1
|
Peeters E, van Genugten EAJ, Heskamp S, de Vries IJM, van Herpen C, Koenen HJPM, Kneilling M, van der Post RS, van Dop WA, Westdorp H, Aarntzen E. Exploring molecular imaging to investigate immune checkpoint inhibitor-related toxicity. J Immunother Cancer 2025; 13:e011009. [PMID: 40341021 PMCID: PMC12060888 DOI: 10.1136/jitc-2024-011009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) boost the endogenous anticancer immunity, evoking long-lasting anticancer responses in a subset of patients with solid tumors. Simultaneously, ICI are also associated with serious toxicities, impacting treatment duration and the quality of life. The proposed processes underlying ICI-related toxicity include T-cell activation and recruitment to non-tumor tissues, involvement of other immune cells and fibroblasts and the host' microbiome composition. However, the exact mechanisms of these processes remain incompletely understood, hindering clinicians' ability to predict and identify ICI-related toxicity in the early stages of treatment. Molecular imaging may play a role as a non-invasive biomarker, providing a tool to study ICI-related toxicity. This review discusses the applications of molecular imaging to answer questions regarding the mechanisms, detection, and prediction of ICI-related toxicity. Potential targets and the current state of development of suitable imaging techniques are discussed.
Collapse
Affiliation(s)
- Eva Peeters
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Sandra Heskamp
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla van Herpen
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Department of Dermatology, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Willemijn A van Dop
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Harm Westdorp
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik Aarntzen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine, Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Kang HS, Lim HK, Jang WY, Cho JY. Anti-Colorectal Cancer Activity of Panax and Its Active Components, Ginsenosides: A Review. Int J Mol Sci 2025; 26:2593. [PMID: 40141242 PMCID: PMC11941759 DOI: 10.3390/ijms26062593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Colorectal cancer (CRC) poses a significant health burden worldwide and necessitates novel treatment approaches with fewer side effects than conventional chemotherapy. Many natural compounds have been tested as possible cancer treatments. Plants in the genus Panax have been widely studied due to their therapeutic potential for various diseases such as inflammatory disorders and cancers. Extracts from plants of genus Panax activate upstream signals, including those related to autophagy and the generation of reactive oxygen species, to induce intrinsic apoptosis in CRC cells. The root extract of Panax notoginseng (P. notoginseng) regulated the gut microbiota to enhance the T-cell-induced immune response against CRC. Protopanaxadiol (PPD)-type ginsenosides, especially Rh2, Rg3, Rb1, and Rb2, significantly reduced proliferation of CRC cells and tumor size in a xenograft mouse model, as well as targeting programmed death (PD)-1 to block the immune checkpoint of CRC cells. Moreover, modified nanocarriers with ginsenosides upregulated drug efficacy, showing that ginsenosides can also be utilized as drug carriers. An increasing body of studies has demonstrated the potential of the genus Panax in curing CRC. Ginsenosides are promising active compounds in the genus Panax, which can also support the activity of conventional cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (H.S.K.); (H.K.L.); (W.Y.J.)
| |
Collapse
|
3
|
Yang M, Hou S, Chen Y, Chen H, Chu M, Liu SB. Emerging insights into intravital imaging, unraveling its role in cancer immunotherapy. Cancer Immunol Immunother 2025; 74:100. [PMID: 39904769 PMCID: PMC11794739 DOI: 10.1007/s00262-025-03944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Cancer immunotherapy has attracted great attention as a potential therapeutic approach for advanced malignancies due to its promising survival benefits. Comprehension of intricate interactions between the tumor microenvironment (TME) and immune checkpoint inhibitors (ICIs) is crucial for optimizing and improving immunotherapies. Currently, several experimental strategies are available to monitor this complexity but most of them fail to facilitate real-time monitoring of the immune response such as cellular phagocytosis and cytolysis. Consequently, the application of intravital imaging has been extensively studied in the domain of cancer immunotherapy. Intravital imaging has been proven to be a powerful real-time imaging modality that provides insights into intratumoral immune responses, cellular metabolic signatures, tumor vasculature, and cellular functions. This review aims to provide a comprehensive overview of the latest research on intravital imaging in cancer immunotherapy, especially addressing how intravital imaging sheds light on essential features of tumor immunity, immune infiltrations, tumor angiogenesis, and aids in the clarification of underlying immunotherapeutic mechanisms. Moreover, a variety of labeling tools, imaging windows and models for real-time visualizations of TME are also summarized. We will also investigate the full potential of using intravital imaging to circumvent the limitations of currently available imaging modalities, which hold promise to advent efficient immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Minfeng Yang
- School of Public Health, Nantong University, Nantong, China
| | - Shiqiang Hou
- The First People's Hospital of Chuzhou, The Affiliated Chuzhou Hospital of Anhui Medical University, Chuzhou, China
| | - Yao Chen
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou, 215009, China
| | - Hongzhao Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, People's Republic of China
| | - Minjie Chu
- School of Public Health, Nantong University, Nantong, China.
| | - Song-Bai Liu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Suzhou, 215009, China.
