1
|
Sang R, Nixdorf S, Hung T, Power C, Deng F, Bui TA, Engel A, Goldys EM, Deng W. Unlocking the in vivo therapeutic potential of radiation-activated photodynamic therapy for locally advanced rectal cancer with lymph node involvement. EBioMedicine 2025; 116:105724. [PMID: 40359628 DOI: 10.1016/j.ebiom.2025.105724] [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: 10/31/2024] [Revised: 03/14/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Rectal cancer is a leading cause of cancer-related mortality worldwide. The recurrence of locally advanced rectal cancer (LARC), particularly in cases involving lymph node-positive tumours, remains a critical challenge in rectal cancer management. In this study, a therapeutic strategy, radiation-activated photodynamic therapy (RA-PDT), for the treatment of LARC with lymph node-positive tumours was developed and evaluated. METHODS RA-PDT was achieved by using a lipid-polymer hybrid nanoplatform loaded with verteporfin (VP) and functionalised with folic acid (FA) as a targeting molecule. Upon receiving a single 4 Gy fraction of radiation, VP was effectively activated, generating sufficient reactive oxygen species (ROS) to induce cancer cell death-however surrounding tissue was less affected and was spared. The efficacy of this strategy was assessed through in vitro cytotoxicity studies in HCT116 cells, as well as in orthotopic and subcutaneous mouse models. In vivo lymph node tumour progression was also evaluated. FINDINGS RA-PDT effectively generated ROS following 4 Gy irradiation and exhibited significant cytotoxicity in HCT116 cells. In vivo, this strategy largely inhibited primary tumour growth in both orthotopic and subcutaneous mouse models while also suppressing lymph node tumour progression. Surrounding tissues were minimally affected, highlighting the precision and safety of this approach. INTERPRETATION RA-PDT demonstrates potential as a safe therapeutic strategy for LARC, paving the way for its clinical translation. FUNDING This study was supported by the Australian National Health and Medical Research Council (GNT1181889), fellowship award (2019/CDF1013) from Cancer Institute NSW, Australia, the Australian Research Council Centre of Excellence for Nanoscale Biophotonics (CE140100003), UNSW SHARP funding, project grant from National Foundation for Medical Research and Innovation, Australia, International Research Training Program Scholarship (IRTP) from Australian Government, PhD Research Scholar Award from Sydney Vital Translational Cancer Research, and Translational Cancer Research Network PhD Scholarship Top-up award.
Collapse
Affiliation(s)
- Rui Sang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Sheri Nixdorf
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Tzongtyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Carl Power
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thuy Anh Bui
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Alexander Engel
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia; Department of Colorectal Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| | - Ewa M Goldys
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
2
|
Zhang X, Wang B, Qi X, Qian Z, Gao X, Cheng Y, Wang X. A Glutathione-Responsive System with Prodrug and Sensitization Strategies for Targeted Therapy of Glioma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2501620. [PMID: 40119786 DOI: 10.1002/smll.202501620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/08/2025] [Indexed: 03/24/2025]
Abstract
Glioblastoma represents a highly aggressive form of malignant tumor within the central nervous system. Although chemotherapy remains the primary therapeutic strategy, its efficacy is often limited. To overcome the limitations associated with chemotherapeutic agents, such as high toxicity and non-specific adverse effects, a novel nanoparticle system comprising cRGD-modified and glutathione (GSH)-responsive polymers, and PEG-ss-Dox and apatinib (AP) (PDOX-AP/cRGD-NPs) is developed. PDOX-AP/cRGD-NPs show effective penetration of the blood-brain barrier (BBB), facilitate targeted delivery to brain tumors, and exhibit controlled drug release. PDOX-AP/cRGD-NPs show more effect in reducing the viability of GL-261, U87-MG, and LN-229 cells, inhibiting clonogenicity, and suppressing anti-apoptotic protein expression than PDOX/cRGD-NPs or AP/cRGD-NPs. Additionally, PDOX-AP/cRGD-NPs substantially increase drug uptake, BBB penetration, apoptosis rates, and the proportion of cells in the G2 phase. In vivo experiments further reveal that cRGD-directed nanoparticles exhibit superior accumulation in glioma regions compared to their non-cRGD-modified counterparts. In the interim, PDOX-AP/cRGD-NPs demonstrate significant efficacy in suppressing both ectopic and orthotopic growth of GL-261 gliomas, as well as orthotopic LN-229 gliomas, thereby markedly extending the median survival duration. This study introduces a promising targeted co-delivery system for combination chemotherapy.
Collapse
Affiliation(s)
- Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xin Qi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
3
|
Xiong S, Huang Z, Mukwaya V, Zhao W, Wang L, Dou H. Cell-Targeting Bio-Catalytic Killer Protocell for High-Order Assembly Guided Cancer Cell Inhibition. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500047. [PMID: 40270292 DOI: 10.1002/smll.202500047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/03/2025] [Indexed: 04/25/2025]
Abstract
The design and construction of synthetic therapeutic protocells capable of engaging in high-order assembly with living cells represent a significant challenge in synthetic biology and bioengineering. Inspired by cell membrane receptor-ligand systems, a protocell bioreactor is developed for targeted cancer cell elimination. This is achieved by constructing orthogonal, polysaccharide-based protocells (polysaccharidosomes, P-somes) through a bottom-up approach that leverages host-guest chemistry. The protocells are assembled via electrostatically-driven self-assembly of β-cyclodextrin (β-CD)-modified amino-dextran on a sacrificial template encapsulating glucose oxidase (GOx). To enable specific cancer cell targeting and catalytic activity, cell-targeting ligands (arginylglycylaspartic acid, cRGD) and catalase-like platinum-gold nanoparticles (Pt-AuNPs) are introduced through host-guest interactions, forming a fully functional, cell-targeting, bio-catalytic killer protocell. These protocells are programmed to spatially couple the GOx/Pt-AuNP catalytic reaction cascade. In the presence of glucose and hydroxyurea, this cascade generates a localized flux of nitric oxide (NO), which is exploited for in vitro cancer cell inhibition. Overall, the results highlight the potential of integrating orthogonal and synergistic tumor inhibition mechanisms within synthetic microcompartments. This platform demonstrates promise for future therapeutic applications, especially in cancer treatment, and represents a step forward in the development of programmable protocell-based therapeutic systems.
Collapse
Affiliation(s)
- Shuhan Xiong
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zeqi Huang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Vincent Mukwaya
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
4
|
Ben-Haim AE, Shalev N, Amalraj AJJ, Zelinger E, Mani KA, Belausov E, Shoval I, Nativ-Roth E, Maria R, Atkins A, Sadashiva R, Koltai H, Mechrez G. Nanocarriers for cancer-targeted delivery based on Pickering emulsions stabilized by casein nanoparticles. Int J Biol Macromol 2025; 298:140822. [PMID: 39929470 DOI: 10.1016/j.ijbiomac.2025.140822] [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: 10/30/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
This study demonstrates the development of stimuli-responsive Pickering emulsions stabilized by casein nanoparticles (CNPs) for targeted drug delivery to colorectal cancer cells (CRC). Encapsulation of a fluorescent dye simulates therapeutic delivery, demonstrating biomedical potential. The oil-in-water nanoemulsions stabilized by CNPs function as nanocarriers sensitive to matrix metalloproteinase-7 (MMP-7), an enzyme overexpressed in CRC cells, enabling precise drug release. Emulsions exhibited strong stability due CNPs forming a robust layer at the oil-water interface, enhancing bioavailability and controlled release. Covalent modifications of CNPs with polyethyleneimine (PEI) or polyacrylic acid (PAA), and pH adjustments optimize the zeta potential, improving surface charge and delivery efficiency. Maximal CNP uptake occurred with PAA-modified CNPs (-20 mV), showing superior interaction with CRC cells compared to pristine (-6.7 mV) and PEI-modified (+30.5, +42.1 mV) CNPs. Confocal microscopy and imaging flow cytometry confirmed that CNP-stabilized emulsions enhance CRC inter-localization compared to dispersed CNPs. Nanoemulsions with the highest CNP uptake showed selective interaction with tumor cells, while minimizing oil droplet uptake, driven by nanoscale dimensions and targeted surface interactions. Enzymatic degradation of CNPs by MMP-7 induces phase separation and targeted release. This dual-functional system, leveraging charge modification and enzymatic responsiveness, highlights CNP-stabilized nanoemulsions as a promising CRC-targeted drug delivery platform.
Collapse
Affiliation(s)
- Avital Ella Ben-Haim
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel; The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot 7610001, Israel
| | - Nurit Shalev
- Institute of Plant Sciences, Agricultural Research Organization (ARO), The Volcani Institute, PO Box 6, Beit Dagan 50250, Israel
| | - Antolin Jesila Jesu Amalraj
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel
| | - Einat Zelinger
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot 7610001, Israel
| | - Karthik Ananth Mani
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel; The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot 7610001, Israel
| | - Eduard Belausov
- Department of Ornamental Plants and Agricultural Biotechnology, Institute of Plant Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel
| | - Irit Shoval
- The Kanbar core facility unit, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Einat Nativ-Roth
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Israel
| | - Raquel Maria
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Israel
| | - Ayelet Atkins
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Rajitha Sadashiva
- Institute of Plant Sciences, Agricultural Research Organization (ARO), The Volcani Institute, PO Box 6, Beit Dagan 50250, Israel
| | - Hinanit Koltai
- Institute of Plant Sciences, Agricultural Research Organization (ARO), The Volcani Institute, PO Box 6, Beit Dagan 50250, Israel
| | - Guy Mechrez
- Department of Food Sciences, Institute of Postharvest and Food Sciences, Agricultural Research Organization (ARO), Volcani Institute, 68 HaMaccabim Road, Rishon Lezion 7505101, Israel.
| |
Collapse
|
5
|
Lim JX, Yong YK, Dewi FRP, Chan SY, Lim V. Nanoscale strategies: doxorubicin resistance challenges and enhancing cancer therapy with advanced nanotechnological approaches. Drug Deliv Transl Res 2025:10.1007/s13346-025-01790-3. [PMID: 39955406 DOI: 10.1007/s13346-025-01790-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 02/17/2025]
Abstract
Doxorubicin (DOX), an anthracycline, is widely used in cancer treatment by interfering RNA and DNA synthesis. Its broad antitumour spectrum makes it an effective therapy for a wide array of cancers. However, the prevailing drug-resistant cancer has proven to be a significant drawback to the success of the conventional chemotherapy regime and DOX has been identified as a major hurdle. Furthermore, the clinical application of DOX has been limited by rapid breakdown, increased toxicity, and decreased half-time life, highlighting an urgent need for more innovative delivery methods. Although advancements have been made, achieving a complete cure for cancer remains elusive. The development of nanoparticles offers a promising avenue for the precise delivery of DOX into the tumour microenvironment, aiming to increase the drug concentration at the target site while reducing side effects. Despite the good aspects of this technology, the classical nanoparticles struggle with issues such as premature drug leakage, low bioavailability, and insufficient penetration into tumours due to an inadequate enhanced permeability and retention (EPR) effect. Recent advancements have focused on creating stimuli-responsive nanoparticles and employing various chemosensitisers, including natural compounds and nucleic acids, fortifying the efficacy of DOX against resistant cancers. The efforts to refine nanoparticle targeting precision to improve DOX delivery are reviewed. This includes using receptor-mediated endocytosis systems to maximise the internalisation of drugs. The potential benefits and drawbacks of these novel techniques constitute significant areas of ongoing study, pointing to a promising path forward in addressing the challenges posed by drug-resistant cancers.
