1
|
Yao Y, Xu Z, Ding H, Yang S, Chen B, Zhou M, Zhu Y, Yang A, Yan X, Liang C, Kou X, Chen B, Huang W, Li Y. Carrier-free nanoparticles-new strategy of improving druggability of natural products. J Nanobiotechnology 2025; 23:108. [PMID: 39953594 PMCID: PMC11827262 DOI: 10.1186/s12951-025-03146-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
There are abundant natural products resources and extensive clinical use experience in China. However, the active components of natural products generally have problems such as poor water solubility and low bioavailability, which limit their druggability. Carrier-free nanoparticles, such as nanocrystals, self-assembled nanoparticles, and extracellular vesicles derived from both animal and plant sources, have great application potential in improving the safety and efficacy of drugs due to their simple and flexible preparation methods, high drug loading capacity and delivery efficiency, as well as long half-life in blood circulation. It has been widely used in biomedical fields such as anti-tumor, anti-bacterial, anti-inflammatory and anti-oxidation. Therefore, based on the natural products that have been used in clinic, this review focuses on the advantages of carrier-free nanoparticles in delivering active compounds, in order to improve the delivery process of natural products in vivo and improve their draggability.
Collapse
Affiliation(s)
- Yaqi Yao
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhenna Xu
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haoran Ding
- College of Pharmacy, Shandong Xiandai University, Jinan, 250104, China
| | - Shenshen Yang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bohan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mengjiao Zhou
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yehan Zhu
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Aihong Yang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xingxu Yan
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Chenrui Liang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaodi Kou
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bo Chen
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yubo Li
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
2
|
Han J, Shen Y, Cao R, Wang W, Duan J, Duan J, Bao C. Active herbal ingredients and drug delivery design for tumor therapy: a review. Chin J Nat Med 2024; 22:1134-1162. [PMID: 39725513 DOI: 10.1016/s1875-5364(24)60741-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Indexed: 12/28/2024]
Abstract
Active herbal ingredients are gaining recognition for their potent anti-tumor efficacy, attributable to various mechanisms including tumor cell inhibition, immune system activation, and tumor angiogenesis inhibition. Recent studies have revealed that numerous anti-tumor herbal ingredients, such as ginsenosides, ursolic acid, oleanolic acid, and Angelica sinensis polysaccharides, can be utilized to develop smart drug carriers like liposomes, micelles, and nanoparticles. These carriers can deliver active herbal ingredients and co-deliver anti-tumor drugs to enhance drug accumulation at tumor sites, thereby improving anti-tumor efficacy. This study provides a comprehensive analysis of the mechanisms by which these active herbal ingredients-derived carriers enhance therapeutic outcomes. Additionally, it highlights the structural properties of these active herbal ingredients, demonstrating how their unique features can be strategically employed to design smart drug carriers with improved anti-tumor efficacy. The insights presented aim to serve as a reference and guide future innovations in the design and application of smart drug carriers for cancer therapy that leverage active herbal ingredients.
Collapse
Affiliation(s)
- Jing Han
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanxi Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruiying Cao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiren Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jialun Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunjie Bao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Wang Q, Zhang C, Zhao Y, Jin Y, Zhou S, Qin J, Zhang W, Hu Y, Chen X, Yang K. Polyprodrug nanomedicine for chemiexcitation-triggered self-augmented cancer chemotherapy and gas therapy. Biomaterials 2024; 309:122606. [PMID: 38776593 DOI: 10.1016/j.biomaterials.2024.122606] [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: 10/31/2023] [Revised: 05/01/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Carbon monoxide (CO) has emerged as a potential antitumor agent by inducing the dysfunction of mitochondria and the apoptosis of cancer cells. However, it remains challenging to deliver appropriate amount of CO into tumor to ensure efficient tumor growth suppression with minimum side effects. Herein we developed a CO prodrug-loaded nanomedicine based on the self-assembly of camptothecin (CPT) polyprodrug amphiphiles. The polyprodrug nanoparticles readily dissociate upon exposure to endogenous H2O2 in the tumor, resulting in rapid release of CPT and generation of high-energy intermediate dioxetanedione. The latter can transfer the energy to neighboring CO prodrugs to activate CO production by chemiexcitation, while CPT promotes the generation of H2O2 in tumors, which in turn facilitates cascade CPT and CO release. As a result, the polyprodrug nanoparticles display remarkable tumor suppression in both subcutaneous and orthotopic breast tumor-bearing mice owing to the self-augmented CPT release and CO generation. In addition, no obvious systemic toxicity was observed in mice treated with the metal-free CO prodrug-loaded nanomedicine, suggesting the good biocompatibility of the polyprodrug nanoparticles. Our work provides new insights into the design and construction of polyprodrug nanomedicines for synergistic chemo/gas therapy.
