1
|
Ling D, Jia X, Wang K, Yan Q, Yuan B, Du L, Li M, Jin Y. Cancer cell membrane-coated bacterial ghosts for highly efficient paclitaxel delivery against metastatic lung cancer. Acta Pharm Sin B 2024; 14:365-377. [PMID: 38261850 PMCID: PMC10792973 DOI: 10.1016/j.apsb.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/02/2023] [Accepted: 07/15/2023] [Indexed: 01/25/2024] Open
Abstract
Chemotherapy is one of the major approaches for the treatment of metastatic lung cancer, although it is limited by the low tumor delivery efficacy of anticancer drugs. Bacterial therapy is emerging for cancer treatment due to its high immune stimulation effect; however, excessively generated immunogenicity will cause serious inflammatory response syndrome. Here, we prepared cancer cell membrane-coated liposomal paclitaxel-loaded bacterial ghosts (LP@BG@CCM) by layer-by-layer encapsulation for the treatment of metastatic lung cancer. The preparation processes were simple, only involving film formation, electroporation, and pore extrusion. LP@BG@CCM owned much higher 4T1 cancer cell toxicity than LP@BG due to its faster fusion with cancer cells. In the 4T1 breast cancer metastatic lung cancer mouse models, the remarkably higher lung targeting of intravenously injected LP@BG@CCM was observed with the almost normalized lung appearance, the reduced lung weight, the clear lung tissue structure, and the enhanced cancer cell apoptosis compared to its precursors. Moreover, several major immune factors were improved after administration of LP@BG@CCM, including the CD4+/CD8a+ T cells in the spleen and the TNF-α, IFN-γ, and IL-4 in the lung. LP@BG@CCM exhibits the optimal synergistic chemo-immunotherapy, which is a promising medication for the treatment of metastatic lung cancer.
Collapse
Affiliation(s)
- Dandan Ling
- Anhui Medical University, Hefei 230032, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xueli Jia
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ke Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Qiucheng Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lina Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Miao Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yiguang Jin
- Anhui Medical University, Hefei 230032, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
2
|
Lee CH, Tang JC, Hendricks NG, Anvari B. Proteomes of Micro- and Nanosized Carriers Engineered from Red Blood Cells. J Proteome Res 2023; 22:896-907. [PMID: 36792548 PMCID: PMC10756254 DOI: 10.1021/acs.jproteome.2c00695] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Red blood cell (RBC)-derived systems offer a potential platform for delivery of biomedical cargos. Although the importance of specific proteins associated with the biodistribution and pharmacokinetics of these particles has been recognized, it remains to be explored whether some of the key transmembrane and cytoskeletal proteins responsible for immune-modulatory effects and mechanical integrity of the particles are retained. Herein, using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and quantitative tandem mass tag mass spectrometry in conjunction with bioinformatics analysis, we have examined the proteomes of micro- and nanosized erythrocyte ghosts doped with indocyanine green and compared them with those of RBCs. We identified a total of 884 proteins in each set of RBCs, micro-, and nanosized particles, of which 8 and 45 proteins were expressed at significantly different relative abundances when comparing micro-sized particles vs RBCs and nanosized particles vs RBCs, respectively. We found greater differences in relative abundances of some mechano-modulatory proteins, such as band 3 and protein 4.2, and immunomodulatory proteins like CD44, CD47, and CD55 in nanosized particles as compared to RBCs. Our findings highlight that the methods utilized in fabricating RBC-based systems can induce substantial effects on their proteomes. Mass spectrometry data are available at ProteomeXchange with the identifier PXD038780.
Collapse
Affiliation(s)
- Chi-Hua Lee
- Department of Biochemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Jack C Tang
- Department of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Nathan G Hendricks
- Institute for Integrative Genome Biology, Proteomics Core, University of California, Riverside, Riverside, California 92521, United States
| | - Bahman Anvari
- Department of Biochemistry, University of California, Riverside, Riverside, California 92521, United States
- Department of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| |
Collapse
|
3
|
Rong R, Raza F, Liu Y, Yuan WE, Su J, Qiu M. Blood cell-based drug delivery systems: a biomimetic platform for antibacterial therapy. Eur J Pharm Biopharm 2022; 177:273-288. [PMID: 35868489 DOI: 10.1016/j.ejpb.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
With the rapid increase in multidrug-resistance against antibiotics, higher doses of antibiotics or more effective antibiotics are needed to treat diseases, which ultimately leads to a decrease in the body's immunity and seriously threatens human health worldwide. The efficiency of antibiotics has been a large challenge for years. To overcome this problem, many carriers are utilized for anti-bacteria, attempting to optimize the delivery of such drugs and transport them safely and directly to the site of disease. Blood cell-based drug delivery systems present several advantages as compared to polymeric delivery system. These blood cells including red blood cells (RBCs), leukocytes, platelets. The blood cells and their membranes can both be used as drug carriers to deliver antibacterial drugs. In addition, blood cells can overcome many physiological/pathological obstacles faced by nanoparticles in vivo and effectively deliver drugs to the site of the disease. In this paper, we review studies on blood cell-based delivery systems used in antibacterial therapy, and analyze different roles in antibacterial therapy, which provide basis for further study in this field.
Collapse
Affiliation(s)
- Ruonan Rong
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Yuhao Liu
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Wei-En Yuan
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China.
| |
Collapse
|
4
|
Song W, Jia P, Zhang T, Dou K, Liu L, Ren Y, Liu F, Xue J, Hasanin MS, Qi H, Zhou Q. Cell membrane-camouflaged inorganic nanoparticles for cancer therapy. J Nanobiotechnology 2022; 20:289. [PMID: 35717234 PMCID: PMC9206402 DOI: 10.1186/s12951-022-01475-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/23/2022] [Indexed: 12/18/2022] Open
Abstract
Inorganic nanoparticles (INPs) have been paid great attention in the field of oncology in recent past years since they have enormous potential in drug delivery, gene delivery, photodynamic therapy (PDT), photothermal therapy (PTT), bio-imaging, driven motion, etc. To overcome the innate limitations of the conventional INPs, such as fast elimination by the immune system, low accumulation in tumor sites, and severe toxicity to the organism, great efforts have recently been made to modify naked INPs, facilitating their clinical application. Taking inspiration from nature, considerable researchers have exploited cell membrane-camouflaged INPs (CMCINPs) by coating various cell membranes onto INPs. CMCINPs naturally inherit the surface adhesive molecules, receptors, and functional proteins from the original cell membrane, making them versatile as the natural cells. In order to give a timely and representative review on this rapidly developing research subject, we highlighted recent advances in CMCINPs with superior unique merits of various INPs and natural cell membranes for cancer therapy applications. The opportunity and obstacles of CMCINPs for clinical translation were also discussed. The review is expected to assist researchers in better eliciting the effect of CMCINPs for the management of tumors and may catalyze breakthroughs in this area.