Collapse
Affiliation(s)
- Jian Xin Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Firli Rahmah Primula Dewi
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Siok Yee Chan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia.
| |
Collapse
|
6
|
Song R, Tan J, Cen J, Li Z, Zhang Y, Hou M, Li R, Tang L, Hu J, Liu S. Optimizing Surface Maleimide/cRGD Ratios Enhances Targeting Efficiency of cRGD-Functionalized Nanomedicines. J Am Chem Soc 2025; 147:2889-2901. [PMID: 39780364 DOI: 10.1021/jacs.4c17178] [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: 01/11/2025]
Abstract
Thiol-maleimide (MI) chemistry is a cornerstone of bioconjugation strategies, particularly in the development of drug delivery systems. The cyclic arginine-glycine-aspartic acid (cRGD) peptide, recognized for its ability to target the integrin αvβ3, is commonly employed to functionalize maleimide-bearing nanoparticles (NPs) for fabricating cRGD-functionalized nanomedicines. However, the impact of cRGD density on tumor targeting efficiency remains poorly understood. In this study, we investigate how varying MI/cRGD ratios affect the biological fate of cRGD-functionalized nanomedicines. Using a model system of nanomedicines self-assembled from phthalocyanine derivatives and PEG-PLA block copolymers, we demonstrate that an optimized cRGD/MI ratio can markedly alter the protein corona composition, leading to increased albumin adsorption, while MI-free cRGD-functionalized nanomedicines attract immunoglobulins and complement proteins. Our findings reveal that higher cRGD densities, contrary to expectations, do not enhance tumor targeting but instead promote sequestration in the liver and spleen. However, the presence of MI moieties can significantly mitigate this sequestration of cRGD-functionalized nanomedicines by promoting the formation of an albumin-rich protein corona on nanomedicines. These insights highlight the capacity of MI moieties in improving the targeting and therapeutic effects of cRGD-functionalized nanomedicines, providing refined strategies to maximize the efficacy of nanomedicines while minimizing off-target effects.
Collapse
Affiliation(s)
- Rundi Song
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Jiajia Tan
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Jie Cen
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Ziwei Li
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Yuben Zhang
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Mingxuan Hou
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Runjie Li
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Liqin Tang
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| |
Collapse
|
7
|
Wang B, Tang D, Cui J, Jiang H, Yu J, Guo Z. RGD-based self-assembling nanodrugs for improved tumor therapy. Front Pharmacol 2024; 15:1477409. [PMID: 39411070 PMCID: PMC11473307 DOI: 10.3389/fphar.2024.1477409] [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: 08/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
RGD-based self-assembling nanodrugs are a promising advancement in targeted cancer therapy, combining the specificity of RGD peptides with the benefits of nanotechnology. These nanodrugs enhance tumor targeting and cellular uptake while reducing off-target effects. RGD peptides facilitate the self-assembly of stable nanostructures, ensuring efficient drug delivery. Despite their potential, challenges such as immunogenicity, stability, tumor heterogeneity, and manufacturing scalability need to be addressed. Future research should focus on improving biocompatibility, advanced targeting strategies, personalized medicine approaches, and innovative manufacturing techniques. Overcoming these challenges will pave the way for the successful clinical translation of RGD-based nanodrugs, offering more effective and safer cancer treatments.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Dongmei Tang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jianqiao Cui
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, Municipal Hospital, Qingdao, China
| | - Zhu Guo
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Chen C, Zhang W, Wang P, Zhang Y, Zhu Y, Li Y, Wang R, Ren F. Thermo-responsive composite nanoparticles based on hydroxybutyl chitosan oligosaccharide: Fabrication, stimulus release and cancer therapy. Int J Biol Macromol 2024; 276:133842. [PMID: 39004251 DOI: 10.1016/j.ijbiomac.2024.133842] [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: 01/24/2024] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Designing thermo-responsive nanocarriers based on biopolymers is fascinating and challenging for cancer therapy. In this study, thermo-responsive composite nanoparticles (CNPs) were prepared using hydroxybutyl chitosan oligosaccharide (HBCOS) and sodium caseinate (SC) via electrostatic interactions and covalent crosslinking. The temperature-responsive behaviors of CNPs were induced by the breakage of hydrogen bonds and the shrinkage of chains in nanoparticles. The CNPs exhibited concentration-independent thermo-responsive behavior, non-adsorption aggregation, and non-hemolysis, suggesting excellent stability and thermo-sensitivity. The initial release rate and final amount of DOX released from CNPs at 42 °C were higher than that at 37 °C, showing a thermo-responsive release, which was also more prominent at lower pH. The release of DOX from CNPs followed first order kinetics based on Fickian diffusion. In vitro cytotoxicity assays confirmed the thermo-responsive antitumor activity of DOX-loaded CNPs as the HT-29 cell viability incubated with DOX-loaded CNPs at 42 °C was significantly lower than that at 37 °C. Cellular uptake experiments proved that DOX-loaded CNPs accumulated in the cytoplasm after being endocytosed and promoted DOX release by increasing environment temperature. This study generated stable thermo-sensitive CNPs based on biopolymers, which can be used as potential nanocarriers for the controlled release of anticancer drugs for cancer therapy.
Collapse
Affiliation(s)
- Chong Chen
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Weibo Zhang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Pengjie Wang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Yan Zhang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China.
| | - Yinhua Zhu
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| |
Collapse
|
9
|
Jiang C, Zhao C, Xu P, Song Q, Tao X, Lin S. Effects of Secondary Structures and pH on the Self-Assembly of Poly(ethylene glycol)- b-polytyrosine. Biomacromolecules 2024; 25:5028-5038. [PMID: 38950188 DOI: 10.1021/acs.biomac.4c00437] [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: 07/03/2024]
Abstract
Different from conventional synthetic polymers, polypeptides exhibit a distinguishing characteristic of adopting specific secondary structures, including random coils, α-helixes, and β-sheets. The conformation determines the rigidity and solubility of polypeptide chains, which further direct the self-assembly and morphology of the nanostructures. We studied the effect of distinct secondary structures on the self-assembly behavior of polytyrosine (PTyr)-derived amphiphilic copolymers. Two block copolymers of enantiopure poly(ethylene glycol)-b-poly(l-tyrosine) (PEG-b-P(l-Tyr)) and racemic poly(ethylene glycol)-b-poly(dl-tyrosine) (PEG-b-P(dl-Tyr)) were synthesized through the ring-opening polymerization of l-tyrosine N-thiocarboxyanhydride (l-Tyr-NTA) and dl-tyrosine N-thiocarboxyanhydride (dl-Tyr-NTA), respectively, by using poly(ethylene glycol) amine as the initiator. PEG44-b-P(l-Tyr)10 adopts a β-sheet conformation and self-assembles into rectangular nanosheets in aqueous solutions, while PEG44-b-P(dl-Tyr)9 is primarily in a random coil conformation with a tiny content of β-sheet structures, which self-assembles into sheaf-like nanofibrils. A pH increase results in the ionization of phenolic hydroxyl groups, which decreases the β-sheet content and increases the random coil content of the PTyr segments. Accordingly, PEG44-b-P(l-Tyr)10 and PEG44-b-P(dl-Tyr)9 self-assemble to form slender nanobelts and twisted nanoribbons, respectively, in alkaline aqueous solutions. The secondary structure-driven self-assembly of PTyr-derived copolymers is promising to construct filamentous nanostructures, which have potential for applications in controlled drug release.
Collapse
Affiliation(s)
- Caixia Jiang
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Chonghao Zhao
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Peng Xu
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qipeng Song
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xinfeng Tao
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shaoliang Lin
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
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
|
Ma R, Ji C, Shen M, Xu S, Fan G, Wu C, Yu Q, Yin L. Development of Small HN Linked Radionuclide Iodine-125 for Nanocarrier Image Tracing in Mouse Model. Int J Nanomedicine 2024; 19:1909-1922. [PMID: 38414522 PMCID: PMC10898482 DOI: 10.2147/ijn.s446564] [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: 10/27/2023] [Accepted: 02/15/2024] [Indexed: 02/29/2024] Open
Abstract
Background Radionuclides have important roles in clinical tumor radiotherapy as they are used to kill tumor cells or as imaging agents for drug tracing. The application of radionuclides has been developing as an increasing number of nanomaterials are used to deliver radionuclides to tumor areas to kill tumor cells. However, promoting the efficient combination of radionuclides and nanocarriers (NCs), enhancing radionuclide loading efficiency, and avoiding environmental pollution caused by radionuclide overuse are important challenges that hinder their further development. Methods In the present study, a new small molecule compound (3-[[(2S)-2-hydroxy-3-(4-hydroxyphenyl)-1-carbonyl] amino]-alanine, abbreviation: HN, molecular formula: C12H16N2O5) was synthesized as a linker between radionuclide iodine-125 (125I) and NCs to enable a more efficient binding between NCs and radionuclides. Results In vitro evidence indicated that the linker was able to bind 125I with higher efficiency (labeling efficiency >80%) than that of tyrosine, as well as various NCs, such as cellulose nanofibers, metal oxide NCs, and graphene oxide. Single-photon emission computed tomography/computed tomography imaging demonstrated the biological distribution of 125I-labeled NCs in different organs/tissues after administration in mice. Conclusion These results showed an improvement in radionuclide labeling efficiency for nanocarriers and provided an approach for nanocarrier image tracing.
