1
|
Ihnatsyeu-Kachan A, Sharko O, Bekish A, Saichuk A, Zhogla V, Abashkin V, Ulashchik E, Shcharbin D, Le Goff W, Kontush A, Guillas I, Shmanai V, Kim S. High-density lipoprotein-like nanoparticles with cationic cholesterol derivatives for siRNA delivery. BIOMATERIALS ADVANCES 2025; 170:214202. [PMID: 39923604 DOI: 10.1016/j.bioadv.2025.214202] [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: 10/12/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
A new approach to siRNA delivery using high-density lipoprotein-like nanoparticles (HDL NPs) was investigated, incorporating oligoamine and cholesterol-derived cationic lipids (CLs) to associate siRNA with the carrier. Newly designed or commercially available compounds, including GL67 and 3-β-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Cholesterol), were tested for siRNA binding, cytotoxicity, and siRNA cellular uptake. GL67 emerged as the most promising CL for siRNA delivery via HDL NPs. While it contributed to substantial siRNA uptake and cytosolic delivery in HepG2 cells, gene silencing remained limited, indicating a need for further optimization. Despite this, the study highlights the potential of positively charged cholesterol derivatives for siRNA delivery using HDL NPs. An analysis of the relationship between CL head group structure and HDL NPs' siRNA binding efficiency and cytotoxicity showed that factors such as oligoamine molecule conjugation site, linker type, amine group ethylation, and alkyl chain length between amine groups are crucial for optimizing CL design. Furthermore, the phospholipid environment surrounding CLs significantly influences HDL NPs' performance, particularly in siRNA cellular uptake. The study also revealed that intracellular siRNA trafficking varies by cell type, emphasizing the importance of customizing HDL NP formulations for specific cells. These insights are important for designing more effective HDL NPs for siRNA therapeutic delivery.
Collapse
Affiliation(s)
- Aliaksei Ihnatsyeu-Kachan
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02841, Republic of Korea; Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Olga Sharko
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Andrei Bekish
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Anastasiia Saichuk
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Victoriya Zhogla
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, 27 Academicheskaya Str., Minsk, 220072, Belarus
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, 27 Academicheskaya Str., Minsk, 220072, Belarus
| | - Egor Ulashchik
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, 27 Academicheskaya Str., Minsk, 220072, Belarus
| | - Wilfried Le Goff
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Anatol Kontush
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Isabelle Guillas
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Vadim Shmanai
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02841, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
2
|
Li Y, Xue X, Yu L, Qian J, Li X, Tian M, Yang J, Deng R, Lu C, Xiao C, Liu Y. Recombinant high-density lipoprotein targeted delivery of celastrol to promote foam cells lipophagy against early atherosclerosis. J Nanobiotechnology 2025; 23:237. [PMID: 40119460 PMCID: PMC11929195 DOI: 10.1186/s12951-025-03327-9] [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: 12/22/2024] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
INTRODUCTION Atherosclerosis serving as the main underlying factor of cardiovascular disease (CVD) remains the primary cause of mortality and morbidity globally, while the deposition of massive cholesterol in macrophage-derived foam cells exerts pivotal roles in the occurrence and progression of atherosclerosis. Celastrol (CEL) is a bioactive ingredient owning potent capability to modulate lipid metabolism, whereas the poor bioavailability and potential toxicity limit its clinical application. OBJECTIVES This study aims to design a CEL-loaded recombinant high-density lipoprotein (rHDL) delivery platform for active targeting, which may effectively promote lipid degradation in foam cells and reversely transport excessive cholesterol to the liver for metabolism in time. METHODS The rHDL loaded with CEL (CEL-rHDL) was prepared by the thin film dispersion method. Then the anti-atherosclerotic efficacy and targeted delivery to foam cells of atherosclerotic lesions were verified both in vitro and in vivo. RNA-sequence was applied to reveal the potential mechanism against early atherosclerosis, which was further validated through several molecular biology experiments. RESULTS The prepared CEL-rHDL increased the targeting efficiency to foam cells of atherosclerotic lesions, mitigated its off-target toxicity, and improved anti-atherosclerotic efficacy. Importantly, CEL-rHDL decreased lipid storage in foam cells by triggering lipophagy via the activation of Ca2+/CaMKKβ/AMPK/mTOR signaling pathway and reverse cholesterol transport (RCT). CONCLUSION A combination of hypolipidemic chemo-intervention with rHDL participated specific and reverse delivery may offer a promising strategy for biocompatible treatment of early atherosclerosis.
Collapse
Affiliation(s)
- Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxia Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jinxiu Qian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jue Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Rongjun Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
3
|
Fang M, Su B, Zhang S, Li F, Guo Y, Chen Q, Wu Y, Liu H, Jiang C, Sun T. Engineered Intelligent Microenvironment Responsive Prodrug Conjugates Navigated by Bioinspired Lipoproteins for Reversing Liver Fibrosis. SMALL METHODS 2025:e2402247. [PMID: 40103435 DOI: 10.1002/smtd.202402247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/23/2025] [Indexed: 03/20/2025]
Abstract
Liver fibrosis (LF) is characterized by excessive production of reactive oxygen species (ROS), abnormal activation of hepatic stellate cells (HSCs), and subsequent extracellular matrix (ECM) deposition. The complexity of multiple interrelated pathways involved in this process makes it challenging for monotherapy to achieve the desired therapeutic effects. To address this issue, this study designs a ROS-activated heterodimer conjugate (VTO) to collaboratively alleviate LF. Additionally, a biomimetic high-density lipoprotein is utilized for encapsulation, resulting in the formation of PL-VTO, which enables natural liver targeting. Once PL-VTO is delivered to the fibrotic liver, it can respond and release both parent drugs upon encountering the high ROS microenvironment, effectively scavenge ROS, induce quiescence of activated HSCs, and reduce collagen deposition, ultimately reversing LF. Overall, this study presents a feasible and versatile nanotherapeutic approach to enhance the prodrug-driven treatment of LF.
Collapse
Affiliation(s)
- Mingzhu Fang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Boyu Su
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shilin Zhang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Fangxin Li
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yun Guo
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuxing Wu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Huiyi Liu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Quzhou Fudan Institute, Quzhou, 324003, China
| |
Collapse
|
4
|
He J, Dai Y, Xu F, Huang X, Gao Y, Liu L, Zhang W, Liu J. High-density lipoprotein-based nanoplatforms for macrophage-targeted diagnosis and therapy of atherosclerosis. Int J Biol Macromol 2025; 306:140826. [PMID: 40010459 DOI: 10.1016/j.ijbiomac.2025.140826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/19/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Atherosclerosis, the primary cause of cardiovascular disease, which has the highest mortality worldwide, is a chronic inflammatory disease mainly induced by excessive lipid accumulation in plaque macrophages. Lipid-laden macrophages are crucial at all stages of atherosclerotic lesion progression and are, thus, regarded as popular therapeutic targets for atherosclerosis. High-density lipoprotein (HDL), an endogenous particle with excellent atherosclerotic plaque-homing properties, is considered a potential therapeutic agent for treating atherosclerosis. Based on the excellent properties of HDL, reconstituted HDL (rHDL), with physiological functions similar to those of its natural counterparts, have been successfully prepared as therapeutics and are also recognized as a potential nanoplatform for delivering drugs or contrast agents to atherosclerotic plaques owing to their high biocompatibility, amphiphilic structure, and macrophage-targeting capability. In this review, we focus on the (a) important role of macrophages in atherosclerotic lesions, (b) biological properties of rHDL as a delivery nanoplatform in atherosclerotic diseases, and (c) multiple applications of rHDL in the diagnosis and treatment of atherosclerosis. We systematically summarize the novel applications of rHDL with unique advantages in atherosclerosis, aiming to provide specific insights and inspire additional innovative research in this field.
Collapse
Affiliation(s)
- Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China; School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Fengfei Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xinya Huang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu Gao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Lisha Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
5
|
Huang J, Peng Y, Wang X, Gu X, Yi Y, Wang W, He Z, Ma Z, Feng Q, Qi W, Hui J, Gong R, Weng W, Jiang G, Gao Y, Lin Y, Li J, Jiang J, Feng J. Temperature induces brain-intake shift of recombinant high-density lipoprotein after traumatic brain injury. J Nanobiotechnology 2024; 22:769. [PMID: 39695696 DOI: 10.1186/s12951-024-03016-z] [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: 07/30/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
Traumatic brain injury (TBI) is one of the leading public health concerns in the world. Therapeutic hypothermia is routinely used in severe TBI, and pathophysiological hyperthermia, frequently observed in TBI patients, has an unclear impact on drug transport in the injured brain due to a lack of study on its effects. We investigated the effect of post-traumatic therapeutic hypothermia at 33°C and pathophysiological hyperthermia at 39°C on brain transport and cell uptake of neuroprotectants after TBI. Recombinant high-density lipoprotein (rHDL), which possesses anti-inflammatory, antioxidant activity, and blood-brain barrier (BBB) permeability, was chosen as the model drug. First, we found that mild hypothermia and hyperthermia impaired rHDL transport to the brain and lesion targeting in controlled cortical impact mice. Second, we investigated the temperature-induced rHDL uptake shift by various brain cell types. Mild hypothermia impeded the uptake of rHDL by endothelial cells, neurons, microglia, and astrocytes. Hyperthermia impeded the uptake of rHDL by endothelial cells and neurons while promoting its uptake by microglia and astrocytes. In an attempt to understand the mechanisms behind the above phenomena, it was found that temperature induced brain-intake shift of rHDL through the regulation of low-density lipoprotein receptor (LDLR) and LDLR-related protein 1 (LRP1) stability in brain cells. We therefore reported the full view of the temperature-induced brain-intake shift of rHDL after TBI for the first time. It would be of help in coordinating pharmacotherapy with temperature management in individualization and precision medicine.
