1
|
Zhang D, Wang H, Chen C, Lu G, Yin Y, Ren M, Huang J. Preparation and identification of a fluorescent probe with CsPbBr 3perovskite quantum dots and CD44v6 specific peptide for gastric cancer imaging. NANOTECHNOLOGY 2024; 36:02LT02. [PMID: 39406258 DOI: 10.1088/1361-6528/ad86c7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Since the sensitivity and accuracy of traditional detection for early gastric cancer diagnosis are still insufficient, it is significant to continuously optimize the optical molecular imaging detection technology based on an endoscopic platform. The signal intensity and stability of traditional chemical fluorescent dyes are low, which hinders the clinical application of molecular imaging detection technology. This work developed a probe based on perovskite quantum dots (PQDs) and peptide ligands. By utilizing CsPbBr3perovskite PQDs modified by azithromycin (AZI), combined with the specific polypeptide ligand of CD44v6, a gastric cancer biomarker, the perovskite-based probe (AZI-PQDs probe) which can specifically identify gastric cancer tumor was prepared. Owing to the high photoluminescence quantum yield of CsPbBr3PQDs, the naked eye can observe the imaging under the excitation of the hand-held ultraviolet light source. AZI-PQDs probe can accurately identify gastric cancer cells, tissues, and xenograft models with experiments ofex vivoandin vivofluorescence imaging detection. It also exhibited low toxicity and immunogenicity, indicating the safety of the probe. This work provides a probe combined with cancer specificity and a reliable fluorescent signal that has the potential for application in gastric cancer optical molecular imaging.
Collapse
Affiliation(s)
- Dan Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Hao Wang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
- Shool of Electronic Information and Artificial Intelligence, Shaanxi University of Science &Technology, Xi'an 710021, People's Republic of China
| | - Chunyang Chen
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
- Shool of Electronic Information and Artificial Intelligence, Shaanxi University of Science &Technology, Xi'an 710021, People's Republic of China
| | - Guifang Lu
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Yan Yin
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Mudan Ren
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Jin Huang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
- Shool of Electronic Information and Artificial Intelligence, Shaanxi University of Science &Technology, Xi'an 710021, People's Republic of China
| |
Collapse
|
2
|
Ribéraud M, Porret E, Pruvost A, Theodoro F, Nguyen AL, Specklin S, Kereselidze D, Denis C, Jego B, Barbe P, Keck M, D'Anfray T, Kuhnast B, Audisio D, Truillet C, Taran F. A cancer immunoprofiling strategy using mass spectrometry coupled with bioorthogonal cleavage. Chem Sci 2024:d4sc04471a. [PMID: 39464609 PMCID: PMC11499955 DOI: 10.1039/d4sc04471a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
The accurate quantification of biomarkers is paramount in modern medicine, particularly in cancer where precise diagnosis is imperative for targeted therapy selection. In this paper we described a multiplexed analysis diagnostic approach based on cleavable MS-tagged antibodies. The technology uses MS-tag isotopologues and the sydnonimine-cyclooctyne click-and-release bioorthogonal reaction. In a proof of concept study, we demonstrated the potential of this approach for cancer cell immunoprofiling in culture cells, tissues and in vivo as well, thereby unveiling promising diagnostic avenues.
Collapse
Affiliation(s)
- Maxime Ribéraud
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Estelle Porret
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | - Alain Pruvost
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Frédéric Theodoro
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Anvi Laëtitia Nguyen
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Simon Specklin
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | | | - Caroline Denis
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | - Benoit Jego
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | - Peggy Barbe
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Mathilde Keck
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Timothée D'Anfray
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | | | - Davide Audisio
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | | | - Frédéric Taran
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| |
Collapse
|
3
|
Jirvankar P, Agrawal S, Chambhare N, Agrawal R. Harnessing Biopolymer Gels for Theranostic Applications: Imaging Agent Integration and Real-Time Monitoring of Drug Delivery. Gels 2024; 10:535. [PMID: 39195064 DOI: 10.3390/gels10080535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Biopolymer gels have gained tremendous potential for therapeutic applications due to their biocompatibility, biodegradability, and ability to adsorb and bind biological fluids, making them attractive for drug delivery and therapy. In this study, the versatility of biopolymer gels is explored in theranostic backgrounds, with a focus on integrating imaging features and facilitating real-time monitoring of drug delivery. Different methods of delivery are explored for incorporating imaging agents into biopolymer gels, including encapsulation, surface functionalization, nanoparticle encapsulation, and layer-by-layer assembly techniques. These methods exhibit the integration of agents and real-time monitoring drug delivery. We summarize the synthesis methods, general properties, and functional mechanisms of biopolymer gels, demonstrating their broad applications as multimodal systems for imaging-based therapeutics. These techniques not only enable multiple imaging but also provide signal enhancement and facilitate imaging targets, increasing the diagnostic accuracy and therapeutic efficacy. In addition, current techniques for incorporating imaging agents into biopolymer gels are discussed, as well as their role in precise drug delivery and monitoring.
Collapse
Affiliation(s)
- Pranita Jirvankar
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Wardha 442001, Maharashtra, India
| | - Surendra Agrawal
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Wardha 442001, Maharashtra, India
| | - Nikhita Chambhare
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Wardha 442001, Maharashtra, India
| | - Rishabh Agrawal
- Bajiraoji Karanjekar College of Pharmacy, Sakoli 441802, Maharashtra, India
| |
Collapse
|
4
|
Keshari KR, Heller DA, Boltyanskiy R, Hricak H, Magaldi T, Overholtzer M. Engineering focusing on cancer. Cancer Cell 2024; 42:1138-1141. [PMID: 38848719 DOI: 10.1016/j.ccell.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 06/09/2024]
Abstract
While cancer research and care have benefited from revolutionary advances in the ability to manipulate and study living systems, the field is limited by a lack of synergy to leverage the power of engineering approaches. Cancer engineering is an emerging subfield of biomedical engineering that unifies engineering and cancer biology to better understand, diagnose, and treat cancer. We highlight cancer engineering's unique challenges, the importance of creating dedicated centers and departments that enable translational collaboration, and educational approaches to arm a new generation of scientists with engineering expertise and a fundamental understanding of cancer biology to transform clinical cancer care.
Collapse
Affiliation(s)
- Kayvan R Keshari
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Molecular Imaging and Bioengineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Daniel A Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Molecular Imaging and Bioengineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Rostislav Boltyanskiy
- Center for Molecular Imaging and Bioengineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hedvig Hricak
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Molecular Imaging and Bioengineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Magaldi
- Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Molecular Imaging and Bioengineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
5
|
Zhang Y, Song X, Xu Z, Lv X, Long Y, Lan X, Lei P. Construction of truncated PSMA as a PET reporter gene for CAR T cell trafficking. J Leukoc Biol 2024; 115:476-482. [PMID: 37943840 DOI: 10.1093/jleuko/qiad127] [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: 04/12/2023] [Revised: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 11/12/2023] Open
Abstract
In solid tumors, there are multiple barriers for a chimeric antigen receptor (CAR) T cell to surmount in order to reach the tumor site. For better understanding whether CAR T cells effectively infiltrate into tumor site, and simultaneously, whether there are off-target effects, real-time monitoring technologies need to be established. Cell-based positron emission tomography reporter genes have been developed to monitor engineered cells in living subjects. In this study, we reported the construction of a novel reporter gene truncated prostate-specific membrane antigen (ΔPSMA) pending for monitoring CAR T cells using 68Ga-PSMA-617 and a method for tracking the distribution of CAR T cells in vivo was developed. Data were provided to demonstrate that ΔPSMA was predominantly localized on the plasma membrane and could take up 68Ga-PSMA-617 in vitro in a time-dependent manner. And the expression of ΔPSMA did not affect CAR expression and cytolytic capacity of CAR T cells. CAR-ΔPSMA T cell xenografts in nude mice were clearly imaged by positron emission tomography 60 min after injection of 68Ga-PSMA-617. PSMA paired with 68Ga-PSMA-617 was capable of identifying approximately 1 × 104 engineered CAR T cells. The ability to image small numbers of CAR T cells in vivo would be helpful to accelerate the translation of cell-based therapies into the clinic, and it may reinforce our understanding of treatment success, failure, and toxicity.
Collapse
Affiliation(s)
- Yirui Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, Hubei, 430030, China
| | - Xiangming Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
- Hubei Province Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
| | - Zhuoshuo Xu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, Hubei, 430030, China
| | - Xiaoying Lv
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
- Hubei Province Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
- Hubei Province Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
- Hubei Province Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan 430022, Hubei Province, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, Hangkong Road, Wuhan, Hubei, 430030, China
| |
Collapse
|
6
|
Kiessling F, Schulz V. Perspectives of Evidence-Based Therapy Management. Nuklearmedizin 2023; 62:314-322. [PMID: 37802059 DOI: 10.1055/a-2159-6949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
BACKGROUND Therapeutics that specifically address biological processes often require a much finer selection of patients and subclassification of diseases. Thus, diagnostic procedures must describe the diseases in sufficient detail to allow selection of appropriate therapy and to sensitively track therapy response. Anatomical features are often not sufficient for this purpose and there is a need to image molecular and pathophysiological processes. METHOD Two imaging strategies can be pursued: molecular imaging attempts to image a few biomarkers that play key roles in pathological processes. Alternatively, patterns describing a biological process can be identified from the synopsis of multiple (non-specific) imaging markers, possibly in combination with omics and other clinical findings. Here, AI-based methods are increasingly being used. RESULTS Both strategies of evidence-based therapy management are explained in this review article and examples and clinical successes are presented. In this context, reviews of clinically approved molecular diagnostics and decision support systems are listed. Furthermore, since reliable, representative, and sufficiently large datasets are further important prerequisites for AI-assisted multiparametric analyses, concepts are presented to make data available in a structured way, e. g., using Generative Adversarial Networks to complement databases with virtual cases and to build completely anonymous reference databases. CONCLUSION Molecular imaging and computer-assisted cluster analysis of diagnostic data are complementary methods to describe pathophysiological processes. Both methods have the potential to improve (evidence-based) the future management of therapies, partly on their own but also in combined approaches. KEY POINTS · Molecular imaging and radiomics provide valuable complementary disease biomarkers.. · Data-driven, model-based, and hybrid model-based integrated diagnostics advance precision medicine.. · Synthetic data generation may become essential in the development process of future AI methods..