Collapse
Affiliation(s)
- Qingfu Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Chen Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Ya Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Yifan Jin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Shen Zhou
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Junde Qin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Wenxin Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, 119074, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore,117597, Singapore; Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Kuikun Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150080, PR China.
| |
Collapse
|
4
|
Cao J, Hong K, Lv C, Jiang W, Chen Q, Wang R, Wang Y. Reduction-sensitive polymeric carrier for the targeted delivery of a quinazoline derivative for enhanced generation of reactive oxygen species against cancer. Biomater Sci 2024; 12:2626-2638. [PMID: 38526801 DOI: 10.1039/d3bm02136j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors and the development of effective therapeutics against HCC is urgently needed. A novel quinazoline derivative 04NB-03 (Qd04) has been proved to be highly effective against HCC without obvious toxic side-effects. However, the poor water solubility and low bioavailability in vivo severely limit its clinical application. In addition, Qd04 kills tumor cells by inducing an accumulation of endogenous reactive oxygen species (ROS), which is highly impeded by the overexpression of glutathione (GSH) in tumor cells. Herein, we designed a disulfide cross-linked polyamino acid micelle to deliver Qd04 for HCC therapy. The disulfide linkage not only endowed a tumor-targeted delivery of Qd04 by responding to tumor cell GSH but also depleted GSH to achieve increased levels of ROS generation, which improved the therapeutic efficiency of Qd04. Both in vitro and in vivo results demonstrated that the synthesized nanodrug exerted good anti-hepatoma effects, which provided a potential application for HCC therapy in clinics.
Collapse
Affiliation(s)
- Jianrong Cao
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Keze Hong
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Chengqi Lv
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Weiting Jiang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Qi Chen
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Rongze Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
5
|
Li Q, Lianghao Y, Shijie G, Zhiyi W, Yuanting T, Cong C, Chun-Qin Z, Xianjun F. Self-assembled nanodrug delivery systems for anti-cancer drugs from traditional Chinese medicine. Biomater Sci 2024; 12:1662-1692. [PMID: 38411151 DOI: 10.1039/d3bm01451g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Traditional Chinese medicine (TCM) is a combination of raw herbs and herbal extracts with a plethora of documented beneficial bioactivities, which has unique advantages in anti-tumor therapy, and many of its major bioactive molecules have been identified in recent years due to advances in chemical separation and structural analysis. However, the major chemical classes of plant-derived bioactive compounds frequently possess chemical properties, including poor water solubility, stability, and bioavailability, that limit their therapeutic application. Alternatively, natural small molecules (NSMs) containing these components possess modifiable groups, multiple action sites, hydrophobic side chains, and a rigid skeleton with self-assembly properties that can be exploited to construct self-assembled nanoparticles with therapeutic effects superior to their individual constituents. For instance, the construction of a self-assembled nanodrug delivery system can effectively overcome the strong hydrophobicity and poor in vivo stability of NSMs, thereby greatly improving their bioavailability and enhancing their anti-tumor efficacy. This review summarizes the self-assembly methods, mechanisms, and applications of a variety of NSMs, including terpenoids, flavonoids, alkaloids, polyphenols, and saponins, providing a theoretical basis for the subsequent research on NSMs and the development of SANDDS.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Yuan Lianghao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Gao Shijie
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Wang Zhiyi
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Tang Yuanting
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Chen Cong
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China.
| | - Zhao Chun-Qin
- Academy of Chinese Medicine Literature and Culture, Key Laboratory of Classical Theory of Traditional Chinese Medicine, Ministry of Education, Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Fu Xianjun
- Marine Traditional Chinese Medicine Research Centre, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, P. R. China.
| |
Collapse
|
6
|
Li DZ, Fu ZD, Liu HY, Pan XD. Facile synthesis and cytotoxicity of substituted uracil-1'( N)-acetic acid and 4-pyridone-1'( N)-acetic acid esters of 20(S)-camptothecins. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:259-268. [PMID: 38347748 DOI: 10.1080/10286020.2023.2300374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/25/2023] [Indexed: 04/08/2024]
Abstract
A series of novel substituted uracil-1'(N)-acetic acid esters (5-9) and 4-pyridone-1'(N)-acetic acid esters (10-11) of 20(S)-camptothecins (CPTs) have been synthesized by the acylation method. All of these new esters were assayed for in vitro cytotoxicity against five human cancer cell lines A549, Bel7402, BGC-823, HCT-8 and A2780. The in vitro bioassay results showed that all the synthesized compounds 5-11 had cytotoxities that were higher than TPT and comparable to CPT on these five tumor cell lines, some of them even showed comparable or superior cytotoxic activity to CPT. The in vitro data exhibited the cytotoxicity of the ester depended on that of its parent compound. The ester 5, 6, 8, 10, 11 even possessed the cytotoxity activity comparable to or even a little better than CPT on A549, HCT-8 and A2780. The compound 11 had the same level of cytoxity on Bel7402 as that of CPT. Here the synthesis and the in vitro antitumor evaluation of a series of novel 20-O-linked substituted uracil-1'(N)-acetic acid and 4-pyridone-1'(N)-acetic acid esters derivatives of CPTs are reported.
