1
|
Li N, Pi C, Zhu S, Li X, Wang L, Shi P, Zuo Y, Zheng W, Jiang J, Yang Y, Zhang Q, Tao L, Chu S, Wei Y, Zhao L. Opportunities for the treatment of atherosclerosis: selectins. Pharmacol Res 2025:107807. [PMID: 40449813 DOI: 10.1016/j.phrs.2025.107807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 05/17/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Despite the widespread availability of selectins for tumor therapy, their contribution to atherosclerosis has long been under-emphasized due to their "cofactor's" status and technological limitations. However, advances in immunohistology, glycomics, and related technologies require us to reassess their relationship. Thus, this review identifies pivotal translational opportunities from the intricate mechanisms and explores the clinical promise of selectins in the diagnosis and treatment of atherosclerosis based on the latest clinical research. This review provides insights into selectin-specific tracers and inhibitors, providing lessons for more precise diagnosis and treatment of patients with atherosclerosis.
Collapse
Affiliation(s)
- Nong Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, People's Republic of China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China; Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Chao Pi
- Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Siying Zhu
- Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Xiumei Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, People's Republic of China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China; Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Liu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, People's Republic of China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China; Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Peng Shi
- Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University
| | - Jun Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Qiong Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, People's Republic of China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China; Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Lei Tao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, People's Republic of China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China; Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College.
| | - Yumeng Wei
- Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China; Central Nervous System Product Research and Development Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China.
| |
Collapse
|
2
|
O’Connor EE, Sullivan EV, Chang L, Hammoud DA, Wilson TW, Ragin AB, Meade CS, Coughlin J, Ances BM. Imaging of Brain Structural and Functional Effects in People With Human Immunodeficiency Virus. J Infect Dis 2023; 227:S16-S29. [PMID: 36930637 PMCID: PMC10022717 DOI: 10.1093/infdis/jiac387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Before the introduction of antiretroviral therapy, human immunodeficiency virus (HIV) infection was often accompanied by central nervous system (CNS) opportunistic infections and HIV encephalopathy marked by profound structural and functional alterations detectable with neuroimaging. Treatment with antiretroviral therapy nearly eliminated CNS opportunistic infections, while neuropsychiatric impairment and peripheral nerve and organ damage have persisted among virally suppressed people with HIV (PWH), suggesting ongoing brain injury. Neuroimaging research must use methods sensitive for detecting subtle HIV-associated brain structural and functional abnormalities, while allowing for adjustments for potential confounders, such as age, sex, substance use, hepatitis C coinfection, cardiovascular risk, and others. Here, we review existing and emerging neuroimaging tools that demonstrated promise in detecting markers of HIV-associated brain pathology and explore strategies to study the impact of potential confounding factors on these brain measures. We emphasize neuroimaging approaches that may be used in parallel to gather complementary information, allowing efficient detection and interpretation of altered brain structure and function associated with suboptimal clinical outcomes among virally suppressed PWH. We examine the advantages of each imaging modality and systematic approaches in study design and analysis. We also consider advantages of combining experimental and statistical control techniques to improve sensitivity and specificity of biotype identification and explore the costs and benefits of aggregating data from multiple studies to achieve larger sample sizes, enabling use of emerging methods for combining and analyzing large, multifaceted data sets. Many of the topics addressed in this article were discussed at the National Institute of Mental Health meeting "Biotypes of CNS Complications in People Living with HIV," held in October 2021, and are part of ongoing research initiatives to define the role of neuroimaging in emerging alternative approaches to identifying biotypes of CNS complications in PWH. An outcome of these considerations may be the development of a common neuroimaging protocol available for researchers to use in future studies examining neurological changes in the brains of PWH.
Collapse
Affiliation(s)
- Erin E O’Connor
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Edith V Sullivan
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
- Center for Health Sciences, SRI International, Menlo Park, California, USA
| | - Linda Chang
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, Maryland, USA
| | - Tony W Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, Nebraska, USA
| | - Ann B Ragin
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Christina S Meade
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jennifer Coughlin
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Beau M Ances
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Translation of focused ultrasound for blood-brain barrier opening in glioma. J Control Release 2022; 345:443-463. [PMID: 35337938 DOI: 10.1016/j.jconrel.2022.03.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022]
Abstract
Survival outcomes for patients with glioblastoma multiforme (GBM) have remained poor for the past 15 years, reflecting a clear challenge in the development of more effective treatment strategies. The efficacy of systemic therapies for GBM is greatly limited by the presence of the blood-brain barrier (BBB), which prevents drug penetration and accumulation in regions of infiltrative tumour, as represented in a consistent portion of GBM lesions. Focused ultrasound (FUS) - a technique that uses low-frequency ultrasound waves to induce targeted temporary disruption of the BBB - promises to improve survival outcomes by enhancing drug delivery and accumulation to infiltrating tumour regions. In this review we discuss the current state of preclinical investigations using FUS to enhance delivery of systemic therapies to intracranial neoplasms. We highlight critical methodological inconsistencies that are hampering clinical translation of FUS and we provide guiding principles for future preclinical studies. Particularly, we focus our attention on the importance of the selection of clinically relevant animal models and to the standardization of methods for FUS delivery, which will be paramount to the successful clinical translation of this promising technology for treatment in GBM patients. We also discuss how preclinical FUS research can benefit the development of GBM immunotherapies.
Collapse
|
4
|
Zheng H, Yuan C, Cai J, Pu W, Wu P, Li C, Li G, Zhang Y, Zhang J, Guo J, Huang D. Early diagnosis of breast cancer lung metastasis by nanoprobe-based luminescence imaging of the pre-metastatic niche. J Nanobiotechnology 2022; 20:134. [PMID: 35292019 PMCID: PMC8922882 DOI: 10.1186/s12951-022-01346-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/02/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Early detection of breast cancer lung metastasis remains highly challenging, due to few metastatic cancer cells at an early stage. Herein we propose a new strategy for early diagnosis of lung metastasis of breast cancer by luminescence imaging of pulmonary neutrophil infiltration via self-illuminating nanoprobes. METHODS Luminescent nanoparticles (LAD NPs) were engineered using a biocompatible, neutrophil-responsive self-illuminating cyclodextrin material and an aggregation-induced emission agent. The chemiluminescence resonance energy transfer (CRET) effect and luminescence properties of LAD NPs were fully characterized. Using mouse peritoneal neutrophils, in vitro luminescence properties of LAD NPs were thoroughly examined. In vivo luminescence imaging and correlation analyses were performed in mice inoculated with 4T1 cancer cells. Moreover, an active targeting nanoprobe was developed by surface decoration of LAD NPs with a neutrophil-targeting peptide, which was also systemically evaluated by in vitro and in vivo studies. RESULTS LAD NPs can generate long-wavelength and persistent luminescence due to the CRET effect. In a mouse model of 4T1 breast cancer lung metastasis, we found desirable correlation between neutrophils and tumor cells in the lungs, demonstrating the effectiveness of early imaging of the pre-metastatic niche by the newly developed LAD NPs. The active targeting nanoprobe showed further enhanced luminescence imaging capability for early detection of pulmonary metastasis. Notably, the targeting nanoprobe-based luminescence imaging strategy remarkably outperformed PET/CT imaging modalities in the examined mouse model. Also, preliminary tests demonstrated good safety of LAD NPs. CONCLUSIONS The neutrophil-targeting imaging strategy based on newly developed luminescence nanoparticles can serve as a promising modality for early diagnosis of lung metastasis of breast cancers.
