1
|
Zhang Q, Wang J, Chen Z, Qin H, Zhang Q, Tian B, Li X. Transcytosis: an effective mechanism to enhance nanoparticle extravasation and infiltration through biological barriers. Biomed Mater 2025; 20:022003. [PMID: 39788078 DOI: 10.1088/1748-605x/ada85e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/09/2025] [Indexed: 01/12/2025]
Abstract
Nanoparticles (NPs)1have been explored as drugs carriers for treating tumors and central nervous system (CNS)2diseases and for oral administration. However, they lack satisfactory clinical efficacy due to poor extravasation and infiltration through biological barriers to target tissues. Most clinical antitumor NPs have been designed based on enhanced permeability and retention effects which are insufficient and heterogeneous in human tumors. The tight junctions33TJs: tight junctionsof the blood-brain barrier44BBB: blood-brain barrierand the small intestinal epithelium severely impede NPs from being transported into the CNS and blood circulation, respectively. By contrast, transcytosis enables NPs to bypass these physiological barriers and enhances their infiltration into target tissues by active transport. Here, we systematically review the mechanisms and putative application of NP transcytosis for targeting tumor and CNS tissues, explore oral NP administration, and propose future research directions in the field of NP transcytosis.
Collapse
Affiliation(s)
- Qianyi Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Jiamian Wang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200120, People's Republic of China
| | - Zhiyang Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Hao Qin
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Qichen Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Bo Tian
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Xilei Li
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
2
|
Peng Y, Zhang M, Yan J, Wang R, Xin Y, Zheng X, Zhu L, Fei W, Zhao M. Emerging bioengineering breakthroughs in precision diagnosis and therapy for endometriosis and adenomyosis. J Mater Chem B 2025; 13:742-762. [PMID: 39717994 DOI: 10.1039/d4tb01755b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Endometriosis and adenomyosis are debilitating gynecological conditions that severely affect the quality of life of women. Traditional diagnostic and treatment methods, including laparoscopic surgery and hormonal therapy, face significant limitations such as incomplete lesion detection, high recurrence rates, and adverse side effects. Emerging bioengineering technologies offer promising solutions for precise diagnosis and therapy of these diseases. Advances in biomarker detection through electrochemical immunosensors, including specific molecular markers like cytokines and growth factors, have improved their early diagnosis. Innovative imaging techniques, such as near-infrared fluorescence imaging, magnetic resonance imaging, and photoacoustic imaging, enhance lesion visualization and surgical precision. In therapeutic applications, bioengineered drug delivery systems enable targeted therapy by modifying drug carriers with ligands targeting highly expressed receptors in endometriotic lesions. Such strategies could improve drug accumulation at target sites and reduce damage to healthy tissues. Integrating external energy (including lasers, focused ultrasound, and magnetic fields) with nanoplatforms offers key benefits for treating endometriosis and adenomyosis, allowing precise delivery of energy-responsive molecules to lesions and minimizing damage to healthy tissues. Additionally, novel approaches, such as immunotherapy, gene therapy, ferroptosis induction, and synthetic lethal activation, offer new avenues for effective treatment of endometriosis and adenomyosis. Significantly, this paper discusses the advantages of precision diagnosis and treatment of endometriosis in preserving the fertility of women of reproductive age. This review highlights the potential of bioengineering breakthroughs to transform the diagnosis and management of endometriosis and adenomyosis, emphasizing their role in advancing precision medicine and improving women's health.
Collapse
Affiliation(s)
- Yujie Peng
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Jingjing Yan
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Rong Wang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Yu Xin
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoling Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Libo Zhu
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Mengdan Zhao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
3
|
Lim YN, Ryu IS, Jung YJ, Helmlinger G, Kim I, Park HW, Kang H, Lee J, Lee HJ, Lee KS, Jang HN, Ha DI, Park J, Won J, Lim KS, Jeon CY, Cho HJ, Min HS, Ryu JH. l-Type amino acid transporter 1-targeting nanoparticles for antisense oligonucleotide delivery to the CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102340. [PMID: 39411247 PMCID: PMC11474373 DOI: 10.1016/j.omtn.2024.102340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024]
Abstract
l-Type amino acid transporter 1 (LAT1)-specific ligands and polyion complexes are used as brain-specific targets to deliver RNA-based drugs across the blood-brain barrier. We characterized an LAT1-targeting antisense oligonucleotide (ASO)-encapsulated nanoparticle, Phe-NPs/ASO. A 25% density of phenylalanine effectively binds to the surface of LAT1-targeting NPs in the GL261-Luc cells, and Phe-NPs/ASO shows higher binding affinity compared to that without phenylalanine by cellular binding assay. To further characterize the blood-brain barrier-targeting effect and tissue distribution following a single-dose intravenous injection in mice, we performed in vivo biodistribution studies using fluorescence imaging. The Phe-NPs/ASOs were detected in the brain tissue 1 h post-intravenous injection at an approximately 64-fold higher ratio than that of the same ASOs administered in the absence of any NP carrier. The brain tissue delivery of ASO-loaded Phe-NPs was also confirmed in a fluorescence imaging study performed in non-human primates. These results demonstrate that Phe-NPs may successfully deliver an ASO to the brain tissue across brain regions. Phe-NPs loaded with RNA-based drugs have the potential to treat diseases of the CNS, including all forms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Na Lim
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - In Soo Ryu
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Yeon-Joo Jung
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Gabriel Helmlinger
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Insun Kim
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Hye Won Park
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Hansol Kang
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Jina Lee
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Hyo Jin Lee
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Kang Seon Lee
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Ha-Na Jang
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Dae-In Ha
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Hyun-Jeong Cho
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, Daejeon 35365, South Korea
| | - Hyun Su Min
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Jin-Hyeob Ryu
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| |
Collapse
|
4
|
Duan M, Cao R, Yang Y, Chen X, Liu L, Ren B, Wang L, Goh BC. Blood-Brain Barrier Conquest in Glioblastoma Nanomedicine: Strategies, Clinical Advances, and Emerging Challenges. Cancers (Basel) 2024; 16:3300. [PMID: 39409919 PMCID: PMC11475686 DOI: 10.3390/cancers16193300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a prevalent type of malignancy within the central nervous system (CNS) that is associated with a poor prognosis. The standard treatment for GBM includes the surgical resection of the tumor, followed by radiotherapy and chemotherapy; yet, despite these interventions, overall treatment outcomes remain suboptimal. The blood-brain barrier (BBB), which plays a crucial role in maintaining the stability of brain tissue under normal physiological conditions of the CNS, also poses a significant obstacle to the effective delivery of therapeutic agents to GBMs. Recent preclinical studies have demonstrated that nanomedicine delivery systems (NDDSs) offer promising results, demonstrating both effective GBM targeting and safety, thereby presenting a potential solution for targeted drug delivery. In this review, we first explore the various strategies employed in preclinical studies to overcome the BBB for drug delivery. Subsequently, the results of the clinical translation of NDDSs are summarized, highlighting the progress made. Finally, we discuss potential strategies for advancing the development of NDDSs and accelerating their translational research through well-designed clinical trials in GBM therapy.