Collapse
Affiliation(s)
- Fabian Kiessling
- Universitätsklinikum Aachen, Lehrstuhl für Experimentelle Molekulare Bildgebung, Aachen, Germany
- Group Aachen, Fraunhofer-Institut für Digitale Medizin MEVIS, Bremen, Germany
| | - Volkmar Schulz
- Universitätsklinikum Aachen, Lehrstuhl für Experimentelle Molekulare Bildgebung, Aachen, Germany
- Group Aachen, Fraunhofer-Institut für Digitale Medizin MEVIS, Bremen, Germany
| |
Collapse
|
7
|
Pan S, Ding A, Li Y, Sun Y, Zhan Y, Ye Z, Song N, Peng B, Li L, Huang W, Shao H. Small-molecule probes from bench to bedside: advancing molecular analysis of drug-target interactions toward precision medicine. Chem Soc Rev 2023; 52:5706-5743. [PMID: 37525607 DOI: 10.1039/d3cs00056g] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Over the past decade, remarkable advances have been witnessed in the development of small-molecule probes. These molecular tools have been widely applied for interrogating proteins, pathways and drug-target interactions in preclinical research. While novel structures and designs are commonly explored in probe development, the clinical translation of small-molecule probes remains limited, primarily due to safety and regulatory considerations. Recent synergistic developments - interfacing novel chemical probes with complementary analytical technologies - have introduced and expedited diverse biomedical opportunities to molecularly characterize targeted drug interactions directly in the human body or through accessible clinical specimens (e.g., blood and ascites fluid). These integrated developments thus offer unprecedented opportunities for drug development, disease diagnostics and treatment monitoring. In this review, we discuss recent advances in the structure and design of small-molecule probes with novel functionalities and the integrated development with imaging, proteomics and other emerging technologies. We further highlight recent applications of integrated small-molecule technologies for the molecular analysis of drug-target interactions, including translational applications and emerging opportunities for whole-body imaging, tissue-based measurement and blood-based analysis.
Collapse
Affiliation(s)
- Sijun Pan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yisi Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yaxin Sun
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yueqin Zhan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Zhenkun Ye
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Ning Song
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Wei Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore 117599, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
| |
Collapse
|
8
|
Schwenck J, Sonanini D, Cotton JM, Rammensee HG, la Fougère C, Zender L, Pichler BJ. Advances in PET imaging of cancer. Nat Rev Cancer 2023:10.1038/s41568-023-00576-4. [PMID: 37258875 DOI: 10.1038/s41568-023-00576-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
Molecular imaging has experienced enormous advancements in the areas of imaging technology, imaging probe and contrast development, and data quality, as well as machine learning-based data analysis. Positron emission tomography (PET) and its combination with computed tomography (CT) or magnetic resonance imaging (MRI) as a multimodality PET-CT or PET-MRI system offer a wealth of molecular, functional and morphological data with a single patient scan. Despite the recent technical advances and the availability of dozens of disease-specific contrast and imaging probes, only a few parameters, such as tumour size or the mean tracer uptake, are used for the evaluation of images in clinical practice. Multiparametric in vivo imaging data not only are highly quantitative but also can provide invaluable information about pathophysiology, receptor expression, metabolism, or morphological and functional features of tumours, such as pH, oxygenation or tissue density, as well as pharmacodynamic properties of drugs, to measure drug response with a contrast agent. It can further quantitatively map and spatially resolve the intertumoural and intratumoural heterogeneity, providing insights into tumour vulnerabilities for target-specific therapeutic interventions. Failure to exploit and integrate the full potential of such powerful imaging data may lead to a lost opportunity in which patients do not receive the best possible care. With the desire to implement personalized medicine in the cancer clinic, the full comprehensive diagnostic power of multiplexed imaging should be utilized.
Collapse
Affiliation(s)
- Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonathan M Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Department of Immunology, IFIZ Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany.
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
9
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Sun R, Zhang Y, Gao Y, Zhao M, Wang A, Zhu J, Cheng X, Shi H. A tumor-targetable NIR probe with photoaffinity crosslinking characteristics for enhanced imaging-guided cancer phototherapy. Chem Sci 2023; 14:2369-2378. [PMID: 36873836 PMCID: PMC9977396 DOI: 10.1039/d2sc06413h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/03/2023] [Indexed: 02/05/2023] Open
Abstract
Spatiotemporally manipulating the in situ immobilization of theranostic agents within cancer cells to improve their bioavailability is highly significant yet challenging in tumor diagnosis and treatment. Herein, as a proof-of concept, we for the first time report a tumor-targetable near-infrared (NIR) probe DACF with photoaffinity crosslinking characteristics for enhanced tumor imaging and therapeutic applications. This probe possesses great tumor-targeting capability, intensive NIR/photoacoustic (PA) signals, and a predominant photothermal effect, allowing for sensitive imaging and effective photothermal therapy (PTT) of tumors. Most notably, upon 405 nm laser illumination, DACF could be covalently immobilized within tumor cells through a photocrosslinking reaction between photolabile diazirine groups and surrounding biomolecules resulting in enhanced tumor accumulation and prolonged retention simultaneously, which significantly facilitates the imaging and PTT efficacy of tumor in vivo. We therefore believe that our current approach would provide a new insight for achieving precise cancer theranostics.
Collapse
Affiliation(s)
- Rui Sun
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Yinjia Gao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Meng Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Jinfeng Zhu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Xiaju Cheng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University Suzhou 215123 P. R. China
| |
Collapse
|
11
|
Mishra A, Kumar D, Gupta K, Lofland G, Sharma AK, Banka DS, Hobbs RF, Dannals RF, Rowe SP, Gabrielson E, Nimmagadda S. Gallium-68-labeled Peptide PET Quantifies Tumor Exposure of PD-L1 Therapeutics. Clin Cancer Res 2023; 29:581-591. [PMID: 36449662 PMCID: PMC9890130 DOI: 10.1158/1078-0432.ccr-22-1931] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/06/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022]
Abstract
PURPOSE Immune checkpoint therapy (ICT) is currently ineffective in a majority of patients. Tumor drug exposure measurements can provide vital insights into mechanisms involved in the resistance of solid tumors to those therapeutics; however, tools to quantify in situ drug exposure are few. We have investigated the potential of programmed death-ligand 1 (PD-L1) pharmacodynamics, quantified using PET, to inform on the tumor exposure of anti-PD-L1 (aPD-L1) therapeutics. EXPERIMENTAL DESIGN To noninvasively quantify PD-L1 levels, we first developed a novel peptide-based gallium-68-labeled binder, [68Ga]Ga-DK223, and evaluated its in vivo distribution, pharmacokinetics, and PD-L1 specificity in preclinical models of triple-negative breast cancer and urothelial carcinoma with variable PD-L1 expression. We then quantified baseline and accessible PD-L1 levels in tumors as a noninvasive pharmacodynamic measure to assess tumor exposure to two aPD-L1 antibodies (avelumab and durvalumab). RESULTS DK223 exhibited a KD of 1.01±0.83 nmol/L for PD-L1 and inhibited the PD-1:PD-L1 interaction in a dose-dependent manner. [68Ga]Ga-DK223 provides high-contrast PET images within 60 minutes of administration and detects PD-L1 in an expression-dependent manner in xenograft models. PD-L1 pharmacodynamics measured using [68Ga]Ga-DK223-PET revealed that avelumab and durvalumab had similar exposure early during therapy, but only durvalumab exhibited sustained exposure at the tumor. CONCLUSIONS [68Ga]Ga-DK223 detected variable PD-L1 levels and exhibited salient features required for clinical translation. [68Ga]Ga-DK223-PET could be useful for quantifying total PD-L1 levels at baseline and accessible PD-L1 levels during therapy to understand drug exposure at the tumor, thus supporting its use for guiding and optimizing ICT.
Collapse
Affiliation(s)
- Akhilesh Mishra
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Chemical & Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Dhiraj Kumar
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kuldeep Gupta
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gabriela Lofland
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ajay Kumar Sharma
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dhanush S. Banka
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F. Hobbs
- Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F. Dannals
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg–Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg–Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Corresponding Author: Sridhar Nimmagadda, Johns Hopkins Medical Institutions, 1550 Orleans Street, CRB II, #492, Baltimore, MD 21287. Phone: 410-502-6244, Fax: 410-614-3147, E-mail:
| |
Collapse
|
12
|
Miyazaki T, Chen S, Florinas S, Igarashi K, Matsumoto Y, Yamasoba T, Xu ZQ, Wu H, Gao C, Kataoka K, Christie RJ, Cabral H. A Hoechst Reporter Enables Visualization of Drug Engagement In Vitro and In Vivo: Toward Safe and Effective Nanodrug Delivery. ACS NANO 2022; 16:12290-12304. [PMID: 35942986 DOI: 10.1021/acsnano.2c03170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Assessment of drug activation and subsequent interaction with targets in living tissues could guide nanomedicine design, but technologies enabling insight into how a drug reaches and binds its target are limited. We show that a Hoechst-based reporter system can monitor drug release and engagement from a nanoparticle delivery system in vitro and in vivo, elucidating differences in target-bound drug distribution related to drug-linker and nanoparticle properties. Drug engagement is defined as chemical detachment of drug or reporter from a nanoparticle and subsequent binding to a subcellular target, which in the case of Hoechst results in a fluorescence signal. Hoechst-based nanoreporters for drug activation contain prodrug elements such as dipeptide linkers, conjugation handles, and nanoparticle modifications such as targeting ligands to determine how nanomedicine design affects distribution of drug engaged with a subcellular target, which is tracked via cellular nuclear fluorescence in situ. Furthermore, the nanoplatform is amenable toward common maleimide-based linkers found in many prodrug-based delivery systems including polymer-, peptide-, and antibody-drug conjugates. Findings from the Hoechst reporter system were applied to develop highly potent, targeted, anticancer micelle nanoparticles delivering a monomethyl auristatin E (MMAE) prodrug comprising the same linkers employed in Hoechst studies. MMAE nanomedicine with the optimal drug-linker resulted in effective tumor growth inhibition in mice without associated acute toxicity, whereas the nonoptimal linker that showed broader drug activation in Hoechst reporter studies resulted in severe toxicity. Our results demonstrate the potential to synergize direct visualization of drug engagement with nanomedicine drug-linker design to optimize safety and efficacy.