Collapse
Affiliation(s)
- Di-Zao Li
- School of Traditional Chinese Medicine, Beijing Key Laboratory of TCM Syndrome and Formula, and Key Laboratory of TCM Syndrome and Formula, Ministry of Education, Beijing University of Chinese Medicine, Beijing 100029, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhao-Di Fu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hong-Yan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xian-Dao Pan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
7
|
Das P, N M, Singh N, Datta P. Supramolecular Nanostructures for the Delivery of Peptides in Cancer Therapy. J Pharmacol Exp Ther 2024; 388:67-80. [PMID: 37827700 DOI: 10.1124/jpet.123.001698] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Supramolecular nanostructured based delivery systems are emerging as a meaningful approach in the treatment of cancer, offering controlled drug release and improved therapeutic efficacy. The self-assembled structures can be small molecules, polymers, peptides, or proteins, which can be used and functionalized to achieve tailored release and target specific cells, tissues, or organs. These structures can improve the solubility and stability of drugs having low aqueous solubility by encapsulating and protecting them from degradation. Alongside, peptides as natural biomolecules have gained increasing attention as potential candidates in cancer treatment because of their biocompatibility, low cytotoxicity, and high specificity toward tumor cells. The amino acid sequences in peptide molecules are tunable, efficiently controlling the morphology of peptide-based self-assembled nanosystems and offering flexibility to form supramolecular nanostructures (SNs). It is evident from the current literature that the supramolecular nanostructures based delivery of peptide for cancer treatment hold great promise for future cancer therapy, offering potential strategies for personalized medicine with improved patient outcomes. SIGNIFICANCE STATEMENT: This review focuses on fundamentals and various drug delivery mechanisms based on SNs. Different SN approaches and recent literature reviews on peptide delivery are also presented to the readers.
Collapse
Affiliation(s)
- Priyanka Das
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Manasa N
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Nidhi Singh
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Pallab Datta
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| |
Collapse
|
8
|
Wang X, Zhuang Y, Wang Y, Jiang M, Yao L. The recent developments of camptothecin and its derivatives as potential anti-tumor agents. Eur J Med Chem 2023; 260:115710. [PMID: 37595544 DOI: 10.1016/j.ejmech.2023.115710] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/07/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
This review article focuses on the research progress made in the structural modifications of camptothecin (CPT), a potent cytotoxic natural alkaloid. CPT possesses a unique 5-fused ring structure and exhibits various beneficial activities such as anti-proliferative, anti-fungal, insecticidal, and anti-SARS-CoV-2 properties. CPT and its analogs, including Topotecan and Irinotecan, have been successfully developed and marketed as topoisomerase I inhibitors. To enhance the therapeutic potential of CPT, researchers have undertaken structural modifications primarily on the A, B, and E rings of the CPT core structure. These modifications aim to improve the efficacy, selectivity, and pharmacokinetic properties of CPT derivatives. The article reviews the advancements in hybridizing CPT with other bioactive compounds, the synthesis of novel CPT analogs, and their associated biological activities. Moreover, the structure-activity relationship (SAR) of these modified CPT derivatives is summarized to gain insights into their structure-function correlations. In addition to discussing the modifications and biological activities of CPT derivatives, the article also touches upon the mechanism of parent drug release. Many CPT derivatives are prodrugs, meaning they require metabolic activation to generate the active form of the drug. It is a resource for researchers interested in developing novel anti-tumor agents based on CPT, addressing the limitations associated with the parent drug, and exploring various aspects of CPT modifications.
Collapse
Affiliation(s)
- Xianzhang Wang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Yumeng Zhuang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Yuankun Wang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Maokai Jiang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Lei Yao
- School of Pharmacy, Yantai University, Yantai, 264005, China.
| |
Collapse
|
9
|
Laskar P, Dhasmana A, Kotnala S, Jaggi M, Yallapu MM, Chauhan SC. Glutathione-Responsive Tannic Acid-Assisted FRET Nanomedicine for Cancer Therapy. Pharmaceutics 2023; 15:1326. [PMID: 37242568 PMCID: PMC10222396 DOI: 10.3390/pharmaceutics15051326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/28/2023] Open
Abstract
In cancer combination therapy, a multimodal delivery vector is used to improve the bioavailability of multiple anti-cancer hydrophobic drugs. Further, targeted delivery of therapeutics along with simultaneous monitoring of the drug release at the tumor site without normal organ toxicity is an emerging and effective strategy for cancer treatment. However, the lack of a smart nano-delivery system limits the application of this therapeutic strategy. To overcome this issue, a PEGylated dual drug, conjugated amphiphilic polymer (CPT-S-S-PEG-CUR), has been successfully synthesized by conjugating two hydrophobic fluorescent anti-cancer drugs, curcumin (CUR) and camptothecin (CPT), through an ester and a redox-sensitive disulfide (-S-S-) linkage, respectively, with a PEG chain via in situ two-step reactions. CPT-S-S-PEG-CUR is spontaneously self-assembled in the presence of tannic acid (TA, a physical crosslinker) into anionic, comparatively smaller-sized (~100 nm), stable nano-assemblies in water in comparison to only polymer due to stronger H-bond formation between polymer and TA. Further, due to the spectral overlap between CPT and CUR and a stable, smaller nano-assembly formation by the pro-drug polymer in water in presence of TA, a successful Fluorescence Resonance Energy Transfer (FRET) signal was generated between the conjugated CPT (FRET donor) and conjugated CUR (FRET acceptor). Interestingly, these stable nano-assemblies showed a preferential breakdown and release of CPT in a tumor-relevant redox environment (in the presence of 50 mM glutathione), leading to the disappearance of the FRET signal. These nano-assemblies exhibited a successful cellular uptake by the cancer cells and an enhanced antiproliferative effect in comparison to the individual drugs in cancer cells (AsPC1 and SW480). Such promising in vitro results with a novel redox-responsive, dual-drug conjugated, FRET pair-based nanosized multimodal delivery vector can be highly useful as an advanced theranostic system towards effective cancer treatment.