Collapse
Affiliation(s)
- Wanli Song
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Pengfei Jia
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Ting Zhang
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Keke Dou
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
| | - Lubin Liu
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Yaping Ren
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Fujun Liu
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Junmiao Xue
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.,School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Mohamed Sayed Hasanin
- Cellulose and Paper Department, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Hongzhao Qi
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.
| | - Qihui Zhou
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China. .,School of Stomatology, Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
5
|
Chen Y, Han L, Qiu X, Wang M, Chen Z, Cai Y, Xin Y, Lv Y, Hu A, Chai D, Li L, Li H, Zheng J, Wang G. Reassembling of albumin-bound paclitaxel mitigates myelosuppression and improves its antitumoral efficacy via neutrophil-mediated targeting drug delivery. Drug Deliv 2022; 29:728-742. [PMID: 35244505 PMCID: PMC8903760 DOI: 10.1080/10717544.2022.2046892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Albumin-bound paclitaxel (abPTX) has been widely used in cancer treatment. However, dose-related side effects, such as myelosuppression, restrict its clinical application. Cell-based targeting drug delivery is a promising way to mitigate systematic side-effects and improve antitumoral efficacy. In this study, we demonstrated that reassembled abPTX could be engulfed by neutrophils in vivo and delivered to tumor site, thus improving therapeutic efficacy and mitigating myelosuppression. First, in vitro analysis confirmed that reassembling of abPTX formed uniform and stable serum albumin nanoparticles (NP-abPTX) with size of 107.5 ± 2.29 nm and reserved the ability to kill tumor cells. Second, we found that NP-abPTX could be engulfed by activated neutrophil in vitro and in vivo but do not affect neutrophils’ function, such as chemotaxis and activation. In a murine tumor model, we further proved that local radiotherapy (RT) induced inflammation activated peripheral neutrophils to capture venous infused NP-abPTX and carry them into tumor tissue. As compared to abPTX, infusion of NP-abPTX dramatically enhanced inhibition of tumor growth treated by local RT and mitigated hematotoxicity. Therefore, our study demonstrated a novel strategy to mitigate side-effects and to improve tumor killing efficacy of abPTX through neutrophil-mediated targeting drug delivery.
Collapse
Affiliation(s)
- Yuxin Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xiaoyan Qiu
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Zheng Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ying Cai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yong Xin
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yanfang Lv
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Ankang Hu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Liantao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
6
|
Resealed erythrocytes: Towards a novel approach for anticancer therapy. J INDIAN CHEM SOC 2021. [DOI: 10.1016/j.jics.2021.100257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
7
|
Javed S, Alshehri S, Shoaib A, Ahsan W, Sultan MH, Alqahtani SS, Kazi M, Shakeel F. Chronicles of Nanoerythrosomes: An Erythrocyte-Based Biomimetic Smart Drug Delivery System as a Therapeutic and Diagnostic Tool in Cancer Therapy. Pharmaceutics 2021; 13:368. [PMID: 33802156 PMCID: PMC7998655 DOI: 10.3390/pharmaceutics13030368] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Recently, drug delivery using natural biological carriers has emerged as one of the most widely investigated topics of research. Erythrocytes, or red blood cells, can act as potential carriers for a wide variety of drugs, including anticancer, antibacterial, antiviral, and anti-inflammatory, along with various proteins, peptides, enzymes, and other macromolecules. The red blood cell-based nanocarrier systems, also called nanoerythrosomes, are nanovesicles poised with extraordinary features such as long blood circulation times, the ability to escape immune system, the ability to release the drug gradually, the protection of drugs from various endogenous factors, targeted and specified delivery of drugs, as well as possessing both therapeutic and diagnostic applications in various fields of biomedical sciences. Their journey over the last two decades is escalating with fast pace, ranging from in vivo to preclinical and clinical studies by encapsulating a number of drugs into these carriers. Being biomimetic nanoparticles, they have enhanced the stability profile of drugs and their excellent site-specific targeting ability makes them potential carrier systems in the diagnosis and therapy of wide variety of tumors including gliomas, lung cancers, breast cancers, colon cancers, gastric cancers, and other solid tumors. This review focuses on the most recent advancements in the field of nanoerythrosomes, as an excellent and promising nanoplatform for the novel drug delivery of various drugs particularly antineoplastic drugs along with their potential as a promising diagnostic tool for the identification of different tumors.
Collapse
Affiliation(s)
- Shamama Javed
- Department of Pharmaceutics, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (S.J.); (M.H.S.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.); (M.K.)
- Department of Pharmaceutical Sciences, College of Pharmacy, Almaarefa University, Riyadh 11597, Saudi Arabia
| | - Ambreen Shoaib
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (A.S.); (S.S.A.)
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia;
| | - Muhammad Hadi Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (S.J.); (M.H.S.)
| | - Saad Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (A.S.); (S.S.A.)
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.); (M.K.)
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.); (M.K.)
| |
Collapse
|
8
|
Karakoçak B, Laradji A, Primeau T, Berezin MY, Li S, Ravi N. Hyaluronan-Conjugated Carbon Quantum Dots for Bioimaging Use. ACS APPLIED MATERIALS & INTERFACES 2021; 13:277-286. [PMID: 33355448 PMCID: PMC8243741 DOI: 10.1021/acsami.0c20088] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 05/28/2023]
Abstract
This work demonstrates the application of hyaluronan-conjugated nitrogen-doped carbon quantum dots (HA-nCQDs) for bioimaging of tumor cells and illustrates their potential use as carriers in targeted drug delivery. Quantum dots are challenging to deliver with specificity, which hinders their application. To facilitate targeted internalization by cancer cells, hyaluronic acid, a natural ligand of CD44 receptors, was covalently grafted on nCQDs. The HA-nCQD conjugate was synthesized by carbodiimide coupling of the amine moieties on nCQDs and the carboxylic acids on HA chains. Conjugated HA-nCQD retained sufficient fluorescence, although with 30% lower quantum efficiency than the original nCQDs. Confocal microscopy showed enhanced internalization of HA-nCQDs, facilitated by CD44 receptors. To demonstrate the specificity of HA-nCQDs toward human tumor cells, patient-derived breast cancer tissue with high-CD44 expression was implanted in adult mice. The tumors were allowed to grow up to 200-250 mm3 prior to the injection of HA-nCQDs. With either local or systemic injection, we achieved a high level of tumor specificity judged by a strong signal-to-noise ratio between the tumor and the surrounding tissue in vivo. Overall, the results show that HA-nCQDs can be used for imaging of CD44-specific tumors in preclinical models of human cancer and potentially used as carriers for targeted drug delivery into CD44-rich cells.
