1
|
Wang X, Hao X, Zhang Y, Wu Q, Zhou J, Cheng Z, Chen J, Liu S, Pan J, Wang Y, Fan JB. Bioinspired Adaptive Microdrugs Enhance the Chemotherapy of Malignant Glioma: Beyond Their Nanodrugs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405165. [PMID: 38758975 DOI: 10.1002/adma.202405165] [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: 04/10/2024] [Revised: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Solid nanoparticle-mediated drug delivery systems are usually confined to nanoscale due to the enhanced permeability and retention effect. However, they remain a great challenge for malignant glioma chemotherapy because of poor drug delivery efficiency and insufficient tumor penetration resulting from the blood-brain barrier/blood-brain tumor barrier (BBB/BBTB). Inspired by biological microparticles (e.g., cells) with excellent adaptive deformation, it is demonstrated that the adaptive microdrugs (even up to 3.0 µm in size) are more efficient than their nanodrugs (less than 200 nm in size) to cross BBB/BBTB and penetrate into tumor tissues, achieving highly efficient chemotherapy of malignant glioma. The distinct delivery of the adaptive microdrugs is mainly attributed to the enhanced interfacial binding and endocytosis via adaptive deformation. As expected, the obtained adaptive microdrugs exhibit enhanced accumulation, deep penetration, and cellular internalization into tumor tissues in comparison with nanodrugs, significantly improving the survival rate of glioblastoma mice. It is believed that the bioinspired adaptive microdrugs enable them to efficiently cross physiological barriers and deeply penetrate tumor tissues for drug delivery, providing an avenue for the treatment of solid tumors.
Collapse
Affiliation(s)
- Xuejiao Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xiangrong Hao
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yangning Zhang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Qun Wu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jiajia Zhou
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, 510515, P. R. China
| | - Zhongman Cheng
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jianping Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
- Department of Radiotherapy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, P. R. China
| | - Sijia Liu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jiahao Pan
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Ying Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jun-Bing Fan
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
2
|
Beach M, Nayanathara U, Gao Y, Zhang C, Xiong Y, Wang Y, Such GK. Polymeric Nanoparticles for Drug Delivery. Chem Rev 2024; 124:5505-5616. [PMID: 38626459 PMCID: PMC11086401 DOI: 10.1021/acs.chemrev.3c00705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
3
|
Liu X, Wang F, Liu L, Li T, Zhong X, Lin H, Zhang Y, Xue W. Functionalized polydopamine nanospheres as in situ spray for photothermal image-guided tumor precise surgical resection. Biosens Bioelectron 2023; 222:114995. [PMID: 36516631 DOI: 10.1016/j.bios.2022.114995] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/18/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022]
Abstract
Surgical resection is a critical procedure for treatment of solid tumor, which commonly suffers from postoperative local recurrence due to the possibility of positive surgical margin. Although the widely used clinical imaging techniques (CT, MRI, PET, etc.) show beneficial effects in providing a macroscopic view of preoperative tumor position, they are still failing to provide intraoperative real-time imaging navigation during the surgery and need oral or intravenous injection contrast agents with risk of adverse effects. In this work, we present a nano-spray assisted photothermal imaging system for in vitro cells discrimination as well as in vivo visualization of tumor position and border that guides real-time precise tumor resection during surgery (even for tiny tumor less than 3 mm). Herein, the nano-spray were prepared by RGD peptide functionalized polydopamine (PDA-RGD) nanospheres with excellent photothermal conversion efficiency (54.27%), stability and reversibility, which target ανβ3 integrin overexpressed tumor cells. Such PDA-RGD serve as nanothermometers that convert and amplify biological signal to intuitive thermal image signal, depicting the tumor margin in situ. In comparison to conventional imaging techniques, our approach through topical spraying together with portable infrared camera has the characteristics of low cost, convenient, no radiation hazard, real-time intraoperative imaging-guidance and avoiding the adverse effects risk of oral or intravenous contrast agent. This technology provides a new universal tool for potentially assisting surgeons' decision in real-time during surgery and aiding to improved outcome.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China; Center for Hybrid Nanostructure (CHyN), Department of Physics, University of Hamburg, Hamburg, 22761, Germany
| | - Fan Wang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Li Liu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Tiantian Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xiangyu Zhong
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Hongsheng Lin
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Yi Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China.
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China; MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Zhou M, Zou X, Cheng K, Zhong S, Su Y, Wu T, Tao Y, Cong L, Yan B, Jiang Y. The role of cell-penetrating peptides in potential anti-cancer therapy. Clin Transl Med 2022; 12:e822. [PMID: 35593206 PMCID: PMC9121317 DOI: 10.1002/ctm2.822] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 12/19/2022] Open
Abstract
Due to the complex physiological structure, microenvironment and multiple physiological barriers, traditional anti-cancer drugs are severely restricted from reaching the tumour site. Cell-penetrating peptides (CPPs) are typically made up of 5-30 amino acids, and can be utilised as molecular transporters to facilitate the passage of therapeutic drugs across physiological barriers. Up to now, CPPs have widely been used in many anti-cancer treatment strategies, serving as an excellent potential choice for oncology treatment. However, their drawbacks, such as the lack of cell specificity, short duration of action, poor stability in vivo, compatibility problems (i.e. immunogenicity), poor therapeutic efficacy and formation of unwanted metabolites, have limited their further application in cancer treatment. The cellular uptake mechanisms of CPPs involve mainly endocytosis and direct penetration, but still remain highly controversial in academia. The CPPs-based drug delivery strategy could be improved by clever design or chemical modifications to develop the next-generation CPPs with enhanced cell penetration capability, stability and selectivity. In addition, some recent advances in targeted cell penetration that involve CPPs provide some new ideas to optimise CPPs.
Collapse
Affiliation(s)
- Meiling Zhou
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Xi Zou
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Kexin Cheng
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Suye Zhong
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Yangzhou Su
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Tao Wu
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Li Cong
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Bin Yan
- Department of Pathology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Yiqun Jiang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
5
|
Yang S, Chen W, Li W, Song J, Gao Y, Si W, Li X, Cui B, Yu T. CD44-targeted pH-responsive micelles for enhanced cellular internalization and intracellular on-demand release of doxorubicin. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:173-184. [PMID: 33620265 DOI: 10.1080/21691401.2021.1884085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Poor cellular uptake and slow intracellular drug release remain the main barriers for the efficient application of micellar delivery system. Taking advantage of the overexpressed CD44 receptor and mild acidic microenvironment of tumour cells, CD44-targeted pH-responsive micelles based on the self-assembly of histidine-hyaluronic acid-dodecylamine (His-HA-DA) were prepared for the delivery of doxorubicin (DOX). These micelles exhibited pH-responsive behaviour with increased particle size, decreased encapsulation efficiency (EE%) of DOX and rapid release of DOX triggered by low pH. Compared with free DOX, DOX/HHD exhibited relatively high cellular uptake mainly via the CD44-mediated endocytosis. The on-demand intracellular release of DOX from DOX/HHD led to improved cytotoxicity. DOX/HHD also showed great penetration efficiency in 3D tumour spheres in vitro. Moreover, these micelles with suitable particle size gained excellent tumour-targeting effects, as well as improved anti-tumour effects and reduced side effects in vivo. In conclusion, these micelles with CD44 targeted and pH-responsive behaviours provide a promising strategy for the efficient delivery of anti-tumour drugs in vivo.
Collapse
Affiliation(s)
- Shudi Yang
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| | - Weiliang Chen
- Pharmaceutical Department, Livzon Research Institute, Livzon Pharmaceutical Group Inc., Zhuhai, China
| | - Wei Li
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Jingcheng Song
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| | - Yue Gao
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| | - Wenhui Si
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| | - Xiaoping Li
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| | - Baowei Cui
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| | - Tongtong Yu
- Suzhou Polytechnic Institute of Agriculture, Suzhou, China
| |
Collapse
|
6
|
Li D, Gao J, Yang C, Li B, Sun J, Yu M, Wang Y, Wang H, Lu Y. cRGDyK-modified procaine liposome inhibits the proliferation and motility of glioma cells via the ERK/p38MAPK pathway. Exp Ther Med 2021; 22:859. [PMID: 34178132 PMCID: PMC8220655 DOI: 10.3892/etm.2021.10291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
Glioma is a common type of primary tumor in the central nervous system. Glioma has been increasing in incidence yearly and is a serious threat to human life and health. The aim of the present study was to prepare liposomes for enhanced penetration of the blood-brain barrier and targeting of glioma. A procaine-loaded liposome modified with the cyclic pentapeptide cRGDyK (Pro/cRGDyK-L) was designed and developed. The particle size, ζ potential, encapsulation efficiency, release profile, stability and hemolysis of Pro/cRGDyK-L were characterized in vitro. The targeting and antitumor effects of Pro/cRGDyK-L were also investigated in vitro and in vivo. The results suggested that the cRGDyK peptide significantly facilitated the ability of liposomes to transfer procaine across the BBB and improved the cellular uptake of procaine by C6 glioma cells. The results further demonstrated that Pro/cRGDyK-L strongly suppressed cell motility, stimulated apoptosis and induced cell cycle arrest. The findings further confirmed that Pro/cRGDyK-L exhibited superior antitumor effects by targeting the ERK/p38MAPK pathway and thereby suppressed tumor growth in mice. In conclusion, the present study indicated the potential of Pro/cRGDyK-L as a means to provide improved therapeutic effects on glioma through the ERK/p38MAPK pathway.