Collapse
Affiliation(s)
- Mengyun Duan
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Ruina Cao
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China;
| | - Yuan Yang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Xiaoguang Chen
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lian Liu
- Department of Pharmacology, Health Science Center, Yangtze University, Jingzhou 434023, China;
| | - Boxu Ren
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Boon-Cher Goh
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
5
|
Miao K, Xia X, Zou Y, Shi B. Small Scale, Big Impact: Nanotechnology-Enhanced Drug Delivery for Brain Diseases. Mol Pharm 2024; 21:3777-3799. [PMID: 39038108 DOI: 10.1021/acs.molpharmaceut.4c00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Central nervous system (CNS) diseases, ranging from brain cancers to neurodegenerative disorders like dementia and acute conditions such as strokes, have been heavily burdening healthcare and have a direct impact on patient quality of life. A significant hurdle in developing effective treatments is the presence of the blood-brain barrier (BBB), a highly selective barrier that prevents most drugs from reaching the brain. The tight junctions and adherens junctions between the endothelial cells and various receptors expressed on the cells make the BBB form a nonfenestrated and highly selective structure that is crucial for brain homeostasis but complicates drug delivery. Nanotechnology offers a novel pathway to circumvent this barrier, with nanoparticles engineered to ferry drugs across the BBB, protect drugs from degradation, and deliver medications to the designated area. After years of development, nanoparticle optimization, including sizes, shapes, surface modifications, and targeting ligands, can enable nanomaterials tailored to specific brain drug delivery settings. Moreover, smart nano drug delivery systems can respond to endogenous and exogenous stimuli that control subsequent drug release. Here, we address the importance of the BBB in brain disease treatment, summarize different delivery routes for brain drug delivery, discuss the cutting-edge nanotechnology-based strategies for brain drug delivery, and further offer valuable insights into how these innovations in nanoparticle technology could revolutionize the treatment of CNS diseases, presenting a promising avenue for noninvasive, targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kaiting Miao
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xue Xia
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yan Zou
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bingyang Shi
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
6
|
Mythri RB, Aishwarya MRB. Biopolymers as promising vehicles for drug delivery to the brain. Drug Metab Rev 2024; 56:46-61. [PMID: 37955126 DOI: 10.1080/03602532.2023.2281855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
The brain is a privileged organ, tightly guarded by a network of endothelial cells, pericytes, and glial cells called the blood brain barrier. This barrier facilitates tight regulation of the transport of molecules, ions, and cells from the blood to the brain. While this feature ensures protection to the brain, it also presents a challenge for drug delivery for brain diseases. It is, therefore, crucial to identify molecules and/or vehicles that carry drugs, cross the blood brain barrier, and reach targets within the central nervous system. Biopolymers are large polymeric molecules obtained from biological sources. In comparison with synthetic polymers, biopolymers are structurally more complex and their 3D architecture makes them biologically active. Researchers are therefore investigating biopolymers as safe and efficient carriers of brain-targeted therapeutic agents. In this article, we bring together various approaches toward achieving this objective with a note on the prospects for biopolymer-based neurotherapeutic/neurorestorative/neuroprotective interventions. Finally, as a representative paradigm, we discuss the potential use of nanocarrier biopolymers in targeting protein aggregation diseases.
Collapse
Affiliation(s)
- Rajeswara Babu Mythri
- Department of Psychology, Christ (Deemed to be University), Dharmaram College Post, Bengaluru, Karnataka, India
| | | |
Collapse
|
7
|
Farahzadi R, Hejazi MS, Molavi O, Pishgahzadeh E, Montazersaheb S, Jafari S. Clinical Significance of Carnitine in the Treatment of Cancer: From Traffic to the Regulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:9328344. [PMID: 37600065 PMCID: PMC10435298 DOI: 10.1155/2023/9328344] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/12/2022] [Accepted: 03/23/2023] [Indexed: 08/22/2023]
Abstract
Metabolic reprogramming is a common hallmark of cancer cells. Cancer cells exhibit metabolic flexibility to maintain high proliferation and survival rates. In other words, adaptation of cellular demand is essential for tumorigenesis, since a diverse supply of nutrients is required to accommodate tumor growth and progression. Diversity of carbon substrates fueling cancer cells indicate metabolic heterogeneity, even in tumors sharing the same clinical diagnosis. In addition to the alteration of glucose and amino acid metabolism in cancer cells, there is evidence that cancer cells can alter lipid metabolism. Some tumors rely on fatty acid oxidation (FAO) as the primary energy source; hence, cancer cells overexpress the enzymes involved in FAO. Carnitine is an essential cofactor in the lipid metabolic pathways. It is crucial in facilitating the transport of long-chain fatty acids into the mitochondria for β-oxidation. This role and others played by carnitine, especially its antioxidant function in cellular processes, emphasize the fine regulation of carnitine traffic within tissues and subcellular compartments. The biological activity of carnitine is orchestrated by specific membrane transporters that mediate the transfer of carnitine and its derivatives across the cell membrane. The concerted function of carnitine transporters creates a collaborative network that is relevant to metabolic reprogramming in cancer cells. Here, the molecular mechanisms relevant to the role and expression of carnitine transporters are discussed, providing insights into cancer treatment.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Saeid Hejazi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elahe Pishgahzadeh
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Fluorescent Multifunctional Organic Nanoparticles for Drug Delivery and Bioimaging: A Tutorial Review. Pharmaceutics 2022; 14:pharmaceutics14112498. [PMID: 36432688 PMCID: PMC9698844 DOI: 10.3390/pharmaceutics14112498] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Fluorescent organic nanoparticles (FONs) are a large family of nanostructures constituted by organic components that emit light in different spectral regions upon excitation, due to the presence of organic fluorophores. FONs are of great interest for numerous biological and medical applications, due to their high tunability in terms of composition, morphology, surface functionalization, and optical properties. Multifunctional FONs combine several functionalities in a single nanostructure (emission of light, carriers for drug-delivery, functionalization with targeting ligands, etc.), opening the possibility of using the same nanoparticle for diagnosis and therapy. The preparation, characterization, and application of these multifunctional FONs require a multidisciplinary approach. In this review, we present FONs following a tutorial approach, with the aim of providing a general overview of the different aspects of the design, preparation, and characterization of FONs. The review encompasses the most common FONs developed to date, the description of the most important features of fluorophores that determine the optical properties of FONs, an overview of the preparation methods and of the optical characterization techniques, and the description of the theoretical approaches that are currently adopted for modeling FONs. The last part of the review is devoted to a non-exhaustive selection of some recent biomedical applications of FONs.