Collapse
Affiliation(s)
- Bedia
Begüm Karakoçak
- Department
of Ophthalmology and Visual Sciences, Washington
University in St. Louis, St. Louis, Missouri 63110, United States
- Veterans
Affairs Medical Center, St. Louis, Missouri 63106, United States
| | - Amine Laradji
- Department
of Ophthalmology and Visual Sciences, Washington
University in St. Louis, St. Louis, Missouri 63110, United States
- Veterans
Affairs Medical Center, St. Louis, Missouri 63106, United States
| | - Tina Primeau
- Department
of Medicine, Washington University School
of Medicine, St. Louis, Missouri 63110, United
States
| | - Mikhail Y. Berezin
- Department
of Radiology, Washington University School
of Medicine, St. Louis, Missouri 63110, United
States
| | - Shunqiang Li
- Department
of Medicine, Washington University School
of Medicine, St. Louis, Missouri 63110, United
States
| | - Nathan Ravi
- Department
of Ophthalmology and Visual Sciences, Washington
University in St. Louis, St. Louis, Missouri 63110, United States
- Veterans
Affairs Medical Center, St. Louis, Missouri 63106, United States
| |
Collapse
|
9
|
Harisa GI, Sherif AY, Youssof AM, Alanazi FK, Salem-Bekhit MM. Bacteriosomes as a Promising Tool in Biomedical Applications: Immunotherapy and Drug Delivery. AAPS PharmSciTech 2020; 21:168. [PMID: 32514657 DOI: 10.1208/s12249-020-01716-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
Bacteriosomes are a member of cell-derived vesicles that are proposed as promising tools in diagnosis, therapy, and drug delivery. These vesicles could be derived from a virus, bacterial cells, and animal cells. Biotechnology techniques were used in bioengineering of cell-derived vesicles in vitro, and in vivo. Bacterial vesicles such as bacterial cells, bacterial ghost, or bacteriosomes are vesicular structures derived from bacteria produced by manipulation of bacterial cells by chemical agents or gene-mediated lysis. Subsequently, bacterial vesicles (bacteriosomes) are non-living, non-denatured bacterial cell envelopes free of the cytoplasm and genetic materials. Gram-negative and Gram-positive bacteria are exploited in the production of bacteriosomes. Bacteriosomes have instinct organs, tissues, cells, as well as subcellular tropism. Moreover, bacteriosomes might be used as immunotherapy and/or drug delivery shuttles. They could act as cargoes for the delivery of small drugs, large therapeutics, and nanoparticles to the specific location. Furthermore, bacteriosomes have nature endosomal escaping ability, hence they could traffic different bio-membranes by endocytosis mechanisms. Therefore, bacterial-derived vesicles could be used in therapy and development of an innovative drug delivery systems. Consequently, utilizing bacteriosomes as drug cargoes enhances the delivery and efficacy of administered therapeutic agents. This review highlighted bacteriosomes in terms of source, engineering, characterization, applications, and limitations.
Collapse
|
10
|
Lu M, Huang Y. Bioinspired exosome-like therapeutics and delivery nanoplatforms. Biomaterials 2020; 242:119925. [PMID: 32151860 DOI: 10.1016/j.biomaterials.2020.119925] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/09/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023]
Abstract
Exosomes have emerged as appealing candidate therapeutic agents and delivery nanoplatforms due to their endogenous features and unique biological properties. However, obstacles such as low isolation yield, considerable complexity and potential safety concerns, and inefficient drug payload substantially hamper their therapeutic applicability. To this end, developing bioinspired exosome-like nanoparticles has become a promising area to overcome certain limitations of their natural counterparts. Synthetically fabrication of exosome-like nanoparticles that harbor only crucial components of exosomes through controllable protocols strongly increases the pharmaceutical acceptability of these vesicles. Assembly of exosome-like nanovesicles derived from producer cells allows for a promising strategy for scale-up production. To improve the loading capability and delivery efficiency of exosomes, hybrid exosome-like nanovesicles and membrane-camouflaged nanoparticles towards better bridging synthetic nanocarriers with natural exosomes could be designed. Building off these observations, herein, efforts are made to give an overview of bioinspired exosome-like therapeutics and delivery nanoplatforms. We briefly recapitulate the recent advance in exosome biology with focus on tailoring exosomes as therapeutics and delivery vehicles. Furthermore, we elaborately discuss the biomimicry methodologies for preparation of exosome-like nanoparticles with special emphasis on offering insights into strategies for rational design of exosome-like biomaterials as effective and safe therapeutics and delivery nanoplatforms.
Collapse
Affiliation(s)
- Mei Lu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, PR China.
| |
Collapse
|
11
|
Zargar SM, Hafshejani DK, Eskandarinia A, Rafienia M, Kharazi AZ. A Review of Controlled Drug Delivery Systems Based on Cells and Cell Membranes. JOURNAL OF MEDICAL SIGNALS & SENSORS 2019; 9:181-189. [PMID: 31544058 PMCID: PMC6743242 DOI: 10.4103/jmss.jmss_53_18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Novel drug delivery systems have ameliorated drugs’ pharmacokinetics and declined undesired ramifications while led to a better patient compliance by extending the time of release. In fact, although there has been a multitude of encouraging achievements in controlled drug release, the application of micro- and nano-carriers is confronted with some challenges such as rapid clearance and inefficient targeting. In addition, since cell systems can be an appropriate alternative to micro- and nano-particles, they have been used as biological carriers. In general, features such as stable release into blood, slow clearance, efficient targeting, and high biocompatibility are the main properties of cells applied as drug carriers. Furthermore, some cells such as erythrocytes, leukocytes, stem cells, and platelets have been used as release systems. Hence, most common cells that were used as aforementioned release systems are going to be presented in this review article.