Collapse
Affiliation(s)
- Dedong Li
- Department of Anesthesiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Jie Gao
- Department of Anesthesiology, Tianjin Haihe Hospital, Tianjin 300350, P.R. China
| | - Chenyi Yang
- Department of Anesthesiology, Tianjin Third Central Hospital, Tianjin 300052, P.R. China
| | - Bo Li
- Department of Anesthesiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Jian Sun
- Department of Anesthesiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Mingdong Yu
- Department of Anesthesiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Ying Wang
- Department of Anesthesiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Haiyun Wang
- Department of Anesthesiology, Tianjin Third Central Hospital, Tianjin 300052, P.R. China
| | - Yuechun Lu
- Department of Anesthesiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
7
|
Shi M, Wang Y, Zhao X, Zhang J, Hu H, Qiao M, Zhao X, Chen D. Stimuli-Responsive and Highly Penetrable Nanoparticles as a Multifunctional Nanoplatform for Boosting Nonsmall Cell Lung Cancer siRNA Therapy. ACS Biomater Sci Eng 2021; 7:3141-3155. [PMID: 34137580 DOI: 10.1021/acsbiomaterials.1c00582] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In cancer therapy, it is acknowledged that large-size nanoparticles stay in the circulation system for a long time, but their permeability to tumor tissues is poor. To address the conflicting need for prolonging circulation time and favorable tumor tissue penetration ability, a charge conversional multifunctional nanoplatform was strategically designed to improve the efficacy of small interfering RNA (siRNA) therapy against nonsmall cell lung cancer (NSCLC). The development of nanodrug delivery systems (NDDSs) was constructed by loading siRNA on polyamidoamine (PAMAM) dendrimers to build small-sized PAM/siRNA via electrostatic interaction and then capped with a pH-triggered copolymer poly(ethylene glycol) methyl ether (mPEG)-poly-l-lysine (PLL)-2,3-dimethylmaleic anhydride (DMA) (shorted as PLM) under physiological conditions. While in the tumor microenvironment, the acidic reaction of the PLM copolymer changes from negative charge to positive charge due to the cleavable amide bond between mPEG-PLL and DMA, leading to large-size nanoparticles (NPs) with a negative charge that turns into a positive charge and small NPs with a high tumor-penetrating ability. All of the in vitro and in vivo studies validated that PLM/PAM/siRNA NPs possess desirable features including excellent biocompatibility, a prolonged circulation time, significant pH sensitivity, high tumor tissue penetration ability, and sufficient endo-/lysosomal escape. Taken together, all results suggest tremendous potential of the gene therapy based on the stimuli-sensitive PLM/PAM/siRNA NPs, providing a profound application prospective treatment strategy in cancer gene therapy.
Collapse
Affiliation(s)
- Menghao Shi
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| | - Yu Wang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| | - Xiufeng Zhao
- Hongqi Hostital, Mudanjiang Medical University, No. 5 Tongxiang Road, Mudanjiang, Heilongjiang Province 157011, P. R. China
| | - Jiulong Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| | - Mingxi Qiao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| | - Xiuli Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning Province 110016, P. R. China
| |
Collapse
|
8
|
Desale K, Kuche K, Jain S. Cell-penetrating peptides (CPPs): an overview of applications for improving the potential of nanotherapeutics. Biomater Sci 2021; 9:1153-1188. [PMID: 33355322 DOI: 10.1039/d0bm01755h] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the field of nanotherapeutics, gaining cellular entry into the cytoplasm of the target cell continues to be an ultimate challenge. There are many physicochemical factors such as charge, size and molecular weight of the molecules and delivery vehicles, which restrict their cellular entry. Hence, to dodge such situations, a class of short peptides called cell-penetrating peptides (CPPs) was brought into use. CPPs can effectively interact with the cell membrane and can assist in achieving the desired intracellular entry. Such strategy is majorly employed in the field of cancer therapy and diagnosis, but now it is also used for other purposes such as evaluation of atherosclerotic plaques, determination of thrombin levels and HIV therapy. Thus, the current review expounds on each of these mentioned aspects. Further, the review briefly summarizes the basic know-how of CPPs, their utility as therapeutic molecules, their use in cancer therapy, tumor imaging and their assistance to nanocarriers in improving their membrane penetrability. The review also discusses the challenges faced with CPPs pertaining to their stability and also mentions the strategies to overcome them. Thus, in a nutshell, this review will assist in understanding how CPPs can present novel possibilities for resolving the conventional issues faced with the present-day nanotherapeutics.
Collapse
Affiliation(s)
- Kalyani Desale
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| |
Collapse
|
9
|
Yu W, Maynard E, Chiaradia V, Arno MC, Dove AP. Aliphatic Polycarbonates from Cyclic Carbonate Monomers and Their Application as Biomaterials. Chem Rev 2021; 121:10865-10907. [DOI: 10.1021/acs.chemrev.0c00883] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Wei Yu
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Edward Maynard
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Viviane Chiaradia
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Maria C. Arno
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Andrew P. Dove
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| |
Collapse
|
10
|
Wang M, Zhang L, Cai Y, Yang Y, Qiu L, Shen Y, Jin J, Zhou J, Chen J. Bioengineered Human Serum Albumin Fusion Protein as Target/Enzyme/pH Three-Stage Propulsive Drug Vehicle for Tumor Therapy. ACS NANO 2020; 14:17405-17418. [PMID: 33202141 DOI: 10.1021/acsnano.0c07610] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Human serum albumin (HSA) has the characteristics of biocompatibility and long circulation, which is widely used as the carrier of insoluble anticancer drugs, but it also has some disadvantages such as weak tumor targeting and uncontrollable drug release. Herein, HSA was modified to improve its biological performance by introducing polyhistidine (pHis), matrix metalloproteinase-2 (MMP-2) digestion, and Arg-Gly-Asp (RGD) peptide at the separated end of HSA through gene fusion technology. The resulting protein expressed by Pichia pastoris could self-assemble into 3RGD-HSA-MMP-18His nanoparticles (RHMH18 NPs) accompanied by loading hydrophobic drug paclitaxel (PTX) into the polyhistidine micelle core. RHMH18 NPs exhibited active tumor targeting in high efficiency owing to the RGD-mediated specific binding toward ανβ3-integrin upregulated on tumor vasculature endothelium, resulting in the enrichment of therapeutic substances in tumor sites. Once reaching the tumor microenvironment, RHMH18 NPs was cut off by MMP-2 to remove the HSA-3RGD moiety, leaving the small and positively charged histidine micelle, which could penetrate the deep part of tumor tissue more effectively. Finally, the histidine micelle escaped from lysosome successfully and released drug in response to pH. The in vivo experiments' results demonstrated that the three-stage propulsion RHMH18 NPs presented superior tumor inhibition activity with minimal side effects, providing potential strategies of protein based drug delivery systems for tumor therapy.
Collapse
Affiliation(s)
- Mingyu Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Li Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Yanfei Cai
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Yang Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Lipeng Qiu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Yiting Shen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Jian Jin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Juan Zhou
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Jinghua Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
11
|
Sebak AA, Gomaa IEO, ElMeshad AN, Farag MH, Breitinger U, Breitinger HG, AbdelKader MH. Distinct Proteins in Protein Corona of Nanoparticles Represent a Promising Venue for Endogenous Targeting - Part II: In vitro and in vivo Kinetics Study. Int J Nanomedicine 2020; 15:9539-9556. [PMID: 33299308 PMCID: PMC7721286 DOI: 10.2147/ijn.s273721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/30/2020] [Indexed: 01/04/2023] Open
Abstract
Introduction Nanoparticles (NPs), upon introduction to the biological systems, become wrapped by serum and cellular proteins constituting the protein corona (PC). This PC contributes largely to the NPs’ interaction with the biological systems and their subsequent functions. On the one hand, PC can decrease the efficiency of targeting by directing the NPs to the reticuloendothelial system (RES) or by masking the active targeting moieties and decreasing their ability to bind to their target receptors. On the other hand, some components of PC have offered hopes for achieving endogenous targeting. Methods In this study, we aimed at the investigation of the role of the PC in determining the behavior of cRGDyk peptide-unconjugated and -conjugated NPs (uNPs and cNPs) exhibiting different physicochemical properties and their interaction with melanoma on in vitro and in vivo levels. Mathematical modeling has been utilized to understand the kinetics of the interaction of NPs with the tumor cells and different organs, respectively. Results Endocytosis and exocytosis were reported to occur simultaneously for the utilized NPs. The balance was largely dependent on the NPs’ physicochemical properties and the role of the PC. In addition, distinct proteins present in the PC (illustrated in the results of the PC analysis in part I) have also determined the patterns of the NPs’ distribution in different organs and tissues of the vascular system, the RES system and the target tumot tissue. Vitronectin (VN) was found to mediate higher accumulation in integrin receptor-expressing melanoma cells, while complement 3 protein (C3) and clusterin (CLU), as an opsonin and dysopsonin, respectively, regulated the balance between the RES uptake and blood circulation. Discussion PC, if properly modulated by tuning NPs’ physicochemical properties, can serve as a potential venue for optimum utilization of NPs in cancer therapy.