Collapse
Affiliation(s)
- Hanwen Zheng
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Chunsen Yuan
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jiajun Cai
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Peng Wu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
- College of Pharmacy and Medical Technology, Hanzhong Vocational and Technical College, Hanzhong, 723000, Shaanxi, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Gang Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Yang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jiawei Guo
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China.
| | - Dingde Huang
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China.
| |
Collapse
|
5
|
Hyodo F, Eto H, Naganuma T, Koyasu N, Elhelaly AE, Noda Y, Kato H, Murata M, Akahoshi T, Hashizume M, Utsumi H, Matsuo M. In Vivo Dynamic Nuclear Polarization Magnetic Resonance Imaging for the Evaluation of Redox-Related Diseases and Theranostics. Antioxid Redox Signal 2022; 36:172-184. [PMID: 34015957 DOI: 10.1089/ars.2021.0087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Significance:In vivo molecular and metabolic imaging is an emerging field in biomedical research that aims to perform noninvasive detection of tissue metabolism in disease states and responses to therapeutic agents. The imbalance in tissue oxidation/reduction (Redox) states is related to the onset and progression of several diseases. Tissue redox metabolism provides biomarkers for early diagnosis and drug treatments. Thus, noninvasive imaging of redox metabolism could be a useful, novel diagnostic tool for diagnosis of redox-related disease and drug discovery. Recent Advances:In vivo dynamic nuclear polarization magnetic resonance imaging (DNP-MRI) is a technique that enables the imaging of free radicals in living animals. DNP enhances the MRI signal by irradiating the target tissue or solution with the free radical molecule's electron paramagnetic resonance frequency before executing pulse sequence of the MRI. In vivo DNP-MRI with redox-sensitive nitroxyl radicals as the DNP redox contrast agent enables the imaging of the redox metabolism on various diseases. Moreover, nitroxyl radicals show antioxidant effects that suppress oxidative stress. Critical Issues: To date, considerable progress has been documented preclinically in the development of animal imaging systems. Here, we review redox imaging of in vivo DNP-MRI with a focus on the recent progress of this system and its uses in patients with redox-related diseases. Future Directions: This technique could have broad applications in the study of other redox-related diseases, such as cancer, inflammation, and neurological disorders, and facilitate the evaluation of treatment response as a theranostic tool. Antioxid. Redox Signal. 36, 172-184.
Collapse
Affiliation(s)
- Fuminori Hyodo
- Department of Radiology, Frontier Science for Imaging, School of Medicine, Gifu University, Gifu, Japan
| | - Hinako Eto
- Center for Advanced Medical Open Innovation, Kyushu University, Fukuoka, Japan
| | | | | | - Abdelazim Elsayed Elhelaly
- Department of Radiology, Frontier Science for Imaging, School of Medicine, Gifu University, Gifu, Japan.,Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Hiroki Kato
- Department of Radiology, Gifu University, Gifu, Japan
| | - Masaharu Murata
- Center for Advanced Medical Open Innovation, Kyushu University, Fukuoka, Japan.,Graduate School of Medicine, Disaster and Emergency Medicine, Kyushu University, Fukuoka, Japan
| | - Tomohiko Akahoshi
- Graduate School of Medicine, Disaster and Emergency Medicine, Kyushu University, Fukuoka, Japan
| | | | - Hideo Utsumi
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | |
Collapse
|
6
|
Prigent K, Vigne J. Advances in Radiopharmaceutical Sciences for Vascular Inflammation Imaging: Focus on Clinical Applications. Molecules 2021; 26:molecules26237111. [PMID: 34885690 PMCID: PMC8659223 DOI: 10.3390/molecules26237111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 01/18/2023] Open
Abstract
Biomedical imaging technologies offer identification of several anatomic and molecular features of disease pathogenesis. Molecular imaging techniques to assess cellular processes in vivo have been useful in advancing our understanding of several vascular inflammatory diseases. For the non-invasive molecular imaging of vascular inflammation, nuclear medicine constitutes one of the best imaging modalities, thanks to its high sensitivity for the detection of probes in tissues. 2-[18F]fluoro-2-deoxy-d-glucose ([18F]FDG) is currently the most widely used radiopharmaceutical for molecular imaging of vascular inflammatory diseases such as atherosclerosis and large-vessel vasculitis. The combination of [18F]FDG and positron emission tomography (PET) imaging has become a powerful tool to identify and monitor non-invasively inflammatory activities over time but suffers from several limitations including a lack of specificity and avid background in different localizations. The use of novel radiotracers may help to better understand the underlying pathophysiological processes and overcome some limitations of [18F]FDG PET for the imaging of vascular inflammation. This review examines how [18F]FDG PET has given us deeper insight into the role of inflammation in different vascular pathologies progression and discusses perspectives for alternative radiopharmaceuticals that could provide a more specific and simple identification of pathologies where vascular inflammation is implicated. Use of these novel PET tracers could lead to a better understanding of underlying disease mechanisms and help inform the identification and stratification of patients for newly emerging immune-modulatory therapies. Future research is needed to realize the true clinical translational value of PET imaging in vascular inflammatory diseases.
Collapse
Affiliation(s)
- Kevin Prigent
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
| | - Jonathan Vigne
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
- CHU de Caen Normandie, Department of Pharmacy, Normandie Université, UNICAEN, 14000 Caen, France
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, 14000 Caen, France
- Correspondence:
| |
Collapse
|
7
|
Wang C, Leach BI, Lister D, Adams SR, Xu H, Hoh C, McConville P, Zhang J, Messer K, Ahrens ET. Metallofluorocarbon Nanoemulsion for Inflammatory Macrophage Detection via PET and MRI. J Nucl Med 2021; 62:1146-1153. [PMID: 33277399 PMCID: PMC8833871 DOI: 10.2967/jnumed.120.255273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is associated with a range of serious human conditions, including autoimmune and cardiovascular diseases and cancer. The ability to image active inflammatory processes greatly enhances our ability to diagnose and treat these diseases at an early stage. We describe molecular compositions enabling sensitive and precise imaging of inflammatory hotspots in vivo. Methods: A functionalized nanoemulsion with a fluorocarbon-encapsulated radiometal chelate (FERM) was developed to serve as a platform for multimodal imaging probe development. The 19F-containing FERM nanoemulsion encapsulates 89Zr in the fluorous oil via a fluorinated hydroxamic acid chelate. Simple mixing of the radiometal with the preformed aqueous nanoemulsion before use yields FERM, a stable in vivo cell tracer, enabling whole-body 89Zr PET and 19F MRI after a single intravenous injection. Results: The FERM nanoemulsion was intrinsically taken up by phagocytic immune cells, particularly macrophages, with high specificity. FERM stability was demonstrated by a high correlation between the 19F and 89Zr content in the blood (correlation coefficient > 0.99). Image sensitivity at a low dose (37 kBq) was observed in a rodent model of acute infection. The versatility of FERM was further demonstrated in models of inflammatory bowel disease and 4T1 tumor. Conclusion: Multimodal detection using FERM yields robust whole-body lesion detection and leverages the strengths of combined PET and 19F MRI. The FERM nanoemulsion has scalable production and is potentially useful for precise diagnosis, stratification, and treatment monitoring of inflammatory diseases.
