51
|
Zhang X, Qiu M, Guo P, Lian Y, Xu E, Su J. Autologous Red Blood Cell Delivery of Betamethasone Phosphate Sodium for Long Anti-Inflammation. Pharmaceutics 2018; 10:pharmaceutics10040286. [PMID: 30567356 PMCID: PMC6320894 DOI: 10.3390/pharmaceutics10040286] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 12/30/2022] Open
Abstract
Although glucocorticoids are highly effective in treating various types of inflammation such as skin disease, rheumatic disease, and allergic disease, their application have been seriously limited for their high incidence of side effects, particularly in long term treatment. To improve efficacy and reduce side effects, we encapsulated betamethasone phosphate (BSP) into biocompatible red blood cells (RBCs) and explored its long acting-effect. BSP was loaded into rat autologous erythrocytes by hypotonic preswelling method, and the loading amount was about 2.5 mg/mL cells. In vitro, BSP loaded RBCs (BSP-RBCs) presented similar morphology, osmotic fragility to native RBCs (NRBCs). After the loading process, the loaded cells can maintain around 70% of Na+/K+-ATPase activity of natural cells. In vivo, a series of tests including survival, pharmacokinetics, and anti-inflammatory effect were carried out to examine the long-acting effect of BSP-RBCs. The results shown that the loaded cells could circulate in plasma for over nine days, the release of BSP can last for over seven days and the anti-inflammatory effect can still be observed on day 5 after injection. Totally, BSP-loaded autologous erythrocytes seem to be a promising sustained releasing delivery system with long anti-inflammatory effect.
Collapse
Affiliation(s)
- Xiumei Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Pengcheng Guo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yumei Lian
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Enge Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
52
|
Yang W, Wei Y, Yang L, Zhang J, Zhong Z, Storm G, Meng F. Granzyme B-loaded, cell-selective penetrating and reduction-responsive polymersomes effectively inhibit progression of orthotopic human lung tumor in vivo. J Control Release 2018; 290:141-149. [PMID: 30312720 DOI: 10.1016/j.jconrel.2018.10.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/30/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
The clinical use of protein therapeutics with intracellular targets is hampered by its in vivo fragility and low cell permeability. Here, we report that cell-selective penetrating and reduction-responsive polymersomes (CPRPs) mediate high-efficiency targeted delivery of granzyme B (GrB) to orthotopic human lung tumor in vivo. Model protein studies using FITC-labeled cytochrome C (FITC-CC) revealed efficient and high protein loading up to 17.2 wt% for CPRPs. FITC-CC-loaded CPRPs exhibited a small size of 82-90 nm, reduction-responsive protein release, as well as greatly enhanced internalization and cytoplasmic protein release in A549 lung cancer cells compared with the non-targeted FITC-CC-loaded RPs control. GrB-loaded CPRPs showed a high potency toward A549 lung cancer cells with a half maximal inhibitory concentration (IC50) of 20.7 nM. Under the same condition, free GrB was essentially non-toxic. Importantly, installing cell-selective penetrating peptide did not alter the circulation time but did enhance tumor accumulation of RPs. Orthotopic A549-Luc lung tumor-bearing nude mice administered with GrB-loaded CPRPs at a dosage of 2.88 nmol GrB equiv./kg showed complete tumor growth inhibition with little body weight loss throughout the treatment period, resulting in significantly improved survival rate over the non-targeted and non-treated controls. These cell-selective penetrating and reduction-responsive polymersomes provide a targeted protein therapy for cancers.
Collapse
Affiliation(s)
- Weijing Yang
- 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, PR China
| | - Yaohua Wei
- 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, PR China; Department of Targeted Therapeutics, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, Enschede 7500AE, The Netherlands
| | - Liang Yang
- 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, PR China
| | - Jian 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, PR 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, PR China.
| | - Gert Storm
- Department of Targeted Therapeutics, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, Enschede 7500AE, The Netherlands
| | - Fenghua 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, PR China.
| |
Collapse
|
53
|
Liu X, Wang C, Liu Z. Protein-Engineered Biomaterials for Cancer Theranostics. Adv Healthc Mater 2018; 7:e1800913. [PMID: 30260583 DOI: 10.1002/adhm.201800913] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/19/2018] [Indexed: 12/18/2022]
Abstract
Proteins are an important class of biomaterials promising a variety of applications such as drug delivery, and imaging or therapy, owing to their biodegradability, biocompatibility, as well as inherent biological activities acting as enzymes, recognizing molecules, or therapeutics by themselves. Over the few past decades, different types of proteins with desired properties have been widely explored for biomedical applications. Many therapeutic proteins have now entered clinical use. This review therefore summarizes various strategies in the engineering of biomaterials for delivery of therapeutic proteins, as well as the recent development of protein-based biomaterials for cancer theranostics.
Collapse
Affiliation(s)
- Xiaowen Liu
- Pharmacology; Department of Basic Medical Sciences; Faculty of Medical Science; Jinan University; Guangzhou Guangdong 510632 China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM); Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices; Soochow University; Suzhou Jiangsu 215123 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM); Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices; Soochow University; Suzhou Jiangsu 215123 China
| |
Collapse
|
54
|
Lang T, Yin Q, Li Y. Progress of Cell-Derived Biomimetic Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800053] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tianqun Lang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; 501 Haike Road Shanghai 201203 China
- School of Pharmacy; University of Chinese Academy of Sciences; Beijing 100049 China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; 501 Haike Road Shanghai 201203 China
- School of Pharmacy; University of Chinese Academy of Sciences; Beijing 100049 China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; 501 Haike Road Shanghai 201203 China
- School of Pharmacy; University of Chinese Academy of Sciences; Beijing 100049 China
| |
Collapse
|
55
|
Abstract
In drug targeting, the urgent need for more effective and less iatrogenic therapies is pushing toward a complete revision of carrier setup. After the era of 'articles used as homing systems', novel prototypes are now emerging. Newly conceived carriers are endowed with better biocompatibility, biodistribution and targeting properties. The biomimetic approach bestows such improved functional properties. Exploiting biological molecules, organisms and cells, or taking inspiration from them, drug vector performances are now rapidly progressing toward the perfect carrier. Following this direction, researchers have refined carrier properties, achieving significant results. The present review summarizes recent advances in biomimetic and bioinspired drug vectors, derived from biologicals or obtained by processing synthetic materials with a biomimetic approach.