Collapse
Affiliation(s)
- Takuya Miyazaki
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), 705-1 Shimoimaizumi, Ebina, Kanagawa 243-0435, Japan
| | - Shaoyi Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Stelios Florinas
- Antibody Discovery and Protein Engineering, AstraZeneca R&D, 1 MedImmune Way, Gaithersburg, Maryland 20878, United States
| | - Kazunori Igarashi
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yu Matsumoto
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ze-Qi Xu
- SynChem, Inc., Elk Grove Village, Illinois 60007, United States
| | - Herren Wu
- Antibody Discovery and Protein Engineering, AstraZeneca R&D, 1 MedImmune Way, Gaithersburg, Maryland 20878, United States
| | - Changshou Gao
- Antibody Discovery and Protein Engineering, AstraZeneca R&D, 1 MedImmune Way, Gaithersburg, Maryland 20878, United States
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - R James Christie
- Antibody Discovery and Protein Engineering, AstraZeneca R&D, 1 MedImmune Way, Gaithersburg, Maryland 20878, United States
- Biologics Engineering, AstraZeneca Oncology R&D, 1 MedImmune Way, Gaithersburg, Maryland 20878, United States
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
13
|
Optimized 68Ga-Labeled Urea-Based PSMA-Targeted PET Tracers for Prostate Cancer. Pharmaceuticals (Basel) 2022; 15:ph15081001. [PMID: 36015149 PMCID: PMC9414910 DOI: 10.3390/ph15081001] [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: 05/31/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeting radiopharmaceuticals have become some of the most promising tools for the diagnosis and therapy prostate cancer (PCa). The structure of existing PSMA-targeted PET tracers still needs to be optimized to improve their pharmacokinetic properties and tumor-to-background ratio. In this study, we modified the structure of a well-studied PSMA tracer, and six novel tracers with variable hydrophilicity and pharmacokinetics were developed and evaluated both in vitro and in vivo. All of the novel tracers showed high hydrophilicity (log p = −2.99 ± 0.33 to −3.49 ± 0.01), rapid clearance rates (elimination half-times = 15.55 to 35.97 min), and high affinity for PSMA (Ki = 8.11 ± 0.49 to 42.40 ± 2.11 nM) in vitro. Specific cell binding and micro-PET experiments showed that [68Ga]Ga-PSMA-Q displayed the highest specific PSMA+ cell uptake (3.75 ± 0.35 IA%/106 at 60 min), tumor uptake (SUVmax = 0.97 ± 0.24 at 60 min p.i.), and tumor-to-muscle ratio (59.33 ± 5.72 at 60 min p.i.), while the tumor-to-muscle ratio was much higher than that of [68Ga]Ga-PSMA-617. The results of this study validate the clinical potential of [68Ga]Ga-PSMA-Q for PET imaging and further targeted therapy of prostate cancer.
Collapse
|
14
|
Chin LK, Li H, Choi JH, Iwamoto Y, Oh J, Min J, Beak SK, Yoo D, Castro CM, Lee D, Im H. Hydrogel Stamping for Rapid, Multiplexed, Point-of-Care Immunostaining of Cells and Tissues. ACS APPLIED MATERIALS & INTERFACES 2022; 14:27613-27622. [PMID: 35671240 DOI: 10.1021/acsami.2c05071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In the era of precision oncology, multicolor fluorescence imaging has become a core technology for multiplexed molecular analysis of cellular and tissue specimens. However, conventional solution-based staining is labor-intensive and time-consuming and requires considerable expertise to yield optimal results, which creates difficulties for employing this technology in resource-limited settings. Here, we report a new immunostaining method based on hydrogel stamping, which is simple, fast, easy to use, and reproducible. We showed that a hydrophilic hydrogel stamp could effectively transfer fluorescent antibodies to targets and withdraw an excess solution when the reaction is completed, obviating the need for extra washing. This unique property allows for quality immunostaining in 5 min for cells using one-eighth of antibody consumption compared to the conventional solution-based method. Furthermore, we implemented fluorescence quenching and immunocycling with hydrogel staining for multiplexed analysis of 9 protein markers at a single cell level. Finally, we applied the immunocycling method to human breast cancer tissue samples and showed quality immunostaining over a large area (∼2 cm2) in 30 min for molecular subtyping of breast cancer. The hydrogel immunostaining could open new opportunities for rapid, automated, and multiplexed profiling in compact point-of-care systems for molecular cancer diagnosis.
Collapse
Affiliation(s)
- Lip Ket Chin
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department of Electrical Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR
| | - Huiyan Li
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- School of Engineering, University of Guelph, 50 Stone Road East, Guelph N1G2W1, Canada
| | - Jae-Hyeok Choi
- Noul Co. Limited, Gyeonggi-do, Yongin 16942, Republic of Korea
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Juhyun Oh
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Jouha Min
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Suk Kyung Beak
- Noul Co. Limited, Gyeonggi-do, Yongin 16942, Republic of Korea
| | - Dahyeon Yoo
- Noul Co. Limited, Gyeonggi-do, Yongin 16942, Republic of Korea
| | - Cesar M Castro
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Dongyoung Lee
- Noul Co. Limited, Gyeonggi-do, Yongin 16942, Republic of Korea
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| |
Collapse
|
15
|
Kiessling F, Schulz V. Perspectives of Evidence-Based Therapy Management. ROFO-FORTSCHR RONTG 2022; 194:728-736. [PMID: 35545101 DOI: 10.1055/a-1752-0839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Therapeutics that specifically address biological processes often require a much finer selection of patients and subclassification of diseases. Thus, diagnostic procedures must describe the diseases in sufficient detail to allow selection of appropriate therapy and to sensitively track therapy response. Anatomical features are often not sufficient for this purpose and there is a need to image molecular and pathophysiological processes. METHOD Two imaging strategies can be pursued: molecular imaging attempts to image a few biomarkers that play key roles in pathological processes. Alternatively, patterns describing a biological process can be identified from the synopsis of multiple (non-specific) imaging markers, possibly in combination with omics and other clinical findings. Here, AI-based methods are increasingly being used. RESULTS Both strategies of evidence-based therapy management are explained in this review article and examples and clinical successes are presented. In this context, reviews of clinically approved molecular diagnostics and decision support systems are listed. Furthermore, since reliable, representative, and sufficiently large datasets are further important prerequisites for AI-assisted multiparametric analyses, concepts are presented to make data available in a structured way, e. g., using Generative Adversarial Networks to complement databases with virtual cases and to build completely anonymous reference databases. CONCLUSION Molecular imaging and computer-assisted cluster analysis of diagnostic data are complementary methods to describe pathophysiological processes. Both methods have the potential to improve (evidence-based) the future management of therapies, partly on their own but also in combined approaches. KEY POINTS · Molecular imaging and radiomics provide valuable complementary disease biomarkers.. · Data-driven, model-based, and hybrid model-based integrated diagnostics advance precision medicine.. · Synthetic data generation may become essential in the development process of future AI methods.. CITATION FORMAT · Kiessling F, Schulz V, . Perspectives of Evidence-Based Therapy Management. Fortschr Röntgenstr 2022; DOI: 10.1055/a-1752-0839.
Collapse
Affiliation(s)
- Fabian Kiessling
- Universitätsklinikum Aachen, Lehrstuhl für Experimentelle Molekulare Bildgebung, Aachen, Germany.,Group Aachen, Fraunhofer-Institut für Digitale Medizin MEVIS, Bremen, Germany
| | - Volkmar Schulz
- Universitätsklinikum Aachen, Lehrstuhl für Experimentelle Molekulare Bildgebung, Aachen, Germany.,Group Aachen, Fraunhofer-Institut für Digitale Medizin MEVIS, Bremen, Germany
| |
Collapse
|
16
|
Andreou C, Weissleder R, Kircher MF. Multiplexed imaging in oncology. Nat Biomed Eng 2022; 6:527-540. [PMID: 35624151 DOI: 10.1038/s41551-022-00891-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/06/2021] [Indexed: 01/24/2023]
Abstract
In oncology, technologies for clinical molecular imaging are used to diagnose patients, establish the efficacy of treatments and monitor the recurrence of disease. Multiplexed methods increase the number of disease-specific biomarkers that can be detected simultaneously, such as the overexpression of oncogenic proteins, aberrant metabolite uptake and anomalous blood perfusion. The quantitative localization of each biomarker could considerably increase the specificity and the accuracy of technologies for clinical molecular imaging to facilitate granular diagnoses, patient stratification and earlier assessments of the responses to administered therapeutics. In this Review, we discuss established techniques for multiplexed imaging and the most promising emerging multiplexing technologies applied to the imaging of isolated tissues and cells and to non-invasive whole-body imaging. We also highlight advances in radiology that have been made possible by multiplexed imaging.
Collapse
Affiliation(s)
- Chrysafis Andreou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Electrical and Computer Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Moritz F Kircher
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.,Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Du Y, Zhang P, Liu W, Tian J. Optical Imaging of Epigenetic Modifications in Cancer: A Systematic Review. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:88-101. [PMID: 36939779 PMCID: PMC9590553 DOI: 10.1007/s43657-021-00041-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/10/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023]
Abstract
Increasing evidence has demonstrated that abnormal epigenetic modifications are strongly related to cancer initiation. Thus, sensitive and specific detection of epigenetic modifications could markedly improve biological investigations and cancer precision medicine. A rapid development of molecular imaging approaches for the diagnosis and prognosis of cancer has been observed during the past few years. Various biomarkers unique to epigenetic modifications and targeted imaging probes have been characterized and used to discriminate cancer from healthy tissues, as well as evaluate therapeutic responses. In this study, we summarize the latest studies associated with optical molecular imaging of epigenetic modification targets, such as those involving DNA methylation, histone modification, noncoding RNA regulation, and chromosome remodeling, and further review their clinical application on cancer diagnosis and treatment. Lastly, we further propose the future directions for precision imaging of epigenetic modification in cancer. Supported by promising clinical and preclinical studies associated with optical molecular imaging technology and epigenetic drugs, the central role of epigenetics in cancer should be increasingly recognized and accepted.
Collapse
Affiliation(s)
- Yang Du
- grid.9227.e0000000119573309CAS Key Laboratory of Molecular Imaging, Beijing 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
- grid.410726.60000 0004 1797 8419The University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Pei Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Molecular Imaging, Beijing 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
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Supportive Care Center and Day Oncology Unit, Peking University Cancer Hospital and Institute, Beijing, 100142 China
| | - Wei Liu
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Supportive Care Center and Day Oncology Unit, Peking University Cancer Hospital and Institute, Beijing, 100142 China
| | - Jie Tian
- grid.9227.e0000000119573309CAS Key Laboratory of Molecular Imaging, Beijing 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
- grid.64939.310000 0000 9999 1211Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191 China
- grid.440736.20000 0001 0707 115XSchool of Life Science and Technology, Xidian University, Xi’an, 710071 Shaanxi China
| |
Collapse
|
18
|
Lu D, Yang T, Tang N, Li C, Song Y, Wang L, Wong WY, Yin SF, Xing Y, Kambe N, Qiu R. A pH-Dependent rhodamine fluorophore with antiproliferative activity of bladder cancer in Vitro/Vivo and apoptosis mechanism. Eur J Med Chem 2022; 236:114293. [DOI: 10.1016/j.ejmech.2022.114293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/04/2022] [Accepted: 03/13/2022] [Indexed: 02/04/2023]
|
19
|
Fundamentals and developments in fluorescence-guided cancer surgery. Nat Rev Clin Oncol 2022; 19:9-22. [PMID: 34493858 DOI: 10.1038/s41571-021-00548-3] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
Fluorescence-guided surgery using tumour-targeted imaging agents has emerged over the past decade as a promising and effective method of intraoperative cancer detection. An impressive number of fluorescently labelled antibodies, peptides, particles and other molecules related to cancer hallmarks have been developed for the illumination of target lesions. New approaches are being implemented to translate these imaging agents into the clinic, although only a few have made it past early-phase clinical trials. For this translational process to succeed, target selection, imaging agents and their related detection systems and clinical implementation have to operate in perfect harmony to enable real-time intraoperative visualization that can benefit patients. Herein, we review key aspects of this imaging cascade and focus on imaging approaches and methods that have helped to shed new light onto the field of intraoperative fluorescence-guided cancer surgery with the singular goal of improving patient outcomes.
