1
|
Babaei M, Abrishami A, Iranpour S, Saljooghi AS, Matin MM. Harnessing curcumin in a multifunctional biodegradable metal-organic framework (bio-MOF) for targeted colorectal cancer theranostics. Drug Deliv Transl Res 2025; 15:1719-1738. [PMID: 39302530 DOI: 10.1007/s13346-024-01707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
Despite significant advancements in managing colorectal cancer (CRC), the issues of efficient diagnosis and targeted therapy remain demanding. To address these challenges and improve treatment outcomes while reducing the cost and side effects, there is a need for more effective theranostic systems that combine diagnostic techniques with therapeutic modalities. This study introduces a pioneering approach for the synthesis of a porous bio-MOF (biodegradable metal-organic framework) using iron as the metal component and curcumin as the pharmaceutical ingredient. Subsequently, the developed drug delivery system was equipped with the anticancer drug doxorubicin (DOX), coated with biocompatible polyethylene glycol (PEG), and targeted with a CRC-specific aptamer (EpCAM). The physicochemical characterization confirmed the successful synthesis of the bio-MOF, demonstrating high encapsulation efficiency and pH-dependent release of DOX. In vitro studies for anticancer activity, cellular uptake, and mechanism of cell death demonstrated that in the case of positive EpCAM HT-29 cells, Apt-PEG-MOF@DOX had enhanced internalization that resulted in massive apoptosis. In vivo studies of the nanoparticles were then conducted in immunocompromised C57BL/6 mice bearing HT-29 tumors. These studies showed that the targeted platform could induce efficient tumor regression with reduced systemic toxicity. The targeted bio-MOF also exhibited MRI imaging properties useful for monitoring tumors. Significantly, the biocompatibility of the introduced bio-MOF was enhanced by pursuing the green synthesis method, which does not engage toxic solvents and strong acids. Overall, this multimodal system acts diversely as a tumor imaging agent and a therapeutic delivery platform suitable for CRC theranostics.
Collapse
Affiliation(s)
- Maryam Babaei
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Abrishami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sonia Iranpour
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
2
|
Driscoll J, Gondaliya P, Zinn DA, Jain R, Yan IK, Dong H, Patel T. Using aptamers for targeted delivery of RNA therapies. Mol Ther 2025; 33:1344-1367. [PMID: 40045577 PMCID: PMC11997499 DOI: 10.1016/j.ymthe.2025.02.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/15/2025] [Accepted: 02/28/2025] [Indexed: 03/21/2025] Open
Abstract
RNA-based treatments that can silence, introduce, or restore gene expression to target human diseases are emerging as a new class of therapeutics. Despite their potential for use in broad applications, their clinical translation has been hampered by a need for delivery to specific cells and tissues. Cell targeting based on the use of aptamers provides an approach for improving their delivery to the desired sites of action. Aptamers are nucleic acid oligonucleotides with structural conformations that provide a robust capacity for the recognition of cell surface molecules and that can be used for directed targeting. Aptamers can be directly conjugated to therapeutic RNA molecules, in the form of aptamer-oligonucleotide chimeras, or incorporated into nanoparticles used as vehicles for the delivery of these therapeutics. Herein, we discuss the use of aptamers for cell-directed RNA therapies, provide an overview of different types of aptamer-targeting RNA therapeutics, and review examples of their therapeutic applications. Challenges associated with manufacturing and scaling up production, and key considerations for their clinical implementation, are also outlined.
Collapse
Affiliation(s)
- Julia Driscoll
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| | - Piyush Gondaliya
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| | - Dylan A Zinn
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| | - Rupesh Jain
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| | - Irene K Yan
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA; Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
3
|
Aulifa DL, Amarilis B, Ichsani LN, Maharani DS, Shabrina AM, Hanifah H, Wulandari RP, Rusdin A, Subra L, Budiman A. A Comprehensive Review: Mesoporous Silica Nanoparticles Greatly Improve Pharmacological Effectiveness of Phytoconstituent in Plant Extracts. Pharmaceuticals (Basel) 2024; 17:1684. [PMID: 39770526 PMCID: PMC11677945 DOI: 10.3390/ph17121684] [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: 10/24/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Medicinal plants are increasingly being explored due to their possible pharmacological properties and minimal adverse effects. However, low bioavailability and stability often limit efficacy, necessitating high oral doses to achieve therapeutic levels in the bloodstream. Mesoporous silica nanoparticles (MSNs) offer a potential solution to these limitations. Due to their large surface area, substantial pore volume, and ability to precisely control pore size. MSNs are also capable of efficiently incorporating a wide range of therapeutic substances, including herbal plant extracts, leading to potential use for drug containment and delivery systems. Therefore, this review aimed to discuss and summarize the successful developments of herbal plant extracts loaded into MSN, focusing on preparation, characterization, and the impact on efficacy. Data were collected from publications on Scopus, PubMed, and Google Scholar databases using the precise keywords "mesoporous silica nanoparticle" and "herbal extract". The results showed that improved phytoconstituent bioavailability, modified release profiles, increased stability, reduced dose and toxicity are the primary benefits of this method. This review offers insights on the significance of integrating MSNs into therapeutic formulations to improve pharmacological characteristics and effectiveness of medicinal plant extracts. Future prospects show favorable potential for therapeutic applications using MSNs combined with herbal medicines for clinical therapy.
Collapse
Affiliation(s)
- Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (R.P.W.); (A.R.)
| | - Bunga Amarilis
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Luthfia Nur Ichsani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Devita Salsa Maharani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Ayunda Myela Shabrina
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Hanifah Hanifah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Rizky Prasiska Wulandari
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (R.P.W.); (A.R.)
| | - Agus Rusdin
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (R.P.W.); (A.R.)
| | - Laila Subra
- Department of Pharmacy, Faculty of Bioeconomic, Food and Health Sciences, Universiti Geomatika Malaysia, Kuala Lumpur 54200, Malaysia;
| | - Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| |
Collapse
|
4
|
Chen G, Huang Y, Yu H, Wang J, Li H, Shen S, Zhou X, Shi K, Sun H. Nanoparticles Fueled by Enzyme for the Treatment of Hyperlipidemic Acute Pancreatitis. ACS Biomater Sci Eng 2024; 10:7176-7190. [PMID: 39412351 PMCID: PMC11559557 DOI: 10.1021/acsbiomaterials.4c00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 11/12/2024]
Abstract
Hyperlipidemic acute pancreatitis (HAP) is a serious inflammatory pancreatic disease commonly seen in patients with disorders of lipid metabolism. Decreasing blood triglyceride levels and proinflammatory factors can alleviate hyperlipidemic pancreatitis. The lipase that enhanced the Brownian motion of mesoporous silica in triglyceride solutions could accelerate decomposition of the lipid and improve the efficiency of absorption. In this study, we developed a mesoporous silica nanoparticle with dual modification of IL-6 aptamer and lipase for the treatment of HAP. The nanoparticle could increase the ability of particles to absorb inflammatory factor IL-6 and decompose triglycerides. For every 10 mg of the dual-modified nanoparticles, the efficiency of capturing IL-6 was approximately 9.67 pg/mL and of decomposing triglycerides was approximately 3.88 mg/mL in the plasma of HAP patients within 2 h. In summary, the mesoporous silica nanoparticle could absorb the IL-6 inflammatory factor through IL-6 aptamers and decompose triglycerides through lipase. Furthermore, based on clinically available plasma exchange technology, combined with our developed dual-modified nanoparticles, we designed an absorption device for the treatment of hyperlipidemic pancreatitis; it works to promote the treatment of hyperlipidemic pancreatitis.
Collapse
Affiliation(s)
- Geer Chen
- Department
of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, PR China
| | - Yunfeng Huang
- Translational
Medicine Laboratory, The First Affiliated
Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Haohui Yu
- Department
of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, PR China
| | - Junru Wang
- Department
of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, PR China
| | - Haobing Li
- Translational
Medicine Laboratory, The First Affiliated
Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Shuqi Shen
- Translational
Medicine Laboratory, The First Affiliated
Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xingjian Zhou
- Translational
Medicine Laboratory, The First Affiliated
Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Keqing Shi
- Translational
Medicine Laboratory, The First Affiliated
Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
- Cixi
Biomedical Research Institute, Wenzhou Medical
University, Wenzhou 325035, Zhejiang, China
| | - Hongwei Sun
- Department
of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, PR China
| |
Collapse
|
5
|
Chang Y, Zheng W, Duan M, Su T, Wang Z, Wu S, Duan N. Construction of Aptamer-Functionalized DNA Hydrogels for Effective Inhibition of Shiga Toxin II Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23533-23543. [PMID: 39388632 DOI: 10.1021/acs.jafc.4c07612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Bacterial infections have been seriously endangering public health and life, making it imperative to explore novel anti-infection strategies for their control. Herein, we constructed a DNA hydrogel encoded with aptamers (Apt-hydrogel) to inhibit Shiga toxin II (Stx2) toxicity, thereby alleviating Escherichia coli (EHEC) infection. The Apt-hydrogel was formed by two Y-shaped DNA scaffolds through rational design, where one end of Y was encoded with an aptamer sequence targeting the B subunit of Shiga toxin II (Stx2B). The Apt-hydrogel not only retained the high affinity of the aptamer but also provided protection for the aptamer, endowing it with better stability and biocompatibility. The results from in vitro and in vivo demonstrated good mediation effects of the Apt-hydrogel on Stx2 toxicity and confirmed its excellent inhibition activity. We hypothesized that the mechanism could be attributed to the high affinity of Apt-hydrogel for Stx2B, which effectively occupies the active site of Stx2B and its receptor Gb3. This interaction enhanced steric hindrance, thereby mediating their interaction and preventing Stx2 from entering the cell to exert toxicity. We anticipate that the novel Apt-hydrogel will expand the usage of aptamers and provide a new dimension for the Apt-hydrogel as a promising blocking assistant to inhibit Shiga toxin infections via a strong steric hindrance effect.
Collapse
Affiliation(s)
- Yuting Chang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wenxiu Zheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Mengxia Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Tingting Su
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhouping Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Shijia Wu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Nuo Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
6
|
Chavan DD, Bhosale RR, Thorat VM, Shete AS, Patil SJ, Tiwari DD. Recent Advances in the Development and Utilization of Nanoparticles for the Management of Malignant Solid Tumors. Cureus 2024; 16:e70312. [PMID: 39469411 PMCID: PMC11513206 DOI: 10.7759/cureus.70312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
The purpose of nanotechnology-based drug delivery systems or novel drug delivery systems is to improve the effectiveness of therapy, and their promising properties have led to their increasing significance in the management of cancer. The researchers have primarily focused on designing novel nanocarriers, like nanoparticles (NPs), that can effectively deliver drugs to target cells and respond specifically to conditions particular to cancer. Whether passive or active targeting, these nanocarriers can deliver therapeutic cargoes to the tumor site to release the drug from the drug delivery systems. The purpose of this study is to provide recent scientific literature and key findings to researchers as well as the scientific community from the medical and pharmaceutical domains by reporting current advancements in the development of NPs for the treatment of different malignant solid tumors, such as colorectal, pancreatic, prostate, and cervical cancer.