Collapse
Affiliation(s)
- Seyed Mohammad Zargar
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Darioush Khodabakhshi Hafshejani
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Asghar Eskandarinia
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohamad Rafienia
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anousheh Zargar Kharazi
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Majumder J, Taratula O, Minko T. Nanocarrier-based systems for targeted and site specific therapeutic delivery. Adv Drug Deliv Rev 2019; 144:57-77. [PMID: 31400350 PMCID: PMC6748653 DOI: 10.1016/j.addr.2019.07.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 01/04/2023]
Abstract
Systemic drug delivery methods such as oral or parenteral administration of free drugs possess relatively low treatment efficiency and marked adverse side effects. The use of nanoparticles for drug delivery in most cases substantially enhances drug efficacy, improves pharmacokinetics and drug release and limits their side effects. However, further enhancement in drug efficacy and significant limitation of adverse side effects can be achieved by specific targeting of nanocarrier-based delivery systems especially in combination with local administration. The present review describes major advantages and limitations of organic and inorganic nanocarriers or living cell-based drug and nucleic acid delivery systems. Among these, different nanoparticles, supramolecular gels, therapeutic cells as living drug carriers etc. have emerged as a new frontier in modern medicine.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; Environmental and Occupational Health Science Institute, Piscataway, NJ 08854, USA.
| |
Collapse
|
13
|
Agnihotri J, Maurya P, Singh S, Saraf SA. Biomimetic Approaches for Targeted Nanomedicine: Current Status and Future Perspectives. CURRENT DRUG THERAPY 2019. [DOI: 10.2174/1574885514666181220092721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background: Cytotherapy products can be described as “living drugs”.
Cytotherapy is the swiftest growing fields in the treatment of cancer, heart diseases, aging
population and neuromuscular ailments. Biomimetic approaches are processes developed
by humans such as devices, substances, or systems that mimic nature or natural processes.
</P><P>
Objective and Method: It aims at developing a base for personalized medicine with
allogeneic, autologous and xenogenic therapies where cells are modified for target
selection. Such drug delivery methods appear to be complex and challenging. Literature
for approximately past two decades was collected and reviewed for the present article.
</P><P>
Results and Conclusion: The opportunities and challenges in cytotherapy have been classified,
discussed and demystified. Various process inputs, materials and process conditions
required in bioprocessing and preservation have been discussed at length. The review
also focuses on the regulatory requirements in India, Europe and U.S.
Collapse
Affiliation(s)
- Jaya Agnihotri
- H.K. College of Pharmacy, Jogeshwari West, Mumbai, India
| | - Priyanka Maurya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), VidyaVihar, Raebareli Road, Lucknow, 226025, India
| | | | - Shubhini A. Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), VidyaVihar, Raebareli Road, Lucknow, 226025, India
| |
Collapse
|
14
|
Li Z, Hu S, Cheng K. Platelets and their biomimetics for regenerative medicine and cancer therapies. J Mater Chem B 2018; 6:7354-7365. [PMID: 31372220 PMCID: PMC6675472 DOI: 10.1039/c8tb02301h] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Platelets, circulating blood cells derived from megakaryocytes, play a key role in various physical activities, including coagulation, hemostasis, the body's innate immune response, and cancer metastasis. By taking advantage of their key traits, researchers have developed strategies to exploit platelets and platelet-mimicking nanoassemblies to treat a number of conditions, including wounds, cancers, and bacterial infections. Compared to traditional polymer, lipsosome, and inorganic nanoparticles-based delivery systems, platelets and platelet-mimicking vehicles hold many advantages. Among these are their enhanced circulation time, their large volumes and surface areas for drug loading or conjugation, and their inherent ability to target some diseases. In this review, we will highlight the recent progress made in the development of disease-targeting platelets- and platelet-mimicking-vehicles as therapeutic platforms.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
15
|
Mac JT, Vankayala R, Patel DK, Wueste S, Anvari B. Erythrocyte-Derived Optical Nanoprobes Doped with Indocyanine Green-Bound Albumin: Material Characteristics and Evaluation for Cancer Cell Imaging. ACS Biomater Sci Eng 2018; 4:3055-3062. [PMID: 33435025 DOI: 10.1021/acsbiomaterials.8b00621] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nanosize structures activated by near-infrared (NIR) photoexcitation can provide an optical platform for the image-guided removal of small tumor nodules. We have engineered nanoparticles derived from erythrocytes that can be doped with NIR fluorophore indocyanine green (ICG). We refer to these constructs as NIR erythrocyte-derived transducers (NETs). The objective of this study was to determine if ICG-bound albumin (IbA), as the doping material, could enhance the fluorescence emission of NETs, and evaluate the capability of these nanoprobes in imaging cancer cells. Erythrocytes were isolated from bovine whole blood and depleted of hemoglobin to form erythrocyte ghosts (EGs). EGs were then extruded through nanosize porous membranes in the presence of 10-100 μm ICG or Iba (1:1 molar ratio) to form ICG- or IbA-doped NETs. The resulting nanosize constructs were characterized for their diameters, zeta-potentials, absorption, and fluorescence emission spectra. We used fluorescence microscopic imaging to evaluate the capability of the constructs in imaging SKOV3 ovarian cancer cells. Based on dynamic light-scattering measurements, ICG- and IbA-doped NETs had similar diameter distributions (Z-average diameter of 236 and 238 nm, respectively) in phosphate-buffered saline supplemented with 10% fetal bovine serum, which remained nearly constant over the course of 2 h at 37 °C. Despite a much-lower loading efficiency of IbA (∼0.7-8%) as compared to ICG (10-45%), the integrated normalized fluorescence emission of IbA-NETs was 2- to 6-fold higher than ICG-doped NETs. IbA-NETs also demonstrated an enhanced capability in fluorescence imaging of SKOV3 ovarian cancer cells, and can serve as potentially effective nanoprobes for the fluorescence imaging of cancerous cells.
Collapse
|
16
|
Gao C, Lin Z, Lin X, He Q. Cell Membrane-Camouflaged Colloid Motors for Biomedical Applications. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800056] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Changyong Gao
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| | - Zhihua Lin
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| | - Xiankun Lin
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| | - Qiang He
- Key Laboratory of Microsystems and Microstructures Manufacturing; State Key Laboratory of Robotics and Systems; Micro/Nano Technology Research Center; Harbin Institute of Technology; 2 Yikuang Street Harbin 150080 China
| |
Collapse
|
17
|
Gao Y, Kraft JC, Yu D, Ho RJY. Recent developments of nanotherapeutics for targeted and long-acting, combination HIV chemotherapy. Eur J Pharm Biopharm 2018; 138:75-91. [PMID: 29678735 DOI: 10.1016/j.ejpb.2018.04.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 01/20/2023]
Abstract
Combination antiretroviral therapy (cART) given orally has transformed HIV from a terminal illness to a manageable chronic disease. Yet despite the recent development of newer and more potent drugs for cART and suppression of virus in blood to undetectable levels, residual virus remains in tissues. Upon stopping cART, virus rebounds and progresses to AIDS. Current oral cART regimens have several drawbacks including (1) challenges in patient adherence due to pill fatigue or side-effects, (2) the requirement of life-long daily drug intake, and (3) limited penetration and retention in cells within lymph nodes. Appropriately designed injectable nano-drug combinations that are long-acting and retained in HIV susceptible cells within lymph nodes may address these challenges. While a number of nanomaterials have been investigated for delivery of HIV drugs and drug combinations, key challenges involve developing and scaling delivery systems that provide a drug combination targeted to HIV host cells and tissues where residual virus persists. With validation of the drug-insufficiency hypothesis in lymph nodes, progress has been made in the development of drug combination nanoparticles that are long-acting and targeted to lymph nodes and cells. Unique drug combination nanoparticles (DcNPs) composed of three HIV drugs-lopinavir, ritonavir, and tenofovir-have been shown to provide enhanced drug levels in lymph nodes; and elevated drug-combination levels in HIV-host cells in the blood and plasma for two weeks. This review summarizes the progress in the development of nanoparticle-based drug delivery systems for HIV therapy. It discusses how injectable nanocarriers may be designed to enable delivery of drug combinations that are long-lasting and target-selective in physiological contexts (in vivo) to provide safe and effective use. Consistent drug combination exposure in the sites of residual HIV in tissues and cells may overcome drug insufficiency observed in patients on oral cART.
