1
|
Li Y, Shen Q, Feng L, Zhang C, Jiang X, Liu F, Pang B. A nanoscale natural drug delivery system for targeted drug delivery against ovarian cancer: action mechanism, application enlightenment and future potential. Front Immunol 2024; 15:1427573. [PMID: 39464892 PMCID: PMC11502327 DOI: 10.3389/fimmu.2024.1427573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/22/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological malignancies in the world and is the leading cause of cancer-related death in women. The complexity and difficult-to-treat nature of OC pose a huge challenge to the treatment of the disease, Therefore, it is critical to find green and sustainable drug treatment options. Natural drugs have wide sources, many targets, and high safety, and are currently recognized as ideal drugs for tumor treatment, has previously been found to have a good effect on controlling tumor progression and reducing the burden of metastasis. However, its clinical transformation is often hindered by structural stability, bioavailability, and bioactivity. Emerging technologies for the treatment of OC, such as photodynamic therapy, immunotherapy, targeted therapy, gene therapy, molecular therapy, and nanotherapy, are developing rapidly, particularly, nanotechnology can play a bridging role between different therapies, synergistically drive the complementary role of differentiated treatment schemes, and has a wide range of clinical application prospects. In this review, nanoscale natural drug delivery systems (NNDDS) for targeted drug delivery against OC were extensively explored. We reviewed the mechanism of action of natural drugs against OC, reviewed the morphological composition and delivery potential of drug nanocarriers based on the application of nanotechnology in the treatment of OC, and discussed the limitations of current NNDDS research. After elucidating these problems, it will provide a theoretical basis for future exploration of novel NNDDS for anti-OC therapy.
Collapse
Affiliation(s)
- Yi Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Shen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Feng
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanlong Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Pang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Goksen S, Varan G, Bilensoy E, Esendagli G. Folate Receptor β (FRβ) Expression on Myeloid Cells and the Impact of Reticuloendothelial System on Folate-Functionalized Nanoparticles' Biodistribution in Cancer. Mol Pharm 2024; 21:4688-4699. [PMID: 39105761 PMCID: PMC11372836 DOI: 10.1021/acs.molpharmaceut.4c00663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
Folate uptake is largely mediated by folate receptor (FR)β, encoded by FOLR2 gene, in myeloid immune cells such as granulocytes, monocytes, and especially in macrophages that constitute the reticuloendothelial system (RES) and infiltrate the tumor microenvironment. Since the myeloid immune compartment dynamically changes during tumorigenesis, it is critical to assess the infiltration status of the tumors by FRβ-expressing myeloid cells to better define the targeting efficacy of folate-functionalized drug delivery systems. On the other hand, clearance by RES is a major limitation for the targeting efficacy of nanoparticles decorated with folate. Therefore, the aims of this study are (i) to determine the amount and subtypes of FRβ+ myeloid cells infiltrating the tumors at different stages, (ii) to compare the amount and subtype of FRβ+ myeloid cells in distinct organs of tumor-bearing and healthy animals, (iii) to test if the cancer-targeting efficacy and biodistribution of a prototypic folate-functionalized nanoparticle associates with the density of FRβ+ myeloid cells. Here, we report that myeloid cell infiltration was enhanced and FRβ was upregulated at distinct stages of tumorigenesis in a mouse breast cancer model. The CD206+ subset of macrophages highly expressed FRβ, prominently both in tumor-bearing and healthy mice. In tumor-bearing mice, the amount of all myeloid cells, but particularly granulocytes, was remarkably increased in the tumor, liver, lungs, spleen, kidneys, lymph nodes, peritoneal cavity, bone marrow, heart, and brain. Compared with macrophages, the level of FRβ was moderate in granulocytes and monocytes. The density of FRβ+ immune cells in the tumor microenvironment was not directly associated with the tumor-targeting efficacy of the folate-functionalized cyclodextrin nanoparticles. The lung was determined as a preferential site of accumulation for folate-functionalized nanoparticles, wherein FRβ+CD206+ macrophages significantly engulfed cyclodextrin nanoparticles. In conclusion, our results demonstrate that the tumor formation augments the FR levels and alters the infiltration and distribution of myeloid immune cells in all organs which should be considered as a major factor influencing the targeting efficacy of nanoparticles for drug delivery.
Collapse
Affiliation(s)
- Sibel Goksen
- Department
of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara 06100, Türkiye
| | - Gamze Varan
- Department
of Vaccine Technology, Hacettepe University
Vaccine Institute, Ankara 06100, Türkiye
| | - Erem Bilensoy
- Department
of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Türkiye
| | - Gunes Esendagli
- Department
of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara 06100, Türkiye
- Department
of Vaccinology, Hacettepe University Vaccine
Institute, Ankara 06100, Türkiye
- Department
of Basic Oncology, Hacettepe University
Cancer Institute, Ankara 06100, Türkiye
| |
Collapse
|
3
|
Jacob EM, Huang J, Chen M. Lipid nanoparticle-based mRNA vaccines: a new frontier in precision oncology. PRECISION CLINICAL MEDICINE 2024; 7:pbae017. [PMID: 39171210 PMCID: PMC11336688 DOI: 10.1093/pcmedi/pbae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
The delivery of lipid nanoparticle (LNP)-based mRNA therapeutics has captured the attention of the vaccine research community as an innovative and versatile tool for treating a variety of human malignancies. mRNA vaccines are now in the limelight as an alternative to conventional vaccines owing to their high precision, low-cost, rapid manufacture, and superior safety profile. Multiple mRNA vaccine platforms have been developed to target several types of cancer, and many have demonstrated encouraging results in animal models and human trials. The effectiveness of these new mRNA vaccines depends on the efficacy and stability of the antigen(s) of interest generated and the reliability of their delivery to antigen-presenting cells (APCs), especially dendritic cells (DCs). In this review, we provide a detailed overview of mRNA vaccines and their delivery strategies and consider future directions and challenges in advancing and expanding this promising vaccine platform to widespread therapeutic use against cancer.
Collapse
Affiliation(s)
- Eden M Jacob
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Ming Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| |
Collapse
|
4
|
Hu X, Ye K, Bo S, Xiao Z, Ma M, Pan J, Zhong X, Zhang D, Mo X, Yu X, Chen M, Luo L, Shi C. Monitoring imatinib decreasing pericyte coverage and HIF-1α level in a colorectal cancer model by an ultrahigh-field multiparametric MRI approach. J Transl Med 2024; 22:712. [PMID: 39085929 PMCID: PMC11293104 DOI: 10.1186/s12967-024-05497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Excessive pericyte coverage promotes tumor growth, and a downregulation may solve this dilemma. Due to the double-edged sword role of vascular pericytes in tumor microenvironment (TME), indiscriminately decreasing pericyte coverage by imatinib causes poor treatment outcomes. Here, we optimized the use of imatinib in a colorectal cancer (CRC) model in high pericyte-coverage status, and revealed the value of multiparametric magnetic resonance imaging (mpMRI) at 9.4T in monitoring treatment-related changes in pericyte coverage and the TME. METHODS CRC xenograft models were evaluated by histological vascular characterizations and mpMRI. Mice with the highest pericyte coverage were treated with imatinib or saline; then, vascular characterizations, tumor apoptosis and HIF-1α level were analyzed histologically, and alterations in the expression of Bcl-2/bax pathway were assessed through qPCR. The effects of imatinib were monitored by dynamic contrast-enhanced (DCE)-, diffusion-weighted imaging (DWI)- and amide proton transfer chemical exchange saturation transfer (APT CEST)-MRI at 9.4T. RESULTS The DCE- parameters provided a good histologic match the tumor vascular characterizations. In the high pericyte coverage status, imatinib exhibited significant tumor growth inhibition, necrosis increase and pericyte coverage downregulation, and these changes were accompanied by increased vessel permeability, decreased microvessel density (MVD), increased tumor apoptosis and altered gene expression of apoptosis-related Bcl-2/bax pathway. Strategically, a 4-day imatinib effectively decreased pericyte coverage and HIF-1α level, and continuous treatment led to a less marked decrease in pericyte coverage and re-elevated HIF-1α level. Correlation analysis confirmed the feasibility of using mpMRI parameters to monitor imatinib treatment, with DCE-derived Ve and Ktrans being most correlated with pericyte coverage, Ve with vessel permeability, AUC with microvessel density (MVD), DWI-derived ADC with tumor apoptosis, and APT CEST-derived MTRasym at 1 µT with HIF-1α. CONCLUSIONS These results provided an optimized imatinib regimen to achieve decreasing pericyte coverage and HIF-1α level in the high pericyte-coverage CRC model, and offered an ultrahigh-field multiparametric MRI approach for monitoring pericyte coverage and dynamics response of the TME to treatment.
Collapse
Affiliation(s)
- Xinpeng Hu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Kunlin Ye
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Shaowei Bo
- Department of Medical Imaging, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Xingang Middle Road No. 466, Guangzhou, 510317, China
| | - Zeyu Xiao
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Mengjie Ma
- Department of Radiology, Guangzhou First People's Hospital, Panfu Road No. 1, Guangzhou, 510080, China
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xing Zhong
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Dong Zhang
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xukai Mo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xiaojun Yu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, West Huangpu Avenue No.601, Guangzhou, 510632, China.
| | - Liangping Luo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
| | - Changzheng Shi
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
| |
Collapse
|
5
|
Paolino D, d'Avanzo N, Canato E, Ciriolo L, Grigoletto A, Cristiano MC, Mancuso A, Celia C, Pasut G, Fresta M. Improved anti-breast cancer activity by doxorubicin-loaded super stealth liposomes. Biomater Sci 2024; 12:3933-3946. [PMID: 38940612 DOI: 10.1039/d4bm00478g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
PEGylation is currently used for the synthesis of stealth liposomes and to enhance the pharmacokinetic and biopharmaceutical properties of payloads. PEGylated dendron phospholipids can decrease the detachment of polyethylene glycol (PEG) from the liposomal surface owing to an increased hydrophobic anchoring effect on the phospholipid bilayer of liposomes and thus generating super stealth liposomes that are suitable for the systemic delivery of anticancer drugs. Herein, doxorubicin hydrochloride-loaded super stealth liposomes were studied for the treatment of breast cancer lung metastasis in an animal model. The results demonstrated that the super stealth liposomes had suitable physicochemical properties for in vivo administration and could significantly increase the efficacy of doxorubicin in breast cancer lung metastasis tumor-bearing mice compared to the free drug. The super stealth liposomes also increased doxorubicin accumulation inside the tumor tissue. The permanence of PEG on the surface of the super stealth liposomes favored the formation of a depot of therapeutic nanocarriers inside the tumor tissue by improving their permanence after stopping treatment. The doxorubicin-loaded super stealth liposomes increased the survival of the mouse tumor model. These promising results demonstrate that the doxorubicin-loaded super stealth liposomes could be an effective nanomedicine to treat metastatic breast cancer.