Collapse
Affiliation(s)
- Jialin Huang
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Yidong Peng
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Xin Wang
- Department of Anesthesiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaokun Gu
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Yao Yi
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
- Department of Ophthalmology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wenye Wang
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Zhenghui He
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Zixuan Ma
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Qiyuan Feng
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Wenlan Qi
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Jiyuan Hui
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ru Gong
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Weiji Weng
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology Shanghai Universities Collaborative Innovation Center for Translational Medicine Shanghai, Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingwei Gao
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Yong Lin
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Jin Li
- Department of Ophthalmology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jiyao Jiang
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Shanghai Institute of Head Trauma, Shanghai, 200127, China.
| | - Junfeng Feng
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Shanghai Institute of Head Trauma, Shanghai, 200127, China.
| |
Collapse
|
6
|
Németh K, Kestecher BM, Ghosal S, Bodnár BR, Kittel Á, Hambalkó S, Kovácsházi C, Giricz Z, Ferdinandy P, Osteikoetxea X, Burkhardt R, Buzas EI, Orsó E. Therapeutic and pharmacological applications of extracellular vesicles and lipoproteins. Br J Pharmacol 2024; 181:4733-4749. [PMID: 39491825 DOI: 10.1111/bph.17336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/15/2024] [Accepted: 07/13/2024] [Indexed: 11/05/2024] Open
Abstract
In recent years, various approaches have been undertaken to eliminate lipoproteins co-isolated with extracellular vesicles, as they were initially regarded as contaminating entities. However, novel discoveries are reshaping our perspective. In body fluids, these distinct particles not only co-exist, but also interactions between them are likely to occur. Extracellular vesicles and lipoproteins can associate with each other, share cargo, influence each other's functions, and jointly have a role in the pathomechanisms of diseases. Additionally, their association carries important implications for therapeutic and pharmacological aspects of lipid-lowering strategies. Extracellular vesicles and lipoprotein particles may have roles in the elimination of each other from the circulation. The objective of this minireview is to delve into these aspects. Here, we show that under certain physiological and pathological conditions, extracellular vesicles and lipoproteins are 'partners' rather than 'strangers' or 'rivals'.
Collapse
Affiliation(s)
- Krisztina Németh
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
| | - Brachyahu M Kestecher
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Sayam Ghosal
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Bernadett R Bodnár
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ágnes Kittel
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HUN-REN, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Xabier Osteikoetxea
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Edit I Buzas
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, Budapest, Hungary
| | - Evelyn Orsó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
Lin J, Yu Z, Gao X. Advanced Noninvasive Strategies for the Brain Delivery of Therapeutic Proteins and Peptides. ACS NANO 2024; 18:22752-22779. [PMID: 39133564 DOI: 10.1021/acsnano.4c06851] [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: 08/28/2024]
Abstract
Recent years have witnessed rapid progress in the discovery of therapeutic proteins and peptides for the treatment of central nervous system (CNS) diseases. However, their clinical applications have been considerably hindered by challenges such as low biomembrane permeability, poor stability, short circulation time, and the formidable blood-brain barrier (BBB). Recently, substantial improvements have been made in understanding the dynamics of the BBB and developing efficient approaches for delivering proteins and peptides to the CNS, especially by using various nanoparticles. Herein, we present an overview of the up-to-date understanding of the BBB under physiological and pathological conditions, emphasizing their effects on brain drug delivery. We summarize advanced strategies and elucidate the underlying mechanisms for delivering proteins and peptides to the brain. We highlight the developments and applications of nanocarriers in treating CNS diseases via BBB crossing. We also provide critical opinions on the limitations and obstacles of the current strategies and put forward prospects for future research.
Collapse
Affiliation(s)
- Jiayuan Lin
- Department of Pharmacology and Chemical Biology, Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
8
|
Tang L, Yin Y, Zhang Z, Fu C, Cao Y, Liu H, Feng J, Gao J, Shang J, Wang W. Size-switchable and dual-targeting nanomedicine for cancer chemoimmunotherapy by potentiating deep tumor penetration and antitumor immunity. CHEMICAL ENGINEERING JOURNAL 2024; 493:152590. [DOI: 10.1016/j.cej.2024.152590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
9
|
He W, Wang M, Zhang X, Wang Y, Zhao D, Li W, Lei F, Peng M, Zhang Z, Yuan Y, Huang Z. Estrogen Induces LCAT to Maintain Cholesterol Homeostasis and Suppress Hepatocellular Carcinoma Development. Cancer Res 2024; 84:2417-2431. [PMID: 38718297 DOI: 10.1158/0008-5472.can-23-3966] [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: 12/13/2023] [Revised: 02/24/2024] [Accepted: 05/01/2024] [Indexed: 08/02/2024]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive disease that occurs predominantly in men. Estrogen elicits protective effects against HCC development. Elucidation of the estrogen-regulated biological processes that suppress HCC could lead to improved prevention and treatment strategies. Here, we performed transcriptomic analyses on mouse and human liver cancer and identified lecithin cholesterol acyltransferase (LCAT) as the most highly estrogen-upregulated gene and a biomarker of favorable prognosis. LCAT upregulation inhibited HCC in vitro and in vivo and mediated estrogen-induced suppression of HCC in an ESR1-dependent manner. LCAT facilitated high-density lipoprotein cholesterol production and uptake via the LDLR and SCARB1 pathways. Consistently, high HDL-C levels corresponded to a favorable prognosis in HCC patients. The enhanced HDL-C absorption induced by LCAT impaired SREBP2 maturation, which ultimately suppressed cholesterol biosynthesis and dampened HCC cell proliferation. HDL-C alone inhibited HCC growth comparably to the cholesterol-lowering drug lovastatin, and SREBF2 overexpression abolished the inhibitory activity of LCAT. Clinical observations and cross-analyses of multiple databases confirmed the correlation of elevated LCAT and HDL-C levels to reduced cholesterol synthesis and improved HCC patient prognosis. Furthermore, LCAT deficiency mimicked whereas LCAT overexpression abrogated the tumor growth-promoting effects of ovariectomy in HCC-bearing female mice. Most importantly, HDL-C and LCAT delayed the development of subcutaneous tumors in nude mice, and HDL-C synergized with lenvatinib to eradicate orthotopic liver tumors. Collectively, this study reveals that estrogen upregulates LCAT to maintain cholesterol homeostasis and to dampen hepatocarcinogenesis. LCAT and HDL-C represent potential prognostic and therapeutic biomarkers for targeting cholesterol homeostasis as a strategy for treating HCC. Significance: Estrogen mediates the sex differences in hepatocellular carcinoma development by reducing cholesterol biosynthesis through activation of an LCAT/HDL-C axis, providing strategies for improving liver cancer prevention, prognosis, and treatment.
Collapse
Affiliation(s)
- Wenzhi He
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, PR China
| | - Min Wang
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuechun Zhang
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yilan Wang
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dongli Zhao
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenhua Li
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fang Lei
- School of Basic Medicine, Institute of Model Animal, Wuhan University, Wuhan, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhonglin Zhang
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, PR China
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yufeng Yuan
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, PR China
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Zan Huang
- Hubei Key Laboratory of Cell Homeostasis, Department of Hepatobiliary and Pancreatic Surgery, College of Life Sciences, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Liu M, Feng Y, Lu Y, Huang R, Zhang Y, Zhao Y, Mo R. Lymph-targeted high-density lipoprotein-mimetic nanovaccine for multi-antigenic personalized cancer immunotherapy. SCIENCE ADVANCES 2024; 10:eadk2444. [PMID: 38478602 PMCID: PMC10936870 DOI: 10.1126/sciadv.adk2444] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/07/2024] [Indexed: 11/02/2024]
Abstract
Cancer vaccines show huge potential for cancer prevention and treatment. However, their efficacy remains limited due to weak immunogenicity regarding inefficient stimulation of cytotoxic T lymphocyte (CTL) responses. Inspired by the unique characteristic and biological function of high-density lipoprotein (HDL), we here develop an HDL-mimicking nanovaccine with the commendable lymph-targeted capacity to potently elicit antitumor immunity using lipid nanoparticle that is co-loaded with specific cancer cytomembrane harboring a collection of tumor-associated antigens and an immune adjuvant. The nanoparticulate impact is explored on the efficiency of lymphatic targeting and dendritic cell uptake. The optimized nanovaccine promotes the co-delivery of antigens and adjuvants to lymph nodes and maintains antigen presentation of dendritic cells, resulting in long-term immune surveillance as the elevated frequency of CTLs within lymphoid organs and tumor tissue. Immunization of nanovaccine suppresses tumor formation and growth and augments the therapeutic efficacy of checkpoint inhibitors notably on the high-stemness melanoma in the mouse models.