Collapse
Affiliation(s)
- Ronglin Ma
- Center for Medical Ultrasound, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| | - Chunya Ji
- Center for Medical Ultrasound, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| | - Mengdan Shen
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| | - Shujuan Xu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| | - Guojia Fan
- Center for Cytotoxicity Testing, Sanitation & Environment Technology Institute, Soochow University, Suzhou, Jiangsu, 215006, People’s Republic of China
| | - Chengcheng Wu
- Center for Medical Ultrasound, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| | - Qiang Yu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| | - Linliang Yin
- Center for Medical Ultrasound, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, People’s Republic of China
| |
Collapse
|
12
|
Rajput H, Nangare S, Khan Z, Patil A, Bari S, Patil P. Design of lactoferrin functionalized carboxymethyl dextran coated egg albumin nanoconjugate for targeted delivery of capsaicin: Spectroscopic and cytotoxicity studies. Int J Biol Macromol 2024; 256:128392. [PMID: 38029917 DOI: 10.1016/j.ijbiomac.2023.128392] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
The increased mortality rates associated with colorectal cancer highlight the pressing need for improving treatment approaches. While capsaicin (CAP) has shown promising anticancer activity, its efficacy is hampered due to low solubility, rapid metabolism, suboptimal bioavailability, and a short half-life. Therefore, this study aimed to prepare a lactoferrin-functionalized carboxymethyl dextran-coated egg albumin nanoconjugate (LF-CMD@CAP-EGA-NCs) for the targeted CAP delivery to enhance its potential for colorectal cancer therapy. Briefly, LF-CMD was synthesized through an esterification reaction involving LF as a receptor and CMD as a shell. Concurrently, CAP was incorporated into an EGA carrier using gelation and hydrophobic interactions. The subsequent production of LF-CMD@CAP-EGA-NCs was achieved through the Maillard reaction. Spectral characterizations confirmed the successful synthesis of smooth and spherical-shaped LF-CMD@CAP-EGA-NCs using LF-CMD and EGA-CAP nanoparticles, with high entrapment efficiency and satisfactory drug content. Furthermore, LF-CMD@CAP-EGA-NCs demonstrated a sustained release of CAP (76.52 ± 1.01 % in 24 h, R2 = 0.9966) in pH 5.8 buffer with anomalous transport (n = 0.68) owing to the shell of the CMD and EGA matrix. The nanoconjugate exhibited enhanced cytotoxicity in HCT116 and LoVo cell lines, which is attributed to the overexpression of LF receptors in colorectal HCT116 cells. Additionally, LF-CMD@CAP-EGA-NCs demonstrated excellent biocompatibility, as observed in the FHC-CRL-1831 cell line. In conclusion, LF-CMD@CAP-EGA-NCs can be considered as a promising approach for targeted delivery of CAP and other anticancer agents in colorectal cancer treatment.
Collapse
Affiliation(s)
- Hrishikesh Rajput
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India; Department of Quality Assurance, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India
| | - Sopan Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India
| | - Zamir Khan
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India
| | - Ashwini Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India
| | - Sanjaykumar Bari
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India; Department of Quality Assurance, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India
| | - Pravin Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur-425405, Dist: Dhule, MS, India.
| |
Collapse
|
13
|
Chen R, Yang J, Mao Y, Zhao X, Cheng R, Deng C, Zhong Z. Antibody-Mediated Nanodrug of Proteasome Inhibitor Carfilzomib Boosts the Treatment of Multiple Myeloma. Biomacromolecules 2023; 24:5371-5380. [PMID: 37801632 DOI: 10.1021/acs.biomac.3c00830] [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: 10/08/2023]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy. For relapsed and refractory MM, a proteasome inhibitor, carfilzomib (CFZ), has become one of the few clinical options. CFZ suffers, nevertheless, metabolic instability and poor bioavailability and may induce severe cardiovascular and renal adverse events. Here, we report that daratumumab (Dar)-decorated polypeptide micelles (Dar-PMs) mediate the targeted delivery of CFZ to CD38-positive MM, effectively boosting its anti-MM efficacy. CFZ-loaded Dar-PMs (Dar-PMs-CFZ) exhibited an average diameter of ca. 80 nm and Dar density-dependent cell endocytosis and anti-MM activity, in which over 6-fold greater inhibitory effect to LP-1 and MM.1S MM cells than nontargeted PMs-CFZ control was achieved at a Dar density of 3.2 (Dar3.2-PMs-CFZ). Interestingly, Dar3.2-PMs-CFZ markedly enhanced the growth inhibition of orthotopic LP-1 MM in mice and significantly extended the median survival time compared with PMs-CFZ and free CFZ (95 days vs 60 and 54 days, respectively). In line with its high MM targetability and anti-MM efficacy, Dar3.2-PMs-CFZ revealed little toxic effects and effectively prevented osteolytic lesions. The antibody-targeted nanodelivery of a proteasome inhibitor appears to be an appealing strategy to treat multiple myeloma.
Collapse
Affiliation(s)
- Ran Chen
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Jiakun Yang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Yumin Mao
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Xiaofei Zhao
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Ru Cheng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| |
Collapse
|
14
|
Jarak I, Isabel Santos A, Helena Pinto A, Domingues C, Silva I, Melo R, Veiga F, Figueiras A. Colorectal cancer cell exosome and cytoplasmic membrane for homotypic delivery of therapeutic molecules. Int J Pharm 2023; 646:123456. [PMID: 37778515 DOI: 10.1016/j.ijpharm.2023.123456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
Colorectal cancer (CRC) is one of the most common causes of death in the world. The multi-drug resistance, especially in metastatic colorectal cancer, drives the development of new strategies that secure a positive outcome and reduce undesirable side effects. Nanotechnology has made an impact in addressing some pharmacokinetic and safety issues related to administration of free therapeutic agents. However, demands of managing complex biointerfacing require equally complex methods for introducing stimuli-responsive or targeting elements. In order to procure a more efficient solution to the overcoming of biological barriers, the physiological functions of cancer cell plasma and exosomal membranes provided the source of highly functionalized coatings. Biomimetic nanovehicles based on colorectal cancer (CRC) membranes imparted enhanced biological compatibility, immune escape and protection to diverse classes of therapeutic molecules. When loaded with therapeutic load or used as a coating for other therapeutic nanovehicles, they provide highly efficient and selective cell targeting and uptake. This review presents a detailed overview of the recent application of homotypic biomimetic nanovehicles in the management of CRC. We also address some of the current possibilities and challenges associated with the CRC membrane biomimetics.
Collapse
Affiliation(s)
- Ivana Jarak
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Porto, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Ana Isabel Santos
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Ana Helena Pinto
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Cátia Domingues
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal; Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
| | - Inês Silva
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Raquel Melo
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Francisco Veiga
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal.
| |
Collapse
|
15
|
Zhang F, Pei G, Huang B, Xu J, Zhang L. Exploring release mechanisms by disrupting π-π stacking regions in stable micelles. J Mater Chem B 2023; 11:9246-9259. [PMID: 37721031 DOI: 10.1039/d3tb01388j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
π-π stacking strategies can enhance the stability performance of delivery platforms but are often restricted by incomplete drug release performance, even with the help of crosslinking strategies. Therefore, there has been considerable interest in enhancing the drug release performance by disrupting the π-π stacking region (structural rearrangements). Herein, we synthesized poly(3-(isobutyloxy)-2-oxopropyl benzoate)-b-poly(2-hydroxybutyl methacrylate)-co-poly((ethylene glycol)methylether methacrylate) [PBOOPMA-b-P(HBMA-co-PEGMA), PHB] and revealed the drug release mechanism of PHB-based micelles. The structural rearrangements derived from the crosslinking strategy were revealed to improve the early release performance by 43-55% using micellar dissolutions. Moreover, the esterase-responsive strategy was elucidated to induce reassembly with 77-79% size variation, intensifying the structural rearrangements, which was also synergistic with the crosslinking strategy. Based on the advantages of improving drug release performance, the esterase-responsive strategy was considered a promising candidate for enhancing late release performance. Meanwhile, it is believed that such responsive modulation (crosslinking, esterase-responsive) in the π-π stacking region will become highly promising for subsequent research. Finally, the biosafety of 95.81% at 400 mg L-1 and drug cytotoxicity of IC50 ≈ 2.5 mg L-1 of PHB-EDE@CPT were also validated, confirming the broad application prospects of PHB-based crosslinked micelles.
Collapse
Affiliation(s)
- Fusheng Zhang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Gongcui Pei
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Baihao Huang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Jianchang Xu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lijuan Zhang
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
16
|
Shaik BB, Katari NK, Jonnalagadda SB. Internal stimuli-responsive nanocarriers for controlled anti-cancer drug release: a review. Ther Deliv 2023; 14:595-613. [PMID: 37877308 DOI: 10.4155/tde-2023-0041] [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] [Indexed: 10/26/2023] Open
Abstract
Cancer disease is one of the most frequent life-threatening, with a high fatality rate worldwide. However, recent immunotherapy studies in various tumours have yielded unsatisfactory outcomes, with just a few individuals experiencing long-term responses. To overcome these issues, nowadays internal stimuli-responsive nanocarriers have been widely exploited to transport a wide range of active substances, including peptides, genes and medicines. These nanosystems could be chemically adjusted to produce target-based drug release at the target location, minimizing pathological and physiological difficulties while increasing therapeutic efficiency. This review highlights the various types of internal stimuli-responsive nanocarriers and applications in cancer diagnosis. This study can provide inspiration and impetus for exploiting more promising internal stimuli-responsive nanosystems for drug delivery.
Collapse
Affiliation(s)
- Baji Baba Shaik
- Department of Chemistry, School of Science, GITAM (Deemed to be) University, Hyderabad, Telangana, 502329, India
- School of Chemistry & Physics, Westville Campus, University of KwaZulu-Natal, P Bag X 54001, Durban, 4000, Kwa-Zulu Natal, South Africa
| | - Naresh Kumar Katari
- Department of Chemistry, School of Science, GITAM (Deemed to be) University, Hyderabad, Telangana, 502329, India
- School of Chemistry & Physics, Westville Campus, University of KwaZulu-Natal, P Bag X 54001, Durban, 4000, Kwa-Zulu Natal, South Africa
| | - Sreekanth B Jonnalagadda
- School of Chemistry & Physics, Westville Campus, University of KwaZulu-Natal, P Bag X 54001, Durban, 4000, Kwa-Zulu Natal, South Africa
| |
Collapse
|
17
|
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: 217] [Impact Index Per Article: 108.5] [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
|
18
|
Li S, Li F, Wan D, Chen Z, Pan J, Liang XJ. A micelle-based stage-by-stage impelled system for efficient doxorubicin delivery. Bioact Mater 2023; 25:783-795. [PMID: 37056277 PMCID: PMC10086681 DOI: 10.1016/j.bioactmat.2022.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022] Open
Abstract
Chemotherapy remains the mainstay of cancer treatment, benefiting millions of patients each year, but the side effects of chemotherapy drugs severely limit their clinical use. Doxorubicin (DOX) can cause various side effects such as heart damage and treatment-related tumors. The effective use of active and passive targeting will improve the clinical application of DOX. Here, TPGS3350 and bioactive peptides were utilized to construct a micelle-based stage-by-stage impelled efficient system (missiles) for DOX delivery (DOX missiles). By taking advantage of the EPR effect, DOX missiles are efficiently enriched at the tumor site. After being cleaved by matrix metalloproteinase2 (MMP2), the peptide (VRGD) targets tumor cells to facilitate uptake of the missiles by the tumor cells via receptor-mediated endocytosis. The intracellular activated caspase-3-catalyzed explosion of DOX missiles further enables efficient tumor killing. This study provides an efficient approach for DOX delivery and toxicity reduction.