Collapse
|
20
|
Pharmacodynamic measures within tumors expose differential activity of PD(L)-1 antibody therapeutics. Proc Natl Acad Sci U S A 2021; 118:2107982118. [PMID: 34508005 DOI: 10.1073/pnas.2107982118] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 01/22/2023] Open
Abstract
Macromolecules such as monoclonal antibodies (mAbs) are likely to experience poor tumor penetration because of their large size, and thus low drug exposure of target cells within a tumor could contribute to suboptimal responses. Given the challenge of inadequate quantitative tools to assess mAb activity within tumors, we hypothesized that measurement of accessible target levels in tumors could elucidate the pharmacologic activity of a mAb and could be used to compare the activity of different mAbs. Using positron emission tomography (PET), we measured the pharmacodynamics of immune checkpoint protein programmed-death ligand 1 (PD-L1) to evaluate pharmacologic effects of mAbs targeting PD-L1 and its receptor programmed cell death protein 1 (PD-1). For PD-L1 quantification, we first developed a small peptide-based fluorine-18-labeled PET imaging agent, [18F]DK222, which provided high-contrast images in preclinical models. We then quantified accessible PD-L1 levels in the tumor bed during treatment with anti-PD-1 and anti-PD-L1 mAbs. Applying mixed-effects models to these data, we found subtle differences in the pharmacodynamic effects of two anti-PD-1 mAbs (nivolumab and pembrolizumab). In contrast, we observed starkly divergent target engagement with anti-PD-L1 mAbs (atezolizumab, avelumab, and durvalumab) that were administered at equivalent doses, correlating with differential effects on tumor growth. Thus, we show that measuring PD-L1 pharmacodynamics informs mechanistic understanding of therapeutic mAbs targeting PD-L1 and PD-1. These findings demonstrate the value of quantifying target pharmacodynamics to elucidate the pharmacologic activity of mAbs, independent of mAb biophysical properties and inclusive of all physiological variables, which are highly heterogeneous within and across tumors and patients.
Collapse
|
21
|
Islam A, Pishesha N, Harmand TJ, Heston H, Woodham AW, Cheloha RW, Bousbaine D, Rashidian M, Ploegh HL. Converting an Anti-Mouse CD4 Monoclonal Antibody into an scFv Positron Emission Tomography Imaging Agent for Longitudinal Monitoring of CD4 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1468-1477. [PMID: 34408009 PMCID: PMC8387391 DOI: 10.4049/jimmunol.2100274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
Immuno-positron emission tomography (PET), a noninvasive imaging modality, can provide a dynamic approach for longitudinal assessment of cell populations of interest. Transformation of mAbs into single-chain variable fragment (scFv)-based PET imaging agents would allow noninvasive tracking in vivo of a wide range of possible targets. We used sortase-mediated enzymatic labeling in combination with PEGylation to develop an anti-mouse CD4 scFv-based PET imaging agent constructed from an anti-mouse CD4 mAb. This anti-CD4 scFv can monitor the in vivo distribution of CD4+ T cells by immuno-PET. We tracked CD4+ and CD8+ T cells in wild-type mice, in immunodeficient recipients reconstituted with monoclonal populations of OT-II and OT-I T cells, and in a B16 melanoma model. Anti-CD4 and -CD8 immuno-PET showed that the persistence of both CD4+ and CD8+ T cells transferred into immunodeficient mice improved when recipients were immunized with OVA in CFA. In tumor-bearing animals, infiltration of both CD4+ and CD8+ T cells increased as the tumor grew. The approach described in this study should be readily applicable to convert clinically useful Abs into the corresponding scFv PET imaging agents.
Collapse
Affiliation(s)
- Ashraful Islam
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Society of Fellows, Harvard University, Cambridge, MA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Thibault J Harmand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Hailey Heston
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Djenet Bousbaine
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA; and
- Department of Radiology, Harvard Medical School, Boston, MA
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA;
- Department of Pediatrics, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Shboul ZA, Diawara N, Vossough A, Chen JY, Iftekharuddin KM. Joint Modeling of RNAseq and Radiomics Data for Glioma Molecular Characterization and Prediction. Front Med (Lausanne) 2021; 8:705071. [PMID: 34490297 PMCID: PMC8416908 DOI: 10.3389/fmed.2021.705071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
RNA sequencing (RNAseq) is a recent technology that profiles gene expression by measuring the relative frequency of the RNAseq reads. RNAseq read counts data is increasingly used in oncologic care and while radiology features (radiomics) have also been gaining utility in radiology practice such as disease diagnosis, monitoring, and treatment planning. However, contemporary literature lacks appropriate RNA-radiomics (henceforth, radiogenomics ) joint modeling where RNAseq distribution is adaptive and also preserves the nature of RNAseq read counts data for glioma grading and prediction. The Negative Binomial (NB) distribution may be useful to model RNAseq read counts data that addresses potential shortcomings. In this study, we propose a novel radiogenomics-NB model for glioma grading and prediction. Our radiogenomics-NB model is developed based on differentially expressed RNAseq and selected radiomics/volumetric features which characterize tumor volume and sub-regions. The NB distribution is fitted to RNAseq counts data, and a log-linear regression model is assumed to link between the estimated NB mean and radiomics. Three radiogenomics-NB molecular mutation models (e.g., IDH mutation, 1p/19q codeletion, and ATRX mutation) are investigated. Additionally, we explore gender-specific effects on the radiogenomics-NB models. Finally, we compare the performance of the proposed three mutation prediction radiogenomics-NB models with different well-known methods in the literature: Negative Binomial Linear Discriminant Analysis (NBLDA), differentially expressed RNAseq with Random Forest (RF-genomics), radiomics and differentially expressed RNAseq with Random Forest (RF-radiogenomics), and Voom-based count transformation combined with the nearest shrinkage classifier (VoomNSC). Our analysis shows that the proposed radiogenomics-NB model significantly outperforms (ANOVA test, p < 0.05) for prediction of IDH and ATRX mutations and offers similar performance for prediction of 1p/19q codeletion, when compared to the competing models in the literature, respectively.
Collapse
Affiliation(s)
- Zeina A. Shboul
- Vision Lab, Department of Electrical & Computer Engineering, Old Dominion University, Norfolk, VA, United States
| | - Norou Diawara
- Department of Mathematics & Statistics, Old Dominion University, Norfolk, VA, United States
| | - Arastoo Vossough
- Department of Radiology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, United States
| | - James Y. Chen
- University of California, San Diego Health System, San Diego, CA, United States
| | - Khan M. Iftekharuddin
- Vision Lab, Department of Electrical & Computer Engineering, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
23
|
Höltke C, Grewer M, Stölting M, Geyer C, Wildgruber M, Helfen A. Exploring the Influence of Different Albumin Binders on Molecular Imaging Probe Distribution. Mol Pharm 2021; 18:2574-2585. [PMID: 34048242 DOI: 10.1021/acs.molpharmaceut.1c00064] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The biodistribution of molecular imaging probes or tracers mainly depends on the chemical nature of the probe and the preferred metabolization and excretion routes. Small molecules have rather short half-lives while antibodies reside inside the organism for a longer period of time. An excretion via kidneys and bladder is faster than a mainly hepatobiliary elimination. To manipulate the biodistribution behavior of probes, different strategies have been pursued, including utilizing serum albumin as an inherent transport mechanism for small molecules. Here, we modified an existing small molecular fluorescent probe targeted to the endothelin-A receptor (ETAR) with three different albumin-binding moieties to search for an optimal modification strategy. A diphenylcyclohexyl (DPCH) group, a p-iodophenyl butyric acid (IPBA), and a fatty acid (FA) group were attached via amino acid linkers. All three modifications result in transient albumin binding of the developed compounds, as concluded from gel electrophoresis investigations. Spectrophotometric measurements applying variable amounts of bovine, murine, and human serum albumin (BSA, MSA, and HSA) reveal distinct variations of absorption and emission intensities and shifts of their maximum wavelengths. Binding to MSA results in the weakest effects, while binding to HSA leads to the strongest. Cell-based in vitro investigations utilizing ETAR-positive HT-1080 fibrosarcoma and ETAR-negative BT-20 breast adenocarcinoma cells support a retained specific target-binding capacity of the modified compounds and different degrees of unspecific binding. In vivo analysis of a HT-1080 xenograft model in nude mice over the course of 1 week by fluorescence reflectance imaging illustrates noticeable differences between the four examined probes. While the IPBA-modified probe shows the highest absolute signal intensity values, the FA-modified probe exhibits the most favorable tumor-to-organ ratios. In summary, reversible binding to albumin enhances the biological half-life of the designed probes substantially and enables near infrared optical imaging of subcutaneous tumors for several days in vivo. Because the unmodified probe already exhibits reasonable results, the attachment of albumin-binding moieties does not lead to a substantially improved imaging outcome in terms of target-to-background ratios. On the other hand, because the implemented transient albumin binding results in an overall higher amount of probe inside tumor lesions, this strategy might be adaptable for theranostic or therapeutic approaches in a future clinical routine.
Collapse
Affiliation(s)
- Carsten Höltke
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Martin Grewer
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Miriam Stölting
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Christiane Geyer
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Moritz Wildgruber
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany.,Department of Radiology, University Hospital, LMU Munich, 80539 Munich, Germany
| | - Anne Helfen
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| |
Collapse
|
24
|
Hyväkkä A, Virtanen V, Kemppainen J, Grönroos TJ, Minn H, Sundvall M. More Than Meets the Eye: Scientific Rationale behind Molecular Imaging and Therapeutic Targeting of Prostate-Specific Membrane Antigen (PSMA) in Metastatic Prostate Cancer and Beyond. Cancers (Basel) 2021; 13:cancers13092244. [PMID: 34067046 PMCID: PMC8125679 DOI: 10.3390/cancers13092244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Prostate-specific membrane antigen (PSMA) is a transmembrane protein that is overexpressed in prostate cancer and correlates with the aggressiveness of the disease. PSMA is a promising target for imaging and therapeutics in prostate cancer patients validated in prospective trials. However, the role of PSMA in prostate cancer progression is poorly understood. In this review, we discuss the biology and scientific rationale behind the use of PSMA and other targets in the detection and theranostics of metastatic prostate cancer. Abstract Prostate cancer is the second most common cancer type in men globally. Although the prognosis for localized prostate cancer is good, no curative treatments are available for metastatic disease. Better diagnostic methods could help target therapies and improve the outcome. Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein that is overexpressed on malignant prostate tumor cells and correlates with the aggressiveness of the disease. PSMA is a clinically validated target for positron emission tomography (PET) imaging-based diagnostics in prostate cancer, and during recent years several therapeutics have been developed based on PSMA expression and activity. The expression of PSMA in prostate cancer can be very heterogeneous and some metastases are negative for PSMA. Determinants that dictate clinical responses to PSMA-targeting therapeutics are not well known. Moreover, it is not clear how to manipulate PSMA expression for therapeutic purposes and develop rational treatment combinations. A deeper understanding of the biology behind the use of PSMA would help the development of theranostics with radiolabeled compounds and other PSMA-based therapeutic approaches. Along with PSMA several other targets have also been evaluated or are currently under investigation in preclinical or clinical settings in prostate cancer. Here we critically elaborate the biology and scientific rationale behind the use of PSMA and other targets in the detection and therapeutic targeting of metastatic prostate cancer.