Collapse
|
56
|
Yang W, Xia Y, Fang Y, Meng F, Zhang J, Cheng R, Deng C, Zhong Z. Selective Cell Penetrating Peptide-Functionalized Polymersomes Mediate Efficient and Targeted Delivery of Methotrexate Disodium to Human Lung Cancer In Vivo. Adv Healthc Mater 2018; 7:e1701135. [PMID: 29280317 DOI: 10.1002/adhm.201701135] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/01/2017] [Indexed: 12/17/2022]
Abstract
It is a long challenge to develop nanomedicines that simultaneously possess tumor cell selectivity and penetration functions. Here, it is reported that selective cell penetrating peptide (RLWMRWYSPRTRAYGC)-functionalized polymersomes (SCPP-PS) mediate efficient and targeted delivery of methotrexate disodium (MTX) to human lung cancer in vivo. SCPP-PS with an SCPP density of 18.7% is self-crosslinked, has a small size (63-65 nm), and high MTX loading (up to 19.4 wt%), shows selective uptake and fast penetration into A549 lung cancer cells, and efficiently releases MTX intracellularly. Interestingly, MTX-loaded SCPP-PS (MTX-SCPP-PS) displays much lower IC50 than those of MTX-PS and free MTX. Installing SCPP to polymersomes has no detrimental effect to their long blood circulation time but significantly increases drug accumulation in A549 tumor (5.3% injected dose per gram at 8 h post injection). Remarkably, SCPP-PS exhibits deep penetration in to A549 tumors. MTX-SCPP-PS completely inhibits tumor progression and significantly improves survival rates in mice bearing A549 lung tumor xenografts as compared to MTX-PS and free MTX groups (median survival time: 75 vs 45 and 38 d, respectively), without causing noticeable adverse effects. These results highlight that functionalization of nanomedicines with SCPP is a feasible strategy to achieve efficient and targeted tumor therapy.
Collapse
Affiliation(s)
- Weijing Yang
- 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
| | - Yifeng Xia
- 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
| | - Yuan Fang
- 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
| | - Fenghua 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
| | - Jian 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
| | - Ru Cheng
- 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
| | - Chao Deng
- 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
|
57
|
Pang L, Qin J, Han L, Zhao W, Liang J, Xie Z, Yang P, Wang J. Exploiting macrophages as targeted carrier to guide nanoparticles into glioma. Oncotarget 2018; 7:37081-37091. [PMID: 27213597 PMCID: PMC5095060 DOI: 10.18632/oncotarget.9464] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 04/24/2016] [Indexed: 11/25/2022] Open
Abstract
The restriction of anti-cancer drugs entry to tumor sites in the brain is a major impediment to the development of new strategies for the treatment of glioma. Based on the finding that macrophages possess an intrinsic homing property enabling them to migrate to tumor sites across the endothelial barriers in response to the excretion of cytokines/chemokines in the diseased tissues, we exploited macrophages as 'Trojan horses' to carry drug-loading nanoparticles (NPs), pass through barriers, and offload them into brain tumor sites. Anticancer drugs were encapsulated in nanoparticles to avoid their damage to the cells. Drug loading NPs was then incubated with RAW264.7 cells in vitro to prepare macrophage-NPs (M-NPs). The release of NPs from M-NPs was very slow in medium of DMEM and 10% FBS and significantly accelerated when LPS and IFN-γ were added to mimic tumor inflammation microenvironment. The viability of macrophages was not affected when the concentration of doxorubicin lower than 25 μg/ml. The improvement of cellular uptake and penetration into the core of glioma spheroids of M-NPs compared with NPs was verified in in vitro studies. The tumor-targeting efficiency of NPs was also significantly enhanced after loading into macrophages in nude mice bearing intracranial U87 glioma. Our results provided great potential of macrophages as an active biocarrier to deliver anticancer drugs to the tumor sites in the brain and improve therapeutic effects of glioma.
Collapse
Affiliation(s)
- Liang Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jing Qin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Limei Han
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Wenjie Zhao
- Shanghai Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Jianming Liang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhongyi Xie
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Pei Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jianxin Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
58
|
Affiliation(s)
- Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
59
|
Saeed H, Ali H, Soudan H, Embaby A, El-Sharkawy A, Farag A, Hussein A, Ataya F. Molecular cloning, structural modeling and production of recombinant Aspergillus terreus l. asparaginase in Escherichia coli. Int J Biol Macromol 2018; 106:1041-1051. [DOI: 10.1016/j.ijbiomac.2017.08.110] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 11/15/2022]
|
60
|
Ngandeu Neubi GM, Opoku-Damoah Y, Gu X, Han Y, Zhou J, Ding Y. Bio-inspired drug delivery systems: an emerging platform for targeted cancer therapy. Biomater Sci 2018; 6:958-973. [DOI: 10.1039/c8bm00175h] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bio-inspired platforms directly derived from biological sources are becoming a rapidly emerging field in the development of future anticancer therapeutics. The various platforms discussed are bacteria-based, virus-inspired, cell-derived, nanostructured lipid nanoparticles, and biomacromolecular drug delivery systems.
Collapse
Affiliation(s)
- Gella Maelys Ngandeu Neubi
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yaw Opoku-Damoah
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Xiaochen Gu
- Faculty of Pharmacy
- University of Manitoba
- Winnipeg
- Canada R3E 0T5
| | - Yue Han
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yang Ding
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
61
|
Dong X, Niu Y, Ding Y, Wang Y, Zhao J, Leng W, Qin L. Formulation and Drug Loading Features of Nano-Erythrocytes. NANOSCALE RESEARCH LETTERS 2017; 12:202. [PMID: 28314369 PMCID: PMC5355415 DOI: 10.1186/s11671-017-1980-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/06/2017] [Indexed: 06/06/2023]
Abstract
Nano erythrocyte ghosts have recently been used as drug carriers of water-soluble APIs due to inherit biological characteristics of good compatibility, low toxicity, and small side-effect. In this study, we developed a novel drug delivery system based on nano erythrocyte ghosts (STS-Nano-RBCs) to transport Sodium Tanshinone IIA sulfonate (STS) for intravenous use in rat. STS-Nano-RBCs were prepared by hypotonic lysis and by extrusion methods, and its biological properties were investigated compared with STS injection. The results revealed that STS-Nano-RBCs have narrow particle size distribution, good drug loading efficiency, and good stability within 21 days. Compared with STS injection, STS-Nano-RBCs extended the drug release time in vitro and in vivo with better repairing effect on oxidative stress-impaired endothelial cells. These results suggest that the nano erythrocyte ghosts system could be used to deliver STS.
Collapse
Affiliation(s)
- Xiaoting Dong
- Department of Pharmaceutics, School of Pharmacy, Guangdong Pharmaceutical University, No. 280 Waihuandong Road, Guangzhou, 510006, China
| | - Yawei Niu
- Department of Pharmaceutics, School of Pharmacy, Guangdong Pharmaceutical University, No. 280 Waihuandong Road, Guangzhou, 510006, China
| | - Yi Ding
- Guangzhou Institute for Drug Control, 23 Xizeng Road, Guangzhou, 510160, China
| | - Yuemin Wang
- Department of Pharmaceutics, School of Pharmacy, Guangdong Pharmaceutical University, No. 280 Waihuandong Road, Guangzhou, 510006, China
| | - Jialan Zhao
- Department of Pharmaceutics, School of Pharmacy, Guangdong Pharmaceutical University, No. 280 Waihuandong Road, Guangzhou, 510006, China
| | - Wei Leng
- Jacobson Pharma Group, 7 Dai Shun Street, Tai Po District, NT, Hong Kong
| | - Linghao Qin
- Department of Pharmaceutics, School of Pharmacy, Guangdong Pharmaceutical University, No. 280 Waihuandong Road, Guangzhou, 510006, China.