Collapse
Affiliation(s)
| | | | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yanan Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | | |
Collapse
|
11
|
Nair M, Chandra A, Krishnan A, Chandra A, Basha R, Orimoloye H, Raut S, Gayathri V, Mudgapalli VV, Vishwanatha JK. Protein and peptide nanoparticles for drug delivery applications. NANOSTRUCTURED MATERIALS FOR BIOMEDICAL APPLICATIONS 2024:339-404. [DOI: 10.1016/b978-0-323-90838-2.00011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Gao C, Liu Y, Zhang TL, Luo Y, Gao J, Chu JJ, Gong BF, Chen XH, Yin T, Zhang J, Yin Y. Biomembrane-Derived Nanoparticles in Alzheimer's Disease Therapy: A Comprehensive Review of Synthetic Lipid Nanoparticles and Natural Cell-Derived Vesicles. Int J Nanomedicine 2023; 18:7441-7468. [PMID: 38090364 PMCID: PMC10712251 DOI: 10.2147/ijn.s436774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Current therapies for Alzheimer's disease used in the clinic predominantly focus on reducing symptoms with limited capability to control disease progression; thus, novel drugs are urgently needed. While nanoparticles (liposomes, high-density lipoprotein-based nanoparticles) constructed with synthetic biomembranes have shown great potential in AD therapy due to their excellent biocompatibility, multifunctionality and ability to penetrate the BBB, nanoparticles derived from natural biomembranes (extracellular vesicles, cell membrane-based nanoparticles) display inherent biocompatibility, stability, homing ability and ability to penetrate the BBB, which may present a safer and more effective treatment for AD. In this paper, we reviewed the synthetic and natural biomembrane-derived nanoparticles that are used in AD therapy. The challenges associated with the clinical translation of biomembrane-derived nanoparticles and future perspectives are also discussed.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Yan Liu
- Department of Clinical Pharmacy, Shanghai Jiao Tong University of Medicine, Shanghai, People’s Republic of China
| | - Ting-Lin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Yi Luo
- Department of Clinical Pharmacy, Shanghai Jiao Tong University of Medicine, Shanghai, People’s Republic of China
- New Drug Discovery and Development, Biotheus Inc., Zhuhai, People’s Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Jian-Jian Chu
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Bao-Feng Gong
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Xiao-Han Chen
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Jian Zhang
- Department of Clinical Pharmacy, Shanghai Jiao Tong University of Medicine, Shanghai, People’s Republic of China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
13
|
Cao Z, Zhang Q, Zhou Z, Xu S, Pan B, Zhang S, Zhang G, Zhi Z, Shi Y, Cui L, Liu P. Construction and application of artificial lipoproteins using adiposomes. J Lipid Res 2023; 64:100436. [PMID: 37648212 PMCID: PMC10518588 DOI: 10.1016/j.jlr.2023.100436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Lipoproteins are complex particles comprised of a neutral lipid core wrapped with a phospholipid monolayer membrane and apolipoproteins on the membrane, which is closely associated with metabolic diseases. To facilitate the elucidation of its formation and dynamics, as well as its applications, we developed an in vitro system in which adiposomes, consisting of a hydrophobic core encircled by a monolayer-phospholipid membrane, were engineered into artificial lipoproteins (ALPs) by recruiting one or more kinds of apolipoproteins, for example, apolipoprotein (Apo) A-I, ApoE, ApoA-IV, and ApoB. In vitro and in vivo studies demonstrated the stability and biological activity of ALPs derived from adiposomes, which resembles native lipoproteins. Of note, adiposomes bearing ApoE were internalized via clathrin-mediated endocytosis following LDLR binding and were delivered to lysosomes. On the other hand, adiposomes bearing ApoA-IV mimicked the existing form of endogenous ApoA-IV and exhibited significant improvement in glucose tolerance in mice. In addition, the construction process was simple, precise, reproducible, as well as easy to adjust for mass production. With this experimental system, different apolipoproteins can be recruited to build ALPs for some biological goals and potential applications in biomedicine.
Collapse
Affiliation(s)
- Zhen Cao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ziyun Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Bin Pan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shuyan Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Infectious Diseases, Beijing, China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Gaoxin Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zelun Zhi
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yumeng Shi
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Liujuan Cui
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
14
|
Wang R, Zhang X, Feng K, Zeng W, Wu J, Sun D, Lu Z, Feng H, Di L. Nanotechnologies meeting natural sources: Engineered lipoproteins for precise brain disease theranostics. Asian J Pharm Sci 2023; 18:100857. [PMID: 37953874 PMCID: PMC10637878 DOI: 10.1016/j.ajps.2023.100857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/03/2023] [Accepted: 09/08/2023] [Indexed: 11/14/2023] Open
Abstract
Biological nanotechnologies have provided considerable opportunities in the management of malignancies with delicate design and negligible toxicity, from preventive and diagnostic to therapeutic fields. Lipoproteins, because of their inherent blood-brain barrier permeability and lesion-homing capability, have been identified as promising strategies for high-performance theranostics of brain diseases. However, the application of natural lipoproteins remains limited owing to insufficient accumulation and complex purification processes, which can be critical for individual therapeutics and clinical translation. To address these issues, lipoprotein-inspired nano drug-delivery systems (nano-DDSs), which have been learned from nature, have been fabricated to achieve synergistic drug delivery involving site-specific accumulation and tractable preparation with versatile physicochemical functions. In this review, the barriers in brain disease treatment, advantages of state-of-the-art lipoprotein-inspired nano-DDSs, and bio-interactions of such nano-DDSs are highlighted. Furthermore, the characteristics and advanced applications of natural lipoproteins and tailor-made lipoprotein-inspired nano-DDSs are summarized. Specifically, the key designs and current applications of lipoprotein-inspired nano-DDSs in the field of brain disease therapy are intensively discussed. Finally, the current challenges and future perspectives in the field of lipoprotein-inspired nano-DDSs combined with other vehicles, such as exosomes, cell membranes, and bacteria, are discussed.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Xinru Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Kuanhan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wei Zeng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jie Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Danni Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Ziyi Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Hao Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| |
Collapse
|
15
|
Yu H, Wu M, Chen S, Song M, Yue Y. Biomimetic nanoparticles for tumor immunotherapy. Front Bioeng Biotechnol 2022; 10:989881. [PMID: 36440446 PMCID: PMC9682960 DOI: 10.3389/fbioe.2022.989881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/11/2023] Open
Abstract
Currently, tumor treatment research still focuses on the cancer cells themselves, but the fact that the immune system plays an important role in inhibiting tumor development cannot be ignored. The activation of the immune system depends on the difference between self and non-self. Unfortunately, cancer is characterized by genetic changes in the host cells that lead to uncontrolled cell proliferation and evade immune surveillance. Cancer immunotherapy aims to coordinate a patient's immune system to target, fight, and destroy cancer cells without destroying the normal cells. Nevertheless, antitumor immunity driven by the autoimmune system alone may be inadequate for treatment. The development of drug delivery systems (DDS) based on nanoparticles can not only promote immunotherapy but also improve the immunosuppressive tumor microenvironment (ITM), which provides promising strategies for cancer treatment. However, conventional nano drug delivery systems (NDDS) are subject to several limitations in clinical transformation, such as immunogenicity and the potential toxicity risks of the carrier materials, premature drug leakage at off-target sites during circulation and drug load content. In order to address these limitations, this paper reviews the trends and progress of biomimetic NDDS and discusses the applications of each biomimetic system in tumor immunotherapy. Furthermore, we review the various combination immunotherapies based on biomimetic NDDS and key considerations for clinical transformation.
Collapse
Affiliation(s)
- Hanqing Yu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Wu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yulin Yue
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Akhtarkhavari T, Bahrami AR, M Matin M. Downregulation of miR-21 as a promising strategy to overcome drug resistance in cancer. Eur J Pharmacol 2022; 932:175233. [PMID: 36038011 DOI: 10.1016/j.ejphar.2022.175233] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Despite tremendous achievements in the field of targeted cancer therapy, chemotherapy is still the main treatment option, which is challenged by acquired drug resistance. Various microRNAs are involved in developing drug-resistant cells. miR-21 is one of the first identified miRNAs involved in this process. Here, we conducted a literature review to categorize different mechanisms employed by miR-21 to drive drug resistance. miR-21 targets various genes involved in many pathways that can justify chemoresistance. It alters cancer cell metabolism and facilitates adaptation to the new environment. It also enhances drug detoxification in cancerous cells and increases genomic instability. We also summarized various strategies applied for the inhibition of miR-21 in order to reverse cancer drug resistance. These strategies include the delivery of antagomiRs, miRZip knockdown vectors, inhibitory small molecules, CRISPR-Cas9 technology, catalytic nucleic acids, artificial DNA and RNA sponges, and nanostructures like mesoporous silica nanoparticles, dendrimers, and exosomes. Furthermore, current challenges and limitations in targeting miR-21 are discussed in this article. Although huge progress has been made in the downregulation of miR-21 in drug-resistant cancer cells, there are still many challenges to be resolved. More research is still required to find the best strategy and timeline for the downregulation of miR-21 and also the most feasible approach for the delivery of this system into the tumor cells. In conclusion, downregulation of miR-21 would be a promising strategy to reverse chemoresistance, but still, more studies are required to clarify the aforementioned issues.
Collapse
Affiliation(s)
- Tara Akhtarkhavari
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran.
| |
Collapse
|
17
|
Tang L, Fei Y, Su Y, Zhang A, Xiao Q, Mei Y, Su Y, Li Y, Li W, Wang T, Shen Y, Wang W. A neurovascular dual-targeting platelet-like bioinspired nanoplatform for ischemic stroke treatment. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
18
|
Johnson AP, Sabu C, Nivitha K, Sankar R, Shirin VA, Henna T, Raphey V, Gangadharappa H, Kotta S, Pramod K. Bioinspired and biomimetic micro- and nanostructures in biomedicine. J Control Release 2022; 343:724-754. [DOI: 10.1016/j.jconrel.2022.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/15/2022]
|
19
|
Zheng C, Zhang W, Wang J, Zhai Y, Xiong F, Cai Y, Gong X, Zhu B, Zhu HH, Wang H, Li Y, Zhang P. Lenvatinib- and vadimezan-loaded synthetic high-density lipoprotein for combinational immunochemotherapy of metastatic triple-negative breast cancer. Acta Pharm Sin B 2022; 12:3726-3738. [PMID: 36176911 PMCID: PMC9513558 DOI: 10.1016/j.apsb.2022.02.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 02/16/2022] [Indexed: 01/16/2023] Open
Abstract
Metastatic triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer. Combination of systemic chemotherapy and immune checkpoint blockade is effective but of limited benefit due to insufficient intratumoral infiltration of cytotoxic T lymphocytes (CTLs) and the accumulation of immunosuppressive cells. Herein, we designed a lenvatinib- and vadimezan-loaded synthetic high-density lipoprotein (LV-sHDL) for combinational immunochemotherapy of metastatic TNBC. The LV-sHDL targeted scavenger receptor class B type 1-overexpressing 4T1 cells in the tumor after intravenous injection. The multitargeted tyrosine kinase inhibitor (TKI) lenvatinib induced immunogenic cell death of the cancer cells, and the stimulator of interferon genes (STING) agonist vadimezan triggered local inflammation to facilitate dendritic cell maturation and antitumor macrophage differentiation, which synergistically improved the intratumoral infiltration of total and active CTLs by 33- and 13-fold, respectively. LV-sHDL inhibited the growth of orthotopic 4T1 tumors, reduced pulmonary metastasis, and prolonged the survival of animals. The efficacy could be further improved when LV-sHDL was used in combination with antibody against programmed cell death ligand 1. This study highlights the combination use of multitargeted TKI and STING agonist a promising treatment for metastatic TNBC.