| |
Collapse
|
4
|
Cai J, Zhang M, Peng J, Wei Y, Zhu W, Guo K, Gao M, Wang H, Wang H, Wang L. Peptide-AIE Nanofibers Functionalized Sutures with Antimicrobial Activity and Subcutaneous Traceability. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400531. [PMID: 38716716 DOI: 10.1002/adma.202400531] [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: 01/11/2024] [Revised: 04/12/2024] [Indexed: 05/18/2024]
Abstract
As one of the most widely used medical devices, sutures face challenges related to surgical site infections (SSIs) and lack of subcutaneous traceability. In the present study, a facile and effective approach using peptide-AIE nanofibers (NFs-K18) to create fluorescent-traceable antimicrobial sutures, which have been applied to four commercially available sutures is developed. The functionalized sutures of PGAS-NFs-K18 and PGLAS-NFs-K18 exhibit fluorescence with excellent penetration from 4 mm chicken breasts. They also demonstrate remarkable stability after 24 h of white light illumination and threading through chicken breasts 10 times. These sutures efficiently generate ROS, resulting in significant suppression of four clinical bacteria, with the highest antimicrobial rate of ≈100%. Moreover, the sutures exhibit favorable hemocompatibility and biocompatibility. In vivo experiments demonstrate that the optimized PGLAS-NFs-K18 suture displays potent antimicrobial activity against MRSA, effectively inhibiting inflammation and promoting tissue healing in both skin wound and abdominal wall wound models, outperforming the commercially available Coated VICRYL Plus Antibacterial suture. Importantly, PGLAS-NFs-K18 exhibits sensitive subcutaneous traceability, allowing for accurate in situ monitoring of its degradation. It is believed that this straightforward strategy offers a new pathway for inhibiting SSIs and monitoring the status of sutures.
Collapse
Affiliation(s)
- Junyi Cai
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Meng Zhang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Jingqi Peng
- The Third General Surgery Department, Xinjiang Uygur Autonomous Region Traditional Chinese Medicine Research Institute The Fourth Affiliated Hospital of Xinjiang Medical University), Urumqi, 830011, China
| | - Yingqi Wei
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Henan Provincial Key Laboratory of Radiation Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wenchao Zhu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Kunzhong Guo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Meng Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Hui Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Huaiming Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Lin Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
5
|
Simunic M, Joshi JT, Merkens H, Colpo N, Kuo HT, Lum JJ, Bénard F. PSMA imaging as a non-invasive tool to monitor inducible gene expression in vivo. EJNMMI Res 2024; 14:3. [PMID: 38177950 PMCID: PMC10767034 DOI: 10.1186/s13550-023-01063-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Affiliation(s)
- Marin Simunic
- Department of Hematology, Clinic for Internal Medicine, Clinical Hospital Centre, Spinciceva 1, 21000, Split, Croatia
| | - Jay T Joshi
- Deeley Research Centre, BC Cancer Research Institute, 2410 Lee Avenue, Victoria, BC, V8R 6V5, Canada
| | - Helen Merkens
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Nadine Colpo
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Hsiou-Ting Kuo
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Julian J Lum
- Deeley Research Centre, BC Cancer Research Institute, 2410 Lee Avenue, Victoria, BC, V8R 6V5, Canada
| | - François Bénard
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
6
|
Kheyrolahzadeh K, Tohidkia MR, Tarighatnia A, Shahabi P, Nader ND, Aghanejad A. Theranostic chimeric antigen receptor (CAR)-T cells: Insight into recent trends and challenges in solid tumors. Life Sci 2023; 328:121917. [PMID: 37422069 DOI: 10.1016/j.lfs.2023.121917] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/15/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Cell therapy has reached significant milestones in various life-threatening diseases, including cancer. Cell therapy using fluorescent and radiolabeled chimeric antigen receptor (CAR)-T cell is a successful strategy for diagnosing or treating malignancies. Since cell therapy approaches have different results in cancers, the success of hematological cancers has yet to transfer to solid tumor therapy, leading to more casualties. Therefore, there are many areas for improvement in the cell therapy platform. Understanding the therapeutic barriers associated with solid cancers through cell tracking and molecular imaging may provide a platform for effectively delivering CAR-T cells into solid tumors. This review describes CAR-T cells' role in treating solid and non-solid tumors and recent advances. Furthermore, we discuss the main obstacles, mechanism of action, novel strategies and solutions to overcome the challenges from molecular imaging and cell tracking perspectives.