| |
Collapse
|
62
|
Pang L, Zhang C, Qin J, Han L, Li R, Hong C, He H, Wang J. A novel strategy to achieve effective drug delivery: exploit cells as carrier combined with nanoparticles. Drug Deliv 2017; 24:83-91. [PMID: 28155538 PMCID: PMC8241159 DOI: 10.1080/10717544.2016.1230903] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/23/2016] [Accepted: 08/28/2016] [Indexed: 12/21/2022] Open
Abstract
Cell-mediated drug delivery systems employ specific cells as drug vehicles to deliver drugs to targeted sites. Therapeutics or imaging agents are loaded into these cells and then released in diseased sites. These specific cells mainly include red blood cells, leukocytes, stem cells and so on. The cell acts as a Trojan horse to transfer the drug from circulating blood to the diseased tissue. In such a system, these cells keep their original properties, which allow them to mimic the migration behavior of specific cells to carry drug to the targeted site after in vivo administration. This strategy elegantly combines the advantages of both carriers, i.e. the adjustability of nanoparticles (NPs) and the natural functions of active cells, which therefore provides a new perspective to challenge current obstacles in drug delivery. This review will describe a fundamental understanding of these cell-based drug delivery systems, and discuss the great potential of combinational application of cell carrier and NPs.
Collapse
Affiliation(s)
- Liang Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Chun Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Jing Qin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Limei Han
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Ruixiang Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Chao Hong
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jianxin Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| |
Collapse
|
63
|
Andón FT, Digifico E, Maeda A, Erreni M, Mantovani A, Alonso MJ, Allavena P. Targeting tumor associated macrophages: The new challenge for nanomedicine. Semin Immunol 2017; 34:103-113. [PMID: 28941641 DOI: 10.1016/j.smim.2017.09.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/23/2022]
Abstract
The engineering of new nanomedicines with ability to target and kill or re-educate Tumor Associated Macrophages (TAMs) stands up as a promising strategy to induce the effective switching of the tumor-promoting immune suppressive microenvironment, characteristic of tumors rich in macrophages, to one that kills tumor cells, is anti-angiogenic and promotes adaptive immune responses. Alternatively, the loading of monocytes/macrophages in blood circulation with nanomedicines, may be used to profit from the high infiltration ability of myeloid cells and to allow the drug release in the bulk of the tumor. In addition, the development of TAM-targeted imaging nanostructures, can be used to study the macrophage content in solid tumors and, hence, for a better diagnosis and prognosis of cancer disease. The major challenges for the effective targeting of TAM with nanomedicines and their application in the clinic have already been identified. These challenges are associated to the undesirable clearance of nanomedicines by, the mononuclear phagocyte system (macrophages) in competing organs (liver, lung or spleen), upon their intravenous injection; and also to the difficult penetration of nanomedicines across solid tumors due to the abnormal vasculature and the excessive extracellular matrix present in stromal tumors. In this review we describe the recent nanotechnology-base strategies that have been developed to target macrophages in tumors.
Collapse
Affiliation(s)
- Fernando Torres Andón
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy; Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), University of Santiago de Compostela, 15706 Campus Vida, Santiago de Compostela, Spain.
| | - Elisabeth Digifico
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy; Humanitas University, Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Akihiro Maeda
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Marco Erreni
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Alberto Mantovani
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy; Humanitas University, Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - María José Alonso
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), University of Santiago de Compostela, 15706 Campus Vida, Santiago de Compostela, Spain; Pharmacy & Pharmaceutical Technology Department, School of Pharmacy, University of Santiago de Compostela, 15705 Campus Vida, Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Paola Allavena
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| |
Collapse
|
64
|
Simulation of the osmosis-based drug encapsulation in erythrocytes. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 47:261-270. [DOI: 10.1007/s00249-017-1255-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/01/2017] [Accepted: 09/11/2017] [Indexed: 12/11/2022]
|
65
|
Cai H, Liang Z, Huang W, Wen L, Chen G. Engineering PLGA nano-based systems through understanding the influence of nanoparticle properties and cell-penetrating peptides for cochlear drug delivery. Int J Pharm 2017; 532:55-65. [PMID: 28870763 DOI: 10.1016/j.ijpharm.2017.08.084] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/15/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022]
Abstract
The properties of poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and penetration enhancers play a deciding role in the inner ear drug delivery of NPs across the round window membrane (RWM). Thus, PLGA nano-based systems with a variety of particle sizes and surface chemistries and those combined with cell-penetrating peptides (CPPs) as penetration enhancers were devised to explore their impact on the cochlear drug delivery in vivo. First, we demonstrated that the properties of NPs dictated the extent of NP cochlear entry by near-infrared fluorescence imaging. NPs with the sizes of 150 and 300nm had faster entry than that of 80nm NPs. At 0.5h, among the NPs unmodified and modified with chitosan (CS), poloxamer 407 (P407) and methoxy polyethylene glycol, CS-PLGA-NPs (positive surface charge) carried payload to the cochlea fastest, whereas P407-PLGA-NPs (surface hydrophilicity) showed the greatest distribution in the cochlea at 24h. Compared to other CPPs (TAT, penetratin and poly(arginine)8), low molecular weight protamine (LMWP) performed an outstanding enhanced NP cellular uptake in HEI-OC1 cells and cochlear entry. More importantly, NPs with optimized properties and CPPs may be combined to improve RWM penetration. For the first time, we confirmed that the combination of P407-PLGA-NPs (mean diameter: 100-200nm) and LMWP provided a synergistic enhancement in NP entry to the organ of Corti and stria vascularis without inducing pathological alteration of cochlear tissues and RWM. Taken together, we propose an effective PLGA nano-based strategy for enhanced drug delivery to the inner ear tissues that combines hydrophilic molecule-modified NPs and CPPs, ultimately opening an avenue for superior inner ear therapy.
Collapse
Affiliation(s)
- Hui Cai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhongping Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wenli Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lu Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Gang Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
66
|
Tzounakas VL, Karadimas DG, Papassideri IS, Seghatchian J, Antonelou MH. Erythrocyte-based drug delivery in Transfusion Medicine: Wandering questions seeking answers. Transfus Apher Sci 2017; 56:626-634. [DOI: 10.1016/j.transci.2017.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
67
|
Macromolecular Conjugate and Biological Carrier Approaches for the Targeted Delivery of Antibiotics. Antibiotics (Basel) 2017; 6:antibiotics6030014. [PMID: 28677631 PMCID: PMC5617978 DOI: 10.3390/antibiotics6030014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/24/2017] [Accepted: 06/29/2017] [Indexed: 01/21/2023] Open
Abstract
For the past few decades, the rapid rise of antibiotic multidrug-resistance has presented a palpable threat to human health worldwide. Meanwhile, the number of novel antibiotics released to the market has been steadily declining. Therefore, it is imperative that we utilize innovative approaches for the development of antimicrobial therapies. This article will explore alternative strategies, namely drug conjugates and biological carriers for the targeted delivery of antibiotics, which are often eclipsed by their nanomedicine-based counterparts. A variety of macromolecules have been investigated as conjugate carriers, but only those most widely studied in the field of infectious diseases (e.g., proteins, peptides, antibodies) will be discussed in detail. For the latter group, blood cells, especially erythrocytes, have been successfully tested as homing carriers of antimicrobial agents. Bacteriophages have also been studied as a candidate for similar functions. Once these alternative strategies receive the amount of research interest and resources that would more accurately reflect their latent applicability, they will inevitably prove valuable in the perennial fight against antibiotic resistance.