Collapse
Affiliation(s)
- Sunfan Li
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387, PR China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
| | - Dong Wan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387, PR China
| | - Zuqin Chen
- Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, PR China
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, PR China
- Department of Radiology, Chinese PAP Guangxi Corps Hospital, Nanning, Guangxi, PR China
| | - Jie Pan
- School of Chemical Engineering and Technology, Tiangong University, Tianjin, 300387, PR China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
19
|
Liu S, Xu Y, Wang J, Wang X, Guan S, Zhang T. Long-circulating gambogic acid-loaded nanodiamond composite nanosystem with inhibition of cell migration for tumor therapy. J Colloid Interface Sci 2023; 646:732-744. [PMID: 37229991 DOI: 10.1016/j.jcis.2023.05.103] [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: 02/02/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Herein, ultra dispersed and stably suspended nanodiamonds (NDs) were demonstrated to have a high load capacity, sustained release, and ability to serve as a biocompatible vehicle for delivery anticancer drugs. NDs with size of 50-100 nm exhibited good biocompatibility in normal human liver (L-02) cells. In particular, 50 nm ND not only promoted the noticeable proliferation of the L-02 cells but also can effectively inhibited the migration of human liver carcinoma (HepG2) cells. The gambogic acid-loaded nanodiamond (ND/GA) complex assembled by π-π stacking exhibits ultrasensitive and apparent suppression efficiency on the proliferation of HepG2 cells through high internalization and less efflux compared to free GA. More importantly, the ND/GA system can significantly increase the intracellular reactive oxygen species (ROS) levels in HepG2 cells and thus induce the cell apoptosis. The increase in intracellular ROS levels causes damage to the mitochondrial membrane potential (MMP) and activates cysteinyl aspartate specific proteinase 3 (Caspase-3) and cysteinyl aspartate specific proteinase 9 (Caspase-9), which leads to the occurrence of apoptosis. In vivo experiments also confirmed that the ND/GA complex has a much higher anti-tumor capability than free GA. Thus, the current ND/GA system is promising for cancer therapy.
Collapse
Affiliation(s)
- Shanshan Liu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; Center of Advanced Analysis and Gene Sequencing, Zhengzhou University, Zhengzhou 450001, China
| | - Yujia Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jianfeng Wang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China.
| | - Xuemin Wang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Shaokang Guan
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Tao Zhang
- School of Materials Science and Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
20
|
Wang M, Li J, Wang D, Xin Y, Liu Z. The effects of mesenchymal stem cells on the chemotherapy of colorectal cancer. Biomed Pharmacother 2023; 160:114373. [PMID: 36753960 DOI: 10.1016/j.biopha.2023.114373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Colorectal cancer (CRC) has been the third commonest cancer in the world. The prognosis of patients with CRC is related to the molecular subtypes and gene mutations, which is prone to recurrence, metastasis, and drug resistance. Mesenchymal stem cells (MSCs) are a group of progenitor ones with the capabilities of self-renewal, multi-directional differentiation, and tissue re-population, which could be isolated from various kinds of tissues and be differentiated into diverse cell types. In recent years, MSCs are applied for mechanisms study of tissue repairing, graft-versus-host disease (GVHD) and autoimmune-related disease, and tumor development, with the advantages of anti-inflammation, multi-lineage differentiation, and homing capability. Integrating the chemotherapy and MSCs therapy might provide a novel treatment approach for CRC patients. In this review, we summarize the current progress in the integrated treatment of integrating the MSCs and chemotherapy for CRC.
Collapse
Affiliation(s)
- Meiqi Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Dongxin Wang
- Department of Anesthesiology, Jilin Cancer Hospital, Jilin, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
21
|
Xie J, Zhao X, Zhang P, Zhang Y, Cheng R, Zhong Z, Deng C. Codelivery of BCL2 and MCL1 Inhibitors Enabled by Phenylboronic Acid-Functionalized Polypeptide Nanovehicles for Synergetic and Potent Therapy of Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204866. [PMID: 36683178 PMCID: PMC10015845 DOI: 10.1002/advs.202204866] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Acute myeloid leukemia (AML) is the most refractory hematologic malignancy characterized by acute onset, rapid progression, and high recurrence rate. Here, codelivery of BCL2 (ABT199) and MCL1 (TW37) inhibitors using phenylboronic acid-functionalized polypeptide nanovehicles to achieve synergetic and potent treatment of AML is adopted. Leveraging the dynamic boronic ester bonds, BN coordination, and π-π stacking, the nanovehicles reveal remarkably efficient and robust drug coencapsulation. ABT199 can induce a series of pro-apoptotic reactions by promoting the dissociation of the pro-apoptotic protein Bim from BCL2, while the released Bim is often captured by MCL1 protein overexpressed in AML. TW37 has a strong inhibitory ability to MCL1, thereby can restrain the depletion of Bim protein. Dual inhibitor-loaded nanoparticles (NPAT) reveal excellent stability, acid/enzyme/H2 O2 -triggered drug release, and significant cytotoxicity toward MOLM-13-Luc and MV-411 AML cells with low half maximal inhibitory concentrations of 1.15 and 7.45 ng mL-1 , respectively. In mice bearing MOLM-13-Luc or MV-411 AML cancer, NPAT reveal significant inhibition of tumor cell infiltration in bone marrow and main organs, potent suppression of tumor growth, and remarkably elevated mouse survival. With facile construction, varying drug combination, superior safety, synergetic efficacy, the phenylboronic acid-functionalized smart nanodrugs hold remarkable potential for AML treatment.
Collapse
Affiliation(s)
- Jiguo Xie
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Xiaofei Zhao
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Peng Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Yueyue Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Ru Cheng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Chao Deng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
22
|
Jain S, John A, George CE, Johnson RP. Tyrosine-Derived Polymers as Potential Biomaterials: Synthesis Strategies, Properties, and Applications. Biomacromolecules 2023; 24:531-565. [PMID: 36702743 DOI: 10.1021/acs.biomac.2c01232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Peptide-based polymers are evolving as promising materials for various biomedical applications. Among peptide-based polymers, polytyrosine (PTyr)-based and l-tyrosine (Tyr)-derived polymers are unique, due to their excellent biocompatibility, degradability, and functional as well as engineering properties. To date, different polymerization techniques (ring-opening polymerization, enzymatic polymerization, condensation polymerization, solution-interfacial polymerization, and electropolymerization) have been used to synthesize various PTyr-based and Tyr-derived polymers. Even though the synthesis starts from Tyr, different synthesis routes yield different polymers (polypeptides, polyarylates, polyurethanes, polycarbonates, polyiminocarbonate, and polyphosphates) with unique functional characteristics, and these polymers have been successfully used for various biomedical applications in the past decades. This Review comprehensively describes the synthesis approaches, classification, and properties of various PTyr-based and Tyr-derived polymers employed in drug delivery, tissue engineering, and biosensing applications.
Collapse
Affiliation(s)
- Supriya Jain
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Alona John
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Christina Elizhabeth George
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| |
Collapse
|
23
|
He Y, Ju Y, Hu Y, Wang B, Che S, Jian Y, Zhuo W, Fu X, Cheng Y, Zheng S, Huang N, Qian Z, Liu J, Zhou P, Gao X. Brd4 proteolysis-targeting chimera nanoparticles sensitized colorectal cancer chemotherapy. J Control Release 2023; 354:155-166. [PMID: 36538950 DOI: 10.1016/j.jconrel.2022.12.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/21/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023]
Abstract
Bromodomain-Containing Protein 4 (BRD4) is a member of the BET family of bromodomains, which participates in gene transcription process and is closely related to tumor progression. We observed the up-regulated expression of BRD4 in colorectal cancer (CRC) after doxorubicin (DOX) treatment, which might be a potential mechanism for DOX resistance. This study constructed the tumor-targeting (cyclo (Arg-Gly-Asp-D-Phe-Lys)-poly(ethylene glycol)-poly(ε-caprolactone)) (cRGD-PEG-PCL) copolymer for co-delivery of DOX and BRD4 PROTAC degrader ARV-825 (ARV-DOX/cRGD-P) for CRC treatment. The ARV-DOX/cRGD-P complexes elicited synergistic anti-tumor effect via cell cycle arrest and the increased cell apoptosis, and mechanism studies implicated the regulation of proliferation- and apoptosis-related pathways in vitro. Moreover, the administration of ARV-DOX/cRGD-P significantly improved anti-tumor activity in subcutaneous colorectal tumors and colorectal intraperitoneal disseminated tumor models in mice by promoting tumor apoptosis, suppressing tumor proliferation and angiogenesis. Taken together, these data reveal that ARV-825 can heighten DOX sensitivity in CRC treatment and BRD4 is a potential therapeutic target for DOX-resistant CRC. The ARV-DOX/cRGD-P preparations have outstanding anti-cancer effects and may be used for clinical treatment of colorectal cancer in the future.
Collapse
Affiliation(s)
- Yihong He
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yan Ju
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yuzhu Hu
- Department of Radiation Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu 610041, China
| | - Siyao Che
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yue Jian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Weiling Zhuo
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xianghui Fu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Songping Zheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ning Huang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jiagang Liu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Peizhi Zhou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
24
|
Meghana MC, Nandhini C, Benny L, George L, Varghese A. A road map on synthetic strategies and applications of biodegradable polymers. Polym Bull (Berl) 2022; 80:1-50. [PMID: 36530484 PMCID: PMC9735231 DOI: 10.1007/s00289-022-04565-9] [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: 05/01/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 12/14/2022]
Abstract
Biodegradable polymers have emerged as fascinating materials due to their non-toxicity, environmentally benign nature and good mechanical strength. The toxic effects of non-biodegradable plastics paved way for the development of sustainable and biodegradable polymers. The engineering of biodegradable polymers employing various strategies like radical ring opening polymerization, enzymatic ring opening polymerization, anionic ring opening polymerization, photo-initiated radical polymerization, chemoenzymatic method, enzymatic polymerization, ring opening polymerization and coordinative ring opening polymerization have been discussed in this review. The application of biodegradable polymeric nanoparticles in the biomedical field and cosmetic industry is considered to be an emerging field of interest. However, this review mainly highlights the applications of selected biodegradable polymers like polylactic acid, poly(ε-caprolactone), polyethylene glycol, polyhydroxyalkanoates, poly(lactide-co-glycolide) and polytrimethyl carbonate in various fields like agriculture, biomedical, biosensing, food packaging, automobiles, wastewater treatment, textile and hygiene, cosmetics and electronic devices.