Collapse
Affiliation(s)
- Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- The Ångström Laboratory, Macromolecular Chemistry, Department of Chemistry, Uppsala University, 751 21 Uppsala, Sweden
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- Cancer Research Institute, Himalayan School of Biosciences, Swami Rama Himalayan University, Dehradun 248016, India
| | - Sudhir Kotnala
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
10
|
Liu Y, Wu Y, Luo Z, Li M. Designing supramolecular self-assembly nanomaterials as stimuli-responsive drug delivery platforms for cancer therapy. iScience 2023; 26:106279. [PMID: 36936787 PMCID: PMC10014307 DOI: 10.1016/j.isci.2023.106279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Stimuli-responsive nanomaterials have attracted substantial interest in cancer therapy, as they hold promise to deliver anticancer agents to tumor sites in a precise and on-demand manner. Interestingly, supramolecular chemistry is a burgeoning discipline that entails the reversible bonding between components at the molecular and nanoscale levels, and the recent advances in this area offer the possibility to design nanotherapeutics with improved controllability and functionality for cancer therapy. Herein, we provide a comprehensive summary of typical non-covalent interaction modes, which primarily include hydrophobic interaction, hydrogel bonding, host-guest interaction, π-π stacking, and electrostatic interaction. Special emphasis is placed on the implications of these interaction modes to design novel stimuli-responsive drug delivery principles and concepts, aiming to enhance the spatial, temporal, and dosage precision of drug delivery to cancer cells. Finally, future perspectives are discussed to highlight current challenges and future opportunities in self-assembly-based stimuli-responsive drug delivery nanotechnologies for cancer therapy.
Collapse
Affiliation(s)
- Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yunyun Wu
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing 400042, China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
11
|
Linear-like polypeptide-based micelle with pH-sensitive detachable PEG to deliver dimeric camptothecin for cancer therapy. Asian J Pharm Sci 2023; 18:100773. [PMID: 36711109 PMCID: PMC9871073 DOI: 10.1016/j.ajps.2022.100773] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 11/23/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Nano drug delivery systems have made significant progress in delivering anticancer drugs camptothecin (CPT). However, many challenges for CPT delivery remain, including low drug loading efficiency, premature drug leakage, and poor cellular internalization. Herein, we report a novel dual-sensitive polypeptide-based micelle with remarkably high drug loading of CPT for cancer therapy. This self-assembled micelle possesses the following essential components for CPT: (1) pH-sensitive PEG (OHC-PEG-CHO) for prolonging blood circulation and allowing biocompatibility by shielding the cationic micelles, which can be detached under the tumor acidic microenvironment and facilitates the cellular uptake; (2) polypeptide polylysine-polyphenylalanine (PKF) synthesized via ring-opening polymerization for micelle formation and CPT analogue loading; (3) dimeric CPT (DCPT) with redox-sensitive linker for increasing CPT loading and ensuring drug release at tumor sites. Interestingly, the linear-like morphology of PEG-PKF/DCPT micelles was able to enhance their cellular internalization when compared with the spherical blank PKF micelles. Also, the anticancer efficacy of DCPT against lung cancer cells was significantly improved by the micelle formation. In conclusion, this work provides a promising strategy facilitating the safety and effective application of CPT in cancer therapy.
Collapse
|
12
|
Gao T, Liu H, Xie Z, Zheng M. Biomimetic nanoprodrugs from fatty acid modified camptothecin and albumin for enhanced pharmacotherapy. J Colloid Interface Sci 2023; 630:385-394. [DOI: 10.1016/j.jcis.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/20/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2022]
|
13
|
Misra S, Singh P, Singh AK, Roy L, Kuila S, Dey S, Mahapatra AK, Nanda J. Tuning of the Supramolecular Helicity of Peptide-Based Gel Nanofibers. J Phys Chem B 2022; 126:10882-10892. [PMID: 36516185 DOI: 10.1021/acs.jpcb.2c06897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Helical supramolecular architectures play important structural and functional roles in biological systems. The helicity of synthetic molecules can be tuned mainly by the chiral manipulation of the system. However, tuning of helicity by the achiral unit of the molecules is less studied. In this work, the helicity of naphthalimide-capped peptide-based gel nanofibers is tuned by the alteration of methylene units present in the achiral amino acid. The inversion of supramolecular helicity has been extensively studied by CD spectroscopy and morphological analysis. The density functional theory (DFT) study indicates that methylene spacers influence the orientation of π-π stacking interactions of naphthalimide units in the self-assembled structure that regulates the helicity. This work illustrates a new approach to tuning the supramolecular chirality of self-assembled biomaterials.