Collapse
Affiliation(s)
- Donatella Paolino
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
- Research Center "ProHealth Translational Hub", Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-88100 Catanzaro, Italy
| | - Nicola d'Avanzo
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
- Research Center "ProHealth Translational Hub", Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-88100 Catanzaro, Italy
| | - Elena Canato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, I-35131 Padua, Italy.
| | - Luigi Ciriolo
- Department of Health Science, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, I-35131 Padua, Italy.
| | - Maria Chiara Cristiano
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-Catanzaro, Italy
| | - Antonia Mancuso
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
- Research Center "ProHealth Translational Hub", Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-88100 Catanzaro, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100, Chieti, Italy.
- Lithuanian University of Health Sciences, Laboratory of Drug Targets Histopathology, Institute of Cardiology, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, I-35131 Padua, Italy.
| | - Massimo Fresta
- Department of Health Science, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
| |
Collapse
|
6
|
Han L, Zhu J, Jones KL, Yang J, Zhai R, Cao J, Hu B. Fabrication and functional application of zein-based core-shell structures: A review. Int J Biol Macromol 2024; 272:132796. [PMID: 38823740 DOI: 10.1016/j.ijbiomac.2024.132796] [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: 08/25/2023] [Revised: 03/07/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Core-shell structures exhibit a number of distinct absorptive properties that make them attractive tools for use in a range of industrial contexts including pharmaceuticals, biotechnology, cosmetics, and food/agriculture. Several recent studies have focused on the development and fabrication of zein-based core-shell structures for a range of functional material deliveries. However, no recent review article has evaluated the fabrication of such core-shell structures for food-based applications. In this paper, we therefore survey current approaches to fabricating different zein-based platforms including particles, fibers, films, and hydrogels that have appeared in a variety of functionally relevant applications. In addition, we highlight certain challenges and future research directions in this field, thereby providing a novel perspective on zein-based core-shell structures.
Collapse
Affiliation(s)
- Lingyu Han
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning 116600, China
| | - Junzhe Zhu
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning 116600, China
| | - Kevin L Jones
- Faculty of Arts, Science and Technology, Wrexham Glyndwr University, Plas Coch, Mold Road, Wrexham LL11 2AW, United Kingdom
| | - Jixin Yang
- Faculty of Arts, Science and Technology, Wrexham Glyndwr University, Plas Coch, Mold Road, Wrexham LL11 2AW, United Kingdom
| | - Ruiyi Zhai
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning 116600, China
| | - Jijuan Cao
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning 116600, China.
| | - Bing Hu
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning 116600, China.
| |
Collapse
|
7
|
Tai Y, Chen M, Wang F, Fan Y, Zhang J, Cai B, Yan L, Luo Y, Li Y. The role of dendritic cells in cancer immunity and therapeutic strategies. Int Immunopharmacol 2024; 128:111548. [PMID: 38244518 DOI: 10.1016/j.intimp.2024.111548] [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: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Dendritic cells (DCs) are asserted as the most potent antigen-presenting cells (APCs) that orchestrate both innate and adaptive immunity, being extremely effective in the induction of robust anti-cancer T cell responses. Hence, the modulation of DCs function represents an attractive target for improving cancer immunotherapy efficacy. A better understanding of the immunobiology of DCs, the interaction among DCs, immune effector cells and tumor cells in tumor microenvironment (TME) and the latest advances in biomedical engineering technology would be required for the design of optimal DC-based immunotherapy. In this review, we focus on elaborating the immunobiology of DCs in healthy and cancer environments, the recent advances in the development of enhancing endogenous DCs immunocompetence via immunomodulators as well as DC-based vaccines. The rapidly developing field of applying nanotechnology to improve DC-based immunotherapy is also highlighted.
Collapse
Affiliation(s)
- Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Man Chen
- Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Fang Wang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, China
| | - Yu Fan
- Department of Urology, National Clinical Research Center for Geriatrics and Organ Transplantation Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yao Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Karimova A, Hajizada S, Shirinova H, Nuriyeva S, Gahramanli L, Yusuf MM, Bellucci S, Reissfelder C, Yagublu V. Surface Modification Strategies for Chrysin-Loaded Iron Oxide Nanoparticles to Boost Their Anti-Tumor Efficacy in Human Colon Carcinoma Cells. J Funct Biomater 2024; 15:43. [PMID: 38391896 PMCID: PMC10889794 DOI: 10.3390/jfb15020043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Enhancing nanoparticles' anti-cancer capabilities as drug carriers requires the careful adjustment of formulation parameters, including loading efficiency, drug/carrier ratio, and synthesis method. Small adjustments to these parameters can significantly influence the drug-loading efficiency of nanoparticles. Our study explored how chitosan and polyethylene glycol (PEG) coatings affect the structural properties, drug-loading efficiency, and anti-cancer efficacy of Fe3O4 nanoparticles (NPs). The loading efficiency of the NPs was determined using FTIR spectrometry and XRD. The quantity of chrysin incorporated into the coated NPs was examined using UV-Vis spectrometry. The effect of the NPs on cell viability and apoptosis was determined by employing the HCT 116 human colon carcinoma cell line. We showed that a two-fold increase in drug concentration did not impact the loading efficiency of Fe3O4 NPs coated with PEG. However, there was a 33 Å difference in the crystallite sizes obtained from chitosan-coated Fe3O4 NPs and drug concentrations of 1:0.5 and 1:2, resulting in decreased system stability. In conclusion, PEG coating exhibited a higher loading efficiency of Fe3O4 NPs compared to chitosan, resulting in enhanced anti-tumor effects. Furthermore, variations in the loaded amount of chrysin did not impact the crystallinity of PEG-coated NPs, emphasizing the stability and regularity of the system.
Collapse
Affiliation(s)
- Aynura Karimova
- Nanoresearch Laboratory, Baku State University, Baku AZ 1148, Azerbaijan
| | - Sabina Hajizada
- Department of Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Habiba Shirinova
- Nanoresearch Laboratory, Baku State University, Baku AZ 1148, Azerbaijan
| | - Sevinj Nuriyeva
- Nanoresearch Laboratory, Baku State University, Baku AZ 1148, Azerbaijan
| | - Lala Gahramanli
- Nanoresearch Laboratory, Baku State University, Baku AZ 1148, Azerbaijan
| | - Mohammed M Yusuf
- Department of Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Stefano Bellucci
- Laboratori Nazionali di Frascati, Istituto Nazionale di Fisica Nucleare, 00044 Frascati, Italy
| | - Christoph Reissfelder
- Department of Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Vugar Yagublu
- Department of Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
9
|
Barmoudeh Z, Fouani MH, Moslemi Z, Azizi M, Doustimotlagh AH, Bardania H. Melatonin and metformin co-loaded nanoliposomes efficiently attenuate liver damage induced by bile duct ligation in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:395-410. [PMID: 37452836 DOI: 10.1007/s00210-023-02613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
In the current study, the therapeutic effectiveness of the metformin (Met) and melatonin (Mel) co-loaded liposomes was investigated on cholestasis induced by bile duct ligation (BDL) in male rats. Histopathological analysis, biochemical analysis, and oxidative stress markers were assayed to determine the therapeutic effect of Met and Mel co-loaded liposomes on cholestasis. Histopathological analysis revealed that the simultaneous administration of Met and Mel, whether in the free (C-Mel-Met) or liposomal (C-Lipo-Mel-Met) forms, reduced inflammation as well as proliferation of bile ducts; however, results were more prominent in the liposomal form of Mel and Met. Additionaly, serum levels of aspartate aminotransferase (AST) were significantly (p < 0.001) higher in (C-Mel-Met) treated rats compared with (BDL) rats; however, (C-Lipo-Mel-Met) treated rats exhibited significant (p < 0.05) lower AST rates in comparison to (BDL) rats. Moreover, a significant (p < 0.0001) drop in bilirubin levels was detected in (C-Lipo-Mel-Met) treated rats in comparison to (BDL) rats; it is noteworthy mentioning that bilirubin levels in (C-Lipo-Mel-Met) treated rats were insignificant in comparison to sham-control (SC) rats. Furthermore, rats concomitantly administered Met and Mel, exhibited significant downregulation in the expression levels of inflammatory cytokine genes such as TNF-α and IL-1 gene expression, where the downregulation was more prominent in the liposomal from. Our findings demonestrate that the concomitant administration of metformin and melatonin in the liposomal form had more therapeutic effect on liver injury than their free forms through improving histological changes, reducing biochemical markers and favoring oxidant- antioxidant balance.