Collapse
Affiliation(s)
- Anniina Hyväkkä
- Institute of Biomedicine, Cancer Research Unit, FICAN West Cancer Center Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (A.H.); (V.V.)
| | - Verneri Virtanen
- Institute of Biomedicine, Cancer Research Unit, FICAN West Cancer Center Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (A.H.); (V.V.)
- Turku Doctoral Programme of Molecular Medicine (TuDMM), University of Turku, FI-20520 Turku, Finland
| | - Jukka Kemppainen
- Turku PET Centre, University of Turku, FI-20521 Turku, Finland;
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, FI-20521 Turku, Finland
- Docrates Cancer Center, FI-00180 Helsinki, Finland
| | - Tove J. Grönroos
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, FI-20520 Turku, Finland;
| | - Heikki Minn
- Department of Oncology, FICAN West Cancer Center, University of Turku and Turku University Hospital, FI-20521 Turku, Finland;
| | - Maria Sundvall
- Institute of Biomedicine, Cancer Research Unit, FICAN West Cancer Center Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (A.H.); (V.V.)
- Department of Oncology, FICAN West Cancer Center, University of Turku and Turku University Hospital, FI-20521 Turku, Finland;
- Correspondence:
| |
Collapse
|
25
|
Li W, Zeng G, Yan J, Liu X, Jiang X, Yang J, Liu J, Sun D. One-pot green synthesis of I@CNDs-Fe 3O 4 hybrid nanoparticles from kelp for multi-modal imaging in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112037. [PMID: 33947537 DOI: 10.1016/j.msec.2021.112037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 12/21/2022]
Abstract
Multi-modal imaging technologies are playing an increasingly important role in biomedical research. However, there remains a demanding challenge to develop biocompatible contrast agents via a simple, green synthetic route for multi-modal imaging. Here we report the synthesis and applications of a new contrast agent for triple-modal imaging, that is, iodine-containing N-doping carbon nano-dots hybridized with Fe3O4 nanoparticles (I@CNDs-Fe3O4). We develop a one-pot, environment friendly hydrothermal method to synthesize the hybrid nanoparticles, primarily using kelp as the bioresource. I@CNDs-Fe3O4 nanoparticles have been demonstrated to exhibit excellent multi-modal imaging capabilities, including wavelength-tunable fluorescent imaging, X-ray attenuation for CT imaging enhancement, and T2-Weighted MR imaging. Importantly, the formulation of the hybrid nanoparticle provides an optimal solution to address the disequilibrium of osmotic pressure caused by the conventional CT imaging contrast agents of iodine compounds. The I@CNDs-Fe3O4 nanoparticles promise important applications in multi-modal imaging technologies in vivo as a versatile and biocompatible contrast agent.
Collapse
Affiliation(s)
- Wenping Li
- Institute of Chemicobiology and Functional Materials, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, Jiangsu Province, China
| | - Ganmin Zeng
- Institute of Chemicobiology and Functional Materials, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, Jiangsu Province, China
| | - Jun Yan
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province, China
| | - Xiaoli Liu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Qixia District, Nanjing, Jiangsu 210023, China.
| | - Xiaohong Jiang
- Institute of Chemicobiology and Functional Materials, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, Jiangsu Province, China
| | - Jiazhi Yang
- Institute of Chemicobiology and Functional Materials, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, Jiangsu Province, China.
| | - Jian Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province, China
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, Jiangsu Province, China
| |
Collapse
|
26
|
Serkova NJ, Glunde K, Haney CR, Farhoud M, De Lille A, Redente EF, Simberg D, Westerly DC, Griffin L, Mason RP. Preclinical Applications of Multi-Platform Imaging in Animal Models of Cancer. Cancer Res 2021; 81:1189-1200. [PMID: 33262127 PMCID: PMC8026542 DOI: 10.1158/0008-5472.can-20-0373] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/10/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
In animal models of cancer, oncologic imaging has evolved from a simple assessment of tumor location and size to sophisticated multimodality exploration of molecular, physiologic, genetic, immunologic, and biochemical events at microscopic to macroscopic levels, performed noninvasively and sometimes in real time. Here, we briefly review animal imaging technology and molecular imaging probes together with selected applications from recent literature. Fast and sensitive optical imaging is primarily used to track luciferase-expressing tumor cells, image molecular targets with fluorescence probes, and to report on metabolic and physiologic phenotypes using smart switchable luminescent probes. MicroPET/single-photon emission CT have proven to be two of the most translational modalities for molecular and metabolic imaging of cancers: immuno-PET is a promising and rapidly evolving area of imaging research. Sophisticated MRI techniques provide high-resolution images of small metastases, tumor inflammation, perfusion, oxygenation, and acidity. Disseminated tumors to the bone and lung are easily detected by microCT, while ultrasound provides real-time visualization of tumor vasculature and perfusion. Recently available photoacoustic imaging provides real-time evaluation of vascular patency, oxygenation, and nanoparticle distributions. New hybrid instruments, such as PET-MRI, promise more convenient combination of the capabilities of each modality, enabling enhanced research efficacy and throughput.
Collapse
Affiliation(s)
- Natalie J Serkova
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- Animal Imaging Shared Resource, University of Colorado Cancer Center, Aurora, Colorado
| | - Kristine Glunde
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology, and the Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chad R Haney
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, Illinois
| | | | | | | | - Dmitri Simberg
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David C Westerly
- Animal Imaging Shared Resource, University of Colorado Cancer Center, Aurora, Colorado
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lynn Griffin
- Department of Radiology, Veterinary Teaching Hospital, Colorado State University, Fort Collins, Colorado
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
27
|
Yang JM, Ghim CM. Photoacoustic Tomography Opening New Paradigms in Biomedical Imaging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1310:239-341. [PMID: 33834440 DOI: 10.1007/978-981-33-6064-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
After the emergence of the ultrasound, X-ray CT, PET, and MRI, photoacoustic tomography (PAT) is now in the phase of its exponential growth, with its expected full maturation being another form of mainstream clinical imaging modality. By combining the high contrast benefit of optical imaging and the high-resolution deep imaging capability of ultrasound, PAT can provide unprecedented anatomical image contrasts at clinically relevant depths as well as enable the use of a variety of functional and molecular imaging information, which is not possible with conventional imaging modalities. With these strengths, PAT has achieved numerous breakthroughs in various biomedical applications and also provided new technical platforms that may be able to resolve unmet issues in clinics. In this chapter, we provide an overview of the development of PAT technology for several major biomedical applications and provide an approximate projection of the future of PAT.
Collapse
Affiliation(s)
- Joon-Mo Yang
- Center for Photoacoustic Medical Instruments, Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea.
| | - Cheol-Min Ghim
- Department of Physics, School of Natural Science, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
28
|
Song X, Zhu W, Ge X, Li R, Li S, Chen X, Song J, Xie J, Chen X, Yang H. A New Class of NIR‐II Gold Nanocluster‐Based Protein Biolabels for In Vivo Tumor‐Targeted Imaging. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202010870] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xiaorong Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China Fuzhou Fujian 350108 China
| | - Wei Zhu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Xiaoguang Ge
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Renfu Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Key Laboratory of Nanomaterials Fujian Institute of Research on the Structure of Matter Chinese Academy of Sciences Fuzhou Fujian 350002 China
| | - Shihua Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Xian Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Jianping Xie
- Department of Chemical and Biomolecular Engineering National University of Singapore 4 Engineering Drive 4 Singapore 117585 Singapore
- Joint School of National University of Singapore and Tianjin University International Campus of Tianjin University, Binhai New City Fuzhou Fujian 350207 China
| | - Xueyuan Chen
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Key Laboratory of Nanomaterials Fujian Institute of Research on the Structure of Matter Chinese Academy of Sciences Fuzhou Fujian 350002 China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China Fuzhou Fujian 350108 China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China Fuzhou Fujian 350108 China
| |
Collapse
|
29
|
Song X, Zhu W, Ge X, Li R, Li S, Chen X, Song J, Xie J, Chen X, Yang H. A New Class of NIR‐II Gold Nanocluster‐Based Protein Biolabels for In Vivo Tumor‐Targeted Imaging. Angew Chem Int Ed Engl 2020; 60:1306-1312. [DOI: 10.1002/anie.202010870] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Xiaorong Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China Fuzhou Fujian 350108 China
| | - Wei Zhu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Xiaoguang Ge
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Renfu Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Key Laboratory of Nanomaterials Fujian Institute of Research on the Structure of Matter Chinese Academy of Sciences Fuzhou Fujian 350002 China
| | - Shihua Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Xian Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
| | - Jianping Xie
- Department of Chemical and Biomolecular Engineering National University of Singapore 4 Engineering Drive 4 Singapore 117585 Singapore
- Joint School of National University of Singapore and Tianjin University International Campus of Tianjin University, Binhai New City Fuzhou Fujian 350207 China
| | - Xueyuan Chen
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Key Laboratory of Nanomaterials Fujian Institute of Research on the Structure of Matter Chinese Academy of Sciences Fuzhou Fujian 350002 China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China Fuzhou Fujian 350108 China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou Fujian 350116 China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China Fuzhou Fujian 350108 China
| |
Collapse
|
30
|
Li W, Wang Y, Rubins D, Bennacef I, Holahan M, Haley H, Purcell M, Gantert L, Hseih S, Judo M, Seghezzi W, Zhang S, van der Veen EL, Lub-de Hooge MN, de Vries EGE, Evelhoch JL, Klimas M, Hostetler ED. PET/CT Imaging of 89Zr-N-sucDf-Pembrolizumab in Healthy Cynomolgus Monkeys. Mol Imaging Biol 2020; 23:250-259. [PMID: 33104972 PMCID: PMC7910264 DOI: 10.1007/s11307-020-01558-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/03/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
Purpose Programmed cell death-1 receptor (PD-1) and its ligand (PD-L1) are the targets for immunotherapy in many cancer types. Although PD-1 blockade has therapeutic effects, the efficacy differs between patients. Factors contributing to this variability are PD-L1 expression levels and immune cells present in tumors. However, it is not well understood how PD-1 expression in the tumor microenvironment impacts immunotherapy response. Thus, imaging of PD-1-expressing immune cells is of interest. This study aims to evaluate the biodistribution of Zirconium-89 (89Zr)-labeled pembrolizumab, a humanized IgG4 kappa monoclonal antibody targeting PD-1, in healthy cynomolgus monkeys as a translational model of tracking PD-1-positive immune cells. Procedures Pembrolizumab was conjugated with the tetrafluorophenol-N-succinyl desferal-Fe(III) ester (TFP-N-sucDf) and subsequently radiolabeled with 89Zr. Four cynomolgus monkeys with no previous exposure to humanized monoclonal antibodies received tracer only or tracer co-injected with pembrolizumab intravenously over 5 min. Thereafter, a static whole-body positron emission tomography (PET) scan was acquired with 10 min per bed position on days 0, 2, 5, and 7. Image-derived standardized uptake values (SUVmean) were quantified by region of interest (ROI) analysis. Results 89Zr-N-sucDf-pembrolizumab was synthesized with high radiochemical purity (> 99 %) and acceptable molar activity (> 7 MBq/nmol). In animals dosed with tracer only, 89Zr-N-sucDf-pembrolizumab distribution in lymphoid tissues such as mesenteric lymph nodes, spleen, and tonsils increased over time. Except for the liver, low radiotracer distribution was observed in all non-lymphoid tissue including the lung, muscle, brain, heart, and kidney. When a large excess of pembrolizumab was co-administered with a radiotracer, accumulation in the lymph nodes, spleen, and tonsils was reduced, suggestive of target-mediated accumulation. Conclusions 89Zr-N-sucDf-pembrolizumab shows preferential uptake in the lymphoid tissues including the lymph nodes, spleen, and tonsils. 89Zr-N-sucDf-pembrolizumab may be useful in tracking the distribution of a subset of immune cells in non-human primates and humans. Trial Registration ClinicalTrials.gov Identifier: NCT02760225 Supplementary Information The online version contains supplementary material available at 10.1007/s11307-020-01558-w.