Collapse
Affiliation(s)
- Aya Ahmed Sebak
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Iman Emam Omar Gomaa
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Aliaa Nabil ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mahmoud Hussien Farag
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Ulrike Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Hans-Georg Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Mahmoud Hashem AbdelKader
- National Institute of Laser Enhanced Sciences (NILES), Cairo University (CU), Giza, Egypt.,European University in Egypt (EUE), New Administrative Capital, Cairo, Egypt
| |
Collapse
|
12
|
Sebak AA, Gomaa IEO, ElMeshad AN, Farag MH, Breitinger U, Breitinger HG, AbdelKader MH. Distinct Proteins in Protein Corona of Nanoparticles Represent a Promising Venue for Endogenous Targeting - Part I: In vitro Release and Intracellular Uptake Perspective. Int J Nanomedicine 2020; 15:8845-8862. [PMID: 33204091 PMCID: PMC7667594 DOI: 10.2147/ijn.s273713] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Protein corona (PC) deposition on nanoparticles (NPs) in biological systems contributes to a great extent to NPs' fates; their targeting potential, the interaction with different biological systems and the subsequent functions. PC - when properly tuned - can serve as a potential avenue for optimization of NPs' use in cancer therapy. Methods Poly-lactic co-glycolic acid (PLGA)-based NPs exhibiting different physicochemical properties were fabricated and characterized. The PC makeup of these NPs were qualitatively and quantitatively analyzed by Western blot and Bradford assay, respectively. The effect of PC on the release of NPs' cargos and the intracellular uptake into B16F10 melanoma cells has been studied. Results The composition of NPs (polymeric PLGA NPs vs lipid-polymer hybrid NPs) and the conjugation of an active targeting ligand (cRGDyk peptide) represented the major determinants of the PC makeup of NPs. The in vitro release of the loaded cargos from the NPs depended on the PC and the presence of serum proteins in the release medium. Higher cumulative release has been recorded in the presence of proteins in the case of peptide conjugated NPs, cNPs, while the unconjugated formulations, uNPs, showed an opposite pattern. NPs intracellular uptake studies revealed important roles of distinct serum and cellular proteins on the extent of NPs' accumulation in melanoma cells. For example, the abundance of vitronectin (VN) protein from serum has been positively related to the intracellular accumulation of the NPs. Conclusion Careful engineering of nanocarriers can modulate the recruitment of some proteins suggesting a potential use for achieving endogenous targeting to overcome the current limitations of targeted delivery of chemotherapeutic agents.
Collapse
Affiliation(s)
- Aya Ahmed Sebak
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Iman Emam Omar Gomaa
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Aliaa Nabil ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Mahmoud Hussien Farag
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Ulrike Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Hans-Georg Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Mahmoud Hashem AbdelKader
- National Institute of Laser Enhanced Sciences (NILES), Cairo University (CU), Giza, Egypt.,European University in Egypt (EUE), New Administrative Capital, Cairo, Egypt
| |
Collapse
|
13
|
Shetab Boushehri MA, Dietrich D, Lamprecht A. Nanotechnology as a Platform for the Development of Injectable Parenteral Formulations: A Comprehensive Review of the Know-Hows and State of the Art. Pharmaceutics 2020; 12:pharmaceutics12060510. [PMID: 32503171 PMCID: PMC7356945 DOI: 10.3390/pharmaceutics12060510] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Within recent decades, the development of nanotechnology has made a significant contribution to the progress of various fields of study, including the domains of medical and pharmaceutical sciences. A substantially transformed arena within the context of the latter is the development and production of various injectable parenteral formulations. Indeed, recent decades have witnessed a rapid growth of the marketed and pipeline nanotechnology-based injectable products, which is a testimony to the remarkability of the aforementioned contribution. Adjunct to the ability of nanomaterials to deliver the incorporated payloads to many different targets of interest, nanotechnology has substantially assisted to the development of many further facets of the art. Such contributions include the enhancement of the drug solubility, development of long-acting locally and systemically injectable formulations, tuning the onset of the drug’s release through the endowment of sensitivity to various internal or external stimuli, as well as adjuvancy and immune activation, which is a desirable component for injectable vaccines and immunotherapeutic formulations. The current work seeks to provide a comprehensive review of all the abovementioned contributions, along with the most recent advances made within each domain. Furthermore, recent developments within the domains of passive and active targeting will be briefly debated.
Collapse
Affiliation(s)
- Maryam A. Shetab Boushehri
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-736428; Fax: +49-228-735268
| | - Dirk Dietrich
- Department of Neurosurgery, University Clinic of Bonn, 53105 Bonn, Germany;
| | - Alf Lamprecht
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- PEPITE EA4267, Institute of Pharmacy, University Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
14
|
Wang H, Cheng L, Ma S, Ding L, Zhang W, Xu Z, Li D, Gao L. Self-Assembled Multiple-Enzyme Composites for Enhanced Synergistic Cancer Starving-Catalytic Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:20191-20201. [PMID: 32295337 DOI: 10.1021/acsami.0c02006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Inspired by the particularity of tumor microenvironments, including acidity and sensibility to reactive oxygen species (ROS), advanced and smart responsive nanomaterials have recently been developed. The present study synthesized tumor-targeted and pH-sensitive supramolecular micelles that self-assembled via host-guest recognition. The micelles consumed intratumoral glucose and lactate via loading with glucose oxidase (GOD) and lactate oxidase (LOD). Intratumoral glucose and lactate were converted into hydrogen peroxide (H2O2) and were sequentially reduced to highly toxic hydroxyl radicals (•OH) via the peroxidase (POD)-like activity of the loaded C-dot nanozymes. Tumor-killing effects were observed via cascade catalytic reactions. After an intravenous injection, the nanocomposite exhibited an excellent tumor-targeted ability with good biocompatibility, which demonstrated its effective antitumor effect. The nanocomposite effectively combined starvation and catalytic therapies and exerted a synergistic anticancer effect with minimal side effects and without external addition.
Collapse
Affiliation(s)
- Huihui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Lu Cheng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Shang Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Liming Ding
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Wei Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225001, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
15
|
Shi M, Zhang J, Huang Z, Chen Y, Pan S, Hu H, Qiao M, Chen D, Zhao X. Stimuli-responsive release and efficient siRNA delivery in non-small cell lung cancer by a poly(l-histidine)-based multifunctional nanoplatform. J Mater Chem B 2020; 8:1616-1628. [PMID: 32010914 DOI: 10.1039/c9tb02764e] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Small interfering RNA (siRNA) has extensive potential for the treatment of non-small cell lung cancer (NSCLC). While both cationic lipids and polymers have demonstrated promise to facilitate siRNA encapsulation, they can also hamper cytosolic siRNA release and induce severe cytotoxicity. To address these issues, a unique polymer hybrid nanoparticle (NP) nanoplatform was developed for multistage siRNA delivery based on both pH-responsive and endo/lysosomal escape characteristics, which was formed via a combination of an electrostatic interactions between the copolymer methoxy poly(ethylene glycol)-poly(l-histidine)-poly(sulfadimethoxine) (mPEG-PHis-PSD, shortened to PHD), dendritic poly-l-lysine (PLL) and PLK1 siRNA (shortened to siPLK1). The biological composition of the proton sponge effect polymer of the PHis chain, which was in position to make efficient endo/lysosomal escape, and the pH-responsive polymer of the PSD fragment, which could accelerate the release of siPLK1. In the present study, the NP illustrated excellent physiochemical properties and rapid endo/lysosomal escape in vitro. Besides this, compared with the PD/PLL/siRNA formulation, the PHD/PLL/siRNA NP indicated higher cellular uptake, and higher cell cytotoxicity in vitro. The in vivo results demonstrated that the PHD/PLL/siRNA NP exhibited the strongest tumor growth inhibition rate and ideal safety compared with the control and other siPLK1-treated formulations, which can be mainly attributed to pH-induced instantaneous dissociation and efficient endo/lysosomal escape arising from the PHD copolymer. Consequently, the above evidence indicates that the PHD/PLL/siRNA NP is a favorable gene delivery system and provides a potential strategy for siRNA delivery.
Collapse
Affiliation(s)
- Menghao Shi
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Jiulong Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Ziyuan Huang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Yuying Chen
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Shuang Pan
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Mingxi Qiao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| | - Xiuli Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Liaoning Province, China.