Collapse
|
68
|
Parayath NN, Amiji MM. Therapeutic targeting strategies using endogenous cells and proteins. J Control Release 2017; 258:81-94. [DOI: 10.1016/j.jconrel.2017.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/02/2017] [Accepted: 05/04/2017] [Indexed: 01/14/2023]
|
69
|
Magdanz V, Medina-Sánchez M, Schwarz L, Xu H, Elgeti J, Schmidt OG. Spermatozoa as Functional Components of Robotic Microswimmers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1606301. [PMID: 28323360 DOI: 10.1002/adma.201606301] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/18/2017] [Indexed: 05/24/2023]
Abstract
In recent years, the combination of synthetic micro- and nanomaterials with spermatozoa as functional components has led to the development of tubular and helical spermbots - microrobotic devices with potential applications in the biomedical and nanotechnological field. Here, the initial advances in this field are discussed and the use of spermatozoa as functional parts in microdevices elaborated. Besides the potential uses of these hybrid robotic microswimmers, the obstacles along the way are discussed, with suggestions for solutions of the encountered challenges also given.
Collapse
Affiliation(s)
- Veronika Magdanz
- Leibniz Institute for Solid State and Materials Research, IFW Dresden e.V., Institute for Integrative Nanosciences, Helmholtzstrasse 20, 01069, Dresden, Germany
| | - Mariana Medina-Sánchez
- Leibniz Institute for Solid State and Materials Research, IFW Dresden e.V., Institute for Integrative Nanosciences, Helmholtzstrasse 20, 01069, Dresden, Germany
| | - Lukas Schwarz
- Leibniz Institute for Solid State and Materials Research, IFW Dresden e.V., Institute for Integrative Nanosciences, Helmholtzstrasse 20, 01069, Dresden, Germany
| | - Haifeng Xu
- Leibniz Institute for Solid State and Materials Research, IFW Dresden e.V., Institute for Integrative Nanosciences, Helmholtzstrasse 20, 01069, Dresden, Germany
| | - Jens Elgeti
- Theoretical Soft Matter and Biophysics, Institute of Complex Systems (ICS-2), 52425, Jülich, Germany
| | - Oliver G Schmidt
- Leibniz Institute for Solid State and Materials Research, IFW Dresden e.V., Institute for Integrative Nanosciences, Helmholtzstrasse 20, 01069, Dresden, Germany
- Chemnitz University of Technology, Reichenhainer Str. 70, 09107, Chemnitz, Germany
- Center for Advancing Electronics Dresden, Dresden University of Technology, Würzburger Str. 46, 01187, Dresden, Germany
| |
Collapse
|
70
|
Agrahari V, Agrahari V, Mitra AK. Next generation drug delivery: circulatory cells-mediated nanotherapeutic approaches. Expert Opin Drug Deliv 2017; 14:285-289. [PMID: 27791407 DOI: 10.1080/17425247.2017.1254614] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Vivek Agrahari
- a Division of Pharmaceutical Sciences , School of Pharmacy, University of Missouri-Kansas City , Kansas City , MO , USA
| | - Vibhuti Agrahari
- a Division of Pharmaceutical Sciences , School of Pharmacy, University of Missouri-Kansas City , Kansas City , MO , USA
| | - Ashim K Mitra
- a Division of Pharmaceutical Sciences , School of Pharmacy, University of Missouri-Kansas City , Kansas City , MO , USA
| |
Collapse
|
71
|
Feni L, Neundorf I. The Current Role of Cell-Penetrating Peptides in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1030:279-295. [PMID: 29081059 DOI: 10.1007/978-3-319-66095-0_13] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cell-penetrating peptides (CPPs) are a heterogeneous class of peptides with the ability to translocate across the plasma membrane and to carry attached cargos inside the cell. Two main entry pathways are discussed, as direct translocation and endocytosis , whereas the latter is often favored when bulky cargos are added to the CPP. Attachment to the CPP can be achieved by means of covalent coupling or non-covalent complex formation, depending on the chemical nature of the cargo. Owing to their striking abilities the further development and application of CPP-based delivery strategies has steadily emerged during the past years. However, one main pitfall when using CPPs is their non-selective uptake in nearly all types of cells. Thus, one particular interest lies in the design of targeting strategies that help to circumvent this drawback but still benefit from the potent delivery abilities of CPPs. The following review aims to summarize some of these very recent concepts and to highlight the current role of CPPs in cancer therapy.
Collapse
Affiliation(s)
- Lucia Feni
- Department of Chemistry, Biochemistry, University of Cologne, Zuelpicherstr. 47a, D-50674, Cologne, Germany
| | - Ines Neundorf
- Department of Chemistry, Biochemistry, University of Cologne, Zuelpicherstr. 47a, D-50674, Cologne, Germany.
| |
Collapse
|
72
|
Abstract
Macromolecules (proteins/peptides) have the potential for the development of new therapeutics. Due to their specific mechanism of action, macromolecules can be administered at relatively low doses compared with small-molecule drugs. Unfortunately, the therapeutic potential and clinical application of macromolecules is hampered by various obstacles including their large size, short in vivo half-life, phagocytic clearance, poor membrane permeability and structural instability. These challenges have encouraged researchers to develop novel strategies for effective delivery of macromolecules. In this review, various routes of macromolecule administration (invasive/noninvasive) are discussed. The advantages/limitations of novel delivery systems and the potential role of nanotechnology for the delivery of macromolecules are elaborated. In addition, fabrication approaches to make nanoformulations in different shapes and sizes are also summarized.
Collapse
|
73
|
Red blood cells: Supercarriers for drugs, biologicals, and nanoparticles and inspiration for advanced delivery systems. Adv Drug Deliv Rev 2016; 106:88-103. [PMID: 26941164 DOI: 10.1016/j.addr.2016.02.007] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/17/2016] [Accepted: 02/19/2016] [Indexed: 12/19/2022]
Abstract
Red blood cells (RBCs) constitute a unique drug delivery system as a biologic or hybrid carrier capable of greatly enhancing pharmacokinetics, altering pharmacodynamics (for example, by changing margination within the intravascular space), and modulating immune responses to appended cargoes. Strategies for RBC drug delivery systems include internal and surface loading, and the latter can be performed both ex vivo and in vivo. A relatively new avenue for RBC drug delivery is their application as a carrier for nanoparticles. Efforts are also being made to incorporate features of RBCs in nanocarriers to mimic their most useful aspects, such as long circulation and stealth features. RBCs have also recently been explored as carriers for the delivery of antigens for modulation of immune response. Therefore, RBC-based drug delivery systems represent supercarriers for a diverse array of biomedical interventions, and this is reflected by several industrial and academic efforts that are poised to enter the clinical realm.
