1
|
Yang X, Hu J, Gao Q, Deng Y, Liu Y, He X, Li C, Yu X, Wan Y, Pi C, Wei Y, Li C. Advances in nano-delivery systems based on diagnosis and theranostics strategy for atherosclerosis. J Drug Target 2025; 33:492-507. [PMID: 39601425 DOI: 10.1080/1061186x.2024.2433560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/21/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerosis (AS) is a chronic systemic inflammatory disease, where early diagnosis and theranostics strategy for AS are crucial for improving outcomes. However, conventional diagnostic techniques are limited in identifying early AS lesions, failing to stop the progression of AS in time. Nano-delivery systems have shown significant potential in AS diagnosis and treatment, offering distinct advantages in plaque identification and enhancing drugs concentration at lesion sites, thereby advancing new-generation theranostics strategy. This review discusses the application of nano-delivery systems based on imaging technology in AS diagnosis, and we further explore recent advancements in combining different imaging technologies with emerging theranostics strategy. In addition, we also discuss the challenges faced by nano-delivery systems for AS diagnosis and theranostics in clinical translation, such as nanoparticle targeting efficiency, cytotoxicity and long-term accumulation, immune clearance and inaccurate disease modelling. Finally, we also provide prospects on nano-delivery systems based on diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Hu
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Quanle Gao
- Department of Neurology, Geriatrics, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Elendu C, Amaechi DC, Elendu TC, Amaechi EC, Elendu ID, Omeludike JC, Omeludike EK, Onubogu NC, Ogelle EC, Meduoye OOM, Oloyede PO, Ezeh CP, Esangbedo IJ, Adigwe AC, Akuma NM, Okafor SU. Essential information about nanotechnology in cardiology. Ann Med Surg (Lond) 2025; 87:748-779. [PMID: 40110293 PMCID: PMC11918598 DOI: 10.1097/ms9.0000000000002867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 03/22/2025] Open
Abstract
Cardiology, as a medical specialty, addresses cardiovascular diseases (CVDs), a leading cause of global mortality. Nanomaterials offer transformative potential across key areas such as drug delivery, stem cell therapy, imaging, and gene delivery. Nanomaterials improve solubility, bioavailability, and targeted delivery in drug delivery, reducing systemic side effects. Examples include gas microbubbles, liposomal preparations, and paramagnetic nanoparticles, which show promise in treating atherosclerosis. Stem cell therapy benefits from nanotechnology through enhanced cell culture conditions and three-dimensional scaffolds that support cardiomyocyte growth and survival. Gold nanoparticles and poly(lactic-co-glycolic acid)-derived microparticles further improve stem cell viability. In imaging, nanomaterials enable advanced visualization techniques such as magnetic resonance imaging with direct labeling and optical tracking via dye-conjugated nanoparticles. In gene delivery, polymeric nanocarriers like polyethyleneimine, dendrimers, and graphene-based materials offer efficient, non-viral alternatives, with magnetic nanoparticles showing promise in targeted applications. Ongoing research highlights the potential of nanomaterials to revolutionize CVD management by improving therapeutic outcomes and enabling precision medicine. These advancements position nanotechnology as a cornerstone of modern cardiology.
Collapse
|
3
|
Soni SS, Rodell CB. Cyclodextrin Nanoparticles and Injectable Polymer-Nanoparticle Hydrogels for Macrophage-Targeted Delivery of Small-Molecule Drugs. Methods Mol Biol 2025; 2902:117-131. [PMID: 40029599 DOI: 10.1007/978-1-0716-4402-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Small-molecule drugs are pillars of modern medicine, used to prevent and treat a plethora of diseases. Their applications include modulating the immune response, where macrophages and other phagocytic innate immune cells are critical therapeutic targets. In comparison to biologics, small-molecule drugs benefit from relative ease of synthesis and structural modification, long shelf life, and low cost. However, these drugs often suffer from poor pharmacokinetics (i.e., rapid renal clearance, nonspecific tissue, and cell biodistribution) and off-target effects that limit their efficacy. We have, therefore, developed a macrophage-targeted nanoparticulate carrier (cyclodextrin nanoparticle [CDNP]) that can house a variety of hydrophobic small-molecule drugs for systemic delivery through the body. This system has further been manipulated to formulate an injectable polymer-nanoparticle (iPNP) hydrogel for local delivery, with the goal of concentrating drug effects at the target site while perpetuating sustained delivery and minimizing off-target effects. Herein, we describe the strategy for preparing these CDNPs and iPNP hydrogels for a wide range of therapeutic applications.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Sharma I, Bhardwaj S, Karwasra R, Kaushik D, Sharma S. The Emergence of Nanotechnology in the Prognosis and Treatment of Myocardial Infarctions. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:35-55. [PMID: 37904554 DOI: 10.2174/1872210517666230721123453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/08/2023] [Accepted: 06/16/2023] [Indexed: 11/01/2023]
Abstract
Myocardial infarction (MI), commonly known as a heart attack, is a critical cardiovascular condition associated with high morbidity and mortality rates worldwide. Despite significant advancements in traditional treatment modalities, there remains a need for innovative approaches to improve the prognosis and treatment outcomes of MI. The emergence of nanotechnology has provided a promising avenue for revolutionizing the management of this life-threatening condition. This manuscript aims to explore the role of nanotechnology in the prognosis and treatment of myocardial infarctions. Nanotechnology offers unique advantages in the field of cardiovascular medicine, including targeted drug delivery, precise imaging and diagnosis, regenerative medicine approaches, biosensors and monitoring, and the integration of therapy and diagnostics (theragnostic). One of the key advantages of nanotechnology is the ability to deliver therapeutic agents directly to the affected site. Nanoparticles can be engineered to carry drugs specifically to damaged heart tissue, enhancing their efficacy while minimizing off-target effects. Additionally, nanoparticles can serve as contrast agents, facilitating high-resolution imaging and accurate diagnosis of infarcted heart tissue. Furthermore, nanotechnology-based regenerative approaches show promise in promoting tissue healing and regeneration after MI. Nanomaterials can provide scaffolding structures or release growth factors to stimulate the growth of new blood vessels and support tissue repair. This regenerative potential holds significant implications for restoring cardiac function and minimizing long-term complications. Nanotechnology also enables real-time monitoring of critical parameters within the heart, such as oxygen levels, pH, and electrical activity, through the utilization of nanoscale devices and sensors. This capability allows for the early detection of complications and facilitates timely interventions. Moreover, the integration of therapy and diagnostics through nanotechnology- based platforms, known as theragnostic, holds tremendous potential. Nanoparticles can simultaneously deliver therapeutic agents while providing imaging capabilities, enabling personalized treatment strategies tailored to individual patients. This manuscript will review the recent advancements, clinical trials, and patents in nanotechnology for the prognosis and treatment of myocardial infarctions. By leveraging nanotechnology's unique properties and applications, researchers and clinicians can develop innovative therapeutic approaches that enhance patient outcomes, improve prognosis, and ultimately revolutionize the management of myocardial infarctions.
Collapse
Affiliation(s)
- Isha Sharma
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, India
| | - Shivani Bhardwaj
- ICAR- Central Institute for Research on Buffaloes Hisar, Haryana, 125001, India
| | - Ritu Karwasra
- Central Council for Research in Unani Medicine, Ministry of Ayush, Govt. of India, New Delhi, 110058, India
| | - Dhirender Kaushik
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, India
| | - Shivkant Sharma
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, India
| |
Collapse
|
5
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024; 42:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
6
|
Tong J, Wang Z, Zhang J, Gao R, Liu X, Liao Y, Guo X, Wei Y. Advanced Applications of Nanomaterials in Atherosclerosis Diagnosis and Treatment: Challenges and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58072-58099. [PMID: 39432384 DOI: 10.1021/acsami.4c13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Atherosclerosis-induced coronary artery disease is a major cause of cardiovascular mortality. Clinically, conservative treatment strategies for atherosclerosis still focus on lifestyle interventions and the use of lipid-lowering and anticoagulant medications. Despite achieving some therapeutic effects, these approaches are limited by low bioavailability, long intervention periods, and significant side effects. With the advancement of nanotechnology, nanomaterials have demonstrated extraordinary potential in the biomedical field. Their excellent biocompatibility, surface modifiability, and high targeting capability not only enable efficient diagnosis of plaque progression but also allow precise drug delivery within atherosclerotic plaques, significantly enhancing drug bioavailability and reducing systemic side effects. Here, we systematically review the current research progress of nanomaterials in the field of atherosclerosis to summarize not only the types of nanomaterials but also their applications in both the diagnosis and treatment of atherosclerosis. Notably, in the context of plaque therapy, we provide a comprehensive overview of current nanomaterial applications based on their targeted therapeutic systems for different cell types within plaques. Additionally, we address the persistent challenge of clinical translation of nanomaterials by summarizing current issues and providing directions for innovation and improvement in nanomaterial design. Overall, we believe that this review systematically summarizes the applications and challenges of biomedical nanomaterials in atherosclerosis diagnosis and therapy, thereby offering insights and references for the development of therapeutic materials for atherosclerosis.