Collapse
Affiliation(s)
- Dhanashri D Chavan
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Malkapur, IND
| | - Rohit R Bhosale
- Department of Pharmaceutics, Krishna Foundation's Jaywant Institute of Pharmacy, Wathar, IND
| | - Vandana M Thorat
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Malkapur, IND
| | - Amol S Shete
- Department of Pharmaceutics, Krishna Institute of Pharmacy, Krishna Vishwa Vidyapeeth (Deemed to be University), Malkapur, IND
| | - Sarika J Patil
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Malkapur, IND
| | - Devkumar D Tiwari
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Malkapur, IND
| |
Collapse
|
7
|
Lian S, Wang Q, Liu Y, Lu Y, Huang L, Deng H, Xie X. Multi-targeted nanoarrays for early broad-spectrum detection of lung cancer based on blood biopsy of tumor exosomes. Talanta 2024; 276:126270. [PMID: 38761662 DOI: 10.1016/j.talanta.2024.126270] [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: 03/21/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Liquid biopsies utilizing tumor exosomes offer a noninvasive approach for cancer diagnosis. However, validation studies consistently report that in the early stages of cancer, the secretion of exosomes by cancer cells is relatively low, while bodily fluids exhibit a high abundance of other interfering biomolecules. Additionally, target mutations or differences in biomarker expression among various lung cancer subtypes may contribute to detection failures. In this study, we propose a targeted nanoarray-based early cancer diagnostic approach for multiple subtypes of lung cancer. The targeted nanoarray was constructed by modifying five targeting aptamers onto mesoporous silica nanoparticles through the conjugation between amino and carboxyl groups. The flow cytometry experiments demonstrated the specific recognition ability of the targeted nanoarray to tumor exosomes in PBS, even at biomarker expression levels as low as 1.5 %. Moreover, the TEM results indicated that the targeted nanoarray could isolate tumor exosomes in the blood of tumor-bearing mice. Furthermore, the targeted nanoarray could detect tumor exosomes in the blood of various lung cancer bearing mice, including at the early stages of cancer, which has just been established for 7 days. Overall, the targeted nanoarray represents a promising tool for the early detection of various subtypes of lung cancer.
Collapse
Affiliation(s)
- Shu Lian
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Qixuan Wang
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China; College of Chemical Engineering, Fuzhou University, 350116, Fuzhou, China
| | - Yuxin Liu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Yusheng Lu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Lu Huang
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China; College of Chemical Engineering, Fuzhou University, 350116, Fuzhou, China.
| | - Haohua Deng
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China.
| | - Xiaodong Xie
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China; College of Chemical Engineering, Fuzhou University, 350116, Fuzhou, China.
| |
Collapse
|
8
|
Meng J, Wang ZG, Zhao X, Wang Y, Chen DY, Liu DL, Ji CC, Wang TF, Zhang LM, Bai HX, Li BY, Liu Y, Wang L, Yu WG, Yin ZT. Silica nanoparticle design for colorectal cancer treatment: Recent progress and clinical potential. World J Clin Oncol 2024; 15:667-673. [PMID: 38946830 PMCID: PMC11212613 DOI: 10.5306/wjco.v15.i6.667] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 06/24/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and the second most common cause of cancer death. Nanotherapies are able to selectively target the delivery of cancer therapeutics, thus improving overall antitumor efficiency and reducing conventional chemotherapy side effects. Mesoporous silica nanoparticles (MSNs) have attracted the attention of many researchers due to their remarkable advantages and biosafety. We offer insights into the recent advances of MSNs in CRC treatment and their potential clinical application value.
Collapse
Affiliation(s)
- Jin Meng
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Zhi-Gang Wang
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Xiu Zhao
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Ying Wang
- Acupuncture and Tuina College, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
| | - De-Yu Chen
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - De-Long Liu
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Cheng-Chun Ji
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Tian-Fu Wang
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Li-Mei Zhang
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian 116001, Liaoning Province, China
| | - Hai-Xia Bai
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Bo-Yang Li
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Yuan Liu
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Lei Wang
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Wei-Gang Yu
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| | - Zhi-Tao Yin
- Department of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang 110000, Liaoning Province, China
| |
Collapse
|
9
|
Mili M, Bachu V, Kuri PR, Singh NK, Goswami P. Improving synthesis and binding affinities of nucleic acid aptamers and their therapeutics and diagnostic applications. Biophys Chem 2024; 309:107218. [PMID: 38547671 DOI: 10.1016/j.bpc.2024.107218] [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: 11/21/2023] [Revised: 02/21/2024] [Accepted: 03/17/2024] [Indexed: 04/22/2024]
Abstract
Nucleic acid aptamers have captivated the attention of analytical and medicinal scientists globally due to their several advantages as recognition molecules over conventional antibodies because of their small size, simple and inexpensive synthesis, broad target range, and high stability in varied environmental conditions. These recognition molecules can be chemically modified to make them resistant to nuclease action in blood serum, reduce rapid renel clearance, improve the target affinity and selectivity, and make them amenable to chemically conjugate with a support system that facilitates their selective applications. This review focuses on the development of efficient aptamer candidates and their application in clinical diagnosis and therapeutic applications. Significant advances have been made in aptamer-based diagnosis of infectious and non-infectious diseases. Collaterally, the progress made in therapeutic applications of aptamers is encouraging, as evident from their use in diagnosing cancer, neurodegenerative diseases, microbial infection, and in imaging. This review also updates the progress on clinical trials of many aptamer-based products of commercial interests. The key development and critical issues on the subject have been summarized in the concluding remarks.
Collapse
Affiliation(s)
- Malaya Mili
- Department of Biosciences and Bioengineering, IIT Guwahati, 781039, Assam, India
| | - Vinay Bachu
- Department of Biosciences and Bioengineering, IIT Guwahati, 781039, Assam, India
| | - Pooja Rani Kuri
- Department of Biosciences and Bioengineering, IIT Guwahati, 781039, Assam, India
| | | | - Pranab Goswami
- Department of Biosciences and Bioengineering, IIT Guwahati, 781039, Assam, India.
| |
Collapse
|
10
|
Moghimipour E, Handali S. Functionalized liposomes as a potential drug delivery systems for colon cancer treatment: A systematic review. Int J Biol Macromol 2024; 269:132023. [PMID: 38697444 DOI: 10.1016/j.ijbiomac.2024.132023] [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: 07/14/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Colon cancer is one of the lethal diseases in the world with approximately 700,000 fatalities annually. Nowadays, due to the side effects of existing methods in the treatment of colon cancer such as radiotherapy and chemotherapy, the use of targeted nanocarriers in cancer treatment has received wide attention, and among them, especially liposomes have been studied a lot. Based on this, anti-tumor drugs hidden in targeted active liposomes can selectively act on cancer cells. In this systematic review, the use of various ligands such as folic acid, transferrin, aptamer, hyaluronic acid and cRGD for active targeting of liposomes to achieve improved drug delivery to colon cancer cells has been reviewed. The original articles published in English in the databases of Science Direct, PubMed and Google scholar from 2012 to 2022 were reviewed. From the total of 26,256 published articles, 19 studies met the inclusion criteria. The results of in vitro and in vivo studies have revealed that targeted liposomes lead to increasing the efficacy of anti-cancer agents on colon cancer cells with reducing side effects compared to free drugs and non-targeted liposomes. To the best of our knowledge, this is the first systematic review showing promising results for improvement treatment of colon cancer using targeted liposomes.
Collapse
Affiliation(s)
- Eskandar Moghimipour
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Abrishami A, Bahrami AR, Nekooei S, Sh Saljooghi A, Matin MM. Hybridized quantum dot, silica, and gold nanoparticles for targeted chemo-radiotherapy in colorectal cancer theranostics. Commun Biol 2024; 7:393. [PMID: 38561432 PMCID: PMC10984983 DOI: 10.1038/s42003-024-06043-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Multimodal nanoparticles, utilizing quantum dots (QDs), mesoporous silica nanoparticles (MSNs), and gold nanoparticles (Au NPs), offer substantial potential as a smart and targeted drug delivery system for simultaneous cancer therapy and imaging. This method entails coating magnetic GZCIS/ZnS QDs with mesoporous silica, loading epirubicin into the pores, capping with Au NPs, PEGylation, and conjugating with epithelial cell adhesion molecule (EpCAM) aptamers to actively target colorectal cancer (CRC) cells. This study showcases the hybrid QD@MSN-EPI-Au-PEG-Apt nanocarriers (size ~65 nm) with comprehensive characterizations post-synthesis. In vitro studies demonstrate the selective cytotoxicity of these targeted nanocarriers towards HT-29 cells compared to CHO cells, leading to a significant reduction in HT-29 cell survival when combined with irradiation. Targeted delivery of nanocarriers in vivo is validated by enhanced anti-tumor effects with reduced side effects following chemo-radiotherapy, along with imaging in a CRC mouse model. This approach holds promise for improved CRC theranostics.
Collapse
Affiliation(s)
- Amir Abrishami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
12
|
Mahmoudian F, Ahmari A, Shabani S, Sadeghi B, Fahimirad S, Fattahi F. Aptamers as an approach to targeted cancer therapy. Cancer Cell Int 2024; 24:108. [PMID: 38493153 PMCID: PMC10943855 DOI: 10.1186/s12935-024-03295-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Conventional cancer treatments can cause serious side effects because they are not specific to cancer cells and can damage healthy cells. Aptamers often are single-stranded oligonucleotides arranged in a unique architecture, allowing them to bind specifically to target sites. This feature makes them an ideal choice for targeted therapeutics. They are typically produced through the systematic evolution of ligands by exponential enrichment (SELEX) and undergo extensive pharmacological revision to modify their affinity, specificity, and therapeutic half-life. Aptamers can act as drugs themselves, directly inhibiting tumor cells. Alternatively, they can be used in targeted drug delivery systems to transport drugs directly to tumor cells, minimizing toxicity to healthy cells. In this review, we will discuss the latest and most advanced approaches to using aptamers for cancer treatment, particularly targeted therapy overcoming resistance to conventional therapies.