Collapse
|
9
|
Mitusova K, Peltek OO, Karpov TE, Muslimov AR, Zyuzin MV, Timin AS. Overcoming the blood-brain barrier for the therapy of malignant brain tumor: current status and prospects of drug delivery approaches. J Nanobiotechnology 2022; 20:412. [PMID: 36109754 PMCID: PMC9479308 DOI: 10.1186/s12951-022-01610-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/18/2022] [Indexed: 01/06/2023] Open
Abstract
Besides the broad development of nanotechnological approaches for cancer diagnosis and therapy, currently, there is no significant progress in the treatment of different types of brain tumors. Therapeutic molecules crossing the blood-brain barrier (BBB) and reaching an appropriate targeting ability remain the key challenges. Many invasive and non-invasive methods, and various types of nanocarriers and their hybrids have been widely explored for brain tumor treatment. However, unfortunately, no crucial clinical translations were observed to date. In particular, chemotherapy and surgery remain the main methods for the therapy of brain tumors. Exploring the mechanisms of the BBB penetration in detail and investigating advanced drug delivery platforms are the key factors that could bring us closer to understanding the development of effective therapy against brain tumors. In this review, we discuss the most relevant aspects of the BBB penetration mechanisms, observing both invasive and non-invasive methods of drug delivery. We also review the recent progress in the development of functional drug delivery platforms, from viruses to cell-based vehicles, for brain tumor therapy. The destructive potential of chemotherapeutic drugs delivered to the brain tumor is also considered. This review then summarizes the existing challenges and future prospects in the use of drug delivery platforms for the treatment of brain tumors.
Collapse
Affiliation(s)
- Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Oleksii O Peltek
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Albert R Muslimov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
- Sirius University of Science and Technology, Olympic Ave 1, Sirius, 354340, Russian Federation
| | - Mikhail V Zyuzin
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation.
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation.
| |
Collapse
|
10
|
Lakshmi BA, Kim YJ. Modernistic and Emerging Developments of Nanotechnology in Glioblastoma-Targeted Theranostic Applications. Int J Mol Sci 2022; 23:ijms23031641. [PMID: 35163563 PMCID: PMC8836088 DOI: 10.3390/ijms23031641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Brain tumors such as glioblastoma are typically associated with an unstoppable cell proliferation with aggressive infiltration behavior and a shortened life span. Though treatment options such as chemotherapy and radiotherapy are available in combating glioblastoma, satisfactory therapeutics are still not available due to the high impermeability of the blood–brain barrier. To address these concerns, recently, multifarious theranostics based on nanotechnology have been developed, which can deal with diagnosis and therapy together. The multifunctional nanomaterials find a strategic path against glioblastoma by adjoining novel thermal and magnetic therapy approaches. Their convenient combination of specific features such as real-time tracking, in-depth tissue penetration, drug-loading capacity, and contrasting performance is of great demand in the clinical investigation of glioblastoma. The potential benefits of nanomaterials including specificity, surface tunability, biodegradability, non-toxicity, ligand functionalization, and near-infrared (NIR) and photoacoustic (PA) imaging are sufficient in developing effective theranostics. This review discusses the recent developments in nanotechnology toward the diagnosis, drug delivery, and therapy regarding glioblastoma.
Collapse
|
11
|
Rabha B, Bharadwaj KK, Pati S, Choudhury BK, Sarkar T, Kari ZA, Edinur HA, Baishya D, Atanase LI. Development of Polymer-Based Nanoformulations for Glioblastoma Brain Cancer Therapy and Diagnosis: An Update. Polymers (Basel) 2021; 13:polym13234114. [PMID: 34883617 PMCID: PMC8659151 DOI: 10.3390/polym13234114] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Brain cancers, mainly high-grade gliomas/glioblastoma, are characterized by uncontrolled proliferation and recurrence with an extremely poor prognosis. Despite various conventional treatment strategies, viz., resection, chemotherapy, and radiotherapy, the outcomes are still inefficient against glioblastoma. The blood–brain barrier is one of the major issues that affect the effective delivery of drugs to the brain for glioblastoma therapy. Various studies have been undergone in order to find novel therapeutic strategies for effective glioblastoma treatment. The advent of nanodiagnostics, i.e., imaging combined with therapies termed as nanotheranostics, can improve the therapeutic efficacy by determining the extent of tumour distribution prior to surgery as well as the response to a treatment regimen after surgery. Polymer nanoparticles gain tremendous attention due to their versatile nature for modification that allows precise targeting, diagnosis, and drug delivery to the brain with minimal adverse side effects. This review addresses the advancements of polymer nanoparticles in drug delivery, diagnosis, and therapy against brain cancer. The mechanisms of drug delivery to the brain of these systems and their future directions are also briefly discussed.
Collapse
Affiliation(s)
- Bijuli Rabha
- Department of Bioengineering & Technology, GUIST, Gauhati University, Guwahati 781014, India; (B.R.); (K.K.B.)
| | - Kaushik Kumar Bharadwaj
- Department of Bioengineering & Technology, GUIST, Gauhati University, Guwahati 781014, India; (B.R.); (K.K.B.)
| | - Siddhartha Pati
- Skills Innovation & Academic Network (SIAN) Institute-Association for Biodiversity Conservation and Research (ABC), Balasore 756001, India;
- NatNov Bioscience Private Limited, Balasore, 756001, India
| | | | - Tanmay Sarkar
- Malda Polytechnic, West Bengal State Council of Technical Education, Govt. of West Bengal, Malda 732102, India;
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata 700032, India
| | - Zulhisyam Abdul Kari
- Faculty of Agro Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Malaysia;
| | - Hisham Atan Edinur
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Debabrat Baishya
- Department of Bioengineering & Technology, GUIST, Gauhati University, Guwahati 781014, India; (B.R.); (K.K.B.)
- Correspondence: (D.B.); (L.I.A.)
| | - Leonard Ionut Atanase
- Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania
- Correspondence: (D.B.); (L.I.A.)
| |
Collapse
|
12
|
Han L, Jiang C. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm Sin B 2021; 11:2306-2325. [PMID: 34522589 PMCID: PMC8424230 DOI: 10.1016/j.apsb.2020.11.023] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Blood–brain barrier (BBB) strictly controls matter exchange between blood and brain, and severely limits brain penetration of systemically administered drugs, resulting in ineffective drug therapy of brain diseases. However, during the onset and progression of brain diseases, BBB alterations evolve inevitably. In this review, we focus on nanoscale brain-targeting drug delivery strategies designed based on BBB evolutions and related applications in various brain diseases including Alzheimer's disease, Parkinson's disease, epilepsy, stroke, traumatic brain injury and brain tumor. The advances on optimization of small molecules for BBB crossing and non-systemic administration routes (e.g., intranasal treatment) for BBB bypassing are not included in this review.