Collapse
Affiliation(s)
- Junran Tong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangfei Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
7
|
Yang Y, Pan J, Wang A, Ma Y, Liu Y, Jiang W. A novel method for the diagnosis of atherosclerosis based on nanotechnology. J Mater Chem B 2024; 12:9144-9154. [PMID: 39177217 DOI: 10.1039/d4tb00900b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Cardiovascular disease (CVD) is a global health concern, presenting significant risks to human health. Atherosclerosis is among the most prevalent CVD, impacting the medium and large arteries in the kidneys, brain, heart, and other vital organs, as well as the lower limbs. As the disease progresses, arterial obstruction can result in heart attacks and strokes. Therefore, patients with atherosclerosis should receive accurate diagnosis and timely therapeutic intervention. With the advancements in nanomedicine, researchers have proposed new research strategies and methods for atherosclerosis imaging. This paper summarizes some current research findings on the use of nanomaterials in diagnosing atherosclerosis and offers insights for optimizing the imaging applications of nanomaterials in the future.
Collapse
Affiliation(s)
- Ying Yang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Jiangpeng Pan
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Aifeng Wang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Yongcheng Ma
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Ying Liu
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| |
Collapse
|
8
|
Maier A, Teunissen AJP, Nauta SA, Lutgens E, Fayad ZA, van Leent MMT. Uncovering atherosclerotic cardiovascular disease by PET imaging. Nat Rev Cardiol 2024; 21:632-651. [PMID: 38575752 PMCID: PMC11324396 DOI: 10.1038/s41569-024-01009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Assessing atherosclerosis severity is essential for precise patient stratification. Specifically, there is a need to identify patients with residual inflammation because these patients remain at high risk of cardiovascular events despite optimal management of cardiovascular risk factors. Molecular imaging techniques, such as PET, can have an essential role in this context. PET imaging can indicate tissue-based disease status, detect early molecular changes and provide whole-body information. Advances in molecular biology and bioinformatics continue to help to decipher the complex pathogenesis of atherosclerosis and inform the development of imaging tracers. Concomitant advances in tracer synthesis methods and PET imaging technology provide future possibilities for atherosclerosis imaging. In this Review, we summarize the latest developments in PET imaging techniques and technologies for assessment of atherosclerotic cardiovascular disease and discuss the relationship between imaging readouts and transcriptomics-based plaque phenotyping.
Collapse
Affiliation(s)
- Alexander Maier
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sheqouia A Nauta
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Soni S, Kim KM, Sarkar B, Rodell CB. Uptake of Cyclodextrin Nanoparticles by Macrophages is Dependent on Particle Size and Receptor-Mediated Interactions. ACS APPLIED BIO MATERIALS 2024; 7:4856-4866. [PMID: 38231485 PMCID: PMC11252246 DOI: 10.1021/acsabm.3c00985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/18/2024]
Abstract
Physiochemical properties of nanoparticles, such as their size and chemical composition, dictate their interaction with professional phagocytes of the innate immune system. Macrophages, in particular, are key regulators of the immune microenvironment that heavily influence particle biodistribution as a result of their uptake. This attribute enables macrophage-targeted delivery, including for phenotypic modulation. Saccharide-based materials, including polyglucose polymers and nanoparticles, are efficient vehicles for macrophage-targeted delivery. Here, we investigate the influence of particle size on cyclodextrin nanoparticle (CDNP) uptake by macrophages and further examine the receptor-mediated interactions that drive macrophage-targeted delivery. We designed and synthesized CDNPs ranging in size from 25 nm to >100 nm in diameter. Increasing particle size was correlated with greater uptake by macrophages in vitro. Both scavenger receptor A1 and mannose receptor were critical mediators of macrophage-targeted delivery, inhibition of which reduced the extent of uptake. Finally, we investigated the cellular bioavailability of drug-loaded CDNPs using a model anti-inflammatory drug, celastrol, which demonstrated that drug bioactivity is improved by CDNP loading relative to free drug alone. This study thus elucidates the interactions between the polyglucose nanoparticles and macrophages, thereby facilitating their application in macrophage-targeted drug delivery that has applications in the context of tissue injury and repair.
Collapse
Affiliation(s)
- Shreya
S. Soni
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Kenneth M. Kim
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
- Department
of Microbiology and Immunology, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Biplab Sarkar
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Christopher B. Rodell
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
10
|
Kou H, Yang H. Molecular imaging nanoprobes and their applications in atherosclerosis diagnosis. Theranostics 2024; 14:4747-4772. [PMID: 39239513 PMCID: PMC11373619 DOI: 10.7150/thno.96037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/06/2024] [Indexed: 09/07/2024] Open
Abstract
Molecular imaging has undergone significant development in recent years for its excellent ability to image and quantify biologic processes at cellular and molecular levels. Its application is of significance in cardiovascular diseases, particularly in diagnosing them at early stages. Atherosclerosis is a complex, chronic, and progressive disease that can lead to serious consequences such as heart strokes or infarctions. Attempts have been made to detect atherosclerosis with molecular imaging modalities. Not only do imaging modalities develop rapidly, but research of relevant nanomaterials as imaging probes has also been increasingly studied in recent years. This review focuses on the latest developments in the design and synthesis of probes that can be utilized in computed tomography, positron emission tomography, magnetic resonance imaging, ultrasound imaging, photoacoustic imaging and combined modalities. The challenges and future developments of nanomaterials for molecular imaging modalities are also discussed.
Collapse
Affiliation(s)
| | - Hu Yang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, MO 65409, United States
| |
Collapse
|
11
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
12
|
Toner YC, Prévot G, van Leent MMT, Munitz J, Oosterwijk R, Verschuur AVD, van Elsas Y, Peric V, Maas RJF, Ranzenigo A, Morla-Folch J, Wang W, Umali M, de Dreu A, Fernandes JC, Sullivan NAT, Maier A, Mason C, Reiner T, Fayad ZA, Mulder WJM, Teunissen AJP, Pérez-Medina C. Macrophage PET imaging in mouse models of cardiovascular disease and cancer with an apolipoprotein-inspired radiotracer. NPJ IMAGING 2024; 2:12. [PMID: 38765879 PMCID: PMC11096117 DOI: 10.1038/s44303-024-00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/06/2024] [Indexed: 05/22/2024]
Abstract
Macrophages are key inflammatory mediators in many pathological conditions, including cardiovascular disease (CVD) and cancer, the leading causes of morbidity and mortality worldwide. This makes macrophage burden a valuable diagnostic marker and several strategies to monitor these cells have been reported. However, such strategies are often high-priced, non-specific, invasive, and/or not quantitative. Here, we developed a positron emission tomography (PET) radiotracer based on apolipoprotein A1 (ApoA1), the main protein component of high-density lipoprotein (HDL), which has an inherent affinity for macrophages. We radiolabeled an ApoA1-mimetic peptide (mA1) with zirconium-89 (89Zr) to generate a lipoprotein-avid PET probe (89Zr-mA1). We first characterized 89Zr-mA1's affinity for lipoproteins in vitro by size exclusion chromatography. To study 89Zr-mA1's in vivo behavior and interaction with endogenous lipoproteins, we performed extensive studies in wildtype C57BL/6 and Apoe-/- hypercholesterolemic mice. Subsequently, we used in vivo PET imaging to study macrophages in melanoma and myocardial infarction using mouse models. The tracer's cell specificity was assessed by histology and mass cytometry (CyTOF). Our data show that 89Zr-mA1 associates with lipoproteins in vitro. This is in line with our in vivo experiments, in which we observed longer 89Zr-mA1 circulation times in hypercholesterolemic mice compared to C57BL/6 controls. 89Zr-mA1 displayed a tissue distribution profile similar to ApoA1 and HDL, with high kidney and liver uptake as well as substantial signal in the bone marrow and spleen. The tracer also accumulated in tumors of melanoma-bearing mice and in the ischemic myocardium of infarcted animals. In these sites, CyTOF analyses revealed that natZr-mA1 was predominantly taken up by macrophages. Our results demonstrate that 89Zr-mA1 associates with lipoproteins and hence accumulates in macrophages in vivo. 89Zr-mA1's high uptake in these cells makes it a promising radiotracer for non-invasively and quantitatively studying conditions characterized by marked changes in macrophage burden.