Collapse
|
74
|
Rossi L, Pierigè F, Antonelli A, Bigini N, Gabucci C, Peiretti E, Magnani M. Engineering erythrocytes for the modulation of drugs' and contrasting agents' pharmacokinetics and biodistribution. Adv Drug Deliv Rev 2016; 106:73-87. [PMID: 27189231 DOI: 10.1016/j.addr.2016.05.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 01/14/2023]
Abstract
Pharmacokinetics, biodistribution, and biological activity are key parameters that determine the success or failure of therapeutics. Many developments intended to improve their in vivo performance, aim at modulating concentration, biodistribution, and targeting to tissues, cells or subcellular compartments. Erythrocyte-based drug delivery systems are especially efficient in maintaining active drugs in circulation, in releasing them for several weeks or in targeting drugs to selected cells. Erythrocytes can also be easily processed to entrap the desired pharmaceutical ingredients before re-infusion into the same or matched donors. These carriers are totally biocompatible, have a large capacity and could accommodate traditional chemical entities (glucocorticoids, immunossuppresants, etc.), biologics (proteins) and/or contrasting agents (dyes, nanoparticles). Carrier erythrocytes have been evaluated in thousands of infusions in humans proving treatment safety and efficacy, hence gaining interest in the management of complex pathologies (particularly in chronic treatments and when side-effects become serious issues) and in new diagnostic approaches.
Collapse
|
75
|
CPP-Assisted Intracellular Drug Delivery, What Is Next? Int J Mol Sci 2016; 17:ijms17111892. [PMID: 27854260 PMCID: PMC5133891 DOI: 10.3390/ijms17111892] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 11/16/2022] Open
Abstract
For the past 20 years, we have witnessed an unprecedented and, indeed, rather miraculous event of how cell-penetrating peptides (CPPs), the naturally originated penetrating enhancers, help overcome the membrane barrier that has hindered the access of bio-macromolecular compounds such as genes and proteins into cells, thereby denying their clinical potential to become potent anti-cancer drugs. By taking the advantage of the unique cell-translocation property of these short peptides, various payloads of proteins, nucleic acids, or even nanoparticle-based carriers were delivered into all cell types with unparalleled efficiency. However, non-specific CPP-mediated cell penetration into normal tissues can lead to widespread organ distribution of the payloads, thereby reducing the therapeutic efficacy of the drug and at the same time increasing the drug-induced toxic effects. In view of these challenges, we present herein a review of the new designs of CPP-linked vehicles and strategies to achieve highly effective yet less toxic chemotherapy in combating tumor oncology.
Collapse
|
76
|
Abstract
Red blood cells (RBCs) are naturally capable of transporting diverse cargoes throughout the circulatory system, both loaded to their surface or within their inner volume. Starting largely from the 1970s, diverse approaches for encapsulation into, and surface coupling onto, RBCs have been investigated as potential drug delivery systems. In the last decade, these efforts have yielded diverse strategies to load drugs and nanocarriers to RBCs, and to optimize their pharmacokinetics, distribution, and effects in the body. Several formulations of donor RBCs encapsulated with enzymes and drugs are currently undergoing clinical trials for treatment of oncologic and neurologic conditions. Newer approaches include design of drugs with an affinity to circulating RBCs, encapsulation into RBCs using membrane permeating compounds, and design of hybrid drug delivery systems combining synthetic components with fragments of RBC membranes. Notwithstanding the growing enthusiasm and optimism in RBC drug delivery, in this article we discuss potentially problematic issues of this biomedical concept, especially impairment of biocompatibility of the carrier RBCs, and other adverse and unintended effects. Rigorous and systematic analysis of the cautionary aspects described in this article should be further developed and extended in order to soberly gauge the risk/benefit balance of any given RBC-based drug delivery application. While there is little doubt that RBC drug delivery will ultimately flourish, focusing research efforts on approaches that are unlikely to cause adverse effects in patients will help to sooner bring this day.
Collapse
Affiliation(s)
- Carlos H Villa
- Division of Transfusion Medicine and Therapeutic Pathology, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety Improvement & DDR Strategies, London, UK
| | - Vladimir Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
77
|
He H, Sun L, Ye J, Liu E, Chen S, Liang Q, Shin MC, Yang VC. Enzyme-triggered, cell penetrating peptide-mediated delivery of anti-tumor agents. J Control Release 2016; 240:67-76. [DOI: 10.1016/j.jconrel.2015.10.040] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/15/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022]
|
78
|
Advances of blood cell-based drug delivery systems. Eur J Pharm Sci 2016; 96:115-128. [PMID: 27496050 DOI: 10.1016/j.ejps.2016.07.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/28/2016] [Accepted: 07/31/2016] [Indexed: 11/22/2022]
Abstract
Blood cells, including erythrocytes, leukocytes and platelets are used as drug carriers in a wide range of applications. They have many unique advantages such as long life-span in circulation (especially erythrocytes), target release capacities (especially platelets), and natural adhesive properties (leukocytes and platelets). These properties make blood cell based delivery systems, as well as their membrane-derived carriers, far superior to other drug delivery systems. Despite the advantages, the further development of blood cell-based delivery systems was hindered by limitations in the source, storage, and mass production. To overcome these problems, synthetic biomaterials that mimic blood cell and nanocrystallization of blood cells have been developed and may represent the future direction for blood cell membrane-based delivery systems. In this paper, we review recent progress of the rising blood cell-based drug delivery systems, and also discuss their challenges and future tendency of development.
Collapse
|
79
|
Leuzzi V, Rossi L, Gabucci C, Nardecchia F, Magnani M. Erythrocyte-mediated delivery of recombinant enzymes. J Inherit Metab Dis 2016; 39:519-30. [PMID: 27026098 DOI: 10.1007/s10545-016-9926-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/29/2016] [Accepted: 03/03/2016] [Indexed: 10/22/2022]
Abstract
The possibility to clone, express and purify recombinant enzymes have originated the opportunity to dispose of a virtually infinite array of proteins that could be used in the clinics to treat several inherited and acquired pathological conditions. However, the direct administration of these recombinant proteins faces some intrinsic difficulties, such as degradation by circulating proteases and/or inactivation by the patient immune system. The use of drug delivery systems may overcome these limitations. Concerning recombinant enzyme therapy, the present review will mainly focus on the exploitation of erythrocytes as a carrier system for enzymes removing potentially noxious metabolites from the circulation, either as limiting treatment strategy for auxotrophic tumours or as a detoxing approach for some intoxication type inherited metabolic disorders. Moreover, the possibility of using RBCs as a potential delivering system addressing the enzymes to the monocyte-macrophages of reticular endothelial system for the treatment of diseases associated with this cell lineage, e.g. lysosome storage diseases, will be briefly discussed.