Collapse
Affiliation(s)
- Chao Wang
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Benjamin I Leach
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Deanne Lister
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Stephen R Adams
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Hongyan Xu
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Carl Hoh
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Patrick McConville
- Department of Radiology, University of California San Diego, La Jolla, California
- Invicro, Inc., Boston, Massachusetts; and
| | - Jing Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Karen Messer
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Eric T Ahrens
- Department of Radiology, University of California San Diego, La Jolla, California;
| |
Collapse
|
8
|
AlAshqar A, Reschke L, Kirschen GW, Borahay MA. Role of inflammation in benign gynecologic disorders: from pathogenesis to novel therapies†. Biol Reprod 2021; 105:7-31. [PMID: 33739368 PMCID: PMC8256101 DOI: 10.1093/biolre/ioab054] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/16/2022] Open
Abstract
Emerging evidence supports the notion that inflammation fosters the development of common benign gynecologic disorders, including uterine leiomyoma, endometriosis, and adenomyosis. Numerous cytokines, chemokines, and growth and transcription factors have indisputable roles in the establishment and maintenance of benign gynecologic disorders by initiating complex cascades that promote proliferation, angiogenesis, and lesion progression. The interaction between inflammation and benign gynecologic disorders is orchestrated by a plethora of factors, including sex steroids, genetics, epigenetics, extracellular matrix, stem cells, cardiometabolic risk factors, diet, vitamin D, and the immune system. The role of inflammation in these disorders is not limited to local pathobiology but also extends to involve clinical sequelae that range from those confined to the reproductive tract, such as infertility and gynecologic malignancies, to systemic complications such as cardiovascular disease. Enhanced understanding of the intricate mechanisms of this association will introduce us to unvisited pathophysiological perspectives and guide future diagnostic and therapeutic implications aimed at reducing the burden of these disorders. Utilization of inflammatory markers, microRNA, and molecular imaging as diagnostic adjuncts may be valuable, noninvasive techniques for prompt detection of benign gynecologic disorders. Further, use of novel as well as previously established therapeutics, such as immunomodulators, hormonal treatments, cardiometabolic medications, and cyclooxygenase-2 and NF-κB inhibitors, can target inflammatory pathways involved in their pathogenesis. In this comprehensive review, we aim to dissect the existing literature on the role of inflammation in benign gynecologic disorders, including the proposed underlying mechanisms and complex interactions, its contribution to clinical sequelae, and the clinical implications this role entails.
Collapse
Affiliation(s)
- Abdelrahman AlAshqar
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Kuwait University, Kuwait City, Kuwait
| | - Lauren Reschke
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| | - Gregory W Kirschen
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| | - Mostafa A Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
9
|
Yan X, Lin W, Liu H, Pu W, Li J, Wu P, Ding J, Luo G, Zhang J. Wavelength-Tunable, Long Lifetime, and Biocompatible Luminescent Nanoparticles Based on a Vitamin E-Derived Material for Inflammation and Tumor Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100045. [PMID: 34031977 DOI: 10.1002/smll.202100045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/09/2021] [Indexed: 06/12/2023]
Abstract
Luminescence imaging is one of the most effective noninvasive strategies for detection and stratification of inflammation and oxidative stress that are closely related to the pathogenesis of numerous acute and chronic diseases. Herein biocompatible nanoparticles based on a peroxalate ester derived from vitamin E (defined as OVE) are developed. In combination with different fluorophores, OVE can generate luminescence systems with emission wavelengths varying from blue to the near-infrared light in its native and nanoparticle forms, in the presence of hydrogen peroxide (H2 O2 ). The OVE-based nanoprobes exhibit high luminescence signals with extremely long lifetime, upon triggering by inflammatory conditions with abnormally elevated H2 O2 . Activated neutrophils and macrophages can be illuminated by this type of luminescent nanoprobes, with luminescence intensities positively correlated with inflammatory cell counts. In mouse models of peritonitis, alcoholic liver injury, drug-induced acute liver injury, and acute lung injury, the developed luminescence nanoprobes enable precision imaging of inflammation and disease progression. Moreover, tumors expressing a high level of H2 O2 can be shined. Importantly, the OVE-based nanoplatform shows excellent in vitro and in vivo biocompatibility.
Collapse
Affiliation(s)
- Xinhao Yan
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- College of Pharmacy and Medical Technology, Hanzhong Vocational and Technical College, Hanzhong, Shaanxi, 723000, China
| | - Wenjie Lin
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Huan Liu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Junhong Li
- College of Pharmacy and Medical Technology, Hanzhong Vocational and Technical College, Hanzhong, Shaanxi, 723000, China
| | - Peng Wu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- College of Pharmacy and Medical Technology, Hanzhong Vocational and Technical College, Hanzhong, Shaanxi, 723000, China
| | - Jun Ding
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Gaoxing Luo
- State Key Lab of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Lab of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
10
|
Zhang K, Sun Y, Wu S, Zhou M, Zhang X, Zhou R, Zhang T, Gao Y, Chen T, Chen Y, Yao X, Watanabe Y, Tian M, Zhang H. Systematic imaging in medicine: a comprehensive review. Eur J Nucl Med Mol Imaging 2021; 48:1736-1758. [PMID: 33210241 DOI: 10.1007/s00259-020-05107-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/08/2020] [Indexed: 01/05/2023]
Abstract
Systematic imaging can be broadly defined as the systematic identification and characterization of biological processes at multiple scales and levels. In contrast to "classical" diagnostic imaging, systematic imaging emphasizes on detecting the overall abnormalities including molecular, functional, and structural alterations occurring during disease course in a systematic manner, rather than just one aspect in a partial manner. Concomitant efforts including improvement of imaging instruments, development of novel imaging agents, and advancement of artificial intelligence are warranted for achievement of systematic imaging. It is undeniable that scientists and radiologists will play a predominant role in directing this burgeoning field. This article introduces several recent developments in imaging modalities and nanoparticles-based imaging agents, and discusses how systematic imaging can be achieved. In the near future, systematic imaging which combines multiple imaging modalities with multimodal imaging agents will pave a new avenue for comprehensive characterization of diseases, successful achievement of image-guided therapy, precise evaluation of therapeutic effects, and rapid development of novel pharmaceuticals, with the final goal of improving human health-related outcomes.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yujie Sun
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing, China
| | - Shuang Wu
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Min Zhou
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Tingting Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuanxue Gao
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Ting Chen
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yao Chen
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xin Yao
- Department of Gastroenterology, The First Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Mei Tian
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
- The College of Biomedical Engineering and Instrument Science of Zhejiang University, Hangzhou, China.
| |
Collapse
|
11
|
Regensburger AP, Brown E, Krönke G, Waldner MJ, Knieling F. Optoacoustic Imaging in Inflammation. Biomedicines 2021; 9:483. [PMID: 33924983 PMCID: PMC8145174 DOI: 10.3390/biomedicines9050483] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/11/2022] Open
Abstract
Optoacoustic or photoacoustic imaging (OAI/PAI) is a technology which enables non-invasive visualization of laser-illuminated tissue by the detection of acoustic signals. The combination of "light in" and "sound out" offers unprecedented scalability with a high penetration depth and resolution. The wide range of biomedical applications makes this technology a versatile tool for preclinical and clinical research. Particularly when imaging inflammation, the technology offers advantages over current clinical methods to diagnose, stage, and monitor physiological and pathophysiological processes. This review discusses the clinical perspective of using OAI in the context of imaging inflammation as well as in current and emerging translational applications.
