for all cancers. Immunotherapy for hepatocellular carcinoma (HCC) has yielded some encouraging results, but the percentage of patients responding to single-agent therapies remains low. Therefore, potential directions for improved immunotherapies include identifying new immune targets and checkpoints and customizing treatment procedures for individual patients. The development of combination therapies for HCC is also crucial and urgent and, thus, further studies are required. Mice have been utilized in immunotherapy research due to several advantages, for example, being low in cost, having high success rates for inducing tumor growth, and so on. Moreover, immune-competent mice are used in immunotherapy research to clarify the role that the immune system plays in cancer growth. In this review paper, the advantages and disadvantages of mouse models for immunotherapy, the equipment that are used for monitoring HCC, and the cell strains used for inducing HCC are reviewed.
Collapse
|
26
|
Li J, Van Valkenburgh J, Hong X, Conti PS, Zhang X, Chen K. Small molecules as theranostic agents in cancer immunology. Theranostics 2019; 9:7849-7871. [PMID: 31695804 PMCID: PMC6831453 DOI: 10.7150/thno.37218] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022] Open
Abstract
With further research into the molecular mechanisms and roles linking immune suppression and restraint of (pre)malignancies, immunotherapies have revolutionized clinical strategies in the treatment of cancer. However, nearly 70% of patients who received immune checkpoint therapeutics showed no response. Complementary and/or synergistic effects may occur when extracellular checkpoint antibody blockades combine with small molecules targeting intracellular signal pathways up/downstream of immune checkpoints or regulating the innate and adaptive immune response. After radiolabeling with radionuclides, small molecules can also be used for estimating treatment efficacy of immune checkpoint blockades. This review not only highlights some significant intracellular pathways and immune-related targets such as the kynurenine pathway, purinergic signaling, the kinase signaling axis, chemokines, etc., but also summarizes some attractive and potentially immunosuppression-related small molecule agents, which may be synergistic with extracellular immune checkpoint blockade. In addition, opportunities for small molecule-based theranostics in cancer immunology will be discussed.
Collapse
Affiliation(s)
- Jindian Li
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Juno Van Valkenburgh
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| | - Xingfang Hong
- Laboratory of Pathogen Biology, School of Basic Medical Sciences, Dali University, Dali 671000, China
| | - Peter S. Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Kai Chen
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| |
Collapse
|
27
|
Huang HF, Zhu H, Li GH, Xie Q, Yang XT, Xu XX, Tian XB, Wan YK, Yang Z. Construction of Anti-hPD-L1 HCAb Nb6 and in Situ 124I Labeling for Noninvasive Detection of PD-L1 Expression in Human Bone Sarcoma. Bioconjug Chem 2019; 30:2614-2623. [PMID: 31535847 DOI: 10.1021/acs.bioconjchem.9b00539] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunotherapy is considered the fourth major treatment mode for cancer following surgery, chemotherapy, and radiotherapy. In recent years, tumor immunotherapy has achieved breakthrough progress; therefore, it is important to screen patients to identify those who will respond to tumor immunotherapy. Here, we report the construction of a novel heavy chain-only antibody (HCAb) and its corresponding 124I-labeled probe. Using phage display technology, we generated a novel anti-hPD-L1-specific HCAb named Nb6 (selected from 95 monoclones) with high affinity for hPD-L1. The positron-emitting 124I-labeled hPD-L1-targeted HCAb probe was prepared for further evaluation, and nonradioactive natural iodine (natI)-labeled anti-hPD-L1 Nb6 was synthesized as a reference compound. 125I-anti-hPD-L1 Nb6 uptake in OS-732 cells in vitro can be blocked by the precursor. The binding affinity of 125I-anti-hPD-L1 Nb6 to OS-732 cell lines was 2.19 nM. For in vivo studies, an osteosarcoma OS-732 tumor-bearing mouse model was successfully constructed. Polymerase chain reaction (PCR) and Western blot analyses were performed to confirm the presence of the hPD-L1 gene and antigen in the tumor tissue of the OS-732 mouse model. Biodistribution showed that uptake of 124I-anti-hPD-L1 Nb6 probes at 24 h was 4.43 ± 0.33% ID/g in OS-732 tumor tissues. Tumor lesions can be clearly delineated on micro-PET (positron emission tomography)/CT (computed tomography) imaging 24 h after injection of 124I-anti-hPD-L1 Nb6, while the blocking group shows substantially decreased uptake on imaging. Pathological staining validated hPD-L1 expression on the surface of the tumor cell membrane; thus, 124I-anti-hPD-L1 Nb6 can be used for in vivo noninvasive PET imaging. When administered in tandem, Nb6 and 124I-anti-hPD-L1 Nb6 may provide a novel strategy to clinically screen patients for hPD-L1 to identify those who would benefit from immunotherapy of malignant tumors such as osteosarcoma.