Collapse
Affiliation(s)
- Yohana C. Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Geoffrey Prévot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Mandy M. T. van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Roderick Oosterwijk
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Anna Vera D. Verschuur
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Yuri van Elsas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vedran Peric
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rianne J. F. Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Martin Umali
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jessica Chimene Fernandes
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Nathaniel A. T. Sullivan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Cardiology and Angiology, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Mason
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Department of Radiology, Weill Cornell Medical College, New York, NY USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Zahi A. Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Willem J. M. Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abraham J. P. Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
13
|
Kim HS, Halabi EA, Enbergs N, Kohler RH, Fei F, Garris CS, Weissleder R. A non-lipid nucleic acid delivery vector with dendritic cell tropism and stimulation. Theranostics 2024; 14:2934-2945. [PMID: 38773971 PMCID: PMC11103498 DOI: 10.7150/thno.95267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/10/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: Nucleic acid constructs are commonly used for vaccination, immune stimulation, and gene therapy, but their use in cancer still remains limited. One of the reasons is that systemic delivery to tumor-associated antigen-presenting cells (dendritic cells and macrophages) is often inefficient, while off-target nucleic acid-sensing immune pathways can stimulate systemic immune responses. Conversely, certain carbohydrate nanoparticles with small molecule payloads have been shown to target these cells efficiently in the tumor microenvironment. Yet, nucleic acid incorporation into such carbohydrate-based nanoparticles has proven challenging. Methods: We developed a novel approach using cross-linked bis succinyl-cyclodextrin (b-s-CD) nanoparticles to efficiently deliver nucleic acids and small-molecule immune enhancer to phagocytic cells in tumor environments and lymph nodes. Our study involved incorporating these components into the nanoparticles and assessing their efficacy in activating antigen-presenting cells. Results: The multi-modality immune stimulators effectively activated antigen-presenting cells and promoted anti-tumor immunity in vivo. This was evidenced by enhanced delivery to phagocytic cells and subsequent immune response activation in tumor environments and lymph nodes. Conclusion: Here, we describe a new approach to incorporating both nucleic acids and small-molecule immune enhancers into cross-linked bis succinyl-cyclodextrin (b-s-CD) nanoparticles for efficient delivery to phagocytic cells in tumor environments and lymph nodes in vivo. These multi-modality immune stimulators can activate antigen-presenting cells and foster anti-tumor immunity. We argue that this strategy can potentially be used to enhance anti-tumor efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ralph Weissleder
- ✉ Corresponding author: R. Weissleder, MD, PhD. Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA, 02114. 617-726-8226;
| |
Collapse
|
14
|
Switala L, Di L, Gao H, Asase C, Klos M, Rengasamy P, Fedyukina D, Maiseyeu A. Engineered nanoparticles promote cardiac tropism of AAV vectors. J Nanobiotechnology 2024; 22:223. [PMID: 38702815 PMCID: PMC11067271 DOI: 10.1186/s12951-024-02485-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.
Collapse
Affiliation(s)
- Lauren Switala
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Lin Di
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Huiyun Gao
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
| | - Courteney Asase
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
| | - Matthew Klos
- Department of Pediatrics, Case Western Reserve University, Cleveland, USA
| | - Palanivel Rengasamy
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
| | - Daria Fedyukina
- Bioheights LLC, Cleveland, USA
- Advanced Research Projects Agency for Health, ARPA-H, Washington, USA
| | - Andrei Maiseyeu
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
15
|
Deleuziere M, Benoist É, Quelven I, Gras E, Amiens C. [ 18F]-Radiolabelled Nanoplatforms: A Critical Review of Their Intrinsic Characteristics, Radiolabelling Methods, and Purification Techniques. Molecules 2024; 29:1537. [PMID: 38611815 PMCID: PMC11013168 DOI: 10.3390/molecules29071537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
A wide range of nano-objects is found in many applications of our everyday life. Recognition of their peculiar properties and ease of functionalization has prompted their engineering into multifunctional platforms that are supposed to afford efficient tools for the development of biomedical applications. However, bridging the gap between bench to bedside cannot be expected without a good knowledge of their behaviour in vivo, which can be obtained through non-invasive imaging techniques, such as positron emission tomography (PET). Their radiolabelling with [18F]-fluorine, a technique already well established and widely used routinely for PET imaging, with [18F]-FDG for example, and in preclinical investigation using [18F]-radiolabelled biological macromolecules, has, therefore, been developed. In this context, this review highlights the various nano-objects studied so far, the reasons behind their radiolabelling, and main in vitro and/or in vivo results obtained thereof. Then, the methods developed to introduce the radioelement are presented. Detailed indications on the chemical steps involved are provided, and the stability of the radiolabelling is discussed. Emphasis is then made on the techniques used to purify and analyse the radiolabelled nano-objects, a point that is rarely discussed despite its technical relevance and importance for accurate imaging. The pros and cons of the different methods developed are finally discussed from which future work can develop.
Collapse
Affiliation(s)
- Maëlle Deleuziere
- SPCMIB, CNRS UMR 5068, Université de Toulouse III Paul Sabatier, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (M.D.); (É.B.)
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Éric Benoist
- SPCMIB, CNRS UMR 5068, Université de Toulouse III Paul Sabatier, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (M.D.); (É.B.)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| | - Emmanuel Gras
- Laboratoire Hétérochimie Fondamentale et Appliquée, UMR 5069, CNRS—Université de Toulouse, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France;
| | - Catherine Amiens
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| |
Collapse
|
16
|
Sarkar B, Arlauckas SP, Cuccarese MF, Garris CS, Weissleder R, Rodell CB. Host-functionalization of macrin nanoparticles to enable drug loading and control tumor-associated macrophage phenotype. Front Immunol 2024; 15:1331480. [PMID: 38545103 PMCID: PMC10965546 DOI: 10.3389/fimmu.2024.1331480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Macrophages are critical regulators of the tumor microenvironment and often present an immuno-suppressive phenotype, supporting tumor growth and immune evasion. Promoting a robust pro-inflammatory macrophage phenotype has emerged as a therapeutic modality that supports tumor clearance, including through synergy with immune checkpoint therapies. Polyglucose nanoparticles (macrins), which possess high macrophage affinity, are useful vehicles for delivering drugs to macrophages, potentially altering their phenotype. Here, we examine the potential of functionalized macrins, synthesized by crosslinking carboxymethyl dextran with L-lysine, as effective carriers of immuno-stimulatory drugs to tumor-associated macrophages (TAMs). Azide groups incorporated during particle synthesis provided a handle for click-coupling of propargyl-modified β-cyclodextrin to macrins under mild conditions. Fluorescence-based competitive binding assays revealed the ability of β-cyclodextrin to non-covalently bind to hydrophobic immuno-stimulatory drug candidates (Keq ~ 103 M-1), enabling drug loading within nanoparticles. Furthermore, transcriptional profiles of macrophages indicated robust pro-inflammatory reprogramming (elevated Nos2 and Il12; suppressed Arg1 and Mrc1 expression levels) for a subset of these immuno-stimulatory agents (UNC2025 and R848). Loading of R848 into the modified macrins improved the drug's effect on primary murine macrophages by three-fold in vitro. Intravital microscopy in IL-12-eYFP reporter mice (24 h post-injection) revealed a two-fold enhancement in mean YFP fluorescence intensity in macrophages targeted with R848-loaded macrins, relative to vehicle controls, validating the desired pro-inflammatory reprogramming of TAMs in vivo by cell-targeted drug delivery. Finally, in an intradermal MC38 tumor model, cyclodextrin-modified macrin NPs loaded with immunostimulatory drugs significantly reduced tumor growth. Therefore, efficient and effective repolarization of tumor-associated macrophages to an M1-like phenotype-via drug-loaded macrins-inhibits tumor growth and may be useful as an adjuvant to existing immune checkpoint therapies.
Collapse
Affiliation(s)
- Biplab Sarkar
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Sean P. Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Michael F. Cuccarese
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Christopher S. Garris
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Pathology, Harvard Medical School, Boston, MA, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Christopher B. Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
17
|
Shah H, Paul G, Yadav AK. Surface-Tailored Nanoplatform for the Diagnosis and Management of Stroke: Current Strategies and Future Outlook. Mol Neurobiol 2024; 61:1383-1403. [PMID: 37707740 DOI: 10.1007/s12035-023-03635-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/02/2023] [Indexed: 09/15/2023]
Abstract
Stroke accounts for one of the top leading reasons for neurological mortality and morbidity around the globe. Both ischemic and hemorrhagic strokes lead to local hypoxia and are brought about by the occlusion or rupturing of the blood vessels. The events taking place after the onset of a stroke include membrane ion pump failure, calcium and glutamate-mediated excitotoxicity, increased ROS production causing DNA damage, mitochondrial dysfunction, oxidative stress, development of brain edema, and microvascular dysfunction. To date, tissue plasminogen activator (tPA) therapy and mechanical removal of blood clots are the only clinically available stroke therapies, approved by Food and Drug Administration (FDA). But because of the narrow therapeutic window of around 4.5 h for tPA therapy and complications like systemic bleeding and anaphylaxis, more clinical trials are ongoing in the same field. Therefore, using nanocarriers with diverse physicochemical properties is a promising strategy in treating and diagnosing stroke as they can efficiently bypass the tight blood-brain barrier (BBB) through mechanisms like receptor-mediated transcytosis and help achieve controlled and targeted drug delivery. In this review, we will mainly focus on the pathophysiology of stroke, BBB alterations following stroke, strategies to target BBB for stroke therapies, different types of nanocarriers currently being used for therapeutic intervention of stroke, and biomarkers as well as imaging techniques used for the detection and diagnosis of stroke.