| |
Collapse
|
16
|
Ferro-Flores G. Targeted Nanomedicines: In the Right Route Towards Improved Therapies. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666181224144500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Guillermina Ferro-Flores
- Laboratorio Nacional de Investigacion y Desarrollo de Radiofarmacos CONACytT Instituto Nacional de Investigaciones Nucleares La Marquesa, 52750, Mexico
| |
Collapse
|
17
|
Xia Y, Tang G, Guo M, Xu T, Chen H, Lin Z, Li Y, Chen Y, Zhu B, Liu H, Cao J. Silencing KLK12 expression via RGDfC-decorated selenium nanoparticles for the treatment of colorectal cancer in vitro and in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110594. [PMID: 32204058 DOI: 10.1016/j.msec.2019.110594] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 02/07/2023]
Abstract
Short interfering RNA (siRNA) has been investigated as a promising modality of cancer treatment due to its capability to target specific target genes for downregulation. However, the successful application of this strategy depends on producing a safe and effective carrier system for delivering siRNA to the tumor. Thus, investigation of siRNA delivery carriers is a fundamental step in the field of siRNA-based therapeutics. In the current research, the surface of selenium nanoparticles (SeNPs) were modified with the tumor-targeted molecular RGDfC peptide with positive charge to synthetize the biocompatible siRNA carrier RGDfC-SeNPs. Subsequently, KLK12-siRNA was loaded onto the surface of RGDfC-SeNPs to create functionalized nanoparticles (RGDfC-Se@siRNA) that we tested for in vitro and in vivo antitumor efficacy. We measured significantly greater particle uptake in HT-29 colorectal cancer cells relative to HUVECs, providing evidence for the targeted delivery of RGDfC-Se@siRNA. We found that RGDfC-Se@siRNA could enter HT-29 cells primarily via clathrin-mediated endocytosis. Further, these particles experienced faster siRNA release in an acidic microenvironment compared to pH 7.4. The results from quantitative PCR and Western blot assays suggested that the target gene of KLK12 in HT-29 cells were obviously silenced by RGDfC-Se@siRNA. The further biological studies showed that treatment with RGDfC-Se@siRNA had ability to suppress the proliferation and migration/invasion of HT-29 cells, and triggered HT-29 cells apoptosis. RGDfC-Se@siRNA could induce the mitochondrial membrane potential (MMP) disruption and enhance the reactive oxygen species (ROS) generation in HT-29 cells, indicating that RGDfC-Se@siRNA induced the HT-29 cells apoptosis possibly by a ROS-mediated mitochondrial dysfunction pathway. Importantly, the in vivo antitumor study also verified that RGDfC-Se@siRNA could significantly suppress the growth of tumor in vivo. In addition, we did not observe any signs of systemic or tissue-specific toxicity after administration of RGDfC-Se@siRNA in mice. As a whole, these findings suggest that RGDfC-Se@siRNA has promising potential as a therapy for colorectal cancer.
Collapse
Affiliation(s)
- Yu Xia
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China.
| | - Guoyi Tang
- Department of Obstetrics Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Min Guo
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Tiantian Xu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Haiyang Chen
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Zhengfang Lin
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yinghua Li
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yi Chen
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Bing Zhu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Hongsheng Liu
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.
| |
Collapse
|
18
|
Hao T, Chen Q, Qi Y, Sun P, Chen D, Jiang W, Liu K, Sun H, Li L, Ding J, Li Z. Biomineralized Gd 2 O 3 @HSA Nanoparticles as a Versatile Platform for Dual-Modal Imaging and Chemo-Phototherapy-Synergized Tumor Ablation. Adv Healthc Mater 2019; 8:e1901005. [PMID: 31738019 DOI: 10.1002/adhm.201901005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/27/2019] [Indexed: 12/25/2022]
Abstract
A great challenge still remains to explore the facile approaches to construct multifunctional nanoparticles for acquiring precise cancer theranostics. Herein, a biocompatible theranostic nanoplatform capable of simultaneous cancer imaging and therapy is attempted by loading of paclitaxel (PTX) and indocyanine green (ICG) molecules into the matrix of Gd2 O3 @human serum albumin (HSA) nanoparticles (PIGH NPs) via hydrophobic interaction. The subsequent in vitro investigations reveal that the PIGH NPs afford uniform particle size, sustained drug release profile, strong longitudinal relaxivity, potent photothermal effect, effective singlet oxygen generation, and ideal resistance to photobleaching. Moreover, the PIGH NPs achieve high cellular uptake, efficient cytoplasmic drug translocation based on singlet oxygen-triggered endolysosomal disruption and prominent cytotoxicity effect against 4T1 cells under 808 nm near-infrared (NIR) irradiation in contrast to PTX/ICG-loaded HSA nanoparticles (PIH NPs) and free PTX/ICG. After intravenous injection, the PIGH NPs exhibit preferable tumor accumulation and achieve effective tumor ablation in 4T1 tumor bearing mouse model with excellent dual near-infrared fluorescence/magnetic resonance (NIRF/MR) imaging guided synergistic chemo-phototherapy. Hence, the PIGH NPs can be utilized as potential theranostic nanosystem for simultaneous cancer imaging and therapy.
Collapse
Affiliation(s)
- Tangna Hao
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
- Department of PharmacyThe Second Affiliated Hospital of Dalian Medical University Dalian 116011 Liaoning P. R. China
| | - Qixian Chen
- School of Life Science and BiotechnologyDalian University of Technology Dalian 116024 Liaoning P. R. China
| | - Yan Qi
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| | - Pengyuan Sun
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| | - Dawei Chen
- School of PharmacyShenyang Pharmaceutical University Shenyang 110016 Liaoning P. R. China
- School of PharmacyMedical College of Soochow University Suzhou 215123 Jiangsu P. R. China
| | - Weiwei Jiang
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| | - Kexin Liu
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| | - Huijun Sun
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| | - Lei Li
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences Changchun 130022 Jilin P. R. China
| | - Zhen Li
- School of PharmacyDalian Medical University Dalian 116044 Liaoning P. R. China
| |
Collapse
|
19
|
Nag OK, Delehanty JB. Active Cellular and Subcellular Targeting of Nanoparticles for Drug Delivery. Pharmaceutics 2019; 11:E543. [PMID: 31635367 PMCID: PMC6836276 DOI: 10.3390/pharmaceutics11100543] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 02/08/2023] Open
Abstract
Nanoparticle (NP)-mediated drug delivery (NMDD) for active targeting of diseases is a primary goal of nanomedicine. NPs have much to offer in overcoming the limitations of traditional drug delivery approaches, including off-target drug toxicity and the need for the administration of repetitive doses. In the last decade, one of the main foci in NMDD has been the realization of NP-mediated drug formulations for active targeted delivery to diseased tissues, with an emphasis on cellular and subcellular targeting. Advances on this front have included the intricate design of targeted NP-drug constructs to navigate through biological barriers, overcome multidrug resistance (MDR), decrease side effects, and improve overall drug efficacy. In this review, we survey advancements in NP-mediated drug targeting over the last five years, highlighting how various NP-drug constructs have been designed to achieve active targeted delivery and improved therapeutic outcomes for critical diseases including cancer, rheumatoid arthritis, and Alzheimer's disease. We conclude with a survey of the current clinical trial landscape for active targeted NP-drug delivery and how we envision this field will progress in the near future.
Collapse
Affiliation(s)
- Okhil K Nag
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Code 6900, 4555 Overlook Ave. SW, Washington, DC 20375, USA.
| | - James B Delehanty
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Code 6900, 4555 Overlook Ave. SW, Washington, DC 20375, USA.
| |
Collapse
|
20
|
Efficient fabrication of reversible pH-induced carboxymethyl chitosan nanoparticles for antitumor drug delivery under weakly acidic microenvironment. Int J Biol Macromol 2019; 126:68-73. [DOI: 10.1016/j.ijbiomac.2018.12.178] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/14/2022]
|
21
|
Sun X, Zhang J, Yang C, Huang Z, Shi M, Pan S, Hu H, Qiao M, Chen D, Zhao X. Dual-Responsive Size-Shrinking Nanocluster with Hierarchical Disassembly Capability for Improved Tumor Penetration and Therapeutic Efficacy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:11865-11875. [PMID: 30830746 DOI: 10.1021/acsami.8b21580] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
It is generally known that, for nanoparticles in cancer therapy, sufficient tumor penetration needs a minor particle size, while long in vivo circulation time needs a larger particle size. It is hard to balance them because they are standing on either side of a seesaw. To address these two different requirements, a dual-responsive size-shrinking nanocluster can self-adaptively respond to a complicated tumor microenvironment and transform its particulate property to overcome sequential in vivo barriers and reach a preferable antitumor activity. The nanocluster (RPSPT@SNCs) could preferentially accumulate into tumor tissue and dissociate under extracellular matrix metalloproteinase-2 (MMP-2) to release small-sized micelle formulations (RPSPTs). RPSPT possesses favorable tumor penetration and tumor targeting capability to deliver the antitumor agent paclitaxel (PTX) into deep regions of solid tumor. The intracellular redox microenvironment can also accelerate drug accumulation. The prepared RPSPT@SNCs possesses enhanced cell cytotoxicity and tumor penetration capability on MCF-7 cells and a favorable antitumor activity on the xenograft tumor mouse model.
Collapse
Affiliation(s)
- Xiaoyan Sun
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Jiulong Zhang
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Chunrong Yang
- College of Pharmacy , Jiamusi University , 148 Xuefu Street , Jiamusi 154007 , Heilongjiang , P.R. China
| | - Ziyuan Huang
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Menghao Shi
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Shuang Pan
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Haiyang Hu
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Mingxi Qiao
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Dawei Chen
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| | - Xiuli Zhao
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , P.R. China
| |
Collapse
|
22
|
Cheng Y, Ji Y. RGD-modified polymer and liposome nanovehicles: Recent research progress for drug delivery in cancer therapeutics. Eur J Pharm Sci 2018; 128:8-17. [PMID: 30471410 DOI: 10.1016/j.ejps.2018.11.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/31/2018] [Accepted: 11/21/2018] [Indexed: 12/19/2022]
Abstract
Over the past few decades, as the demand for cancer treatment has increased, more rational treatment options (considering size, mode of administration, biocompatibility, efficacy, etc.) and plenty of specifically active targeted nanovehicles have been developed. Integrin receptors targeting are one of the most frequently used approaches because of its highly expressed in cancer cells. In particular, the arginine-glycine-aspartic acid (RGD) peptide and its derivatives have been widely used as ligands for integrin to increase direct targeting capabilies. Polymers as well as liposomes are commonly used as nanovehicles for drug delivery. A variety of work is focused on the RGD-modified polymer and liposome nanovehicles for cancer therapeutics. The goal of this article is to review the published literature in recent years concerning the RGD-modified liposome and polymer nanovehicles to highlight its successful designs for improving cancer therapy and discuss the current challenges as well as the possible development prospects.