Collapse
Affiliation(s)
- Zahra Barmoudeh
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohamad Hassan Fouani
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Moslemi
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahdokht Azizi
- Clinical Research Development Unit, Imamsajad Hospital, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Amir Hossein Doustimotlagh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
10
|
Kandi MTS, Meshkat SS, Hosseinzadeh S, Behroozsarand A. Synthesis & characterization of novel MIP with RAFT polymerization of (2-hydroxy ethyl methacrylate)/chitosan as a nanocarrier for drug delivery applications. Int J Biol Macromol 2023; 250:126052. [PMID: 37517749 DOI: 10.1016/j.ijbiomac.2023.126052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/17/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
The primary function of the drug delivery system is to transfer various drugs to certain parts of the body. The drug is transferred reliably in the molecularity imprinted system, based on adjusting the drug release mechanism to control the drug amount and treatment duration. Molecular Imprinting Technology can provide an efficient polymer system to detect bioactive molecules and has high adsorption capacity as drug delivery carriers. This study developed a nanocarrier of molecular imprinted polymer (MIP) of poly (2-hydroxy ethyl methacrylate)/chitosan nanocomposite and also evaluated their performance for drug loading and release in the buffer blood medium. Nanocarriers were prepared based on the RAFT polymerization technique, and cefixime was applied to evaluate the load and release of drugs in nanocarriers. Crucial parameters such as the ratio of imprinted to functional monomer and also the ratio of functional monomer to cross-link should be assessed to obtain the best performance of the MIP. Each of these parameters was studied for four different ratios. TEM analysis showed that the particle size of optimum MIP was between 15 and 20 nm. The specific surface area of CH-CEMIP (chitosan-cefixime MIP) and CEMIP (cefixime MIP) samples were 7.53 and 6.32 m2/g, respectively. Comparing these amounts and the specific surface area of the (Non-Imprinted) NIP sample (1.87 m2/g) indicated that special diagnostic pores were generated. In the drug loading process, the CH-CEMIP sample with 82 mg/g could link to the drug more than CEMIP and NIP samples due to its high selectivity property. Furthermore, according to the drug release experiments, the CH-CEMIP sample performed better in 250 h as 84 % of cefixime in this duration was released slowly and steadily.
Collapse
|
11
|
Hicks MR, Liu X, Young CS, Saleh K, Ji Y, Jiang J, Emami MR, Mokhonova E, Spencer MJ, Meng H, Pyle AD. Nanoparticles systemically biodistribute to regenerating skeletal muscle in DMD. J Nanobiotechnology 2023; 21:303. [PMID: 37641124 PMCID: PMC10463982 DOI: 10.1186/s12951-023-01994-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 07/09/2023] [Indexed: 08/31/2023] Open
Abstract
Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.
Collapse
Affiliation(s)
- Michael R Hicks
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Xiangsheng Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Courtney S Young
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- MyoGene Bio, San Diego, CA, USA
| | - Kholoud Saleh
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ying Ji
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Michael R Emami
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ekaterina Mokhonova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melissa J Spencer
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Huan Meng
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China.
| | - April D Pyle
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Xie Q, Tang J, Guo S, Zhao Q, Li S. Recent Progress of Preparation Strategies in Organic Nanoparticles for Cancer Phototherapeutics. Molecules 2023; 28:6038. [PMID: 37630290 PMCID: PMC10459389 DOI: 10.3390/molecules28166038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/27/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Phototherapy has the advantages of being a highly targeted, less toxic, less invasive, and repeatable treatment, compared with conventional treatment methods such as surgery, chemotherapy, and radiotherapy. The preparation strategies are significant in order to determine the physical and chemical properties of nanoparticles. However, choosing appropriate preparation strategies to meet applications is still challenging. This review summarizes the recent progress of preparation strategies in organic nanoparticles, mainly focusing on the principles, methods, and advantages of nanopreparation strategies. In addition, typical examples of cancer phototherapeutics are introduced in detail to inform the choice of appropriate preparation strategies. The relative future trend and outlook are preliminarily proposed.
Collapse
Affiliation(s)
| | | | | | - Qi Zhao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (Q.X.); (J.T.); (S.G.)
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (Q.X.); (J.T.); (S.G.)
| |
Collapse
|
13
|
Dai Z, Zhang Y, Meng Y, Li S, Suonan Z, Sun Y, Ji J, Shen Q, Zheng H, Xue Y. Targeted delivery of nutraceuticals derived from food for the treatment of obesity and its related complications. Food Chem 2023; 418:135980. [PMID: 36989644 DOI: 10.1016/j.foodchem.2023.135980] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Nutraceuticals which are abundant in foods have attracted much attention due to their bioactive activities of anti-obesity, anti-hyperlipidemia and anti-atherosclerosis. Unfortunately, the poor bioavailability severely undermines their envisioned benefits. Therefore, there is an urgent need to develop suitable delivery systems to promote the benefits of their biological activity. Targeted drug delivery system (TDDS) is a novel drug delivery system that can selectively concentrate drugs on targets in the body, improve the bioavailability of agents and reduce side effects. This emerging drug delivery system provides a new strategy for the treatment of obesity with nutraceuticals and would be a promising alternative to be widely used in the food field. This review summarizes the recent studies on the application in the targeted delivery of nutraceuticals for treating obesity and its related complications, especially the available receptors and their corresponding ligands for TDDS and the evaluation methods of the targeting ability.
Collapse
|
14
|
Yang J, Dong X, Li B, Chen T, Yu B, Wang X, Dou X, Peng B, Hu Q. Poria cocos polysaccharide-functionalized graphene oxide nanosheet induces efficient cancer immunotherapy in mice. Front Bioeng Biotechnol 2023; 10:1050077. [PMID: 36727039 PMCID: PMC9885324 DOI: 10.3389/fbioe.2022.1050077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction: Tumor vaccines that induce robust humoral and cellular immune responses have attracted tremendous interest for cancer immunotherapy. Despite the tremendous potential of tumor vaccines as an effective approach for cancer treatment and prevention, a major challenge in achieving sustained antitumor immunity is inefficient antigen delivery to secondary lymphoid organs, even with adjuvant aid. Methods: Herein, we present antigen/adjuvant integrated nanocomplexes termed nsGO/PCP/OVA by employing graphene oxide nanosheet (nsGO) as antigen nanocarriers loaded with model antigen ovalbumin (OVA) and adjuvant, Poria cocos polysaccharides (PCP). We evaluated the efficacy of nsGO/PCP/OVA in activating antigen-specific humoral as well as cellular immune responses and consequent tumor prevention and rejection in vivo. Results: The optimally formed nsGO/PCP/OVA was approximately 120-150 nm in diameter with a uniform size distribution. Nanoparticles can be effectively engulfed by dendritic cells (DCs) through receptor-mediated endocytosis, induced the maturation of DCs and improved the delivery efficiency both in vitro and in vivo. The nsGO/PCP/OVA nanoparticles also induced a significant enhancement of OVA antigen-specific Th1 and Th2 immune responses in vivo. In addition, vaccination with nsGO/PCP/OVA not only significantly suppressed tumor growth in prophylactic treatments, but also achieved a therapeutic effect in inhibiting the growth of already-established tumors. Conclusion: Therefore, this potent nanovaccine platform with nanocarrier nsGO and PCP as adjuvants provides a promising strategy for boosting anti-tumor immunity for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinning Yang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China
| | - Xiaoxiao Dong
- Institute of Medical Biotechnology, Chinese Academy of Medical Sciences, Beijing, China
| | - Boye Li
- Civil Aviation Medicine Center, Civil Aviation Administration of China, Beijing, China
| | - Tian Chen
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China
| | - Boyang Yu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China
| | - Xiaoli Wang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| | - Xiangnan Dou
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| | - Bo Peng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| | - Qin Hu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, China,Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, China,*Correspondence: Xiaoli Wang, ; Xiangnan Dou, ; Bo Peng, ; Qin Hu,
| |
Collapse
|
15
|
Bal-Öztürk A, Tietilu ŞD, Yücel O, Erol T, Akgüner ZP, Darıcı H, Alarcin E, Emik S. Hyperbranched polymer-based nanoparticle drug delivery platform for the nucleus-targeting in cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
16
|
Bezbaruah R, Chavda VP, Nongrang L, Alom S, Deka K, Kalita T, Ali F, Bhattacharjee B, Vora L. Nanoparticle-Based Delivery Systems for Vaccines. Vaccines (Basel) 2022; 10:1946. [PMID: 36423041 PMCID: PMC9694785 DOI: 10.3390/vaccines10111946] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
Vaccination is still the most cost-effective way to combat infectious illnesses. Conventional vaccinations may have low immunogenicity and, in most situations, only provide partial protection. A new class of nanoparticle-based vaccinations has shown considerable promise in addressing the majority of the shortcomings of traditional and subunit vaccines. This is due to recent breakthroughs in chemical and biological engineering, which allow for the exact regulation of nanoparticle size, shape, functionality, and surface characteristics, resulting in improved antigen presentation and robust immunogenicity. A blend of physicochemical, immunological, and toxicological experiments can be used to accurately characterize nanovaccines. This narrative review will provide an overview of the current scenario of the nanovaccine.
Collapse
Affiliation(s)
- Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Lawandashisha Nongrang
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | - Kangkan Deka
- Department of Pharmacognosy, NETES Institute of Pharmaceutical Science, Mirza, Guwahati 781125, Assam, India
| | - Tutumoni Kalita
- Department of Pharmaceutical Chemistry, Girijananda Chowdhury Institute of Pharmaceutical Sciences, Azara, Guwahati 781017, Assam, India
| | - Farak Ali
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
- Department of Pharmaceutical Chemistry, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | | |
Collapse
|
17
|
Nanocarriers: A Reliable Tool for the Delivery of Anticancer Drugs. Pharmaceutics 2022; 14:pharmaceutics14081566. [PMID: 36015192 PMCID: PMC9415391 DOI: 10.3390/pharmaceutics14081566] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/26/2022] Open
Abstract
Nanomedicines have gained popularity due to their potential therapeutic applications, especially cancer treatment. Targeted nanoparticles can deliver drugs directly to cancer cells and enable prolonged drug release, reducing off-target toxicity and increasing therapeutic efficacy. However, translating nanomedicines from preclinical to clinical settings has been difficult. Rapid advancements in nanotechnology promise to enhance cancer therapies. Nanomedicine offers advanced targeting and multifunctionality. Nanoparticles (NPs) have several uses nowadays. They have been studied as drug transporters, tumor gene delivery agents, and imaging contrast agents. Nanomaterials based on organic, inorganic, lipid, or glycan substances and synthetic polymers have been used to enhance cancer therapies. This review focuses on polymeric nanoparticle delivery strategies for anticancer nanomedicines.