Collapse
Affiliation(s)
- Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Azin Ahmari
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Radiation Oncology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shiva Shabani
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Infectious Diseases, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Bahman Sadeghi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shohreh Fahimirad
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran.
| | - Fahimeh Fattahi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Weisany W, Yousefi S, Soufiani SP, Pashang D, McClements DJ, Ghasemlou M. Mesoporous silica nanoparticles: A versatile platform for encapsulation and delivery of essential oils for food applications. Adv Colloid Interface Sci 2024; 325:103116. [PMID: 38430728 DOI: 10.1016/j.cis.2024.103116] [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: 11/21/2023] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 03/05/2024]
Abstract
Essential oils (EOs) are biologically active and volatile substances that have found widespread applications in the food, cosmetics, and pharmaceutical industries. However, there are some challenges to their commercial utilization due to their high volatility, susceptibility to degradation, and hydrophobicity. In their free form, EOs can quickly evaporate, as well as undergo degradation reactions like oxidation, isomerization, dehydrogenation, or polymerization when exposed to light, heat, or air. Encapsulating EOs within mesoporous silica nanoparticles (MSNPs) could overcome these limitations and thereby broaden their usage. MSNPs may endow protection and slow-release properties to EOs, thereby extending their stability, enhancing their efficacy, and improving their dispersion in aqueous environments. This review explores and compares the design and development of different MSNP-based nanoplatforms to encapsulate, protect, and release EOs. Initially, a brief overview of the various types of available MSNPs, their properties, and their synthesis methods is given to better understand their roles as carriers for EOs. Several encapsulation technologies are then examined, including solvent-based and solvent-free methods. The suitability of each technology for EO encapsulation, as well as its impact on their stability and release, is discussed in detail. Opportunities and challenges for using EO-loaded MSNPs as preservatives, flavor enhancers, and antimicrobial agents in the food industry are then highlighted. Overall, this review aims to bridge a knowledge gap by providing a thorough understanding of EO encapsulation within MSNPs, which should facilitate the application of this technology in the food industry.
Collapse
Affiliation(s)
- Weria Weisany
- Department of Agriculture and Food Science, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Shima Yousefi
- Department of Agriculture and Food Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Solmaz Pourbarghi Soufiani
- Department of Agriculture and Food Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Danial Pashang
- Department of Agriculture and Food Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - David Julian McClements
- Biopolymers & Colloids Research Laboratory, Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Mehran Ghasemlou
- School of Science, STEM College, RMIT University, Melbourne, VIC 3083, Australia; Centre for Sustainable Bioproducts, Deakin University, Waurn Ponds, VIC 3216, Australia.
| |
Collapse
|
14
|
Qi Q, Shen Q, Geng J, An W, Wu Q, Wang N, Zhang Y, Li X, Wang W, Yu C, Li L. Stimuli-responsive biodegradable silica nanoparticles: From native structure designs to biological applications. Adv Colloid Interface Sci 2024; 324:103087. [PMID: 38278083 DOI: 10.1016/j.cis.2024.103087] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/24/2023] [Accepted: 01/05/2024] [Indexed: 01/28/2024]
Abstract
Due to their inherent advantages, silica nanoparticles (SiNPs) have greatly potential applications as bioactive materials in biosensors/biomedicine. However, the long-term and nonspecific accumulation in healthy tissues may give rise to toxicity, thereby impeding their widespread clinical application. Hence, it is imperative and noteworthy to develop biodegradable and clearable SiNPs for biomedical purposes. Recently, the design of multi-stimuli responsive SiNPs to improve degradation efficiency under specific pathological conditions has increased their clinical trial potential as theranostic nanoplatform. This review comprehensively summaries the rational design and recent progress of biodegradable SiNPs under various internal and external stimuli for rapid in vivo degradation and clearance. In addition, the factors that affect the biodegradation of SiNPs are also discussed. We believe that this systematic review will offer profound stimulus and timely guide for further research in the field of SiNP-based nanosensors/nanomedicine.
Collapse
Affiliation(s)
- Qianhui Qi
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Qian Shen
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211800, China
| | - Jiaying Geng
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211800, China
| | - Weizhen An
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211800, China
| | - Qiong Wu
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211800, China
| | - Nan Wang
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yu Zhang
- Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xue Li
- Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Wei Wang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE) and School of Flexible Electronics (Future Technologies), Nanjing Tech University, Nanjing 211800, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, China.
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| |
Collapse
|
15
|
Park D, Lee SJ, Park JW. Aptamer-Based Smart Targeting and Spatial Trigger-Response Drug-Delivery Systems for Anticancer Therapy. Biomedicines 2024; 12:187. [PMID: 38255292 PMCID: PMC10813750 DOI: 10.3390/biomedicines12010187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
In recent years, the field of drug delivery has witnessed remarkable progress, driven by the quest for more effective and precise therapeutic interventions. Among the myriad strategies employed, the integration of aptamers as targeting moieties and stimuli-responsive systems has emerged as a promising avenue, particularly in the context of anticancer therapy. This review explores cutting-edge advancements in targeted drug-delivery systems, focusing on the integration of aptamers and stimuli-responsive platforms for enhanced spatial anticancer therapy. In the aptamer-based drug-delivery systems, we delve into the versatile applications of aptamers, examining their conjugation with gold, silica, and carbon materials. The synergistic interplay between aptamers and these materials is discussed, emphasizing their potential in achieving precise and targeted drug delivery. Additionally, we explore stimuli-responsive drug-delivery systems with an emphasis on spatial anticancer therapy. Tumor microenvironment-responsive nanoparticles are elucidated, and their capacity to exploit the dynamic conditions within cancerous tissues for controlled drug release is detailed. External stimuli-responsive strategies, including ultrasound-mediated, photo-responsive, and magnetic-guided drug-delivery systems, are examined for their role in achieving synergistic anticancer effects. This review integrates diverse approaches in the quest for precision medicine, showcasing the potential of aptamers and stimuli-responsive systems to revolutionize drug-delivery strategies for enhanced anticancer therapy.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|
16
|
Zhang J, Shang J, Tang X, Xu X. TfR Aptamer-Functionalized MSNs for Enhancing Targeted Cellular Uptake and Therapy of Cancer Cells. ACS OMEGA 2023; 8:48975-48983. [PMID: 38162791 PMCID: PMC10753727 DOI: 10.1021/acsomega.3c06562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024]
Abstract
Mesoporous silica nanoparticles (MSNs), as novel nanocarriers for drug delivery in cancer treatment, have attracted widespread concern because of their rich pore structure, large pore capacity, ease of modification, and biocompatibility. However, the limitation of nontargeting and low uptake efficiency hindered their further application. Considering the overexpression of the transferrin receptor (TfR) on most cancer cell membranes, herein, we propose a strategy to effectively enhance the cellular internalization of MSNs by arming them with the TfR aptamer. Cellular fluorescent imaging and flow cytometry analysis demonstrated that TfR aptamer-functionalized MSNs exhibited superior cellular internalization compared to unmodified or random sequence-modified MSNs toward three different cancer cell lines, including MCF-7, HeLa, and A549. Furthermore, TfR aptamer-functionalized MSNs displayed enhanced drug delivery efficiency compared with MSNs at equivalent doses and incubation times. These results suggested that TfR aptamer-functionalized MSNs have the potential for enhanced delivery of therapeutic agents into TfR-positive cancer cells to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Jiajia Zhang
- School
of Nursing and Health Management, Wuhan
Donghu University, Wuhan 430212, China
- Key
Laboratory for Green Chemical Process of Ministry of Education, Hubei
Key Lab of Novel Reaction & Green Chemical Technology, School
of Chemical Engineering and Pharmacy, Wuhan
Institute of Technology, Wuhan 430205, China
| | - Jing Shang
- Key
Laboratory for Green Chemical Process of Ministry of Education, Hubei
Key Lab of Novel Reaction & Green Chemical Technology, School
of Chemical Engineering and Pharmacy, Wuhan
Institute of Technology, Wuhan 430205, China
| | - Xiuhui Tang
- School
of Electrical and Information Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Xuemei Xu
- Key
Laboratory for Green Chemical Process of Ministry of Education, Hubei
Key Lab of Novel Reaction & Green Chemical Technology, School
of Chemical Engineering and Pharmacy, Wuhan
Institute of Technology, Wuhan 430205, China
| |
Collapse
|
17
|
Li S, Coffinier Y, Lagadec C, Cleri F, Nishiguchi K, Fujiwara A, Kim SH, Clément N. Single-Cell Electrochemical Aptasensor Array. ACS Sens 2023; 8:2921-2926. [PMID: 37431846 DOI: 10.1021/acssensors.3c00570] [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: 07/12/2023]
Abstract
Despite several demonstrations of electrochemical devices with limits of detection (LOD) of 1 cell/mL, the implementation of single-cell bioelectrochemical sensor arrays has remained elusive due to the challenges of scaling up. In this study, we show that the recently introduced nanopillar array technology combined with redox-labeled aptamers targeting epithelial cell adhesion molecule (EpCAM) is perfectly suited for such implementation. Combining nanopillar arrays with microwells determined for single cell trapping directly on the sensor surface, single target cells are successfully detected and analyzed. This first implementation of a single-cell electrochemical aptasensor array, based on Brownian-fluctuating redox species, opens new opportunities for large-scale implementation and statistical analysis of early cancer diagnosis and cancer therapy in clinical settings.
Collapse
Affiliation(s)
- Shuo Li
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo 4-6-1 Komaba, Meguro-ku 153-8505, Tokyo, Japan
| | - Yannick Coffinier
- IEMN, CNRS UMR8520, Univ. Lille Avenue Poincare, BP 60069, Villeneuve d'Ascq cedex 59652, France
| | - Chann Lagadec
- Univ. Lille, CNRS, Inserm, CHU Lille, Centre Oscar Lambret, UMR9020 - UMR-S 1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille F-59000, France
| | - Fabrizio Cleri
- IEMN, CNRS UMR8520, Univ. Lille Avenue Poincare, BP 60069, Villeneuve d'Ascq cedex 59652, France
| | - Katsuhiko Nishiguchi
- NTT Basic Research Laboratories, NTT Corporation, 3-1, Morinosato-Wakamiya, Atsugi-shi 243-0198, Japan
| | - Akira Fujiwara
- NTT Basic Research Laboratories, NTT Corporation, 3-1, Morinosato-Wakamiya, Atsugi-shi 243-0198, Japan
| | - Soo Hyeon Kim
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo 4-6-1 Komaba, Meguro-ku 153-8505, Tokyo, Japan
| | - Nicolas Clément
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo 4-6-1 Komaba, Meguro-ku 153-8505, Tokyo, Japan
| |
Collapse
|
18
|
Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther 2023; 8:293. [PMID: 37544972 PMCID: PMC10404590 DOI: 10.1038/s41392-023-01536-y] [Citation(s) in RCA: 213] [Impact Index Per Article: 106.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/08/2023] Open
Abstract
Cancer remains a highly lethal disease in the world. Currently, either conventional cancer therapies or modern immunotherapies are non-tumor-targeted therapeutic approaches that cannot accurately distinguish malignant cells from healthy ones, giving rise to multiple undesired side effects. Recent advances in nanotechnology, accompanied by our growing understanding of cancer biology and nano-bio interactions, have led to the development of a series of nanocarriers, which aim to improve the therapeutic efficacy while reducing off-target toxicity of the encapsulated anticancer agents through tumor tissue-, cell-, or organelle-specific targeting. However, the vast majority of nanocarriers do not possess hierarchical targeting capability, and their therapeutic indices are often compromised by either poor tumor accumulation, inefficient cellular internalization, or inaccurate subcellular localization. This Review outlines current and prospective strategies in the design of tumor tissue-, cell-, and organelle-targeted cancer nanomedicines, and highlights the latest progress in hierarchical targeting technologies that can dynamically integrate these three different stages of static tumor targeting to maximize therapeutic outcomes. Finally, we briefly discuss the current challenges and future opportunities for the clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Dahua Fan
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China.