Collapse
Affiliation(s)
- Adrian P. Regensburger
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Loschgestr. 15, D-91054 Erlangen, Germany;
| | - 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
| | - Gerhard Krönke
- Department of Medicine 3, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, D-91054 Erlangen, Germany;
| | - Maximilian J. Waldner
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, D-91054 Erlangen, Germany;
| | - Ferdinand Knieling
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Loschgestr. 15, D-91054 Erlangen, Germany;
| |
Collapse
|
12
|
Advancing Biomarker Development Through Convergent Engagement: Summary Report of the 2nd International Danube Symposium on Biomarker Development, Molecular Imaging and Applied Diagnostics; March 14-16, 2018; Vienna, Austria. Mol Imaging Biol 2021; 22:47-65. [PMID: 31049831 DOI: 10.1007/s11307-019-01361-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Here, we report on the outcome of the 2nd International Danube Symposium on advanced biomarker development that was held in Vienna, Austria, in early 2018. During the meeting, cross-speciality participants assessed critical aspects of non-invasive, quantitative biomarker development in view of the need to expand our understanding of disease mechanisms and the definition of appropriate strategies both for molecular diagnostics and personalised therapies. More specifically, panelists addressed the main topics, including the current status of disease characterisation by means of non-invasive imaging, histopathology and liquid biopsies as well as strategies of gaining new understanding of disease formation, modulation and plasticity to large-scale molecular imaging as well as integrative multi-platform approaches. Highlights of the 2018 meeting included dedicated sessions on non-invasive disease characterisation, development of disease and therapeutic tailored biomarkers, standardisation and quality measures in biospecimens, new therapeutic approaches and socio-economic challenges of biomarker developments. The scientific programme was accompanied by a roundtable discussion on identification and implementation of sustainable strategies to address the educational needs in the rapidly evolving field of molecular diagnostics. The central theme that emanated from the 2nd Donau Symposium was the importance of the conceptualisation and implementation of a convergent approach towards a disease characterisation beyond lesion-counting "lumpology" for a cost-effective and patient-centric diagnosis, therapy planning, guidance and monitoring. This involves a judicious choice of diagnostic means, the adoption of clinical decision support systems and, above all, a new way of communication involving all stakeholders across modalities and specialities. Moreover, complex diseases require a comprehensive diagnosis by converging parameters from different disciplines, which will finally yield to a precise therapeutic guidance and outcome prediction. While it is attractive to focus on technical advances alone, it is important to develop a patient-centric approach, thus asking "What can we do with our expertise to help patients?"
Collapse
|
13
|
Gordon O, Miller RJ, Thompson JM, Ordonez AA, Klunk MH, Dikeman DA, Joyce DP, Ruiz-Bedoya CA, Miller LS, Jain SK. Rabbit model of Staphylococcus aureus implant-associated spinal infection. Dis Model Mech 2020; 13:dmm.045385. [PMID: 32586832 PMCID: PMC7406311 DOI: 10.1242/dmm.045385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/16/2020] [Indexed: 12/22/2022] Open
Abstract
Post-surgical implant-associated spinal infection is a devastating complication commonly caused by Staphylococcus aureus. Biofilm formation is thought to reduce penetration of antibiotics and immune cells, contributing to chronic and difficult-to-treat infections. A rabbit model of a posterior-approach spinal surgery was created, in which bilateral titanium pedicle screws were interconnected by a plate at the level of lumbar vertebra L6 and inoculated with a methicillin-resistant S.aureus (MRSA) bioluminescent strain. In vivo whole-animal bioluminescence imaging (BLI) and ex vivo bacterial cultures demonstrated a peak in bacterial burden by day 14, when wound dehiscence occurred. Structures suggestive of biofilm, visualized by scanning electron microscopy, were evident up to 56 days following infection. Infection-induced inflammation and bone remodeling were also monitored using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) and computed tomography (CT). PET imaging signals were noted in the soft tissue and bone surrounding the implanted materials. CT imaging demonstrated marked bone remodeling and a decrease in dense bone at the infection sites. This rabbit model of implant-associated spinal infection provides a valuable preclinical in vivo approach to investigate the pathogenesis of implant-associated spinal infections and to evaluate novel therapeutics. Summary: A model of post-surgical methicillin-resistant Staphylococcus aureus implant-associated spinal infection was created in rabbits, recapitulating acute infection as well as chronic low-burden infection, with structures suggestive of biofilm formation and bone remodeling.
Collapse
Affiliation(s)
- Oren Gordon
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - John M Thompson
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alvaro A Ordonez
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mariah H Klunk
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dustin A Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel P Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Camilo A Ruiz-Bedoya
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Immunology, Janssen Research and Development, Spring House, PA 19477, USA
| | - Sanjay K Jain
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA .,Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
14
|
Methods of Granulocyte Isolation from Human Blood and Labeling with Multimodal Superparamagnetic Iron Oxide Nanoparticles. Molecules 2020; 25:molecules25040765. [PMID: 32053865 PMCID: PMC7070653 DOI: 10.3390/molecules25040765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/17/2020] [Accepted: 02/02/2020] [Indexed: 02/08/2023] Open
Abstract
This in vitro study aimed to find the best method of granulocyte isolation for subsequent labeling with multimodal nanoparticles (magnetic and fluorescent properties) to enable detection by optical and magnetic resonance imaging (MRI) techniques. The granulocytes were obtained from venous blood samples from 12 healthy volunteers. To achieve high purity and yield, four different methods of granulocyte isolation were evaluated. The isolated granulocytes were labeled with multimodal superparamagnetic iron oxide nanoparticles (M-SPIONs) coated with dextran, and the iron load was evaluated qualitatively and quantitatively by MRI, near-infrared fluorescence (NIRF) and inductively coupled plasma mass spectrometry (ICP-MS). The best method of granulocyte isolation was Percoll with Ficoll, which showed 95.92% purity and 94% viability. After labeling with M-SPIONs, the granulocytes showed 98.0% purity with a yield of 3.5 × 106 cells/mL and more than 98.6% viability. The iron-loading value in the labeled granulocytes, as obtained by MRI, was 6.40 ± 0.18 pg/cell. Similar values were found with the ICP-MS and NIRF imaging techniques. Therefore, our study shows that it is possible to isolate granulocytes with high purity and yield and labeling with M-SPIONs provides a high internalized iron load and low toxicity to cells. Therefore, these M-SPION-labeled granulocytes could be a promising candidate for future use in inflammation/infection detection by optical and MRI techniques.
Collapse
|
15
|
Abstract
Molecular imaging is an emerging technology that enables the noninvasive visualization, characterization, and quantification of molecular events within living subjects. Positron emission tomography (PET) is a clinically available molecular imaging tool with significant potential to study pathogenesis of infections in humans. Molecular imaging is an emerging technology that enables the noninvasive visualization, characterization, and quantification of molecular events within living subjects. Positron emission tomography (PET) is a clinically available molecular imaging tool with significant potential to study pathogenesis of infections in humans. PET enables dynamic assessment of infectious processes within the same subject with high temporal and spatial resolution and obviates the need for invasive tissue sampling, which is difficult in patients and generally limited to a single time point, even in animal models. This review presents current state-of-the-art concepts on the application of molecular imaging for infectious diseases and details how PET imaging can facilitate novel insights into infectious processes, ongoing development of pathogen-specific imaging, and simultaneous in situ measurements of intralesional antimicrobial pharmacokinetics in multiple compartments, including privileged sites. Finally, the potential clinical applications of this promising technology are also discussed.
Collapse
|
16
|
Perkins LA, Anderson CJ, Novelli EM. Targeting P-Selectin Adhesion Molecule in Molecular Imaging: P-Selectin Expression as a Valuable Imaging Biomarker of Inflammation in Cardiovascular Disease. J Nucl Med 2019; 60:1691-1697. [PMID: 31601694 DOI: 10.2967/jnumed.118.225169] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
P-selectin is an adhesion molecule translocated to the surface of endothelial cells and platelets under inflammatory stimuli, and its potential as a biomarker in inflammatory conditions has driven preclinical studies to investigate its application for molecular imaging of inflammation. Clinical imaging of P-selectin expression for disease characterization could have an important role in stratifying patients and determining treatment strategies. The objective of this review is to outline the role of P-selectin in cardiovascular inflammatory conditions and its translation as an early inflammatory biomarker for several molecular imaging modalities for diagnostic purposes and therapeutic planning.