Collapse
Affiliation(s)
- Wenping Li
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA.
| | - Yuchuan Wang
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Daniel Rubins
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Idriss Bennacef
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Marie Holahan
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Hyking Haley
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Mona Purcell
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Liza Gantert
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - SuChun Hseih
- PPDM Bioanalysis, MRL South San Francisco, 213 East Grand Blvd, South San Francisco, CA, 94080, USA
| | - Michael Judo
- PPDM Bioanalysis, MRL South San Francisco, 213 East Grand Blvd, South San Francisco, CA, 94080, USA
| | - Wolfgang Seghezzi
- PPDM Bioanalysis, MRL South San Francisco, 213 East Grand Blvd, South San Francisco, CA, 94080, USA
| | - Shuli Zhang
- PPDM Bioanalysis, MRL South San Francisco, 213 East Grand Blvd, South San Francisco, CA, 94080, USA
| | - Elly L van der Veen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Jeffrey L Evelhoch
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Michael Klimas
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Eric D Hostetler
- Translational Biomarkers, Merck & Co., Inc., WP 44D, 770 Sumneytown Pike, West Point, PA, 19486, USA
| |
Collapse
|
31
|
Woodham AW, Zeigler SH, Zeyang EL, Kolifrath SC, Cheloha RW, Rashidian M, Chaparro RJ, Seidel RD, Garforth SJ, Dearling JL, Mesyngier M, Duddempudi PK, Packard AB, Almo SC, Ploegh HL. In vivo detection of antigen-specific CD8 + T cells by immuno-positron emission tomography. Nat Methods 2020; 17:1025-1032. [PMID: 32929269 PMCID: PMC7541633 DOI: 10.1038/s41592-020-0934-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/23/2020] [Indexed: 12/27/2022]
Abstract
The immune system's ability to recognize peptides on major histocompatibility molecules contributes to the eradication of cancers and pathogens. Tracking these responses in vivo could help evaluate the efficacy of immune interventions and improve mechanistic understanding of immune responses. For this purpose, we employ synTacs, which are dimeric major histocompatibility molecule scaffolds of defined composition. SynTacs, when labeled with positron-emitting isotopes, can noninvasively image antigen-specific CD8+ T cells in vivo. Using radiolabeled synTacs loaded with the appropriate peptides, we imaged human papillomavirus-specific CD8+ T cells by positron emission tomography in mice bearing human papillomavirus-positive tumors, as well as influenza A virus-specific CD8+ T cells in the lungs of influenza A virus-infected mice. It is thus possible to visualize antigen-specific CD8+ T-cell populations in vivo, which may serve prognostic and diagnostic roles.
Collapse
Affiliation(s)
- Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stad H Zeigler
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ella L Zeyang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stephen C Kolifrath
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jason L Dearling
- Division of Nuclear Medicine, Department of Radiology, Children's Hospital Boston, Boston, MA, USA
| | - Maia Mesyngier
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Alan B Packard
- Nuclear Medicine and Molecular Imaging, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Mi P, Miyata K, Kataoka K, Cabral H. Clinical Translation of Self‐Assembled Cancer Nanomedicines. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000159] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center West China Hospital, Sichuan University No. 17 People's South Road Chengdu 610041 China
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| | - Kazunori Kataoka
- Institute for Future Initiatives The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐0033 Japan
- Innovation Center of NanoMedicine Kawasaki Institute of Industrial Promotion 3‐25‐14, Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo, Bunkyo‐ku Tokyo 113‐8656 Japan
| |
Collapse
|
33
|
Wu L, Zou H, Wang H, Zhang S, Liu S, Jiang Y, Chen J, Li Y, Shao M, Zhang R, Li X, Dong J, Yang L, Wang K, Zhu X, Sun X. Update on the development of molecular imaging and nanomedicine in China: Optical imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1660. [PMID: 32725869 DOI: 10.1002/wnan.1660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/11/2020] [Accepted: 05/18/2020] [Indexed: 12/22/2022]
Abstract
Molecular imaging has received increased attention worldwide, including in China, because it offers noninvasive characterization of widely diverse clinically significant pathologies. To achieve these goals, nanomedicine has evolved into a broad interdisciplinary field with flexible designs to accommodate and concentrate imaging and therapeutic payloads into pathological cells through selective binding to disease specific cell membrane biomarkers. This concept of personalized medicine reflects the vision of "magic bullets" proposed by German biochemist Paul Ehrlich over 100 years ago. As happening worldwide, Chinese scientists are contributing to this tsunami of science and technologies through impactful national programs and international research collaborations. This review provides a comprehensive update of Chinese innovations to address intractable unmet medical need in China and worldwide in the optical sciences. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Lina Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Hongyan Zou
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Hongbin Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | | | - Shuang Liu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Ying Jiang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jing Chen
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Yingbo Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Mengping Shao
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Ruixin Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xiaona Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jing Dong
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lili Yang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Kai Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, China
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Lesniak WG, Mease RC, Chatterjee S, Kumar D, Lisok A, Wharram B, Kalagadda VR, Emens LA, Pomper MG, Nimmagadda S. Development of [ 18F]FPy-WL12 as a PD-L1 Specific PET Imaging Peptide. Mol Imaging 2020; 18:1536012119852189. [PMID: 31187691 PMCID: PMC6563393 DOI: 10.1177/1536012119852189] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Expression of programmed cell death ligand 1 (PD-L1) within tumors is an important biomarker for guiding immune checkpoint therapies; however, immunohistochemistry-based methods of detection fail to provide a comprehensive picture of PD-L1 levels in an entire patient. To facilitate quantification of PD-L1 in the whole body, we developed a peptide-based, high-affinity PD-L1 imaging agent labeled with [18F]fluoride for positron emission tomography (PET) imaging. The parent peptide, WL12, and the nonradioactive analog of the radiotracer, 19FPy-WL12, inhibit PD-1/PD-L1 interaction at low nanomolar concentrations (half maximal inhibitory concentration [IC50], 26-32 nM). The radiotracer, [18F]FPy-WL12, was prepared by conjugating 2,3,5,6-tetrafluorophenyl 6-[18F]fluoronicotinate ([18F]FPy-TFP) to WL12 and assessed for specificity in vitro in 6 cancer cell lines with varying PD-L1 expression. The uptake of the radiotracer reflected the PD-L1 expression assessed by flow cytometry. Next, we performed the in vivo evaluation of [18F]FPy-WL12 in mice bearing cancer xenografts by PET imaging, ex vivo biodistribution, and blocking studies. In vivo data demonstrated a PD-L1-specific uptake of [18F]FPy-WL12 in tumors that is reduced in mice receiving a blocking dose. The majority of [18F]FPy-WL12 radioactivity was localized in the tumors, liver, and kidneys indicating the need for optimization of the labeling strategy to improve the in vivo pharmacokinetics of the radiotracer.
Collapse
Affiliation(s)
- Wojciech G Lesniak
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Ronnie C Mease
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA.,2 Sidney Kimmel Comprehensive Cancer Center, Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA
| | - Samit Chatterjee
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Dhiraj Kumar
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Ala Lisok
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Bryan Wharram
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Venkateswara Rao Kalagadda
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Leisha A Emens
- 2 Sidney Kimmel Comprehensive Cancer Center, Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA
| | - Martin G Pomper
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA.,2 Sidney Kimmel Comprehensive Cancer Center, Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA
| | - Sridhar Nimmagadda
- 1 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA.,2 Sidney Kimmel Comprehensive Cancer Center, Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
35
|
Yu Z, Eich C, Cruz LJ. Recent Advances in Rare-Earth-Doped Nanoparticles for NIR-II Imaging and Cancer Theranostics. Front Chem 2020; 8:496. [PMID: 32656181 PMCID: PMC7325968 DOI: 10.3389/fchem.2020.00496] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Fluorescence imaging in the second near infrared window (NIR-II, 1,000-1,700 nm) has been widely used in cancer diagnosis and treatment due to its high spatial resolution and deep tissue penetration depths. In this work, recent advances in rare-earth-doped nanoparticles (RENPs)-a novel kind of NIR-II nanoprobes-are presented. The main focus of this study is on the modification of RENPs and their applications in NIR-II in vitro and in vivo imaging and cancer theranostics. Finally, the perspectives and challenges of NIR-II RENPs are discussed.
Collapse
Affiliation(s)
| | | | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging Group, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
36
|
Ko J, Oh J, Ahmed MS, Carlson JCT, Weissleder R. Ultra-fast cycling for multiplexed cellular fluorescence imaging. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 132:6906-6913. [PMID: 34366494 PMCID: PMC8340598 DOI: 10.1002/ange.201915153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Indexed: 11/11/2022]
Abstract
Rapid analysis of single and scant cell populations is essential in modern diagnostics, yet existing methods are often limited and slow. Here we describe an ultra-fast, highly efficient cycling method for the analysis of single cells based on unique linkers for tetrazine (Tz) / trans-cyclooctene (TCO) mediated quenching. Surprisingly, the quenching reaction rates were more than 3 orders of magnitude faster (t1/2 < 1 sec) than predicted. This allowed multi-cycle staining and immune cell profiling within an hour, leveraging the accelerated kinetics to open new diagnostic possibilities for rapid cellular analyses.