Collapse
Affiliation(s)
- Hai-Feng Huang
- Guizhou University School of Medicine , Guizhou University , Guiyang 550025 , Guizhou , P. R. China.,Department of Orthopedics , Guizhou Provincial People's Hospital , Guiyang 550002 , Guizhou , P. R. China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine , Peking University Cancer Hospital & Institute , Beijing 100142 , P. R. China
| | - Guang-Hui Li
- Shanghai Novamab Biopharmaceuticals Co., Ltd. , Shanghai 201203 , P. R. China
| | - Quan Xie
- Guizhou University School of Medicine , Guizhou University , Guiyang 550025 , Guizhou , P. R. China
| | - Xian-Teng Yang
- Guizhou University School of Medicine , Guizhou University , Guiyang 550025 , Guizhou , P. R. China.,Department of Orthopedics , Guizhou Provincial People's Hospital , Guiyang 550002 , Guizhou , P. R. China
| | - Xiao-Xia Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine , Peking University Cancer Hospital & Institute , Beijing 100142 , P. R. China
| | - Xiao-Bin Tian
- Clinical Medical College of Guizhou Medical University , Guiyang 550025 , Guizhou , P. R. China
| | - Ya-Kun Wan
- Shanghai Novamab Biopharmaceuticals Co., Ltd. , Shanghai 201203 , P. R. China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine , Peking University Cancer Hospital & Institute , Beijing 100142 , P. R. China
| |
Collapse
|
28
|
Noninvasive Imaging of the Immune Checkpoint LAG-3 Using Nanobodies, from Development to Pre-Clinical Use. Biomolecules 2019; 9:biom9100548. [PMID: 31569553 PMCID: PMC6843898 DOI: 10.3390/biom9100548] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibition (ICI) is a promising cancer therapy, which has progressed rapidly from a preclinical concept to clinical implementation. Commonly considered targets in ICI are CTLA-4, PD-1/PD-L1, and LAG-3, and the list grows. As ICI is generally only beneficial for a subset of patients, there is a need to select patients that are eligible for therapy as well as to monitor therapy response. There is growing interest to do this noninvasively, by molecular imaging with target-specific tracers. To this day, noninvasive imaging has focused on CTLA-4 and PD-1/PD-L1, while there is no noninvasive tool available to accurately assess LAG-3 expression in vivo. In this proof-of-concept study, we developed nanobodies, the smallest functional fragments from camelid heavy chain-only antibodies, to noninvasively evaluate mouse LAG-3 expression using single photon emission computed tomography (SPECT)/CT imaging. The in vitro characterization of 114 nanobodies led to the selection of nine nanobodies binding to mouse LAG-3. The injection of 99mTechnetium-labeled nanobodies in healthy mice showed specific uptake in immune peripheral organs like the spleen and lymph nodes, which was not observed in LAG-3 gene knock-out mice. Moreover, nanobody uptake could be visualized using SPECT/CT and correlated to the presence of LAG-3 as assessed in flow cytometry and immunohistochemistry. SPECT/CT scans of tumor bearing mice further confirmed the diagnostic potential of the nanobodies. These findings substantiate the approach to use nanobodies as a tool to image inhibitory immune checkpoints in the tumor environment.
Collapse
|
29
|
Wei H, Jiang H, Song B. Role of medical imaging for immune checkpoint blockade therapy: From response assessment to prognosis prediction. Cancer Med 2019; 8:5399-5413. [PMID: 31385454 PMCID: PMC6745848 DOI: 10.1002/cam4.2464] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/29/2019] [Accepted: 07/15/2019] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint blockade (ICB) represents a promising approach in cancer therapy. Owing to the peculiar biologic mechanisms of anticancer activity, checkpoint blockers are accompanied with distinctive response patterns and toxicity profiles. Medical imaging is the cornerstone for response assessment to immunotherapy and plays a critical role in monitoring of immune-related adverse events (irAEs). Imaging-based biomarkers have shown tremendous potential for the prediction of therapeutic efficacies and clinical outcomes in patients treated with checkpoint inhibitors. In this article, the landscape of current response assessment systems for immunotherapy was reviewed with a special focus on the latest advances in the assessment of responses to ICB. Emerging imaging biomarkers were discussed along with the challenges regarding their clinical transformation. In addition, the biological mechanisms and clinical applications of ICB and irAEs were also within the scope of this review.
Collapse
Affiliation(s)
- Hong Wei
- Department of RadiologySichuan University West China HospitalChengduSichuan ProvinceChina
| | - Hanyu Jiang
- Department of RadiologySichuan University West China HospitalChengduSichuan ProvinceChina
| | - Bin Song
- Department of RadiologySichuan University West China HospitalChengduSichuan ProvinceChina
| |
Collapse
|
30
|
Radiofrequency ablation of liver metastasis: potential impact on immune checkpoint inhibitor therapy. Eur Radiol 2019; 29:5045-5051. [PMID: 30963271 DOI: 10.1007/s00330-019-06189-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/09/2019] [Accepted: 03/20/2019] [Indexed: 12/31/2022]
Abstract
Percutaneous radiofrequency ablation (RFA), a generally accepted alternative therapy for patients with liver metastases, is a minimally invasive approach with a favorable safety profile and a lower rate of major complications. The use of RFA or combined RFA plus resection can produce total tumor clearance in patients with unresectable liver metastases. However, the relatively high rate of local tumor progression has prevented the widespread use of RFA. Furthermore, its efficacy is controversial because there have been no comparisons for its effect on overall survival compared with standard options such as systemic chemotherapy. Meanwhile, immunotherapy has become a major research focus for oncology based on the recent successes reported for immune checkpoint inhibitors for melanoma, non-small cell lung cancer, gastric cancer, and other cancers. Immune checkpoints negatively regulate T cell function, and inhibition prevents the blockade of the immune system by cancer cells to prevent their destruction. Unfortunately, only some patients (< 25%) respond to immuno-oncology drugs, whereas other patients acquire resistance. However, RFA can induce massive necrotic cell death which might activate immunity and the presentation of cryptic antigens to induce tumor-specific T cell response. Because RFA can induce the rapid release of large amounts of tumor antigens, it can potentially stimulate transient immune responses to much tumor antigens. Combination therapies have induced synergistic enhancement of anticancer immune response in preclinical studies, indicating great promise for the future of oncologic treatment.Key Points • Only some patients respond to immuno-oncology drugs. • RFA causes the release of large amounts of cellular debris, a source of tumor antigens that elicit immune responses against tumors. • Combination RFA for liver metastases and immune checkpoint inhibitor therapies might synergistically enhance antitumor immunity.
Collapse
|