Collapse
Affiliation(s)
- Souvik Misra
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri 734013, West Bengal, India.,Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, P.O. Botanic Garden, Howrah 711103, West Bengal, India
| | - Pijush Singh
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India
| | - Ajeet Kumar Singh
- Institute of Chemical Technology Mumbai - IOC Odisha Campus Bhubaneswar, IIT Kharagpur Extension Centre, Bhubaneswar 751013, India
| | - Lisa Roy
- Institute of Chemical Technology Mumbai - IOC Odisha Campus Bhubaneswar, IIT Kharagpur Extension Centre, Bhubaneswar 751013, India
| | - Soumen Kuila
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri 734013, West Bengal, India
| | - Sukantha Dey
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri 734013, West Bengal, India
| | - Ajit K Mahapatra
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, P.O. Botanic Garden, Howrah 711103, West Bengal, India
| | - Jayanta Nanda
- Department of Chemistry, University of North Bengal, Raja Rammohanpur, Siliguri 734013, West Bengal, India
| |
Collapse
|
14
|
Tailoring carrier-free nanocombo of small-molecule prodrug for combinational cancer therapy. J Control Release 2022; 352:256-275. [PMID: 36272660 DOI: 10.1016/j.jconrel.2022.10.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
The outcomes of monotherapy could not satisfy clinical cancer treatment owing to the challenges of tumor heterogeneity, multi-drug resistance, tumor metastasis and relapse. In response, the significance of combinational cancer therapy has been highlighted. Traditional combinational schemes usually utilize "free" drug for multi drug administration, independently. The diverse pharmacokinetics and biodistribution greatly hinder the antitumor effects and cause systematic toxicity. To tackle the hinderance, various nanoparticulate drug delivery systems (Nano-DDSs) have been developed. However, conventional Nano-DDSs encapsulate drugs into carrier materials through noncovalent interactions, resulting in low drug loading, fixed multi drug encapsulation ratio, chemical instability and carrier-associated toxicity. Recently, carrier-free nanocombos based on self-assembling small-molecule prodrugs (SPNCs) have emerged as a versatile Nano-DDSs for multiple drug delivery. Benefited by the self-assembly capability, SPNCs could be facilely fabricated with distinct merits of ultra-high drug loading, adjustable drug ratio and negligible carrier-associated toxicity. Herein, we summarize the latest trends of SPNCs. First, a basic review on self-assembling small-molecule prodrugs is presented. Additionally, facile techniques to prepare SPNCs are introduced. Furthermore, advanced combinational therapies based on SPNCs are spotlighted with special emphasis on synergistic mechanisms. Finally, future prospects and challenges are discussed.
Collapse
|
15
|
Zhou H, Wang Y, Hou Y, Zhang Z, Wang Q, Tian X, Lu H. Co‐delivery of Cisplatin and Chlorin e6 by Poly(phosphotyrosine) for Synergistic Chemotherapy and Photodynamic Therapy. CHINESE J CHEM 2022. [DOI: 10.1002/cjoc.202200334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Haisen Zhou
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| | - Yaoyi Wang
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| | - Yingqin Hou
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| | - Zhengkui Zhang
- Department of General Surgery Peking University First Hospital Beijing 100034 People's Republic of China
| | - Qi Wang
- Department of General Surgery Peking University First Hospital Beijing 100034 People's Republic of China
| | - Xiaodong Tian
- Department of General Surgery Peking University First Hospital Beijing 100034 People's Republic of China
| | - Hua Lu
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| |
Collapse
|
16
|
Nano-bio interactions: A major principle in the dynamic biological processes of nano-assemblies. Adv Drug Deliv Rev 2022; 186:114318. [PMID: 35533787 DOI: 10.1016/j.addr.2022.114318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/12/2022] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Controllable nano-assembly with stimuli-responsive groups is emerging as a powerful strategy to generate theranostic nanosystems that meet unique requirements in modern medicine. However, this prospective field is still in a proof-of-concept stage due to the gaps in our understanding of complex-(nano-assemblies)-complex-(biosystems) interactions. Indeed, stimuli-responsive assembly-disassembly is, in and of itself, a process of nano-bio interactions, the key steps for biological fate and functional activity of nano-assemblies. To provide a comprehensive understanding of these interactions in this review, we first propose a 4W1H principle (Where, When, What, Which and How) to delineate the relevant dynamic biological processes, behaviour and fate of nano-assemblies. We further summarize several key parameters that govern effective nano-bio interactions. The effects of these kinetic parameters on ADMET processes (absorption, distribution, metabolism, excretion and transformation) are then discussed. Furthermore, we provide an overview of the challenges facing the evaluation of nano-bio interactions of assembled nanodrugs. We finally conclude with future perspectives on safe-by-design and application-driven-design of nano-assemblies. This review will highlight the dynamic biological and physicochemical parameters of nano-bio interactions and bridge discrete concepts to build a full spectrum understanding of the biological outcomes of nano-assemblies. These principles are expected to pave the way for future development and clinical translation of precise, safe and effective nanomedicines with intelligent theranostic features.