Collapse
Affiliation(s)
- Keyvan Kheyrolahzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Tarighatnia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, United States of America
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Wang S, Wang Y, Xu B, Qin T, Lv Y, Yan H, Shao Y, Fang Y, Zheng S, Qiu Y. Biodistribution of 89Zr-oxine-labeled human bone marrow-derived mesenchymal stem cells by micro-PET/computed tomography imaging in Sprague-Dawley rats. Nucl Med Commun 2022; 43:834-846. [PMID: 35438673 PMCID: PMC9177155 DOI: 10.1097/mnm.0000000000001562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To develop a method for labeling human bone marrow mesenchymal stem cells (hMSCs) with 89Zr-oxine to characterize the biodistribution characteristics of hMSCs in normal Sprague-Dawley (SD) rats in real-time by micro-PET-computed tomography (micro-PET/CT) imaging. METHODS 89Zr-oxine complex was synthesized from 89Zr-oxalate and 8-hydroxyquinoline (oxine). After hMSCs were labeled with the 89Zr-oxine complex, the radioactivity retention, viability, proliferation, apoptosis, differentiation, morphology, and phenotype of labeled cells were assessed. The biodistribution of 89Zr-oxine-labeled hMSCs in SD rats was tracked in real-time by micro-PET/CT imaging. RESULTS The cell labeling efficiency was 52.6 ± 0.01%, and 89Zr-oxine was stably retained in cells (66.7 ± 0.9% retention on 7 days after labeling). Compared with the unlabeled hMSCs, 89Zr-oxine labeling did not affect the biological characteristics of cells. Following intravenous administration in SD rats, labeled hMSCs mainly accumulated in the liver (7.35 ± 1.41% ID/g 10 days after labeling, n = 6) and spleen (8.48 ± 1.20% ID/g 10 days after labeling, n = 6), whereas intravenously injected 89Zr-oxalate mainly accumulated in the bone (4.47 ± 0.35% ID/g 10 days after labeling, n = 3). CONCLUSION 89Zr-oxine labeling and micro-PET/CT imaging provide a useful and non-invasive method of assessing the biodistribution of cell therapy products in SD rats. The platform provides a foundation for us to further understand the mechanism of action and migration dynamics of cell therapy products.
Collapse
Affiliation(s)
- Shuzhe Wang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science
- Toxicology Department of China Academy of Pharmaceutical Industry Shanghai InnoStar Biotechnology Co. Ltd, Shanghai
| | - Yan Wang
- Toxicology Department of China Academy of Pharmaceutical Industry Shanghai InnoStar Biotechnology Co. Ltd, Shanghai
| | - Bohua Xu
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Tian Qin
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Yupeng Lv
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Heng Yan
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Yifei Shao
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Yangyang Fang
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Shaoqiu Zheng
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
- Jiangxi University of Traditional Chinese Medicine, Nanchang
- Yangtze Delta Advanced Research Institute, Yangtze Delta Pharmaceutical College Nantong, Jiangsu, China
| | - Yunliang Qiu
- Toxicology Department of China Academy of Pharmaceutical Industry Shanghai InnoStar Biotechnology Co. Ltd, Shanghai
| |
Collapse
|
8
|
Sandker GGW, Adema G, Molkenboer-Kuenen J, Wierstra P, Bussink J, Heskamp S, Aarntzen EHJG. PD-L1 Antibody Pharmacokinetics and Tumor Targeting in Mouse Models for Infectious Diseases. Front Immunol 2022; 13:837370. [PMID: 35359962 PMCID: PMC8960984 DOI: 10.3389/fimmu.2022.837370] [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: 12/16/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background Programmed death-ligand 1 (PD-L1) regulates immune homeostasis by promoting T-cell exhaustion. It is involved in chronic infections and tumor progression. Nuclear imaging using radiolabeled anti-PD-L1 antibodies can monitor PD-L1 tissue expression and antibody distribution. However, physiological PD-L1 can cause rapid antibody clearance from blood at imaging doses. Therefore, we hypothesized that inflammatory responses, which can induce PD-L1 expression, affect anti-PD-L1 antibody distribution. Here, we investigated the effects of three different infectious stimuli on the pharmacokinetics and tumor targeting of radiolabeled anti-PD-L1 antibodies in tumor-bearing mice. Materials/Methods Anti-mouse-PD-L1 and isotype control antibodies were labelled with indium-111 ([111In]In-DTPA-anti-mPD-L1 and [111In]In-DTPA-IgG2a, respectively). We evaluated the effect of inflammatory responses on the pharmacokinetics of [111In]In-DTPA-anti-mPD-L1 in RenCa tumor-bearing BALB/c mice in three conditions: lipopolysaccharide (LPS), local Staphylococcus aureus, and heat-killed Candida albicans. After intravenous injection of 30 or 100 µg of [111In]In-DTPA-anti-mPD-L1 or [111In]In-DTPA-IgG2a, blood samples were collected 1, 4, and 24 h p.i. followed by microSPECT/CT and ex vivo biodistribution analyses. PD-L1 expression, neutrophil, and macrophage infiltration in relevant tissues were evaluated immunohistochemically. Results In 30 µg of [111In]In-DTPA-anti-mPD-L1 injected tumor-bearing mice the LPS-challenge significantly increased lymphoid organ uptake compared with vehicle controls (spleen: 49.9 ± 4.4%ID/g versus 21.2 ± 6.9%ID/g, p < 0.001), resulting in lower blood levels (3.6 ± 1.6%ID/g versus 11.5 ± 7.2%ID/g; p < 0.01) and reduced tumor targeting (8.1 ± 4.5%ID/g versus 25.2 ± 5.2%ID/g, p < 0.001). Local S. aureus infections showed high PD-L1+ neutrophil influx resulting in significantly increased [111In]In-DTPA-anti-mPD-L1 uptake in affected muscles (8.6 ± 2.6%ID/g versus 1.7 ± 0.8%ID/g, p < 0.001). Heat-killed Candida albicans (Hk-C. albicans) challenge did not affect pharmacokinetics. Increasing [111In]In-DTPA-anti-mPD-L1 dose to 100 µg normalized blood clearance and tumor uptake in LPS-challenged mice, although lymphoid organ uptake remained higher. Infectious stimuli did not affect [111In]In-DTPA-IgG2a pharmacokinetics. Conclusions This study shows that anti-PD-L1 antibody pharmacokinetics and tumor targeting can be significantly altered by severe inflammatory responses, which can be compensated for by increasing the tracer dose. This has implications for developing clinical PD-L1 imaging protocols in onco-immunology. We further demonstrate that radiolabeled anti-PD-L1 antibodies can be used to evaluate PD-L1 expression changes in a range of infectious diseases. This supports the exploration of using these techniques to assess hosts' responses to infectious stimuli.