Collapse
|
18
|
Howard RL, Bernardi F, Leff M, Abele E, Allbritton NL, Harris DM. Passive Control of Silane Diffusion for Gradient Application of Surface Properties. MICROMACHINES 2021; 12:1360. [PMID: 34832772 PMCID: PMC8620173 DOI: 10.3390/mi12111360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/30/2022]
Abstract
Liquid lithography represents a robust technique for fabricating three-dimensional (3D) microstructures on a two-dimensional template. Silanization of a surface is often a key step in the liquid lithography process and is used to alter the surface energy of the substrate and, consequently, the shape of the 3D microfeatures produced. In this work, we present a passive technique that allows for the generation of silane gradients along the length of a substrate. The technique relies on a secondary diffusion chamber with a single opening, leading to a directional introduction of silane to the substrate via passive diffusion. The secondary chamber geometry influences the deposited gradient, which is shown to be well captured by Monte Carlo simulations that incorporate the passive diffusion and grafting processes. The technique ultimately allows the user to generate a range of substrate wettabilities on a single chip, enhancing throughput for organ-on-a-chip applications by mimicking the spatial variability of tissue topographies present in vivo.
Collapse
Affiliation(s)
- Riley L. Howard
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Francesca Bernardi
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Matthew Leff
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Emma Abele
- School of Engineering, Brown University, Providence, RI 02912, USA; (E.A.); (D.M.H.)
| | - Nancy L. Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| | - Daniel M. Harris
- School of Engineering, Brown University, Providence, RI 02912, USA; (E.A.); (D.M.H.)
| |
Collapse
|
19
|
Thurner GC, Haybaeck J, Debbage P. Targeting Drug Delivery in the Elderly: Are Nanoparticles an Option for Treating Osteoporosis? Int J Mol Sci 2021; 22:8932. [PMID: 34445639 PMCID: PMC8396227 DOI: 10.3390/ijms22168932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles bearing specific targeting groups can, in principle, accumulate exclusively at lesion sites bearing target molecules, and release therapeutic agents there. However, practical application of targeted nanoparticles in the living organism presents challenges. In particular, intravasally applied nanoparticles encounter physical and physiological barriers located in blood vessel walls, blocking passage from the blood into tissue compartments. Whereas small molecules can pass out of the blood, nanoparticles are too large and need to utilize physiological carriers enabling passage across endothelial walls. The issues associated with crossing blood-tissue barriers have limited the usefulness of nanoparticles in clinical applications. However, nanoparticles do not encounter blood-tissue barriers if their targets are directly accessible from the blood. This review focuses on osteoporosis, a disabling and common disease for which therapeutic strategies are limited. The target sites for therapeutic agents in osteoporosis are located in bone resorption pits, and these are in immediate contact with the blood. There are specific targetable biomarkers within bone resorption pits. These present nanomedicine with the opportunity to treat a major disease by use of simple nanoparticles loaded with any of several available effective therapeutics that, at present, cannot be used due to their associated side effects.
Collapse
Affiliation(s)
- Gudrun C. Thurner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria;
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria;
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Paul Debbage
- Department of Anatomy, Histology and Embryology, Medical University of Innsbruck, Müllerstraße 59, 6020 Innsbruck, Austria
| |
Collapse
|
20
|
Wang Y, Zhang Z, Zheng C, Zhao X, Zheng Y, Liu Q, Liu Y, Shi L. Multistage Adaptive Nanoparticle Overcomes Biological Barriers for Effective Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100578. [PMID: 34190401 DOI: 10.1002/smll.202100578] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/31/2021] [Indexed: 06/13/2023]
Abstract
Drug delivery systems (DDS) are extensively studied to improve the solubility, stability, pharmacokinetic, and biodistribution of chemotherapeutics. However, the drug delivery efficiency of traditional DDS is often limited by the complicated biological barriers in vivo. Herein, a multistage adaptive nanoparticle (MAN) that simultaneously overcomes multiple biological barriers to achieve tumor-targeted drug delivery with high efficiency is presented. MAN has a core-shell structure, in which both the core and the shell are made of responsive polymers. This structure allows MAN to present different surface properties to adapt to its surrounding biological microenvironment, thereby achieving enhanced stability in blood circulation, improved tumor accumulation and cellular internalization in tumor tissues, and effective release of drug in cells. With these unique characteristics, the MAN loaded with docetaxel achieves effective tumor suppression with reduced systemic toxicity. Furthermore, MAN can load almost any hydrophobic drugs, providing a general strategy for the tumor-targeted delivery of hydrophobic drugs to overcome the multiple biological barriers and improve the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Ying Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Zhanzhan Zhang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Chunxiong Zheng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xinzhi Zhao
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yadan Zheng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Qi Liu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yang Liu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Linqi Shi
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
21
|
Kucuksayan E, Bozkurt F, Yilmaz MT, Sircan-Kucuksayan A, Hanikoglu A, Ozben T. A new combination strategy to enhance apoptosis in cancer cells by using nanoparticles as biocompatible drug delivery carriers. Sci Rep 2021; 11:13027. [PMID: 34158544 PMCID: PMC8219778 DOI: 10.1038/s41598-021-92447-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/27/2021] [Indexed: 12/21/2022] Open
Abstract
Some experimental and clinical studies have been conducted for the usage of chemotherapeutic drugs encapsulated into nanoparticles (NPs). However, no study has been conducted so far on the co-encapsulation of doxorubicin (Dox) and epoxomicin (Epo) into NPs as biocompatible drug delivery carriers. Therefore, we investigated if co-encapsulation of doxorubicin (Dox) and/or epoxomicin (Epo) into NPs enhance their anticancer efficiency and prevent drug resistance and toxicity to normal cells. We synthesized Dox and/or Epo loaded poly (lactic-co-glycolic acid) (PLGA) NPs using a multiple emulsion solvent evaporation technique and characterized them in terms of their particle size and stability, surface, molecular, thermal, encapsulation efficiency and in vitro release properties. We studied the effects of drug encapsulated NPs on cellular accumulation, intracellular drug levels, oxidative stress status, cellular viability, drug resistance, 20S proteasome activity, cytosolic Nuclear Factor Kappa B (NF-κB-p65), and apoptosis in breast cancer and normal cells. Our results proved that the nanoparticles we synthesized were thermally stable possessing higher encapsulation efficiency and particle stability. Thermal, morphological and molecular analyses demonstrated the presence of Dox and/or Epo within NPs, indicating that they were successfully loaded. Cell line assays proved that Dox and Epo loaded NPs were less cytotoxic to single-layer normal HUVECs than free Dox and Epo, suggesting that the NPs would be biocompatible drug delivery carriers. The apoptotic index of free Dox and Epo increased 50% through their encapsulation into NPs, proving combination strategy to enhance apoptosis in breast cancer cells. Our results demonstrated that the co-encapsulation of Dox and Epo within NPs would be a promising treatment strategy to overcome multidrug resistance and toxicity to normal tissues that can be studied in further in vivo and clinical studies in breast cancer.
Collapse
Affiliation(s)
- Ertan Kucuksayan
- Faculty of Medicine, Department of Medical Biochemistry, Alanya Alaaddin Keykubat University (ALKU), Antalya, 07490, Turkey.,Chemical and Metallurgical Engineering Faculty, Department of Food Engineering, Yildiz Technical University, Istanbul, Turkey.,Faculty of Medicine, Department of Medical Biochemistry, Akdeniz University, Antalya, Turkey
| | - Fatih Bozkurt
- Chemical and Metallurgical Engineering Faculty, Department of Food Engineering, Yildiz Technical University, Istanbul, Turkey.,Faculty of Engineering and Architecture, Department of Food Engineering, Mus Alparslan University, Mus, Turkey
| | - Mustafa Tahsin Yilmaz
- Chemical and Metallurgical Engineering Faculty, Department of Food Engineering, Yildiz Technical University, Istanbul, Turkey.,Faculty of Engineering, Department of Industrial Engineering, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Aslinur Sircan-Kucuksayan
- Faculty of Medicine, Department of Biophysics, Alanya Alaaddin Keykubat University (ALKU), Antalya, 07490, Turkey
| | - Aysegul Hanikoglu
- Faculty of Medicine, Department of Medical Biochemistry, Akdeniz University, Antalya, Turkey
| | - Tomris Ozben
- Faculty of Medicine, Department of Medical Biochemistry, Akdeniz University, Antalya, Turkey.
| |
Collapse
|
22
|
Rodríguez-Castejón J, Alarcia-Lacalle A, Gómez-Aguado I, Vicente-Pascual M, Solinís Aspiazu MÁ, del Pozo-Rodríguez A, Rodríguez-Gascón A. α-Galactosidase A Augmentation by Non-Viral Gene Therapy: Evaluation in Fabry Disease Mice. Pharmaceutics 2021; 13:771. [PMID: 34064206 PMCID: PMC8224287 DOI: 10.3390/pharmaceutics13060771] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/28/2022] Open
Abstract
Fabry disease (FD) is a monogenic X-linked lysosomal storage disorder caused by a deficiency in the lysosomal enzyme α-Galactosidase A (α-Gal A). It is a good candidate to be treated with gene therapy, in which moderately low levels of enzyme activity should be sufficient for clinical efficacy. In the present work we have evaluated the efficacy of a non-viral vector based on solid lipid nanoparticles (SLN) to increase α-Gal A activity in an FD mouse model after intravenous administration. The SLN-based vector incremented α-Gal A activity to about 10%, 15%, 20% and 14% of the levels of the wild-type in liver, spleen, heart and kidney, respectively. In addition, the SLN-based vector significantly increased α-Gal A activity with respect to the naked pDNA used as a control in plasma, heart and kidney. The administration of a dose per week for three weeks was more effective than a single-dose administration. Administration of the SLN-based vector did not increase liver transaminases, indicative of a lack of toxicity. Additional studies are necessary to optimize the efficacy of the system; however, these results reinforce the potential of lipid-based nanocarriers to treat FD by gene therapy.