Collapse
Affiliation(s)
- Hinal Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER) Raebareli (An Institute of National Importance Under Dept. of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Gajanan Paul
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER) Raebareli (An Institute of National Importance Under Dept. of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER) Raebareli (An Institute of National Importance Under Dept. of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
18
|
Cheng J, Huang H, Chen Y, Wu R. Nanomedicine for Diagnosis and Treatment of Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304294. [PMID: 37897322 PMCID: PMC10754137 DOI: 10.1002/advs.202304294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/11/2023] [Indexed: 10/30/2023]
Abstract
With the changing disease spectrum, atherosclerosis has become increasingly prevalent worldwide and the associated diseases have emerged as the leading cause of death. Due to their fascinating physical, chemical, and biological characteristics, nanomaterials are regarded as a promising tool to tackle enormous challenges in medicine. The emerging discipline of nanomedicine has filled a huge application gap in the atherosclerotic field, ushering a new generation of diagnosis and treatment strategies. Herein, based on the essential pathogenic contributors of atherogenesis, as well as the distinct composition/structural characteristics, synthesis strategies, and surface design of nanoplatforms, the three major application branches (nanodiagnosis, nanotherapy, and nanotheranostic) of nanomedicine in atherosclerosis are elaborated. Then, state-of-art studies containing a sequence of representative and significant achievements are summarized in detail with an emphasis on the intrinsic interaction/relationship between nanomedicines and atherosclerosis. Particularly, attention is paid to the biosafety of nanomedicines, which aims to pave the way for future clinical translation of this burgeoning field. Finally, this comprehensive review is concluded by proposing unresolved key scientific issues and sharing the vision and expectation for the future, fully elucidating the closed loop from atherogenesis to the application paradigm of nanomedicines for advancing the early achievement of clinical applications.
Collapse
Affiliation(s)
- Jingyun Cheng
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Hui Huang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| | - Rong Wu
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
19
|
Ju J, Xu D, Mo X, Miao J, Xu L, Ge G, Zhu X, Deng H. Multifunctional polysaccharide nanoprobes for biological imaging. Carbohydr Polym 2023; 317:121048. [PMID: 37364948 DOI: 10.1016/j.carbpol.2023.121048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023]
Abstract
Imaging and tracking biological targets or processes play an important role in revealing molecular mechanisms and disease states. Bioimaging via optical, nuclear, or magnetic resonance techniques enables high resolution, high sensitivity, and high depth imaging from the whole animal down to single cells via advanced functional nanoprobes. To overcome the limitations of single-modality imaging, multimodality nanoprobes have been engineered with a variety of imaging modalities and functionalities. Polysaccharides are sugar-containing bioactive polymers with superior biocompatibility, biodegradability, and solubility. The combination of polysaccharides with single or multiple contrast agents facilitates the development of novel nanoprobes with enhanced functions for biological imaging. Nanoprobes constructed with clinically applicable polysaccharides and contrast agents hold great potential for clinical translations. This review briefly introduces the basics of different imaging modalities and polysaccharides, then summarizes the recent progress of polysaccharide-based nanoprobes for biological imaging in various diseases, emphasizing bioimaging with optical, nuclear, and magnetic resonance techniques. The current issues and future directions regarding the development and applications of polysaccharide nanoprobes are further discussed.
Collapse
Affiliation(s)
- Jingxuan Ju
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Danni Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuan Mo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqian Miao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Hongping Deng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
20
|
Zhang J, Guo Y, Bai Y, Wei Y. Application of biomedical materials in the diagnosis and treatment of myocardial infarction. J Nanobiotechnology 2023; 21:298. [PMID: 37626396 PMCID: PMC10463704 DOI: 10.1186/s12951-023-02063-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Myocardial infarction (MI) is a cardiovascular emergency and the leading cause of death worldwide. Inflammatory and immune responses are initiated immediately after MI, leading to myocardial death, scarring, and ventricular remodeling. Current therapeutic approaches emphasize early restoration of ischemic myocardial reperfusion, but there is no effective treatment for the pathological changes of infarction. Biomedical materials development has brought new hope for MI diagnosis and treatment. Biomedical materials, such as cardiac patches, hydrogels, nano biomaterials, and artificial blood vessels, have played an irreplaceable role in MI diagnosis and treatment. They improve the accuracy and efficacy of MI diagnosis and offer further possibilities for reducing inflammation, immunomodulation, inhibiting fibrosis, and cardiac regeneration. This review focuses on the advances in biomedical materials applications in MI diagnosis and treatment. The current studies are outlined in terms of mechanisms of action and effects. It is addressed how biomedical materials application can lessen myocardial damage, encourage angiogenesis, and enhance heart function. Their clinical transformation value and application prospect are discussed.
Collapse
Affiliation(s)
- Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yishan Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, 256600, China
| | - Yu Bai
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100000, China.
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, P.R. China.
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
21
|
Algoet M, Janssens S, Himmelreich U, Gsell W, Pusovnik M, Van den Eynde J, Oosterlinck W. Myocardial ischemia-reperfusion injury and the influence of inflammation. Trends Cardiovasc Med 2023; 33:357-366. [PMID: 35181472 DOI: 10.1016/j.tcm.2022.02.005] [Citation(s) in RCA: 182] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/20/2022]
Abstract
Acute myocardial infarction is caused by a sudden coronary artery occlusion and leads to ischemia in the corresponding myocardial territory which generally results in myocardial necrosis. Without restoration of coronary perfusion, myocardial scar formation will cause adverse remodelling of the myocardium and heart failure. Successful introduction of percutaneous coronary intervention and surgical coronary artery bypass grafting made it possible to achieve early revascularisation/reperfusion, hence limiting the ischemic zone of myocardium. However, reperfusion by itself paradoxically triggers an exacerbated and accelerated injury in the myocardium, called ischemia-reperfusion (I/R) injury. This mechanism is partially driven by inflammation through multiple interacting pathways. In this review we summarize the current insights in mechanisms of I/R injury and the influence of altered inflammation. Multiple pharmacological and interventional therapeutic strategies (ischemic conditioning) have proven to be beneficial during I/R in preclinical models but were notoriously unsuccessful upon clinical translation. In this review we focus on common mechanisms of I/R injury, altered inflammation and potential therapeutic strategies. We hypothesize that a dual approach may be of value because I/R injury patients are predestined with multiple comorbidities and systemic low-grade inflammation, which requires targeted intervention before other strategies can be fully effective.
Collapse
Affiliation(s)
- Michiel Algoet
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Matic Pusovnik
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jef Van den Eynde
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Helen B. Taussig Heart Center, The Johns Hopkins Hospital and School of Medicine, Baltimore, United States
| | | |
Collapse
|
22
|
Soni SS, D'Elia AM, Rodell CB. Control of the post-infarct immune microenvironment through biotherapeutic and biomaterial-based approaches. Drug Deliv Transl Res 2023; 13:1983-2014. [PMID: 36763330 PMCID: PMC9913034 DOI: 10.1007/s13346-023-01290-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/11/2023]
Abstract
Ischemic heart failure (IHF) is a leading cause of morbidity and mortality worldwide, for which heart transplantation remains the only definitive treatment. IHF manifests from myocardial infarction (MI) that initiates tissue remodeling processes, mediated by mechanical changes in the tissue (loss of contractility, softening of the myocardium) that are interdependent with cellular mechanisms (cardiomyocyte death, inflammatory response). The early remodeling phase is characterized by robust inflammation that is necessary for tissue debridement and the initiation of repair processes. While later transition toward an immunoregenerative function is desirable, functional reorientation from an inflammatory to reparatory environment is often lacking, trapping the heart in a chronically inflamed state that perpetuates cardiomyocyte death, ventricular dilatation, excess fibrosis, and progressive IHF. Therapies can redirect the immune microenvironment, including biotherapeutic and biomaterial-based approaches. In this review, we outline these existing approaches, with a particular focus on the immunomodulatory effects of therapeutics (small molecule drugs, biomolecules, and cell or cell-derived products). Cardioprotective strategies, often focusing on immunosuppression, have shown promise in pre-clinical and clinical trials. However, immunoregenerative therapies are emerging that often benefit from exacerbating early inflammation. Biomaterials can be used to enhance these therapies as a result of their intrinsic immunomodulatory properties, parallel mechanisms of action (e.g., mechanical restraint), or by enabling cell or tissue-targeted delivery. We further discuss translatability and the continued progress of technologies and procedures that contribute to the bench-to-bedside development of these critically needed treatments.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA.