Collapse
Affiliation(s)
- Gerwin G W Sandker
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gosse Adema
- Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janneke Molkenboer-Kuenen
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Wierstra
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
9
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
10
|
Immune Checkpoint Inhibitor Therapy for Bone Metastases: Specific Microenvironment and Current Situation. J Immunol Res 2021; 2021:8970173. [PMID: 34877360 PMCID: PMC8645368 DOI: 10.1155/2021/8970173] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/19/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022] Open
Abstract
The treatment of bone metastases is a thorny issue. Immunotherapy may be one of the few hopes for patients with unresectable bone metastases. Immune checkpoint inhibitors are the most commonly used immunotherapy drugs currently. In this review, the characteristics and interaction of bone metastases and their immune microenvironment were systematically discussed, and the relevant research progress of the immunological mechanism of tumor bone metastasis was reviewed. On this basis, we expounded the clinical application of immune checkpoint inhibitors for bone metastasis of common tumors, including non-small-cell lung cancer, renal cell carcinoma, prostate cancer, melanoma, and breast cancer. Then, the deficiencies and limitations in current researches were summarized. In-depth basic research on bone metastases and optimization of clinical treatment is needed.
Collapse
|
11
|
Van Hoeck J, Vanhove C, De Smedt SC, Raemdonck K. Non-invasive cell-tracking methods for adoptive T cell therapies. Drug Discov Today 2021; 27:793-807. [PMID: 34718210 DOI: 10.1016/j.drudis.2021.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/26/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022]
Abstract
Adoptive T cell therapies (ACT) have demonstrated groundbreaking results in blood cancers and melanoma. Nevertheless, their significant cost, the occurrence of severe adverse events, and their poor performance in solid tumors are important hurdles hampering more widespread applicability. In vivo cell tracking allows instantaneous and non-invasive monitoring of the distribution, tumor homing, persistence, and redistribution to other organs of infused T cells in patients. Furthermore, cell tracking could aid in the clinical management of patients, allowing the detection of non-responders or severe adverse events at an early stage. This review provides a concise overview of the main principles and potential of cell tracking, followed by a discussion of the clinically relevant labeling strategies and their application in ACT.
Collapse
Affiliation(s)
- Jelter Van Hoeck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Christian Vanhove
- Infinity Lab, Medical Imaging and Signal Processing Group-IBiTech, Faculty of Engineering and Architecture, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
12
|
Krekorian M, Sandker GGW, Cortenbach KRG, Tagit O, van Riessen NK, Raavé R, Srinivas M, Figdor CG, Heskamp S, Aarntzen EHJG. Characterization of Intrinsically Radiolabeled Poly(lactic- co-glycolic acid) Nanoparticles for ex Vivo Autologous Cell Labeling and in Vivo Tracking. Bioconjug Chem 2021; 32:1802-1811. [PMID: 34161070 PMCID: PMC8377710 DOI: 10.1021/acs.bioconjchem.1c00271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/11/2021] [Indexed: 02/04/2023]
Abstract
With the advent of novel immunotherapies, interest in ex vivo autologous cell labeling for in vivo cell tracking has revived. However, current clinically available labeling strategies have several drawbacks, such as release of radiolabel over time and cytotoxicity. Poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) are clinically used biodegradable carriers of contrast agents, with high loading capacity for multimodal imaging agents. Here we show the development of PLGA-based NPs for ex vivo cell labeling and in vivo cell tracking with SPECT. We used primary amine-modified PLGA polymers (PLGA-NH2) to construct NPs similar to unmodified PLGA NPs. PLGA-NH2 NPs were efficiently radiolabeled without chelator and retained the radionuclide for 2 weeks. Monocyte-derived dendritic cells labeled with [111In]In-PLGA-NH2 showed higher specific activity than those labeled with [111In]In-oxine, with no negative effect on cell viability. SPECT/CT imaging showed that radiolabeled THP-1 cells accumulated at the Staphylococcus aureus infection site in mice. In conclusion, PLGA-NH2 NPs are able to retain 111In, independent of chelator presence. Furthermore, [111In]In-PLGA-NH2 allows cell labeling with high specific activity and no loss of activity over prolonged time intervals. Finally, in vivo tracking of ex vivo labeled THP-1 cells was demonstrated in an infection model using SPECT/CT imaging.