Collapse
Key Words
- AD, Alzheimer's disease
- AMT, alpha-methyl-l-tryptophan
- Aβ, amyloid beta
- BACE1, β-secretase 1
- BBB, blood–brain barrier
- BDNF, brain derived neurotrophic factor
- BTB, blood–brain tumor barrier
- Blood–brain barrier
- Brain diseases
- Brain-targeting
- CMT, carrier-mediated transportation
- DTPA-Gd, Gd-diethyltriaminepentaacetic acid
- Drug delivery systems
- EPR, enhanced permeability and retention
- GLUT1, glucose transporter-1
- Gd, gadolinium
- ICAM-1, intercellular adhesion molecule-1
- KATP, ATP-sensitive potassium channels
- KCa, calcium-dependent potassium channels
- LAT1, L-type amino acid transporter 1
- LDL, low density lipoprotein
- LDLR, LDL receptor
- LFA-1, lymphocyte function associated antigen-1
- LRP1, LDLR-related protein 1
- MFSD2A, major facilitator superfamily domain-containing protein 2a
- MMP9, metalloproteinase-9
- MRI, magnetic resonance imaging
- NPs, nanoparticles
- Nanoparticles
- P-gp, P-glycoprotein
- PD, Parkinson's disease
- PEG, polyethyleneglycol
- PEG-PLGA, polyethyleneglycol-poly(lactic-co-glycolic acid)
- PLGA, poly(lactic-co-glycolic acid)
- PSMA, prostate-specific membrane antigen
- RAGE, receptor for advanced glycosylation end products
- RBC, red blood cell
- RMT, receptor-mediated transcytosis
- ROS, reactive oxygen species
- TBI, traumatic brain injury
- TJ, tight junction
- TfR, transferrin receptor
- VEGF, vascular endothelial growth factor
- ZO1, zona occludens 1
- siRNA, short interfering RNA
- tPA, tissue plasminogen activator
Collapse
Affiliation(s)
- Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Corresponding author. Tel./fax: +86 512 65882089.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 200032, China
| |
Collapse
|
13
|
Yang T, Feng J, Zhang Q, Wu W, Mo H, Huang L, Zhang W. l-Carnitine conjugated chitosan-stearic acid polymeric micelles for improving the oral bioavailability of paclitaxel. Drug Deliv 2020; 27:575-584. [PMID: 32306775 PMCID: PMC7191914 DOI: 10.1080/10717544.2020.1748762] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022] Open
Abstract
A delivery system based on l-carnitine (LC) conjugated chitosan (CS)-stearic acid polymeric micelles has been developed for improving the oral bioavailability of paclitaxel (PTX) through targeting intestinal organic cation/carnitine transporter 2 (OCTN2). Stearic acid grafted chitosan (CS-SA), as micelle skeleton material, was synthesized by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC)-mediated coupling reaction. The PTX-loaded micelles were prepared by solvent evaporation-hydration method, and the ligand LC was conjugated onto the micelle surface by anchoring its derivative stearoyl group to the lipophilic core of micelle. The modified polymeric micelles showed regular spherical shapes with small particle size of 157.1 ± 5.2 nm and high drug loading capacity of 15.96 ± 0.20 wt%, and the micelle stability in water was supported by low critical micelle concentration of 14.31 ± 0.21 μg/ml. The drug-loaded micelles presented a slow and incomplete in vitro release, and the pharmacokinetic studies indicated the micelle carriers increased the relative bioavailability of PTX to 165.8% against the commercial formulation. The enhancement effect on intestinal absorption was also confirmed by the intracellular uptake of Caco-2 cells. The proposed micelle carrier system manifested a prospective tool for oral drug delivery.
Collapse
Affiliation(s)
- Tan Yang
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
| | - Jianfang Feng
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
- Department of Pharmacy, Guangxi University of Chinese Medicine, Nanning, PR China
| | - Qian Zhang
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
| | - Wei Wu
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
| | - Hailan Mo
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
| | - Lanzhen Huang
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
| | - Wei Zhang
- Department of Pharmacy, Guilin Medical University, Guilin, PR China
| |
Collapse
|
14
|
Wang X, Qiu Y, Wang M, Zhang C, Zhang T, Zhou H, Zhao W, Zhao W, Xia G, Shao R. Endocytosis and Organelle Targeting of Nanomedicines in Cancer Therapy. Int J Nanomedicine 2020; 15:9447-9467. [PMID: 33268987 PMCID: PMC7701161 DOI: 10.2147/ijn.s274289] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Nanomedicines (NMs) have played an increasing role in cancer therapy as carriers to efficiently deliver therapeutics into tumor cells. For this application, the uptake of NMs by tumor cells is usually a prerequisite to deliver the cargo to intracellular locations, which mainly relies on endocytosis. NMs can enter cells through a variety of endocytosis pathways. Different endocytosis pathways exhibit different intracellular trafficking routes and diverse subcellular localizations. Therefore, a comprehensive understanding of endocytosis mechanisms is necessary for increasing cellular entry efficiency and to trace the fate of NMs after internalization. This review focuses on endocytosis pathways of NMs in tumor cells, mainly including clathrin- and caveolae-mediated endocytosis pathways, involving effector molecules, expression difference of those molecules between normal and tumor cells, as well as the intracellular trafficking route of corresponding endocytosis vesicles. Then, the latest strategies for NMs to actively employ endocytosis are described, including improving tumor cellular uptake of NMs by receptor-mediated endocytosis, transporter-mediated endocytosis and enabling drug activity by changing intracellular routes. Finally, active targeting strategies towards intracellular organelles are also mentioned. This review will be helpful not only in explicating endocytosis and the trafficking process of NMs and elucidating anti-tumor mechanisms inside the cell but also in rendering new ideas for the design of highly efficacious and cancer-targeted NMs.
Collapse
Affiliation(s)
- Xiaowei Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yuhan Qiu
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mengyan Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Conghui Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Tianshu Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Huimin Zhou
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenxia Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Rongguang Shao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
15
|
Kou L, Sun R, Xiao S, Cui X, Sun J, Ganapathy V, Yao Q, Chen R. OCTN2-targeted nanoparticles for oral delivery of paclitaxel: differential impact of the polyethylene glycol linker size on drug delivery in vitro, in situ, and in vivo. Drug Deliv 2020; 27:170-179. [PMID: 31913724 PMCID: PMC6968687 DOI: 10.1080/10717544.2019.1710623] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Targeted nanocarriers have shown great promise in drug delivery because of optimized drug
behavior and improved therapeutic efficacy. How to improve the targeting efficiency of
nanocarriers for the maximum possible drug delivery is a critical issue. Here we developed
L-carnitine-conjugated nanoparticles targeting the carnitine transporter OCTN2 on
enterocytes for improved oral absorption. As a variable, we introduced various lengths of
the polyethylene glycol linker (0, 500, 1000, and 2000) between the nanoparticle surface
and the ligand (CNP, C5NP, C10NP and C20NP) to improve the ligand flexibility, and
consequently for more efficient interaction with the transporter, to enhance the oral
delivery of the cargo load into cells. An increased absorption was observed in cellular
uptake in vitro and in intestinal perfusion assay in
situ when the polyethylene glycol was introduced to link L-carnitine to the
nanoparticles; the highest absorption was achieved with C10NP. In contrast, the linker
decreased the absorption efficiency in vivo. As the presence or absence
of the mucus layer was the primary difference between in vitro/in
situ versus in vivo, the presence of this layer was the likely
reason for this differential effect. In summary, the size of the polyethylene glycol
linker improved the absorption in vitro and in situ, but
interfered with the absorption in vivo. Even though this strategy of
increasing the ligand flexibility with the variable size of the polyethylene glycol failed
to increase oral absorption in vivo, this approach is likely to be useful
for enhanced cellular uptake following intravenous administration of the nanocarriers.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rui Sun
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shuyi Xiao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao Cui
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Jiang X, Zheng YW, Bao S, Zhang H, Chen R, Yao Q, Kou L. Drug discovery and formulation development for acute pancreatitis. Drug Deliv 2020; 27:1562-1580. [PMID: 33118404 PMCID: PMC7598990 DOI: 10.1080/10717544.2020.1840665] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acute pancreatitis is a sudden inflammation and only last for a short time, but might lead to a life-threatening emergency. Traditional drug therapy is an essential supportive method for acute pancreatitis treatment, yet, failed to achieve satisfactory therapeutic outcomes. To date, it is still challenging to develop therapeutic medicine to redress the intricate microenvironment promptly in the inflamed pancreas, and more importantly, avoid multi-organ failure. The understanding of the acute pancreatitis, including the causes, mechanism, and severity judgment, could help the scientists bring up more effective intervention and treatment strategies. New formulation approaches have been investigated to precisely deliver therapeutics to inflammatory lesions in the pancreas, and some even could directly attenuate the pancreatic damages. In this review, we will briefly introduce the involved pathogenesis and underlying mechanisms of acute pancreatitis, as well as the traditional Chinese medicine and the new drug option. Most of all, we will summarize the drug delivery strategies to reduce inflammation and potentially prevent the further development of pancreatitis, with an emphasis on the bifunctional nanoparticles that act as both drug delivery carriers and therapeutics.