Collapse
Affiliation(s)
- M. C. Meghana
- Department of Chemistry, CHRIST (Deemed to be University), Hosur Road, Bengaluru, 560029 India
| | - C. Nandhini
- Department of Chemistry, CHRIST (Deemed to be University), Hosur Road, Bengaluru, 560029 India
| | - Libina Benny
- Department of Chemistry, CHRIST (Deemed to be University), Hosur Road, Bengaluru, 560029 India
| | - Louis George
- Department of Chemistry, CHRIST (Deemed to be University), Hosur Road, Bengaluru, 560029 India
| | - Anitha Varghese
- Department of Chemistry, CHRIST (Deemed to be University), Hosur Road, Bengaluru, 560029 India
| |
Collapse
|
25
|
Nguyen VD, Kim HY, Choi YH, Park JO, Choi E. Tumor-derived extracellular vesicles for the active targeting and effective treatment of colorectal tumors in vivo. Drug Deliv 2022; 29:2621-2631. [PMID: 35941835 PMCID: PMC9367655 DOI: 10.1080/10717544.2022.2105444] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Colorectal cancer remains one of the main causes of cancer-related deaths worldwide. Although numerous nanomedicine formulations have been developed to tackle the disease, their low selectivity still limits effective therapeutic outcomes. In this study, we isolated extracellular vesicles (EVs) from CT26 colorectal cancer cells and 4T1 murine mammary carcinoma cells, loaded them with the chemotherapeutic agent (doxorubicin, DOX). Then we evaluated the cellular uptake of the extracellular vesicles both in 2D monolayer and 3D tumor spheroid setups using confocal laser scanning microscope and flow cytometry. In vivo tumor homing of the extracellular vesicles was verified on CT26 tumor bearing BALB/c mice using in vivo imaging system. Finally, in vivo therapeutic effects were evaluated and compared using the same animal models treated with five doses of EV formulations. CT26-EV-DOX exhibited excellent biocompatibility, a high drug-loading capacity, controlled drug release behavior, and a high capability for targeting colorectal cancer cells. In particular, we verified that CT26-EV-DOX could preferentially be up taken by their parent cells and could effectively target and penetrate 3D tumor spheroids resembling colorectal tumors in vivo in comparison with their 4T1 derived EV partner. Additionally, treatment of colorectal tumor-bearing BALB/c mice with of CT26-EV-DOX significantly inhibited the growth of the tumors during the treatment course. The developed CT26-EV-DOX nanoparticles may present a novel and effective strategy for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Van Du Nguyen
- School of Mechanical Engineering, Chonnam National University, Buk-gu, Gwangju, Korea.,Korea Institute of Medical Microrobotics, Buk-gu, Gwangju, Korea
| | - Ho Yong Kim
- Korea Institute of Medical Microrobotics, Buk-gu, Gwangju, Korea
| | - You Hee Choi
- Korea Institute of Medical Microrobotics, Buk-gu, Gwangju, Korea
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, Buk-gu, Gwangju, Korea
| | - Eunpyo Choi
- School of Mechanical Engineering, Chonnam National University, Buk-gu, Gwangju, Korea.,Korea Institute of Medical Microrobotics, Buk-gu, Gwangju, Korea
| |
Collapse
|
26
|
Chen S, Wang Z, Liu L, Li Y, Ni X, Yuan H, Wang C. Redox homeostasis modulation using theranostic AIE nanoparticles results in positive-feedback drug accumulation and enhanced drug penetration to combat drug-resistant cancer. Mater Today Bio 2022; 16:100396. [PMID: 36060105 PMCID: PMC9434132 DOI: 10.1016/j.mtbio.2022.100396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
Drug-resistant cancers usually have multiple barriers to compromise the effect of therapies, of which multidrug-resistance (MDR) phenotype as the intracellular barrier and dense tumor matrix as the extracellular barrier, significantly contribute to the poor anticancer performance of current drug delivery systems (DDS). Here in this study, we fabricated a novel aggregation-induced emission (AIE)-active polymer capable of self-assembling into ultrasmall nanoparticles (∼20 nm) with D-alpha Tocopheryl Polyethylene Glycol Succinate (TPGS), for dual-encapsulating of doxorubicin (Dox) and sulforaphane (SFN) (AT/Dox/SFN). It revealed that redox homeostasis modulation of MDR cells (MCF-7/Adr) using AT/Dox/SFN can trigger mitochondria damage and ATP deficiency, which reverse the MDR phenotype of MCF-7/Adr cells to afford enhanced cellular uptake of both drug and DDS in a positive-feedback manner. The enhanced cellular drug accumulation further initiates the “neighboring effect” for improved drug penetration. Using this strategy, the growth of in vivo MCF-7/Adr tumors can be effectively inhibited at a low dosage (1/5) of doxorubicin (Dox) as compared to free Dox. In summary, we offer a new approach to overcome both the intracellular and extracellular barriers of drug-resistant cancers and elucidate the potential action mechanisms, which are beneficial for better cancer management. Redox homeostasis modulation in MDR cancer cell results in positive-feedback drug accumulation and enhanced drug penetration. Mitochondria damage and neighboring effect is responsible for MDR reversal and enhanced drug penetration, respectively. AT/Dox/SFN effectively inhibits in vivo MCF-7/Adr tumors at a low dosage (1/5) of doxorubicin (Dox) as compared to free Dox.
Collapse
Affiliation(s)
- Shaoqing Chen
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu 213003, China
| | - Ziyu Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, China
| | - Li Liu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Yuting Li
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu 213003, China
- Corresponding author. Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang, China
- Corresponding author.
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
- Corresponding author.
| |
Collapse
|
27
|
Li J, Khalenkow D, Volodkin D, Lapanje A, Skirtach AG, Parakhonskiy BV. Surface enhanced Raman scattering (SERS)-active bacterial detection by Layer-by-Layer (LbL) assembly all-nanoparticle microcapsules. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
28
|
Liu Y, Xie J, Zhao X, Zhang Y, Zhong Z, Deng C. A polymeric IDO inhibitor based on poly(ethylene glycol)- b-poly(L-tyrosine- co-1-methyl-D-tryptophan) enables facile trident cancer immunotherapy. Biomater Sci 2022; 10:5731-5743. [PMID: 36039890 DOI: 10.1039/d2bm01181f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO), with an immunoregulatory effect related to tryptophan metabolism, has emerged as an attractive target for cancer immunotherapy. Here, a polymeric IDO inhibitor based on the poly(ethylene glycol)-b-poly(L-tyrosine-co-1-methyl-D-tryptophan) copolymer (PEG-b-P(Tyr-co-1-MT)) was developed for facile trident cancer immunotherapy. PEG-b-P(Tyr-co-1-MT) could self-assemble into nanoparticles (NPs), which were subject to enzyme degradation and capable of retarding the metabolism of L-tryptophan (TRP) to L-kynurenine (KYN) in B16F10 cancer cells. Notably, cRGD-functionalized NPs showed efficient encapsulation and an enzyme-responsive release of doxorubicin (DOX) and the BET bromodomain inhibitor JQ1. DOX in drug-loaded nanoparticles (cRGD-NPDJ) could activate immunization by inducing the discernible immunogenic cell death (ICD) of cancer cells and promoting the secretion of interferon-γ (IFN-γ), which besides activating the antitumor cellular immunity often upregulates the expression of PD-L1 and IDO to accelerate tumor progression. The encapsulated JQ1 and polymeric 1-MT in cRGD-NPDJ could reverse the expression by disrupting the binding of BET proteins with chromatin and elevating the TRP/KYN ratio. In B16F10 tumor-bearing C57BL/6 mice, cRGD-NPDJ displayed significantly increased CD8+ T cells, matured dendritic cells (mDCs), and cytokines (IFN-γ, TNF-α), as well as reduced regulatory T cells and downregulated PD-L1 expression at tumor sites, generating immune cascade reactions and a distinct improvement of the tumor microenvironment (TME), leading to significant tumor suppression and survival prolongation. The polymeric IDO inhibitor provides a facile strategy for the co-delivery of chemotherapeutics and inhibitors for efficient and safe combination cancer immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Jiguo Xie
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Xiaofei Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Yueyue Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Chao Deng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
29
|
Nechaeva AM, Artyukhov AA, Luss AL, Shtilman MI, Svistunova AY, Motyakin MV, Levina II, Krivoborodov EG, Toropygin I, Chistyakov EM, Tsatsakis AM, Gurevich L, Mezhuev YO. The Synthesis and Properties of a New Carrier for Paclitaxel and Doxorubicin Based on the Amphiphilic Copolymer of N‐vinyl‐2‐pyrrolidone and Acrylic Acid. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202200081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Affiliation(s)
- Anna M. Nechaeva
- Mendeleev University of Chemical Technology of Russia Moscow 125047 Russia
| | | | - Anna L. Luss
- Mendeleev University of Chemical Technology of Russia Moscow 125047 Russia
| | | | | | - Mikhail V. Motyakin
- Emanuel Institute of Biochemical Physics Russian Academy of Sciences Moscow 119334 Russia
- Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences Moscow 119991 Russia
| | - Irina I. Levina
- Emanuel Institute of Biochemical Physics Russian Academy of Sciences Moscow 119334 Russia
| | | | - IlyaYu Toropygin
- V.N. Orekhovich Research Institute of Biomedical Chemistry Russian Academy of Medical Sciences Moscow 119832 Russia
| | | | - Aristides M. Tsatsakis
- Center of Toxicology Science & Research Division of Morphology Medical School University of Crete Voutes Campus Heraklion Crete 71003 Greece
| | - Leonid Gurevich
- Department of Materials and Production Aalborg University Skjernvej 4A Aalborg 9220 Denmark
| | | |
Collapse
|
30
|
He M, Zhang Z, Jiao Z, Yan M, Miao P, Wei Z, Leng X, Li Y, Fan J, Sun W, Peng X. Redox-responsive phenyl-functionalized polylactide micelles for enhancing Ru complexes delivery and phototherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Liu F, Wang D, Zhang M, Ma L, Yu CY, Wei H. Synthesis of enzyme-responsive theranostic amphiphilic conjugated bottlebrush copolymers for enhanced anticancer drug delivery. Acta Biomater 2022; 144:15-31. [PMID: 35306183 DOI: 10.1016/j.actbio.2022.03.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/09/2022] [Accepted: 03/13/2022] [Indexed: 12/17/2022]
Abstract
Synthesis of polyfluorene (PF) based theranostic amphiphilic copolymers with simultaneously high drug loading efficiency and tumor microenvironment-specific responsiveness for promoted intracellular drug release and enhanced cancer therapy has been rarely reported likely due to the lack of efficient synthetic approaches to integrate these desirable properties. In this work, we recorded the successful preparation of well-defined theranostic amphiliphilic bottlebrush copolymers composing of fluorescent backbone of PF and tunable enzyme-degradable side chains of polytyrosine (PTyr) and POEGMA by integrating Suzuki coupling, NCA ROP and ATRP techniques. Notably, the resulting copolymer, PF25-g-(PTyr26-b-(POEGMA28)2 (P4) with two branched POEGMA brushes tethered to one PTyr termini for each unit could form steady unimolecular micelles with higher fluorescence quantum yield of 18.3% in aqueous and greater entrapment efficiency (EE) of 91.0% for DOX ascribed to the efficient π-π stacking interactions between PTyr blocks and drug molecules and the unique structure of branched hydrophilic brushes with a moderate chain length. DOX@P4 micelles revealed visualization of intracellular trafficking and accelerated drug release due to the enzyme-triggered degradation of PTyr blocks with proteinase K and subsequent deshielding of POEGMA corona for micelle destruction. In vitro and In vivo animal study further verified the intensive therapeutic efficiency with attenuated systematic toxicity. Taken together, we provided a universal strategy toward multifunctional polymeric delivery vehicles based on conjugated PF and biocompatible and degradable polypeptide by integratied Suzuki coupling and NCA ROP, and identified the branched structure of hydrophilic brushes for better performance of bottlebrush copolymers-based micelles for drug delivery applications. STATEMENT OF SIGNIFICANCE: Synthesis of polyfluorene (PF)-based theranostic amphiphilic copolymers with simultaneously high drug loading efficiency and tumor microenvironment-specific responsiveness for promoted intracellular drug release and enhanced cancer therapy has been rarely reported likely due to the lack of efficient synthetic approaches to integrate these desirable properties. We reported herein successful preparation of enzyme-responsive theranostic amphiliphilic bottlebrush copolymers with simultaneously high drug loading efficiency and tumor microenvironment-specific responsiveness for enhanced chemotherapy in vivo. This study therefore not only developed a universal strategy for the construction of multifunction polymeric vehicles based on the conjugated polymer of PF and degradable polypeptide by integrated Suzuki coupling and NCA ROP, but also emphasized the better stability of micelles endowed by the branched hydrophilic brushes than linear ones.