Collapse
Affiliation(s)
- Yu Cheng
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
23
|
Li J, Han Y, Lu Y, Song B, Zhao M, Hu H, Chen D. A novel disulfide bond-mediated cleavable RGD-modified PAMAM nanocomplex containing nuclear localization signal HMGB1 for enhancing gene transfection efficiency. Int J Nanomedicine 2018; 13:7135-7153. [PMID: 30464464 PMCID: PMC6228086 DOI: 10.2147/ijn.s182445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Polyamidoamine (PAMAM) dendrimers modified by polyethylene glycol (PEG) have frequently been investigated as a delivery carrier for gene therapy. However, modification of PAMAM with PEG using covalent linkage significantly reduces the cellular uptake rate and the transfection efficiency. How to conquer these barriers becomes a burning question in gene delivery. MATERIALS AND METHODS The present study constructed an effective disulfide bond-mediated cleavable RGD modified gene delivery system to overcome the aforementioned limitations. The disulfide bond was introduced between PAMAM dendrimers and PEG chains to realize the cleavage of PEG from the carrier system, whereas the arginine-glycine-aspartate (RGD) peptide was expected to promote the cellular uptake rate. A high mobility group Box 1 (HMGB1) protein containing nuclear localization signal (NLS) was simultaneously introduced to further promote gene expression efficiency. A pDNA/HMGB1/PAMAM-SS-PEG-RGD (DHP) nanocomplex was prepared via electrostatic interaction and characterized. RESULTS The results showed that DHP generated small particles and was able to condense and protect pDNA against degradation. In addition, the RGD peptide could significantly promote the cellular uptake of a nanocomplex. Intracellular trafficking and in vitro expression study indicated that the DHP nanocomplex escaped from lysosomes and the disulfide bonds between PAMAM and PEG cleaved due to the high concentration of GSH in the cytoplasm, pDNA consequently became exclusively located in the nucleus under the guidance of HMGB1, thereby promoting the red fluorescence protein (RFP) expression. Importantly, an in vivo antitumor activity study demonstrated that the DHP nanocomplex had higher antitumor activity than any other reference preparation. CONCLUSION All these results confirm that DHP could be a new strategy for improving the transfection and expression efficiency in gene delivery.
Collapse
Affiliation(s)
- Ji Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| | - Yuting Han
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| | - Yue Lu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| | - Baohui Song
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China, ;
| |
Collapse
|
24
|
Sun H, Dong Y, Feijen J, Zhong Z. Peptide-decorated polymeric nanomedicines for precision cancer therapy. J Control Release 2018; 290:11-27. [DOI: 10.1016/j.jconrel.2018.09.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/27/2018] [Accepted: 09/30/2018] [Indexed: 01/12/2023]
|
25
|
Yao P, Zhang Y, Meng H, Sun H, Zhong Z. Smart Polymersomes Dually Functionalized with cRGD and Fusogenic GALA Peptides Enable Specific and High-Efficiency Cytosolic Delivery of Apoptotic Proteins. Biomacromolecules 2018; 20:184-191. [DOI: 10.1021/acs.biomac.8b01243] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Peili Yao
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Hao Meng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. 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, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
26
|
Majumder P. Integrin-Mediated Delivery of Drugs and Nucleic Acids for Anti-Angiogenic Cancer Therapy: Current Landscape and Remaining Challenges. Bioengineering (Basel) 2018; 5:bioengineering5040076. [PMID: 30241287 PMCID: PMC6315429 DOI: 10.3390/bioengineering5040076] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/12/2018] [Accepted: 09/16/2018] [Indexed: 01/19/2023] Open
Abstract
Angiogenesis, sprouting of new blood vessels from pre-existing vasculatures, plays a critical role in regulating tumor growth. Binding interactions between integrin, a heterodimeric transmembrane glycoprotein receptor, and its extracellular matrix (ECM) protein ligands govern the angiogenic potential of tumor endothelial cells. Integrin receptors are attractive targets in cancer therapy due to their overexpression on tumor endothelial cells, but not on quiescent blood vessels. These receptors are finding increasing applications in anti-angiogenic therapy via targeted delivery of chemotherapeutic drugs and nucleic acids to tumor vasculatures. The current article attempts to provide a retrospective account of the past developments, highlight important contemporary contributions and unresolved set-backs of this emerging field.
Collapse
Affiliation(s)
- Poulami Majumder
- Division of Lipid Science and Technology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.
- Chemical Biology Laboratory, National Cancer Institute, 376 Boyles St, Frederick, MD 21702, USA.
| |
Collapse
|
27
|
Zhang J, Yang C, Pan S, Shi M, Li J, Hu H, Qiao M, Chen D, Zhao X. Eph A10-modified pH-sensitive liposomes loaded with novel triphenylphosphine-docetaxel conjugate possess hierarchical targetability and sufficient antitumor effect both in vitro and in vivo. Drug Deliv 2018. [PMID: 29513049 PMCID: PMC6058733 DOI: 10.1080/10717544.2018.1446475] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Mitochondrial-targeting therapy was considered to be a promising approach for the efficient treatment of cancer while positive charge induced nonspecific cytotoxicity severely limits its application. To overcome this drawback, a novel mitochondria targeted conjugate triphenylphosphine-docetaxel (TD) has been synthesized successfully and incorporated it into liposomes (EPSLP/TD), which possessed excellent pH-sensitive characteristic, EphA 10 mediated active targetability as well as mitochondria-targeting capability. EPSLP/TD was characterized to have a small particle size, high-encapsulation efficiency and excellent pH-sensitive characteristic. Compared with DTX-loaded liposomes (EPSLP/DTX), EPSLP/TD possessed higher cytotoxicity against MCF-7 cell line. Mitochondrial-targeting assay demonstrated mitochondria-targeting moiety triphenylphosphine (TPP) could efficiently deliver DTX to mitochondria. Western immunoblotting assay indicated that EPSLP/TD could efficiently deliver antitumor drug to mitochondria and induce cell apoptosis via mitochondria-mediated apoptosis pathway. In vivo antitumor study demonstrated EPSLP/TD owed excellent in vivo antitumor activity. Histological assay demonstrated EPSLP/TD showed strongly apoptosis inducing effect, anti-proliferation effect and anti-angiogenesis effect. This work investigated the potential of hierarchical targeting pH-sensitive liposomes is a suitable carrier to activate mitochondria-mediated apoptosis pathway for cancer therapy.
Collapse
Affiliation(s)
- Jiulong Zhang
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| | - Chunrong Yang
- b College Pharmacy , Jiamusi University , Jiamusi , Heilongjiang , PR China
| | - Shuang Pan
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| | - Menghao Shi
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| | - Jie Li
- c Mudanjiang Medical University , Mudanjiang , Heilongjiang , PR China
| | - Haiyang Hu
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| | - Mingxi Qiao
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| | - Dawei Chen
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| | - Xiuli Zhao
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , Liaoning , PR China
| |
Collapse
|
28
|
Wu W, Luo L, Wang Y, Wu Q, Dai HB, Li JS, Durkan C, Wang N, Wang GX. Endogenous pH-responsive nanoparticles with programmable size changes for targeted tumor therapy and imaging applications. Theranostics 2018; 8:3038-3058. [PMID: 29896301 PMCID: PMC5996358 DOI: 10.7150/thno.23459] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/06/2018] [Indexed: 12/20/2022] Open
Abstract
Nanotechnology-based antitumor drug delivery systems, known as nanocarriers, have demonstrated their efficacy in recent years. Typically, the size of the nanocarriers is around 100 nm. It is imperative to achieve an optimum size of these nanocarriers which must be designed uniquely for each type of delivery process. For pH-responsive nanocarriers with programmable size, changes in pH (~6.5 for tumor tissue, ~5.5 for endosomes, and ~5.0 for lysosomes) may serve as an endogenous stimulus improving the safety and therapeutic efficacy of antitumor drugs. This review focuses on current advanced pH-responsive nanocarriers with programmable size changes for anticancer drug delivery. In particular, pH-responsive mechanisms for nanocarrier retention at tumor sites, size reduction for penetrating into tumor parenchyma, escaping from endo/lysosomes, and swelling or disassembly for drug release will be highlighted. Additional trends and challenges of employing these nanocarriers in future clinical applications are also addressed.
Collapse
Affiliation(s)
- Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Qi Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Han-Bin Dai
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Jian-Shu Li
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Gui-Xue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| |
Collapse
|
29
|
Wen Y, Dong H, Wang K, Li Y, Li Y. Self-Templated, Green-Synthetic, Size-Controlled Protein Nanoassembly as a Robust Nanoplatform for Biomedical Application. ACS APPLIED MATERIALS & INTERFACES 2018; 10:11457-11466. [PMID: 29508606 DOI: 10.1021/acsami.7b19201] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Despite inherent advantages over their synthetic polymer-based counterparts, protein nanoparticles remain unsatisfactory in fabrication owing to their low size control, usage of toxic cross-linkers, or organic solvents. This partially contributes to the marginal benefits of Abraxane in clinics. Herein, a green-synthetic, size-controlled approach was developed to generate stable albumin nanoparticles. Physically packed ovalbumin nanoclusters were temporally formed in heat, which was then used as the template to form protein nanoparticles chemically stabilized by the intermolecular disulfide network. Exposure of embedded free thiols within a hydrophobic albumin structure and oxidation of them into disulfides (2-3 fold reduction of thiols groups in this process) were identified as key factors during the process. The fact that the structure was stable in sodium dodecyl sulfate treatment (hydrophobic destroyer) while disassembling fast in reduction condition (to cleave disulfide) validated the disulfide cross-linked mechanism. The developed approach is facile and reproducible with precision size control (from tens to hundreds of nanometers). The approach can be extended to other proteins such as bovine serum albumin, underscoring the potential universal applicability. Further study demonstrated that the resultant protein nanoparticles can be a robust nanoplatform for extensive biomedical applications including drug delivery (doxorubicin encapsulation of 5.7%), target bioconjugation, or robust immune adjuvant effect.