Collapse
Affiliation(s)
- Vincenzo Leuzzi
- Department of Child and Adolescent Neuropsychiatry, SAPIENZA University of Rome, Via deiSabelli 108, 00185, Rome, Italy.
| | - Luigia Rossi
- Department of Biomolecular Science, University of Urbino, Via Saffi 2, 61029, Urbino, PU, Italy
| | - Claudia Gabucci
- Department of Biomolecular Science, University of Urbino, Via Saffi 2, 61029, Urbino, PU, Italy
| | - Francesca Nardecchia
- Department of Child and Adolescent Neuropsychiatry, SAPIENZA University of Rome, Via deiSabelli 108, 00185, Rome, Italy
- Department of Physiology and Pharmacology, SAPIENZA University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Mauro Magnani
- Department of Biomolecular Science, University of Urbino, Via Saffi 2, 61029, Urbino, PU, Italy
| |
Collapse
|
80
|
Shegokar R, Sawant S, Al Shaal L. Applications of Cell-Based Drug Delivery Systems: Use of Single Cell Assay. SERIES IN BIOENGINEERING 2016. [DOI: 10.1007/978-3-662-49118-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
81
|
Bourgeaux V, Lanao JM, Bax BE, Godfrin Y. Drug-loaded erythrocytes: on the road toward marketing approval. Drug Des Devel Ther 2016; 10:665-76. [PMID: 26929599 PMCID: PMC4755692 DOI: 10.2147/dddt.s96470] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Erythrocyte drug encapsulation is one of the most promising therapeutic alternative approaches for the administration of toxic or rapidly cleared drugs. Drug-loaded erythrocytes can operate through one of the three main mechanisms of action: extension of circulation half-life (bioreactor), slow drug release, or specific organ targeting. Although the clinical development of erythrocyte carriers is confronted with regulatory and development process challenges, industrial development is expanding. The manufacture of this type of product can be either centralized or bedside based, and different procedures are employed for the encapsulation of therapeutic agents. The major challenges for successful industrialization include production scalability, process validation, and quality control of the released therapeutic agents. Advantages and drawbacks of the different manufacturing processes as well as success key points of clinical development are discussed. Several entrapment technologies based on osmotic methods have been industrialized. Companies have already achieved many of the critical clinical stages, thus providing the opportunity in the future to cover a wide range of diseases for which effective therapies are not currently available.
Collapse
Affiliation(s)
- Vanessa Bourgeaux
- ERYTECH Pharma, Lyon, France
- Correspondence: Vanessa Bourgeaux, ERYTECH Pharma, 60 Avenue Rockefeller, Bâtiment Adénine, 69008 Lyon, France, Tel +33 478 781 572, Fax +33 478 789 309, Email
| | - José M Lanao
- Department of Pharmacy and Pharmaceutical Technology, University of Salamanca, Salamanca, Spain
| | - Bridget E Bax
- Cardiovascular and Cell Sciences Research Institute, St George’s University of London, London, UK
| | | |
Collapse
|
82
|
Terracciano M, Shahbazi MA, Correia A, Rea I, Lamberti A, De Stefano L, Santos HA. Surface bioengineering of diatomite based nanovectors for efficient intracellular uptake and drug delivery. NANOSCALE 2015; 7:20063-20074. [PMID: 26568517 DOI: 10.1039/c5nr05173h] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Diatomite is a natural porous silica material of sedimentary origin. Due to its peculiar properties, it can be considered as a valid surrogate of synthetic porous silica for nano-based drug delivery. In this work, we exploit the potential of diatomite nanoparticles (DNPs) for drug delivery with the aim of developing a successful dual-biofunctionalization method by polyethylene glycol (PEG) coverage and cell-penetrating peptide (CPP) bioconjugation, to improve the physicochemical and biological properties of the particles, to enhance the intracellular uptake in cancer cells, and to increase the biocompatibility of 3-aminopropyltriethoxysilane (APT) modified-DNPs. DNPs-APT-PEG-CPP showed hemocompatibility for up to 200 μg mL(-1) after 48 h of incubation with erythrocytes, with a hemolysis value of only 1.3%. The cytotoxicity of the modified-DNPs with a concentration up to 200 μg mL(-1) and incubation with MCF-7 and MDA-MB-231 breast cancer cells for 24 h, demonstrated that PEGylation and CPP-bioconjugation can strongly reduce the cytotoxicity of DNPs-APT. The cellular uptake of the modified-DNPs was also evaluated using the above mentioned cancer cell lines, showing that the CPP-bioconjugation can considerably increase the DNP cellular uptake. Moreover, the dual surface modification of DNPs improved both the loading of a poorly water-soluble anticancer drug, sorafenib, with a loading degree up to 22 wt%, and also enhanced the drug release profiles in aqueous solutions. Overall, this work demonstrates that the biofunctionalization of DNPs is a promising platform for drug delivery applications in cancer therapy as a result of its enhanced stability, biocompatibility, cellular uptake, and drug release profiles.
Collapse
Affiliation(s)
- Monica Terracciano
- Institute for Microelectronics and Microsystems, National Research Council, Naples, 80131, Italy.