Collapse
Affiliation(s)
- Fangjun Liu
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Dun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China
| | - Miao Zhang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Liwei Ma
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China.
| | - Hua Wei
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China.
| |
Collapse
|
32
|
Myat YY, Ngawhirunpat T, Rojanarata T, Opanasopit P, Bradley M, Patrojanasophon P, Pornpitchanarong C. Synthesis of Polyethylene Glycol Diacrylate/Acrylic Acid Nanoparticles as Nanocarriers for the Controlled Delivery of Doxorubicin to Colorectal Cancer Cells. Pharmaceutics 2022; 14:479. [PMID: 35335856 PMCID: PMC8950920 DOI: 10.3390/pharmaceutics14030479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Doxorubicin (Dox) is known for its potential to deliver desirable anticancer effects against various types of cancer including colorectal cancer. However, the adverse effects are serious. This study aimed to synthesize polyethylene glycol diacrylate (PEGDA)/acrylic acid (AA)-based nanoparticles (PEGDA/AA NPs) for Dox delivery to colorectal cancer cells. The NPs were synthesized using free-radical polymerization reaction using the monomers PEGDA and AA with their physical properties, drug loading and release, biocompatibility, and anticancer effect evaluated. The NPs were spherical with a size of around 230 nm, with a 48% Dox loading efficiency and with loading capacity of 150 µg/mg. Intriguingly, the NPs had the ability to prolong the release of Dox in vitro over 24 h and were non-toxic to intestinal epithelial cells. Dox-loaded PEGDA/AA NPs (Dox-NPs) were able to effectively kill the colorectal cancer cell line (HT-29) with the Dox-NPs accumulating inside the cell and killing the cell through the apoptosis pathway. Overall, the synthesized PEGDA/AA NPs exhibit considerable potential as a drug delivery carrier for colon cancer-directed, staged-release therapy.
Collapse
Affiliation(s)
- Yin Yin Myat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; (Y.Y.M.); (T.N.); (T.R.); (P.O.); (P.P.)
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; (Y.Y.M.); (T.N.); (T.R.); (P.O.); (P.P.)
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; (Y.Y.M.); (T.N.); (T.R.); (P.O.); (P.P.)
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; (Y.Y.M.); (T.N.); (T.R.); (P.O.); (P.P.)
| | - Mark Bradley
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK;
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; (Y.Y.M.); (T.N.); (T.R.); (P.O.); (P.P.)
| | - Chaiyakarn Pornpitchanarong
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; (Y.Y.M.); (T.N.); (T.R.); (P.O.); (P.P.)
| |
Collapse
|
33
|
Huang X, Wu W, Jing D, Yang L, Guo H, Wang L, Zhang W, Pu F, Shao Z. Engineered exosome as targeted lncRNA MEG3 delivery vehicles for osteosarcoma therapy. J Control Release 2022; 343:107-117. [PMID: 35077741 DOI: 10.1016/j.jconrel.2022.01.026] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/27/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022]
Abstract
Exosomes as nanosized membrane vesicles, could targeted deliver therapeutic agents by modification with target ligands. Exosome-derived non-coding RNAs play a vital role in the development of tumors. Previous evidences reveal that long non-coding RNA maternally expressed gene 3 (lncRNA MEG3) has anti-tumor properties. Whereas, the inhibitory effects of exosome-derived lncRNA MEG3 in osteosarcoma (OS) remain largely unknown. In this study, we utilize the engineering technology to combine exosome and lncRNA for tumor-targeting therapy of OS. We elucidated the anti-OS effects of lncRNA MEG3, and then prepared the c(RGDyK)-modified and MEG3-loaded exosomes (cRGD-Exo-MEG3). The engineered exosomes cRGD-Exo-MEG3 could deliver more efficiently to OS cells both in vitro and in vivo. In this way, cRGD-Exo-MEG3 facilitate the anti-OS effects of MEG3 significantly, with the help of enhanced tumor-targeting therapy. This study elucidates that engineered exosomes as targeted lncRNA MEG3 delivery vehicles have potentially therapeutic effects for OS.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Wei Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Doudou Jing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingkai Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haoyu Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lutong Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Feifei Pu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
34
|
Liu H, Zhang R, Zhang D, Zhang C, Zhang Z, Fu X, Luo Y, Chen S, Wu A, Zeng W, Qu K, Zhang H, Wang S, Shi H. Cyclic RGD-Decorated Liposomal Gossypol AT-101 Targeting for Enhanced Antitumor Effect. Int J Nanomedicine 2022; 17:227-244. [PMID: 35068931 PMCID: PMC8766252 DOI: 10.2147/ijn.s341824] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction (-)-Gossypol (AT-101), the (-)-enantiomer of the natural compound gossypol, has shown significant inhibitory effects on various types of cancers such as osteosarcoma, myeloma, glioma, lung cancer, and prostate cancer. However, the clinical application of (-)-gossypol was often hindered by its evident side effects and the low bioavailability via oral administration, which necessitated the development of suitable (-)-gossypol preparations to settle the problems. In this study, injectable cyclic RGD (cRGD)-decorated liposome (cRGD-LP) was prepared for tumor-targeted delivery of (-)-gossypol. Methods The cRGD-LP was prepared based on cRGD-modified lipids. For comparison, a non-cRGD-containing liposome (LP) with a similar chemical composition to cRGD-LP was specially designed. The physicochemical properties of (-)-gossypol-loaded cRGD-LP (Gos/cRGD-LP) were investigated in terms of the drug loading efficiency, particle size, morphology, drug release, and so on. The inhibitory effect of Gos/cRGD-LP on the proliferation of tumor cells in vitro was evaluated using different cell lines. The biodistribution of cRGD-LP in vivo was investigated via the near-infrared (NIR) fluorescence imaging technique. The antitumor effect of Gos/cRGD-LP in vivo was evaluated in PC-3 tumor-bearing nude mice. Results Gos/cRGD-LP had an average particle size of about 62 nm with a narrow size distribution, drug loading efficiency of over 90%, and sustained drug release for over 96 h. The results of NIR fluorescence imaging demonstrated the enhanced tumor targeting of cRGD-LP in vivo. Moreover, Gos/cRGD-LP showed a significantly enhanced inhibitory effect on PC-3 tumors in mice, with a tumor inhibition rate of over 74% and good biocompatibility. Conclusion The incorporation of cRGD could significantly enhance the tumor-targeting effect of the liposomes and improve the antitumor effect of the liposomal (-)-gossypol in vivo, which indicated the potential of Gos/cRGD-LP that warrants further investigation for clinical applications of this single-isomer drug.
Collapse
Affiliation(s)
- Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
- Correspondence: Hao Liu School of Pharmacy, Southwest Medical University, No. 1 Section 1, Xiang Lin Road, Longmatan District, Luzhou City, Sichuan Province, 646000, People’s Republic of ChinaTel +86 830 3162291 Email
| | - Ruirui Zhang
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Chun Zhang
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Xiujuan Fu
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Yu Luo
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Siwei Chen
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Ailing Wu
- Department of Anesthesiology, The First People’s Hospital of Neijiang, Neijiang, Sichuan, People’s Republic of China
| | - Weiling Zeng
- Department of Scientific Research, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Kunyan Qu
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Hao Zhang
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Sijiao Wang
- School of Pharmacy, Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
| | - Houyin Shi
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou City, Sichuan, People’s Republic of China
- Houyin Shi Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182 Chunhui Road, Longmatan District, Luzhou City, Sichuan Province, 646000, People’s Republic of ChinaTel +86 830 3162209 Email
| |
Collapse
|
35
|
Conejos-Sánchez I, Đorđević S, Medel M, Vicent MJ. Polypeptides as building blocks for image-guided nanotherapies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
36
|
Raza F, Siyu L, Zafar H, Kamal Z, Zheng B, Su J, Qiu M. Recent Advances in Gelatin-Based Nanomedicine for Targeted Delivery of Anti-Cancer Drugs. Curr Pharm Des 2021; 28:380-394. [PMID: 34727851 DOI: 10.2174/1381612827666211102100118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/29/2021] [Accepted: 09/25/2021] [Indexed: 11/22/2022]
Abstract
Nanoparticles based on natural polymers are utilized for the development of a wide range of drug delivery systems (DDS) in the current era. Gelatin-based nanoparticles, for example, are a remarkable cancer therapy with high efficacy and specificity. This paper reviews the recent advancements in gelatin-based nanomedicine for use in cancer therapeutics. Due to the characteristics features of gelatin, such as biocompatibility, biodegradability, stability, and good surface properties, these nanoparticles provide high therapeutic potency in cancer nanomedicine. The surface of gelatin can be modified in a number of ways using various ligands to explore the platform for the development of a more novel DDS. Various methods are available for the preparation of gelatin nanomedicine discussed in this review. In addition, various cross-linkers to stabilized nanocarriers and stimuli base gelatin nanoparticles are reviewed. Furthermore, recent advances and research in gelatin-based nanomedicine are discussed. Also, some drawbacks and challenges are evaluated. In general, this paper paves the pathway to identify the details about the gelatin-based DDS for cancer therapy.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| | - Liu Siyu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| | - Zul Kamal
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| | - Bo Zheng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240. China
| |
Collapse
|
37
|
Zhang Z, Zhang Q, Xie J, Zhong Z, Deng C. Enzyme-responsive micellar JQ1 induces enhanced BET protein inhibition and immunotherapy of malignant tumors. Biomater Sci 2021; 9:6915-6926. [PMID: 34524279 DOI: 10.1039/d1bm00724f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bromodomain and extra-terminal (BET) proteins are attractive targets for treating various malignancies including melanoma. The inhibition of BET bromodomains, e.g. with JQ1, is found to downregulate the expression of both c-MYC oncoprotein and programmed cell death ligand 1 (PD-L1), which play a crucial role in tumor growth and the immunosuppressive tumor microenvironment, respectively. The BET bromodomain inhibitors like JQ1 though exhibiting high selectivity and affinity show usually low bioavailability and efficacy in vivo due to fast clearance and inferior uptake by tumor cells. The therapeutic effect of JQ1 might further be lowered by drug resistance. Here, enzyme-responsive micellar JQ1 (mJQ1) was fabricated from a poly(ethylene glycol)-b-poly(L-tyrosine) copolypeptide to enhance JQ1 delivery and the immunotherapy of malignant melanoma. The in vitro results showed that mJQ1 induced clearly better repression of c-MYC and PD-L1 proteins, cell cycle arrest, cell inhibition, and apoptotic activity than free JQ1 in B16F10 cancer cells. The intratumoral administration of mJQ1 at 2.5 mg of JQ1 equiv. per kg was found to show better inhibition of B16F10 tumors in C57BL/6 mice than the intraperitoneal administration of free JQ1 at 50 mg kg-1. In particular, when combined with radiotherapy, mJQ1 effectively suppressed tumor growth and brought about strong local and systemic antitumor immunity as evidenced by elevated CD8+ T cells and increased ratios of CD8+ T cells to Tregs, affording significantly improved survival of B16F10 tumor-bearing mice than their JQ1 counterparts and marked growth suppression of distant tumors. The great potency of enzyme-responsive micellar JQ1 makes it interesting for immunotherapy of various tumors.