Collapse
Affiliation(s)
- Yu Gao
- Cancer Metastasis Alert and Prevention Center, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Department of Pharmaceutics, University of Washington, Seattle, WA 98195, United States
| | - John C Kraft
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, United States
| | - Danni Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, United States
| | - Rodney J Y Ho
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, United States; Department of Bioengineering, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
18
|
Zhang K, Cao Y, Kuang Y, Liu M, Chen Y, Wang Z, Hong S, Wang J, Pei R. Gd 2O 3 and GH combined with red blood cells to improve the sensitivity of contrast agents for cancer targeting MR imaging. Biomater Sci 2018; 5:46-49. [PMID: 27840866 DOI: 10.1039/c6bm00627b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Herein, we fabricated efficient MR imaging probes by incorporating gadolinium oxide nanoparticles (Gd2O3) and gadolinium hybrid nanoparticles (GH) within RBCs. The Gd2O3 and GH encapsulated in the RBCs exhibited high relaxation rates and revealed high sensitivity for T1 MR imaging.
Collapse
Affiliation(s)
- Kunchi Zhang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yi Cao
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Ye Kuang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Min Liu
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yang Chen
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Zhili Wang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Shanni Hong
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Jine Wang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Renjun Pei
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
19
|
Martinez JO, Molinaro R, Hartman KA, Boada C, Sukhovershin R, De Rosa E, Kuri D, Zhang S, Evangelopoulos M, Carter AM, Bibb JA, Cooke JP, Tasciotti E. Biomimetic nanoparticles with enhanced affinity towards activated endothelium as versatile tools for theranostic drug delivery. Theranostics 2018; 8:1131-1145. [PMID: 29464004 PMCID: PMC5817115 DOI: 10.7150/thno.22078] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/09/2017] [Indexed: 12/30/2022] Open
Abstract
Activation of the vascular endothelium is characterized by increased expression of vascular adhesion molecules and chemokines. This activation occurs early in the progression of several diseases and triggers the recruitment of leukocytes. Inspired by the tropism of leukocytes, we investigated leukocyte-based biomimetic nanoparticles (i.e., leukosomes) as a novel theranostic platform for inflammatory diseases. Methods: Leukosomes were assembled by combining phospholipids and membrane proteins from leukocytes. For imaging applications, phospholipids modified with rhodamine and gadolinium were used. Leukosomes incubated with antibodies blocking lymphocyte function-associated antigen 1 (LFA-1) and CD45 were administered to explore their roles in targeting inflammation. In addition, relaxometric assessment of NPs was evaluated. Results: Liposomes and leukosomes were both spherical in shape with sizes ranging from 140-170 nm. Both NPs successfully integrated 8 and 13 µg of rhodamine and gadolinium, respectively, and demonstrated less than 4% variation in physicochemical features. Leukosomes demonstrated a 16-fold increase in breast tumor accumulation relative to liposomes. Furthermore, quantification of leukosomes in tumor vessels demonstrated a 4.5-fold increase in vessel lumens and a 14-fold increase in vessel walls. Investigating the targeting mechanism of action revealed that blockage of LFA-1 on leukosomes resulted in a 95% decrease in tumor accumulation. Whereas blockage of CD45 yielded a 60% decrease in targeting and significant increases in liver and spleen accumulation. In addition, when administered in mice with atherosclerotic plaques, leukosomes exhibited a 4-fold increase in the targeting of inflammatory vascular lesions. Lastly, relaxometric assessment of NPs demonstrated that the incorporation of membrane proteins into leukosomes did not impact the r1 and r2 relaxivities of the NPs, demonstrating 6 and 30 mM-1s-1, respectively. Conclusion: Our study demonstrates the ability of leukosomes to target activated vasculature and exhibit superior accumulation in tumors and vascular lesions. The versatility of the phospholipid backbone within leukosomes permits the incorporation of various contrast agents. Furthermore, leukosomes can potentially be loaded with therapeutics possessing diverse physical properties and thus warrant further investigation toward the development of powerful theranostic agents.
Collapse
|
20
|
Jia Y, Sheng Z, Hu D, Yan F, Zhu M, Gao G, Wang P, Liu X, Wang X, Zheng H. Highly penetrative liposome nanomedicine generated by a biomimetic strategy for enhanced cancer chemotherapy. Biomater Sci 2018; 6:1546-1555. [DOI: 10.1039/c8bm00256h] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Biomimetic liposome nanomedicine with deep tumor penetration and specific homotypic targeting ability enhanced cancer chemotherapy.
Collapse
|
21
|
Wang C, Wen D, Gu Z. Cellular Bioparticulates with Therapeutics for Cancer Immunotherapy. Bioconjug Chem 2017; 29:702-708. [DOI: 10.1021/acs.bioconjchem.7b00619] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
|
23
|
Molinaro R, Corbo C, Martinez JO, Taraballi F, Evangelopoulos M, Minardi S, Yazdi I, Zhao P, De Rosa E, Sherman M, De Vita A, Furman NT, Wang X, Parodi A, Tasciotti E. Biomimetic proteolipid vesicles for targeting inflamed tissues. NATURE MATERIALS 2016; 15:1037-46. [PMID: 27213956 PMCID: PMC5127392 DOI: 10.1038/nmat4644] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/13/2016] [Indexed: 05/13/2023]
Abstract
A multitude of micro- and nanoparticles have been developed to improve the delivery of systemically administered pharmaceuticals, which are subject to a number of biological barriers that limit their optimal biodistribution. Bioinspired drug-delivery carriers formulated by bottom-up or top-down strategies have emerged as an alternative approach to evade the mononuclear phagocytic system and facilitate transport across the endothelial vessel wall. Here, we describe a method that leverages the advantages of bottom-up and top-down strategies to incorporate proteins derived from the leukocyte plasma membrane into lipid nanoparticles. The resulting proteolipid vesicles-which we refer to as leukosomes-retained the versatility and physicochemical properties typical of liposomal formulations, preferentially targeted inflamed vasculature, enabled the selective and effective delivery of dexamethasone to inflamed tissues, and reduced phlogosis in a localized model of inflammation.