Collapse
Affiliation(s)
- Chao Zheng
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen Zhang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jinming Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengqin Xiong
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Gong
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Hao Wang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- Corresponding authors. Tel./fax: +86 21 31779066 (Hao Wang); +86 21 20231979 (Yaping Li); +86 21 20231979 (Pengcheng Zhang).
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
- Corresponding authors. Tel./fax: +86 21 31779066 (Hao Wang); +86 21 20231979 (Yaping Li); +86 21 20231979 (Pengcheng Zhang).
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Corresponding authors. Tel./fax: +86 21 31779066 (Hao Wang); +86 21 20231979 (Yaping Li); +86 21 20231979 (Pengcheng Zhang).
| |
Collapse
|
20
|
Abstract
Hybrid membranes comprised of diblock copolymers, and phospholipids have gained interest due to their unique properties that result from blending natural and synthetic components. The integration of membrane proteins into these synthetic membranes is an important step towards creating biomembrane systems for uses such as artificial cellular systems, biosensors, and drug delivery vehicles. Here, we outline a technique to create hybrid membranes composed of phospholipids and diblock copolymers. Next, we describe how membrane proteins can be co-translationally integrated into hybrid lipid/polymer membranes using a cell-free reaction. We then outline a method to monitor insertion and folding of a membrane-embedded channel protein into the hybrid membrane using a fluorescent-protein reporter and dye release assay, respectively. This method is expected to be applicable for a wide range of membrane proteins that do not require chaperones for co-translational integration into vesicles and provides a generalized protocol for expressing a membrane protein into a membrane mimetic.
Collapse
Affiliation(s)
- Miranda L Jacobs
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
| | - Neha P Kamat
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
21
|
Zheng A, Dubuis G, Georgieva M, Ferreira CSM, Serulla M, Del Carmen Conde Rubio M, Trofimenko E, Mercier T, Decosterd L, Widmann C. HDLs extract lipophilic drugs from cells. J Cell Sci 2021; 135:273878. [PMID: 34981808 PMCID: PMC8919334 DOI: 10.1242/jcs.258644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022] Open
Abstract
High-density lipoproteins (HDLs) prevent cell death induced by a variety of cytotoxic drugs. The underlying mechanisms are however still poorly understood. Here, we present evidence that HDLs efficiently protect cells against thapsigargin (TG), a sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) inhibitor, by extracting the drug from cells. Drug efflux could also be triggered to some extent by low-density lipoproteins and serum. HDLs did not reverse the non-lethal mild ER stress response induced by low TG concentrations or by SERCA knockdown, but HDLs inhibited the toxic SERCA-independent effects mediated by high TG concentrations. HDLs could extract other lipophilic compounds, but not hydrophilic substances. This work shows that HDLs utilize their capacity of loading themselves with lipophilic compounds, akin to their ability to extract cellular cholesterol, to reduce the cell content of hydrophobic drugs. This can be beneficial if lipophilic xenobiotics are toxic but may be detrimental to the therapeutic benefit of lipophilic drugs such as glibenclamide. Summary: HDLs, akin to their capacity for extracting cholesterol, can remove lipophilic compounds from cells, thus protecting the cells when these compounds are toxic.
Collapse
Affiliation(s)
- Adi Zheng
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Gilles Dubuis
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Maria Georgieva
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | | | - Marc Serulla
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | | | - Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Thomas Mercier
- Laboratory of Clinical Pharmacology, Lausanne University Hospital (CHUV) and University of Lausanne, Switzerland
| | - Laurent Decosterd
- Laboratory of Clinical Pharmacology, Lausanne University Hospital (CHUV) and University of Lausanne, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| |
Collapse
|
22
|
Asghar S, Khan IU, Salman S, Khalid SH, Ashfaq R, Vandamme TF. Plant-derived nanotherapeutic systems to counter the overgrowing threat of resistant microbes and biofilms. Adv Drug Deliv Rev 2021; 179:114019. [PMID: 34699940 DOI: 10.1016/j.addr.2021.114019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/03/2021] [Accepted: 10/19/2021] [Indexed: 12/17/2022]
Abstract
Since antiquity, the survival of human civilization has always been threatened by the microbial infections. An alarming surge in the resistant microbial strains against the conventional drugs is quite evident in the preceding years. Furthermore, failure of currently available regimens of antibiotics has been highlighted by the emerging threat of biofilms in the community and hospital settings. Biofilms are complex dynamic composites rich in extracellular polysaccharides and DNA, supporting plethora of symbiotic microbial life forms, that can grow on both living and non-living surfaces. These enforced structures are impervious to the drugs and lead to spread of recurrent and non-treatable infections. There is a strong realization among the scientists and healthcare providers to work out alternative strategies to combat the issue of drug resistance and biofilms. Plants are a traditional but rich source of effective antimicrobials with wider spectrum due to presence of multiple constituents in perfect synergy. Other than the biocompatibility and the safety profile, these phytochemicals have been repeatedly proven to overcome the non-responsiveness of resistant microbes and films via multiple pathways such as blocking the efflux pumps, better penetration across the cell membranes or biofilms, and anti-adhesive properties. However, the unfavorable physicochemical attributes and stability issues of these phytochemicals have hampered their commercialization. These issues of the phytochemicals can be solved by designing suitably constructed nanoscaled structures. Nanosized systems can not only improve the physicochemical features of the encapsulated payloads but can also enhance their pharmacokinetic and therapeutic profile. This review encompasses why and how various types of phytochemicals and their nanosized preparations counter the microbial resistance and the biofouling. We believe that phytochemical in tandem with nanotechnological innovations can be employed to defeat the microbial resistance and biofilms. This review will help in better understanding of the challenges associated with developing such platforms and their future prospects.
Collapse
|
23
|
Zhang Q, He J, Xu F, Huang X, Wang Y, Zhang W, Liu J. Supramolecular copolymer modified statin-loaded discoidal rHDLs for atherosclerotic anti-inflammatory therapy by cholesterol efflux and M2 macrophage polarization. Biomater Sci 2021; 9:6153-6168. [PMID: 34346410 DOI: 10.1039/d1bm00610j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Foam cells with the pro-inflammatory macrophage phenotype (M1) play an essential role in atherosclerosis progression. Either cellular cholesterol removal or drug intervention was reported to polarize M1 into the anti-inflammatory phenotype (M2) for atherosclerosis regression. These might be realized simultaneously by drug-loaded discoidal reconstituted high-density lipoproteins (d-rHDLs) with the functions of cellular cholesterol efflux and targeted drug delivery on macrophages. However, cholesterol reception can drive the remodelling of d-rHDLs, which serves to release drugs specifically in the atherosclerotic plaque but might incur premature drug leakage in blood circulation. Given that, the proposed strategy is to inhibit the remodelling behaviour of the carrier in blood circulation and responsively accelerate it under the atherosclerotic microenvironmental stimulus. Herein, atorvastatin calcium-loaded d-rHDL was modified by a PEGylated ferrocene/β-cyclodextrin supramolecular copolymer (PF/TC) to construct ROS-responsive PF/TC-AT-d-rHDL, which is expected to possess plasma stability and biosafety as well as triggered drug release by cholesterol efflux promotion. As a result, PF/TC-AT-d-rHDL could responsively dissemble into β-cyclodextrin modified AT-d-rHDL under the ROS-triggered dissociation of PF/TC, therefore exhibiting increased cholesterol efflux from the cholesterol donor and drug release through the remodelling behaviour of the carrier in vitro. Moreover, PF/TC-AT-d-rHDL enhanced cellular cholesterol removal in foam cells after response to ROS, inhibiting intracellular lipid deposition compared with other d-rHDL carriers. Interestingly, cellular drug uptake was significantly promoted upon cellular cholesterol removal by restoring the permeability and fluidity of foam cell membranes as indicated by flow cytometry and fluorescence polarization analysis, respectively. Importantly, compared with untreated foam cells, PF/TC-AT-d-rHDL obviously increased the ratio of M2/M1 by 6.3-fold, which was even higher than the effect of PF/TC-d-rHDL (3.4-fold) and free drugs (1.9-fold), revealing that PF/TC-AT-d-rHDL synergistically promoted the M2 polarization of macrophages. Accordingly, PF/TC-AT-d-rHDL boosted the secretion of anti-inflammatory cytokines and inhibited that of inflammatory cytokines. Collectively, PF/TC-AT-d-rHDL exerted synergistic M2 polarization effects on foam cells for atherosclerotic immunomodulatory therapy via responsively mediating cholesterol efflux and delivering drugs.