Collapse
Affiliation(s)
- Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| | - Ana Alarcia-Lacalle
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| | - Itziar Gómez-Aguado
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| | - Mónica Vicente-Pascual
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| | - María Ángeles Solinís Aspiazu
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| | - Ana del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Centro de investigación Lascaray ikergunea, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (J.R.-C.); (A.A.-L.); (I.G.-A.); (M.V.-P.); (M.Á.S.A.)
| |
Collapse
|
23
|
Oh HJ, Kim J, Kim H, Choi N, Chung S. Microfluidic Reconstitution of Tumor Microenvironment for Nanomedical Applications. Adv Healthc Mater 2021; 10:e2002122. [PMID: 33576178 DOI: 10.1002/adhm.202002122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Indexed: 12/17/2022]
Abstract
Nanoparticles have an extensive range of diagnostic and therapeutic applications in cancer treatment. However, their current clinical translation is slow, mainly due to the failure to develop preclinical evaluation techniques that can draw similar conclusions to clinical outcomes by adequately mimicking nanoparticle behavior in complicated tumor microenvironments (TMEs). Microfluidic methods offer significant advantages over conventional in vitro methods to resolve these challenges by recapitulating physiological cues of the TME such as the extracellular matrix, shear stress, interstitial flow, soluble factors, oxygen, and nutrient gradients. The methods are capable of de-coupling microenvironmental features, spatiotemporal controlling of experimental sequences, and high throughput readouts in situ. This progress report highlights the recent achievements of microfluidic models to reconstitute the physiological microenvironment, especially for nanomedical tools for cancer treatment.
Collapse
Affiliation(s)
- Hyun Jeong Oh
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Jaehoon Kim
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Hyunho Kim
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- Division of Bio‐Medical Science & Technology KIST School Korea University of Science and Technology (UST) Seoul 34113 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| |
Collapse
|
24
|
Chin AL, Wang X, Tong R. Aliphatic Polyester-Based Materials for Enhanced Cancer Immunotherapy. Macromol Biosci 2021; 21:e2100087. [PMID: 33909344 DOI: 10.1002/mabi.202100087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/07/2021] [Indexed: 12/19/2022]
Abstract
Poly(lactic acid) (PLA) and its copolymer, poly(lactic-co-glycolic acid) (PLGA), based aliphatic polyesters have been extensively used for biomedical applications, such as drug delivery system and tissue engineering, thanks to their biodegradability, benign toxicity, renewability, and adjustable mechanical properties. A rapidly growing field of cancer research, the development of therapeutic cancer vaccines or treatment modalities is aimed to deliver immunomodulatory signals that control the quality of immune responses against tumors. Herein, the progress and applications of PLA and PLGA are reviewed in delivering immunotherapeutics to treat cancers.
Collapse
Affiliation(s)
- Ai Lin Chin
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Xiaoqian Wang
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Rong Tong
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| |
Collapse
|
25
|
Ho W, Gao M, Li F, Li Z, Zhang X, Xu X. Next-Generation Vaccines: Nanoparticle-Mediated DNA and mRNA Delivery. Adv Healthc Mater 2021; 10:e2001812. [PMID: 33458958 PMCID: PMC7995055 DOI: 10.1002/adhm.202001812] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/06/2020] [Indexed: 01/07/2023]
Abstract
Nucleic acid vaccines are a method of immunization aiming to elicit immune responses akin to live attenuated vaccines. In this method, DNA or messenger RNA (mRNA) sequences are delivered to the body to generate proteins, which mimic disease antigens to stimulate the immune response. Advantages of nucleic acid vaccines include stimulation of both cell-mediated and humoral immunity, ease of design, rapid adaptability to changing pathogen strains, and customizable multiantigen vaccines. To combat the SARS-CoV-2 pandemic, and many other diseases, nucleic acid vaccines appear to be a promising method. However, aid is needed in delivering the fragile DNA/mRNA payload. Many delivery strategies have been developed to elicit effective immune stimulation, yet no nucleic acid vaccine has been FDA-approved for human use. Nanoparticles (NPs) are one of the top candidates to mediate successful DNA/mRNA vaccine delivery due to their unique properties, including unlimited possibilities for formulations, protective capacity, simultaneous loading, and delivery potential of multiple DNA/mRNA vaccines. This review will summarize the many varieties of novel NP formulations for DNA and mRNA vaccine delivery as well as give the reader a brief synopsis of NP vaccine clinical trials. Finally, the future perspectives and challenges for NP-mediated nucleic acid vaccines will be explored.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Mingzhu Gao
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Fengqiao Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Zhongyu Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
- Department of Biomedical EngineeringNew Jersey Institute of Technology323 Dr Martin Luther King Jr BlvdNewarkNJ07102USA
| |
Collapse
|
26
|
Antifouling Strategies of Nanoparticles for Diagnostic and Therapeutic Application: A Systematic Review of the Literature. NANOMATERIALS 2021; 11:nano11030780. [PMID: 33803884 PMCID: PMC8003124 DOI: 10.3390/nano11030780] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Nanoparticles (NPs) are promising platforms for the development of diagnostic and therapeutic tools. One of the main hurdle to their medical application and translation into the clinic is the fact that they accumulate in the spleen and liver due to opsonization and scavenging by the mononuclear phagocyte system. The “protein corona” controls the fate of NPs in vivo and becomes the interface with cells, influencing their physiological response like cellular uptake and targeting efficiency. For these reasons, the surface properties play a pivotal role in fouling and antifouling behavior of particles. Therefore, surface engineering of the nanocarriers is an extremely important issue for the design of useful diagnostic and therapeutic systems. In recent decades, a huge number of studies have proposed and developed different strategies to improve antifouling features and produce NPs as safe and performing as possible. However, it is not always easy to compare the various approaches and understand their advantages and disadvantages in terms of interaction with biological systems. Here, we propose a systematic study of literature with the aim of summarizing current knowledge on promising antifouling coatings to render NPs more biocompatible and performing for diagnostic and therapeutic purposes. Thirty-nine studies from 2009 were included and investigated. Our findings have shown that two main classes of non-fouling materials (i.e., pegylated and zwitterionic) are associated with NPs and their applications are discussed here highlighting pitfalls and challenges to develop biocompatible tools for diagnostic and therapeutic uses. In conclusion, although the complexity of biofouling strategies and the field is still young, the collective data selected in this review indicate that a careful tuning of surface moieties is a pivotal step to lead NPs through their future clinical applications.
Collapse
|
27
|
Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:746. [PMID: 33809633 PMCID: PMC8000772 DOI: 10.3390/nano11030746] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
To achieve the promise of stimuli-responsive drug delivery systems for the treatment of cancer, they should (1) avoid premature clearance; (2) accumulate in tumors and undergo endocytosis by cancer cells; and (3) exhibit appropriate stimuli-responsive release of the payload. It is challenging to address all of these requirements simultaneously. However, the numerous proof-of-concept studies addressing one or more of these requirements reported every year have dramatically expanded the toolbox available for the design of drug delivery systems. This review highlights recent advances in the targeting and stimuli-responsiveness of drug delivery systems. It begins with a discussion of nanocarrier types and an overview of the factors influencing nanocarrier biodistribution. On-demand release strategies and their application to each type of nanocarrier are reviewed, including both endogenous and exogenous stimuli. Recent developments in stimuli-responsive targeting strategies are also discussed. The remaining challenges and prospective solutions in the field are discussed throughout the review, which is intended to assist researchers in overcoming interdisciplinary knowledge barriers and increase the speed of development. This review presents a nanocarrier-based drug delivery systems toolbox that enables the application of techniques across platforms and inspires researchers with interdisciplinary information to boost the development of multifunctional therapeutic nanoplatforms for cancer therapy.
Collapse
Affiliation(s)
| | - Edward Davis
- Materials Engineering Program, Mechanical Engineering Department, Auburn University, 101 Wilmore Drive, Auburn, AL 36830, USA;
| |
Collapse
|
28
|
Lin M, Zhang J, Wan H, Yan C, Xia F. Rationally Designed Multivalent Aptamers Targeting Cell Surface for Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9369-9389. [PMID: 33146988 DOI: 10.1021/acsami.0c15644] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Specific interactions between ligands and receptors on cell surface play an important role in the cell biological process. Nucleic acid aptamers as commonly used ligands enable specific recognition and tight binding to membrane protein receptors for modulation of cell fate. Therefore, molecular probes with aptamers can be applied for cancer diagnosis and targeted therapy by targeting overexpression membrane proteins of cancer cells. However, because of their fast degradation and rapid glomerulus clearance in vivo, the applications of aptamers in physiological conditions remain challenged. Inspired by natural multivalent interactions, many approaches have been developed to construct multivalent aptamers to improve the performance of aptamers in complex matrices with higher binding affinity, more stability, and longer circulation time. In this review, we first introduce the aptamer generation from purified protein-based SELEX and whole cell-based SELEX for targeting the cell surface. We then highlight the approaches to fabricate multivalent aptamers and discuss their properties. By integrating different materials (including inorganic nanomaterials, diacyllipid, polymeric nanoparticles, and DNA nanostructures) as scaffolds with an interface modification technique, we have summarized four kinds of multivalent aptamers. After that, representative applications in biosensing and targeted therapy are illustrated to show the elevated performance of multivalent aptamers. In addition, we analyze the challenges and opportunities for the clinical practices of multivalent aptamers.
Collapse
Affiliation(s)
- Meihua Lin
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Jian Zhang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Hao Wan
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Chengyang Yan
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| |
Collapse
|
29
|
Elliott RO, He M. Unlocking the Power of Exosomes for Crossing Biological Barriers in Drug Delivery. Pharmaceutics 2021; 13:pharmaceutics13010122. [PMID: 33477972 PMCID: PMC7835896 DOI: 10.3390/pharmaceutics13010122] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/25/2022] Open
Abstract
Since the 2013 Nobel Prize was awarded for the discovery of vesicle trafficking, a subgroup of nanovesicles called exosomes has been driving the research field to a new regime for understanding cellular communication. This exosome-dominated traffic control system has increased understanding of many diseases, including cancer metastasis, diabetes, and HIV. In addition to the important diagnostic role, exosomes are particularly attractive for drug delivery, due to their distinctive properties in cellular information transfer and uptake. Compared to viral and non-viral synthetic systems, the natural, cell-derived exosomes exhibit intrinsic payload and bioavailability. Most importantly, exosomes easily cross biological barriers, obstacles that continue to challenge other drug delivery nanoparticle systems. Recent emerging studies have shown numerous critical roles of exosomes in many biological barriers, including the blood–brain barrier (BBB), blood–cerebrospinal fluid barrier (BCSFB), blood–lymph barrier (BlyB), blood–air barrier (BAB), stromal barrier (SB), blood–labyrinth barrier (BLaB), blood–retinal barrier (BRB), and placental barrier (PB), which opens exciting new possibilities for using exosomes as the delivery platform. However, the systematic reviews summarizing such discoveries are still limited. This review covers state-of-the-art exosome research on crossing several important biological barriers with a focus on the current, accepted models used to explain the mechanisms of barrier crossing, including tight junctions. The potential to design and engineer exosomes to enhance delivery efficacy, leading to future applications in precision medicine and immunotherapy, is discussed.