Collapse
Affiliation(s)
- Jina Ko
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Juhyun Oh
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Maaz S. Ahmed
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
| | - Jonathan C. T. Carlson
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
37
|
Ko J, Oh J, Ahmed MS, Carlson JCT, Weissleder R. Ultra-fast Cycling for Multiplexed Cellular Fluorescence Imaging. Angew Chem Int Ed Engl 2020; 59:6839-6846. [PMID: 32004403 DOI: 10.1002/anie.201915153] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/08/2020] [Indexed: 12/26/2022]
Abstract
Rapid analysis of single and scant cell populations is essential in modern diagnostics, yet existing methods are often limited and slow. Herein, we describe an ultra-fast, highly efficient cycling method for the analysis of single cells based on unique linkers for tetrazine (Tz)/trans-cyclooctene (TCO)-mediated quenching. Surprisingly, the quenching reaction rates were more than 3 orders of magnitude faster (t1/2 <1 s) than predicted. This allowed multi-cycle staining and immune cell profiling within an hour, leveraging the accelerated kinetics to open new diagnostic possibilities for rapid cellular analyses.
Collapse
Affiliation(s)
- Jina Ko
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Juhyun Oh
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Maaz S Ahmed
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Jonathan C T Carlson
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
38
|
Li J, Chekkoury A, Prakash J, Glasl S, Vetschera P, Koberstein-Schwarz B, Olefir I, Gujrati V, Omar M, Ntziachristos V. Spatial heterogeneity of oxygenation and haemodynamics in breast cancer resolved in vivo by conical multispectral optoacoustic mesoscopy. LIGHT, SCIENCE & APPLICATIONS 2020; 9:57. [PMID: 32337021 PMCID: PMC7154032 DOI: 10.1038/s41377-020-0295-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 02/10/2020] [Accepted: 03/19/2020] [Indexed: 05/11/2023]
Abstract
The characteristics of tumour development and metastasis relate not only to genomic heterogeneity but also to spatial heterogeneity, associated with variations in the intratumoural arrangement of cell populations, vascular morphology and oxygen and nutrient supply. While optical (photonic) microscopy is commonly employed to visualize the tumour microenvironment, it assesses only a few hundred cubic microns of tissue. Therefore, it is not suitable for investigating biological processes at the level of the entire tumour, which can be at least four orders of magnitude larger. In this study, we aimed to extend optical visualization and resolve spatial heterogeneity throughout the entire tumour volume. We developed an optoacoustic (photoacoustic) mesoscope adapted to solid tumour imaging and, in a pilot study, offer the first insights into cancer optical contrast heterogeneity in vivo at an unprecedented resolution of <50 μm throughout the entire tumour mass. Using spectral methods, we resolve unknown patterns of oxygenation, vasculature and perfusion in three types of breast cancer and showcase different levels of structural and functional organization. To our knowledge, these results are the most detailed insights of optical signatures reported throughout entire tumours in vivo, and they position optoacoustic mesoscopy as a unique investigational tool linking microscopic and macroscopic observations.
Collapse
Affiliation(s)
- Jiao Li
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, No.92, Weijin Road, Nankai District, 300072 Tianjin, China
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Andrei Chekkoury
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Jaya Prakash
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
- Department of Instrumentation and Applied Physics, Indian Institute of Science Bangalore, CV Raman Rd, Bengaluru, 560012 Karnataka India
| | - Sarah Glasl
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Paul Vetschera
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Benno Koberstein-Schwarz
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Ivan Olefir
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Vipul Gujrati
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Murad Omar
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, TranslaTUM, Technische Universität München, Ismaningerstr. 22, D-81675 Munich, Germany
| |
Collapse
|
39
|
Hu Z, Chen WH, Tian J, Cheng Z. NIRF Nanoprobes for Cancer Molecular Imaging: Approaching Clinic. Trends Mol Med 2020; 26:469-482. [PMID: 32359478 DOI: 10.1016/j.molmed.2020.02.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/13/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
Near-IR fluorescence imaging (NIRFI) is a highly promising technique for improving cancer theranostics in the era of precision medicine. Through the combination with cutting-edge bionanotechnologies, the potential of NIRFI can be greatly broadened. A variety of novel NIRF nanoprobes has been developed with ultimate goals of addressing unmet medical needs. Here, we present recent breakthroughs on the fundamental aspects of NIRFI, such as imaging at long wavelengths (1000-1700 nm), and the use of new approaches (X-rays, chemiluminescence, radioluminescence, etc.) for the excitation of novel nanoprobes. Within two decades, research on NIRF nanoprobes has translated to clinical trials and it will further translate to cancer management.
Collapse
Affiliation(s)
- Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing 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, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Wen-Hua Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China; Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Bio-X Program, and Stanford Cancer Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing 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, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; School of Life Science and Technology, Xidian University, Xian 710071, PR China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, PR China.
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Bio-X Program, and Stanford Cancer Center, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
40
|
Ng TS, Garlin MA, Weissleder R, Miller MA. Improving nanotherapy delivery and action through image-guided systems pharmacology. Theranostics 2020; 10:968-997. [PMID: 31938046 PMCID: PMC6956809 DOI: 10.7150/thno.37215] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in the translation of therapeutic nanoparticles (TNPs) into the clinic, the field continues to face challenges in predictably and selectively delivering nanomaterials for the treatment of solid cancers. The concept of enhanced permeability and retention (EPR) has been coined as a convenient but simplistic descriptor of high TNP accumulation in some tumors. However, in practice EPR represents a number of physiological variables rather than a single one (including dysfunctional vasculature, compromised lymphatics and recruited host cells, among other aspects of the tumor microenvironment) — each of which can be highly heterogenous within a given tumor, patient and across patients. Therefore, a clear need exists to dissect the specific biophysical factors underlying the EPR effect, to formulate better TNP designs, and to identify patients with high-EPR tumors who are likely to respond to TNP. The overall pharmacology of TNP is governed by an interconnected set of spatially defined and dynamic processes that benefit from a systems-level quantitative approach, and insights into the physiology have profited from the marriage between in vivo imaging and quantitative systems pharmacology (QSP) methodologies. In this article, we review recent developments pertinent to image-guided systems pharmacology of nanomedicines in oncology. We first discuss recent developments of quantitative imaging technologies that enable analysis of nanomaterial pharmacology at multiple spatiotemporal scales, and then examine reports that have adopted these imaging technologies to guide QSP approaches. In particular, we focus on studies that have integrated multi-scale imaging with computational modeling to derive insights about the EPR effect, as well as studies that have used modeling to guide the manipulation of the EPR effect and other aspects of the tumor microenvironment for improving TNP action. We anticipate that the synergistic combination of imaging with systems-level computational methods for effective clinical translation of TNPs will only grow in relevance as technologies increase in resolution, multiplexing capability, and in the ability to examine heterogeneous behaviors at the single-cell level.
Collapse
|
41
|
Abstract
Image analysis in clinical research has evolved at fast pace in the last decade. This review discusses basic concepts ranging from immunohistochemistry to advanced techniques such as multiplex imaging, digital pathology, flow cytometry and intravital microscopy. Tissue imaging
ex vivo is still one of the gold-standards in the field due to feasibility. We describe here different protocols and applications of digital analysis providing basic and clinical researchers with an overview on how to analyse tissue images.
In vivo imaging is not easily accessible to researchers; however, it provides invaluable dynamic information. Overall, we discuss a plethora of techniques that - when combined - constitute a powerful platform for basic and translational cancer research.
Collapse
Affiliation(s)
- Oscar Maiques
- Barts Cancer Institute, John Vane Science Building, Charterhouse Square, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Mirella Georgouli
- Oncology Cell Therapy RU, GlaxoSmithKline, Stevenage, London, SG1 2NY, UK
| | - Victoria Sanz-Moreno
- Barts Cancer Institute, John Vane Science Building, Charterhouse Square, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
42
|
Sellmyer MA, Richman SA, Lohith K, Hou C, Weng CC, Mach RH, O'Connor RS, Milone MC, Farwell MD. Imaging CAR T Cell Trafficking with eDHFR as a PET Reporter Gene. Mol Ther 2019; 28:42-51. [PMID: 31668558 DOI: 10.1016/j.ymthe.2019.10.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 01/11/2023] Open
Abstract
Cell-based therapeutics have considerable promise across diverse medical specialties; however, reliable human imaging of the distribution and trafficking of genetically engineered cells remains a challenge. We developed positron emission tomography (PET) probes based on the small-molecule antibiotic trimethoprim (TMP) that can be used to image the expression of the Escherichia coli dihydrofolate reductase enzyme (eDHFR) and tested the ability of [18F]-TMP, a fluorine-18 probe, to image primary human chimeric antigen receptor (CAR) T cells expressing the PET reporter gene eDHFR, yellow fluorescent protein (YFP), and Renilla luciferase (rLuc). Engineered T cells showed an approximately 50-fold increased bioluminescent imaging signal and 10-fold increased [18F]-TMP uptake compared to controls in vitro. eDHFR-expressing anti-GD2 CAR T cells were then injected into mice bearing control GD2- and GD2+ tumors. PET/computed tomography (CT) images acquired on days 7 and 13 demonstrated early residency of CAR T cells in the spleen followed by on-target redistribution to the GD2+ tumors. This was corroborated by autoradiography and anti-human CD8 immunohistochemistry. We found a high sensitivity of detection for identifying tumor-infiltrating CD8 CAR T cells, ∼11,000 cells per mm3. These data suggest that the [18F]-TMP/eDHFR PET pair offers important advantages that could better allow investigators to monitor immune cell trafficking to tumors in patients.