Collapse
Affiliation(s)
- Massis Krekorian
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
- Department
of Medical Imaging, Radboud Institute for
Molecular Life Sciences, Radboud university Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Gerwin G. W. Sandker
- Department
of Medical Imaging, Radboud Institute for
Molecular Life Sciences, Radboud university Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Kimberley R. G. Cortenbach
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Oya Tagit
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - N. Koen van Riessen
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
- Cenya
Imaging BV, Tweede Kostverlorenkade
11H, 1052 RK Amsterdam, The Netherlands
| | - René Raavé
- Department
of Medical Imaging, Radboud Institute for
Molecular Life Sciences, Radboud university Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Mangala Srinivas
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
- Cenya
Imaging BV, Tweede Kostverlorenkade
11H, 1052 RK Amsterdam, The Netherlands
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department
of Medical Imaging, Radboud Institute for
Molecular Life Sciences, Radboud university Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Erik H. J. G. Aarntzen
- Department
of Medical Imaging, Radboud Institute for
Molecular Life Sciences, Radboud university Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
13
|
Made to Measure: Patient-Tailored Treatment of Multiple Sclerosis Using Cell-Based Therapies. Int J Mol Sci 2021; 22:ijms22147536. [PMID: 34299154 PMCID: PMC8304207 DOI: 10.3390/ijms22147536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, there is still no cure for multiple sclerosis (MS), which is an autoimmune and neurodegenerative disease of the central nervous system. Treatment options predominantly consist of drugs that affect adaptive immunity and lead to a reduction of the inflammatory disease activity. A broad range of possible cell-based therapeutic options are being explored in the treatment of autoimmune diseases, including MS. This review aims to provide an overview of recent and future advances in the development of cell-based treatment options for the induction of tolerance in MS. Here, we will focus on haematopoietic stem cells, mesenchymal stromal cells, regulatory T cells and dendritic cells. We will also focus on less familiar cell types that are used in cell therapy, including B cells, natural killer cells and peripheral blood mononuclear cells. We will address key issues regarding the depicted therapies and highlight the major challenges that lie ahead to successfully reverse autoimmune diseases, such as MS, while minimising the side effects. Although cell-based therapies are well known and used in the treatment of several cancers, cell-based treatment options hold promise for the future treatment of autoimmune diseases in general, and MS in particular.
Collapse
|
14
|
Van Tilbeurgh M, Lemdani K, Beignon AS, Chapon C, Tchitchek N, Cheraitia L, Marcos Lopez E, Pascal Q, Le Grand R, Maisonnasse P, Manet C. Predictive Markers of Immunogenicity and Efficacy for Human Vaccines. Vaccines (Basel) 2021; 9:579. [PMID: 34205932 PMCID: PMC8226531 DOI: 10.3390/vaccines9060579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines represent one of the major advances of modern medicine. Despite the many successes of vaccination, continuous efforts to design new vaccines are needed to fight "old" pandemics, such as tuberculosis and malaria, as well as emerging pathogens, such as Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination aims at reaching sterilizing immunity, however assessing vaccine efficacy is still challenging and underscores the need for a better understanding of immune protective responses. Identifying reliable predictive markers of immunogenicity can help to select and develop promising vaccine candidates during early preclinical studies and can lead to improved, personalized, vaccination strategies. A systems biology approach is increasingly being adopted to address these major challenges using multiple high-dimensional technologies combined with in silico models. Although the goal is to develop predictive models of vaccine efficacy in humans, applying this approach to animal models empowers basic and translational vaccine research. In this review, we provide an overview of vaccine immune signatures in preclinical models, as well as in target human populations. We also discuss high-throughput technologies used to probe vaccine-induced responses, along with data analysis and computational methodologies applied to the predictive modeling of vaccine efficacy.
Collapse
Affiliation(s)
- Matthieu Van Tilbeurgh
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Katia Lemdani
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Nicolas Tchitchek
- Unité de Recherche i3, Inserm UMR-S 959, Bâtiment CERVI, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Lina Cheraitia
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Ernesto Marcos Lopez
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Quentin Pascal
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Pauline Maisonnasse
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Caroline Manet
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| |
Collapse
|
15
|
Xiao Z, Puré E. Imaging of T-cell Responses in the Context of Cancer Immunotherapy. Cancer Immunol Res 2021; 9:490-502. [PMID: 33941536 DOI: 10.1158/2326-6066.cir-20-0678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/18/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022]
Abstract
Immunotherapy, which promotes the induction of cytotoxic T lymphocytes and enhances their infiltration into and function within tumors, is a rapidly expanding and evolving approach to treating cancer. However, many of the critical denominators for inducing effective anticancer immune responses remain unknown. Efforts are underway to develop comprehensive ex vivo assessments of the immune landscape of patients prior to and during response to immunotherapy. An important complementary approach to these efforts involves the development of noninvasive imaging approaches to detect immune targets, assess delivery of immune-based therapeutics, and evaluate responses to immunotherapy. Herein, we review the merits and limitations of various noninvasive imaging modalities (MRI, PET, and single-photon emission tomography) and discuss candidate targets for cellular and molecular imaging for visualization of T-cell responses at various stages along the cancer-immunity cycle in the context of immunotherapy. We also discuss the potential use of these imaging strategies in monitoring treatment responses and predicting prognosis for patients treated with immunotherapy.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
16
|
Arlauckas S, Oh N, Li R, Weissleder R, Miller MA. Macrophage imaging and subset analysis using single-cell RNA sequencing. Nanotheranostics 2021; 5:36-56. [PMID: 33391974 PMCID: PMC7738942 DOI: 10.7150/ntno.50185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages have been associated with drug response and resistance in diverse settings, thus raising the possibility of using macrophage imaging as a companion diagnostic to inform personalized patient treatment strategies. Nanoparticle-based contrast agents are especially promising because they efficiently deliver fluorescent, magnetic, and/or radionuclide labels by leveraging the intrinsic capacity of macrophages to accumulate nanomaterials in their role as professional phagocytes. Unfortunately, current clinical imaging modalities are limited in their ability to quantify broad molecular programs that may explain (a) which particular cell subsets a given imaging agent is actually labeling, and (b) what mechanistic role those cells play in promoting drug response or resistance. Highly multiplexed single-cell approaches including single-cell RNA sequencing (scRNAseq) have emerged as resources to help answer these questions. In this review, we query recently published scRNAseq datasets to support companion macrophage imaging, with particular focus on using dextran-based nanoparticles to predict the action of anti-cancer nanotherapies and monoclonal antibodies.