Collapse
Affiliation(s)
- Lydia A Perkins
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carolyn J Anderson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Enrico M Novelli
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
Han SS, Jin Z, Lee BS, Han JS, Choi JJ, Park SJ, Chung HM, Mukhtar AS, Moon SH, Kang SW. Reproducible hindlimb ischemia model based on photochemically induced thrombosis to evaluate angiogenic effects. Microvasc Res 2019; 126:103912. [PMID: 31433972 DOI: 10.1016/j.mvr.2019.103912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/28/2019] [Accepted: 08/17/2019] [Indexed: 01/04/2023]
Abstract
Critical limb ischemia is one of the most common types of peripheral arterial disease. Preclinical development of ischemia therapeutics relies on the availability of a relevant and reproducible in vivo disease model. Thus, establishing appropriate animal disease models is essential for the development of new therapeutic strategies. Currently, the most commonly employed model of hindlimb ischemia is the surgical induction method with ligation of the femoral artery and its branches after skin incision. However, the efficiency of the method is highly variable depending on the availability of skilled technicians. In addition, after surgical procedures, animals can quickly and spontaneously recover from damage, limiting observations of the therapeutic effect of potential agents. The aim of this study was to develop a hindlimb ischemia mouse model with similarities to human ischemic disease. To that end, a photochemical reaction was used to induce thrombosis in the hindlimb. After the photochemical reaction was induced by light irradiation, thrombotic plugs and adjacent red blood cell stasis were observed in hindlimb vessels in the light-irradiated zone. Additionally, the photochemically induced thrombosis maintained the ischemic condition and did not cause notable side effects in mice.
Collapse
Affiliation(s)
- Sang-Soo Han
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, Republic of Korea; Applied Bioresources Research Division, Freshwater Bioresources Utilization Bureau, Nakdonggang National Institute of Biological Resource (NNIBR), Sangju, Republic of Korea
| | - Zhen Jin
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Byoung-Seok Lee
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Ji-Seok Han
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Jong-Jin Choi
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Soon-Jung Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | | | - Sung-Hwan Moon
- Department of Medicine, School of Medicine, Konkuk University, Seoul, Republic of Korea; Research Institute, T&R Biofab Co. Ltd, 237, Siheung 15073, Republic of Korea.
| | - Sun-Woong Kang
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, Republic of Korea; Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
18
|
Sinharay S, Tu TW, Kovacs ZI, Schreiber-Stainthorp W, Sundby M, Zhang X, Papadakis GZ, Reid WC, Frank JA, Hammoud DA. In vivo imaging of sterile microglial activation in rat brain after disrupting the blood-brain barrier with pulsed focused ultrasound: [18F]DPA-714 PET study. J Neuroinflammation 2019; 16:155. [PMID: 31345243 PMCID: PMC6657093 DOI: 10.1186/s12974-019-1543-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 12/26/2022] Open
Abstract
Background Magnetic resonance imaging (MRI)-guided pulsed focused ultrasound combined with the infusion of microbubbles (pFUS+MB) induces transient blood-brain barrier opening (BBBO) in targeted regions. pFUS+MB, through the facilitation of neurotherapeutics’ delivery, has been advocated as an adjuvant treatment for neurodegenerative diseases and malignancies. Sterile neuroinflammation has been recently described following pFUS+MB BBBO. In this study, we used PET imaging with [18F]-DPA714, a biomarker of translocator protein (TSPO), to assess for neuroinflammatory changes following single and multiple pFUS+MB sessions. Methods Three groups of Sprague-Dawley female rats received MRI-guided pFUS+MB (Optison™; 5–8 × 107 MB/rat) treatments to the left frontal cortex and right hippocampus. Group A rats were sonicated once. Group B rats were sonicated twice and group C rats were sonicated six times on weekly basis. Passive cavitation detection feedback (PCD) controlled the peak negative pressure during sonication. We performed T1-weighted scans immediately after sonication to assess efficiency of BBBO and T2*-weighted scans to evaluate for hypointense voxels. [18F]DPA-714 PET/CT scans were acquired after the BBB had closed, 24 h after sonication in group A and within an average of 10 days from the last sonication in groups B and C. Ratios of T1 enhancement, T2* values, and [18F]DPA-714 percent injected dose/cc (%ID/cc) values in the targeted areas to the contralateral brain were calculated. Histological assessment for microglial activation/astrocytosis was performed. Results In all groups, [18F]DPA-714 binding was increased at the sonicated compared to non-sonicated brain (%ID/cc ratios > 1). Immunohistopathology showed increased staining for microglial and astrocytic markers in the sonicated frontal cortex compared to contralateral brain and to a lesser extent in the sonicated hippocampus. Using MRI, we documented BBB disruption immediately after sonication with resolution of BBBO 24 h later. We found more T2* hypointense voxels with increasing number of sonications. In a longitudinal group of animals imaged after two and after six sonications, there was no cumulative increase of neuroinflammation on PET. Conclusion Using [18F]DPA-714 PET, we documented in vivo neuroinflammatory changes in association with pFUS+MB. Our protocol (utilizing PCD feedback to minimize damage) resulted in neuroinflammation visualized 24 h post one sonication. Our findings were supported by immunohistochemistry showing microglial activation and astrocytosis. Experimental sonication parameters intended for BBB disruption should be evaluated for neuroinflammatory sequelae prior to implementation in clinical trials. Electronic supplementary material The online version of this article (10.1186/s12974-019-1543-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanhita Sinharay
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.,University of Texas, MD Anderson Cancer Center, Houston, USA
| | - Tsang-Wei Tu
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology, Howard University, Washington DC, USA
| | - Zsofia I Kovacs
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Institute for Biomedical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - William Schreiber-Stainthorp
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Xiang Zhang
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Georgios Z Papadakis
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.,Department of Radiology, University of Crete and Department of Medical Imaging Heraklion University Hospital, Crete, Greece
| | - William C Reid
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Dima A Hammoud
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.
| |
Collapse
|
19
|
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: 60] [Impact Index Per Article: 10.0] [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
|
20
|
The beginning of the end for conventional RECIST - novel therapies require novel imaging approaches. Nat Rev Clin Oncol 2019; 16:442-458. [PMID: 30718844 DOI: 10.1038/s41571-019-0169-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Owing to improvements in our understanding of the biological principles of tumour initiation and progression, a wide variety of novel targeted therapies have been developed. Developments in biomedical imaging, however, have not kept pace with these improvements and are still mainly designed to determine lesion size alone, which is reflected in the Response Evaluation Criteria in Solid Tumors (RECIST). Imaging approaches currently used for the evaluation of treatment responses in patients with solid tumours, therefore, often fail to detect successful responses to novel targeted agents and might even falsely suggest disease progression, a scenario known as pseudoprogression. The ability to differentiate between responders and nonresponders early in the course of treatment is essential to allowing the early adjustment of treatment regimens. Various imaging approaches targeting a single dedicated tumour feature, as described in the hallmarks of cancer, have been successful in preclinical investigations, and some have been evaluated in pilot clinical trials. However, these approaches have largely not been implemented in clinical practice. In this Review, we describe current biomedical imaging approaches used to monitor responses to treatment in patients receiving novel targeted therapies, including a summary of the most promising future approaches and how these might improve clinical practice.