Collapse
Affiliation(s)
- Zhenqi Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Qiang Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Jiguo Xie
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Chao Deng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
38
|
Li Q, Cen B, Huang W, Chen J, Chen Z, Pang J, Fu W, He S, Ji A. [Development and functional validation of a nano-delivery system of miR-16/polypeptide targeting ovarian cancer cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:736-746. [PMID: 34134962 DOI: 10.12122/j.issn.1673-4254.2021.05.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To develop a nano-delivery system for targeted delivery of miR-16/polypeptide for enhancing cisplatin sensitivity of ovarian cancer. OBJECTIVE R9-SS-R9 and cRGD-R9-SS-R9 peptides were synthesized and self-assembled with miR-16 molecules to form a nano-delivery system. The stability, particle size, potential and morphology of the nanoparticles were determined by agarose gel electrophoresis, particle size potentiometer and transmission electron microscopy. CCK-8 assay was used to assess the toxicity of the polypeptides in ovarian cancer cells. Stem loop qRT-PCR and living cell imaging were used to verify the uptake efficiency and intracellular distribution of the nanoparticles. Flow cytometry and Western blotting were performed to verify the effect of the nanoparticles for enhancing cisplatin sensitivity of ovarian cancer cells and explore the possible mechanism. OBJECTIVE R9-SS-R9/miR-16 and cRGD-R9-SS-R9/miR-16 nanoparticles were successfully prepared. The nanoparticles, with a particle size below 150 nm, a dispersity index less than 0.1 and a potential of about 40 mV, showed a good serum stability. The polypeptide material had no obvious cytotoxicity. The miR-16/polypeptide nanoparticles could be efficiently absorbed by human ovarian cancer cells and were distributed in the cytoplasm. The nanoparticles significantly increased the intracellular expression level of miR-16 (P < 0.001) and decreased the expression of Bcl-2 and Chk-1 proteins in ovarian cancer cells, thus enabling miR-16 to promote apoptosis and enhance cisplatin sensitivity of the cells. OBJECTIVE We successfully prepared a miR-16/polypeptide nano-delivery system for targeted delivery of miR-16 to ovarian cancer cells for enhancing cisplatin sensitivity of the cancer cells.
Collapse
Affiliation(s)
- Q Li
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China.,Department of Pharmacy, Nanhai Hospital Affiliated to Southern Medical University, Foshan 528200, China
| | - B Cen
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 510095, China
| | - W Huang
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China
| | - J Chen
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China
| | - Z Chen
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China
| | - J Pang
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China
| | - W Fu
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China
| | - S He
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - A Ji
- Department of Pharmacy, Nanhai Hospital Affiliated to Southern Medical University, Foshan 528200, China
| |
Collapse
|
39
|
Sang R, Stratton B, Engel A, Deng W. Liposome technologies towards colorectal cancer therapeutics. Acta Biomater 2021; 127:24-40. [PMID: 33812076 DOI: 10.1016/j.actbio.2021.03.055] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth most common deadly cancer worldwide. After treatment with curative intent recurrence rates vary with staging 0-13% in Stage 1, 11-61% in S2 and 28-73% in Stage 3. The toxicity to healthy tissues from chemotherapy and radiotherapy and drug resistance severely affect the quality of life and cancer specific outcomes of CRC patients. To overcome some of these limitations, many efforts have been made to develop nanomaterial-based drug delivery systems. Among these nanocarriers, liposomes represented one of the most successful candidates in delivering targeted oncological treatment, improving safety profile and therapeutic efficacy of encapsulated drugs. In this review we will discuss liposome design with a particular focus on the targeting feature and triggering functions. We will also summarise the recent advances in liposomal delivery system for CRC treatment in both the preclinical and clinical studies. We will finally provide our perspectives on the liposome technology development for the future clinical translation. STATEMENT OF SIGNIFICANCE: Conventional treatments for colorectal cancer (CRC) severely affect the therapeutic effects for advanced patients. With the development of nanomedicines, liposomal delivery system appears to be one of the most promising nanocarriers for CRC treatment. In last three years several reviews in this area have been published focusing on the preclinical research and drug delivery function, which is a fairly narrow focus in the field of liposome technology for CRC therapy. Our review presented the most recent advances of the liposome technology (both clinical and preclinical applications) for CRC with strong potential for further clinical translation. We believe it will attract lots of attention from various audiences, including researchers, clinicians and the industry.
Collapse
|
40
|
He M, Chen F, Shao D, Weis P, Wei Z, Sun W. Photoresponsive metallopolymer nanoparticles for cancer theranostics. Biomaterials 2021; 275:120915. [PMID: 34102525 DOI: 10.1016/j.biomaterials.2021.120915] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022]
Abstract
Over the past decades, transition metal complexes have been successfully used in anticancer phototherapies. They have shown promising properties in many different areas including photo-induced ligand exchange or release, rich excited state behavior, and versatile biochemical properties. When encorporated into polymeric frameworks and become part of nanostructures, photoresponsive metallopolymer nanoparticles (MPNs) show enhanced water solubility, extended blood circulation and increased tumor-specific accumulation, which greatly improves the tumor therapeutic effects compared to low-molecule-weight metal complexes. In this review, we aim to present the recent development of photoresponsive MPNs as therapeutic nanomedicines. This review will summarize four major areas separately, namely platinum-containing polymers, zinc-containing polymers, iridium-containing polymers and ruthenium-containing polymers. Representative MPNs of each type are discussed in terms of their design strategies, fabrication methods, and working mechanisms. Current challenges and future perspectives in this field are also highlighted.
Collapse
Affiliation(s)
- Maomao He
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116024, China
| | - Fangman Chen
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 510630, China
| | - Dan Shao
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 510630, China
| | - Philipp Weis
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Zhiyong Wei
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116024, China.
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
41
|
Wang X, Song Z, Wei S, Ji G, Zheng X, Fu Z, Cheng J. Polypeptide-based drug delivery systems for programmed release. Biomaterials 2021; 275:120913. [PMID: 34217020 DOI: 10.1016/j.biomaterials.2021.120913] [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: 01/08/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023]
Abstract
Recent years have seen increasing interests in the use of ring-opening polymerization of α-amino acid N-carboxyanhydrides (NCAs) to prepare synthetic polypeptides, a class of biocompatible and versatile materials, for various biomedical applications. Because of their rich side-chain functionalities, diverse hydrophilicity/hydrophobicity profiles, and the capability of forming stable secondary structures, polypeptides can assemble into a variety of well-organized nano-structures that have unique advantages in drug delivery and controlled release. Herein, we review the design and use of polypeptide-based drug delivery system derived from NCA chemistry, and discuss the future perspectives of this exciting and important biomaterial area that may potentially change the landscape of next-generation therapeutics and diagnosis. Given the high significance of precise control over release for polypeptide-based systems, we specifically focus on the versatile designs of drug delivery systems capable of programmed release, through the changes in the chemical and physical properties controlled by the built-in molecular structures of polypeptides.
Collapse
Affiliation(s)
- Xu Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China; Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States; Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| | - Shiqi Wei
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Guonan Ji
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Xuetao Zheng
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Zihuan Fu
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States; Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States.
| |
Collapse
|
42
|
Ginghină O, Hudiță A, Zaharia C, Tsatsakis A, Mezhuev Y, Costache M, Gălățeanu B. Current Landscape in Organic Nanosized Materials Advances for Improved Management of Colorectal Cancer Patients. MATERIALS (BASEL, SWITZERLAND) 2021; 14:2440. [PMID: 34066710 PMCID: PMC8125868 DOI: 10.3390/ma14092440] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 12/24/2022]
Abstract
Globally, colorectal cancer (CRC) ranks as one of the most prevalent types of cancers at the moment, being the second cause of cancer-related deaths. The CRC chemotherapy backbone is represented by 5-fluorouracil, oxaliplatin, irinotecan, and their combinations, but their administration presents several serious disadvantages, such as poor bioavailability, lack of tumor specificity, and susceptibility to multidrug resistance. To address these limitations, nanomedicine has arisen as a powerful tool to improve current chemotherapy since nanosized carriers hold great promise in improving the stability and solubility of the drug payload and enhancing the active concentration of the drug that reaches the tumor tissue, increasing, therefore, the safety and efficacy of the treatment. In this context, the present review offers an overview of the most recent advances in the development of nanosized drug-delivery systems as smart therapeutic tools in CRC management and highlights the emerging need for improving the existing in vitro cancer models to reduce animal testing and increase the success of nanomedicine in clinical trials.
Collapse
Affiliation(s)
- Octav Ginghină
- Department of Surgery, “Sf. Ioan” Emergency Clinical Hospital, 13 Vitan Barzesti Street, 042122 Bucharest, Romania;
- Department II, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy Bucharest, 17-21 Calea Plevnei Street, 010232 Bucharest, Romania
| | - Ariana Hudiță
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (M.C.); (B.G.)
| | - Cătălin Zaharia
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania;
| | - Aristidis Tsatsakis
- Department of Toxicology and Forensic Sciences, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Yaroslav Mezhuev
- Center of Biomaterials, D Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, 125047 Moscow, Russia;
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (M.C.); (B.G.)