Collapse
Affiliation(s)
- Sean Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Nuri Oh
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Lau D, Garçon F, Chandra A, Lechermann LM, Aloj L, Chilvers ER, Corrie PG, Okkenhaug K, Gallagher FA. Intravital Imaging of Adoptive T-Cell Morphology, Mobility and Trafficking Following Immune Checkpoint Inhibition in a Mouse Melanoma Model. Front Immunol 2020; 11:1514. [PMID: 32793206 PMCID: PMC7387409 DOI: 10.3389/fimmu.2020.01514] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Efficient T-cell targeting, infiltration and activation within tumors is crucial for successful adoptive T-cell therapy. Intravital microscopy is a powerful tool for the visualization of T-cell behavior within tumors, as well as spatial and temporal heterogeneity in response to immunotherapy. Here we describe an experimental approach for intravital imaging of adoptive T-cell morphology, mobility and trafficking in a skin-flap tumor model, following immune modulation with immune checkpoint inhibitors (ICIs) targeting PD-L1 and CTLA-4. A syngeneic model of ovalbumin and mCherry-expressing amelanotic mouse melanoma was used in conjunction with adoptively transferred OT-1+ cytotoxic T-cells expressing GFP to image antigen-specific live T-cell behavior within the tumor microenvironment. Dynamic image analysis of T-cell motility showed distinct CD8+ T-cell migration patterns and morpho-dynamics within different tumor compartments in response to ICIs: this approach was used to cluster T-cell behavior into four groups based on velocity and meandering index. The results showed that most T-cells within the tumor periphery demonstrated Lévy-like trajectories, consistent with tumor cell searching strategies. T-cells adjacent to tumor cells had reduced velocity and appeared to probe the local environment, consistent with cell-cell interactions. An increased number of T-cells were detected following treatment, traveling at lower mean velocities than controls, and demonstrating reduced displacement consistent with target engagement. Histogram-based analysis of immunofluorescent images from harvested tumors showed that in the ICI-treated mice there was a higher density of CD31+ vessels compared to untreated controls and a greater infiltration of T-cells towards the tumor core, consistent with increased cellular trafficking post-treatment.
Collapse
Affiliation(s)
- Doreen Lau
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Fabien Garçon
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Anita Chandra
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Luigi Aloj
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Edwin R. Chilvers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Pippa G. Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ferdia A. Gallagher
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Ashmore-Harris C, Iafrate M, Saleem A, Fruhwirth GO. Non-invasive Reporter Gene Imaging of Cell Therapies, including T Cells and Stem Cells. Mol Ther 2020; 28:1392-1416. [PMID: 32243834 PMCID: PMC7264441 DOI: 10.1016/j.ymthe.2020.03.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/15/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Cell therapies represent a rapidly emerging class of new therapeutics. They are intended and developed for the treatment of some of the most prevalent human diseases, including cancer, diabetes, and for regenerative medicine. Currently, they are largely developed without precise assessment of their in vivo distribution, efficacy, or survival either clinically or preclinically. However, it would be highly beneficial for both preclinical cell therapy development and subsequent clinical use to assess these parameters in situ to enable enhancements in efficacy, applicability, and safety. Molecular imaging can be exploited to track cells non-invasively on the whole-body level and can enable monitoring for prolonged periods in a manner compatible with rapidly expanding cell types. In this review, we explain how in vivo imaging can aid the development and clinical translation of cell-based therapeutics. We describe the underlying principles governing non-invasive in vivo long-term cell tracking in the preclinical and clinical settings, including available imaging technologies, reporter genes, and imaging agents as well as pitfalls related to experimental design. Our emphasis is on adoptively transferred T cell and stem cell therapies.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Madeleine Iafrate
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Adeel Saleem
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK; Department of Haematological Medicine, King's College Hospital, London SE5 9RS, UK
| | - Gilbert O Fruhwirth
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK.