Collapse
Affiliation(s)
- Rebekah Omarkhail Elliott
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA;
| | - Mei He
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA;
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Correspondence:
| |
Collapse
|
30
|
Ahmad MZ, Rizwanullah M, Ahmad J, Alasmary MY, Akhter MH, Abdel-Wahab BA, Warsi MH, Haque A. Progress in nanomedicine-based drug delivery in designing of chitosan nanoparticles for cancer therapy. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2020.1869737] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Md. Rizwanullah
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | | | | | - Basel A. Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine, Assiut University, Assiut, Egypt
| | - Musarrat Husain Warsi
- Department of Pharmaceutics, College of Pharmacy, Taif University, Taif, Kingdom of Saudi Arabia
| | - Anzarul Haque
- Department of Pharmacognosy, Prince Sattam bin Abdulaziz University College of Pharmacy, Alkharj Al-Kharj, Kingdom of Saudi Arabia
| |
Collapse
|
31
|
Ren S, Song L, Tian Y, Zhu L, Guo K, Zhang H, Wang Z. Emodin-Conjugated PEGylation of Fe 3O 4 Nanoparticles for FI/MRI Dual-Modal Imaging and Therapy in Pancreatic Cancer. Int J Nanomedicine 2021; 16:7463-7478. [PMID: 34785894 PMCID: PMC8579871 DOI: 10.2147/ijn.s335588] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/08/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) remains a difficult tumor to diagnose and treat. It is often diagnosed as advanced by reason of the anatomical structure of the deep retroperitoneal layer of the pancreas, lack of typical symptoms and effective screening methods to detect this malignancy, resulting in a low survival rate. Emodin (EMO) is an economical natural product with effective treatment and few side effects of cancer treatment. Magnetic nanoparticles (MNPs) can achieve multiplexed imaging and targeted therapy by loading a wide range of functional materials such as fluorescent dyes and therapeutic agents. PURPOSE The purpose of this study was to design and evaluate a multifunctional theranostic nanoplatform for PC diagnosis and treatment. METHODS In this study, we successfully developed EMO-loaded, Cy7-functionalized, PEG-coated Fe3O4 (Fe3O4-PEG-Cy7-EMO). Characteristics including morphology, hydrodynamic size, zeta potentials, stability, and magnetic properties of Fe3O4-PEG-Cy7-EMO were evaluated. Fluorescence imaging (FI)/magnetic resonance imaging (MRI) and therapeutic treatment were examined in vitro and in vivo. RESULTS Fe3O4-PEG-Cy7-EMO nanoparticles had a core size of 9.9 ± 1.2 nm, which showed long-time stability and FI/MRI properties. Bio-transmission electron microscopy (bio-TEM) results showed that Fe3O4-PEG-Cy7-EMO nanoparticles were endocytosed into BxPC-3 cells, while few were observed in hTERT-HPNE cells. Prussian blue staining also confirmed that BxPC-3 cells have a stronger phagocytic ability as compared to hTERT-HPNE cells. Additionally, Fe3O4-PEG-Cy7-EMO had a stronger inhibition effect on BxPC-3 cells than Fe3O4-PEG and EMO. The hemolysis experiment proved that Fe3O4-PEG-Cy7-EMO can be used in vivo experiments. In vivo analysis demonstrated that Fe3O4-PEG-Cy7-EMO enabled FI/MRI dual-modal imaging and targeted therapy in pancreatic tumor xenografted mice. CONCLUSION Fe3O4-PEG-Cy7-EMO may serve as a potential theranostic nanoplatform for PC.
Collapse
Affiliation(s)
- Shuai Ren
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
- Correspondence: Shuai Ren; Zhongqiu Wang Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, Jiangsu Province, 210029, People’s Republic of China Email ;
| | - Lina Song
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
| | - Ying Tian
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
| | - Li Zhu
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
| | - Kai Guo
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
| | - Huifeng Zhang
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
| | - Zhongqiu Wang
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, People’s Republic of China
| |
Collapse
|
32
|
Perros F, Ghigna MR, Loisel F, Chemla D, Decante B, de Montpreville V, Montani D, Humbert M, Fadel E, Mercier O, Boulate D. Description, Staging and Quantification of Pulmonary Artery Angiophagy in a Large Animal Model of Chronic Thromboembolic Pulmonary Hypertension. Biomedicines 2020; 8:biomedicines8110493. [PMID: 33187154 PMCID: PMC7696066 DOI: 10.3390/biomedicines8110493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 11/30/2022] Open
Abstract
Angiophagy has been described as a non-fibrinolytic mechanism of pulmonary artery (PA) patency restoration after distal (<50 µm in diameter) pulmonary embolism in mice. We hypothesized that angiophagy could achieve muscularized PA patency restoration after pulmonary embolism in piglets and humans. Angiophagy was defined by pathological assessment as the moving of an embolic specimen from the lumen to the interstitium according to three stages in a pig model of chronic thromboembolic pulmonary hypertension (CTEPH) 6 to 10 weeks after embolization with enbucrilate: the embolic specimen is (I) covered by endothelial cells, (II) covered by endothelial cells and smooth muscle cells, and (III) located in the adventitia. In animals, we observed the three stages of the pulmonary angiophagy of enbucrilate emboli in <300 µm PA. Stages II and III were observed in 300 to 1000 μm PA, and only Stage I was observed in larger-diameter PA (>1000 μm). In lung samples from patients with histories of pulmonary embolisms, we observed PA angiophagy stigma for embolic specimens derived from blood clots and from bone marrow emboli. This study provides an original pathological description and staging of PA angiophagy in a large animal model of CTEPH and in humans after pulmonary embolism.
Collapse
Affiliation(s)
- Frédéric Perros
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Maria-Rosa Ghigna
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Research and Innovation Unit, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; (F.L.); (B.D.)
- Department of Pathology, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France;
| | - Fanny Loisel
- Research and Innovation Unit, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; (F.L.); (B.D.)
| | - Denis Chemla
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- Department of Physiology, Hôpital Bicêtre, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Benoit Decante
- Research and Innovation Unit, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; (F.L.); (B.D.)
| | | | - David Montani
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Elie Fadel
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Research and Innovation Unit, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; (F.L.); (B.D.)
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
| | - Olaf Mercier
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Research and Innovation Unit, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; (F.L.); (B.D.)
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
| | - David Boulate
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (F.P.); (M.-R.G.); (D.C.); (D.M.); (M.H.); (E.F.); (O.M.)
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Research and Innovation Unit, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; (F.L.); (B.D.)
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
- Correspondence: ; Tel.: +33-140-948-725; Fax: +33-140-948-718
| |
Collapse
|
33
|
Sun X, Ni N, Ma Y, Wang Y, Leong DT. Retooling Cancer Nanotherapeutics' Entry into Tumors to Alleviate Tumoral Hypoxia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003000. [PMID: 32803846 DOI: 10.1002/smll.202003000] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/20/2020] [Indexed: 06/11/2023]
Abstract
Anti-hypoxia cancer nanomedicine (AHCN) holds exciting potential in improving oxygen-dependent therapeutic efficiencies of malignant tumors. However, most studies regarding AHCN focus on optimizing structure and function of nanomaterials with presupposed successful entry into tumor cells. From such a traditional perspective, the main barrier that AHCN needs to overcome is mainly the tumor cell membrane. However, such an oversimplified perspective would neglect that real tumors have many biological, physiological, physical, and chemical defenses preventing the current state-of-the-art AHCNs from even reaching the targeted tumor cells. Fortunately, in recent years, some studies are beginning to intentionally focus on overcoming physiological barriers to alleviate hypoxia. In this Review, the limitations behind the traditional AHCN delivery mindset are addressed and the key barriers that need to be surmounted before delivery to cancer cells and some good ways to improve cell membrane attachment, internalization, and intracellular retention are summarized. It is aimed to contribute to Review literature on this emerging topic through refreshing perspectives based on this work and what is also learnt from others. This Review would therefore assist AHCNs researchers to have a quick overview of the essential information and glean thought-provoking ideas to advance this sub-field in cancer nanomedicine.
Collapse
Affiliation(s)
- Xiao Sun
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yanling Ma
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yan Wang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
34
|
Chen Q, He Y, Wang Y, Li C, Zhang Y, Guo Q, Zhang Y, Chu Y, Liu P, Chen H, Zhou Z, Zhou W, Zhao Z, Li X, Sun T, Jiang C. Penetrable Nanoplatform for "Cold" Tumor Immune Microenvironment Reeducation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000411. [PMID: 32995118 PMCID: PMC7503208 DOI: 10.1002/advs.202000411] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/05/2020] [Indexed: 05/08/2023]
Abstract
Lack of tumor-infiltration lymphocytes (TILs) and resistances by overexpressed immunosuppressive cells (principally, myeloid-derived suppressor cells (MDSCs)) in tumor milieu are two major challenges hindering the effectiveness of immunotherapy for "immune-cold" tumors. In addition, the natural physical barrier existing in solid cancer also limits deeper delivery of drugs. Here, a tumor-targeting and light-responsive-penetrable nanoplatform (Apt/PDGs^s@pMOF) is developed to elicit intratumoral infiltration of cytotoxic T cells (CTLs) and reeducate immunosuppressive microenvironment simultaneously. In particular, porphyrinic metal-organic framework (pMOF)-based photodynamic therapy (PDT) induces tumor immunogenic cell death (ICD) to promote CTLs intratumoral infiltration and hot "immune-cold" tumor. Upon being triggered by PDT, the nearly 10 nm adsorbed drug-loaded dendrimer de-shields from the nanoplatform and spreads into the deeper tumor, eliminating MDSCs and reversing immunosuppression, eventually reinforcing immune response. Meanwhile, the designed nanoplatform also has a systemic MDSC inhibition effect and moderate improvement of overall antitumor immune responses, resulting in effective suppression of distal tumors within less significant immune-related adverse effects (irAEs) induced.