Collapse
Affiliation(s)
- Ya Wen
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO) , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Haiqing Dong
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO) , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Kun Wang
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO) , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Yan Li
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO) , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Yongyong Li
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO) , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| |
Collapse
|
30
|
Xia Y, Xu T, Wang C, Li Y, Lin Z, Zhao M, Zhu B. Novel functionalized nanoparticles for tumor-targeting co-delivery of doxorubicin and siRNA to enhance cancer therapy. Int J Nanomedicine 2017; 13:143-159. [PMID: 29317822 PMCID: PMC5743186 DOI: 10.2147/ijn.s148960] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human homeobox protein (Nanog) is highly expressed in most cancer cells and has gradually emerged as an excellent target in cancer therapy, owing to its regulation of cancer cell proliferation, metastasis and apoptosis. In this study, we prepared tumor-targeting functionalized selenium nanoparticles (RGDfC-SeNPs) to load chemotherapeutic doxorubicin (DOX) and Nanog siRNA. Herein, RGDfC peptide was used as a tumor-targeting moiety which could specifically bind to αvβ3 integrins overexpressed on various cancer cells. The sizes of RGDfC-SeNPs@DOX nanoparticles (~12 nm) were confirmed by both dynamic light scattering and transmission electron microscopy. The chemical structure of RGDfC-SeNPs@DOX was characterized via Fourier-transform infrared spectroscopy. The RGDfC-SeNPs@DOX was compacted with siRNA (anti-Nanog) by electrostatic interaction to fabricate the RGDfC-SeNPs@DOX/siRNA complex. The RGDfC-SeNPs@DOX/siRNA complex nanoparticles could efficiently enter into HepG2 cells via clathrin-associated endocytosis, and showed high gene transfection efficiency that resulted in enhanced gene silencing. The in vivo biodistribution experiment indicated that RGDfC-SeNPs@DOX/siRNA nanoparticles were capable of specifically accumulating in the tumor site. Furthermore, treatment with RGDfC-SeNPs@DOX/siRNA resulted in a more significant anticancer activity than the free DOX, RGDfC-SeNPs@DOX or RGDfC-SeNPs/siRNA in vitro and in vivo. In summary, this study shows a novel type of DOX and siRNA co-delivery system, thereby providing an alternative route for cancer treatment.
Collapse
Affiliation(s)
- Yu Xia
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Tiantian Xu
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Changbing Wang
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Yinghua Li
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Zhengfang Lin
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Mingqi Zhao
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Bing Zhu
- Central Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
31
|
Duro-Castano A, Gallon E, Decker C, Vicent MJ. Modulating angiogenesis with integrin-targeted nanomedicines. Adv Drug Deliv Rev 2017; 119:101-119. [PMID: 28502767 DOI: 10.1016/j.addr.2017.05.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
Targeting angiogenesis-related pathologies, which include tumorigenesis and metastatic processes, has become an attractive strategy for the development of efficient guided nanomedicines. In this respect, integrins are cell-adhesion molecules involved in angiogenesis signaling pathways and are overexpressed in many angiogenic processes. Therefore, they represent specific biomarkers not only to monitor disease progression but also to rationally design targeted nanomedicines. Arginine-glycine-aspartic (RGD) containing peptides that bind to specific integrins have been widely utilized to provide ligand-mediated targeting capabilities to small molecules, peptides, proteins, and antibodies, as well as to drug/imaging agent-containing nanomedicines, with the final aim of maximizing their therapeutic index. Within this review, we aim to cover recent and relevant examples of different integrin-assisted nanosystems including polymeric nanoconstructs, liposomes, and inorganic nanoparticles applied in drug/gene therapy as well as imaging and theranostics. We will also critically address the overall benefits of integrin-targeting.
Collapse
Affiliation(s)
- Aroa Duro-Castano
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Elena Gallon
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Caitlin Decker
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
32
|
Sun Y, Kang C, Liu F, Zhou Y, Luo L, Qiao H. RGD Peptide-Based Target Drug Delivery of Doxorubicin Nanomedicine. Drug Dev Res 2017; 78:283-291. [PMID: 28815721 DOI: 10.1002/ddr.21399] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preclinical Research Doxorubicin (DOX) is commonly used for the treatment of breast cancer and lymphoma. However, its clinical use has been severely limited due to cardiotoxicity, requiring the development of safer and more efficient pharmaceutical formulations of DOX. Advances in nanotechnology have provided new ways to administer chemotherapeutic drugs like DOX are conveyed into the body and to tumor sites. These Nanotechnology approaches have aided in the selective accumulation of DOX into tumor sites via the enhanced permeability and retention. However, the absence of active targeting ligands still hinders the effective delivery of DOX. Among all active targeting ligands developed to date, RGD peptide (Arginylglycylaspartic acid) occupies a unique position owing to its inherent safety, biocompatibility, and targeting ability. Accordingly, modification of DOX with RGD ligand is anticipated to improve transport of DOX into tumor cells. In this review, we discuss using RGD peptide for improving the therapeutic efficacy of DOX nanomedicine. Drug Dev Res 78 : 283-291, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, California, 95758
| | - Chen Kang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242
| | - Fei Liu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - You Zhou
- College of Biotechnology, Southwest University, Chongqing, 400715, China
| | - Lei Luo
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Hongzhi Qiao
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
33
|
Xia Y, Lin Z, Li Y, Zhao M, Wang C, Guo M, Zhang B, Zhu B. Targeted delivery of siRNA using RGDfC-conjugated functionalized selenium nanoparticles for anticancer therapy. J Mater Chem B 2017; 5:6941-6952. [PMID: 32264343 DOI: 10.1039/c7tb01315a] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lack of biocompatible and effective delivery carriers is a significant shortcoming for siRNA-mediated cancer therapy. To overcome these limitations, selenium nanoparticles (SeNPs) have been proposed for siRNA transfection vehicles. In this study, we synthesized novel RGDfC peptide modified selenium nanoparticles (RGDfC-SeNPs) as a gene vehicle, which was expected to improve the tumor-targeted delivery activity. RGDfC-SeNPs were compacted with siRNAs (anti-Oct4) by electrostatic interaction, which was capable of protecting siRNA from degradation. RGDfC-SeNPs exhibited excellent ability to deliver siRNA into HepG2 cells. siRNA transfection assay showed that RGDfC-SeNPs presented a higher gene silencing efficacy than conventional lipofectamine 2000. The cytotoxicity of RGDfC-SeNPs/siRNA on normal cells was lower than that on tumor cells, indicating that RGDfC-SeNPs/siRNA exhibited selectivity between normal and cancer cells. Additionally, Oct4 knockdown mediated by the selenium nanoparticle transfection arrested HepG2 cells mainly at the G2/M phase and significantly induced HepG2 cell apoptosis. Western blotting results showed that RGDfC-SeNPs/siRNA might trigger Wnt/β-catenin signaling, and further activate a BCL-2 apoptosis-related signaling pathway to advance HepG2 cell apoptosis. In vivo biodistribution experiments indicated that RGDfC-SeNPs/siRNA nanoparticles were specifically targeted to the HepG2 tumors. Most importantly, RGDfC-SeNPs/siRNA inhibited tumor growth significantly and induced HepG2 cell apoptosis via silencing the Oct4 gene. In addition, the results of H&E staining demonstrated that RGDfC-SeNPs/siRNA had negligible toxicity on the major organs of mice. In a word, this study provides a novel strategy for the design of biocompatible and effective siRNA delivery vehicles in cancer therapy.
Collapse
Affiliation(s)
- Yu Xia
- Central Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Chen WL, Li F, Tang Y, Yang SD, Li JZ, Yuan ZQ, Liu Y, Zhou XF, Liu C, Zhang XN. Stepwise pH-responsive nanoparticles for enhanced cellular uptake and on-demand intracellular release of doxorubicin. Int J Nanomedicine 2017; 12:4241-4256. [PMID: 28652730 PMCID: PMC5473598 DOI: 10.2147/ijn.s129748] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Physicochemical properties, including particle size, zeta potential, and drug release behavior, affect targeting efficiency, cellular uptake, and antitumor effect of nanocarriers in a formulated drug-delivery system. In this study, a novel stepwise pH-responsive nanodrug delivery system was developed to efficiently deliver and significantly promote the therapeutic effect of doxorubicin (DOX). The system comprised dimethylmaleic acid-chitosan-urocanic acid and elicited stepwise responses to extracellular and intracellular pH. The nanoparticles (NPs), which possessed negative surface charge under physiological conditions and an appropriate nanosize, exhibited advantageous stability during blood circulation and enhanced accumulation in tumor sites via enhanced permeability and retention effect. The tumor cellular uptake of DOX-loaded NPs was significantly promoted by the first-step pH response, wherein surface charge reversion of NPs from negative to positive was triggered by the slightly acidic tumor extracellular environment. After internalization into tumor cells, the second-step pH response in endo/lysosome acidic environment elicited the on-demand intracellular release of DOX from NPs, thereby increasing cytotoxicity against tumor cells. Furthermore, stepwise pH-responsive NPs showed enhanced antiproliferation effect and reduced systemic side effect in vivo. Hence, the stepwise pH-responsive NPs provide a promising strategy for efficient delivery of antitumor agents.