| |
Collapse
|
23
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Derlin T, Spencer BA, Mamach M, Abdelhafez Y, Nardo L, Badawi RD, Cherry SR, Bengel FM. Exploring Vessel Wall Biology In Vivo by Ultrasensitive Total-Body PET. J Nucl Med 2023; 64:416-422. [PMID: 36175139 PMCID: PMC10071799 DOI: 10.2967/jnumed.122.264550] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ultrasensitive, high-resolution, extended-field-of-view total-body (TB) PET using the first-of-its-kind 194-cm axial-field-of-view uEXPLORER may facilitate the interrogation of biologic hallmarks of hitherto difficult-to-evaluate low-signal vessel wall pathology in cardiovascular disease. Methods: Healthy volunteers were imaged serially for up to 12 h after a standard dose of 18F-FDG (n = 15) or for up to 3 h after injection of a very low dose (about 5% of a standard dose; n = 15). A cohort undergoing standard 18F-FDG PET (n = 15) on a conventional scanner with a 22-cm axial field of view served as a comparison group. Arterial wall signal, crosstalk with hematopoietic and lymphoid organs, and image quality were analyzed using standardized techniques. Results: TB PET depicted the large vessel walls with excellent quality. The arterial wall could be imaged with high contrast up to 12 h after tracer injection. Ultralow-dose TB 18F-FDG images yielded a vessel wall signal and target-to-background ratio comparable to those of conventional-dose, short-axial-field-of-view PET. Crosstalk between vessel wall and lymphoid organs was identified with better accuracy in both TB PET cohorts than in conventional PET. Conclusion: TB PET enables detailed assessment of in vivo vessel wall biology and its crosstalk with other organs over an extended time window after tracer injection or at an ultralow tracer dose. These initial observations support the feasibility of serial imaging in low-risk populations and will stimulate future mechanistic studies or therapy monitoring in atherosclerosis and other vessel wall pathologies.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany;
| | - Benjamin A Spencer
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Martin Mamach
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany; and
| | - Yasser Abdelhafez
- Department of Radiology, University of California, Davis, Davis, California
| | - Lorenzo Nardo
- Department of Radiology, University of California, Davis, Davis, California
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, Davis, California
- Department of Radiology, University of California, Davis, Davis, California
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, Davis, California
- Department of Radiology, University of California, Davis, Davis, California
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
25
|
Senders ML, Calcagno C, Tawakol A, Nahrendorf M, Mulder WJM, Fayad ZA. PET/MR imaging of inflammation in atherosclerosis. Nat Biomed Eng 2023; 7:202-220. [PMID: 36522465 DOI: 10.1038/s41551-022-00970-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
Myocardial infarction, stroke, mental disorders, neurodegenerative processes, autoimmune diseases, cancer and the human immunodeficiency virus impact the haematopoietic system, which through immunity and inflammation may aggravate pre-existing atherosclerosis. The interplay between the haematopoietic system and its modulation of atherosclerosis has been studied by imaging the cardiovascular system and the activation of haematopoietic organs via scanners integrating positron emission tomography and resonance imaging (PET/MRI). In this Perspective, we review the applicability of integrated whole-body PET/MRI for the study of immune-mediated phenomena associated with haematopoietic activity and cardiovascular disease, and discuss the translational opportunities and challenges of the technology.
Collapse
Affiliation(s)
- Max L Senders
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ahmed Tawakol
- Cardiology Division and Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Willem J M Mulder
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands.
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
Goel M, Mackeyev Y, Krishnan S. Radiolabeled nanomaterial for cancer diagnostics and therapeutics: principles and concepts. Cancer Nanotechnol 2023; 14:15. [PMID: 36865684 PMCID: PMC9968708 DOI: 10.1186/s12645-023-00165-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
In the last three decades, radiopharmaceuticals have proven their effectiveness for cancer diagnosis and therapy. In parallel, the advances in nanotechnology have fueled a plethora of applications in biology and medicine. A convergence of these disciplines has emerged more recently with the advent of nanotechnology-aided radiopharmaceuticals. Capitalizing on the unique physical and functional properties of nanoparticles, radiolabeled nanomaterials or nano-radiopharmaceuticals have the potential to enhance imaging and therapy of human diseases. This article provides an overview of various radionuclides used in diagnostic, therapeutic, and theranostic applications, radionuclide production through different techniques, conventional radionuclide delivery systems, and advancements in the delivery systems for nanomaterials. The review also provides insights into fundamental concepts necessary to improve currently available radionuclide agents and formulate new nano-radiopharmaceuticals.
Collapse
Affiliation(s)
- Muskan Goel
- Amity School of Applied Sciences, Amity University, Gurugram, Haryana 122413 India
| | - Yuri Mackeyev
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center, Houston, TX 77030 USA
| | - Sunil Krishnan
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center, Houston, TX 77030 USA
| |
Collapse
|
27
|
Tu S, He W, Han J, Wu A, Ren W. Advances in imaging and treatment of atherosclerosis based on organic nanoparticles. APL Bioeng 2022; 6:041501. [PMCID: PMC9726224 DOI: 10.1063/5.0127835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 10/31/2022] [Indexed: 12/09/2022] Open
Abstract
Atherosclerosis, a systemic chronic inflammatory disease, can lead to thrombosis and vascular occlusion, thereby inducing a series of serious vascular diseases. Currently, distinguishing unstable plaques early and achieving more effective treatment are the two main clinical concerns in atherosclerosis. Organic nanoparticles have great potential in atherosclerotic imaging and treatment, showing superior biocompatibility, drug-loading capacity, and synthesis. This article illustrates the process of atherosclerosis onset and the key targeted cells, then systematically summarizes recent progress made in organic nanoparticle-based imaging of different types of targeted cells and therapeutic methods for atherosclerosis, including optical and acoustic-induced therapy, drug delivery, gene therapy, and immunotherapy. Finally, we discuss the major impediments that need to be addressed in future clinical practice. We believe this article will help readers to develop a comprehensive and in-depth understanding of organic nanoparticle-based atherosclerotic imaging and treatment, thus advancing further development of anti-atherosclerosis therapies.
Collapse
Affiliation(s)
| | - Wenming He
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province 315020, China,Authors to whom correspondence should be addressed:; ; and
| | | | - Aiguo Wu
- Authors to whom correspondence should be addressed:; ; and
| | - Wenzhi Ren
- Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
28
|
Parry R, Majeed K, Pixley F, Hillis GS, Francis RJ, Schultz CJ. Unravelling the role of macrophages in cardiovascular inflammation through imaging: a state-of-the-art review. Eur Heart J Cardiovasc Imaging 2022; 23:e504-e525. [PMID: 35993316 PMCID: PMC9671294 DOI: 10.1093/ehjci/jeac167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death and disability for patients across the world. Our understanding of atherosclerosis as a primary cholesterol issue has diversified, with a significant dysregulated inflammatory component that largely remains untreated and continues to drive persistent cardiovascular risk. Macrophages are central to atherosclerotic inflammation, and they exist along a functional spectrum between pro-inflammatory and anti-inflammatory extremes. Recent clinical trials have demonstrated a reduction in major cardiovascular events with some, but not all, anti-inflammatory therapies. The recent addition of colchicine to societal guidelines for the prevention of recurrent cardiovascular events in high-risk patients with chronic coronary syndromes highlights the real-world utility of this class of therapies. A highly targeted approach to modification of interleukin-1-dependent pathways shows promise with several novel agents in development, although excessive immunosuppression and resulting serious infection have proven a barrier to implementation into clinical practice. Current risk stratification tools to identify high-risk patients for secondary prevention are either inadequately robust or prohibitively expensive and invasive. A non-invasive and relatively inexpensive method to identify patients who will benefit most from novel anti-inflammatory therapies is required, a role likely to be fulfilled by functional imaging methods. This review article outlines our current understanding of the inflammatory biology of atherosclerosis, upcoming therapies and recent landmark clinical trials, imaging modalities (both invasive and non-invasive) and the current landscape surrounding functional imaging including through targeted nuclear and nanobody tracer development and their application.
Collapse
Affiliation(s)
- Reece Parry
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| | - Kamran Majeed
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Waikato District Health Board, Hamilton 3204, New Zealand
| | - Fiona Pixley
- School of Biomedical Sciences, Pharmacology and Toxicology, University of Western Australia, Perth 6009, Australia
| | - Graham Scott Hillis
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| | - Roslyn Jane Francis
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth 6009, Australia
| | - Carl Johann Schultz
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| |
Collapse
|
29
|
Li X, Wu M, Li J, Guo Q, Zhao Y, Zhang X. Advanced targeted nanomedicines for vulnerable atherosclerosis plaque imaging and their potential clinical implications. Front Pharmacol 2022; 13:906512. [PMID: 36313319 PMCID: PMC9606597 DOI: 10.3389/fphar.2022.906512] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis plaques caused by cerebrovascular and coronary artery disease have been the leading cause of death and morbidity worldwide. Precise assessment of the degree of atherosclerotic plaque is critical for predicting the risk of atherosclerosis plaques and monitoring postinterventional outcomes. However, traditional imaging techniques to predict cardiocerebrovascular events mainly depend on quantifying the percentage reduction in luminal diameter, which would immensely underestimate non-stenotic high-risk plaque. Identifying the degree of atherosclerosis plaques still remains highly limited. vNanomedicine-based imaging techniques present unique advantages over conventional techniques due to the superior properties intrinsic to nanoscope, which possess enormous potential for characterization and detection of the features of atherosclerosis plaque vulnerability. Here, we review recent advancements in the development of targeted nanomedicine-based approaches and their applications to atherosclerosis plaque imaging and risk stratification. Finally, the challenges and opportunities regarding the future development and clinical translation of the targeted nanomedicine in related fields are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuening Zhang
- Department of Radiology, Tianjin Medical University Second Hospital, Tianjin, China
| |
Collapse
|
30
|
Hu PP, Luo SX, Fan XQ, Li D, Tong XY. Macrophage-targeted nanomedicine for the diagnosis and management of atherosclerosis. Front Pharmacol 2022; 13:1000316. [PMID: 36160452 PMCID: PMC9501673 DOI: 10.3389/fphar.2022.1000316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular diseases, such as myocardial infarction and stroke, which account for the highest death toll worldwide. Macrophage is the major contributor to atherosclerosis progression, and therefore, macrophage-associated pathological process is considered an extremely important target for the diagnosis and treatment of atherosclerosis. However, the existing clinical strategies still have many bottlenecks and challenges in atherosclerosis’s early detection and management. Nanomedicine, using various nanoparticles/nanocarriers for medical purposes, can effectively load therapeutic agents, significantly improve their stability and accurately deliver them to the atherosclerotic plaques. In this review, we summarized the latest progress of the macrophage-targeted nanomedicine in the diagnosis and treatment of atherosclerosis, and their potential applications and clinical benefits are also discussed.