| |
Collapse
|
19
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
20
|
Ashmore-Harris C, Fruhwirth GO. The clinical potential of gene editing as a tool to engineer cell-based therapeutics. Clin Transl Med 2020; 9:15. [PMID: 32034584 PMCID: PMC7007464 DOI: 10.1186/s40169-020-0268-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
The clinical application of ex vivo gene edited cell therapies first began a decade ago with zinc finger nuclease editing of autologous CD4+ T-cells. Editing aimed to disrupt expression of the human immunodeficiency virus co-receptor gene CCR5, with the goal of yielding cells resistant to viral entry, prior to re-infusion into the patient. Since then the field has substantially evolved with the arrival of the new editing technologies transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR), and the potential benefits of gene editing in the arenas of immuno-oncology and blood disorders were quickly recognised. As the breadth of cell therapies available clinically continues to rise there is growing interest in allogeneic and off-the-shelf approaches and multiplex editing strategies are increasingly employed. We review here the latest clinical trials utilising these editing technologies and consider the applications on the horizon.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapy and Cancer Group, Dept of Imaging Chemistry & Biology, School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK
- Centre for Stem Cells & Regenerative Medicine, School of Basic and Medical Biosciences, Guy's Hospital, KCL, London, SE1 9RT, UK
| | - Gilbert O Fruhwirth
- Imaging Therapy and Cancer Group, Dept of Imaging Chemistry & Biology, School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK.
| |
Collapse
|
21
|
|
22
|
Ashmore-Harris C, Blackford SJ, Grimsdell B, Kurtys E, Glatz MC, Rashid TS, Fruhwirth GO. Reporter gene-engineering of human induced pluripotent stem cells during differentiation renders in vivo traceable hepatocyte-like cells accessible. Stem Cell Res 2019; 41:101599. [PMID: 31707210 PMCID: PMC6905152 DOI: 10.1016/j.scr.2019.101599] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/15/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
Primary hepatocyte transplantation (HTx) is a safe cell therapy for patients with liver disease, but wider application is circumvented by poor cell engraftment due to limitations in hepatocyte quality and transplantation strategies. Hepatocyte-like cells (HLCs) derived from human induced pluripotent stem cells (hiPSC) are considered a promising alternative but also require optimisation of transplantation and are often transplanted prior to full maturation. Whole-body in vivo imaging would be highly beneficial to assess engraftment non-invasively and monitor the transplanted cells in the short and long-term. Here we report a lentiviral transduction approach designed to engineer hiPSC-derived HLCs during differentiation. This strategy resulted in the successful production of sodium iodide symporter (NIS)-expressing HLCs that were functionally characterised, transplanted into mice, and subsequently imaged using radionuclide tomography.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK; Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Guy's Hospital, KCL, London SE1 9RT, UK
| | - Samuel Ji Blackford
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Guy's Hospital, KCL, London SE1 9RT, UK
| | - Benjamin Grimsdell
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK; Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Shepherd's House, King's College London, SE1 1UL, UK
| | - Ewelina Kurtys
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK
| | - Marlies C Glatz
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK
| | - Tamir S Rashid
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Guy's Hospital, KCL, London SE1 9RT, UK; Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Gilbert O Fruhwirth
- Imaging Therapy and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK.
| |
Collapse
|
23
|
Fiordelisi MF, Cavaliere C, Auletta L, Basso L, Salvatore M. Magnetic Resonance Imaging for Translational Research in Oncology. J Clin Med 2019; 8:jcm8111883. [PMID: 31698697 PMCID: PMC6912299 DOI: 10.3390/jcm8111883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The translation of results from the preclinical to the clinical setting is often anything other than straightforward. Indeed, ideas and even very intriguing results obtained at all levels of preclinical research, i.e., in vitro, on animal models, or even in clinical trials, often require much effort to validate, and sometimes, even useful data are lost or are demonstrated to be inapplicable in the clinic. In vivo, small-animal, preclinical imaging uses almost the same technologies in terms of hardware and software settings as for human patients, and hence, might result in a more rapid translation. In this perspective, magnetic resonance imaging might be the most translatable technique, since only in rare cases does it require the use of contrast agents, and when not, sequences developed in the lab can be readily applied to patients, thanks to their non-invasiveness. The wide range of sequences can give much useful information on the anatomy and pathophysiology of oncologic lesions in different body districts. This review aims to underline the versatility of this imaging technique and its various approaches, reporting the latest preclinical studies on thyroid, breast, and prostate cancers, both on small laboratory animals and on human patients, according to our previous and ongoing research lines.