Collapse
|
17
|
Li G, Sun B, Li Y, Luo C, He Z, Sun J. Small-Molecule Prodrug Nanoassemblies: An Emerging Nanoplatform for Anticancer Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101460. [PMID: 34342126 DOI: 10.1002/smll.202101460] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/21/2021] [Indexed: 06/13/2023]
Abstract
The antitumor efficiency and clinical translation of traditional nanomedicines is mainly restricted by low drug loading, complex preparation technology, and potential toxicity caused by the overused carrier materials. In recent decades, small-molecule prodrug nanoassemblies (SMP-NAs), which are formed by the self-assembly of prodrugs themselves, have been widely investigated with distinct advantages of ultrahigh drug-loading and negligible excipients-trigged adverse reaction. Benefited from the simple preparation process, SMP-NAs are widely used for chemotherapy, phototherapy, immunotherapy, and tumor diagnosis. In addition, combination therapy based on the accurate co-delivery behavior of SMP-NAs can effectively address the challenges of tumor heterogeneity and multidrug resistance. Recent trends in SMP-NAs are outlined, and the corresponding self-assembly mechanisms are discussed in detail. Besides, the smart stimuli-responsive SMP-NAs and the combination therapy based on SMP-NAs are summarized, with special emphasis on the structure-function relationships. Finally, the outlooks and potential challenges of SMP-NAs in cancer therapy are highlighted.
Collapse
Affiliation(s)
- Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yaqiao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
18
|
Constructing nanocomplexes by multicomponent self-assembly for curing orthotopic glioblastoma with synergistic chemo-photothermal therapy. Biomaterials 2021; 279:121193. [PMID: 34700227 DOI: 10.1016/j.biomaterials.2021.121193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/18/2022]
Abstract
The blood-brain barrier (BBB) is one of the major limitations of glioblastoma therapy in the clinic. Nanodrugs have shown great potential for glioblastoma therapy. Herein, we purposefully developed a multicomponent self-assembly nanocomplex with very high drug loading content for curing orthotopic glioblastoma with synergistic chemo-photothermal therapy. The nanocomplex consisted of self-assembled pH-responsive nanodrugs derived from amino acid-conjugated camptothecin (CPT) and canine dyes (IR783) coated with peptide Angiopep-2-conjugated copolymer of Ang-PEG-g-PLL. Specifically, the carrier-free nanocomplex exhibited a high drug loading content (up to 62%), good biocompatibility, and effective glioma accumulation ability. Moreover, the nanocomplex displayed good stability and pH-responsive behavior ex vivo. Both in vitro and in vivo results revealed that the nanocomplex could effectively cross the BBB and target glioma cells. Furthermore, the combination of chemotherapy and photothermal therapy of the nanocomplex achieved a better therapeutic effect, longer survival time, and minimized toxic side effects in orthotopic glioblastoma tumor-bearing nude mice. Overall, we modified the chemotherapeutic drug CPT so that it could self-assemble with other molecules into nanoparticles, which providing an alternative for the preparation of the carrier-free nanodrugs. The results highlighted the potential of self-assembly nanodrugs as a novel platform for effective glioblastoma therapy.
Collapse
|
19
|
Concurrent impairment of nucleus and mitochondria for synergistic inhibition of cancer metastasis. Int J Pharm 2021; 608:121077. [PMID: 34487811 DOI: 10.1016/j.ijpharm.2021.121077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022]
Abstract
Cancer metastasis, which increases the mortality in a short period of time, has been considered as the main challenge in tumor treatment. However, tumor growth suppression also should not be ignored in cancer metastasis treatment. Recently, accumulating evidences have suggested that mitochondria play an important role in mitigating caner metastasis. Nucleus, as the repository of genetic information, plays a key role in cell proliferation. However, it remains elusive that the concurrent impairment of nucleus and mitochondria may achieve better anti-tumor and anti-metastatic effects. Here, we designed a mitochondria-penetrating peptide modified doxorubicin (MPP-Dox) loaded N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer conjugates (PM), as well as a nuclear accumulating HPMA copolymer Dox conjugates (PN) by the nuclear tendency of Dox. After co-delivering the two copolymers (abbreviation for PMN), PM promoted cell apoptosis and inhibited tumor metastasis by damaging mitochondria, whereas PN suppressed cell proliferation and promoted apoptosis by destroying nucleus. Importantly, PM and PN complemented each other as expected. The mitochondrial dysfunction and tumor metastasis inhibition of PM was improved by PN, while cell proliferation suppression and apoptosis by nucleus destroying of PN was enhanced by PM. As a result, tumor growth of breast cancer 4T1 cells in vivo was significantly restrained and lung metastasis was potently decreased and almost eradicated, fully reflecting the advantages of organelle targeting combination therapy. As a consequence, our work showed that concurrent impairment of nucleus and mitochondria was feasible and beneficial to metastatic cancer treatment.