Collapse
Affiliation(s)
- Qiqi Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Fengfei Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Xinya Huang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Yanyan Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China.
| |
Collapse
|
24
|
Rawal S, Patel M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. NANO-MICRO LETTERS 2021; 13:142. [PMID: 34138386 PMCID: PMC8196938 DOI: 10.1007/s40820-021-00630-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/23/2021] [Indexed: 05/03/2023]
Abstract
Lung cancer is a complex thoracic malignancy developing consequential to aberrations in a myriad of molecular and biomolecular signaling pathways. It is one of the most lethal forms of cancers accounting to almost 1.8 million new annual incidences, bearing overall mortality to incidence ratio of 0.87. The dismal prognostic scenario at advanced stages of the disease and metastatic/resistant tumor cell populations stresses the requisite of advanced translational interdisciplinary interventions such as bionanotechnology. This review article deliberates insights and apprehensions on the recent prologue of nanobioengineering and bionanotechnology as an approach for the clinical management of lung cancer. The role of nanobioengineered (bio-nano) tools like bio-nanocarriers and nanobiodevices in secondary prophylaxis, diagnosis, therapeutics, and theranostics for lung cancer management has been discussed. Bioengineered, bioinspired, and biomimetic bio-nanotools of considerate translational value have been reviewed. Perspectives on existent oncostrategies, their critical comparison with bio-nanocarriers, and issues hampering their clinical bench side to bed transformation have also been summarized.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India
| | - Mayur Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
25
|
Wang R, Yan H, Yu A, Ye L, Zhai G. Cancer targeted biomimetic drug delivery system. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
26
|
Asrorov AM, Gu Z, Li F, Liu L, Huang Y. Biomimetic camouflage delivery strategies for cancer therapy. NANOSCALE 2021; 13:8693-8706. [PMID: 33949576 DOI: 10.1039/d1nr01127h] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer remains a significant challenge despite the progress in developing different therapeutic approaches. Nanomedicine has been explored as a promising novel cancer therapy. Recently, biomimetic camouflage strategies have been investigated to change the bio-fate of therapeutics and target cancer cells while reducing the unwanted exposure on normal tissues. Endogenous components (e.g., proteins, polysaccharides, and cell membranes) have been used to develop anticancer drug delivery systems. These biomimetic systems can overcome biological barriers and enhance tumor cell-specific uptake. The tumor-targeting mechanisms include ligand-receptor interactions and stimuli-responsive (e.g., pH-sensitive and light-sensitive) delivery. Drug delivery carriers composed of endogenous components represent a promising approach for improving cancer treatment efficacy. In this paper, different biomimetic drug delivery strategies for cancer treatment are reviewed with a focus on the discussion of their advantages and potential applications.
Collapse
Affiliation(s)
- Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China. and Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, 83, M. Ulughbek Street, Tashkent 100125, Uzbekistan
| | - Zeyun Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Feng Li
- Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA.
| | - Lingyun Liu
- First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China. and Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan 528437, China and NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
| |
Collapse
|
27
|
Mei Y, Tang L, Xiao Q, Zhang Z, Zhang Z, Zang J, Zhou J, Wang Y, Wang W, Ren M. Reconstituted high density lipoprotein (rHDL), a versatile drug delivery nanoplatform for tumor targeted therapy. J Mater Chem B 2021; 9:612-633. [PMID: 33306079 DOI: 10.1039/d0tb02139c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
rHDL is a synthesized drug delivery nanoplatform exhibiting excellent biocompatibility, which possesses most of the advantages of HDL. rHDL shows almost no toxicity and can be degraded to non-toxic substances in vivo. The severe limitation of the application of various antitumor agents is mainly due to their low bioavailability, high toxicity, poor stability, etc. Favorably, antitumor drug-loaded rHDL nanoparticles (NPs), which are known as an important drug delivery system (DDS), help to change the situation a lot. This DDS shows an outstanding active-targeting ability towards tumor cells and improves the therapeutic effect during antitumor treatment while overcoming the shortcomings mentioned above. In the following text, we will mainly focus on the various applications of rHDL in tumor targeted therapy by describing the properties, preparation, receptor active-targeting ability and antitumor effects of antineoplastic drug-loaded rHDL NPs.
Collapse
Affiliation(s)
- Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Amaral M, Pereiro AB, Gaspar MM, Reis CP. Recent advances in ionic liquids and nanotechnology for drug delivery. Nanomedicine (Lond) 2020; 16:63-80. [PMID: 33356551 DOI: 10.2217/nnm-2020-0340] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In drug discovery and drug development, it is estimated that around 40% of commercialized and 90% of under-study drugs have inadequate pharmaceutical properties, severely impairing its therapeutic efficacy. Thus, there is a strong demand to find strategies to enhance the delivery of such drugs. Ionic liquids are a novel class of liquids composed of a combination of organic salts that are of particular interest alone or in combination with drug delivery systems. This review is focused on the recent efforts using ionic liquids in drug solubility, formulation and drug delivery with specific emphasis on nanotechnology. The latest developments using hybrid delivery systems obtained upon the combination of drug delivery systems and ionic liquids will also be addressed.
Collapse
Affiliation(s)
- Mariana Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, 1649-003, Portugal
| | - Ana B Pereiro
- LAQV, REQUIMTE, Department of Chemistry, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, 2829-516, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, 1649-003, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, 1649-003, Portugal.,IBEB, Institute of Biophysics & Biomedical Engineering, Faculdade de Ciências, Universidade de Lisboa, Lisboa, 1749-016, Portugal
| |
Collapse
|
29
|
Dogra A, Narang RS, Narang JK. Recent Advances in Nanotherapeutic Interventions for the Treatment of Alzheimer's Disease. Curr Pharm Des 2020; 26:2257-2279. [PMID: 32321393 DOI: 10.2174/1381612826666200422092620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 03/06/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD), with impairment of learning and memory as the common clinical manifestations, is one of the most challenging diseases affecting individuals, their families and society as a whole. The fact that its prevalence is escalating rapidly, with the total number of AD patients estimated to reach 115.4 million by 2050, has made the disease a very challenging ailment worldwide. Several biological barriers like the bloodbrain barrier (BBB), drug efflux by P-glycoprotein and the blood-cerebrospinal fluid barrier restrict the delivery of conventional AD drugs to the central nervous system (CNS), thereby limiting their effectiveness. In order to overcome the above physiological barriers, the development of nanomedicines has been extensively explored. The present review provides an insight into the pathophysiology of AD and risk factors associated with AD. Besides, various nanoformulations reported in the literature for the diagnosis and treatments of AD have been classified and summarised. The patented nanoformulations for AD and details of nanoformulations which are in clinical trials are also mentioned. The review would be helpful to researchers and scientific community by providing them with information related to the recent advances in nanointerventions for the diagnosis and treatment of AD, which they can further explore for better management of the disease. However, although the nanotherapeutics for managing AD have been extensively explored, the factors which hinder their commercialisation, the toxicity concern being one of them, need to be addressed so that effective nanotherapeutics for AD can be developed for clinical use.
Collapse
Affiliation(s)
- Anmol Dogra
- Department of Pharmaceutics, Khalsa College of Pharmacy, Amritsar, Punjab, India.,I.K. Gujral Punjab Technical University, Kapurthala, Punjab, India
| | - R S Narang
- Department of Oral & Maxillofacial Pathology and Microbiology, Sri Guru Ram Das Institute of Dental Sciences and Research, Amritsar, Punjab, India
| | - Jasjeet K Narang
- Department of Pharmaceutics, Khalsa College of Pharmacy, Amritsar, Punjab, India
| |
Collapse
|
30
|
Krishna KV, Saha RN, Dubey SK. Biophysical, Biochemical, and Behavioral Implications of ApoE3 Conjugated Donepezil Nanomedicine in a Aβ 1-42 Induced Alzheimer's Disease Rat Model. ACS Chem Neurosci 2020; 11:4139-4151. [PMID: 33251785 DOI: 10.1021/acschemneuro.0c00430] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder and is the most common type of dementia. Amyloid β (Aβ) plaques play an important role in the pathophysiology of AD. However, the existing therapeutic strategies are not effective for the management of both Aβ-induced neurotoxicity and Aβ fibrils clearance in biological conditions. Herein, we have developed lipoprotein conjugated polymeric nanoparticles that can boost the clearance rate of Aβ fibrils and mitigate Aβ-induced neurotoxicity in AD rat. These nanoparticles were designed by loading donepezil in an amphiphilic polymer with a lipoprotein (ApoE3) integrated over the surface. Polymeric nanoparticles were prepared by a nanoprecipitation method, and ApoE3 was conjugated to the polymer layer by polysorbate 80. In the present study, we intended to examine the protective effect of ApoE3 nanoparticles against Aβ-induced neurotoxicity both in vitro and in vivo to evaluate if these can reduce the Aβ fibril formation and cognitive and behavioral deficits observed in AD induced rats. In the in vitro study, neurotoxicity induced by Aβ1-42 in human neuroblastoma (SH-SY5Y) cells was found to be significantly reduced upon treatment with ApoE3 donepezil nanoparticles. The presence of the ApoE3 significantly modified the morphology of Aβ fibrils and also inhibited the formation Aβ oligomers. Moreover, in the in vivo study, following treatment, AD induced rats were tested on Morris water maze (MWM) and passive avoidance task for their cognitive ability and sacrificed for biochemical estimations. From our observations, ApoE3 donepezil nanoparticles exhibited neuroprotection in the Aβ1-42 induced model by mitigating the pathological features and cognitive impairments. Thus, we anticipate that the nanosized lipoprotein carriers will possibly offer a rational therapeutic strategy in the formulation development of AD.
Collapse
Affiliation(s)
- Kowthavarapu Venkata Krishna
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, 345055 Pilani, Rajasthan, India
| | - Ranendra Narayan Saha
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, 345055 Pilani, Rajasthan, India
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Dubai Campus, Dubai International Academic City, P.O. Box 345055, Dubai, United Arab Emirates
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, 345055 Pilani, Rajasthan, India
| |
Collapse
|
31
|
Pedersbæk D, Simonsen JB. A systematic review of the biodistribution of biomimetic high-density lipoproteins in mice. J Control Release 2020; 328:792-804. [PMID: 32971201 DOI: 10.1016/j.jconrel.2020.09.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/18/2022]
Abstract
For the past two decades, biomimetic high-density lipoproteins (b-HDL) have been used for various drug delivery applications. The b-HDL mimic the endogenous HDL, and therefore possess many attractive features for drug delivery, including high biocompatibility, biodegradability, and ability to transport and deliver their cargo (e.g. drugs and/or imaging agents) to specific cells and tissues that are recognized by HDL. The b-HDL designs reported in the literature often differ in size, shape, composition, and type of incorporated cargo. However, there exists only limited insight into how the b-HDL design dictates their biodistribution. To fill this gap, we conducted a comprehensive systematic literature search of biodistribution studies using various designs of apolipoprotein A-I (apoA-I)-based b-HDL (i.e. b-HDL with apoA-I, apoA-I mutants, or apoA-I mimicking peptides). We carefully screened 679 papers (search hits) for b-HDL biodistribution studies in mice, and ended up with 24 relevant biodistribution profiles that we compared according to b-HDL design. We show similarities between b-HDL biodistribution studies irrespectively of the b-HDL design, whereas the biodistribution of the b-HDL components (lipids and scaffold) differ significantly. The b-HDL lipids primarily accumulate in liver, while the b-HDL scaffold primarily accumulates in the kidney. Furthermore, both b-HDL lipids and scaffold accumulate well in the tumor tissue in tumor-bearing mice. Finally, we present essential considerations and strategies for b-HDL labeling, and discuss how the b-HDL biodistribution can be tuned through particle design and administration route. Our meta-analysis and discussions provide a detailed overview of the fate of b-HDL in mice that is highly relevant when applying b-HDL for drug delivery or in vivo imaging applications.