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yajun Wang
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China
| | | | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
19
|
Kianpour M, Huang CW, Vejvisithsakul PP, Wang JY, Li CF, Shiao MS, Pan CT, Shiue YL. Aptamer/doxorubicin-conjugated nanoparticles target membranous CEMIP2 in colorectal cancer. Int J Biol Macromol 2023; 245:125510. [PMID: 37353120 DOI: 10.1016/j.ijbiomac.2023.125510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
The objectives were to identify the functional domains of a potential oncoprotein, cell migration inducing hyaluronidase 2 (CEMIP2), evaluate its expression levels and roles in colorectal cancer (CRC), and develop an aptamer-based nanoparticle for targeted therapy. Data mining on TCGA identified that CEMIP2 might play oncogenic roles in CRC. In a local cohort, CEMIP2 mRNA levels significantly stepwise increase in CRC patients with higher stages, and high CEMIP2 confers worse disease-free survival. In addition, CEMIP2 mRNA levels significantly correlated to hyaluronan levels in sera from CRC patients. Deletion mapping identified that CEMIP2 containing G8 and PANDER-like domains preserved hyaluronidase activity and oncogenic roles, including cell proliferation, anchorage-independent cell growth, cell migration and invasion, and human umbilical vein endothelial cell (HUVEC) tube formation in CRC-derived cells. A customized monoclonal mouse anti-human CEMIP2 antibody probing the PANDER-like domain (anti-289307) counteracted CEMIP2-mediated carcinogenesis in vitro. Cell-SELEX pinpointed an aptamer, aptCEMIP2(101), specifically interacted with the full-length CEMIP2, potentially involving its 3D structure. Treatments with aptCEMIP2(101) significantly reduced CEMIP2-mediated tumorigenesis in vitro. Mesoporous silica nanoparticles (MSN) carrying atpCEMIP2(101) and Dox were fabricated. Dox@MSN, MSN-aptCEMIP2(101), and Dox@MSN-aptCEMIP2(101) significantly suppressed tumorigenesis in vitro compared to the Mock, while Dox@MSN-aptCEMIP2(101) showed substantially higher effects compared to Dox@MSN and MSN-aptCEMIP2(101) in CRC-derived cells. Our study identified a novel oncogene and developed an effective aptamer-based targeted therapeutic strategy.
Collapse
Affiliation(s)
- Maryam Kianpour
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Ching-Wen Huang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Pichpisith Pierre Vejvisithsakul
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Jaw-Yuan Wang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; Pingtung Hospital, Ministry of Health and Welfare, Pingtung 900214, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Chien-Feng Li
- Department of Medical Research, Chi Mei Medical Center, Tainan 71004, Taiwan; Department of Pathology, Chi Mei Medical Center, Tainan 71004, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan
| | - Meng-Shin Shiao
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Cheng-Tang Pan
- Institute of Precision Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|
20
|
Abbasi M, Sohail M, Minhas MU, Mahmood A, Shah SA, Munir A, Kashif MUR. Folic acid-decorated alginate nanoparticles loaded hydrogel for the oral delivery of diferourylmethane in colorectal cancer. Int J Biol Macromol 2023; 233:123585. [PMID: 36758757 DOI: 10.1016/j.ijbiomac.2023.123585] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/25/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
The disease-related suffering in colorectal cancer remains prevalent despite advancements in the field of drug delivery. Chemotherapy-related side effects and non-specificity remain a challenge in drug delivery. The great majority of hydrophobic drugs cannot be successfully delivered to the colon orally mainly due to poor solubility, low bioavailability, pH differences, and food interactions. Polymeric nanoparticles are potential drug delivery candidates but there are numerous limitations to their usefulness in colon cancer. The nanoparticles are removed from the body rapidly by p-glycoprotein efflux, inactivation, or breakdown by enzymes limiting their efficiency. Furthermore, there is a lack of selectivity in targeting cancer cells; nanoparticles may also target healthy cells, resulting in toxicity and adverse effects. The study aimed to use nanoparticles for specific targeting of the colorectal tumor cells via the oral route of administration without adverse effects. Folic acid (FA), a cancer-targeting ligand possessing a high affinity for folate receptors overexpressed in colorectal cancers was conjugated to sodium alginate- nanoparticles by NH2-linkage. The folic-acid conjugated nanoparticles (FNPs) were delivered to the colon by a pH-sensitive hydrogel synthesized by the free radical polymerization method to provide sustained drug release. The developed system referred to as the "Hydrogel-Nano (HN) drug delivery system," was specifically capable of delivering diferourylmethane to the colon. The HN system was characterized by DLS, FTIR, XRD, TGA, DSC, and SEM. The FNPs size, polydispersity index, and zeta potential were measured. The folic acid-conjugation to nanoparticles' surface was studied by UV-visible spectroscopy using Beer-Lambert's law. In-vitro studies, including sol-gel, porosity, drug loading, entrapment efficiency, etc., revealed promising results. The swelling and release studies showed pH-dependent release of the drug in colonic pH 7.4. Cellular uptake and cytotoxicity studies performed on FR-overexpressed Hela cell lines and FR-negative A-549 cell lines showed facilitated uptake of nanoparticles by folate receptors. A threefold increase in Cmax and prolongation of the mean residence time (MRT) to 14.52 +/- 0.217 h indicated sustained drug release by the HN system. The findings of the study can provide a sufficient ground that the synergistic approach of the HN system can deliver hydrophobic drugs to colorectal cancer cells via the oral route, but further in-vivo animal cancer model studies are required.
Collapse
Affiliation(s)
- Mudassir Abbasi
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| | - Muhammad Sohail
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan; Faculty of Pharmacy, Cyprus International University, Nicosia, 99258, North Cyprus.
| | | | - Arshad Mahmood
- Collage of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Syed Ahmed Shah
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan; Department of Pharmaceutical Sciences, The Superior University, Lahore 54600, Pakistan
| | - Abubakar Munir
- Department of Pharmaceutical Sciences, The Superior University, Lahore 54600, Pakistan
| | - Mehboob-Ur-Rehman Kashif
- Department of Pharmacy, COMSATS University, Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| |
Collapse
|
21
|
He S, Du Y, Tao H, Duan H. Advances in aptamer-mediated targeted delivery system for cancer treatment. Int J Biol Macromol 2023; 238:124173. [PMID: 36965552 DOI: 10.1016/j.ijbiomac.2023.124173] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Aptamers with high affinity and specificity for certain targets have rapidly become a novel class of targeted ligands applicated in drug delivery. Based on the excellent characteristics of aptamers, different aptamer-mediated drug delivery systems have been developed, including aptamer-drug conjugate (ApDC), aptamer-siRNA, and aptamer-functionalized nanoparticle systems for the effective treatment of cancer, which can reduce potential toxicity and improve therapeutic efficacy. In this review, we summarize the recent progress of aptamer-mediated delivery systems in cancer therapy, and discuss the application prospects and existing problems of innovative approaches based on aptamer therapy. Overall, this review aims to better understand the current aptamer-based targeted delivery applications through in-depth analysis to improve efficacy and develop new therapeutic methods which can ultimately improve treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Tao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Huaiyu Duan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
22
|
Fan R, Tao X, Zhai X, Zhu Y, Li Y, Chen Y, Dong D, Yang S, Lv L. Application of aptamer-drug delivery system in the therapy of breast cancer. Biomed Pharmacother 2023; 161:114444. [PMID: 36857912 DOI: 10.1016/j.biopha.2023.114444] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Despite significant treatment advances, breast cancer remains the leading cause of cancer death in women. From the current treatment situation, in addition to developing chemoresistant tumours, distant organ metastasis, and recurrences, patients with breast cancer often have a poor prognosis. Aptamers as "chemical antibodies" may be a way to resolve this dilemma. Aptamers are single-stranded, non-coding oligonucleotides (DNA or RNA), resulting their many advantages, including stability for long-term storage, simplicity of synthesis and function, and low immunogenicity, a high degree of specificity and antidote. Aptamers have gained popularity as a method for diagnosing and treating specific tumors in recent years. This article introduces the application of ten different aptamer delivery systems in the treatment and diagnosis of breast cancer, and systematically reviews their latest research progress in breast cancer treatment and diagnosis. It provides a new direction for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Rui Fan
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunming Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanwei Chen
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
23
|
Torabi M, Aghanejad A, Savadi P, Barzegari A, Omidi Y, Barar J. Fabrication of mesoporous silica nanoparticles for targeted delivery of sunitinib to ovarian cancer cells. BIOIMPACTS : BI 2023; 13:255-267. [PMID: 37431477 PMCID: PMC10329750 DOI: 10.34172/bi.2023.25298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/06/2022] [Accepted: 07/23/2022] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Mesoporous silica nanoparticles (MSNPs) are considered innovative multifunctional structures for targeted drug delivery owing to their outstanding physicochemical characteristics. METHODS MSNPs were fabricated using the sol-gel method, and polyethylene glycol-600 (PEG600) was used for MSNPs modification. Subsequently, sunitinib (SUN) was loaded into the MSNPs, MSNP-PEG and MSNP-PEG/SUN were grafted with mucin 16 (MUC16) aptamers. The nanosystems (NSs) were characterized using FT-IR, TEM, SEM, DLS, XRD, BJH, and BET. Furthermore, the biological impacts of MSNPs were evaluated on the ovarian cancer cells by MTT assay and flow cytometry analysis. RESULTS The results revealed that the MSNPs have a spherical shape with an average dimension, pore size, and surface area of 56.10 nm, 2.488 nm, and 148.08 m2g-1, respectively. The cell viability results showed higher toxicity of targeted MSNPs in MUC16 overexpressing OVCAR-3 cells as compared to the SK-OV-3 cells; that was further confirmed by the cellular uptake results. The cell cycle analysis exhibited that the induction of sub-G1 phase arrest mostly occurred in MSNP-PEG/SUN-MUC16 treated OVCAR-3 cells and MSNP-PEG/SUN treated SK-OV-3 cells. DAPI staining showed apoptosis induction upon exposure to targeted MSNP in MUC16 positive OVCAR-3 cells. CONCLUSION According to our results, the engineered NSs could be considered an effective multifunctional targeted drug delivery platform for the mucin 16 overexpressing cells.