Collapse
Affiliation(s)
- R. Molinaro
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - C. Corbo
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- EINGE–Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
- IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - J. O. Martinez
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - F. Taraballi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Pain Therapy Service, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - M. Evangelopoulos
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - S. Minardi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - I.K. Yazdi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - P. Zhao
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - E. De Rosa
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - M. Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555
| | - A. De Vita
- Osteoncology and Rare Tumors Center, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Italy
| | - N.E. Toledano Furman
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - X. Wang
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - A. Parodi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- IRCCS SDN, Via Gianturco 113, 80143 Naples, Italy
| | - E. Tasciotti
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- To whom correspondence should be addressed: Dr. Ennio Tasciotti, Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030,
| |
Collapse
|
24
|
Hagenhoff A, Bruns CJ, Zhao Y, von Lüttichau I, Niess H, Spitzweg C, Nelson PJ. Harnessing mesenchymal stem cell homing as an anticancer therapy. Expert Opin Biol Ther 2016; 16:1079-92. [PMID: 27270211 DOI: 10.1080/14712598.2016.1196179] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) are non-hematopoietic progenitor cells that have been exploited as vehicles for cell-based cancer therapy. The general approach is based on the innate potential of adoptively applied MSC to undergo facilitated recruitment to malignant tissue. MSC from different tissue sources have been engineered using a variety of therapy genes that have shown efficacy in solid tumor models. AREAS COVERED In this review we will focus on the current developments of MSC-based gene therapy, in particular the diverse approaches that have been used for MSCs-targeted tumor therapy. We also discuss some outstanding issues and general prospects for their clinical application. EXPERT OPINION The use of modified mesenchymal stem cells as therapy vehicles for the treatment of solid tumors has progressed to the first generation of clinical trials, but the general field is still in its infancy. There are many questions that need to be addressed if this very complex therapy approach is widely applied in clinical settings. More must be understood about the mechanisms underlying tumor tropism and we need to identify the optimal source of the cells used. Outstanding issues also include the therapy transgenes used, and which tumor types represent viable targets for this therapy.
Collapse
Affiliation(s)
- Anna Hagenhoff
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Christiane J Bruns
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Yue Zhao
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Irene von Lüttichau
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Hanno Niess
- c Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery , University of Munich , Munich , Germany
| | - Christine Spitzweg
- d Department of Internal Medicine II , University of Munich , Munich , Germany
| | - Peter J Nelson
- e Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV , University of Munich , Munich , Germany
| |
Collapse
|
25
|
Xu P, Wang R, Wang X, Ouyang J. Recent advancements in erythrocytes, platelets, and albumin as delivery systems. Onco Targets Ther 2016; 9:2873-84. [PMID: 27274282 PMCID: PMC4876107 DOI: 10.2147/ott.s104691] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In the past few years, nanomaterial-based drug delivery systems have been applied to enhance the efficacy of therapeutics and to alleviate negative effects through the controlled delivery of targeting and releasing agents. However, few drug carriers can achieve high targeting efficacy, even when targeting modalities and surface markers are introduced. Immunological problems have also limited their wide applications. Biological drug delivery systems, such as erythrocytes, platelets, and albumin, have been extensively investigated because of their unique properties. In this review, erythrocytes, platelets, and albumin are described as efficient drug delivery systems. Their properties, applications, advantages, and limitations in disease treatment are explained. This review confirms that these systems can be used to facilitate a specific, biocompatible, and smart drug delivery.
Collapse
Affiliation(s)
- Peipei Xu
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Ruju Wang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Medical School, Southeast University, Nanjing, People's Republic of China
| | - Xiaohui Wang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Jian Ouyang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| |
Collapse
|
26
|
Mac JT, Nuñez V, Burns JM, Guerrero YA, Vullev VI, Anvari B. Erythrocyte-derived nano-probes functionalized with antibodies for targeted near infrared fluorescence imaging of cancer cells. BIOMEDICAL OPTICS EXPRESS 2016; 7:1311-22. [PMID: 27446657 PMCID: PMC4929643 DOI: 10.1364/boe.7.001311] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/02/2016] [Accepted: 03/10/2016] [Indexed: 05/20/2023]
Abstract
Constructs derived from mammalian cells are emerging as a new generation of nano-scale platforms for clinical imaging applications. Herein, we report successful engineering of hybrid nano-structures composed of erythrocyte-derived membranes doped with FDA-approved near infrared (NIR) chromophore, indocyanine green (ICG), and surface-functionalized with antibodies to achieve molecular targeting. We demonstrate that these constructs can be used for targeted imaging of cancer cells in vitro. These erythrocyte-derived optical nano-probes may provide a potential platform for clinical translation, and enable molecular imaging of cancer biomarkers.
Collapse
Affiliation(s)
- Jenny T. Mac
- Department of Biochemistry, University of California Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Vicente Nuñez
- Department of Bioengineering, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Joshua M. Burns
- Department of Bioengineering, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Yadir A. Guerrero
- Department of Bioengineering, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Valentine I. Vullev
- Department of Biochemistry, University of California Riverside, 900 University Ave., Riverside, CA 92521, USA
- Department of Bioengineering, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Bahman Anvari
- Department of Biochemistry, University of California Riverside, 900 University Ave., Riverside, CA 92521, USA
- Department of Bioengineering, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| |
Collapse
|
27
|
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
|
28
|
Lui PPY. Stem cell technology for tendon regeneration: current status, challenges, and future research directions. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:163-74. [PMID: 26715856 PMCID: PMC4685888 DOI: 10.2147/sccaa.s60832] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tendon injuries are a common cause of physical disability. They present a clinical challenge to orthopedic surgeons because injured tendons respond poorly to current treatments without tissue regeneration and the time required for rehabilitation is long. New treatment options are required. Stem cell-based therapies offer great potential to promote tendon regeneration due to their high proliferative, synthetic, and immunomodulatory activities as well as their potential to differentiate to the target cell types and undergo genetic modification. In this review, I first recapped the challenges of tendon repair by reviewing the anatomy of tendon. Next, I discussed the advantages and limitations of using different types of stem cells compared to terminally differentiated cells for tendon tissue engineering. The safety and efficacy of application of stem cells and their modified counterparts for tendon tissue engineering were then summarized after a systematic literature search in PubMed. The challenges and future research directions to enhance, optimize, and standardize stem cell-based therapies for augmenting tendon repair were then discussed.