Collapse
|
20
|
Ren H, Wu L, Tan L, Bao Y, Ma Y, Jin Y, Zou Q. Self-assembly of amino acids toward functional biomaterials. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:1140-1150. [PMID: 34760429 PMCID: PMC8551877 DOI: 10.3762/bjnano.12.85] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/30/2021] [Indexed: 06/13/2023]
Abstract
Biomolecules, such as proteins and peptides, can be self-assembled. They are widely distributed, easy to obtain, and biocompatible. However, the self-assembly of proteins and peptides has disadvantages, such as difficulty in obtaining high quantities of materials, high cost, polydispersity, and purification limitations. The difficulties in using proteins and peptides as functional materials make it more complicate to arrange assembled nanostructures at both microscopic and macroscopic scales. Amino acids, as the smallest constituent of proteins and the smallest constituent in the bottom-up approach, are the smallest building blocks that can be self-assembled. The self-assembly of single amino acids has the advantages of low synthesis cost, simple modeling, excellent biocompatibility and biodegradability in vivo. In addition, amino acids can be assembled with other components to meet multiple scientific needs. However, using these simple building blocks to design attractive materials remains a challenge due to the simplicity of the amino acids. Most of the review articles about self-assembly focus on large molecules, such as peptides and proteins. The preparation of complicated materials by self-assembly of amino acids has not yet been evaluated. Therefore, it is of great significance to systematically summarize the literature of amino acid self-assembly. This article reviews the recent advances in amino acid self-assembly regarding amino acid self-assembly, functional amino acid self-assembly, amino acid coordination self-assembly, and amino acid regulatory functional molecule self-assembly.
Collapse
Affiliation(s)
- Huan Ren
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Lifang Wu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Lina Tan
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yanni Bao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuchen Ma
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yong Jin
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qianli Zou
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
21
|
Nguyen A, Böttger R, Li SD. Recent trends in bioresponsive linker technologies of Prodrug-Based Self-Assembling nanomaterials. Biomaterials 2021; 275:120955. [PMID: 34130143 DOI: 10.1016/j.biomaterials.2021.120955] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Prodrugs are designed to improve pharmaceutical properties of potent compounds and represent a central approach in drug development. The success of the prodrug strategy relies on incorporation of a reversible linkage facilitating controlled release of the parent drug. While prodrug approaches enhance pharmacokinetic properties over their parent drug, they still face challenges in absorption, distribution, metabolism, elimination, and toxicity (ADMET). Conjugating a drug to a carrier molecule such as a polymer can create an amphiphile that self-assembles into nanoparticles. These nanoparticles display prolonged blood circulation and passive targeting ability. Furthermore, the drug release can be tailored using a variety of linkers between the parent drug and the carrier molecule. In this review, we introduce the concept of self-assembling prodrugs and summarize different approaches for controlling the drug release with a focus on the linker technology. We also summarize recent clinical trials, discuss the emerging challenges, and provide our perspective on the utility and future potential of this technology.
Collapse
Affiliation(s)
- Anne Nguyen
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
22
|
Guo Z, Hu Y, Zhao M, Hao K, He P, Tian H, Chen X, Chen M. Prodrug-Based Versatile Nanomedicine with Simultaneous Physical and Physiological Tumor Penetration for Enhanced Cancer Chemo-Immunotherapy. NANO LETTERS 2021; 21:3721-3730. [PMID: 33891423 DOI: 10.1021/acs.nanolett.0c04772] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chemo-immunotherapy combination effect remains to be a great challenge due to the poor tumor penetration of therapeutic agents that resulted from condensed extracellular matrix (ECM), T cell-related immune escape, and thus the potential recurrence. Herein, a helix self-assembly camptothecin (CPT) prodrug with simultaneous physical and physiological tumor penetration was constructed to realize effective chemo-immunotherapy. Specifically, CPT was modified with arginine to self-assemble into nanofibers to physically improve tumor penetration. Two plasmids, pshPD-L1 and pSpam1 for expressing small hairpin RNA PD-L1 and hyaluronidase, respectively, were loaded to down-regulate tumor surface PD-L1 expression for converting anergic state of T cells into the tumor-reactive T cells and produce hyaluronidase to physiologically degrade ECM for further enhanced tumor penetration. Moreover, the degraded ECM could also increase immune cells' infiltration into tumor sites, which may exert a synergistic antitumor immunity combined with immune checkpoint inhibition. Such a nanomedicine could cause significant inhibition of primary, distant tumors, and effective prevention of tumor recurrence.