| | | | | | | | | | | | | |
Collapse
|
83
|
Dual-functional bio-derived nanoparticulates for apoptotic antitumor therapy. Biomaterials 2015; 72:90-103. [DOI: 10.1016/j.biomaterials.2015.08.051] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/25/2015] [Accepted: 08/28/2015] [Indexed: 11/22/2022]
|
84
|
Hunault-Berger M, Leguay T, Huguet F, Leprêtre S, Deconinck E, Ojeda-Uribe M, Bonmati C, Escoffre-Barbe M, Bories P, Himberlin C, Chevallier P, Rousselot P, Reman O, Boulland ML, Lissandre S, Turlure P, Bouscary D, Sanhes L, Legrand O, Lafage-Pochitaloff M, Béné MC, Liens D, Godfrin Y, Ifrah N, Dombret H. A Phase 2 study of L-asparaginase encapsulated in erythrocytes in elderly patients with Philadelphia chromosome negative acute lymphoblastic leukemia: The GRASPALL/GRAALL-SA2-2008 study. Am J Hematol 2015; 90:811-8. [PMID: 26094614 DOI: 10.1002/ajh.24093] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 11/11/2022]
Abstract
PURPOSE The GRASPALL/GRAALL-SA2-2008 Phase II trial evaluated the safety and efficacy of L-asparaginase encapsulated within erythrocytes (GRASPA®) in patients ≥ 55 years with Philadelphia chromosome-negative acute lymphoblastic leukemia. FINDINGS Thirty patients received escalating doses of GRASPA® on Day 3 and 6 of induction Phases 1 and 2. The primary efficacy endpoint was asparagine depletion < 2 µmol/L for at least 7 days. This was reached in 85 and 71% of patients with 100 and 150 IU/kg respectively but not with 50 IU/kg. Grade 3/4 infection, hypertransaminasemia, hyperbilirubinemia and deep vein thrombosis occurred in 77, 20, 7, and 7% of patients, respectively. No allergic reaction or clinical pancreatitis was observed despite 17% of Grade 3/4 lipase elevation. Anti-asparaginase antibodies were detected in 50% of patients and related to a reduction in the duration of asparagine depletion during induction Phase 2 without decrease of encapsulated L-asparaginase activity. Complete remission rate was 70%. With a median follow-up of 42 months, median overall survival was 15.8 and 9.7 months, in the 100 and 150 IU/kg cohorts respectively. CONCLUSIONS The addition of GRASPA®, especially at the 100 IU/kg dose level, is feasible in elderly patients without excessive toxicity and associated with durable asparagine depletion. (clinicaltrials.gov identifier NCT01523782).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Pierre Bories
- Haematology Department of Strasbourg; Strasbourg France
| | | | | | | | | | | | | | | | | | | | | | | | | | - David Liens
- Haematology Department of ERYTECH Pharma; France
| | - Yann Godfrin
- Haematology Department of ERYTECH Pharma; France
| | | | - Hervé Dombret
- Haematology Department of Saint-Louis AP-HP Paris; France
| | | |
Collapse
|
85
|
Tan S, Wu T, Zhang D, Zhang Z. Cell or cell membrane-based drug delivery systems. Theranostics 2015; 5:863-81. [PMID: 26000058 PMCID: PMC4440443 DOI: 10.7150/thno.11852] [Citation(s) in RCA: 313] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/18/2015] [Indexed: 01/14/2023] Open
Abstract
Natural cells have been explored as drug carriers for a long period. They have received growing interest as a promising drug delivery system (DDS) until recently along with the development of biology and medical science. The synthetic materials, either organic or inorganic, are found to be with more or less immunogenicity and/or toxicity. The cells and extracellular vesicles (EVs), are endogenous and thought to be much safer and friendlier. Furthermore, in view of their host attributes, they may achieve different biological effects and/or targeting specificity, which can meet the needs of personalized medicine as the next generation of DDS. In this review, we summarized the recent progress in cell or cell membrane-based DDS and their fabrication processes, unique properties and applications, including the whole cells, EVs and cell membrane coated nanoparticles. We expect the continuing development of this cell or cell membrane-based DDS will promote their clinic applications.
Collapse
Affiliation(s)
- Songwei Tan
- 1. Tongji School of Pharmacy
- 2. National Engineering Research Center for Nanomedicine
- 3. Hubei Engineering Research Center for Novel DDS, Huazhong University of Science and Technology, Wuhan 430030, P R China
| | | | | | - Zhiping Zhang
- 1. Tongji School of Pharmacy
- 2. National Engineering Research Center for Nanomedicine
- 3. Hubei Engineering Research Center for Novel DDS, Huazhong University of Science and Technology, Wuhan 430030, P R China
| |
Collapse
|
86
|
Su Y, Xie Z, Kim GB, Dong C, Yang J. Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 2015; 1:201-217. [PMID: 25984572 PMCID: PMC4428174 DOI: 10.1021/ab500179h] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Drug delivery systems, particularly nanomaterial-based drug delivery systems, possess a tremendous amount of potential to improve diagnostic and therapeutic effects of drugs. Controlled drug delivery targeted to a specific disease is designed to significantly improve the pharmaceutical effects of drugs and reduce their side effects. Unfortunately, only a few targeted drug delivery systems can achieve high targeting efficiency after intravenous injection, even with the development of numerous surface markers and targeting modalities. Thus, alternative drug and nanomedicine targeting approaches are desired. Circulating cells, such as erythrocytes, leukocytes, and stem cells, present innate disease sensing and homing properties. Hence, using living cells as drug delivery carriers has gained increasing interest in recent years. This review highlights the recent advances in the design of cell-mediated drug delivery systems and targeting mechanisms. The approaches of drug encapsulation/conjugation to cell-carriers, cell-mediated targeting mechanisms, and the methods of controlled drug release are elaborated here. Cell-based "live" targeting and delivery could be used to facilitate a more specific, robust, and smart payload distribution for the next-generation drug delivery systems.
Collapse
Affiliation(s)
- Yixue Su
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Zhiwei Xie
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Gloria B. Kim
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Cheng Dong
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institutes, the Huck Institutes of Life Sciences, The Pennsylvania State University, W340 Millennium Science Complex, University Park, PA 16802
| |
Collapse
|
87
|
PTD-Modified ATTEMPTS for Enhanced Toxin-based Cancer Therapy: An In Vivo Proof-of-Concept Study. Pharm Res 2015; 32:2690-703. [PMID: 25701313 DOI: 10.1007/s11095-015-1653-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/10/2015] [Indexed: 01/10/2023]
Abstract
PURPOSE To investigate the feasibility of applying PTD-modified ATTEMPTS (Antibody Targeted Triggered Electrically Modified Prodrug-Type Strategy) for enhanced toxin therapy for the treatment of cancer. METHODS A heparin-functionalized murine anti-CEA monoclonal antibody (mAb), T84.66-heparin (T84.66-Hep), was chemically synthesized and characterized for specific binding to CEA overexpressed cells. The T84.66-Hep was then applied to the PTD-modified ATTEMPTS approach and the crucial features of the drug delivery system (DDS), 'antibody targeting' and 'heparin/protamine-based prodrug', were evaluated in vitro to examine whether it could selective delivery a PTD-modified toxin, recombinant TAT-gelonin chimera (TAT-Gel), to CEA high expression cancer cells (LS174T). Furthermore, the feasibility of the drug delivery system (DDS) was assessed in vivo by biodistribution and efficacy studies using LS174T s.c. xenograft tumor bearing mice. RESULTS T84.66-Hep displayed specific binding, but limited internalization (35% after 48 h incubation) to CEA high expression LS174T cells over low expression HCT116 cells. When mixed together with TAT-Gel, the T84.66-Hep formed a strong yet reversible complex. This complex formation provided an effective means of active tumor targeting of TAT-Gel, by 1) directing the TAT-Gel to CEA overexpressed tumor cells and 2) preventing nonspecific cell transduction to non-targeted normal cells. The cell transduction of TAT-Gel could, however, be efficiently reversed by addition of protamine. Feasibility of in vivo tumor targeting and "protamine-induced release" of TAT-Gel from the T84.66-Hep counterpart was confirmed by biodistribution and preliminary efficacy studies. CONCLUSIONS This study successfully demonstrated in vitro and in vivo the applicability of PTD-modified ATTEMPTS for toxin-based cancer therapy.
Collapse
|
88
|
Sun Y, Wang Y, Cui Y, Zou W, Tan Y, Liang J, Fan Y, Zhang X. DOX-encapsulated intelligent PAA-g-PEG/PEG–Fa polymeric micelles for intensifying antitumor therapeutic effect via active-targeted tumor accumulation. J Mater Chem B 2015; 3:5478-5489. [DOI: 10.1039/c5tb00438a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Reduction-breakable active targeting polymeric micelles as drug delivery systems could improve delivery efficiency by tumor-specific recognition.