Collapse
Affiliation(s)
- Pauline Po Yee Lui
- Headquarter, Hospital Authority, Hong Kong SAR, People's Republic of China
| |
Collapse
|
29
|
Cell permeable peptide conjugated nanoerythrosomes of fasudil prolong pulmonary arterial vasodilation in PAH rats. Eur J Pharm Biopharm 2015; 88:1046-55. [PMID: 25460151 DOI: 10.1016/j.ejpb.2014.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/01/2014] [Accepted: 10/16/2014] [Indexed: 11/23/2022]
Abstract
In this study, we tested the hypothesis that a cell permeable peptide, CARSKNKDC (CAR), conjugated nanoerythrosomes (NERs) containing fasudil, a rho-kinase (ROCK) inhibitor, produces prolonged pulmonary preferential vasodilation. CAR conjugated NERs containing fasudil were prepared by hypotonic lysis and extrusion method, and optimized for various physicochemical properties in-vitro. The formulations were then used to study the hemodynamic efficacy in a monocrotaline-induced rodent model of pulmonary arterial hypertension (PAH). CAR-NERs-Fasudil was spherical in shape with an average vesicle size and entrapment efficiency of 161.3 ± 1.37 nm and 48.81 ± 1.96%, respectively. Formulations were stable for ~3 weeks when stored at 4 °C and the drug was released in a controlled fashion for >48 h. The uptake of CAR-NERs-Fasudil by TGF-b activated pulmonary arterial smooth muscle cell was ~1.5-fold greater than the uptake of NERs-Fasudil. CAR-NERs-Fasudil inhibited ROCK activity and 5-hydroxytryptamine induced cell proliferation. In terms of reduction of pulmonary arterial pressure, intratracheal administration of CAR-NERs-Fasudil was ~2-fold more specific to the lungs compared with plain fasudil. Overall,CAR peptide grafted nanoerythrosomes offers a new platform for improving the therapeutic efficacy ofa rho-kinase inhibitor, fasudil, without affecting peripheral vasodilation.
Collapse
|
30
|
Barshtein G, Livshits L, Shvartsman LD, Shlomai NO, Yedgar S, Arbell D. Polystyrene Nanoparticles Activate Erythrocyte Aggregation and Adhesion to Endothelial Cells. Cell Biochem Biophys 2015; 74:19-27. [DOI: 10.1007/s12013-015-0705-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Drug delivery in aortic valve tissue engineering. J Control Release 2014; 196:307-23. [DOI: 10.1016/j.jconrel.2014.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/07/2014] [Accepted: 10/09/2014] [Indexed: 01/08/2023]
|
32
|
Erythrocyte-derived photo-theranostic agents: hybrid nano-vesicles containing indocyanine green for near infrared imaging and therapeutic applications. Sci Rep 2014; 3:2180. [PMID: 23846447 PMCID: PMC3709166 DOI: 10.1038/srep02180] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/24/2013] [Indexed: 01/30/2023] Open
Abstract
Development of theranostic nano-constructs may enable diagnosis and treatment of diseases at high spatial resolution. Some key requirements for clinical translation of such constructs are that they must be non-toxic, non-immunogenic, biodegradable, with extended circulating lifetime. Cell-based structures, particularly those derived from erythrocytes, are promising candidate carrier systems to satisfy these requirements. One particular type of theranostic materials utilize light-sensitive agents that once photo-activated can provide diagnostic imaging capability, and elicit therapeutic effects. Here we demonstrate the first successful engineering of hybrid nano-scale constructs derived from membranes of hemoglobin-depleted erythrocytes that encapsulate the near infrared chromophore, indocyanine green. We show the utility of the constructs as photo-theranostic agents in fluorescence imaging and photothermal destruction of human cells. These erythrocyte-mimicking nano-structures can be derived autologously, and may have broad applications in personal nanomedicine ranging from imaging and photo-destruction of cancerous tissues to vascular abnormalities, and longitudinal evaluations of therapeutic interventions.
Collapse
|
33
|
Zarrin A, Foroozesh M, Hamidi M. Carrier erythrocytes: recent advances, present status, current trends and future horizons. Expert Opin Drug Deliv 2014; 11:433-47. [PMID: 24456118 DOI: 10.1517/17425247.2014.880422] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Carrier erythrocytes, thanks to their main advantages, including biocompatibility, biodegradability, immunocompatibility, simple and well-known structure and physiology, availability for sampling and versatility in loading and use, have been studied as cellular carriers for delivery of drugs and other bioactive agents for more than three decades. Based on this body of knowledge and recent advances in this field, and with the help of novel multidisciplinary sciences and technologies, it seems that this field is becoming renowned and experiencing an outstanding turning point in its developmental history. AREAS COVERED In this trendy and timely review, following a short historical review of the story of erythrocytes from oxygen delivery to drug delivery and evaluation of the present status of these biocarriers, recent advances and current experimental, technological and clinical trends, as well as future horizons, and, in particular, translation-prone strategies, are going to be discussed in detail. EXPERT OPINION Despite the challenging developmental history of carrier erythrocytes, they now stand closer to clinical use and market entrance due to their unique advantages in drug delivery, proven by recently reported success stories in late-stage clinical trials and progresses made in biotechnology, nanotechnology and biomaterials fields. Translation-prone approaches, like in vivo loading of circulating erythrocytes or semiautomatic loading of erythrocytes, and more realistic study designs by focusing on clinical needs that have not been responded to or erythrocyte biology/fate-inspired study design are among the main trends being focused on by pioneer research groups active in this field of drug delivery.