Collapse
Affiliation(s)
- Ping Ping Hu
- Chongqing Engineering Research Center for Pharmacodynamics Evaluation, College of Pharmacy, Chongqing Medical University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| | - Shuang Xue Luo
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiao Qing Fan
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Di Li
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Yong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| |
Collapse
|
31
|
Deng H, Li Xu, Ju J, Mo X, Ge G, Zhu X. Multifunctional nanoprobes for macrophage imaging. Biomaterials 2022; 290:121824. [PMID: 36209580 DOI: 10.1016/j.biomaterials.2022.121824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/28/2022] [Accepted: 09/24/2022] [Indexed: 11/30/2022]
|
32
|
Independent Association of Thyroid Dysfunction and Inflammation Predicts Adverse Events in Patients with Heart Failure via Promoting Cell Death. J Cardiovasc Dev Dis 2022; 9:jcdd9090290. [PMID: 36135435 PMCID: PMC9503390 DOI: 10.3390/jcdd9090290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 12/03/2022] Open
Abstract
Thyroid dysfunction and inflammation are individually implicated in the increased risk of heart failure. Given the regulatory role of thyroid hormones on immune cells, this study aimed to investigate their joint association in heart failure. Patients with pre-existing heart failure were enrolled when hospitalized between July 2019 and September 2021. Thyroid function and inflammatory markers were measured at the enrollment. The composite of all-cause mortality or rehospitalization for heart failure were studied in the following year. Among 451 participants (mean age 66.1 years, 69.4% male), 141 incident primary endpoints were observed during a median follow-up of 289 days. TT3 and FT3 levels were negatively correlated with BNP levels (r: −0.40, p < 0.001; r: −0.40, p < 0.001, respectively) and NT-proBNP levels (r: −0.39, p < 0.001; r: −0.39, p < 0.001). Multivariate COX regression analysis revealed that FT3 (adjusted HR: 0.677, 95% CI: 0.551−0.832) and NLR (adjusted HR: 1.073, 95% CI: 1.036−1.111) were associated with adverse event, and similar results for TT3 (adjusted HR: 0.320, 95% CI: 0.181−0.565) and NLR (adjusted HR: 1.072, 95% CI: 1.035−1.110). Restricted cubic splines analysis indicated a linear relationship between T3 level and adverse events. Mechanistically, primary cardiomyocytes showed strong resistance to TNF-α induced apoptosis under optimal T3 concentrations, as evidenced by TUNEL staining, flow cytometry analysis, and LDH release assay as well as increased expression of Bcl-2. Thyroid dysfunction and inflammation are independently associated with cardiovascular risk in heart failure patients, which may concurrently contribute to the ongoing cardiomyocyte loss in the disease progression.
Collapse
|
33
|
Lv W, Liu Y, Li S, Lv L, Lu H, Xin H. Advances of nano drug delivery system for the theranostics of ischemic stroke. J Nanobiotechnology 2022; 20:248. [PMID: 35641956 PMCID: PMC9153106 DOI: 10.1186/s12951-022-01450-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/05/2022] [Indexed: 02/07/2023] Open
Abstract
From the global perspective, stroke refers to a highly common cause of disability and death. Ischemic stroke (IS), attributed to blood vessel blockage, preventing the flow of blood to brain, acts as the most common form of stroke. Thus far, thrombolytic therapy is the only clinical treatment for IS with the approval from the FDA. Moreover, the physiology barrier complicates therapeutically and diagnostically related intervention development of IS. Accordingly, developing efficient and powerful curative approaches for IS diagnosis and treatment is urgently required. The advent of nanotechnology has brought dawn and hope to better curative and imaging forms for the management of IS. This work reviews the recent advances and challenges correlated with the nano drug delivery system for IS therapy and diagnosis. The overview of the current knowledge of the important molecular pathological mechanisms in cerebral ischemia and how the drugs cross the blood brain barrier will also be briefly summarized.
Collapse
Affiliation(s)
- Wei Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Yijiao Liu
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Shengnan Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Lingyan Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Hongdan Lu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China.
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
34
|
Xu R, Tomeh MA, Ye S, Zhang P, Lv S, You R, Wang N, Zhao X. Novel microfluidic swirl mixers for scalable formulation of curcumin loaded liposomes for cancer therapy. Int J Pharm 2022; 622:121857. [PMID: 35623489 DOI: 10.1016/j.ijpharm.2022.121857] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/15/2022]
Abstract
Liposomes have been widely used in nanomedicine for the delivery of hydrophobic and hydrophilic anticancer agents. The most common applications of these formulations are vaccines and anticancer formulations (e.g., mRNA, small molecule drugs). However, large-scale production with precise control of size and size distribution of the lipid-based drug delivery systems (DDSs) is one of the major challenges in the pharmaceutical industry. In this study, we used newly designed microfluidic swirl mixers with simple 3D mixing chamber structures to prepare liposomes at a larger scale (up to 320 mL/min or 20 L/h) than the commercially available devices. This design demonstrated high productivity and better control of liposome size and polydispersity index (PDI) than conventional liposome preparation methods. The microfluidic swirl mixer devices were used to produce curcumin-loaded liposomes under different processing conditions which were later characterized and studied in vitro to evaluate their efficiency as DDSs. The obtained results demonstrated that the liposomes can effectively deliver curcumin into cancer cells. Therefore, the microfluidic swirl mixers are promising devices for reproducible and scalable manufacturing of DDSs.
Collapse
Affiliation(s)
- Ruicheng Xu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Mhd Anas Tomeh
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Siyuan Ye
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Peng Zhang
- School of Materials Science and Engineering, Changzhou University, Changzhou 213164, China
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Rongrong You
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Nan Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
35
|
Cai D, Gao W, Li Z, Zhang Y, Xiao L, Xiao Y. Current Development of Nano-Drug Delivery to Target Macrophages. Biomedicines 2022; 10:1203. [PMID: 35625939 PMCID: PMC9139084 DOI: 10.3390/biomedicines10051203] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are the most important innate immune cells that participate in various inflammation-related diseases. Therefore, macrophage-related pathological processes are essential targets in the diagnosis and treatment of diseases. Since nanoparticles (NPs) can be preferentially taken up by macrophages, NPs have attracted most attention for specific macrophage-targeting. In this review, the interactions between NPs and the immune system are introduced to help understand the pharmacokinetics and biodistribution of NPs in immune cells. The current design and strategy of NPs modification for specific macrophage-targeting are investigated and summarized.
Collapse
Affiliation(s)
- Donglin Cai
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China;
| | - Wendong Gao
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
| | - Zhelun Li
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China;
| | - Yufeng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China;
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Yin Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical & Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (D.C.); (W.G.); (Z.L.)
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, Brisbane, QLD 4059, Australia
| |
Collapse
|
36
|
Baharoon B, Shaik A, El-Hamidy SM, Eid El-Araby R, Batawi AH, Abdel Salam M. Influence of halloysite nanotubes on the efficiency of Asparaginase against mice Ehrlich solid carcinoma. Saudi J Biol Sci 2022; 29:3626-3634. [PMID: 35844382 PMCID: PMC9280262 DOI: 10.1016/j.sjbs.2022.02.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/25/2022] Open
Abstract
Herein, the impact of the halloysite nanotubes to suppress the side effects of Asparaginase (ANase) cellular proliferation was investigated. Methods: A total of 100 adult male mice was employed. These mice were divided into four equal groups; Group 1 (control), Group 2 (ESC group) of a single dose of 0.15 ml Ehrlich cells (2 × 106) intraperitoneal infusion(IP), Group 3 (ESC + ANase group) received six doses equal treatments of Intratumoral (IT) 0.07 ml Aspragnase (7 mg/kg) over two weeks. For two weeks, Group 4 (ESC + ASNase + HNTs) received an IT administration of 0.07 ml Asparaginase stocked on Halloysite nanotubes (HNTs) (30 mg/kg) three times per week. A blood specimen was collected, and the liver was removed to be investigated histologically. Results: TEM measurements for the Halloysite nanoclay showed their tubular cylindrical shape with a mean diameter of 50 nm and an average length of 1 μm, whereas The X-ray diffraction pattern of the Halloysite nanoclay showed their characteristic peaks. ESC increases the serum levels of aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, and bilirubin than control and other groups, even as albumin and total protein were decreasing. After using Halloysite Nanotube, the rates of these variables were enhanced up to 75%. The hepatocytes histological studies showed protection against Ehrlich Solid carcinoma-induced degenerative, necrotic, and inflammatory changes up to 70%. In conclusion, halloysite nanotubes have demonstrated effective removal of Ehrlich solid carcinoma in mice using an ASNase delivery system. It promoted the ASNase to inhibit the adverse effect of ANase's on the liver and remove the tumour cells.