Collapse
|
24
|
Tracers for non-invasive radionuclide imaging of immune checkpoint expression in cancer. EJNMMI Radiopharm Chem 2019; 4:29. [PMID: 31696402 PMCID: PMC6834817 DOI: 10.1186/s41181-019-0078-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/21/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract Immunotherapy with checkpoint inhibitors demonstrates impressive improvements in the treatment of several types of cancer. Unfortunately, not all patients respond to therapy while severe immune-related adverse effects are prevalent. Currently, patient stratification is based on immunotherapy marker expression through immunohistochemical analysis on biopsied material. However, expression can be heterogeneous within and between tumor lesions, amplifying the sampling limitations of biopsies. Analysis of immunotherapy target expression by non-invasive quantitative molecular imaging with PET or SPECT may overcome this issue. In this review, an overview of tracers that have been developed for preclinical and clinical imaging of key immunotherapy targets, such as programmed cell death-1, programmed cell death ligand-1, IDO1 and cytotoxic T lymphocyte-associated antigen-4 is presented. We discuss important aspects to consider when developing such tracers and outline the future perspectives of molecular imaging of immunotherapy markers. Graphical abstract Current techniques in immune checkpoint imaging and its potential for future applications ![]()
Collapse
|
25
|
Krekorian M, Fruhwirth GO, Srinivas M, Figdor CG, Heskamp S, Witney TH, Aarntzen EHJG. Imaging of T-cells and their responses during anti-cancer immunotherapy. Theranostics 2019; 9:7924-7947. [PMID: 31656546 PMCID: PMC6814447 DOI: 10.7150/thno.37924] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has proven to be an effective approach in a growing number of cancers. Despite durable clinical responses achieved with antibodies targeting immune checkpoint molecules, many patients do not respond. The common denominator for immunotherapies that have successfully been introduced in the clinic is their potential to induce or enhance infiltration of cytotoxic T-cells into the tumour. However, in clinical research the molecules, cells and processes involved in effective responses during immunotherapy remain largely obscure. Therefore, in vivo imaging technologies that interrogate T-cell responses in patients represent a powerful tool to boost further development of immunotherapy. This review comprises a comprehensive analysis of the in vivo imaging technologies that allow the characterisation of T-cell responses induced by anti-cancer immunotherapy, with emphasis on technologies that are clinically available or have high translational potential. Throughout we discuss their respective strengths and weaknesses, providing arguments for selecting the optimal imaging options for future research and patient management.
Collapse
Affiliation(s)
- Massis Krekorian
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Gilbert O Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Timothy H Witney
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Erik H J G Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Preclinical Molecular Imaging for Precision Medicine in Breast Cancer Mouse Models. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:8946729. [PMID: 31598114 PMCID: PMC6778915 DOI: 10.1155/2019/8946729] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/28/2019] [Accepted: 07/25/2019] [Indexed: 12/18/2022]
Abstract
Precision and personalized medicine is gaining importance in modern clinical medicine, as it aims to improve diagnostic precision and to reduce consequent therapeutic failures. In this regard, prior to use in human trials, animal models can help evaluate novel imaging approaches and therapeutic strategies and can help discover new biomarkers. Breast cancer is the most common malignancy in women worldwide, accounting for 25% of cases of all cancers and is responsible for approximately 500,000 deaths per year. Thus, it is important to identify accurate biomarkers for precise stratification of affected patients and for early detection of responsiveness to the selected therapeutic protocol. This review aims to summarize the latest advancements in preclinical molecular imaging in breast cancer mouse models. Positron emission tomography (PET) imaging remains one of the most common preclinical techniques used to evaluate biomarker expression in vivo, whereas magnetic resonance imaging (MRI), particularly diffusion-weighted (DW) sequences, has been demonstrated as capable of distinguishing responders from nonresponders for both conventional and innovative chemo- and immune-therapies with high sensitivity and in a noninvasive manner. The ability to customize therapies is desirable, as this will enable early detection of diseases and tailoring of treatments to individual patient profiles. Animal models remain irreplaceable in the effort to understand the molecular mechanisms and patterns of oncologic diseases.
Collapse
|
27
|
Castello A, Lopci E. Response assessment of bone metastatic disease: seeing the forest for the trees RECIST, PERCIST, iRECIST, and PCWG-2. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:150-158. [PMID: 31286751 DOI: 10.23736/s1824-4785.19.03193-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor response is often used as a surrogate marker for survival practically in all clinical trials. Therefore, robust and valid response criteria during the course of trials are fundamental for the assessment of response to therapy. This aspect, however, becomes particularly challenging when it comes to bone metastases. In the era of targeted therapies and immune-checkpoint inhibitors (ICI), response assessment by morphologic-based criteria cannot detect the real tumor response and, consequently, fail to demonstrate the actual clinical benefit. This review will focus on some of the most common morphologic and metabolic response criteria and their application for bone lesions, highlighting relative strengths and weaknesses as well as potential future methods in the era of target therapies and immunotherapy with ICI.
Collapse
Affiliation(s)
- Angelo Castello
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy -
| |
Collapse
|
28
|
The beginning of the end for conventional RECIST - novel therapies require novel imaging approaches. Nat Rev Clin Oncol 2019; 16:442-458. [PMID: 30718844 DOI: 10.1038/s41571-019-0169-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Owing to improvements in our understanding of the biological principles of tumour initiation and progression, a wide variety of novel targeted therapies have been developed. Developments in biomedical imaging, however, have not kept pace with these improvements and are still mainly designed to determine lesion size alone, which is reflected in the Response Evaluation Criteria in Solid Tumors (RECIST). Imaging approaches currently used for the evaluation of treatment responses in patients with solid tumours, therefore, often fail to detect successful responses to novel targeted agents and might even falsely suggest disease progression, a scenario known as pseudoprogression. The ability to differentiate between responders and nonresponders early in the course of treatment is essential to allowing the early adjustment of treatment regimens. Various imaging approaches targeting a single dedicated tumour feature, as described in the hallmarks of cancer, have been successful in preclinical investigations, and some have been evaluated in pilot clinical trials. However, these approaches have largely not been implemented in clinical practice. In this Review, we describe current biomedical imaging approaches used to monitor responses to treatment in patients receiving novel targeted therapies, including a summary of the most promising future approaches and how these might improve clinical practice.
Collapse
|