Collapse
Affiliation(s)
- Dennis Pedersbæk
- Technical University of Denmark, Department of Health Technology, 2800 Kgs. Lyngby, Denmark
| | - Jens B Simonsen
- Technical University of Denmark, Department of Health Technology, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
32
|
Revilla G, Cedó L, Tondo M, Moral A, Pérez JI, Corcoy R, Lerma E, Fuste V, Reddy ST, Blanco-Vaca F, Mato E, Escolà-Gil JC. LDL, HDL and endocrine-related cancer: From pathogenic mechanisms to therapies. Semin Cancer Biol 2020; 73:134-157. [PMID: 33249202 DOI: 10.1016/j.semcancer.2020.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/19/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Cholesterol is essential for a variety of functions in endocrine-related cells, including hormone and steroid production. We have reviewed the progress to date in research on the role of the main cholesterol-containing lipoproteins; low-density lipoprotein (LDL) and high-density lipoprotein (HDL), and their impact on intracellular cholesterol homeostasis and carcinogenic pathways in endocrine-related cancers. Neither LDL-cholesterol (LDL-C) nor HDL-cholesterol (HDL-C) was consistently associated with endocrine-related cancer risk. However, preclinical studies showed that LDL receptor plays a critical role in endocrine-related tumor cells, mainly by enhancing circulating LDL-C uptake and modulating tumorigenic signaling pathways. Although scavenger receptor type BI-mediated uptake of HDL could enhance cell proliferation in breast, prostate, and ovarian cancer, these effects may be counteracted by the antioxidant and anti-inflammatory properties of HDL. Moreover, 27-hydroxycholesterol a metabolite of cholesterol promotes tumorigenic processes in breast and epithelial thyroid cancer. Furthermore, statins have been reported to reduce the incidence of breast, prostate, pancreatic, and ovarian cancer in large clinical trials, in part because of their ability to lower cholesterol synthesis. Overall, cholesterol homeostasis deregulation in endocrine-related cancers offers new therapeutic opportunities, but more mechanistic studies are needed to translate the preclinical findings into clinical therapies.
Collapse
Affiliation(s)
- Giovanna Revilla
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, C/ Antoni M. Claret 167, 08025 Barcelona, Spain
| | - Lídia Cedó
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Mireia Tondo
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Antonio Moral
- Department of General Surgery, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, C/ Antoni M. Claret 167, 08025 Barcelona, Spain
| | - José Ignacio Pérez
- Department of General Surgery, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Rosa Corcoy
- Departament de Medicina, Universitat Autònoma de Barcelona, C/ Antoni M. Claret 167, 08025 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain; Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Enrique Lerma
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Department of Anatomic Pathology, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Victoria Fuste
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Department of Anatomic Pathology, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain
| | - Srivinasa T Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Francisco Blanco-Vaca
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, C/ Sant Quintí 89, 08041 Barcelona, Spain.
| | - Eugènia Mato
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, C/ Sant Quintí 77, 08041 Barcelona Spain.
| |
Collapse
|
33
|
Delk SC, Chattopadhyay A, Escola-Gil JC, Fogelman AM, Reddy ST. Apolipoprotein mimetics in cancer. Semin Cancer Biol 2020; 73:158-168. [PMID: 33188891 DOI: 10.1016/j.semcancer.2020.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/10/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022]
Abstract
Peptides have many advantages over traditional therapeutics, including small molecules and other biologics, because of their low toxicity and immunogenicity, while still exhibiting efficacy. This review discusses the benefits and mechanism of action of apolipoprotein mimetic peptides in tumor biology and their potential utility in treating various cancers. Among lipoproteins in the circulation, high-density lipoprotein (HDL) and its constituents including apolipoprotein A-I (apoA-I; the predominant protein in HDL), apoJ, and apoE, harbor anti-tumorigenic activities. Peptides that mimic apoA-I function have been developed through molecular mimicry of the amphipathic α-helices of apoA-I. Oral apoA-I mimetic peptides remodel HDL, promote cholesterol efflux, sequester oxidized lipids, and activate anti-inflammatory processes. ApoA-I and apoJ mimetic peptides ameliorate various metrics of cancer progression and have demonstrated efficacy in preclinical models in the inhibition of ovarian, colon, breast, and metastatic lung cancers. Apolipoprotein mimetic peptides are poorly absorbed when administered orally and rapidly degraded when injected into the circulation. The small intestine is the major site of action for apoA-I mimetic peptides and recent studies suggest that modulation of immune cells in the lamina propria of the small intestine is, in part, a potential mechanism of action. Finally, several recent studies underscore the use of reconstituted HDL as target-specific nanoparticles carrying poorly soluble or unstable therapeutics to tumors even across the blood-brain barrier. Preclinical studies suggest that these versatile recombinant lipoprotein based nanoparticles and apolipoprotein mimetics can serve as safe, novel drug delivery, and therapeutic agents for the treatment of a number of cancers.
Collapse
Affiliation(s)
- Samuel C Delk
- Molecular Toxicology Interdepartmental Degree Program, Fielding School of Public Health, University of California, Los Angeles, CA, 90095, USA; Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Arnab Chattopadhyay
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Joan Carles Escola-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Sant Quintí 77, 08041, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Monforte de Lemos 3-5, 28029, Madrid, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Antoni M. Claret 167, 08025, Barcelona, Spain
| | - Alan M Fogelman
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Srinivasa T Reddy
- Molecular Toxicology Interdepartmental Degree Program, Fielding School of Public Health, University of California, Los Angeles, CA, 90095, USA; Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA; Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
34
|
Wünsch A, Mulac D, Langer K. Lipoprotein imitating nanoparticles: Lecithin coating binds ApoE and mediates non-lysosomal uptake leading to transcytosis over the blood-brain barrier. Int J Pharm 2020; 589:119821. [DOI: 10.1016/j.ijpharm.2020.119821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022]
|
35
|
He W, Xing X, Wang X, Wu D, Wu W, Guo J, Mitragotri S. Nanocarrier‐Mediated Cytosolic Delivery of Biopharmaceuticals. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201910566] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/16/2020] [Indexed: 01/04/2025]
Abstract
AbstractBiopharmaceuticals have emerged to play a vital role in disease treatment and have shown promise in the rapidly expanding pharmaceutical market due to their high specificity and potency. However, the delivery of these biologics is hindered by various physiological barriers, owing primarily to the poor cell membrane permeability, low stability, and increased size of biologic agents. Since many biological drugs are intended to function by interacting with intracellular targets, their delivery to intracellular targets is of high relevance. In this review, the authors summarize and discuss the use of nanocarriers for intracellular delivery of biopharmaceuticals via endosomal escape and, especially, the routes of direct cytosolic delivery by means including the caveolae‐mediated pathway, contact release, intermembrane transfer, membrane fusion, direct translocation, and membrane disruption. Strategies with high potential for translation are highlighted. Finally, the authors conclude with the clinical translation of promising carriers and future perspectives.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xuyang Xing
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xiaoling Wang
- School of Biomass Science and Engineering Sichuan University Chengdu 610065 China
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of Ministry of Education of China School of Pharmacy Fudan University Shanghai 201203 China
| | - Junling Guo
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| |
Collapse
|
36
|
Busatto S, Walker SA, Grayson W, Pham A, Tian M, Nesto N, Barklund J, Wolfram J. Lipoprotein-based drug delivery. Adv Drug Deliv Rev 2020; 159:377-390. [PMID: 32791075 PMCID: PMC7747060 DOI: 10.1016/j.addr.2020.08.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/01/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Lipoproteins (LPs) are circulating heterogeneous nanoparticles produced by the liver and intestines. LPs play a major role in the transport of dietary and endogenous lipids to target cells through cell membrane receptors or cell surface-bound lipoprotein lipase. The stability, biocompatibility, and selective transport of LPs make them promising delivery vehicles for various therapeutic and imaging agents. This review discusses isolation, manufacturing, and drug loading techniques used for LP-based drug delivery, as well as recent applications for diagnosis and treatment of cancer, atherosclerosis, and other life-threatening diseases.
Collapse
Affiliation(s)
- Sara Busatto
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Sierra A Walker
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Whisper Grayson
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Anthony Pham
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ming Tian
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nicole Nesto
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Jacqueline Barklund
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
37
|
Chen L, Song Q, Chen Y, Meng S, Zheng M, Huang J, Zhang Q, Jiang J, Feng J, Chen H, Jiang G, Gao X. Tailored Reconstituted Lipoprotein for Site-Specific and Mitochondria-Targeted Cyclosporine A Delivery to Treat Traumatic Brain Injury. ACS NANO 2020; 14:6636-6648. [PMID: 32464051 DOI: 10.1021/acsnano.9b09186] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The secondary damage in traumatic brain injury (TBI) can lead to lifelong disabilities, bringing enormous economic and psychological burden to patients and their families. Mitochondria, as the core mediator of the secondary injury cascade reaction in TBI, is an important target to prevent the spread of cell death and dysfunction. Thus, therapeutics that can accumulate at the damaged sites and subsequently rescue the functions of mitochondria would largely improve the outcome of TBI. Cyclosporine A (CsA), which can maintain the integrity of mitochondrial function, is among the most promising neuroprotective therapeutics for TBI treatment. However, the clinical application of CsA in TBI is largely hindered because of its poor access to the targets. Here, to realize targeted intracellular CsA delivery, we designed a lipoprotein biomimetic nanocarrier by incorporating CsA in the core and decorating a matrix metalloproteinase-9 activatable cell-penetrating peptide onto the surface of the lipoprotein-mimic nanocarrier. This CsA-loaded tailored reconstituted lipoprotein efficiently accumulated at the damaged brain sites, entered the target cells, bound to the membrane of mitochondria, more efficiently reduced neuronal damage, alleviated neuroinflammation, and rescued memory deficits at the dose 1/16 of free CsA in a controlled cortical impact injury mice model. The findings provide strong evidence that the secondary damages in TBI can be well controlled through targeted CsA delivery and highlight the potential of a lipoprotein biomimetic nanocarrier as a flexible nanoplatform for the management of TBI.