Collapse
Affiliation(s)
- Zhaopei Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yingying Hu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Mengyao Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Kai Hao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Pan He
- School of Materials Science and Engineering, Changchun University of Science and Technology, Changchun 130022, China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| |
Collapse
|
23
|
Gao F, Zhou J, Sun Y, Yang C, Zhang S, Wang R, Tan W. Programmable Repurposing of Existing Drugs as Pharmaceutical Elements for the Construction of Aptamer-Drug Conjugates. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9457-9463. [PMID: 33356116 DOI: 10.1021/acsami.0c18846] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Converting marketed drug molecules into phosphoramidites may present a potential strategy to facilitate the development of aptamer-drug conjugates (ApDCs) by a DNA synthesizer in a programmable way; however, quite limited methods were reported. Herein, we demonstrated a general approach by repurposing camptothecin (CPT) species. Commonly used inactive ingredients in pharmaceuticals are investigated and selected as a bonding moiety, from which 2-mercaptoethoxy ethanol and thioglycerol were efficiently incorporated with CPT to give the precursors. Cell viability and molecular docking results of the precursors supported that incorporation of the bonding moiety would not interrupt the inhibitory activity. Therefore, corresponding phosphoramidites were prepared as pharmaceutical elements, and a series of ApDCs were constructed automatically by solid-phase synthesis. Biological studies revealed that CPT elements could be specifically delivered to HCT116 cells by an aptamer and released inside cells. This kind of programmable repurposing may take advantage of established safety data and efficacy of existing drugs resulting in a faster development of ApDCs.
Collapse
Affiliation(s)
- Fei Gao
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Translation Medicine, Shanghai University, Shanghai 200444, China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cai Yang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiyan Zhang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruowen Wang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
24
|
Triple stimuli-responsive supramolecular nanoassembly with mitochondrial targetability for chemophotothermal therapy. J Control Release 2020; 327:35-49. [DOI: 10.1016/j.jconrel.2020.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/27/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022]
|
25
|
Karaosmanoglu S, Zhou M, Shi B, Zhang X, Williams GR, Chen X. Carrier-free nanodrugs for safe and effective cancer treatment. J Control Release 2020; 329:805-832. [PMID: 33045313 DOI: 10.1016/j.jconrel.2020.10.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
Clinical applications of many anti-cancer drugs are restricted due to their hydrophobic nature, requiring use of harmful organic solvents for administration, and poor selectivity and pharmacokinetics resulting in off-target toxicity and inefficient therapies. A wide variety of carrier-based nanoparticles have been developed to tackle these issues, but such strategies often fail to encapsulate drug efficiently and require significant amounts of inorganic and/or organic nanocarriers which may cause toxicity problems in the long term. Preparation of nano-formulations for the delivery of water insoluble drugs without using carriers is thus desired, requiring elegantly designed strategies for products with high quality, stability and performance. These strategies include simple self-assembly or involving chemical modifications via coupling drugs together or conjugating them with various functional molecules such as lipids, carbohydrates and photosensitizers. During nanodrugs synthesis, insertion of redox-responsive linkers and tumor targeting ligands endows them with additional characteristics like on-target delivery, and conjugation with immunotherapeutic reagents enhances immune response alongside therapeutic efficacy. This review aims to summarize the methods of making carrier-free nanodrugs from hydrophobic drug molecules, evaluating their performance, and discussing the advantages, challenges, and future development of these strategies.
Collapse
Affiliation(s)
- Sena Karaosmanoglu
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK
| | - Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Bingyang Shi
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xiujuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, PR China.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK.
| |
Collapse
|
26
|
Wang S, Zhang F, Yu G, Wang Z, Jacobson O, Ma Y, Tian R, Deng H, Yang W, Chen ZY, Chen X. Zwitterionic-to-cationic charge conversion polyprodrug nanomedicine for enhanced drug delivery. Am J Cancer Res 2020; 10:6629-6637. [PMID: 32550894 PMCID: PMC7295052 DOI: 10.7150/thno.47849] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/09/2020] [Indexed: 12/21/2022] Open
Abstract
Zwitterionic surface modification is a promising strategy for nanomedicines to achieve prolonged circulation time and thus effective tumor accumulation. However, zwitterion modified nanoparticles suffer from reduced cellular internalization efficiency. Methods: A polyprodrug-based nanomedicine with zwitterionic-to-cationic charge conversion ability (denoted as ZTC-NMs) was developed for enhanced chemotherapeutic drug delivery. The polyprodrug consists of pH-responsive poly(carboxybetaine)-like zwitterionic segment and glutathione-responsive camptothecin prodrug segment. Results: The ZTC-NMs combine the advantages of zwitterionic surface and polyprodrug. Compared with conventional zwitterionic surface, the ZTC-NMs can respond to tumor microenvironment and realize ZTC surface charge conversion, thus improve cellular internalization efficiency of the nanomedicines. Conclusions: This ZTC method offers a strategy to promote the drug delivery efficiency and therapeutic efficacy, which is promising for the development of cancer nanomedicines.
Collapse
|