Collapse
Affiliation(s)
- Mitra Torabi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouria Savadi
- Di.S.T.A.Bi.F., University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
24
|
Torabi M, Aghanejad A, Savadi P, Barzegari A, Omidi Y, Barar J. Targeted Delivery of Sunitinib by MUC-1 Aptamer-Capped Magnetic Mesoporous Silica Nanoparticles. Molecules 2023; 28:molecules28010411. [PMID: 36615606 PMCID: PMC9824472 DOI: 10.3390/molecules28010411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Magnetic mesoporous silica nanoparticles (MMSNPs) are being widely investigated as multifunctional novel drug delivery systems (DDSs) and play an important role in targeted therapy. Here, magnetic cores were synthesized using the thermal decomposition method. Further, to improve the biocompatibility and pharmacokinetic behavior, mesoporous silica was synthesized using the sol-gel process to coat the magnetic cores. Subsequently, sunitinib (SUN) was loaded into the MMSNPs, and the particles were armed with amine-modified mucin 1 (MUC-1) aptamers. The MMSNPs were characterized using FT-IR, TEM, SEM, electrophoresis gel, DLS, and EDX. MTT assay, flow cytometry analysis, ROS assessment, and mitochondrial membrane potential analysis evaluated the nanoparticles' biological impacts. The physicochemical analysis revealed that the engineered MMSNPs have a smooth surface and spherical shape with an average size of 97.6 nm. The biological in vitro analysis confirmed the highest impacts of the targeted MMSNPs in MUC-1 overexpressing cells (OVCAR-3) compared to the MUC-1 negative MDA-MB-231 cells. In conclusion, the synthesized MMSNP-SUN-MUC-1 nanosystem serves as a unique multifunctional targeted delivery system to combat the MUC-1 overexpressing ovarian cancer cells.
Collapse
Affiliation(s)
- Mitra Torabi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 516664-14766, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Correspondence: (A.A.); or (J.B.); Tel./Fax: +98-41-33367929 (A.A.); +1-(954)-262-1878 (J.B.)
| | - Pouria Savadi
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (Di.S.T.A.Bi.F.), University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 516664-14766, Iran
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
- Correspondence: (A.A.); or (J.B.); Tel./Fax: +98-41-33367929 (A.A.); +1-(954)-262-1878 (J.B.)
| |
Collapse
|
25
|
Yin X, He Z, Ge W, Zhao Z. Application of aptamer functionalized nanomaterials in targeting therapeutics of typical tumors. Front Bioeng Biotechnol 2023; 11:1092901. [PMID: 36873354 PMCID: PMC9978196 DOI: 10.3389/fbioe.2023.1092901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/01/2023] [Indexed: 02/22/2023] Open
Abstract
Cancer is a major cause of human death all over the world. Traditional cancer treatments include surgery, radiotherapy, chemotherapy, immunotherapy, and hormone therapy. Although these conventional treatment methods improve the overall survival rate, there are some problems, such as easy recurrence, poor treatment, and great side effects. Targeted therapy of tumors is a hot research topic at present. Nanomaterials are essential carriers of targeted drug delivery, and nucleic acid aptamers have become one of the most important targets for targeted tumor therapy because of their high stability, high affinity, and high selectivity. At present, aptamer-functionalized nanomaterials (AFNs), which combine the unique selective recognition characteristics of aptamers with the high-loading performance of nanomaterials, have been widely studied in the field of targeted tumor therapy. Based on the reported application of AFNs in the biomedical field, we introduce the characteristics of aptamer and nanomaterials, and the advantages of AFNs first. Then introduce the conventional treatment methods for glioma, oral cancer, lung cancer, breast cancer, liver cancer, colon cancer, pancreatic cancer, ovarian cancer, and prostate cancer, and the application of AFNs in targeted therapy of these tumors. Finally, we discuss the progress and challenges of AFNs in this field.
Collapse
Affiliation(s)
- Xiujuan Yin
- Department of Radiology, Shaoxing People's Hospital, Shaoxing, China.,Key Laboratory of Functional Molecular Imaging of Tumor and Interventional Diagnosis and Treatment of Shaoxing City, Shaoxing, China
| | - Zhenqiang He
- Clinical Medical College of Hebei University, Baoding, China.,Department of Radiology, Hebei University Affiliated Hospital, Baoding, China
| | - Weiying Ge
- Department of Radiology, Hebei University Affiliated Hospital, Baoding, China
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital, Shaoxing, China.,Key Laboratory of Functional Molecular Imaging of Tumor and Interventional Diagnosis and Treatment of Shaoxing City, Shaoxing, China.,Medical College of Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Aptamers Enhance Oncolytic Viruses' Antitumor Efficacy. Pharmaceutics 2022; 15:pharmaceutics15010151. [PMID: 36678780 PMCID: PMC9864469 DOI: 10.3390/pharmaceutics15010151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
Oncolytic viruses are highly promising for cancer treatment because they target and lyse tumor cells. These genetically engineered vectors introduce therapeutic or immunostimulatory genes into the tumor. However, viral therapy is not always safe and effective. Several problems are related to oncolytic viruses' targeted delivery to the tumor and immune system neutralization in the bloodstream. Cryoprotection and preventing viral particles from aggregating during storage are other critical issues. Aptamers, short RNA, or DNA oligonucleotides may help to crawl through this bottleneck. They are not immunogenic, are easily synthesized, can be chemically modified, and are not very demanding in storage conditions. It is possible to select an aptamer that specifically binds to any target cell, oncolytic virus, or molecule using the SELEX technology. This review comprehensively highlights the most important research and methodological approaches related to oncolytic viruses and nucleic acid aptamers. Here, we also analyze possible future research directions for combining these two methodologies to improve the effectiveness of cancer virotherapy.
Collapse
|
27
|
Pei Z, Chen S, Ding L, Liu J, Cui X, Li F, Qiu F. Current perspectives and trend of nanomedicine in cancer: A review and bibliometric analysis. J Control Release 2022; 352:211-241. [PMID: 36270513 DOI: 10.1016/j.jconrel.2022.10.023] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022]
Abstract
The limitations of traditional cancer treatments are driving the creation and development of new nanomedicines. At present, with the rapid increase of research on nanomedicine in the field of cancer, there is a lack of intuitive analysis of the development trend, main authors and research hotspots of nanomedicine in the field of cancer, as well as detailed elaboration of possible research hotspots. In this review, data collected from the Web of Science Core Collection database between January 1st, 2000, and December 31st, 2021, were subjected to a bibliometric analysis. The co-authorship, co-citation, and co-occurrence of countries, institutions, authors, literature, and keywords in this subject were examined using VOSviewer, Citespace, and a well-known online bibliometrics platform. We collected 19,654 published papers, China produced the most publications (36.654%, 7204), followed by the United States (29.594%, 5777), and India (7.780%, 1529). An interesting fact is that, despite China having more publications than the United States, the United States still dominates this field, having the highest H-index and the most citations. Acs Nano, Nano Letters, and Biomaterials are the top three academic publications that publish articles on nanomedicine for cancer out of a total of 7580 academic journals. The most significant increases were shown for the keywords "cancer nanomedicine", "tumor microenvironment", "nanoparticles", "prodrug", "targeted nanomedicine", "combination", and "cancer immunotherapy" indicating the promising area of research. Meanwhile, the development prospects and challenges of nanomedicine in cancer are also discussed and provided some solutions to the major obstacles.
Collapse
Affiliation(s)
- Zerong Pei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuting Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Liqin Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xinyi Cui
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
28
|
Lee J, Bae J, Kwak D, Kim H, Kim J, Phyu Hlaing S, Saparbayeva A, Hee Lee E, Yoon IS, Kim MS, Ryong Moon H, Yoo JW. 5-Fluorouracil crystal-incorporated, pH-responsive, and release-modulating PLGA/Eudragit FS hybrid microparticles for local colorectal cancer-targeted chemotherapy. Int J Pharm 2022; 630:122443. [PMID: 36503847 DOI: 10.1016/j.ijpharm.2022.122443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
5-Fluorouracil (5-FU) is a widely used chemotherapeutic agent for colorectal cancer (CRC) owing to its potent anticancer effects. However, severe systemic side effects and poor drug accumulation in the CRC tissues limit its efficacy. This study aimed to develop 5-FU crystal-incorporated, pH-responsive, and release-modulating poly(d,l-lactide-co-glycolide)/Eudragit FS hybrid microparticles (5FU-EPMPs) for the local CRC-targeted chemotherapy. Approximately 150 μm 5FU-EPMPs were fabricated via the S/O/W emulsion solvent evaporation method, with 7.93 ± 0.24% and 87.23 ± 2.64% 5-FU loading and encapsulation efficiencies, respectively. Drug release profiles in a simulated pH environment of the gastrointestinal tract revealed that premature 5-FU release in the stomach and small intestine was prevented, thereby minimizing systemic 5-FU absorption. After reaching the colon, 5-FU was continuously released for >15 h, allowing long-term exposure of CRC tissues to sufficient 5-FU concentrations. Furthermore, in a CRC mouse model, the 5FU-EPMPs showed potent inhibition of tumor growth without signs of systemic toxicity. Thus, the 5FU-EPMPs represent a promising drug delivery system for local CRC-targeted chemotherapy.
Collapse
Affiliation(s)
- Juho Lee
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Junhwan Bae
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Dongmin Kwak
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Hyunwoo Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Jihyun Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Shwe Phyu Hlaing
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Aruzhan Saparbayeva
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Eun Hee Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - In-Soo Yoon
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Min-Soo Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Hyung Ryong Moon
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Jin-Wook Yoo
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
29
|
Wei Z, Zhou Y, Wang R, Wang J, Chen Z. Aptamers as Smart Ligands for Targeted Drug Delivery in Cancer Therapy. Pharmaceutics 2022; 14:2561. [PMID: 36559056 PMCID: PMC9781707 DOI: 10.3390/pharmaceutics14122561] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Undesirable side effects and multidrug tolerance are the main holdbacks to the treatment of cancer in conventional chemotherapy. Fortunately, targeted drug delivery can improve the enrichment of drugs at the target site and reduce toxicity to normal tissues and cells. A targeted drug delivery system is usually composed of a nanocarrier and a targeting component. The targeting component is called a "ligand". Aptamers have high target affinity and specificity, which are identified as attractive and promising ligands. Therefore, aptamers have potential application in the development of smart targeting systems. For instance, aptamers are able to efficiently recognize tumor markers such as nucleolin, mucin, and epidermal growth factor receptor (EGFR). Besides, aptamers can also identify glycoproteins on the surface of tumor cells. Thus, the aptamer-mediated targeted drug delivery system has received extensive attention in the application of cancer therapy. This article reviews the application of aptamers as smart ligands for targeted drug delivery in cancer therapy. Special interest is focused on aptamers as smart ligands, aptamer-conjugated nanocarriers, aptamer targeting strategy for tumor microenvironment (TME), and aptamers that are specified to crucial cancer biomarkers for targeted drug delivery.