Collapse
Key Words
- ALB, Albumin
- ALP, Alkaline Phosphatase
- ALT, Aniline Aminotransferase
- ASNase, Asparaginase
- AST, Aspartate Aminotransferase
- Asparaginase
- BCP, bromocresol purple
- BD, Bile Duct
- CV, Central Vein
- DDS, Drug Delivery Systems
- EAC, Ehrlich ascites carcinoma
- ESC, Ehrlich Solid Carcenoma
- HNTs, Halloysite Nanotubes
- Halloysite nanotubes, Cancer
- Histopathology
- IFCC, international federation of clinical chemistry
- IM, Intramuscularly
- IP, Intraperitoneal
- IT, Intratumorally
- KAU, King Abdulaziz University
- KFMRC, King Fahd Medical Research Center
- Liver
- Liver functions
- PV, Portal Vein
- TBIL, Total Bilirubin
- TEM, Transmission Electron Microscope
- TP, Total protein
- XRD, X-Ray Diffraction
Collapse
Affiliation(s)
- B.M.M. Baharoon
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A.M. Shaik
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Dr. Najla Bint Saud Al Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salim M. El-Hamidy
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Dr. Najla Bint Saud Al Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rady Eid El-Araby
- Central Lab, Theodor Bilharz Research Institute (TBRI) Ministry of Scientific Research, Egypt
| | - Ashwaq H. Batawi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed Abdel Salam
- Department of Chemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
37
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
38
|
Chen W, Schilperoort M, Cao Y, Shi J, Tabas I, Tao W. Macrophage-targeted nanomedicine for the diagnosis and treatment of atherosclerosis. Nat Rev Cardiol 2022; 19:228-249. [PMID: 34759324 PMCID: PMC8580169 DOI: 10.1038/s41569-021-00629-x] [Citation(s) in RCA: 249] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Nanotechnology could improve our understanding of the pathophysiology of atherosclerosis and contribute to the development of novel diagnostic and therapeutic strategies to further reduce the risk of cardiovascular disease. Macrophages have key roles in atherosclerosis progression and, therefore, macrophage-associated pathological processes are important targets for both diagnostic imaging and novel therapies for atherosclerosis. In this Review, we highlight efforts in the past two decades to develop imaging techniques and to therapeutically manipulate macrophages in atherosclerotic plaques with the use of rationally designed nanoparticles. We review the latest progress in nanoparticle-based imaging modalities that can specifically target macrophages. Using novel molecular imaging technology, these modalities enable the identification of advanced atherosclerotic plaques and the assessment of the therapeutic efficacy of medical interventions. Additionally, we provide novel perspectives on how macrophage-targeting nanoparticles can deliver a broad range of therapeutic payloads to atherosclerotic lesions. These nanoparticles can suppress pro-atherogenic macrophage processes, leading to improved resolution of inflammation and stabilization of plaques. Finally, we propose future opportunities for novel diagnostic and therapeutic strategies and provide solutions to challenges in this area for the purpose of accelerating the clinical translation of nanomedicine for the treatment of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Wei Chen
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Liu C, Fan Z, He D, Chen H, Zhang S, Guo S, Zheng B, Cen H, Zhao Y, Liu H, Wang L. Designer Functional Nanomedicine for Myocardial Repair by Regulating the Inflammatory Microenvironment. Pharmaceutics 2022; 14:pharmaceutics14040758. [PMID: 35456592 PMCID: PMC9025700 DOI: 10.3390/pharmaceutics14040758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction is a major global health problem, and the repair of damaged myocardium is still a major challenge. Myocardial injury triggers an inflammatory response: immune cells infiltrate into the myocardium while activating myofibroblasts and vascular endothelial cells, promoting tissue repair and scar formation. Fragments released by cardiomyocytes become endogenous “danger signals”, which are recognized by cardiac pattern recognition receptors, activate resident cardiac immune cells, release thrombin factors and inflammatory mediators, and trigger severe inflammatory responses. Inflammatory signaling plays an important role in the dilation and fibrosis remodeling of the infarcted heart, and is a key event driving the pathogenesis of post-infarct heart failure. At present, there is no effective way to reverse the inflammatory microenvironment in injured myocardium, so it is urgent to find new therapeutic and diagnostic strategies. Nanomedicine, the application of nanoparticles for the prevention, treatment, and imaging of disease, has produced a number of promising applications. This review discusses the treatment and challenges of myocardial injury and describes the advantages of functional nanoparticles in regulating the myocardial inflammatory microenvironment and overcoming side effects. In addition, the role of inflammatory signals in regulating the repair and remodeling of infarcted hearts is discussed, and specific therapeutic targets are identified to provide new therapeutic ideas for the treatment of myocardial injury.
Collapse
Affiliation(s)
- Chunping Liu
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhijin Fan
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China;
| | - Dongyue He
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Huiqi Chen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Shihui Zhang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Sien Guo
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Bojun Zheng
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Huan Cen
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Yunxuan Zhao
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; (C.L.); (D.H.); (H.C.); (S.Z.); (S.G.); (B.Z.); (H.C.); (Y.Z.)
| | - Hongxing Liu
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510230, China
- Correspondence: (H.L.); (L.W.)
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
- Correspondence: (H.L.); (L.W.)
| |
Collapse
|
40
|
Hu Q, Fang Z, Ge J, Li H. Nanotechnology for cardiovascular diseases. Innovation (N Y) 2022; 3:100214. [PMID: 35243468 PMCID: PMC8866095 DOI: 10.1016/j.xinn.2022.100214] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/30/2022] [Accepted: 01/30/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases have become the major killers in today's world, among which coronary artery diseases (CADs) make the greatest contributions to morbidity and mortality. Although state-of-the-art technologies have increased our knowledge of the cardiovascular system, the current diagnosis and treatment modalities for CADs still have limitations. As an emerging cross-disciplinary approach, nanotechnology has shown great potential for clinical use. In this review, recent advances in nanotechnology in the diagnosis of CADs will first be elucidated. Both the sensitivity and specificity of biosensors for biomarker detection and molecular imaging strategies, such as magnetic resonance imaging, optical imaging, nuclear scintigraphy, and multimodal imaging strategies, have been greatly increased with the assistance of nanomaterials. Second, various nanomaterials, such as liposomes, polymers (PLGA), inorganic nanoparticles (AuNPs, MnO2, etc.), natural nanoparticles (HDL, HA), and biomimetic nanoparticles (cell-membrane coating) will be discussed as engineered as drug (chemicals, proteins, peptides, and nucleic acids) carriers targeting pathological sites based on their optimal physicochemical properties and surface modification potential. Finally, some of these nanomaterials themselves are regarded as pharmaceuticals for the treatment of atherosclerosis because of their intrinsic antioxidative/anti-inflammatory and photoelectric/photothermal characteristics in a complex plaque microenvironment. In summary, novel nanotechnology-based research in the process of clinical transformation could continue to expand the horizon of nanoscale technologies in the diagnosis and therapy of CADs in the foreseeable future.
Collapse
Affiliation(s)
- Qinqin Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zheyan Fang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
41
|
Gonciar D, Mocan T, Agoston-Coldea L. Nanoparticles Targeting the Molecular Pathways of Heart Remodeling and Regeneration. Pharmaceutics 2022; 14:pharmaceutics14040711. [PMID: 35456545 PMCID: PMC9028351 DOI: 10.3390/pharmaceutics14040711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiovascular diseases are the main cause of death worldwide, a trend that will continue to grow over the next decade. The heart consists of a complex cellular network based mainly on cardiomyocytes, but also on endothelial cells, smooth muscle cells, fibroblasts, and pericytes, which closely communicate through paracrine factors and direct contact. These interactions serve as valuable targets in understanding the phenomenon of heart remodeling and regeneration. The advances in nanomedicine in the controlled delivery of active pharmacological agents are remarkable and may provide substantial contribution to the treatment of heart diseases. This review aims to summarize the main mechanisms involved in cardiac remodeling and regeneration and how they have been applied in nanomedicine.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| | - Teodora Mocan
- Physiology Department, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca 400162, Romania
- Correspondence:
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| |
Collapse
|
42
|
Zeng Z, Gao H, Chen C, Xiao L, Zhang K. Bioresponsive Nanomaterials: Recent Advances in Cancer Multimodal Imaging and Imaging-Guided Therapy. Front Chem 2022; 10:881812. [PMID: 35372260 PMCID: PMC8971282 DOI: 10.3389/fchem.2022.881812] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/04/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer is a serious health problem which increasingly causes morbidity and mortality worldwide. It causes abnormal and uncontrolled cell division. Traditional cancer treatments include surgery, chemotherapy, radiotherapy and so on. These traditional therapies suffer from high toxicity and arouse safety concern in normal area and have difficulty in accurately targeting tumour. Recently, a variety of nanomaterials could be used for cancer diagnosis and therapy. Nanomaterials have several advantages, e.g., high concentration in tumour via targeting design, reduced toxicity in normal area and controlled drug release after various rational designs. They can combine with many types of biomaterials in order to improve biocompatibility. In this review, we outlined the latest research on the use of bioresponsive nanomaterials for various cancer imaging modalities (magnetic resonance imaging, positron emission tomography and phototacoustic imaging) and imaging-guided therapy means (chemotherapy, radiotherapy, photothermal therapy and photodynamic therapy), followed by discussing the challenges and future perspectives of this bioresponsive nanomaterials in biomedicine.