Collapse
Affiliation(s)
- Qinjun Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Yongqing He
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Yu Wang
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Chao Li
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Yujie Zhang
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Qin Guo
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Yiwen Zhang
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Yongchao Chu
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Peixin Liu
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Hongyi Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Zheng Zhou
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Wenxi Zhou
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Zhenhao Zhao
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Xiaomin Li
- Department of Chemistry and Laboratory of Advanced MaterialsFudan UniversityShanghai200433P. R. China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education)State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyResearch Center on Aging and MedicineFudan UniversityShanghai201203P. R. China
| |
Collapse
|
35
|
Emerging Trends in Nanotheranostics. Nanobiomedicine (Rij) 2020. [DOI: 10.1007/978-981-32-9898-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
36
|
Li S, Li H, Xu X, Saw PE, Zhang L. Nanocarrier-mediated antioxidant delivery for liver diseases. Theranostics 2020; 10:1262-1280. [PMID: 31938064 PMCID: PMC6956819 DOI: 10.7150/thno.38834] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Liver is the principal detoxifying organ and metabolizes various compounds that produce free radicals (FR) constantly. To maintain the oxidative/antioxidative balance in the liver, antioxidants would scavenge FR by preventing tissue damage through FR formation, scavenging, or by enhancing their decomposition. The disruption of this balance therefore leads to oxidative stress and in turn leads to the onset of various diseases. Supplying the liver with exogeneous antioxidants is an effective way to recreate the oxidative/antioxidative balance in the liver homeostasis. Nevertheless, due to the short half-life and instability of antioxidants in circulation, the methodology for delivering antioxidants to the liver needs to be improved. Nanocarrier mediated delivery of antioxidants proved to be an ingenious way to safely and efficiently deliver a high payload of antioxidants into the liver for circumventing liver diseases. The objective of this review is to provide an overview of the role of reactive oxygen species (oxidant) and ROS scavengers (antioxidant) in liver diseases. Subsequently, current nanocarrier mediated antioxidant delivery methods for liver diseases are discussed.
Collapse
Affiliation(s)
- Senlin Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Huiru Li
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Lei Zhang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| |
Collapse
|
37
|
Abstract
Nanomedicines have historically struggled to find clinical relevance and to achieve translation successfully. In this Perspective, we discuss possible reasons for this difficulty and highlight several key features of nanomedicines that are often overlooked by biomedical engineers. We present the notion of clinical multifunctionality as distinct from multifunctionality as it is traditionally described at the nanoscale and emphasize its importance through examples of nanomedicines that have demonstrated emergent clinical multifunctionality once translated. We also describe a phenomenon in which clinical multifunctionality results in diagonal translation after a nanomedicine is adopted by clinicians to serve as a solution for a clinical problem that it was not designed to solve. Biomedical engineers can take advantage of these phenomena to assist in achieving clinical translation of new nanomedicines.
Collapse
Affiliation(s)
- Keegan Guidolin
- Department of Surgery , University of Toronto , Toronto M5G 1L7 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5G 1L7 , Canada
- Princess Margaret Cancer Centre , Toronto M5G 1L7 , Canada
| | - Gang Zheng
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5G 1L7 , Canada
- Department of Medical Biophysics , University of Toronto , Toronto M5G 1L7 , Canada
- Princess Margaret Cancer Centre , Toronto M5G 1L7 , Canada
| |
Collapse
|
38
|
Neumann PR, Crossley DL, Turner M, Ingleson M, Green M, Rao J, Dailey LA. In Vivo Optical Performance of a New Class of Near-Infrared-Emitting Conjugated Polymers: Borylated PF8-BT. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46525-46535. [PMID: 31746180 DOI: 10.1021/acsami.9b17022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Borylated poly(fluorene-benzothiadiazoles) (PF8-BT) are π-conjugated polymers (CPs) with deep-red/near-infrared (NIR) absorption and emission profiles suitable for in vivo optical imaging. A fully borylated PF8-BT derivative (P4) was encapsulated in pegylated poly(lactic-co-glycolic acid) (PEG-PLGA) nanoparticles and compared with a reference NIR-emitting CP (PCPDTBT) or indocyanine green (ICG). All formulations satisfied quality requirements for parenterally administered diagnostics. P4 nanoparticles had higher quantum yield (2.3%) than PCPCDTBT (0.01%) or ICG nanoparticles (1.1%). The signal/background ratios (SBRs) of CP systems P4 and PCPDTBT in a phantom mouse (λem = 820 nm) increased linearly with fluorophore mass (12.5-100 μg/mL), while the SBRs of ICG decreased above 25 μg/mL. P4 nanoparticles experienced <10% photobleaching over 10 irradiations (PCPDTBT: ∼25% and ICG: >44%). In a mouse tumor xenograft model, P4 nanoparticles showed a 5-fold higher SBR than PCPDTBT particles with fluorophore accumulation in the liver > spleen > tumor. Blood chemistry and tissue histology showed no abnormalities compared to untreated animals after a single administration.
Collapse
Affiliation(s)
- Paul Robert Neumann
- Department of Pharmaceutical Technology and Biopharmaceutics , Martin-Luther-University Halle-Wittenberg , Halle/Saale 06120 , Germany
| | - Daniel L Crossley
- Department of Chemical Sciences , University of Huddersfield , Huddersfield HD1 3DH , U.K
| | - Michael Turner
- School of Chemistry , University of Manchester , Manchester M13 9PL , U.K
| | - Michael Ingleson
- School of Chemistry , University of Edinburgh , Edinburgh EH9 3FJ , U.K
| | - Mark Green
- Department of Physics , King's College London , London WC2R 2LS , U.K
| | - Jianghong Rao
- Department of Radiology and Chemistry , Stanford University , Stanford , California 94305 , United States
| | - Lea Ann Dailey
- Department of Pharmaceutical Technology and Biopharmaceutics , Martin-Luther-University Halle-Wittenberg , Halle/Saale 06120 , Germany
| |
Collapse
|
39
|
Abstract
The poor pharmacokinetic parameters and low solubility of many anticancer therapeutics have warranted the use of drug-delivery systems such as liposomes. Overcoming some drawbacks of the conventional liposomes, targeted liposomal delivery by longer circulation time by addition of poly(ethylene glycol) to the liposomal surface and further adding specific ligands to achieve ligand selective retention and uptake has been introduced. PEGylated liposomes are the only second-generation liposomal formulations in clinical use and are now being challenged with the allergenic response they pose even in the treatment of naive patients. This article will review the challenges and hindrances in the use of long circulating liposomes and explore the opportunities to overcome this issue.
Collapse
|
40
|
Mao B, Li L, Yan M, Wong CKC, Silvestrini B, Li C, Ge R, Lian Q, Cheng CY. F5-Peptide and mTORC1/rpS6 Effectively Enhance BTB Transport Function in the Testis-Lesson From the Adjudin Model. Endocrinology 2019; 160:1832-1853. [PMID: 31157869 PMCID: PMC6637795 DOI: 10.1210/en.2019-00308] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/26/2019] [Indexed: 01/04/2023]
Abstract
During spermatogenesis, the blood-testis barrier (BTB) undergoes cyclic remodeling that is crucial to support the transport of preleptotene spermatocytes across the immunological barrier at stage VIII to IX of the epithelial cycle. Studies have shown that this timely remodeling of the BTB is supported by several endogenously produced barrier modifiers across the seminiferous epithelium, which include the F5-peptide and the ribosomal protein S6 [rpS6; a downstream signaling molecule of the mammalian target of rapamycin complex 1 (mTORC1)] signaling protein. Herein, F5-peptide and a quadruple phosphomimetic (and constitutively active) mutant of rpS6 [i.e., phosphorylated (p-)rpS6-MT] that are capable of inducing reversible immunological barrier remodeling, by making the barrier "leaky" transiently, were used for their overexpression in the testis to induce BTB opening. We sought to examine whether this facilitated the crossing of the nonhormonal male contraceptive adjudin at the BTB when administered by oral gavage, thereby effectively improving its BTB transport to induce germ cell adhesion and aspermatogenesis. Indeed, it was shown that combined overexpression of F5-peptide and p-rpS6-MT and a low dose of adjudin, which by itself had no noticeable effects on spermatogenesis, was capable of perturbing the organization of actin- and microtubule (MT)-based cytoskeletons through changes in the spatial expression of actin- and MT-binding/regulatory proteins to the corresponding cytoskeleton. These findings thus illustrate the possibility of delivering drugs to any target organ behind a blood-tissue barrier by modifying the tight junction permeability barrier using endogenously produced barrier modifiers based on findings from this adjudin animal model.
Collapse
Affiliation(s)
- Baiping Mao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linxi Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Yan
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York
- Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Chris K C Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | | | - Chao Li
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingquan Lian
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Correspondence: C. Yan Cheng, PhD, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, New York 10065. E-mail:
| |
Collapse
|
41
|
Bi H, Xue J, Jiang H, Gao S, Yang D, Fang Y, Shi K. Current developments in drug delivery with thermosensitive liposomes. Asian J Pharm Sci 2019; 14:365-379. [PMID: 32104466 PMCID: PMC7032122 DOI: 10.1016/j.ajps.2018.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/26/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022] Open
Abstract
Thermosensitive liposomes (TSLs) have been an important research area in the field of tumor targeted chemotherapy. Since the first TSLs appeared that using 1,2-dipalmitoyl-sn-glyce-ro-3-phosphocholine (DPPC) as the primary liposomal lipid, many studies have been done using this type of liposome from basic and practical aspects. While TSLs composed of DPPC enhance the cargo release near the phase transition temperature, it has been shown that many factors affect their temperature sensitivity. Thus numerous attempts have been undertaken to develop new TSLs for improving their thermal response performance. The main objective of this review is to introduce the development and recent update of innovative TSLs formulations, including combination of radiofrequency ablation (RFA), high-intensity focused ultrasound (HIFU), magnetic resonance imaging (MRI) and alternating magnetic field (AMF). In addition, various factors affecting the design of TSLs, such as lipid composition, surfactant, size and serum components are also discussed.