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (M.C.); (B.G.)
| |
Collapse
|
43
|
Ma W, Yang H, Hu Y, Chen L. Fabrication of
PEGylated
porphyrin/reduced graphene oxide/doxorubicin nanoplatform for tumour combination therapy. POLYM INT 2021. [DOI: 10.1002/pi.6216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Weiqian Ma
- Department of Chemistry Northeast Normal University Changchun PR China
| | - Huailin Yang
- Department of Chemistry Northeast Normal University Changchun PR China
| | - Yanfang Hu
- Fachgruppe Chemie Institut für Organische Chemie, RWTH Aachen Aachen Germany
| | - Li Chen
- Department of Chemistry Northeast Normal University Changchun PR China
| |
Collapse
|
44
|
Abstract
Multifunctional nanoparticles have been identified as a promising drug-delivery system for sustainable drug release. The structural and size tunability and disease-targeting ability of nanoparticles have made them more suitable for multiple drug loading and delivery, thereby enhancing therapeutic results through synergistic effects. Nanoparticulate carriers with specific features such as target specificity and stimuli-responsiveness enable selective drug delivery with lower potential side effects. In this review we have classified the recently published articles on polymeric and inorganic nanoparticle-mediated drug delivery into three different categories based on functionality and discussed their efficiency for drug delivery and their therapeutic outcomes in preclinical models. Most of the drug-loaded nanodelivery systems discussed have demonstrated negligible or very low systemic toxicity throughout the experimental period in animal models compared with free drug administration. In addition, some challenges associated with the translation of nanoparticle-based drug carrier responses to clinical application are highlighted.
Collapse
|
45
|
Vinchhi P, Patel MM. Triumph against cancer: invading colorectal cancer with nanotechnology. Expert Opin Drug Deliv 2021; 18:1169-1192. [PMID: 33567909 DOI: 10.1080/17425247.2021.1889512] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Recent statistics have reported colorectal cancer (CRC) as the second leading cause of cancer-associated deaths in the world. Early diagnosis of CRC may help to reduce the mortality and associated complications. However, the conventional diagnostic techniques often lead to misdiagnosis, fail to differentiate benign from malignant tissue or diagnose only at an advanced stage. For the treatment of CRC, surgery, chemotherapy, immunotherapy, and radiotherapy have been employed. However, the quality of living of the CRC patients is highly compromised after employing current therapeutic approaches owing to the toxicity issues and relapse. AREA COVERED This review accentuates the molecular mechanisms involved in the pathogenesis, stages of CRC, conventional approaches for diagnosis and therapy of CRC and the issues confronted thereby. It provides an outlook on the advantages of employing nanotechnology-based approaches for prevention, early diagnosis, and treatment of CRC. EXPERT OPINION Employing nanotechnology-based approaches has demonstrated promising outcomes in the prevention, diagnosis, and treatment of CRC. Nanotechnology-based approaches can surmount the major drawbacks of traditional diagnostic and therapeutic approaches. Nanotechnology bestows the advantage of early detection of CRC which helps to undertake instant steps for offering efficient therapy and reducing the mortality rates. For the treatment of CRC, nanocarriers offer the benefit of achieving controlled drug release, improved drug bioavailability, enhanced tumor targetability and reduced adverse effects.
Collapse
Affiliation(s)
- Preksha Vinchhi
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Mayur M Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
46
|
Ahmad E, Ali A, Fatima MT, Nimisha, Apurva, Kumar A, Sumi MP, Sattar RSA, Mahajan B, Saluja SS. Ligand decorated biodegradable nanomedicine in the treatment of cancer. Pharmacol Res 2021; 167:105544. [PMID: 33722711 DOI: 10.1016/j.phrs.2021.105544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
Cancer is one of the major global health problems, responsible for the second-highest number of deaths. The genetic and epigenetic changes in the oncogenes or tumor suppressor genes alter the regulatory pathways leading to its onset and progression. Conventional methods are used in appropriate combinations for the treatment. Surgery effectively treats localized tumors; however, it fails to treat metastatic tumors, leading to a spread in other organs, causing a high recurrence rate and death. Among the different strategies, the nanocarriers-based approach is highly sought for, but its nonspecific delivery can cause a profound side effect on healthy cells. Targeted nanomedicine has the advantage of targeting cancer cells specifically by interacting with the receptors overexpressed on their surface, overcoming its non-specificity to target healthy cells. Nanocarriers prepared from biodegradable and biocompatible materials are decorated with different ligands by encapsulating therapeutic or diagnostic agents or both to target cancer cells overexpressing the receptors. Scientists are now utilizing a theranostic approach to simultaneously evaluate nanocarrier bio-distribution and its effect on the treatment regime. Herein, we have summarized the recent 5-year efforts in the development of the ligands decorated biodegradable nanocarriers, as a targeted nanomedicine approach, which has been highly promising in the treatment of cancer.
Collapse
Affiliation(s)
- Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Science, Patna 810507, India
| | - Munazza Tamkeen Fatima
- Department of Pharmaceutical Science, College of Pharmacy, QU health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Mamta P Sumi
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Bhawna Mahajan
- Department of Biochemistry, Govind Ballabh Pant, Postgraduate Institute of Medical, Education and Research (GIPMER), New Delhi 110002, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India; Department of GI Surgery, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India.
| |
Collapse
|
47
|
Wang Y, Shen N, Wang Y, Zhang Y, Tang Z, Chen X. Self-Amplifying Nanotherapeutic Drugs Homing to Tumors in a Manner of Chain Reaction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2002094. [PMID: 33382144 DOI: 10.1002/adma.202002094] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/26/2020] [Indexed: 06/12/2023]
Abstract
Active tumor-targeting drug delivery has great potency in cancer therapy. However, the targeting efficiency of traditional active tumor-targeting nanotherapeutic drugs is limited by the scarcity of their accessible targets/receptors in tumors. Here, a novel self-amplifying tumor-targeting strategy with a chain reaction mechanism is developed. A coagulation targeting peptide (GNQEQVSPLTLLKXC, termed A15)-decorated poly(L-glutamic acid)-graft-maleimide poly(ethylene glycol)/combretastatin A4 conjugate (A15-PLG-CA4) is prepared to obtain a self-amplifying nanotherapeutic platform homing to tumors. After administration to tumor-bearing mice, A15-PLG-CA4 starts a chain reaction cycle consisting of intratumoral hemorrhage, target FXIIIa amplification, blood clot binding, and CA4 release in tumors. In this way, A15-PLG-CA4 increases the level of its accessible targets (FXIIIa) in a manner of chain reaction. The FXIIIa activity at 8 h is 4.1-fold more than the one at 0 h in the C26 tumors treated with A15-PLG-CA4. The total CA4 concentration at 24 h is 2.9-fold more than the control. A15-PLG-CA4 shows a significantly higher antitumor effect against large C26 tumors (≈500 mm3 ) thanks to the remarkable tumor-targeting ability compared with the control. Therefore, this report highlights the potential of the self-amplifying tumor-targeting strategy in the development of next generation active tumor-targeting nanotherapeutic drugs for tumor therapy.
Collapse
Affiliation(s)
- Yue Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Ying Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
48
|
Gao Y, Li X, Zeng C, Liu C, Hao Q, Li W, Zhang K, Zhang W, Wang S, Zhao H, Fan D, Li M, Zhang Y, Zhang W, Zhang C. CD63 + Cancer-Associated Fibroblasts Confer Tamoxifen Resistance to Breast Cancer Cells through Exosomal miR-22. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002518. [PMID: 33173749 PMCID: PMC7610308 DOI: 10.1002/advs.202002518] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Indexed: 05/21/2023]
Abstract
Tamoxifen remains the most effective treatment for estrogen receptor α (ERα)-positive breast cancer. However, many patients still develop resistance to tamoxifen in association with metastatic recurrence, which presents a tremendous clinical challenge. To better understand tamoxifen resistance from the perspective of the tumor microenvironment, the whole microenvironment landscape is charted by single-cell RNA sequencing and a new cancer-associated fibroblast (CAF) subset, CD63+ CAFs, is identified that promotes tamoxifen resistance in breast cancer. Furthermore, it is discovered that CD63+ CAFs secrete exosomes rich in miR-22, which can bind its targets, ERα and PTEN, to confer tamoxifen resistance on breast cancer cells. Additionally, it is found that the packaging of miR-22 into CD63+ CAF-derived exosomes is mediated by SFRS1. Furthermore, CD63 induces STAT3 activation to maintain the phenotype and function of CD63+ CAFs. Most importantly, the pharmacological blockade of CD63+ CAFs with a CD63-neutralizing antibody or cRGD-miR-22-sponge nanoparticles enhances the therapeutic effect of tamoxifen in breast cancer. In summary, the study reveals a novel subset of CD63+ CAFs that induces tamoxifen resistance in breast cancer via exosomal miR-22, suggesting that CD63+ CAFs may be a novel therapeutic target to enhance tamoxifen sensitivity.
Collapse
Affiliation(s)
- Yuan Gao
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Xiaoju Li
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Cheng Zeng
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
- Institute of Material MedicalSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Chenlin Liu
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Qiang Hao
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Weina Li
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Kuo Zhang
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Wangqian Zhang
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Shuning Wang
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Huadong Zhao
- Department of General SurgeryTangdu HospitalThe Fourth Military Medical UniversityXi'an710038P. R. China
| | - Dong Fan
- Department of General SurgeryTangdu HospitalThe Fourth Military Medical UniversityXi'an710038P. R. China
| | - Meng Li
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Yingqi Zhang
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Wei Zhang
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Cun Zhang
- The State Key Laboratory of Cancer BiologyBiotechnology CenterSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032P. R. China
| |
Collapse
|
49
|
Biodegradable polyurethane PMeOx-PU(SS)-PMeOx micelles with redox and pH-sensitivity for efficient delivery of doxorubicin. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.110054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
50
|
Robust and smart polypeptide-based nanomedicines for targeted tumor therapy. Adv Drug Deliv Rev 2020; 160:199-211. [PMID: 33137364 DOI: 10.1016/j.addr.2020.10.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Nanomedicines based on synthetic polypeptides are among the most versatile and advanced platforms for tumor therapy. Notably, several polypeptide-based nanodrugs are currently under human clinical assessments. The previous (pre)clinical studies clearly show that dynamic stability (i.e. stable in circulation while destabilized in tumor) of nanomedicines plays a vital role in their anti-tumor performance. Various strategies have recently been developed to design dynamically stabilized polypeptide-based nanomedicines by e.g. crosslinking the nanovehicles with acid, reactive oxygen species (ROS), glutathione (GSH), or photo-sensitive linkers, inter-crosslinking between vehicles and drugs, introducing π-π stacking or lipid-lipid interactions in the nanovehicles, chemically conjugating drugs to vehicles, and forming unimolecular micelles. Interestingly, these robust and smart nanodrugs have demonstrated improved tumor targetability, anti-tumor efficacy, as well as safety profiles in different tumor models. In this review, representative strategies to robust and smart polypeptide-based nanomedicines for targeted treatment of varying malignancies are highlighted. The exciting development of dynamic nanomedicines will foresee further increasing clinical translation in the future.
Collapse
|