Collapse
Affiliation(s)
- Xue Jiang
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ya-Wen Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shihui Bao
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hailin Zhang
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Children's Respiration Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- Municipal Key Laboratory of Paediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Kou L, Yao Q, Zhang H, Chu M, Bhutia YD, Chen R, Ganapathy V. Transporter-Targeted Nano-Sized Vehicles for Enhanced and Site-Specific Drug Delivery. Cancers (Basel) 2020; 12:E2837. [PMID: 33019627 PMCID: PMC7599460 DOI: 10.3390/cancers12102837] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Nano-devices are recognized as increasingly attractive to deliver therapeutics to target cells. The specificity of this approach can be improved by modifying the surface of the delivery vehicles such that they are recognized by the target cells. In the past, cell-surface receptors were exploited for this purpose, but plasma membrane transporters also hold similar potential. Selective transporters are often highly expressed in biological barriers (e.g., intestinal barrier, blood-brain barrier, and blood-retinal barrier) in a site-specific manner, and play a key role in the vectorial transfer of nutrients. Similarly, selective transporters are also overexpressed in the plasma membrane of specific cell types under pathological states to meet the biological needs demanded by such conditions. Nano-drug delivery systems could be strategically modified to make them recognizable by these transporters to enhance the transfer of drugs across the biological barriers or to selectively expose specific cell types to therapeutic drugs. Here, we provide a comprehensive review and detailed evaluation of the recent advances in the field of transporter-targeted nano-drug delivery systems. We specifically focus on areas related to intestinal absorption, transfer across blood-brain barrier, tumor-cell selective targeting, ocular drug delivery, identification of the transporters appropriate for this purpose, and details of the rationale for the approach.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China;
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Department of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang 325035, China
| | - Hailin Zhang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Department of Children’s Respiration Disease, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Maoping Chu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Yangzom D. Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China;
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
| | - Vadivel Ganapathy
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China;
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| |
Collapse
|
18
|
Console L, Scalise M, Mazza T, Pochini L, Galluccio M, Giangregorio N, Tonazzi A, Indiveri C. Carnitine Traffic in Cells. Link With Cancer. Front Cell Dev Biol 2020; 8:583850. [PMID: 33072764 PMCID: PMC7530336 DOI: 10.3389/fcell.2020.583850] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Metabolic flexibility is a peculiar hallmark of cancer cells. A growing number of observations reveal that tumors can utilize a wide range of substrates to sustain cell survival and proliferation. The diversity of carbon sources is indicative of metabolic heterogeneity not only across different types of cancer but also within those sharing a common origin. Apart from the well-assessed alteration in glucose and amino acid metabolisms, there are pieces of evidence that cancer cells display alterations of lipid metabolism as well; indeed, some tumors use fatty acid oxidation (FAO) as the main source of energy and express high levels of FAO enzymes. In this metabolic pathway, the cofactor carnitine is crucial since it serves as a “shuttle-molecule” to allow fatty acid acyl moieties entering the mitochondrial matrix where these molecules are oxidized via the β-oxidation pathway. This role, together with others played by carnitine in cell metabolism, underlies the fine regulation of carnitine traffic among different tissues and, within a cell, among different subcellular compartments. Specific membrane transporters mediate carnitine and carnitine derivatives flux across the cell membranes. Among the SLCs, the plasma membrane transporters OCTN2 (Organic cation transport novel 2 or SLC22A5), CT2 (Carnitine transporter 2 or SLC22A16), MCT9 (Monocarboxylate transporter 9 or SLC16A9) and ATB0, + [Sodium- and chloride-dependent neutral and basic amino acid transporter B(0+) or SLC6A14] together with the mitochondrial membrane transporter CAC (Mitochondrial carnitine/acylcarnitine carrier or SLC25A20) are the most acknowledged to mediate the flux of carnitine. The concerted action of these proteins creates a carnitine network that becomes relevant in the context of cancer metabolic rewiring. Therefore, molecular mechanisms underlying modulation of function and expression of carnitine transporters are dealt with furnishing some perspective for cancer treatment.
Collapse
Affiliation(s)
- Lara Console
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Tiziano Mazza
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Lorena Pochini
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Nicola Giangregorio
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Bari, Italy
| | - Annamaria Tonazzi
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Bari, Italy
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Bari, Italy
| |
Collapse
|
19
|
Kou L, Jiang X, Huang H, Lin X, Zhang Y, Yao Q, Chen R. The role of transporters in cancer redox homeostasis and cross-talk with nanomedicines. Asian J Pharm Sci 2020; 15:145-157. [PMID: 32373196 PMCID: PMC7193452 DOI: 10.1016/j.ajps.2020.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/03/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor cell usually exhibits high levels of reactive oxygen species and adaptive antioxidant system due to the metabolic, genetic, and microenvironment-associated alterations. The altered redox homeostasis can promote tumor progression, development, and treatment resistance. Several membrane transporters are involved in the resetting redox homeostasis and play important roles in tumor progression. Therefore, targeting the involved transporters to disrupt the altered redox balance emerges as a viable strategy for cancer therapy. In addition, nanomedicines have drawn much attention in the past decades. Using nanomedicines to target or reset the redox homeostasis alone or combined with other therapies has brought convincing data in cancer treatment. In this review, we will introduce the altered redox balance in cancer metabolism and involved transporters, and highlight the recent advancements of redox-modulating nanomedicines for cancer treatment.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Jiang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huirong Huang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinlu Lin
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Youting Zhang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Chashan, Wenzhou 325035, China
- Corresponding author. Wenzhou Medical University, University Town, Wenzhou 325035, China. Tel: +86 18958969225
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Corresponding author. Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou 325027, China. Tel: +86 13806890233
| |
Collapse
|
20
|
Darrigues E, Nima ZA, Griffin RJ, Anderson JM, Biris AS, Rodriguez A. 3D cultures for modeling nanomaterial-based photothermal therapy. NANOSCALE HORIZONS 2020; 5:400-430. [PMID: 32118219 DOI: 10.1039/c9nh00628a] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Photothermal therapy (PTT) is one of the most promising techniques for cancer tumor ablation. Nanoparticles are increasingly being investigated for use with PTT and can serve as theranostic agents. Based on the ability of near-infrared nano-photo-absorbers to generate heat under laser irradiation, PTT could prove advantageous in certain situations over more classical cancer therapies. To analyze the efficacy of nanoparticle-based PTT, preclinical in vitro studies typically use 2D cultures, but this method cannot completely mimic the complex tumor organization, bioactivity, and physiology that all control the complex penetration depth, biodistribution, and tissue diffusion parameters of nanomaterials in vivo. To fill this knowledge gap, 3D culture systems have been explored for PTT analysis. These models provide more realistic microenvironments that allow spatiotemporal oxygen gradients and cancer cell adaptations to be considered. This review highlights the work that has been done to advance 3D models for cancer microenvironment modeling, specifically in the context of advanced, functionalized nanoparticle-directed PTT.