Collapse
|
21
|
Wang C, Keliher E, Zeller MWG, Wojtkiewicz GR, Aguirre AD, Buckbinder L, Kim HY, Chen J, Maresca K, Ahmed MS, Motlagh NJ, Nahrendorf M, Chen JW. An activatable PET imaging radioprobe is a dynamic reporter of myeloperoxidase activity in vivo. Proc Natl Acad Sci U S A 2019; 116:11966-11971. [PMID: 31123149 PMCID: PMC6575581 DOI: 10.1073/pnas.1818434116] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Myeloperoxidase (MPO) is a critical proinflammatory enzyme implicated in cardiovascular, neurological, and rheumatological diseases. Emerging therapies targeting inflammation have raised interest in tracking MPO activity in patients. We describe 18F-MAPP, an activatable MPO activity radioprobe for positron emission tomography (PET) imaging. The activated radioprobe binds to proteins and accumulates at sites of MPO activity. The radioprobe 18F-MAPP has a short blood half-life, remains stable in plasma, does not demonstrate cytotoxicity, and crosses the intact blood-brain barrier. The 18F-MAPP imaging detected sites of elevated MPO activity in living mice embedded with human MPO and in mice induced with chemical inflammation or myocardial infarction. The 18F-MAPP PET imaging noninvasively differentiated varying amounts of MPO activity, competitive inhibition, and MPO deficiency in living animals, confirming specificity and showing that the radioprobe can quantify changes in in vivo MPO activity. The radiosynthesis has been optimized and automated, an important step in translation. These data indicate that 18F-MAPP is a promising translational candidate to noninvasively monitor MPO activity and inflammation in patients.
Collapse
Affiliation(s)
- Cuihua Wang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Edmund Keliher
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Matthias W G Zeller
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Gregory R Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Aaron D Aguirre
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | | | - Hye-Yeong Kim
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Jianqing Chen
- Pfizer World Wide Research and Development, Cambridge, MA 02139
| | - Kevin Maresca
- Pfizer World Wide Research and Development, Cambridge, MA 02139
| | - Maaz S Ahmed
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Negin Jalali Motlagh
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - John W Chen
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114;
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114
| |
Collapse
|
22
|
Wang H, Jordan VC, Ramsay IA, Sojoodi M, Fuchs BC, Tanabe KK, Caravan P, Gale EM. Molecular Magnetic Resonance Imaging Using a Redox-Active Iron Complex. J Am Chem Soc 2019; 141:5916-5925. [PMID: 30874437 PMCID: PMC6726119 DOI: 10.1021/jacs.9b00603] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We introduce a redox-active iron complex, Fe-PyC3A, as a biochemically responsive MRI contrast agent. Switching between Fe3+-PyC3A and Fe2+-PyC3A yields a full order of magnitude relaxivity change that is field-independent between 1.4 and 11.7 T. The oxidation of Fe2+-PyC3A to Fe3+-PyC3A by hydrogen peroxide is very rapid, and we capitalized on this behavior for the molecular imaging of acute inflammation, which is characterized by elevated levels of reactive oxygen species. Injection of Fe2+-PyC3A generates strong, selective contrast enhancement of inflamed pancreatic tissue in a mouse model (caerulein/LPS model). No significant signal enhancement is observed in normal pancreatic tissue (saline-treated mice). Importantly, signal enhancement of the inflamed pancreas correlates strongly and significantly with ex vivo quantitation of the pro-inflammatory biomarker myeloperoxidase. This is the first example of using metal ion redox for the MR imaging of pathologic change in vivo. Redox-active Fe3+/2+ complexes represent a new design paradigm for biochemically responsive MRI contrast agents.
Collapse
Affiliation(s)
- Huan Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, 149 Thirteenth Street, Charlestown, Massachusetts 02129, United States
| | - Veronica Clavijo Jordan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, 149 Thirteenth Street, Charlestown, Massachusetts 02129, United States
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Ian A. Ramsay
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, 149 Thirteenth Street, Charlestown, Massachusetts 02129, United States
| | - Mozhdeh Sojoodi
- Division of Surgical Oncology, Massachusetts General Hospital/Harvard Medical School, WRN401, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Bryan C. Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital/Harvard Medical School, WRN401, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Kenneth K. Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital/Harvard Medical School, WRN401, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, 149 Thirteenth Street, Charlestown, Massachusetts 02129, United States
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Eric M. Gale
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, 149 Thirteenth Street, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
23
|
Cao Q, Huang Q, Mohan C, Li C. Small-Animal PET/CT Imaging of Local and Systemic Immune Response Using 64Cu-αCD11b. J Nucl Med 2019; 60:1317-1324. [PMID: 30796172 DOI: 10.2967/jnumed.118.220350] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023] Open
Abstract
Current noninvasive imaging methods for monitoring immune response were largely developed for interrogation of the local reaction. This study developed the radiotracer 64Cu-labeled anti-CD11b (64Cu-αCD11b) for longitudinal assessment of local and systemic immune response involving mobilization of CD11b+ myeloid cells by small-animal PET/CT. Methods: Acute or chronic inflammation in the ears of BALB/c mice was induced by 12-o-tetradecanoylphorbol-13-acetate. Acute lung inflammation was induced by intratracheal lipopolysaccharide inoculation. αCD11b was conjugated with p-SCN-Bn-DOTA followed by labeling with 64Cu. PET/CT and biodistribution were evaluated at different times after intravenous injection of 64Cu-αCD11b. Cell populations from bone marrow (BM) and spleen were analyzed by flow cytometry. Results: 64Cu-αCD11b was primarily taken up by BM and spleen in control mice. In comparison, 64Cu-αCD11b uptake was significantly reduced in the BM and spleen of CD11b-knockout mice, indicating that 64Cu-αCD11b selectively homed to CD11b+ myeloid cells in vivo. In mice with ear inflammation, for the local inflammatory response, 64Cu-αCD11b PET/CT revealed significantly higher 64Cu-αCD11b uptake in the inflamed ears in the acute inflammation phase than the chronic phase, consistent with markedly increased infiltration of CD11b+ cells into the inflammatory lesions at the acute phase. Moreover, imaging of 64Cu-αCD11b also showed the difference in mouse systemic response for different inflammatory stages. Compared with uptake in control mice, BM 64Cu-αCD11b uptake in mice with ear inflammation was significantly lower in the acute phase and higher in the chronic phase, reflecting an initial mobilization of CD11b+ cells from the BM to the inflammatory foci followed by a compensatory regeneration of CD11b+ myeloid cells in the BM. Similarly, in mice with lung inflammation, 64Cu-αCD11b PET/CT readily detected acute lung inflammation and recruitment of CD11b+ myeloid cells from the BM. Immunohistochemistry staining and flow cytometry results confirmed the noninvasive imaging of PET/CT. Conclusion: 64Cu-αCD11b PET/CT successfully tracked ear and pulmonary inflammation in mice and differentiated acute from chronic inflammation at the local and systemic levels. 64Cu-αCD11b PET/CT is a robust quantitative method for imaging of local and systemic immune responses.
Collapse
Affiliation(s)
- Qizhen Cao
- Department of Cancer Systems Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; and
| | - Qian Huang
- Department of Cancer Systems Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; and
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, Texas
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; and
| |
Collapse
|
24
|
A Non-Peptidic S100A9 Specific Ligand for Optical Imaging of Phagocyte Activity In Vivo. Mol Imaging Biol 2019; 20:407-416. [PMID: 29185197 DOI: 10.1007/s11307-017-1148-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Non-invasive assessment of inflammatory activity in the course of various diseases is a largely unmet clinical challenge. An early feature of inflammation is local secretion of the alarmin S100A8/A9 by activated phagocytes. We here evaluate a novel S100A9-targeted small molecule tracer Cy5.5-CES271 for in vivo optical imaging of inflammatory activity in exemplary disease models. PROCEDURES Dynamics of Cy5.5-CES271 was characterized in a model of irritant contact dermatitis by sequential fluorescence reflectance imaging (FRI) up to 24 h postinjection (p.i.). Specificity of Cy5.5-CES271 binding to S100A9 in vivo was examined by blocking studies and by employing S100A9-/- mice. Finally, S100A9 secretion in acute lung inflammation was assessed by Cy5.5-CES271 and FRI of explanted lungs. RESULTS In ear inflammation, we were able to non-invasively follow the time course of S100A9 expression using Cy5.5-CES271 and FRI over 24 h p.i. (peak activity at 3 h p.i.). Specificity of imaging could be shown by a significant signal reduction after predosing and using S100A9-/- mice. In acute lung injury, local and systemic S100A8/A9 levels increased over time and correlated significantly with FRI signal levels in explanted lungs. CONCLUSIONS Cy5.5-CES271 shows significant accumulation in models of inflammatory diseases and specific binding to S100A9 in vivo. This study, for the first time, demonstrates the potential of a small molecule non-peptidic tracer enabling imaging of S100A9 as a marker of local phagocyte activity in inflammatory scenarios suggesting this compound class for translational attempts.