Collapse
Affiliation(s)
- Zeng Zeng
- Orthopedic Surgery Department, Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Huali Gao
- Orthopedic Surgery Department, Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - CongXian Chen
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Lianbo Xiao
- Orthopedic Surgery Department, Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kun Zhang
- Central Laboratory, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
43
|
Broad-Spectrum Theranostics and Biomedical Application of Functionalized Nanomaterials. Polymers (Basel) 2022; 14:polym14061221. [PMID: 35335551 PMCID: PMC8956086 DOI: 10.3390/polym14061221] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology is an important branch of science in therapies known as “nanomedicine” and is the junction of various fields such as material science, chemistry, biology, physics, and optics. Nanomaterials are in the range between 1 and 100 nm in size and provide a large surface area to volume ratio; thus, they can be used for various diseases, including cardiovascular diseases, cancer, bacterial infections, and diabetes. Nanoparticles play a crucial role in therapy as they can enhance the accumulation and release of pharmacological agents, improve targeted delivery and ultimately decrease the intensity of drug side effects. In this review, we discussthe types of nanomaterials that have various biomedical applications. Biomolecules that are often conjugated with nanoparticles are proteins, peptides, DNA, and lipids, which can enhance biocompatibility, stability, and solubility. In this review, we focus on bioconjugation and nanoparticles and also discuss different types of nanoparticles including micelles, liposomes, carbon nanotubes, nanospheres, dendrimers, quantum dots, and metallic nanoparticles and their crucial role in various diseases and clinical applications. Additionally, we review the use of nanomaterials for bio-imaging, drug delivery, biosensing tissue engineering, medical devices, and immunoassays. Understandingthe characteristics and properties of nanoparticles and their interactions with the biological system can help us to develop novel strategies for the treatment, prevention, and diagnosis of many diseases including cancer, pulmonary diseases, etc. In this present review, the importance of various kinds of nanoparticles and their biomedical applications are discussed in much detail.
Collapse
|
44
|
Karam M, Fahs D, Maatouk B, Safi B, Jaffa AA, Mhanna R. Polymeric nanoparticles in the diagnosis and treatment of myocardial infarction: Challenges and future prospects. Mater Today Bio 2022; 14:100249. [PMID: 35434594 PMCID: PMC9006854 DOI: 10.1016/j.mtbio.2022.100249] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/26/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of morbidity and mortality worldwide. Despite extensive efforts to provide early diagnosis and adequate treatment regimens, detection of MI still faces major limitations and pathological MI complications continue to threaten the recovery of survivors. Polymeric nanoparticles (NPs) represent novel noninvasive drug delivery systems for the diagnosis and treatment of MI and subsequent prevention of fatal heart failure. In this review, we cover the recent advances in polymeric NP-based diagnostic and therapeutic approaches for MI and their application as multifunctional theranostic tools. We also discuss the in vivo behavior and toxicity profile of polymeric NPs, their application in noninvasive imaging, passive, and active drug delivery, and use in cardiac regenerative therapy. We conclude with the challenges faced with polymeric nanosystems and suggest future efforts needed for clinical translation.
Collapse
Affiliation(s)
- Mia Karam
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Duaa Fahs
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Batoul Maatouk
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Brouna Safi
- Department of Chemical Engineering, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
| | - Ayad A. Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Lebanon
| |
Collapse
|
45
|
|
46
|
Tiwari A, Elgrably B, Saar G, Vandoorne K. Multi-Scale Imaging of Vascular Pathologies in Cardiovascular Disease. Front Med (Lausanne) 2022; 8:754369. [PMID: 35071257 PMCID: PMC8766766 DOI: 10.3389/fmed.2021.754369] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease entails systemic changes in the vasculature. The endothelial cells lining the blood vessels are crucial in the pathogenesis of cardiovascular disease. Healthy endothelial cells direct the blood flow to tissues as vasodilators and act as the systemic interface between the blood and tissues, supplying nutrients for vital organs, and regulating the smooth traffic of leukocytes into tissues. In cardiovascular diseases, when inflammation is sensed, endothelial cells adjust to the local or systemic inflammatory state. As the inflamed vasculature adjusts, changes in the endothelial cells lead to endothelial dysfunction, altered blood flow and permeability, expression of adhesion molecules, vessel wall inflammation, thrombosis, angiogenic processes, and extracellular matrix production at the endothelial cell level. Preclinical multi-scale imaging of these endothelial changes using optical, acoustic, nuclear, MRI, and multimodal techniques has progressed, due to technical advances and enhanced biological understanding on the interaction between immune and endothelial cells. While this review highlights biological processes that are related to changes in the cardiac vasculature during cardiovascular diseases, it also summarizes state-of-the-art vascular imaging techniques. The advantages and disadvantages of the different imaging techniques are highlighted, as well as their principles, methodologies, and preclinical and clinical applications with potential future directions. These multi-scale approaches of vascular imaging carry great potential to further expand our understanding of basic vascular biology, to enable early diagnosis of vascular changes and to provide sensitive diagnostic imaging techniques in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Betsalel Elgrably
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Saar
- Biomedical Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Katrien Vandoorne
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
47
|
Thanh Nguyen TD, Marasini R, Aryal S. Re-engineered imaging agent using biomimetic approaches. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1762. [PMID: 34698438 PMCID: PMC8758533 DOI: 10.1002/wnan.1762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023]
Abstract
Recent progress in biomedical technology, the clinical bioimaging, has a greater impact on the diagnosis, treatment, and prevention of disease, especially by early intervention and precise therapy. Varieties of organic and inorganic materials either in the form of small molecules or nano-sized materials have been engineered as a contrast agent (CA) to enhance image resolution among different tissues for the detection of abnormalities such as cancer and vascular occlusion. Among different innovative imaging agents, contrast agents coupled with biologically derived endogenous platform shows the promising application in the biomedical field, including drug delivery and bioimaging. Strategy using biocomponents such as cells or products of cells as a delivery system predominantly reduces the toxic behavior of its cargo, as these systems reduce non-specific distribution by navigating its cargo toward the targeted location. The hypothesis is that depending on the original biological role of the naïve cell, the contrast agents carried by such a system can provide corresponding natural designated behavior. Therefore, by combining properties of conventional synthetic molecules and nanomaterials with endogenous cell body, new solutions in the field of bioimaging to overcome biological barriers have been offered as innovative bioengineering. In this review, we will discuss the engineering of cell and cell-derived components as a delivery system for various contrast agents to achieve clinically relevant contrast for diagnosis and study underlining mechanism of disease progression. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Tuyen Duong Thanh Nguyen
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ramesh Marasini
- Department of Chemistry, Nanotechnology Innovation Center of Kansas State, Kansas State Univeristy, Manhattan, KS
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas 75799, USA
| |
Collapse
|
48
|
Soni SS, Rodell CB. Polymeric materials for immune engineering: Molecular interaction to biomaterial design. Acta Biomater 2021; 133:139-152. [PMID: 33484909 DOI: 10.1016/j.actbio.2021.01.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022]
Abstract
Biomaterials continue to evolve as complex engineered tools for interactively instructing biological systems, aiding in the understanding and treatment of various disease states through intimate biological interaction. The immune response to polymeric materials is a critical area of study, as it governs the body's response to biomaterial implants, drug delivery vehicles, and even therapeutic drug formulations. Importantly, the development of the immune response to polymeric biomaterials spans length scales - from single molecular interactions to the complex sensing of bulk biophysical properties, all of which coordinate a tissue- and systems-level response. In this review, we specifically discuss a bottom-up approach to designing biomaterials that use molecular-scale interactions to drive immune response to polymers and discuss how these interactions can be leveraged for biomaterial design. STATEMENT OF SIGNIFICANCE: The immune system is an integral controller of (patho)physiological processes, affecting nearly all aspects of human health and disease. Polymeric biomaterials, whether biologically derived or synthetically produced, can potentially alter the behavior of immune cells due to their molecular-scale interaction with individual cells, as well as their interpretation at the bulk scale. This article reviews common mechanisms by which immune cells interact with polymers at the molecular level and discusses how these interactions are being leveraged to produce the next generation of biocompatible and immunomodulatory materials.
Collapse
|
49
|
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
|
50
|
Zhang M, Gao S, Yang D, Fang Y, Lin X, Jin X, Liu Y, Liu X, Su K, Shi K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm Sin B 2021; 11:2265-2285. [PMID: 34522587 PMCID: PMC8424218 DOI: 10.1016/j.apsb.2021.03.033] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
The administration of nanoparticles (NPs) first faces the challenges of evading renal filtration and clearance of reticuloendothelial system (RES). After that, NPs infiltrate through the expanded endothelial space and penetrated the dense stroma of tumor microenvironment to tumor cells. As long as possible to prolong the time of NPs remaining in tumor tissue, NPs release active agent and induce pharmacological action. This review provides a comprehensive summary of the physical and chemical properties of NPs and the influence of various biological factors in tumor microenvironment, and discusses how to improve the final efficacy through adjusting the characteristics and structure of NPs. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Shi
- Corresponding author. Tel./fax: +86 24 43520557.
| |
Collapse
|