Collapse
Affiliation(s)
- Wei-Liang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Fang Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Yan Tang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Shu-di Yang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Ji-Zhao Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Zhi-Qiang Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Yang Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Xiao-Feng Zhou
- Department of Ultrasound, Changshu Hospital of Traditional Chinese Medicine, Changshu
| | - Chun Liu
- Department of Pharmacy, The Hospital of Suzhou People's Hospital Affiliated to Nanjing Medical University, Suzhou, People's Republic of China
| | - Xue-Nong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| |
Collapse
|
35
|
Zhou H, Sun H, Lv S, Zhang D, Zhang X, Tang Z, Chen X. Legumain-cleavable 4-arm poly(ethylene glycol)-doxorubicin conjugate for tumor specific delivery and release. Acta Biomater 2017; 54:227-238. [PMID: 28315495 DOI: 10.1016/j.actbio.2017.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 10/20/2022]
Abstract
Traditional chemotherapy strategy exists undesirable toxic side-effects to normal tissues due to the low selectively to cancer cells of micromolecule cytotoxic drugs. One considered method to realizing the targeted delivery and increasing the specificity to tumor tissues of the cytotoxic drug is to transporting and discharging it through an environment-sensitive mechanism. In this study, a novel enzyme-sensitive polymer-doxorubicin conjugate was designed to delivery chemotherapeutic drug in a tumor-specific behavior and selectively activated in tumor tissue. Briefly, doxorubicin (DOX) was conjugated to carboxyl-terminated 4-arm poly(ethylene glycol) through a tetrapeptide linker, alanine-alanine-asparagine-leucine (AANL), which was one of the substrates of legumain, an asparaginyl endopeptidase that was found presented in plants, mammals and also highly expressed in human tumor tissues. Hereinafter, the polymer-DOX conjugate was termed as 4-arm PEG-AANL-DOX. Dynamic laser scattering (DLS) and transmission electron microscopy (TEM) measurements indicated that the 4-arm PEG-AANL-DOX could self-assemble into micelles in aqueous solution. Drug release and in vitro cytotoxicity studies revealed that the 4-arm PEG-AANL-DOX could be cleaved by legumain. Ex vivo DOX fluorescence imaging measurements demonstrated that the 4-arm PEG-AANL-DOX had an improved tumor-targeting delivery as compared with the free DOX·HCl. In vivo studies on nude mice bearing MDA-MB-435 tumors revealed that the 4-arm PEG-AANL-DOX had a comparable anticancer efficacy with the free DOX·HCl but without DOX-related toxicities to normal tissues as measured by body weight change and histological assessments, indicating that the 4-arm PEG-AANL-DOX had an improved therapeutic index for cancer therapy. STATEMENT OF SIGNIFICANCE Herein we describe the construction of a novel tumor environment-sensitive delivery system through the instruction of a legumain-cleavable linkage to a polymer-DOX conjugate (4-arm PEG-AANL-DOX). This particular design strategy allows for polymer-DOX conjugates to be delivered in a tumor-specific manner and selectively activable in tumor microenvironment so that it can combine the advantages of tumor-specific delivery and tumor intracellular microenvironment-triggered release systems.
Collapse
|
36
|
Chen Y, Li H, Deng Y, Sun H, Ke X, Ci T. Near-infrared light triggered drug delivery system for higher efficacy of combined chemo-photothermal treatment. Acta Biomater 2017; 51:374-392. [PMID: 28088668 DOI: 10.1016/j.actbio.2016.12.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 11/28/2016] [Accepted: 12/05/2016] [Indexed: 12/28/2022]
Abstract
The combination of chemotherapy and photothermal therapy is a promising strategy for cancer treatment. In the present study, indocyanine green (ICG), a widely used near-infrared (NIR) dye in photothermal therapy, and chemotherapeutic drug-doxorubicin (DOX) were loaded within the nanoparticles of novel designed arylboronic ester and cholesterol modified hyaluronic acid (PPE-Chol1-HA), denoted as PCH-DI. We take advantage of reactive oxygen species (ROS) production capability of ICG and ROS-sensitivity of arylboronic ester to realize controllable drug release. It was confirmed that PCH-DI exhibited remarkable photothermal effect and light-triggered faster release of DOX with NIR laser irradiation. DOX in PCH-DI/Laser group exhibited the most efficient nucleus binding toward HCT-116 colon cells in vitro. Furthermore, enhanced cytotoxicity and promoted tumor growth suppression effect of PCH-DI on HCT-116 tumor xenograft nude mice and AOM-induced murine orthotopic colorectal cancer model was achieved under NIR laser irradiation. Thus, the co-delivery system based on PCH appears to be a promising platform for the combined chemo-photothermal therapy in tumor treatment. STATEMENT OF SIGNIFICANCE In case of chemo-photothermal combination therapy, the synchronism of treatments plays an important role in achieving expected antitumor efficiency. In this study, a light triggered ROS mediated drug delivery system was developed with the help of ROS-sensitive moieties of arylboronic ester and ROS producer of ICG. We innovatively make use of the ROS production capability of ICG under NIR laser irradiation to promote a faster release of DOX resulting from swelling of PCH-DI due to the presence of arylboronic ester. Intracellular ROS detection demonstrated that ROS level of PCH-I increased under irradiation. Moreover, the faster release behavior of DOX from PCH-DI with NIR laser irradiation was confirmed by the in vitro drug release and cellular uptake study. Meanwhile, local hyperthermia was verified by photothermal effect tests. Therefore, the synchronism of the combination therapy was achieved via light triggered faster release of DOX (chemo-therapy) and local hyperthermia (thermal-therapy) using PCH-DI under irradiation. It was reasonable to attribute the efficient anti-tumor efficiency of PCH-DI both in vitro and in vivo to the enhanced synergistic effect of chemo-photothermal combination therapy with realization of synchronism. To this end, this novel co-delivery system has provided a promising solution for achieving the synchronism of treatment to strengthen the efficiency of combination therapy.
Collapse
|
37
|
Systematic evaluation of multifunctional paclitaxel-loaded polymeric mixed micelles as a potential anticancer remedy to overcome multidrug resistance. Acta Biomater 2017; 50:381-395. [PMID: 27956367 DOI: 10.1016/j.actbio.2016.12.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/20/2016] [Accepted: 12/07/2016] [Indexed: 01/13/2023]
Abstract
Multidrug resistance (MDR) of tumor cells is becoming the main reason for the failure of chemotherapy and P-glycoprotein (P-gp) mediated drug efflux has demonstrated to be the key factor for MDR. To address this issue, a novel pH-responsive mixed micelles drug delivery system composed of dextran-g-poly(lactide-co-glycolide)-g-histidine (HDP) and folate acid-D-α-tocopheryl polyethylene glycol 2000 (FA-TPGS2K) copolymers has been designed for the delivery of antitumor agent, paclitaxel (PTX) via FA-receptor mediated cell endocytosis, into PTX-resistant breast cancer MCF-7 cells (MCF-7/PTX). PTX-loaded FA-TPGS2K/HDP mixed micelles were characterized to have a small size distribution, high loading content and excellent pH-responsive drug release profiles. Compared with HDP micelles, FA-TPGS2K/HDP mixed micelles showed a higher cytotoxicity against MCF-7 and MCF-7/PTX cells due to the synergistic effect of FA-receptor mediated cell endocytosis, pH-responsive drug release and TPGS mediated P-gp inhibition. P-gp expression level, ATP content and mitochondrial membrane potential change have been measured, the results indicated blank FA-TPGS2K/HDP mixed micelles could inhibit the P-gp activity by reducing the mitochondrial membrane potential and depleting ATP content but not down-regulating the P-gp expression. In vivo antitumor activities demonstrated FA-TPGS2K/HDP mixed micelles could reach higher antitumor activity compared with HDP micelles for MCF-7/PTX tumor cells. Histological assay also indicated that FA-TPGS2K/HDP mixed micelles showed strongly apoptosis inducing effect, anti-proliferation effect and anti-angiogenesis effect. All these evidences demonstrated this pH-sensitive FA-TPGS2K/HDP micelle-based drug delivery system is a promising approach for overcoming MDR. STATEMENT OF SIGNIFICANCE In this work, a novel FA-TPGS2K copolymer has been synthesized and used it to construct mixed micelles with HDP copolymer to overcome MDR effect. Furthermore, a series in vitro and in vivo evaluations have been made, which supported enough evidences for the efficient delivery of antitumor drug to MDR cells.