Collapse
Affiliation(s)
- Yong Sun
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yaning Wang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yani Cui
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Wen Zou
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yanfei Tan
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Jie Liang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
89
|
Ben-Dov N, Korenstein R. The uptake of HIV Tat peptide proceeds via two pathways which differ from macropinocytosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:869-77. [PMID: 25542781 DOI: 10.1016/j.bbamem.2014.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 11/16/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022]
Abstract
Cell penetrating peptides (CPPs) have been extensively studied as vectors for cellular delivery of therapeutic molecules, yet the identity of their uptake routes remained unclear and is still under debate. In this study we provide new insights into CPP entry routes by quantitatively measuring the intracellular uptake of FAM-labeled Tat-peptide under rigorous kinetic and thermal conditions. The uptake of Tat-peptide between 4 and 15°C corresponds to Q10=1.1, proceeding through a prompt (<5 min), temperature-independent process, suggesting direct membrane translocation. At longer durations, Tat rate of uptake shows linear dependence on temperature with Q10=1.44, accompanied by activation energy Ea=4.45 Kcal/mole. These values are significantly lower than those we found for the macropinocytosis probe dextran (Q10=2.2 and Ea=7.2 Kcal/mole) which possesses an exponential dependence on temperature, characteristic of endocytosis processes. Tat-peptide and dextran do not interfere with each other's uptake rate and the ratio of Tat-peptide uptake to its extracellular concentration is ~15 times higher than that for dextran. In addition, Phloretin, a modulator of cell membrane dipole potential, is shown to increase dextran uptake but to reduce that of Tat. We conclude that the uptake of Tat differs from that of dextran in all parameters. Tat uptake proceeds by dual entry routes which differ by their energy dependence.
Collapse
Affiliation(s)
- Nadav Ben-Dov
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel.
| | - Rafi Korenstein
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel.
| |
Collapse
|
90
|
15 years of ATTEMPTS: a macromolecular drug delivery system based on the CPP-mediated intracellular drug delivery and antibody targeting. J Control Release 2014; 205:58-69. [PMID: 25483423 DOI: 10.1016/j.jconrel.2014.12.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/18/2014] [Accepted: 12/01/2014] [Indexed: 01/22/2023]
Abstract
Traditionally, any drug intended for combating the tumor would distribute profoundly to other organs and tissues as lack of targeting specificity, thus resulting in limited therapeutic effects toward the tumor but severe drug-induced toxic side effects. To prevail over this obstacle of drug-induced systemic toxicity, a novel approach termed "ATTEMPTS" (antibody targeted triggered electrically modified prodrug type strategy) was designed, which directly introduces both of the targeting and prodrug features onto the protein drugs. The ATTEMPTS system is composed of the antibody targeting component consisting of antibodies linked with heparin, and the cell penetrating peptide (CPP) modified drug component. The two components mentioned above self-assembled into a tight complex via the charge to charge interaction between the anionic heparin and cationic CPP. Once accumulated at the targeting site, the CPP modified drug is released from the blockage by a second triggering agent, while remaining inactive in the circulation during tumor targeting thus aborting its effect on normal tissues. We utilized the heparin-induced inhibition on the cell-penetrating activity of CPP to create the prodrug feature, and subsequently the protamine-induced reversal of heparin inhibition to resume cell transduction of the protein drug via the CPP function. Our approach is the first known system to overcome this selectivity issue, enabling CPP-mediated cellular drug delivery to be practically applicable clinically. In this review, we thoroughly discussed the historical and novel progress of the "ATTEMPTS" system.
Collapse
|
91
|
Sun W, Lu Y, Gu Z. Advances in Anticancer Protein Delivery Using Micro-/ Nanoparticles. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2014; 31:1204-1222. [PMID: 27642232 PMCID: PMC5026193 DOI: 10.1002/ppsc.201400140] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Indexed: 04/14/2023]
Abstract
Proteins exhibiting anticancer activities, especially those capable of discriminately killing cancer cells, have attracted increasing interest in developing protein-based anticancer therapeutics. This progress report surveys recent advances in delivering anticancer proteins directly to tumor tissue for inducing apoptosis/necrosis or indirectly to antigen presenting cells for provoking immune responses. Protein delivery carriers such as inorganic particles, lipid particles, polymeric particles, DNA/protein based biomacromolecular particles as well as cell based carriers are reviewed with comments on their advantages and limitations. Future challenges and opportunities are also discussed.
Collapse
Affiliation(s)
- Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yue Lu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
92
|
Low molecular weight protamine (LMWP): A nontoxic protamine substitute and an effective cell-penetrating peptide. J Control Release 2014; 193:63-73. [DOI: 10.1016/j.jconrel.2014.05.056] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/21/2014] [Accepted: 05/27/2014] [Indexed: 01/07/2023]
|
93
|
Shin MC, Zhao J, Zhang J, Huang Y, He H, Wang M, Min KA, Yang VC. Recombinant TAT-gelonin fusion toxin: synthesis and characterization of heparin/protamine-regulated cell transduction. J Biomed Mater Res A 2014; 103:409-419. [PMID: 24733757 DOI: 10.1002/jbm.a.35188] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 03/31/2014] [Indexed: 11/05/2022]
Abstract
Protein toxins, such as gelonin, are highly desirable anti-cancer drug candidates due to their unparalleled potency and repetitive reaction mechanism in inhibiting protein translation. However, for its potential application in cancer therapy, there remains the cell membrane barrier that allows permeation of only small molecules, which must be overcome. To address this challenge, we conjugated gelonin with a protein transduction domain (PTD), the TAT peptide, via genetic recombination. The chimeric TAT-gelonin fusion protein (TAT-Gel) retained equipotent N-glycosidase activity yet displayed greater cell uptake than unmodified recombinant gelonin (rGel), thereby yielding a significantly augmented cytotoxic activity. Remarkably, TAT-Gel displayed up to 177-fold lower IC₅₀ (avg. 54.3 nM) than rGel (avg. IC₅₀ : 3640 nM) in tested cell lines. This enhanced cytotoxicity, however, also raised potential toxicity concerns due to the non-selectivity of PTD in its mediated cell transduction. To solve this problem, we investigated the plausibility of regulating the cell transduction of TAT-Gel via a reversible masking using heparin and protamine. Here, we demonstrated, both in vitro and in vivo, that the cell transduction of TAT-Gel can be completely curbed with heparin and yet this heparin block can be efficiently reversed by the addition of protamine. This reversible tight regulation of the cell transduction of TAT-Gel by heparin and protamine sheds light of possible application of TAT-Gel in achieving a highly effective yet safe drug therapy for the treatment of tumors.
Collapse
Affiliation(s)
- Meong Cheol Shin
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Jingwen Zhao
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Jian Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Mei Wang
- College of Pharmacy, Xinjiang Medical University, 393 Xinyi Road, Urumqi 830011, China
| | - Kyoung Ah Min
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Victor C Yang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| |
Collapse
|