Collapse
Affiliation(s)
- Mark A Sellmyer
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Sarah A Richman
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katheryn Lohith
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Catherine Hou
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chi-Chang Weng
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roddy S O'Connor
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael D Farwell
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
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
|
44
|
Li X, Lu S, Xiong Z, Hu Y, Ma D, Lou W, Peng C, Shen M, Shi X. Light-Addressable Nanoclusters of Ultrasmall Iron Oxide Nanoparticles for Enhanced and Dynamic Magnetic Resonance Imaging of Arthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901800. [PMID: 31592427 PMCID: PMC6774037 DOI: 10.1002/advs.201901800] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Indexed: 05/29/2023]
Abstract
Design of novel nanoplatforms with single imaging elements for dynamic and enhanced T 1/T 2-weighted magnetic resonance (MR) imaging of diseases still remains significantly challenging. Here, a facile strategy to synthesize light-addressable ultrasmall Fe3O4 nanoparticles (NPs) that can form nanoclusters (NCs) under laser irradiation for enhanced and dynamic T 1/T 2-weighted MR imaging of inflammatory arthritis is reported. Citric acid-stabilized ultrasmall Fe3O4 NPs synthesized via a solvothermal approach are linked with both the arthritis targeting ligand folic acid (FA) and light-addressable unit diazirine (DA) via polyethylene glycol (PEG) spacer. The formed ultrasmall Fe3O4-PEG-(DA)-FA NPs are cytocompatible, display FA-mediated targeting specificity to arthritis-associated macrophage cells, and can form NCs upon laser irradiation to have tunable r 1 and r 2 relaxivities by varying the laser irradiation duration. With these properties owned, the designed Fe3O4-PEG-(DA)-FA NPs can be used for T 1-weighted MR imaging of arthritis without lasers and enhanced dual-mode T 1/T 2-weighted MR imaging of arthritis under laser irradiation due to the formation of NCs that have extended accumulation within the arthritis region and limited intravasation back to the blood circulation. The designed light-addressable Fe3O4-PEG-(DA)-FA NPs may be used as a promising platform for dynamic and precision T 1/T 2-weighted MR imaging of other diseases.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of ChemistryChemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Shiyi Lu
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of ChemistryChemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Zuogang Xiong
- Cancer Center, China‐France Joint Laboratory for Healthcare TheranosticsShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
| | - Yong Hu
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of ChemistryChemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Dan Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of ChemistryChemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Wenqi Lou
- Cancer Center, China‐France Joint Laboratory for Healthcare TheranosticsShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
| | - Chen Peng
- Cancer Center, China‐France Joint Laboratory for Healthcare TheranosticsShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
- Ninghai First HospitalNingbo315600P. R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of ChemistryChemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsInternational Joint Laboratory for Advanced Fiber and Low‐Dimension MaterialsCollege of ChemistryChemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| |
Collapse
|
45
|
Chen Z, Deán-Ben XL, Liu N, Gujrati V, Gottschalk S, Ntziachristos V, Razansky D. Concurrent fluorescence and volumetric optoacoustic tomography of nanoagent perfusion and bio-distribution in solid tumors. BIOMEDICAL OPTICS EXPRESS 2019; 10:5093-5102. [PMID: 31646032 PMCID: PMC6788589 DOI: 10.1364/boe.10.005093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 05/08/2023]
Abstract
Intravenously administered liposomes and other nano-sized particles are known to passively accumulate in solid tumors via the enhanced permeability and retention (EPR) effect, which is extensively explored toward the improvement of diagnosis and drug delivery in oncology. Agent extravasation into tumors is often hampered by the mononuclear phagocytic and renal systems, which sequester and/or eliminate most of the nanoparticles from the body. Dynamic imaging of the tumor microcirculation and bolus perfusion can thus facilitate optimization of the nanoparticle delivery. When it comes to non-invasive visualization of rapid biological dynamics in whole tumors, the currently available pre-clinical imaging modalities are commonly limited by shallow penetration, lack of suitable contrast or otherwise insufficient spatial or temporal resolution. Herein, we demonstrate the unique capabilities of a combined epi-fluorescence and optoacoustic tomography (FLOT) system for characterizing contrast agent dynamics in orthotopic breast tumors in mice. A liposomal indocyanine green (Lipo-ICG) preparation was administered intravenously with the time-lapse data continuously acquired during and after the injection procedure. In addition to the highly sensitive detection of the fluorescence agent by the epi-fluorescence modality, the volumetric multi-spectral optoacoustic tomography readings further enabled resolving deep-seated vascular structures with high spatial resolution and hence provided accurate readings of the dynamic bio-distribution of nanoparticles in the entire tumor in 3D. The synergetic combination of the two modalities can become a powerful tool in cancer research and potentially aid the diagnosis, staging and treatment guidance of certain types of cancer in the clinical setting.
Collapse
Affiliation(s)
- Zhenyue Chen
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
| | - Xosé Luis Deán-Ben
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
| | - Nian Liu
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
- Faculty of Medicine, Technical University of Munich, Germany
| | - Vipul Gujrati
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
| | - Sven Gottschalk
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
| | - Vasilis Ntziachristos
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
- Faculty of Medicine, Technical University of Munich, Germany
| | - Daniel Razansky
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
- Institute for Biological and Medical Imaging, Helmholtz Center Munich, Neuherberg, Germany
- Faculty of Medicine, Technical University of Munich, Germany
| |
Collapse
|
46
|
Huang D, He B, Mi P. Calcium phosphate nanocarriers for drug delivery to tumors: imaging, therapy and theranostics. Biomater Sci 2019; 7:3942-3960. [PMID: 31414096 DOI: 10.1039/c9bm00831d] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Calcium phosphate (CaP) was engineered as a drug delivery nanocarrier nearly 50 years ago due to its biocompatibility and biodegradability. In recent years, several approaches have been developed for the preparation of size-controllable, stable and multifunctional CaP nanocarriers, and several targeting moieties have also been decorated on the surface of these nanocarriers for active targeting. The CaP nanocarriers have been utilized for loading probes, nucleic acids, anticancer drugs and photosensitizers for cancer imaging, therapy and theranostics. Herein, we reviewed the recent advances in the preparation strategies of CaP nanocarriers and the applications of these nanocarriers in tumor diagnosis, gene delivery, drug delivery and theranostics and finally provided perspectives.
Collapse
Affiliation(s)
- Dan Huang
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Number 17, 3rd Section, Renmin South Road, Chengdu, Sichuan 610041, P.R. China.
| | - Bin He
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Number 17, 3rd Section, Renmin South Road, Chengdu, Sichuan 610041, P.R. China.
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Number 17, 3rd Section, Renmin South Road, Chengdu, Sichuan 610041, P.R. China.
| |
Collapse
|
47
|
Feng GK, Ye JC, Zhang WG, Mei Y, Zhou C, Xiao YT, Li XL, Fan W, Wang F, Zeng MS. Integrin α6 targeted positron emission tomography imaging of hepatocellular carcinoma in mouse models. J Control Release 2019; 310:11-21. [PMID: 31400382 DOI: 10.1016/j.jconrel.2019.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/13/2019] [Accepted: 08/03/2019] [Indexed: 12/20/2022]
Abstract
Integrin α6 emerges to be a diagnostic biomarker for hepatocellular carcinoma (HCC). Here, we translated our previously identified integrin α6 targeted peptide RWY into a positron emission tomography (PET) tracer 18F-RWY for the detection of HCC lesions in following four HCC mouse models including subcutaneous, orthotopic, genetically engineered and chemical induced HCC mice. 18F-RWY produced high PET signals in liver tumor tissues that were reduced by blocking studies using nonradiolabeled RWY peptide. We compared the integrin α6 targeted PET tracer 18F-RWY with the integrin αvβ3-targeted PET tracer 18F-3PRGD2 and the clinical PET tracer 18F-FDG in chemical induced HCC mice. Among 12 HCC identified by enhanced magnetic resonance imaging (MRI) with hepatocellular specific gadoxetate disodium Gd-EOB-DTPA, the sensitivities of 18F-RWY, 18F-3PRGD2 and 18F-FDG were approximately 92%, 73% and 50% while the tumor-to-liver ratios were 4.36 ± 1.41, 1.97 ± 0.43 and 1.63 ± 0.23 respectively. Additionally, PET imaging with the integrin α6 targeted 18F-RWY enabled to visualize small HCC lesions with diameters approximately 0.2 cm that was hard to be distinguished from surround hepatic vascular by enhanced MRI with Gd-EOB-DTPA. These findings potentiate the use of integrin α6 targeted PET tracer 18F-RWY for the detection of HCC.
Collapse
Affiliation(s)
- Guo-Kai Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jia-Cong Ye
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wei-Guang Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yan Mei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chao Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yi-Tai Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xin-Ling Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wei Fan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Fan Wang
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
48
|
Brown E, Brunker J, Bohndiek SE. Photoacoustic imaging as a tool to probe the tumour microenvironment. Dis Model Mech 2019; 12:dmm039636. [PMID: 31337635 PMCID: PMC6679374 DOI: 10.1242/dmm.039636] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The tumour microenvironment (TME) is a complex cellular ecosystem subjected to chemical and physical signals that play a role in shaping tumour heterogeneity, invasion and metastasis. Studying the roles of the TME in cancer progression would strongly benefit from non-invasive visualisation of the tumour as a whole organ in vivo, both preclinically in mouse models of the disease, as well as in patient tumours. Although imaging techniques exist that can probe different facets of the TME, they face several limitations, including limited spatial resolution, extended scan times and poor specificity from confounding signals. Photoacoustic imaging (PAI) is an emerging modality, currently in clinical trials, that has the potential to overcome these limitations. Here, we review the biological properties of the TME and potential of existing imaging methods that have been developed to analyse these properties non-invasively. We then introduce PAI and explore the preclinical and clinical evidence that support its use in probing multiple features of the TME simultaneously, including blood vessel architecture, blood oxygenation, acidity, extracellular matrix deposition, lipid concentration and immune cell infiltration. Finally, we highlight the future prospects and outstanding challenges in the application of PAI as a tool in cancer research and as part of a clinical oncologist's arsenal.
Collapse
Affiliation(s)
- Emma Brown
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Joanna Brunker
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
49
|
Dong P, Wang X, Zheng J, Zhang X, Li Y, Wu H, Li L. Recent Advances in Targeting Nuclear Molecular Imaging Driven by Tetrazine Bioorthogonal Chemistry. Curr Med Chem 2019; 27:3924-3943. [PMID: 31267851 DOI: 10.2174/1386207322666190702105829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 02/05/2023]
Abstract
Molecular imaging techniques apply sophisticated technologies to monitor, directly or indirectly, the spatiotemporal distribution of molecular or cellular processes for biomedical, diagnostic, or therapeutic purposes. For example, Single-Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET) imaging, the most representative modalities of molecular imaging, enable earlier and more accurate diagnosis of cancer and cardiovascular diseases. New possibilities for noninvasive molecular imaging in vivo have emerged with advances in bioorthogonal chemistry. For example, tetrazine-related Inverse Electron Demand Diels-Alder (IEDDA) reactions can rapidly generate short-lived radioisotope probes in vivo that provide strong contrast for SPECT and PET. Here, we review pretargeting strategies for molecular imaging and novel radiotracers synthesized via tetrazine bioorthogonal chemistry. We systematically describe advances in direct radiolabeling and pretargeting approaches in SPECT and PET using metal and nonmetal radioisotopes based on tetrazine bioorthogonal reactions, and we discuss prospects for the future of such contrast agents.
Collapse
Affiliation(s)
- Ping Dong
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xueyi Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junwei Zheng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoyang Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Haoxing Wu
- Huaxi MR Research Center, Department of Radiology, West China Hospital and West China School of Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
50
|
Xia Y, Xu C, Zhang X, Ning P, Wang Z, Tian J, Chen X. Liposome-based probes for molecular imaging: from basic research to the bedside. NANOSCALE 2019; 11:5822-5838. [PMID: 30888379 DOI: 10.1039/c9nr00207c] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Molecular imaging is very important in disease diagnosis and prognosis. Liposomes are excellent carriers for different types of molecular imaging probes. In this work, we summarize current developments in liposome-based probes used for molecular imaging and their applications in image-guided drug delivery and tumour surgery, including computed tomography (CT), ultrasound imaging (USI), magnetic resonance imaging (MRI), positron emission tomography (PET), fluorescence imaging (FLI) and photoacoustic imaging (PAI). We also summarized liposome-based multimodal imaging probes and new targeting strategies for liposomes. This work will offer guidance for the design of liposome-based imaging probes for future clinical applications.
Collapse
Affiliation(s)
- Yuqiong Xia
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China.
| | | | | | | | | | | | | |
Collapse
|