Collapse
Affiliation(s)
- Emilie Darrigues
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S University Avenue, Little Rock, AR 72204, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Zhang L, Sui C, Yang W, Luo Q. Amino acid transporters: Emerging roles in drug delivery for tumor-targeting therapy. Asian J Pharm Sci 2020; 15:192-206. [PMID: 32373199 PMCID: PMC7193455 DOI: 10.1016/j.ajps.2019.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/22/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
Amino acid transporters, which play a vital role in transporting amino acids for the biosynthesis of mammalian cells, are highly expressed in types of tumors. Increasing studies have shown the feasibility of amino acid transporters as a component of tumor-targeting therapy. In this review, we focus on tumor-related amino acid transporters and their potential use in tumor-targeting therapy. Firstly, the expression characteristics of amino acid transporters in cancer and their relationship with tumor growth are reviewed. Secondly, the recognition requirements are discussed, focusing on the "acid-base" properties, conformational isomerism and structural analogues. Finally, recent developments in amino acid transporter-targeting drug delivery strategies are highlighted, including prodrugs and nanocarriers, with special attention to the latest findings of molecular mechanisms and targeting efficiency of transporter-mediated endocytosis. We aim to offer related clues that might lead to valuable tumor-targeting strategies by the utilization of amino acid transporters.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Chengguang Sui
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wenhan Yang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| |
Collapse
|
22
|
Tjakra M, Wang Y, Vania V, Hou Z, Durkan C, Wang N, Wang G. Overview of Crosstalk Between Multiple Factor of Transcytosis in Blood Brain Barrier. Front Neurosci 2020; 13:1436. [PMID: 32038141 PMCID: PMC6990130 DOI: 10.3389/fnins.2019.01436] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Blood brain barrier (BBB) conserves unique regulatory system to maintain barrier tightness while allowing adequate transport between neurovascular units. This mechanism possess a challenge for drug delivery, while abnormality may result in pathogenesis. Communication between vascular and neural system is mediated through paracellular and transcellular (transcytosis) pathway. Transcytosis itself showed dependency with various components, focusing on caveolae-mediated. Among several factors, intense communication between endothelial cells, pericytes, and astrocytes is the key for a normal development. Regulatory signaling pathway such as VEGF, Notch, S1P, PDGFβ, Ang/Tie, and TGF-β showed interaction with the transcytosis steps. Recent discoveries showed exploration of various factors which has been proven to interact with one of the process of transcytosis, either endocytosis, endosomal rearrangement, or exocytosis. As well as providing a hypothetical regulatory pathway between each factors, specifically miRNA, mechanical stress, various cytokines, physicochemical, basement membrane and junctions remodeling, and crosstalk between developmental regulatory pathways. Finally, various hypotheses and probable crosstalk between each factors will be expressed, to point out relevant research application (Drug therapy design and BBB-on-a-chip) and unexplored terrain.
Collapse
Affiliation(s)
- Marco Tjakra
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Vicki Vania
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Zhengjun Hou
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
23
|
Shakeri S, Ashrafizadeh M, Zarrabi A, Roghanian R, Afshar EG, Pardakhty A, Mohammadinejad R, Kumar A, Thakur VK. Multifunctional Polymeric Nanoplatforms for Brain Diseases Diagnosis, Therapy and Theranostics. Biomedicines 2020; 8:E13. [PMID: 31941057 PMCID: PMC7168063 DOI: 10.3390/biomedicines8010013] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022] Open
Abstract
The blood-brain barrier (BBB) acts as a barrier to prevent the central nervous system (CNS) from damage by substances that originate from the blood circulation. The BBB limits drug penetration into the brain and is one of the major clinical obstacles to the treatment of CNS diseases. Nanotechnology-based delivery systems have been tested for overcoming this barrier and releasing related drugs into the brain matrix. In this review, nanoparticles (NPs) from simple to developed delivery systems are discussed for the delivery of a drug to the brain. This review particularly focuses on polymeric nanomaterials that have been used for CNS treatment. Polymeric NPs such as polylactide (PLA), poly (D, L-lactide-co-glycolide) (PLGA), poly (ε-caprolactone) (PCL), poly (alkyl cyanoacrylate) (PACA), human serum albumin (HSA), gelatin, and chitosan are discussed in detail.
Collapse
Affiliation(s)
- Shahryar Shakeri
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman 7631818356, Iran;
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey;
| | - Rasoul Roghanian
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan 81746, Iran;
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran;
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran;
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
| | - Vijay Kumar Thakur
- Enhanced Composites and Structures Center, School of Aerospace, Transport and Manufacturing, Cranfield University, Bedfordshire MK43 0AL, UK
| |
Collapse
|
24
|
Israel LL, Galstyan A, Holler E, Ljubimova JY. Magnetic iron oxide nanoparticles for imaging, targeting and treatment of primary and metastatic tumors of the brain. J Control Release 2020; 320:45-62. [PMID: 31923537 DOI: 10.1016/j.jconrel.2020.01.009] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Abstract
Magnetic nanoparticles in general, and iron oxide nanoparticles in particular, have been studied extensively during the past 20 years for numerous biomedical applications. The main applications of these nanoparticles are in magnetic resonance imaging (MRI), magnetic targeting, gene and drug delivery, magnetic hyperthermia for tumor treatment, and manipulation of the immune system by macrophage polarization for cancer treatment. Recently, considerable attention has been paid to magnetic particle imaging (MPI) because of its better sensitivity compared to MRI. In recent years, MRI and MPI have been combined as a dual or multimodal imaging method to enhance the signal in the brain for the early detection and treatment of brain pathologies. Because magnetic and iron oxide nanoparticles are so diverse and can be used in multiple applications such as imaging or therapy, they have attractive features for brain delivery. However, the greatest limitations for the use of MRI/MPI for imaging and treatment are in brain delivery, with one of these limitations being the brain-blood barrier (BBB). This review addresses the current status, chemical compositions, advantages and disadvantages, toxicity and most importantly the future directions for the delivery of iron oxide based substances across the blood-brain barrier for targeting, imaging and therapy of primary and metastatic tumors of the brain.