Collapse
Affiliation(s)
| | | | | | - Jin Wang
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Zhenhua Chen
- Jiangxi Province Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| |
Collapse
|
30
|
Hu X, Zhang D, Zeng Z, Huang L, Lin X, Hong S. Aptamer-Based Probes for Cancer Diagnostics and Treatment. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111937. [PMID: 36431072 PMCID: PMC9695321 DOI: 10.3390/life12111937] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/23/2022] [Accepted: 11/12/2022] [Indexed: 11/22/2022]
Abstract
Aptamers are single-stranded DNA or RNA oligomers that have the ability to generate unique and diverse tertiary structures that bind to cognate molecules with high specificity. In recent years, aptamer researches have witnessed a huge surge, owing to its unique properties, such as high specificity and binding affinity, low immunogenicity and toxicity, and simplicity of synthesis with negligible batch-to-batch variation. Aptamers may bind to targets, such as various cancer biomarkers, making them applicable for a wide range of cancer diagnosis and treatment. In cancer diagnostic applications, aptamers are used as molecular probes instead of antibodies. They have the potential to detect various cancer-associated biomarkers. For cancer therapeutic purposes, aptamers can serve as therapeutic or delivery agents. The chemical stabilization and modification strategies for aptamers may expand their serum half-life and shelf life. However, aptamer-based probes for cancer diagnosis and therapy still face several challenges for successful clinical translation. A deeper understanding of nucleic acid chemistry, tissue distribution, and pharmacokinetics is required in the development of aptamer-based probes. This review summarizes their application in cancer diagnostics and treatments based on different localization of target biomarkers, as well as current challenges and future prospects.
Collapse
|
31
|
Moradi M, Mohabatkar H, Behbahani M, Dini G. Application of G-quadruplex aptamer conjugated MSNs to deliver ampicillin for suppressing S. aureus biofilm on mice bone. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
32
|
Ghasemii K, Darroudi M, Rahimmanesh I, Ghomi M, Hassanpour M, Sharifi E, Yousefiasl S, Ahmadi S, Zarrabi A, Borzacchiello A, Rabiee M, Paiva-Santos AC, Rabiee N. Advances in aptamer-based drug delivery vehicles for cancer therapy. BIOMATERIALS ADVANCES 2022; 140:213077. [PMID: 35952549 DOI: 10.1016/j.bioadv.2022.213077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
Overall, aptamers are special classes of nucleic acid-based macromolecules that are beginning to investigate because of their capability of avidity binding to a specific target for clinical use. Taking advantage of target-specific medicine led to more effective therapeutic and limitation of side effects of drugs. Herein, we discuss several aptamers and their binding capability and capacity for selecting tumor biomarkers and usage of them as targeting ligands for the functionalization of nanomaterials. We review recent applications based on aptamers and several nanoparticles to rise efficacy and develop carrier systems such as graphene oxide, folic acid, gold, mesopores silica, and various polymers and copolymer, polyethylene glycol, cyclodextrin, chitosan. The nanocarriers have been characterized by particle size, zeta potential, aptamer conjugation, and drug encapsulation efficiency. Hydrodynamic diameter and Zeta potential can used in order to monitor aptamers' crosslinking, in-vitro drug release, intracellular delivery of nanocarriers, and cellular cytotoxicity assay. Also, they are studied for cellular uptake and internalization to types of cancer cell lines such as colorectal, breast, prostate, leukemia and etc. The results are investigated in in-vivo cytotoxicity assay and cell viability assay. Targeted cancer therapy seems a good and promising strategy to overcome the systemic toxicity of chemotherapy.
Collapse
Affiliation(s)
- Kousar Ghasemii
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, Iran
| | - Mahdieh Darroudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan 36716-41167, Iran
| | - Mahnaz Hassanpour
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Esmaeel Sharifi
- Institute for Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Satar Yousefiasl
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Science, Istinye University, Sariyer 34396, Istanbul, Turkey
| | - Assunta Borzacchiello
- Institute for Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, 80125 Naples, Italy
| | - Mohammad Rabiee
- Biomaterial group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Navid Rabiee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea; School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia.
| |
Collapse
|
33
|
Li S, Coffinier Y, Lagadec C, Cleri F, Nishiguchi K, Fujiwara A, Fujii T, Kim SH, Clément N. Redox-labelled electrochemical aptasensors with nanosupported cancer cells. Biosens Bioelectron 2022; 216:114643. [PMID: 36030742 DOI: 10.1016/j.bios.2022.114643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/31/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022]
Abstract
The transfer of redox-labelled bioelectrochemical sensors from proteins to cells is not straightforward because of the cell downward force issue on the surface of the sensors. In this paper, 20-nm-thick nanopillars are introduced to overcome this issue, in a well-controlled manner. We show on both molecular dynamics simulations and experiments that suspending cells a few nanometers above an electrode surface enables redox-labelled tethered DNA aptamer probes to move freely, while remaining at an interaction distance from a target membrane protein, i. e. epithelial cell adhesion molecule (EpCAM), which is typically overexpressed in cancer cells. By this nanopillar configuration, the interaction of aptamer with cancer cells is clearly observable, with 13 cells as the lower limit of detection. Nanoconfinement induced by the gap between the electrode surface and the cell membrane appears to improve the limit of detection and to lower the melting temperature of DNA aptamer hairpins, offering an additional degree of freedom to optimize molecular recognition mechanisms. This novel nanosupported electrochemical DNA cell sensor scheme including Brownian-fluctuating redox species opens new opportunities for the design of all-electrical sensors using redox-labelled probes.
Collapse
Affiliation(s)
- S Li
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo, 4-6-1 Komaba, Meguro-ku Tokyo, 153-8505, Japan.
| | - Y Coffinier
- IEMN, CNRS UMR8520, Univ. Lille Avenue Poincaré, BP 60069, Villeneuve D'Ascq Cedex, 59652, France
| | - C Lagadec
- Univ. Lille, CNRS, Inserm, CHU Lille, Centre Oscar Lambret, UMR9020 - UMR-S 1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - F Cleri
- IEMN, CNRS UMR8520, Univ. Lille Avenue Poincaré, BP 60069, Villeneuve D'Ascq Cedex, 59652, France
| | - K Nishiguchi
- NTT Basic Research Laboratories, NTT Corporation, 3-1, Morinosato-Wakamiya, Atsugi-shi, 243-0198, Japan
| | - A Fujiwara
- NTT Basic Research Laboratories, NTT Corporation, 3-1, Morinosato-Wakamiya, Atsugi-shi, 243-0198, Japan
| | - T Fujii
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo, 4-6-1 Komaba, Meguro-ku Tokyo, 153-8505, Japan
| | - S-H Kim
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo, 4-6-1 Komaba, Meguro-ku Tokyo, 153-8505, Japan
| | - N Clément
- IIS, LIMMS/CNRS-IIS IRL2820, The Univ. of Tokyo, 4-6-1 Komaba, Meguro-ku Tokyo, 153-8505, Japan.
| |
Collapse
|
34
|
Vallet-Regí M, Schüth F, Lozano D, Colilla M, Manzano M. Engineering mesoporous silica nanoparticles for drug delivery: where are we after two decades? Chem Soc Rev 2022; 51:5365-5451. [PMID: 35642539 PMCID: PMC9252171 DOI: 10.1039/d1cs00659b] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Collapse
Affiliation(s)
- María Vallet-Regí
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ferdi Schüth
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Daniel Lozano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Montserrat Colilla
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
35
|
Mohaghegh S, Tarighatnia A, Omidi Y, Barar J, Aghanejad A, Adibkia K. Multifunctional Magnetic Nanoparticles for MRI-guided Co-delivery of Erlotinib and L-Asparaginase to Ovarian Cancer. J Microencapsul 2022; 39:394-408. [PMID: 35748819 DOI: 10.1080/02652048.2022.2094487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AIM(S) The use of magnetic nanoparticles (MNPs) in biomedical applications has been wildly opted due to their unique properties. The objective of this study was to evaluate the effects of aptamer-armed MNPs in ovarian cancer treatment and as T2 weighted MRI contrast agent. METHODS Here, we designed MNPs loaded with erlotinib (ERL/SPION-Val-PEG) and conjugated them with anti-mucin16 (MUC16) aptamer to introduce new image-guided nanoparticles (NPs) for targeted drug delivery as well as non-invasive magnetic resonance imaging (MRI) contrast agents. Also, the combination of our nanosystem (NS) along with L-Asparaginase (L-ASPN) led to synergistic effects in terms of reducing cell viability in ovarian cancer cells, which could suggest a novel combination therapy. RESULTS The mean size of our NS was about 63.4 ± 3.4 nm evaluated by DLS analysis and its morphology was confirmed using TEM. Moreover, the functional groups, as well as magnetic properties of our NS, were examined by FT-IR and VSM tests, respectively. The loading efficacy of erlotinib on MNPs was about 80% and its release reached 70.85% over 7 days in the pH value of 5.4. The MR images and flow cytometry results revealed that the cellular uptake of ERL/SPION-Val-PEG-MUC16 NPs in cells with MUC16 overexpression was considerably higher than unarmed NPs. In addition, T2-weight MR images of ovarian cancer-bearing mice indicated significant signal intensity changes at the tumor site 4 h after intravenous injection compared to the non-target MNPs. CONCLUSIONS Our data suggest ERL/SPION-Val-PEG NPs as an image-guided co-drug delivery system for ovarian cancer.
Collapse
Affiliation(s)
- Seraj Mohaghegh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Tarighatnia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Florida, USA
| | - Jaleh Barar
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Ahmadi A, Sokunbi M, Patel T, Chang MW, Ahmad Z, Singh N. Influence of Critical Parameters on Cytotoxicity Induced by Mesoporous Silica Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2016. [PMID: 35745355 PMCID: PMC9228019 DOI: 10.3390/nano12122016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 02/01/2023]
Abstract
Mesoporous Silica Nanoparticles (MSNs) have received increasing attention in biomedical applications due to their tuneable pore size, surface area, size, surface chemistry, and thermal stability. The biocompatibility of MSNs, although generally believed to be satisfactory, is unclear. Physicochemical properties of MSNs, such as diameter size, morphology, and surface charge, control their biological interactions and toxicity. Experimental conditions also play an essential role in influencing toxicological results. Therefore, the present study includes studies from the last five years to statistically analyse the effect of various physicochemical features on MSN-induced in-vitro cytotoxicity profiles. Due to non-normally distributed data and the presence of outliers, a Kruskal-Wallis H test was conducted on different physicochemical characteristics, including diameter sizes, zeta-potential measurements, and functionalisation of MSNs, based on the viability results, and statistical differences were obtained. Subsequently, pairwise comparisons were performed using Dunn's procedure with a Bonferroni correction for multiple comparisons. Other experimental parameters, such as type of cell line used, cell viability measurement assay, and incubation time, were also explored and analysed for statistically significant results.