Collapse
Affiliation(s)
- Lepei Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yaoxing Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Shuang Meng
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Mengna Zheng
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Jialin Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
- Department of Neurological Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, China
| | - Qian Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Jiyao Jiang
- Department of Neurological Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, China
| | - Junfeng Feng
- Department of Neurological Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
- Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201210, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
38
|
Datta P, Ray S. Nanoparticulate formulations of radiopharmaceuticals: Strategy to improve targeting and biodistribution properties. J Labelled Comp Radiopharm 2020; 63:333-355. [PMID: 32220029 DOI: 10.1002/jlcr.3839] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/17/2020] [Accepted: 03/08/2020] [Indexed: 02/06/2023]
Abstract
Application of nanotechnology principles in drug delivery has created opportunities for treatment of several diseases. Nanotechnology offers the advantage of overcoming the adverse biopharmaceutics or pharmacokinetic properties of drug molecules, to be determined by the transport properties of the particles themselves. Through the manipulation of size, shape, charge, and type of nanoparticle delivery system, variety of distribution profiles may be obtained. However, there still exists greater need to derive and standardize definitive structure property relationships for the distribution profiles of the delivery system. When applied to radiopharmaceuticals, the delivery systems assume greater significance. For the safety and efficacy of both diagnostics and therapeutic radiopharmaceuticals, selective localization in target tissue is even more important. At the same time, the synthesis and fabrication reactions of radiolabelled nanoparticles need to be completed in much shorter time. Moreover, the extensive understanding of the several interesting optical and magnetic properties of materials in nanoscale provides for achieving multiple objectives in nuclear medicine. This review discusses the various nanoparticle systems, which are applied for radionuclides and analyses the important bottlenecks that are required to be overcome for their more widespread clinical adaptation.
Collapse
Affiliation(s)
- Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, India
| | | |
Collapse
|
39
|
Tan T, Wang Y, Wang J, Wang Z, Wang H, Cao H, Li J, Li Y, Zhang Z, Wang S. Targeting peptide-decorated biomimetic lipoproteins improve deep penetration and cancer cells accessibility in solid tumor. Acta Pharm Sin B 2020; 10:529-545. [PMID: 32140397 PMCID: PMC7049576 DOI: 10.1016/j.apsb.2019.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022] Open
Abstract
The limited penetration of nanoparticles and their poor accessibility to cancer cell fractions in tumor remain essential challenges for effective anticancer therapy. Herein, we designed a targeting peptide-decorated biomimetic lipoprotein (termed as BL-RD) to enable their deep penetration and efficient accessibility to cancer cell fractions in a tumor, thereby improving the combinational chemo-photodynamic therapy of triple negative breast cancer. BL-RD was composed of phospholipids, apolipoprotein A1 mimetic peptide (PK22), targeting peptide-conjugated cytotoxic mertansine (RM) and photodynamic agents of DiIC18(5) (DiD). The counterpart biomimetic lipoprotein system without RM (termed as BL-D) was fabricated as control. Both BL-D and BL-RD were nanometer-sized particles with a mean diameter of less than 30 nm and could be efficiently internalized by cancer cells. After intravenous injection, they can be specifically accumulated at tumor sites. When comparing to the counterpart BL-D, BL-RD displayed superior capability to permeate across the tumor mass, extravasate from tumor vasculature to distant regions and efficiently access the cancer cell fractions in a solid tumor, thus producing noticeable depression of the tumor growth. Taken together, BL-RD can be a promising delivery nanoplatform with prominent tumor-penetrating and cancer cells-accessing capability for effective tumor therapy.
Collapse
Key Words
- 4T1-GFP, 4T1 cancer cells with stable expression of green fluorescence protein
- ApoA1, apolipoprotein A1
- BL-D, biomimetic lipoprotein system without targeting peptide
- BL-RD, targeting peptide decorated biomimetic lipoprotein system
- CAF, cancer-associated fibroblasts
- CLSM, confocal laser scanning microscopy
- Cancer therapy
- DAPI, 4′,6-diamidino-2-phenylindole
- DCFH-DA, 2′,7′-dichlorodihydrofluorescein diacetate
- DOPC, 1,2-dioleoyl-sn-glycero-3-phosphocholine
- DiD, DiIC18(5)
- Drug delivery
- EC, endothelial cells
- ECM, extracellular matrix
- EE, encapsulation efficiency
- FBS, fetal bovine serum
- GSH, glutathione
- H&E staining, hematoxylin-eosin staining
- HDL, high density lipoprotein
- HPLC, high performance liquid chromatography
- IC50, half-inhibitory concentration
- Lipo-D, liposome system without targeting peptide
- Lipo-RD, targeting peptide decorated biomimetic lipoprotein system
- Lipoprotein
- MCS, multicellular spheroids
- MTT, thiazolyl blue tetrazolium bromide
- Nanoparticles
- PBS, phosphate buffered solution
- PDT, photodynamic therapy
- RM, targeting peptide-conjugated cytotoxic mertansine
- ROS, reactive oxygen species
- SOSG, singlet oxygen sensor green
- TAM, tumor-associated macrophage
- TEM, transmission electronic microscope
- TGI, tumor growth index
- Tumor penetration
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Tao Tan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuqi Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwan Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hong Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haiqiang Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| | - Siling Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors. Tel./fax: +86 21 20231979.
| |
Collapse
|
40
|
Singh A, Myklebust NN, Furevik SMV, Haugse R, Herfindal L. Immunoliposomes in Acute Myeloid Leukaemia Therapy: An Overview of Possible Targets and Obstacles. Curr Med Chem 2019; 26:5278-5292. [PMID: 31099318 DOI: 10.2174/0929867326666190517114450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/30/2022]
Abstract
Acute Myeloid Leukaemia (AML) is the neoplastic transformation of Hematopoietic Stem Cells (HSC) and relapsed disease is a major challenge in the treatment. Despite technological advances in the field of medicine and our heightened knowledge regarding the pathogenesis of AML, the initial therapy of "7+3" Cytarabine and Daunorubicin has remained mainly unchanged since 1973. AML is a disease of the elderly, and increased morbidity in this patient group does not allow the full use of the treatment and drug-resistant relapse is common. Nanocarriers are drug-delivery systems that can be used to transport drugs to the bone marrow and target Leukemic Stem Cells (LSC), conferring less side-effects compared to the free-drug alternative. Nanocarriers also can be used to favour the transport of drugs that otherwise would not have been used clinically due to toxicity and poor efficacy. Liposomes are a type of nanocarrier that can be used as a dedicated drug delivery system, which can also have active ligands on the surface in order to interact with antigens on the target cells or tissues. In addition to using small molecules, it is possible to attach antibodies to the liposome surface, generating so-called immunoliposomes. By using immunoliposomes as a drug-delivery system, it is possible to minimize the toxic side effects caused by the chemotherapeutic drug on healthy organs, and at the same time direct the drugs towards the remaining AML blasts and stem cells. This article aims to explore the possibilities of using immunoliposomes as a drug carrier in AML therapy. Emphasis will be on possible target molecules on the AML cells, leukaemic stem cells, as well as bone marrow constituents relevant to AML therapy. Further, some conditions and precautions that must be met for immunoliposomes to be used in AML therapy will be discussed.
Collapse
Affiliation(s)
- Aditi Singh
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Sarah Marie Vie Furevik
- Hospital pharmacies enterprise, Western Norway, Bergen, Norway.,Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ragnhild Haugse
- Hospital pharmacies enterprise, Western Norway, Bergen, Norway.,Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars Herfindal
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
41
|
Distribution of Psychotropic Drugs Into Lipoproteins. Ther Drug Monit 2019; 41:766-771. [PMID: 31725695 DOI: 10.1097/ftd.0000000000000660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM The aim of this pilot study was to investigate whether psychotropic drugs frequently analyzed in a routine therapeutic drug monitoring laboratory bind to low-density lipoproteins/very-low-density lipoproteins (LDL/VLDL) in human serum. METHODS Drug concentrations in 20 serum sample pools containing one psychotropic drug each, and in the LDL/VLDL fractions extracted from the same samples, were measured by triple quadrupole liquid chromatography tandem mass spectrometry. The membrane permeability of the drugs was measured using a Parallel Artificial Membrane Permeability Assay. RESULTS Of the 20 antidepressants, antipsychotics, and antiepileptics examined, 7 drugs were detected in both the pooled serum samples and in the LDL/VLDL fraction. Binding of drugs to LDL/VLDL significantly correlated with high octanol: water partition coefficient (logP), high degree of protein binding, and a low polar surface area. The drugs found in LDL/VLDL, with the exception of aripiprazole, were also characterized by high or intermediate membrane permeability. CONCLUSIONS The present results indicate that psychotropic drugs with certain characteristics bind to LDL/VLDL in blood. This further implies that lipoproteins could play an important role in drug transport.