Collapse
Affiliation(s)
- Abdolhossein Zarrin
- Shiraz University of Medical Sciences, Medicinal and Natural Products Chemistry Research Center , Shiraz , Iran
| | | | | |
Collapse
|
34
|
Gupta N, Patel B, Ahsan F. Nano-engineered erythrocyte ghosts as inhalational carriers for delivery of fasudil: preparation and characterization. Pharm Res 2014; 31:1553-65. [PMID: 24449438 DOI: 10.1007/s11095-013-1261-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/05/2013] [Indexed: 01/29/2023]
Abstract
PURPOSE Nanoerythrosomes (NERs), an engineered derivative of erythrocytes, have long been used as drug delivery carriers. These cell based carriers are biocompatible and biodegradable, and they exhibit efficient drug loading, targeting specificity and prolonged biological half-life. In this study, we have evaluated the feasibility of NERs as inhalable carriers for delivery of fasudil, an investigational drug for the treatment of pulmonary arterial hypertension. METHODS We prepared NERs by hypotonic lysis of erythrocytes derived from rat blood followed by extrusion through polycarbonate membranes. The formulations were optimized and characterized for size, morphology, entrapment efficiency, stability, cellular uptake and in-vitro release profiles followed by monitoring of drug absorption and safety evaluation after intratracheal administration of fasudil-loaded NERs into rats. RESULTS NERs were spherical in shape with an average size of 154.1 ± 1.31 nm and the drug loading efficiency was 48.76 ± 2.18%. Formulations were stable when stored at 4°C for 3 weeks. When incubated with rat pulmonary arterial smooth muscle cells (PASM), a significant amount of NERs was taken up by PASM cells. The drug encapsulated in NERs inhibited the rho-kinase activity upto 50%, which was comparable with the plain fasudil. A ~6-8 fold increase in the half-life of fasudil was observed when encapsulated in NERs. CONCLUSION This study suggests that nanoerythrosomes can be used as cell derived carriers for inhalational delivery of fasudil.
Collapse
Affiliation(s)
- Nilesh Gupta
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, Texas, 79106, USA
| | | | | |
Collapse
|
35
|
Preethy S, John S, Ganesh JS, Srinivasan T, Terunuma H, Iwasaki M, Abraham SJ. Age-old wisdom concerning cell-based therapies with added knowledge in the stem cell era: our perspectives. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2014; 6:13-8. [PMID: 24426785 PMCID: PMC3850297 DOI: 10.2147/sccaa.s41798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Among the various strategies providing a cure for illness, cell-based therapies have caught the attention of the world with the advent of the “stem cell” era. Our inherent understanding indicates that stem cells have been in existence since the birth of multicellular organisms. However, the formal discovery of stem cells in the last century, followed by their intricate and extensive analysis, has led to clinical and translational efforts with the aim of using them in the treatment of conditions which don’t have a definitive therapeutic strategy, has fueled our interest and expectations. Technological advances in our ability to study their cellular components in depth, along with surface markers and other finer constituents, that were unknown until last century, have improved our understanding, leading to several novel applications. This has created a need to establish guidelines, and in that process, there are expressed understandings and views which describe cell therapy along lines similar to that of biologic products, drugs, and devices. However, the age-old wisdom of using cells as tools for curing illness should not be misled by recent knowledge, to make cell therapy using highly complex stem cells equal to factory-synthesized and reproducible chemical compounds, drugs, or devices. This article analyses the differences between these two entities from various perspectives.
Collapse
Affiliation(s)
- Senthilkumar Preethy
- Nichi-In Centre for Regenerative Medicine, Chennai, India ; Hope Foundation Trust, Chennai, India
| | - Sudhakar John
- Nichi-In Centre for Regenerative Medicine, Chennai, India
| | | | | | | | | | - Samuel J Abraham
- Nichi-In Centre for Regenerative Medicine, Chennai, India ; Yamanashi University School of Medicine, Chuo, Japan
| |
Collapse
|
36
|
Bioinspired drug delivery systems. Curr Opin Biotechnol 2013; 24:1167-73. [DOI: 10.1016/j.copbio.2013.02.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/03/2013] [Accepted: 02/11/2013] [Indexed: 01/21/2023]
|
37
|
Christ GJ, Saul JM, Furth ME, Andersson KE. The pharmacology of regenerative medicine. Pharmacol Rev 2013; 65:1091-133. [PMID: 23818131 DOI: 10.1124/pr.112.007393] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regenerative medicine is a rapidly evolving multidisciplinary, translational research enterprise whose explicit purpose is to advance technologies for the repair and replacement of damaged cells, tissues, and organs. Scientific progress in the field has been steady and expectations for its robust clinical application continue to rise. The major thesis of this review is that the pharmacological sciences will contribute critically to the accelerated translational progress and clinical utility of regenerative medicine technologies. In 2007, we coined the phrase "regenerative pharmacology" to describe the enormous possibilities that could occur at the interface between pharmacology, regenerative medicine, and tissue engineering. The operational definition of regenerative pharmacology is "the application of pharmacological sciences to accelerate, optimize, and characterize (either in vitro or in vivo) the development, maturation, and function of bioengineered and regenerating tissues." As such, regenerative pharmacology seeks to cure disease through restoration of tissue/organ function. This strategy is distinct from standard pharmacotherapy, which is often limited to the amelioration of symptoms. Our goal here is to get pharmacologists more involved in this field of research by exposing them to the tools, opportunities, challenges, and interdisciplinary expertise that will be required to ensure awareness and galvanize involvement. To this end, we illustrate ways in which the pharmacological sciences can drive future innovations in regenerative medicine and tissue engineering and thus help to revolutionize the discovery of curative therapeutics. Hopefully, the broad foundational knowledge provided herein will spark sustained conversations among experts in diverse fields of scientific research to the benefit of all.
Collapse
Affiliation(s)
- George J Christ
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | | | | | | |
Collapse
|
38
|
Zhumadilov Z. Global initiative for interdisciplinary approach to improve innovative clinical research and treatment outcomes in geriatrics: biological cell-based targeted drug delivery systems for geriatrics. Rejuvenation Res 2013; 16:212-23. [PMID: 23496161 DOI: 10.1089/rej.2013.1408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
At the intersection of the late 20(th) century and early 21(st) century, a worldwide challenge began to emerge--how can the quality of life be improved for a steadily increasing elderly population. It is well known that elderly patients show increased susceptibility to infections and a higher incidence of co-morbidity rates. Older adults frequently demonstrate pharmacokinetic and pharmacodynamic changes promoting adverse drug reactions and complications. Analysis of world literature and practical observations indicate that new approaches are required in gerontology and geriatric medicine due to recent significant advances in biomedical science. Global interdisciplinary approaches to improve medical science and medical care services for growing elderly population are indicated. This global, interdisciplinary initiative should integrate select, tangible clinical results achieved in leading research centers and universities that are applicable in the field of geriatrics and helpful to geriatricians. Among past scientific and clinically significant study results in the field of biomedicine, one must consider targeted drug delivery systems (DDS), which are designed to minimize drug side effects, increase the efficacy of drugs, and prolong and target drug interactions with particular pathological foci in sick patients. Many review articles focus on various methods of drug encapsulation and pharmacokinetics, but not on developing clinical modalities. This article attempts to further the discussion with researchers and clinicians from various fields, as well as to encourage comprehensive and elderly patient-oriented research focused on clinical implementation of DDS, especially erythrocyte-based DDS.
Collapse
|