Collapse
Affiliation(s)
- Amirsadra Ahmadi
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| | - Moses Sokunbi
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| | - Trisha Patel
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, Jordanstown Campus, University of Ulster, Newtownabbey BT37 0QB, UK;
| | - Zeeshan Ahmad
- Leicester School of Pharmaceutical Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK;
| | - Neenu Singh
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| |
Collapse
|
37
|
Abbasi M, Sohail M, Minhas MU, Iqbal J, Mahmood A, Shaikh AJ. Folic acid-functionalized nanoparticles-laden biomaterials for the improved oral delivery of hydrophobic drug in colorectal cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
38
|
Guo L, Zhang Y, Wang Y, Xie M, Dai J, Qu Z, Zhou M, Cao S, Shi J, Wang L, Zuo X, Fan C, Li J. Directing Multivalent Aptamer-Receptor Binding on the Cell Surface with Programmable Atom-Like Nanoparticles. Angew Chem Int Ed Engl 2022; 61:e202117168. [PMID: 35226386 DOI: 10.1002/anie.202117168] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Indexed: 11/08/2022]
Abstract
Multivalent interactions of biomolecules play pivotal roles in physiological and pathological settings. Whereas the directionality of the interactions is crucial, the state-of-the-art synthetic multivalent ligand-receptor systems generally lack programmable approaches for orthogonal directionality. Here, we report the design of programmable atom-like nanoparticles (aptPANs) to direct multivalent aptamer-receptor binding on the cell interface. The positions of the aptamer motifs can be prescribed on tetrahedral DNA frameworks to realize atom-like orthogonal valence and direction, enabling the construction of multivalent molecules with fixed aptamer copy numbers but different directionality. These directional-yet-flexible aptPAN molecules exhibit the adaptability to the receptor distribution on cell surfaces. We demonstrate the high-affinity tumor cell binding with a linear aptPAN oligomer (≈13-fold improved compared to free aptamers), which leads to ≈50 % suppression of cell growth.
Collapse
Affiliation(s)
- Linjie Guo
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yueyue Zhang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Wang
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mo Xie
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Jiangbing Dai
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Zhibei Qu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Mo Zhou
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Shuting Cao
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Jiye Shi
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Lihua Wang
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China.,Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Jiang Li
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.,The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China.,School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| |
Collapse
|
39
|
Porrang S, Davaran S, Rahemi N, Allahyari S, Mostafavi E. How Advancing are Mesoporous Silica Nanoparticles? A Comprehensive Review of the Literature. Int J Nanomedicine 2022; 17:1803-1827. [PMID: 35498391 PMCID: PMC9043011 DOI: 10.2147/ijn.s353349] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/31/2022] [Indexed: 12/12/2022] Open
Abstract
The application of mesoporous silica nanoparticles (MSNs) is ubiquitous in various sciences. MSNs possess unique features, including the diversity in manufacturing by different synthesis methods and from different sources, structure controllability, pore design capabilities, pore size tunability, nanoparticle size distribution adjustment, and the ability to create diverse functional groups on their surface. These characteristics have led to various types of MSNs as a unique system for drug delivery. In this review, first, the synthesis of MSNs by different methods via using different sources were studied. Then, the parameters affecting their physicochemical properties and functionalization have been discussed. Finally, the last decade's novel strategies, including surface functionalization, drug delivery, and cancer treatment, based on the MSNs in drug delivery and cancer therapy have been addressed.
Collapse
Affiliation(s)
- Sahar Porrang
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, Iran
- Environmental Engineering Research Centre, Sahand University of Technology, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Centre for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader Rahemi
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, Iran
- Environmental Engineering Research Centre, Sahand University of Technology, Tabriz, Iran
| | - Somaiyeh Allahyari
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, Iran
- Environmental Engineering Research Centre, Sahand University of Technology, Tabriz, Iran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
40
|
Guo L, Zhang Y, Wang Y, Xie M, Dai J, Qu Z, Zhou M, Cao S, Shi J, Wang L, Zuo X, Fan C, Li J. Directing Multivalent Aptamer‐Receptor Binding on the Cell Surface with Programmable Atom‐Like Nanoparticles. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202117168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Linjie Guo
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
- University of Chinese Academy of Sciences Beijing 100049 China
- The Interdisciplinary Research Center Shanghai Synchrotron Radiation Facility Zhangjiang Laboratory Shanghai Advanced Research Institute Chinese Academy of Sciences Shanghai 201210 China
| | - Yueyue Zhang
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Yue Wang
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Mo Xie
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Jiangbing Dai
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Zhibei Qu
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine Shanghai Jiao Tong University 200240 Shanghai China
| | - Mo Zhou
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Shuting Cao
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Jiye Shi
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Lihua Wang
- The Interdisciplinary Research Center Shanghai Synchrotron Radiation Facility Zhangjiang Laboratory Shanghai Advanced Research Institute Chinese Academy of Sciences Shanghai 201210 China
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University Shanghai 200241 China
| | - Xiaolei Zuo
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine Shanghai Jiao Tong University 200240 Shanghai China
| | - Jiang Li
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
- The Interdisciplinary Research Center Shanghai Synchrotron Radiation Facility Zhangjiang Laboratory Shanghai Advanced Research Institute Chinese Academy of Sciences Shanghai 201210 China
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine Shanghai Jiao Tong University 200240 Shanghai China
| |
Collapse
|
41
|
Design and synthesis of targeted star-shaped micelle for guided delivery of camptothecin: In vitro and in vivo evaluation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112529. [PMID: 34857308 DOI: 10.1016/j.msec.2021.112529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
This study aimed to synthesize a star-shaped micelle using 3-azido-2,2-bis(azidomethyl)propan-1-ol (pentaerythritol triazide) core, as an initiator for the synthesis of three-arm polylactic acid (PLA) block. Then, the ends of the PLA arms were converted to PLA triazide followed by conjugation to the three alkyne-PEG-maleamide through click reaction. The maleamide ends were available for coupling with sulfhydryl-modified DNA aptamer against epithelial cell adhesion molecule in order to offer targeted delivery of encapsulated drug, camptothecin to the site of action. The successful synthesis of the star-shaped polymers was confirmed via1HNMR. Hydrophobic anti-cancer drug, camptothecin was encapsulated into the micelles core implementing solvent switching method providing loading content (LC%) and encapsulation efficiency (EE%) of 3.7 ± 0.4 and 73.7 ± 8.2, respectively. The size of both non-targeted and aptamer-targeted micelles was determined to be 154 and 192 nm, respectively with polydispersity index below 0.3. In vitro drug release evaluation at 37 °C, pH 7.4 showed a controlled release pattern for camptothecin during 72 h. In vitro cytotoxicity of the prepared non-targeted and targeted micelles was carried out on human colorectal adenocarcinoma (HT29) and mouse colon carcinoma (C26) as EpCAM positive cell lines and Chinese hamster ovary (CHO) as EpCAM negative cell line. The results verified significantly higher cytotoxicity of the targeted micelles on HT29 and C26 cell lines, while no obvious difference was observed between targeted and non-targeted formulation on CHO cell line. The in vivo therapeutic efficiency investigation on BALB/c C26 tumor-bearing mice showed superior capability of the targeted formulation on tumor suppression and survival rate of the treated mice. The developed platform exhibited excellent characteristics to diminish camptothecin drawbacks and its adverse effects while considerably increasing its therapeutic index.
Collapse
|
42
|
Du Y, Liu D, Du Y. Recent advances in hepatocellular carcinoma therapeutic strategies and imaging-guided treatment. J Drug Target 2021; 30:287-301. [PMID: 34727794 DOI: 10.1080/1061186x.2021.1999963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant cancer in the world, which greatly threatens human health. However, the routine treatment strategies for HCC have failed to specifically eradicate the tumorigenic cells, leading to the occurrence of metastasis and recurrence. To improve treatment efficacies, the development of novel effective technologies is urgently required. Recently, nanotechnologies have gained the extensive attention in cancer targeted therapy, which could provide a promising way for HCC clinical practice. However, a successful cancer management depends on accurate diagnosis of the tumour along with precise therapeutic protocol, thereby predicting the tumour response to existing therapies. The synergistic effect of targeted therapeutic systems and imaging approaches (also called 'imaging-guided cancer treatment') may establish a more effective platform for individual cancer care. This review outlines the recent advanced nano-targeted and -traceable therapeutic strategies for HCC management. The multifunctional nano agents that have both diagnosis and therapy abilities are highlighted. Finally, we conclude with our perspectives on the future development and challenges of HCC nanotheranostics.
Collapse
Affiliation(s)
- Yan Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Di Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
Iranpour S, Bahrami AR, Nekooei S, Sh Saljooghi A, Matin MM. Improving anti-cancer drug delivery performance of magnetic mesoporous silica nanocarriers for more efficient colorectal cancer therapy. J Nanobiotechnology 2021; 19:314. [PMID: 34641857 PMCID: PMC8507230 DOI: 10.1186/s12951-021-01056-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/21/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Improving anti-cancer drug delivery performance can be achieved through designing smart and targeted drug delivery systems (DDSs). For this aim, it is important to evaluate overexpressed biomarkers in the tumor microenvironment (TME) for optimizing DDSs. MATERIALS AND METHODS Herein, we designed a novel DDS based on magnetic mesoporous silica core-shell nanoparticles (SPION@MSNs) in which release of doxorubicin (DOX) at the physiologic pH was blocked with gold gatekeepers. In this platform, we conjugated heterofunctional polyethylene glycol (PEG) onto the outer surface of nanocarriers to increase their biocompatibility. At the final stage, an epithelial cell adhesion molecule (EpCAM) aptamer as an active targeting moiety was covalently attached (Apt-PEG-Au@NPs-DOX) for selective drug delivery to colorectal cancer (CRC) cells. The physicochemical properties of non-targeted and targeted nanocarriers were fully characterized. The anti-cancer activity, cellular internalization, and then the cell death mechanism of prepared nanocarriers were determined and compared in vitro. Finally, tumor inhibitory effects, biodistribution and possible side effects of the nanocarriers were evaluated in immunocompromised C57BL/6 mice bearing human HT-29 tumors. RESULTS Nanocarriers were successfully synthesized with a mean final size diameter of 58.22 ± 8.54 nm. Higher cytotoxicity and cellular uptake of targeted nanocarriers were shown in the EpCAM-positive HT-29 cells as compared to the EpCAM-negative CHO cells, indicating the efficacy of aptamer as a targeting agent. In vivo results in a humanized mouse model showed that targeted nanocarriers could effectively increase DOX accumulation in the tumor site, inhibit tumor growth, and reduce the adverse side effects. CONCLUSION These results suggest that corporation of a magnetic core, gold gatekeeper, PEG and aptamer can strongly improve drug delivery performance and provide a theranostic DDS for efficient CRC therapy.