Collapse
|
42
|
Chen YX, Wei CX, Lyu YQ, Chen HZ, Jiang G, Gao XL. Biomimetic drug-delivery systems for the management of brain diseases. Biomater Sci 2019; 8:1073-1088. [PMID: 31728485 DOI: 10.1039/c9bm01395d] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acting as a double-edged sword, the blood-brain barrier (BBB) is essential for maintaining brain homeostasis by restricting the entry of small molecules and most macromolecules from blood. However, it also largely limits the brain delivery of most drugs. Even if a drug can penetrate the BBB, its accumulation in the intracerebral pathological regions is relatively low. Thus, an optimal drug-delivery system (DDS) for the management of brain diseases needs to display BBB permeability, lesion-targeting capability, and acceptable safety. Biomimetic DDSs, developed by directly utilizing or mimicking the biological structures and processes, provide promising approaches for overcoming the barriers to brain drug delivery. The present review summarizes the biological properties and biomedical applications of the biomimetic DDSs including the cell membrane-based DDS, lipoprotein-based DDS, exosome-based DDS, virus-based DDS, protein template-based DDS and peptide template-based DDS for the management of brain diseases.
Collapse
Affiliation(s)
- Yao-Xing Chen
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Chen-Xuan Wei
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Ying-Qi Lyu
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China. and Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201210, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Xiao-Ling Gao
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
43
|
Pedersbæk D, Kræmer MK, Kempen PJ, Ashley J, Braesch-Andersen S, Andresen TL, Simonsen JB. The Composition of Reconstituted High-Density Lipoproteins (rHDL) Dictates the Degree of rHDL Cargo- and Size-Remodeling via Direct Interactions with Endogenous Lipoproteins. Bioconjug Chem 2019; 30:2634-2646. [PMID: 31487985 DOI: 10.1021/acs.bioconjchem.9b00552] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The application of reconstituted high-density lipoproteins (rHDL) as a drug-carrier has during the past decade been established as a promising approach for effective receptor-mediated drug delivery, and its ability to target tumors has recently been confirmed in a clinical trial. The rHDL mimics the endogenous HDL, which is known to be highly dynamic and undergo extensive enzyme-mediated remodulations. Hence, to reveal the physiological rHDL stability, a thorough characterization of the dynamics of rHDL in biologically relevant environments is needed. We employ a size-exclusion chromatography (SEC) method to evaluate the dynamics of discoidal rHDL in fetal bovine serum (FBS), where we track both the rHDL lipids (by the fluorescence from lipid-conjugated fluorophores) and apoA-I (by human apoA-I ELISA). We show by using lipoprotein depleted FBS and isolated lipoproteins that rHDL lipids can be transferred to endogenous lipoproteins via direct interactions in a nonenzymatic process, resulting in rHDL compositional- and size-remodeling. This type of dynamics could lead to misinterpretations of fluorescence-based rHDL uptake studies due to desorption of labile lipophilic fluorophores or off-target side effects due to desorption of incorporated drugs. Importantly, we show how the degree of rHDL remodeling can be controlled by the compositional design of the rHDL. Understanding the correlation between the molecular properties of the rHDL constituents and their collective dynamics is essential for improving the rHDL-based drug delivery platform. Taken together, our work highlights the need to carefully consider the compositional design of rHDL and test its stability in a biological relevant environment, when developing rHDL for drug delivery purposes.
Collapse
Affiliation(s)
- Dennis Pedersbæk
- Technical University of Denmark , Department of Health Technology , 2800 Kongens Lyngby , Denmark
| | - Martin Kisha Kræmer
- Technical University of Denmark , Department of Health Technology , 2800 Kongens Lyngby , Denmark
| | - Paul Joseph Kempen
- Technical University of Denmark , Department of Health Technology , 2800 Kongens Lyngby , Denmark
| | - Jon Ashley
- Technical University of Denmark , Department of Health Technology , 2800 Kongens Lyngby , Denmark
| | | | - Thomas L Andresen
- Technical University of Denmark , Department of Health Technology , 2800 Kongens Lyngby , Denmark
| | - Jens B Simonsen
- Technical University of Denmark , Department of Health Technology , 2800 Kongens Lyngby , Denmark
| |
Collapse
|
44
|
Kornmueller K, Vidakovic I, Prassl R. Artificial High Density Lipoprotein Nanoparticles in Cardiovascular Research. Molecules 2019; 24:E2829. [PMID: 31382521 PMCID: PMC6695986 DOI: 10.3390/molecules24152829] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Lipoproteins are endogenous nanoparticles which are the major transporter of fats and cholesterol in the human body. They play a key role in the regulatory mechanisms of cardiovascular events. Lipoproteins can be modified and manipulated to act as drug delivery systems or nanocarriers for contrast agents. In particular, high density lipoproteins (HDL), which are the smallest class of lipoproteins, can be synthetically engineered either as nascent HDL nanodiscs or spherical HDL nanoparticles. Reconstituted HDL (rHDL) particles are formed by self-assembly of various lipids and apolipoprotein AI (apo-AI). A variety of substances including drugs, nucleic acids, signal emitting molecules, or dyes can be loaded, making them efficient nanocarriers for therapeutic applications or medical diagnostics. This review provides an overview about synthesis techniques, physicochemical properties of rHDL nanoparticles, and structural determinants for rHDL function. We discuss recent developments utilizing either apo-AI or apo-AI mimetic peptides for the design of pharmaceutical rHDL formulations. Advantages, limitations, challenges, and prospects for clinical translation are evaluated with a special focus on promising strategies for the treatment and diagnosis of atherosclerosis and cardiovascular diseases.
Collapse
Affiliation(s)
- Karin Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Ivan Vidakovic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Ruth Prassl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria.
| |
Collapse
|
45
|
Yang G, Chen S, Zhang J. Bioinspired and Biomimetic Nanotherapies for the Treatment of Infectious Diseases. Front Pharmacol 2019; 10:751. [PMID: 31333467 PMCID: PMC6624236 DOI: 10.3389/fphar.2019.00751] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
There are still great challenges for the effective treatment of infectious diseases, although considerable achievement has been made by using antiviral and antimicrobial agents varying from small-molecule drugs, peptides/proteins, to nucleic acids. The nanomedicine approach is emerging as a new strategy capable of overcoming disadvantages of molecular therapeutics and amplifying their anti-infective activities, by localized delivery to infection sites, reducing off-target effects, and/or attenuating resistance development. Nanotechnology, in combination with bioinspired and biomimetic approaches, affords additional functions to nanoparticles derived from synthetic materials. Herein, we aim to provide a state-of-the-art review on recent progress in biomimetic and bioengineered nanotherapies for the treatment of infectious disease. Different biomimetic nanoparticles, derived from viruses, bacteria, and mammalian cells, are first described, with respect to their construction and biophysicochemical properties. Then, the applications of diverse biomimetic nanoparticles in anti-infective therapy are introduced, either by their intrinsic activity or by loading and site-specifically delivering various molecular drugs. Bioinspired and biomimetic nanovaccines for prevention and/or therapy of infectious diseases are also highlighted. At the end, major translation issues and future directions of this field are discussed.
Collapse
Affiliation(s)
- Guoyu Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
46
|
Yang J, Liu X, Fu Y, Song Y. Recent advances of microneedles for biomedical applications: drug delivery and beyond. Acta Pharm Sin B 2019; 9:469-483. [PMID: 31193810 PMCID: PMC6543086 DOI: 10.1016/j.apsb.2019.03.007] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/29/2019] [Accepted: 02/16/2019] [Indexed: 12/22/2022] Open
Abstract
The microneedle (MN), a highly efficient and versatile device, has attracted extensive scientific and industrial interests in the past decades due to prominent properties including painless penetration, low cost, excellent therapeutic efficacy, and relative safety. The robust microneedle enabling transdermal delivery has a paramount potential to create advanced functional devices with superior nature for biomedical applications. In this review, a great effort has been made to summarize the advance of microneedles including their materials and latest fabrication method, such as three-dimensional printing (3DP). Importantly, a variety of representative biomedical applications of microneedles such as disease treatment, immunobiological administration, disease diagnosis and cosmetic field, are highlighted in detail. At last, conclusions and future perspectives for development of advanced microneedles in biomedical fields have been discussed systematically. Taken together, as an emerging tool, microneedles have showed profound promise for biomedical applications.
Collapse
|
47
|
Diblock copolymers enhance folding of a mechanosensitive membrane protein during cell-free expression. Proc Natl Acad Sci U S A 2019; 116:4031-4036. [PMID: 30760590 PMCID: PMC6410776 DOI: 10.1073/pnas.1814775116] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Membrane protein folding is a critical step that underlies proper cellular function as well as the design of technologies like vesicle-based biosensors and artificial cells. Membrane composition is known to play a role in membrane protein folding; however, the specific mechanical properties of membranes that govern protein folding remain unclear. Using a highly elastic nonnatural amphiphile, we highlight the importance of a membrane mechanical property, membrane elasticity, on the spontaneous insertion and folding of a model α-helical membrane protein. Through this study, we gain a deeper understanding of cellular membrane protein folding and offer a potential approach to improve the production of membrane proteins through optimizing the mechanical properties of synthetic scaffolds present in cell-free reactions. The expression and integration of membrane proteins into vesicle membranes is a critical step in the design of cell-mimetic biosensors, bioreactors, and artificial cells. While membrane proteins have been integrated into a variety of nonnatural membranes, the effects of the chemical and physical properties of these vesicle membranes on protein behavior remain largely unknown. Nonnatural amphiphiles, such as diblock copolymers, provide an interface that can be synthetically controlled to better investigate this relationship. Here, we focus on the initial step in a membrane protein’s life cycle: expression and folding. We observe improvements in both the folding and overall production of a model mechanosensitive channel protein, the mechanosensitive channel of large conductance, during cell-free reactions when vesicles containing diblock copolymers are present. By systematically tuning the membrane composition of vesicles through incorporation of a poly(ethylene oxide)-b-poly(butadiene) diblock copolymer, we show that membrane protein folding and production can be improved over that observed in traditional lipid vesicles. We then reproduce this effect with an alternate membrane-elasticizing molecule, C12E8. Our results suggest that global membrane physical properties, specifically available membrane surface area and the membrane area expansion modulus, significantly influence the folding and yield of a membrane protein. Furthermore, our results set the stage for explorations into how nonnatural membrane amphiphiles can be used to both study and enhance the production of biological membrane proteins.
Collapse
|
48
|
|