Collapse
Key Words
- (DPPC), 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine
- (DPPGOG), 1,2-dipalmitoyl-sn-glycero-3-phosphoglyceroglycerol
- (DSPC), 1,2-distearoyl-sn-glycero-3-phosphocholine
- (DSPE-mPEG2000), 1,2-distearoyl-sn-glycero-3-phosphatiylethanol-amine-N-[methoxy(polyethyleneglycol)-2000]
- (LTSLs), lyso-lipid temperature sensitive liposomes
- (MPPC), 1-myristoyl-2-palmitoyl-sn-glycero-3-phosphatidylcholine
- (MSPC), 1-stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine
- (P-lyso-PC), lysophosphatidylcholine
- (P188), 1-palmitoyl-2-stearoyl-sn-glycero-3-phosphatidylcholinex
- (P188), HO-(C2H4O)a-(C3H6O)b-(C2H4O)c-H, a=80, b=27, c=80
- Content release rate
- Drug delivery
- Hyperthermia
- Smart liposomes
- Thermosensitive liposomes
- Tumor chemotherapy
- fTSLs, fast release TSLs
- sTSLs, slow release TSLs
Collapse
Affiliation(s)
- Hongshu Bi
- Institute of New Drug Development, Liaoning Yaolian Pharmaceutical Co., Ltd., Benxi, Liaoning 117004, China
| | - Jianxiu Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Hong Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Shan Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Dongjuan Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Yan Fang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Kai Shi
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| |
Collapse
|
42
|
Jauffred L, Samadi A, Klingberg H, Bendix PM, Oddershede LB. Plasmonic Heating of Nanostructures. Chem Rev 2019; 119:8087-8130. [PMID: 31125213 DOI: 10.1021/acs.chemrev.8b00738] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The absorption of light by plasmonic nanostructures and their associated temperature increase are exquisitely sensitive to the shape and composition of the structure and to the wavelength of light. Therefore, much effort is put into synthesizing novel nanostructures for optimized interaction with the incident light. The successful synthesis and characterization of high quality and biocompatible plasmonic colloidal nanoparticles has fostered numerous and expanding applications, especially in biomedical contexts, where such particles are highly promising for general drug delivery and for tomorrow's cancer treatment. We review the thermoplasmonic properties of the most commonly used plasmonic nanoparticles, including solid or composite metallic nanoparticles of various dimensions and geometries. Common methods for synthesizing plasmonic particles are presented with the overall goal of providing the reader with a guide for designing or choosing nanostructures with optimal thermoplasmonic properties for a given application. Finally, the biocompatibility and biological tolerance of structures are critically discussed along with novel applications of plasmonic nanoparticles in the life sciences.
Collapse
Affiliation(s)
| | - Akbar Samadi
- Niels Bohr Institute , University of Copenhagen , Copenhagen , Denmark
| | - Henrik Klingberg
- Niels Bohr Institute , University of Copenhagen , Copenhagen , Denmark
| | | | - Lene B Oddershede
- Niels Bohr Institute , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
43
|
Manthe RL, Rappaport JA, Long Y, Solomon M, Veluvolu V, Hildreth M, Gugutkov D, Marugan J, Zheng W, Muro S. δ-Tocopherol Effect on Endocytosis and Its Combination with Enzyme Replacement Therapy for Lysosomal Disorders: A New Type of Drug Interaction? J Pharmacol Exp Ther 2019; 370:823-833. [PMID: 31101681 DOI: 10.1124/jpet.119.257345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022] Open
Abstract
Induction of lysosomal exocytosis alleviates lysosomal storage of undigested metabolites in cell models of lysosomal disorders (LDs). However, whether this strategy affects other vesicular compartments, e.g., those involved in endocytosis, is unknown. This is important both to predict side effects and to use this strategy in combination with therapies that require endocytosis for intracellular delivery, such as lysosomal enzyme replacement therapy (ERT). We investigated this using δ-tocopherol as a model previously shown to induce lysosomal exocytosis and cell models of type A Niemann-Pick disease, a LD characterized by acid sphingomyelinase (ASM) deficiency and sphingomyelin storage. δ-Tocopherol and derivative CF3-T reduced net accumulation of fluid phase, ligands, and polymer particles via phagocytic, caveolae-, clathrin-, and cell adhesion molecule (CAM)-mediated pathways, yet the latter route was less affected due to receptor overexpression. In agreement, δ-tocopherol lowered uptake of recombinant ASM by deficient cells (known to occur via the clathrin pathway) and via targeting intercellular adhesion molecule-1 (associated to the CAM pathway). However, the net enzyme activity delivered and lysosomal storage attenuation were greater via the latter route. Data suggest stimulation of exocytosis by tocopherols is not specific of lysosomes and affects endocytic cargo. However, this effect was transient and became unnoticeable several hours after tocopherol removal. Therefore, induction of exocytosis in combination with therapies requiring endocytic uptake, such as ERT, may represent a new type of drug interaction, yet this strategy could be valuable if properly timed for minimal interference.
Collapse
Affiliation(s)
- Rachel L Manthe
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Jeffrey A Rappaport
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Yan Long
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Melani Solomon
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Vinay Veluvolu
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Michael Hildreth
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Dencho Gugutkov
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Juan Marugan
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Wei Zheng
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Silvia Muro
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| |
Collapse
|
44
|
Ashour DS. Ivermectin: From theory to clinical application. Int J Antimicrob Agents 2019; 54:134-142. [PMID: 31071469 DOI: 10.1016/j.ijantimicag.2019.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
Abstract
Approximately 250 million people have been using ivermectin (IVM) annually to combat many parasitic diseases including filariasis, onchocerciasis, strongyloidiasis, scabies and pediculosis. Many clinical studies have proven its efficacy against these diseases and have reported the optimum dose and duration of treatment. Moreover, its antiparasitic range has increased to cover more parasitic infections, but it still requires further exploration, e.g. for trichinosis and myiasis. Furthermore, IVM showed high efficacy in killing vectors of disease-causing parasites such as mosquitoes, sandflies and tsetse flies. The World Health Organization (WHO) has managed many control programmes involving the use of IVM to achieve elimination of onchocerciasis and lymphatic filariasis and to reduce malaria transmission. However, IVM is not exempt from the possibility of resistance and, certainly, its intensive use has led to the emergence of resistance in some parasites. Recent research is investigating the possibility of novel drug delivery systems for IVM that increase its potential to treat a new range of diseases and to overcome the possibility of drug resistance. This review highlights the most common human uses of IVM, with special reference to the new and promising properties of IVM.
Collapse
Affiliation(s)
- Dalia S Ashour
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt.
| |
Collapse
|
45
|
Al-Qubaisi MS, Rasedee A, Flaifel MH, Eid EE, Hussein-Al-Ali S, Alhassan FH, Salih AM, Hussein MZ, Zainal Z, Sani D, Aljumaily AH, Saeed MI. Characterization of thymoquinone/hydroxypropyl-β-cyclodextrin inclusion complex: Application to anti-allergy properties. Eur J Pharm Sci 2019; 133:167-182. [DOI: 10.1016/j.ejps.2019.03.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
|
46
|
Ma L, Le P, Kohli M, Smith AM. Nanomedicine in Cancer. Bioanalysis 2019. [DOI: 10.1007/978-3-030-01775-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
47
|
Lin G, Li L, Panwar N, Wang J, Tjin SC, Wang X, Yong KT. Non-viral gene therapy using multifunctional nanoparticles: Status, challenges, and opportunities. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
48
|
Akhtar MJ, Ahamed M, Alhadlaq HA. Challenges facing nanotoxicology and nanomedicine due to cellular diversity. Clin Chim Acta 2018; 487:186-196. [PMID: 30291894 DOI: 10.1016/j.cca.2018.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/26/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022]
Abstract
This review examines the interaction of nanomaterials (NMs) with cells from the perspective of major cellular differentiations. The structure and composition of cells reflect their role and function in a particular organ or environment. The normal differentiated-state and diseased cells may respond to NMs very differently. This review progresses with due care on nanotoxicology while emphasizing the potential of NMs in treating stress-associated disorders, including cancer and degeneration. The striking potential of NMs in inducing ROS, scavenging ROS, depleting cellular antioxidants, replenishing antioxidants, mimicking antioxidant enzyme activity, and modulating the immune system all show their considerable potential in treating cancer and other aging-associated disorders. It is now clear that NMs become more active and versatile when they come into contact with biological machinery, surprisingly in some cases, in a manner dependent on cell type. The mechanisms leading to the contrasting bioresponse of NMs ranging from toxicity to anticancer and from cell survival to carcinogenicity followed by their immuno-modulating potential show NMs to be a highly promising agent in biomedical therapy. This first-of-its-kind article seeks the challenges to be addressed that could provide a solid rationale in translating the promises of nanomedicine. A thorough understanding of normal and cancer biology could help to minimize the gap between basic and translational research in nanotechnology-based therapy.
Collapse
Affiliation(s)
- Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia..
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Hisham A Alhadlaq
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia.; Department of Physics and Astronomy, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
49
|
Bioinspired and biomimetic systems for advanced drug and gene delivery. J Control Release 2018; 287:142-155. [DOI: 10.1016/j.jconrel.2018.08.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 12/15/2022]
|
50
|
Exploring the role of polymeric conjugates toward anti-cancer drug delivery: Current trends and future projections. Int J Pharm 2018; 548:500-514. [DOI: 10.1016/j.ijpharm.2018.06.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
|