Collapse
Affiliation(s)
- Sonia Iranpour
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran. .,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran. .,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
44
|
Zhong J, Ding J, Deng L, Xiang Y, Liu D, Zhang Y, Chen X, Yang Q. Selection of DNA Aptamers Recognizing EpCAM-Positive Prostate Cancer by Cell-SELEX for in vitro and in vivo MR Imaging. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3985-3996. [PMID: 34584404 PMCID: PMC8464308 DOI: 10.2147/dddt.s322854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/06/2021] [Indexed: 11/23/2022]
Abstract
Background The sensitive and specific detection of pathogenic cells is important in tumor diagnosis at an early stage. Aptamers are short single-stranded oligonucleotides evolved from systematic evolution of ligands by exponential enrichment (SELEX). It has been proved that aptamers can interact with cognate target molecules with high affinity and specificity and have great potential in the development of medical imaging at molecular level. Purpose To select epithelial cell adhesion molecule (EpCAM) specific aptamers targeting prostate cancer and further to conjugate aptamers with GoldMag nanoparticles (a typical iron oxide core/gold shell structure) to construct magnetic molecular probes for medical imaging. Methods EpCAM-specific aptamers were selected by Cell-SELEX. The enrichment of specific aptamer candidates was monitored by flow cytometric analysis. Aptamers were further conjugated with GoldMag nanoparticles to construct magnetic molecular probes. The affinity and specificity of aptamer candidates and aptamer-conjugated GoldMag nanoparticles were evaluated. The MR imaging of aptamer-conjugated GoldMag nanoparticles to prostate cancer was further explored in vitro and in vivo. Results After 12 rounds of selection, aptamer candidates Eppc6 and Eppc14 could specifically target three types of prostate cancer cells, revealing a high affinity of Eppc6 and Eppc14. Moreover, aptamer-conjugated GoldMag nanoparticles not only exhibited good affinity to different prostate cancer cells but also produced strong T2WI signal intensity reduction distinguished from peritumoral tissue in MRI, indicating that the molecular probes possess both the affinity properties of EpCAM-specific aptamer and the superparamagnetic features of iron oxide. Conclusion Our study indicates that aptamer Eppc6 and Eppc14 can recognize prostate cancer cells and tissues. The aptamer-conjugated GoldMag nanoparticles constructed in the study can be used as a molecular imaging agent for detection of PCa in MRI.
Collapse
Affiliation(s)
- Jinman Zhong
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Jianke Ding
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, People's Republic of China
| | - Lei Deng
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Ying Xiang
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Duoduo Liu
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Yanyan Zhang
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Xin Chen
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Quanxin Yang
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| |
Collapse
|
45
|
Abstract
Multiple myeloma is the second most common hematological malignancy in adults, accounting for 2% of all cancer-related deaths in the UK. Current chemotherapy-based regimes are insufficient, as most patients relapse and develop therapy resistance. This review focuses on current novel antibody- and aptamer-based therapies aiming to overcome current therapy limitations, as well as their respective limitations and areas of improvement. The use of computer modeling methods, as a tool to study and improve ligand-receptor alignments for the use of novel therapy development will also be discussed, as it has become a rapid, reliable and comparatively inexpensive method of investigation.
Collapse
|
46
|
Application of smart nanoparticles as a potential platform for effective colorectal cancer therapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213949] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Rastegari E, Hsiao YJ, Lai WY, Lai YH, Yang TC, Chen SJ, Huang PI, Chiou SH, Mou CY, Chien Y. An Update on Mesoporous Silica Nanoparticle Applications in Nanomedicine. Pharmaceutics 2021; 13:1067. [PMID: 34371758 PMCID: PMC8309088 DOI: 10.3390/pharmaceutics13071067] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023] Open
Abstract
The efficient and safe delivery of therapeutic drugs, proteins, and nucleic acids are essential for meaningful therapeutic benefits. The field of nanomedicine shows promising implications in the development of therapeutics by delivering diagnostic and therapeutic compounds. Nanomedicine development has led to significant advances in the design and engineering of nanocarrier systems with supra-molecular structures. Smart mesoporous silica nanoparticles (MSNs), with excellent biocompatibility, tunable physicochemical properties, and site-specific functionalization, offer efficient and high loading capacity as well as robust and targeted delivery of a variety of payloads in a controlled fashion. Such unique nanocarriers should have great potential for challenging biomedical applications, such as tissue engineering, bioimaging techniques, stem cell research, and cancer therapies. However, in vivo applications of these nanocarriers should be further validated before clinical translation. To this end, this review begins with a brief introduction of MSNs properties, targeted drug delivery, and controlled release with a particular emphasis on their most recent diagnostic and therapeutic applications.
Collapse
Grants
- MOST 108-2320-B-010 -019 -MY3; MOST 109-2327-B-010-007 Ministry of Science and Technology
- MOHW108-TDU-B-211-133001, MOHW109-TDU-B-211-114001 Ministry of Health and Welfare
- VN109-16 VGH, NTUH Joint Research Program
- VTA107-V1-5-1, VTA108-V1-5-3, VTA109-V1-4-1 VGH, TSGH, NDMC, AS Joint Research Program
- IBMS-CRC109-P04 AS Clinical Research Center
- the "Cancer Progression Research Center, National Yang-Ming University" from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan the "Cancer Progression Research Center, National Yang-Ming University" from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan
- and the Ministry of Education through the SPROUT Project- Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B) of National Chiao Tung University and, Taiwan. and the Ministry of Education through the SPROUT Project- Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B) of National Chiao Tung University and, Taiwan.
Collapse
Affiliation(s)
- Elham Rastegari
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Yun-Hsien Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Tien-Chun Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Shih-Jen Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Pin-I Huang
- Department of Oncology, Taipei Veterans General Hospital, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chung-Yuan Mou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| |
Collapse
|
48
|
Ying K, Bai B, Gao X, Xu Y, Wang H, Xie B. Orally Administrable Therapeutic Nanoparticles for the Treatment of Colorectal Cancer. Front Bioeng Biotechnol 2021; 9:670124. [PMID: 34307319 PMCID: PMC8293278 DOI: 10.3389/fbioe.2021.670124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/14/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common and lethal human malignancies worldwide; however, the therapeutic outcomes in the clinic still are unsatisfactory due to the lack of effective and safe therapeutic regimens. Orally administrable and CRC-targetable drug delivery is an attractive approach for CRC therapy as it improves the efficacy by local drug delivery and reduces systemic toxicity. Currently, chemotherapy remains the mainstay modality for CRC therapy; however, most of chemo drugs have low water solubility and are unstable in the gastrointestinal tract (GIT), poor intestinal permeability, and are susceptible to P-glycoprotein (P-gp) efflux, resulting in limited therapeutic outcomes. Orally administrable nanoformulations hold the great potential for improving the bioavailability of poorly permeable and poorly soluble therapeutics, but there are still limitations associated with these regimes. This review focuses on the barriers for oral drug delivery and various oral therapeutic nanoparticles for the management of CRC.
Collapse
Affiliation(s)
- Kangkang Ying
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC), Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingjun Bai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xing Gao
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuzi Xu
- Department of Oral Implantology and Prosthodontics, The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Hangxiang Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC), Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
| | - Binbin Xie
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission (NHC), Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
49
|
Trzeciak K, Chotera-Ouda A, Bak-Sypien II, Potrzebowski MJ. Mesoporous Silica Particles as Drug Delivery Systems-The State of the Art in Loading Methods and the Recent Progress in Analytical Techniques for Monitoring These Processes. Pharmaceutics 2021; 13:pharmaceutics13070950. [PMID: 34202794 PMCID: PMC8309060 DOI: 10.3390/pharmaceutics13070950] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
Conventional administration of drugs is limited by poor water solubility, low permeability, and mediocre targeting. Safe and effective delivery of drugs and therapeutic agents remains a challenge, especially for complex therapies, such as cancer treatment, pain management, heart failure medication, among several others. Thus, delivery systems designed to improve the pharmacokinetics of loaded molecules, and allowing controlled release and target specific delivery, have received considerable attention in recent years. The last two decades have seen a growing interest among scientists and the pharmaceutical industry in mesoporous silica nanoparticles (MSNs) as drug delivery systems (DDS). This interest is due to the unique physicochemical properties, including high loading capacity, excellent biocompatibility, and easy functionalization. In this review, we discuss the current state of the art related to the preparation of drug-loaded MSNs and their analysis, focusing on the newest advancements, and highlighting the advantages and disadvantages of different methods. Finally, we provide a concise outlook for the remaining challenges in the field.
Collapse
|
50
|
Yan J, Gao T, Lu Z, Yin J, Zhang Y, Pei R. Aptamer-Targeted Photodynamic Platforms for Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:27749-27773. [PMID: 34110790 DOI: 10.1021/acsami.1c06818] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Achieving controlled and accurate delivery of photosensitizers (PSs) into tumor sites is a major challenge in conventional photodynamic therapy (PDT). Aptamer is a short oligonucleotide sequence (DNA or RNA) with a folded three-dimensional structure, which can selectively bind to specific small molecules, proteins, or the whole cells. Aptamers could act as ligands and be modified onto PSs or nanocarriers, enabling specific recognition and binding to tumor cells or their membrane proteins. The resultant aptamer-modified PSs or PSs-containing nanocarriers generate amounts of reactive oxygen species with light irradiation and obtain superior photodynamic therapeutic efficiency in tumors. Herein, we overview the recent progress in the designs and applications of aptamer-targeted photodynamic platforms for tumor therapy. First, we focus on the progress on the rational selection of aptamers and summarize the applications of aptamers which have been applied for targeted tumor diagnosis and therapy. Then, aptamer-targeted photodynamic therapies including various aptamer-PSs, aptamer-nanocarriers containing PSs, and aptamer-nano-photosensitizers are highlighted. The aptamer-targeted synergistically therapeutic platforms including PDT, photothermal therapy, and chemotherapy, as well as the imaging-guided theranostics, are also discussed. Finally, we offer an insight into the development trends and future perspectives of aptamer-targeted photodynamic platforms for tumor therapy.
Collapse
Affiliation(s)
- Jincong Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Tian Gao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Zhongzhong Lu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
| | - Ye Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| |
Collapse
|