Collapse
Affiliation(s)
- Liron L Israel
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Anna Galstyan
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Eggehard Holler
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Julia Y Ljubimova
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA.
| |
Collapse
|
25
|
Juraszek B, Nałęcz KA. SLC22A5 (OCTN2) Carnitine Transporter-Indispensable for Cell Metabolism, a Jekyll and Hyde of Human Cancer. Molecules 2019; 25:molecules25010014. [PMID: 31861504 PMCID: PMC6982704 DOI: 10.3390/molecules25010014] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 12/26/2022] Open
Abstract
Oxidation of fatty acids uses l-carnitine to transport acyl moieties to mitochondria in a so-called carnitine shuttle. The process of β-oxidation also takes place in cancer cells. The majority of carnitine comes from the diet and is transported to the cell by ubiquitously expressed organic cation transporter novel family member 2 (OCTN2)/solute carrier family 22 member 5 (SLC22A5). The expression of SLC22A5 is regulated by transcription factors peroxisome proliferator-activated receptors (PPARs) and estrogen receptor. Transporter delivery to the cell surface, as well as transport activity are controlled by OCTN2 interaction with other proteins, such as PDZ-domain containing proteins, protein phosphatase PP2A, caveolin-1, protein kinase C. SLC22A5 expression is altered in many types of cancer, giving an advantage to some of them by supplying carnitine for β-oxidation, thus providing an alternative to glucose source of energy for growth and proliferation. On the other hand, SLC22A5 can also transport several chemotherapeutics used in clinics, leading to cancer cell death.
Collapse
|
26
|
Kou L, Xiao S, Sun R, Bao S, Yao Q, Chen R. Biomaterial-engineered intra-articular drug delivery systems for osteoarthritis therapy. Drug Deliv 2019; 26:870-885. [PMID: 31524006 PMCID: PMC6758706 DOI: 10.1080/10717544.2019.1660434] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is a progressive and degenerative disease, which is no longer confined to the elderly. So far, current treatments are limited to symptom relief, and no valid OA disease-modifying drugs are available. Additionally, OA relative joint is challenging for drug delivery, since the drugs experience rapid clearance in joint, showing a poor bioavailability. Existing therapeutic drugs, like non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, are not conducive for long-term use due to adverse effects. Though supplementations, including chondroitin sulfate and glucosamine, have shown beneficial effects on joint tissues in OA, their therapeutic use is still debatable. New emerging agents, like Kartogenin (KGN) and Interleukin-1 receptor antagonist (IL-1 ra), without a proper formulation, still will not work. Therefore, it is urgent to establish a suitable and efficient drug delivery system for OA therapy. In this review, we pay attention to various types of drug delivery systems and potential therapeutic drugs that may escalate OA treatments.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuyi Xiao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rui Sun
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shihui Bao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
27
|
The solute carrier transporters and the brain: Physiological and pharmacological implications. Asian J Pharm Sci 2019; 15:131-144. [PMID: 32373195 PMCID: PMC7193445 DOI: 10.1016/j.ajps.2019.09.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/17/2019] [Accepted: 09/27/2019] [Indexed: 02/05/2023] Open
Abstract
Solute carriers (SLCs) are the largest family of transmembrane transporters that determine the exchange of various substances, including nutrients, ions, metabolites, and drugs across biological membranes. To date, the presence of about 287 SLC genes have been identified in the brain, among which mutations or the resultant dysfunctions of 71 SLC genes have been reported to be correlated with human brain disorders. Although increasing interest in SLCs have focused on drug development, SLCs are currently still under-explored as drug targets, especially in the brain. We summarize the main substrates and functions of SLCs that are expressed in the brain, with an emphasis on selected SLCs that are important physiologically, pathologically, and pharmacologically in the blood-brain barrier, astrocytes, and neurons. Evidence suggests that a fraction of SLCs are regulated along with the occurrences of brain disorders, among which epilepsy, neurodegenerative diseases, and autism are representative. Given the review of SLCs involved in the onset and procession of brain disorders, we hope these SLCs will be screened as promising drug targets to improve drug delivery to the brain.
Collapse
|
28
|
Emerging transporter-targeted nanoparticulate drug delivery systems. Acta Pharm Sin B 2019; 9:49-58. [PMID: 30766777 PMCID: PMC6361857 DOI: 10.1016/j.apsb.2018.10.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/11/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
Transporter-targeted nanoparticulate drug delivery systems (nano-DDS) have emerged as promising nanoplatforms for efficient drug delivery. Recently, great progress in transporter-targeted strategies has been made, especially with the rapid developments in nanotherapeutics. In this review, we outline the recent advances in transporter-targeted nano-DDS. First, the emerging transporter-targeted nano-DDS developed to facilitate oral drug delivery are reviewed. These include improvements in the oral absorption of protein and peptide drugs, facilitating the intravenous-to-oral switch in cancer chemotherapy. Secondly, the recent advances in transporter-assisted brain-targeting nano-DDS are discussed, focusing on the specific transporter-based targeting strategies. Recent developments in transporter-mediated tumor-targeting drug delivery are also discussed. Finally, the possible transport mechanisms involved in transporter-mediated endocytosis are highlighted, with special attention to the latest findings of the interactions between membrane transporters and nano-DDS.
Collapse
|
29
|
Pochini L, Galluccio M, Scalise M, Console L, Indiveri C. OCTN: A Small Transporter Subfamily with Great Relevance to Human Pathophysiology, Drug Discovery, and Diagnostics. SLAS DISCOVERY 2018; 24:89-110. [PMID: 30523710 DOI: 10.1177/2472555218812821] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OCTN is a small subfamily of membrane transport proteins that belongs to the larger SLC22 family. Two of the three members of the subfamily, namely, OCTN2 and OCTN1, are present in humans. OCTN2 plays a crucial role in the absorption of carnitine from diet and in its distribution to tissues, as demonstrated by the occurrence of severe pathologies caused by malfunctioning or altered expression of this transporter. These findings suggest avoiding a strict vegetarian diet during pregnancy and in childhood. Other roles of OCTN2 are related to the traffic of carnitine derivatives in many tissues. The role of OCTN1 is still unclear, despite the identification of some substrates such as ergothioneine, acetylcholine, and choline. Plausibly, the transporter acts on the control of inflammation and oxidative stress, even though knockout mice do not display phenotypes. A clear role of both transporters has been revealed in drug interaction and delivery. The polyspecificity of the OCTNs is at the base of the interactions with drugs. Interestingly, OCTN2 has been recently exploited in the prodrug approach and in diagnostics. A promising application derives from the localization of OCTN2 in exosomes that represent a noninvasive diagnostic tool.