Collapse
|
25
|
Dual-Modal In Vivo Fluorescence/Photoacoustic Microscopy Imaging of Inflammation Induced by GFP-Expressing Bacteria. SENSORS 2019; 19:s19020238. [PMID: 30634579 PMCID: PMC6359259 DOI: 10.3390/s19020238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/10/2018] [Accepted: 12/10/2018] [Indexed: 01/05/2023]
Abstract
In this study, dual-modal fluorescence and photoacoustic microscopy was performed for noninvasive and functional in vivo imaging of inflammation induced by green fluorescent protein (GFP) transfected bacteria in mice ear. Our imaging results demonstrated that the multimodal imaging technique is able to monitor the tissue immunovascular responses to infections with molecular specificity. Our study also indicated that the combination of photoacoustic and fluorescence microscopy imaging can simultaneously track the biochemical changes including the bacterial distribution and morphological change of blood vessels in the biological tissues with high resolution and enhanced sensitivity. Consequently, the developed method paves a new avenue for improving the understanding of the pathology mechanism of inflammation.
Collapse
|
26
|
Xu X, An H, Zhang D, Tao H, Dou Y, Li X, Huang J, Zhang J. A self-illuminating nanoparticle for inflammation imaging and cancer therapy. SCIENCE ADVANCES 2019; 5:eaat2953. [PMID: 30662940 PMCID: PMC6326751 DOI: 10.1126/sciadv.aat2953] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 12/04/2018] [Indexed: 05/05/2023]
Abstract
Nanoparticles have been extensively used for inflammation imaging and photodynamic therapy of cancer. However, the major translational barriers to most nanoparticle-based imaging and therapy applications are the limited depth of tissue penetration, inevitable requirement of external irradiation, and poor biocompatibility of the nanoparticles. To overcome these critical limitations, we synthesized a sensitive, specific, biodegradable luminescent nanoparticle that is self-assembled from an amphiphilic polymeric conjugate with a luminescent donor (luminol) and a fluorescent acceptor [chlorin e6 (Ce6)] for in vivo luminescence imaging and photodynamic therapy in deep tissues. Mechanistically, reactive oxygen species (ROS) and myeloperoxidase generated in inflammatory sites or the tumor microenvironment trigger bioluminescence resonance energy transfer and the production of singlet oxygen (1O2) from the nanoparticle, enabling in vivo imaging and cancer therapy, respectively. This self-illuminating nanoparticle shows an excellent in vivo imaging capability with suitable tissue penetration and resolution in diverse animal models of inflammation. It is also proven to be a selective, potent, and safe antitumor nanomedicine that specifically kills cancer cells via in situ 1O2 produced in the tumor microenvironment, which contains a high level of ROS.
Collapse
Affiliation(s)
- Xiaoqiu Xu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Huijie An
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Third Military Medical University, Chongqing 400038, China
| | - Hui Tao
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Yin Dou
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Xiaohui Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Jun Huang
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Corresponding author.
| |
Collapse
|
27
|
Hanly JG, Kozora E, Beyea SD, Birnbaum J. Review: Nervous System Disease in Systemic Lupus Erythematosus: Current Status and Future Directions. Arthritis Rheumatol 2018; 71:33-42. [PMID: 29927108 DOI: 10.1002/art.40591] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 06/19/2018] [Indexed: 12/17/2022]
Abstract
The American College of Rheumatology's case definitions for 19 neuropsychiatric syndromes in systemic lupus erythematosus (SLE) constitute a comprehensive classification of nervous system events in this disease. However, additional strategies are needed to determine whether a neuropsychiatric syndrome is attributable to SLE versus a competing comorbidity. Cognitive function is a clinical surrogate of overall brain health, with applications in both diagnosis and determination of clinical outcomes. Ischemic and inflammatory mechanisms are both key components of the immunopathogenesis of neuropsychiatric SLE (NPSLE), including abnormalities of the blood-brain barrier and autoantibody-mediated production of proinflammatory cytokines. Advances in neuroimaging provide a platform to assess novel disease mechanisms in a noninvasive way. The convergence of more rigorous clinical characterization, validation of biomarkers, and brain neuroimaging provides opportunities to determine the efficacy of novel targeted therapies in the treatment of NPSLE.
Collapse
Affiliation(s)
- John G Hanly
- Queen Elizabeth II Health Sciences Centre and Dalhousie University, Halifax, Nova Scotia, Canada
| | - Elizabeth Kozora
- National Jewish Health, Denver, Colorado, and University of Colorado School of Medicine, Aurora
| | - Steven D Beyea
- Dalhousie University, Biomedical Translational Imaging Centre, Izaak Walton Killam Health Centre and Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
| | - Julius Birnbaum
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
28
|
Helfen A, Roth J, Ng T, Eisenblaetter M. In Vivo Imaging of Pro- and Antitumoral Cellular Components of the Tumor Microenvironment. J Nucl Med 2018; 59:183-188. [PMID: 29217734 DOI: 10.2967/jnumed.117.198952] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022] Open
Abstract
Tumor development and growth, as well as metastatic spread, are strongly influenced by various, mostly innate, immune cells, which are recruited to the tumor site and driven to establish a specific tumor-supportive microenvironment. The contents of this microenvironment, such as myeloid cells, are a major factor in the overall prognosis of malignant disease, addressed by a constantly growing armament of therapeutic interventions targeting tumor-supportive immune cells. Current clinical imaging has long ignored the growing need for diagnostic approaches addressing these microenvironmental contents-approaches enabling a sensitive and specific classification of tumor immune crosstalk and the resulting tumor-associated immune cell activity. In this focus article we review the present status of, and promising developments in, the in vivo molecular imaging of tumor immune components designed to allow for inferences to be made on the cross-talk between tumor cells and the immune system. Current imaging modalities based on the infiltrating cell types are briefly discussed.
Collapse
Affiliation(s)
- Anne Helfen
- Department of Clinical Radiology, University Hospital Muenster, Muenster, Germany
| | - Johannes Roth
- Institute of Immunology, University Hospital Muenster, Muenster, Germany
| | - Tony Ng
- Richard Dimbleby Department of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
- Breast Cancer Now Research Unit, Department of Research Oncology, Guy's Hospital, King's College London, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom; and
| | - Michel Eisenblaetter
- Department of Clinical Radiology, University Hospital Muenster, Muenster, Germany
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
29
|
Exploring Alternative Radiolabeling Strategies for Sialic Acid-Binding Immunoglobulin-Like Lectin 9 Peptide: [ 68Ga]Ga- and [ 18F]AlF-NOTA-Siglec-9. Molecules 2018; 23:molecules23020305. [PMID: 29385091 PMCID: PMC6017478 DOI: 10.3390/molecules23020305] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022] Open
Abstract
Amino acid residues 283–297 from sialic acid-binding immunoglobulin-like lectin 9 (Siglec-9) form a cyclic peptide ligand targeting vascular adhesion protein-1 (VAP-1). VAP-1 is associated with the transfer of leukocytes from blood to tissues upon inflammation. Therefore, analogs of Siglec-9 peptide are good candidates for visualizing inflammation non-invasively using positron emission tomography (PET). Gallium-68-labeled 1,4,7,10-tetraazacyclododecane-N,N′,N″,N‴-tetraacetic acid (DOTA)-conjugated Siglec-9 has been evaluated extensively for this purpose. Here, we explored two alternative strategies for radiolabeling Siglec-9 peptide using a 1,4,7-triazacyclononane-triacetic acid (NOTA)-chelator to bind [68Ga]Ga or [18F]AlF. The radioligands were evaluated by in vivo PET imaging and ex vivo γ-counting of turpentine-induced sterile skin/muscle inflammation in Sprague-Dawley rats. Both tracers showed clear accumulation in the inflamed tissues. The whole-body biodistribution patterns of the tracers were similar.