Collapse
|
38
|
Cordeiro RA, Santo D, Farinha D, Serra A, Faneca H, Coelho JFJ. High transfection efficiency promoted by tailor-made cationic tri-block copolymer-based nanoparticles. Acta Biomater 2017; 47:113-123. [PMID: 27744070 DOI: 10.1016/j.actbio.2016.10.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 10/03/2016] [Accepted: 10/10/2016] [Indexed: 12/25/2022]
Abstract
Cationic polymer-based vectors have been considered a promising strategy in gene therapy area due to their inherent ability to condense genetic material and successfully transfect cells. However, they usually exhibit high cytotoxicity. In this work, it is proposed the use of a tailor-made gene carrier based on a tri-block copolymer of poly[2-(dimethylamino)ethyl methacrylate] and poly(β-amino ester) (PDMAEMA-b-PβAE-b-PDMAEMA), the influence of the PβAE length being assessed. For this purpose, three different block copolymers were prepared varying the molecular weight of this segment. The obtained materials were characterized by NMR and SEC analyzes. Different polyplexes formulations were prepared and evaluated in terms of physicochemical characterization (ethidium bromide intercalation assay, agarose gel electrophoresis assay, dynamic light scattering, zeta potential analyzes and atomic force microscopy) and biological activity (cytotoxicity, and luciferase and green fluorescent protein expression in Hela and COS-7 cell lines). Among the developed nanosystems, the best polyplex formulation revealed between 40- and 60-fold higher transgene expression, in HeLa and COS-7 cell lines, and much lower cytotoxicity than that observed with branched PEI and TurboFect™. Moreover, these nanosystems present suitable physicochemical properties for gene delivery namely reduced mean diameter and high DNA protection. The results reported here show the enormous potential of this block copolymer as gene carrier. STATEMENT OF SIGNIFICANCE Syntheses of PDMAEMA-b-PβAE-b-PDMAEMA block copolymers for an extremely effective non-viral vector. The block copolymer PDMAEMA3000-b-PβAE12000-b-PDMAEMA3000-based polyplexes at 100/1N/P ratio exhibits between 40- and 60-fold higher transgene expression in HeLa and COS-7 cell lines than commonly used polymeric non-viral vectors, namely branched PEI (known as the gold standard) and TurboFect™ (commercial available).
Collapse
Affiliation(s)
- Rosemeyre A Cordeiro
- CEMUC, Department of Chemical Engineering, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Daniela Santo
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Dina Farinha
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Arménio Serra
- CEMUC, Department of Chemical Engineering, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Henrique Faneca
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal.
| | - Jorge F J Coelho
- CEMUC, Department of Chemical Engineering, University of Coimbra, 3030-790 Coimbra, Portugal.
| |
Collapse
|
39
|
Ng KE, Amin MCIM, Katas H, Amjad MW, Butt AM, Kesharwani P, Iyer AK. pH-Responsive Triblock Copolymeric Micelles Decorated with a Cell-Penetrating Peptide Provide Efficient Doxorubicin Delivery. NANOSCALE RESEARCH LETTERS 2016; 11:539. [PMID: 27921280 PMCID: PMC5138181 DOI: 10.1186/s11671-016-1755-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 11/25/2016] [Indexed: 05/10/2023]
Abstract
This study developed novel triblock pH-responsive polymeric micelles (PMs) using cholic acid-polyethyleneimine-poly-L-arginine (CA-PEI-pArg) copolymers. PEI provided pH sensitivity, while the hydrophilic cell-penetrating pArg peptide promoted cellular PM internalization. The copolymers self-assembled into PMs in aqueous solution at above the critical micelle concentration (2.98 × 10-7 M) and encapsulated doxorubicin in the core region, with a 34.2% (w/w) entrapment efficiency. PMs showed pH-dependent swelling, increasing in size by almost sevenfold from pH 7.4 to 5.0. Doxorubicin release was pH-dependent, with about 65% released at pH 5.0, and 32% at pH 7.4. Cellular uptake, assessed by confocal microscopy and flow cytometry, was enhanced by using doxorubicin-loaded CA-PEI-pArg PMs, as compared to free doxorubicin and DOX-loaded CA-PEI PMs. Moreover, 24-h incubation of these PMs with a human breast cancer cell line produced greater cytotoxicity than free doxorubicin. These results indicate that pH-responsive CA-PEI-pArg micelles could provide a versatile delivery system for targeted cancer therapy using hydrophobic drugs. Graphical of CA-PEI-pArg polymeric micelles as a pH-responsive drug delivery system.
Collapse
Affiliation(s)
- Khen Eng Ng
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia.
| | - Haliza Katas
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Muhammad Wahab Amjad
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Adeel Masood Butt
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Arun K Iyer
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| |
Collapse
|
40
|
Chen Q, Long M, Qiu L, Zhu M, Li Z, Qiao M, Hu H, Zhao X, Chen D. Decoration of pH-sensitive copolymer micelles with tumor-specific peptide for enhanced cellular uptake of doxorubicin. Int J Nanomedicine 2016; 11:5415-5427. [PMID: 27799766 PMCID: PMC5077130 DOI: 10.2147/ijn.s111950] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
To improve the targeting efficacy of hyaluronic acid (HA)-based micelles, pH-sensitive mixed micelles based on HA-g-poly(L-histidine) (PHis) and d-α-tocopheryl polyethylene glycol 2000 copolymers were prepared and decorated with human epidermal growth factor receptor 2 (Her2) peptide, a tumor cell-specific peptide ligand, on their surface. The doxorubicin-loaded micelles (HA-PHis/peptide–d-α-tocopheryl polyethylene glycol 2000 mixed micelles [PHTM]) were characterized to have a unimodal size distribution and pH-dependent drug release pattern. In vitro tumor targeting studies demonstrated that PHTM exhibited the pronounced cytotoxicity and efficient internalization in MDA-MB-231 cells overexpressing CD44 and Her2 receptors. In vivo investigation into micelles in MDA-MB-231 tumor-bearing mice confirmed that PTHM could reach the tumor site more effectively and exert excellent tumor killing activity. In general, Her2 peptide decoration can enhance the selective cytotoxicity and antitumor activity of HA-based micelles.
Collapse
Affiliation(s)
- Qing Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Miaomiao Long
- Department of Research and Development, Nanjing Chia Tai Tianqing Pharmaceutical Group Co. Ltd, Nanjing
| | - Lipeng Qiu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, People's Republic of China
| | - Mengqin Zhu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, People's Republic of China
| | - Zhen Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Mingxi Qiao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Xiuli Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| |
Collapse
|
41
|
Alam S, Panda JJ, Mukherjee TK, Chauhan VS. Short peptide based nanotubes capable of effective curcumin delivery for treating drug resistant malaria. J Nanobiotechnology 2016; 14:26. [PMID: 27044333 PMCID: PMC4820878 DOI: 10.1186/s12951-016-0179-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/23/2016] [Indexed: 01/14/2023] Open
Abstract
Background Curcumin (Ccm) has shown immense potential as an antimalarial agent; however its low solubility and less bioavailability attenuate the in vivo efficacy of this potent compound. In order to increase Ccm’s bioavailability, a number of organic/inorganic polymer based nanoparticles have been investigated. However, most of the present day nano based delivery systems pose a conundrum with respect to their complex synthesis procedures, poor in vivo stability and toxicity issues. Peptides due to their high biocompatibility could act as excellent materials for the synthesis of nanoparticulate drug delivery systems. Here, we have investigated dehydrophenylalanine (ΔPhe) di-peptide based self-assembled nanoparticles for the efficient delivery of Ccm as an antimalarial agent. The self-assembly and curcumin loading capacity of different ΔPhe dipeptides, phenylalanine–α,β-dehydrophenylalanine (FΔF), arginine-α,β-dehydrophenylalanine (RΔF), valine-α,β-dehydrophenylalanine (VΔF) and methonine-α,β-dehydrophenylalanine (MΔF) were investigated for achieving enhanced and effective delivery of the compound for potential anti-malarial therapy. Results FΔF, RΔF, VΔF and MΔF peptides formed different types of nanoparticles like nanotubes and nanovesicles under similar assembling conditions. Out of these, F∆F nanotubes showed maximum curcumin loading capacity of almost 68 % W/W. Ccm loaded F∆F nanotubes (Ccm-F∆F) showed comparatively higher (IC50, 3.0 µM) inhibition of Plasmodium falciparum (Indo strain) as compared to free Ccm (IC50, 13 µM). Ccm-F∆F nano formulation further demonstrated higher inhibition of parasite growth in malaria infected mice as compared to free Ccm. The dipeptide nanoparticles were highly biocompatible and didn’t show any toxic effect on mammalian cell lines and normal blood cells. Conclusion This work provides a proof of principle of using highly biocompatible short peptide based nanoparticles for entrapment and in vivo delivery of Ccm leading to an enhancement in its efficacy as an antimalarial agent.
Collapse
Affiliation(s)
- Shadab Alam
- International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Mohali, Punjab, 160062, India
| | | | - Virander Singh Chauhan
- International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.
| |
Collapse
|
42
|
Zhang J, Luo Y, Zhao X, Li X, Li K, Chen D, Qiao M, Hu H, Zhao X. Co-delivery of doxorubicin and the traditional Chinese medicine quercetin using biotin–PEG2000–DSPE modified liposomes for the treatment of multidrug resistant breast cancer. RSC Adv 2016. [DOI: 10.1039/c6ra24173e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
At present, multidrug resistance (MDR) in cancer therapy is an international problem, which is caused mostly by the overexpressed P-glycoprotein (P-gp) efflux pump.
Collapse
Affiliation(s)
- Jiulong Zhang
- Department of Traditional Chinese Materia Medica
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Yue Luo
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Xiufeng Zhao
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Xiaowei Li
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Kexin Li
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Dawei Chen
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Mingxi Qiao
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Haiyang Hu
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| | - Xiuli Zhao
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- P. R. China
| |
Collapse
|