Collapse
Affiliation(s)
- Lorena Pochini
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lara Console
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy.,2 CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
30
|
Lu Q, Cai X, Zhang X, Li S, Song Y, Du D, Dutta P, Lin Y. Synthetic Polymer Nanoparticles Functionalized with Different Ligands for Receptor-mediated Transcytosis across Blood-Brain Barrier. ACS APPLIED BIO MATERIALS 2018; 1:1687-1694. [PMID: 31815251 DOI: 10.1021/acsabm.8b00502] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Polymeric nanoparticles have been investigated as biocompatible and promising nano-carriers to deliver drugs across the blood-brain barrier (BBB). However, most of the polymeric nanoparticles cannot be observed without attaching them with fluorescent dyes. Generally complex synthesis process is required to attach fluorescent dye tracing molecules with drug carrier nanoparticles. In this paper, we synthesized a novel fluorescent polymer based on poly [Triphenylamine-4-vinyl-(P-methoxy-benzene)] (TEB). This polymer was prepared from TEB polymer through coprecipitation. Furthermore, three types of ligands, transferrin (TfR), lactoferrin (LfR) and lipoprotein (LRP), were covalently attached on the nanoparticle surface to improve the BBB transport efficiency. All of prepared TEB-based nanoparticles were biocompatible, exhibited excellent fluorescence properties and could be observed in vivo. The transcellular transportation of these TEB-based nanoparticles across the BBB was evaluated by observing the fluorescent intensity. The translocation study was performed in an in vitro BBB model that were constructed based on mouse cerebral endothelial cells (bEnd.3). The results showed that ligand-coated TEB nanoparticles can be transported across BBB with high efficiencies (up to 29.02%). This is the first time that the fluorescent TEB nanoparticles were applied as nano-carriers for transport across the BBB. Such fluorescent polymeric nanoparticles have the potential applications in brain imaging or drug delivery.
Collapse
Affiliation(s)
- Qian Lu
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Xiaoli Cai
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Xian Zhang
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Suiqiong Li
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Yang Song
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Dan Du
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Prashanta Dutta
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| | - Yuehe Lin
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99163, United States
| |
Collapse
|
31
|
Kou L, Sun R, Ganapathy V, Yao Q, Chen R. Recent advances in drug delivery via the organic cation/carnitine transporter 2 (OCTN2/SLC22A5). Expert Opin Ther Targets 2018; 22:715-726. [PMID: 30016594 DOI: 10.1080/14728222.2018.1502273] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Transporters in the plasma membrane have been exploited successfully for the delivery of drugs in the form of prodrugs and nanoparticles. Organic cation/carnitine transporter 2 (OCTN2, SLC22A5) has emerged as a viable target for drug delivery. OCTN2 is a Na+-dependent high-affinity transporter for L-carnitine and a Na+-independent transporter for organic cations. OCTN2 is expressed in the blood-brain barrier, heart, liver, kidney, intestinal tract and placenta and plays an essential role in L-carnitine homeostasis in the body. Areas covered: In recent years, several studies have been reported in the literature describing the utility of OCTN2 to enhance the delivery of drugs, prodrugs and nanoparticles. Here we summarize the salient features of OCTN2 in terms of its role in the cellular uptake of its physiological substrate L-carnitine in physiological and pathological context; the structural requirements for recognition and the recent advances in OCTN2-targeted drug delivery systems, including prodrugs and nanoparticles, are discussed. Expert opinion: This transporter has great potential to be utilized as a target for drug delivery to improve oral absorption of drugs in the intestinal tract. It also has potential to facilitate the transfer of drugs across the biological barriers such as the blood-brain barrier, blood-retinal barrier, and maternal-fetal barrier.
Collapse
Affiliation(s)
- Longfa Kou
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Rui Sun
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Vadivel Ganapathy
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China.,b Department of Cell Biology and Biochemistry , School of Medicine, Texas Tech University Health Sciences Center , Lubbock , TX , USA
| | - Qing Yao
- c School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , China
| | - Ruijie Chen
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| |
Collapse
|
32
|
Kou L, Bhutia YD, Yao Q, He Z, Sun J, Ganapathy V. Transporter-Guided Delivery of Nanoparticles to Improve Drug Permeation across Cellular Barriers and Drug Exposure to Selective Cell Types. Front Pharmacol 2018; 9:27. [PMID: 29434548 PMCID: PMC5791163 DOI: 10.3389/fphar.2018.00027] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/10/2018] [Indexed: 12/17/2022] Open
Abstract
Targeted nano-drug delivery systems conjugated with specific ligands to target selective cell-surface receptors or transporters could enhance the efficacy of drug delivery and therapy. Transporters are expressed differentially on the cell-surface of different cell types, and also specific transporters are expressed at higher than normal levels in selective cell types under pathological conditions. They also play a key role in intestinal absorption, delivery via non-oral routes (e.g., pulmonary route and nasal route), and transfer across biological barriers (e.g., blood–brain barrier and blood–retinal barrier. As such, the cell-surface transporters represent ideal targets for nano-drug delivery systems to facilitate drug delivery to selective cell types under normal or pathological conditions and also to avoid off-target adverse side effects of the drugs. There is increasing evidence in recent years supporting the utility of cell-surface transporters in the field of nano-drug delivery to increase oral bioavailability, to improve transfer across the blood–brain barrier, and to enhance delivery of therapeutics in a cell-type selective manner in disease states. Here we provide a comprehensive review of recent advancements in this interesting and important area. We also highlight certain key aspects that need to be taken into account for optimal development of transporter-assisted nano-drug delivery systems.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Yangzom D Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Qing Yao
- Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhonggui He
- Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Jin Sun
- Municipal Key Laboratory of Biopharmaceutics, Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
33
|
Luo Q, Jiang M, Kou L, Zhang L, Li G, Yao Q, Shang L, Chen Y. Ascorbate-conjugated nanoparticles for promoted oral delivery of therapeutic drugs via sodium-dependent vitamin C transporter 1 (SVCT1). ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:198-208. [PMID: 29260899 DOI: 10.1080/21691401.2017.1417864] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Qiuhua Luo
- a Department of Pharmacy , the First Affiliated Hospital of China Medical University , Shenyang , China.,b Department of the First Clinical Pharmacy , China Medical University , Shenyang , China
| | - Mingyan Jiang
- a Department of Pharmacy , the First Affiliated Hospital of China Medical University , Shenyang , China
| | - Longfa Kou
- c Municipal Key Laboratory of Biopharmaceutics, School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , China
| | - Ling Zhang
- d Department of Biotherapy , Cancer Research Institute, the First Affiliated Hospital of China Medical University , Shenyang , China
| | - Guyue Li
- a Department of Pharmacy , the First Affiliated Hospital of China Medical University , Shenyang , China
| | - Qing Yao
- c Municipal Key Laboratory of Biopharmaceutics, School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , China
| | - Lei Shang
- e College of Basic Medical Sciences , Shenyang Medical college , Shenyang , China
| | - Ying Chen
- a Department of Pharmacy , the First Affiliated Hospital of China Medical University , Shenyang , China
| |
Collapse
|