Collapse
|
30
|
Prospective of 68Ga Radionuclide Contribution to the Development of Imaging Agents for Infection and Inflammation. CONTRAST MEDIA & MOLECULAR IMAGING 2018. [PMID: 29531507 PMCID: PMC5817300 DOI: 10.1155/2018/9713691] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the last decade, the utilization of 68Ga for the development of imaging agents has increased considerably with the leading position in the oncology. The imaging of infection and inflammation is lagging despite strong unmet medical needs. This review presents the potential routes for the development of 68Ga-based agents for the imaging and quantification of infection and inflammation in various diseases and connection of the diagnosis to the treatment for the individualized patient management.
Collapse
|
31
|
Affiliation(s)
- Greg M Pontone
- a Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,b Department of Neurology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Kelly A Mills
- b Department of Neurology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Gwenn S Smith
- a Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,c Russell H. Morgan Department of Radiology and Radiological Sciences , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
32
|
Anderson CJ, Lewis JS. Current status and future challenges for molecular imaging. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2017; 375:rsta.2017.0023. [PMID: 29038378 DOI: 10.1098/rsta.2017.0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
Molecular imaging (MI), used in its wider sense of biology at the molecular level, is a field that lies at the intersection of molecular biology and traditional medical imaging. As advances in medicine have exponentially expanded over the last few decades, so has our need to better understand the fundamental behaviour of living organisms in a non-invasive and timely manner. This commentary draws from topics the authors addressed in their presentations at the 2017 Royal Society Meeting 'Challenges for chemistry in molecular imaging', as well as a discussion of where MI is today and where it is heading in the future.This article is part of the themed issue 'Challenges for chemistry in molecular imaging'.
Collapse
Affiliation(s)
- Carolyn J Anderson
- Departments of Medicine, Radiology, Bioengineering, and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jason S Lewis
- Department of Radiology and the Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
33
|
Karachaliou CE, Triantis C, Liolios C, Palamaris L, Zikos C, Tsitsilonis OE, Kalbacher H, Voelter W, Loudos G, Papadopoulos M, Pirmettis I, Livaniou E. In vivo biodistribution and imaging studies with a 99mTc-radiolabeled derivative of the C-terminus of prothymosin alpha in mice bearing experimentally-induced inflammation. Eur J Pharm Biopharm 2017; 113:188-197. [PMID: 28087377 DOI: 10.1016/j.ejpb.2016.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/15/2016] [Accepted: 12/14/2016] [Indexed: 10/20/2022]
Abstract
Prothymosin alpha (ProTα) is a highly conserved mammalian polypeptide (109 amino acids in man) exerting in vitro and in vivo immunoenhancing activities. Recently, our team has developed a 99mTc-radiolabeled derivative of the C-terminal bioactive decapeptide of ProTα ([99mTc]C1) and employed it in in vitro studies, the results of which support the existence of binding sites on human neutrophils that recognize [99mTc]C1, intact ProTα as well as the C-terminal decapeptide of ProTα and presumably involve Toll-like receptor 4. In the present work, [99mTc]C1 was administered to Swiss albino mice with experimentally-induced inflammation for in vivo biodistribution and imaging studies, in parallel with a suitable negative control, which differs from [99mTc]C1 only in bearing a scrambled version of the ProTα decapeptide. The biodistribution data obtained with [99mTc]C1 demonstrated fast clearance of radioactivity from blood, heart, lungs, normal muscle, and predominantly urinary excretion. Most importantly, slow clearance of radioactivity from the inflammation focus was observed, resulting in a high ratio of inflamed/normal muscle tissue (9.15 at 30min post injection, which remained practically stable up to 2h). The inflammation-targeting capacity of [99mTc]C1 was confirmed by imaging studies and might be attributed to neutrophils, which are recruited at the inflamed areas and bear binding sites for [99mTc]C1. In this respect, apart from being a valuable tool for further studies on ProTα in in vitro and in vivo systems, [99mTc]C1 merits further evaluation as a radiopharmaceutical for specific imaging of inflammation foci.
Collapse
Affiliation(s)
- Chrysoula-Evangelia Karachaliou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Charalampos Triantis
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Christos Liolios
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Lazaros Palamaris
- Department of Medical Instruments Technology, Technological Educational Institute, Athens 12243, Greece
| | - Christos Zikos
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Ourania E Tsitsilonis
- Division of Animal and Human Physiology, Department of Biology, University of Athens, Athens 15784, Greece
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen 72076, Germany
| | - Wolfgang Voelter
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen 72076, Germany
| | - George Loudos
- Department of Medical Instruments Technology, Technological Educational Institute, Athens 12243, Greece
| | - Minas Papadopoulos
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Evangelia Livaniou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece.
| |
Collapse
|
34
|
Zhang X, Liu F, Slikker W, Wang C, Paule MG. Minimally invasive biomarkers of general anesthetic-induced developmental neurotoxicity. Neurotoxicol Teratol 2016; 60:95-101. [PMID: 27784630 DOI: 10.1016/j.ntt.2016.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/29/2016] [Accepted: 10/21/2016] [Indexed: 12/22/2022]
Abstract
The association of general anesthesia with developmental neurotoxicity, while nearly impossible to study in pediatric populations, is clearly demonstrable in a variety of animal models from rodents to nonhuman primates. Nearly all general anesthetics tested have been shown to cause abnormal brain cell death in animals when administered during periods of rapid brain growth. The ability to repeatedly assess in the same subjects adverse effects induced by general anesthetics provides significant power to address the time course of important events associated with exposures. Minimally-invasive procedures provide the opportunity to bridge the preclinical/clinical gap by providing the means to more easily translate findings from the animal laboratory to the human clinic. Positron Emission Tomography or PET is a tool with great promise for realizing this goal. PET for small animals (microPET) is providing valuable data on the life cycle of general anesthetic induced neurotoxicity. PET radioligands (annexin V and DFNSH) targeting apoptotic processes have demonstrated that a single bout of general anesthesia effected during a vulnerable period of CNS development can result in prolonged apoptotic signals lasting for several weeks in the rat. A marker of cellular proliferation (FLT) has demonstrated in rodents that general anesthesia-induced inhibition of neural progenitor cell proliferation is evident when assessed a full 2weeks after exposure. Activated glia express Translocator Protein (TSPO) which can be used as a marker of presumed neuroinflammatory processes and a PET ligand for the TSPO (FEPPA) has been used to track this process in both rat and nonhuman primate models. It has been shown that single bouts of general anesthesia can result in elevated TSPO expression lasting for over a week. These examples demonstrate the utility of specific PET tracers to inform, in a minimally-invasive fashion, processes associated with general anesthesia-induced developmental neurotoxicity. The fact that PET procedures are also used clinically suggests an opportunity to confirm in humans what has been repeatedly observed in animals.
Collapse
|