51
|
Li S, Su J, Cai W, Liu JX. Nanomaterials Manipulate Macrophages for Rheumatoid Arthritis Treatment. Front Pharmacol 2021; 12:699245. [PMID: 34335264 PMCID: PMC8316763 DOI: 10.3389/fphar.2021.699245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/09/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, progressive, and systemic inflammatory autoimmune disease, characterized by synovial inflammation, synovial lining hyperplasia and inflammatory cell infiltration, autoantibody production, and cartilage/bone destruction. Macrophages are crucial effector cells in the pathological process of RA, which can interact with T, B, and fibroblast-like synovial cells to produce large amounts of cytokines, chemokines, digestive enzymes, prostaglandins, and reactive oxygen species to accelerate bone destruction. Therefore, the use of nanomaterials to target macrophages has far-reaching therapeutic implications for RA. A number of limitations exist in the current clinical therapy for patients with RA, including severe side effects and poor selectivity, as well as the need for frequent administration of therapeutic agents and high doses of medication. These challenges have encouraged the development of targeting drug delivery systems and their application in the treatment of RA. Recently, obvious therapeutic effects on RA were observed following the use of various types of nanomaterials to manipulate macrophages through intravenous injection (active or passive targeting), oral administration, percutaneous absorption, intraperitoneal injection, and intra-articular injection, which offers several advantages, such as high-precision targeting of the macrophages and synovial tissue of the joint. In this review, the mechanisms involved in the manipulation of macrophages by nanomaterials are analyzed, and the prospect of clinical application is also discussed. The objective of this article was to provide a reference for the ongoing research concerning the treatment of RA based on the targeting of macrophages.
Collapse
Affiliation(s)
- Shuang Li
- Hunan Province Key Laboratory of Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China.,College Pharmacy, Jiamusi University, Jiamusi, China
| | - Jin Su
- College Pharmacy, Jiamusi University, Jiamusi, China
| | - Wei Cai
- Hunan Province Key Laboratory of Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Jian-Xin Liu
- Hunan Province Key Laboratory of Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
52
|
Martín-Sabroso C, Fraguas-Sánchez AI, Raposo-González R, Torres-Suárez AI. Perspectives in Breast and Ovarian Cancer Chemotherapy by Nanomedicine Approach: Nanoformulations in Clinical Research. Curr Med Chem 2021; 28:3271-3286. [PMID: 32814522 DOI: 10.2174/0929867327666200819115403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Breast and ovarian carcinomas represent major health problems in women worldwide. Chemotherapy constitutes the main treatment strategy, and the use of nanocarriers, a good tool to improve it. Several nanoformulations have already been approved, and others are under clinical trials for the treatment of both types of cancers. OBJECTIVE This review focuses on the analysis of the nanoformulations that are under clinical research in the treatment of these neoplasms. RESULTS Currently, there are 6 nanoformulations in clinical trials for breast and ovarian carcinomas, most of them in phase II and phase III. In the case of breast cancer treatment, these nanomedicines contain paclitaxel; and, for ovarian cancer, nanoformulations containing paclitaxel or camptothecin analogs are being evaluated. The nanoencapsulation of these antineoplastics facilitates their administration and reduces their systemic toxicity. Nevertheless, the final approval and commercialization of nanoformulations may be limited by other aspects like lack of correlation between the efficacy results evaluated at in vitro and in vivo levels, difficulty in producing large batches of nanoformulations in a reproducible manner and high production costs compared to conventional formulations of antineoplastics. However, these challenges are not insurmountable and the number of approved nanoformulations for cancer therapy is growing. CONCLUSION Reviewed nanoformulations have shown, in general, excellent results, demonstrating a good safety profile, a higher maximum tolerated dose and a similar or even slightly better antitumor efficacy compared to the administration of free drugs, reinforcing the use of nano-chemotherapy in both breast and ovarian tumors.
Collapse
Affiliation(s)
- Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramon y Cajal s/n., 28040 Madrid, Spain
| | - Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramon y Cajal s/n., 28040 Madrid, Spain
| | - Rafaela Raposo-González
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramon y Cajal s/n., 28040 Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Pl Ramon y Cajal s/n., 28040 Madrid, Spain
| |
Collapse
|
53
|
AlSawaftah N, Pitt WG, Husseini GA. Dual-Targeting and Stimuli-Triggered Liposomal Drug Delivery in Cancer Treatment. ACS Pharmacol Transl Sci 2021; 4:1028-1049. [PMID: 34151199 PMCID: PMC8205246 DOI: 10.1021/acsptsci.1c00066] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 12/31/2022]
Abstract
The delivery of chemotherapeutics to solid tumors using smart drug delivery systems (SDDSs) takes advantage of the unique physiology of tumors (i.e., disordered structure, leaky vasculature, abnormal extracellular matrix (ECM), and limited lymphatic drainage) to deliver anticancer drugs with reduced systemic side effects. Liposomes are the most promising of such SDDSs and have been well investigated for cancer therapy. To improve the specificity, bioavailability, and anticancer efficacy of liposomes at the diseased sites, other strategies such as targeting ligands and stimulus-sensitive liposomes have been developed. This review highlights relevant surface functionalization techniques and stimuli-mediated drug release for enhanced delivery of anticancer agents at tumor sites, with a special focus on dual functionalization and design of multistimuli responsive liposomes.
Collapse
Affiliation(s)
- Nour AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| | - William G. Pitt
- Chemical
Engineering Department, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| |
Collapse
|
54
|
Alves RC, Schulte ZM, Luiz MT, Bento da Silva P, Frem RCG, Rosi NL, Chorilli M. Breast Cancer Targeting of a Drug Delivery System through Postsynthetic Modification of Curcumin@N 3-bio-MOF-100 via Click Chemistry. Inorg Chem 2021; 60:11739-11744. [PMID: 34101467 DOI: 10.1021/acs.inorgchem.1c00538] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Metal-organic frameworks (MOFs) offer many opportunities for applications across biology and medicine. Their wide range of chemical composition makes toxicologically acceptable formulation possible, and their high level of functionality enables possible applications as delivery systems for therapeutics agents. Surface modifications have been used in drug delivery systems to minimize their interaction with the bulk, improving their specificity as targeted carriers. Herein, we discuss a strategy to achieve a tumor-targeting drug-loaded MOF using "click" chemistry to anchor functional folic acid (FA) molecules on the surface of N3-bio-MOF-100. Using curcumin (CCM) as an anticancer drug, we observed drug loading encapsulation efficiencies (DLEs) of 24.02 and 25.64% after soaking N3-bio-MOF-100 in CCM solutions for 1 day and 3 days, respectively. The success of postsynthetic modification of FA was confirmed by 1H NMR spectroscopy, Fourier transform infrared spectroscopy (FTIR), and liquid chromatography-mass spectrometry (LC-MS). The stimuli-responsive drug release studies demonstrated an increase of CCM released under acidic microenvironments. Moreover, the cell viability assay was performed on the 4T1 (breast cancer) cell line in the presence of CCM@N3-bio-MOF-100 and CCM@N3-bio-MOF-100/FA carriers to confirm its biological compatibility. In addition, a cellular uptake study was conducted to evaluate the targeting of tumor cells.
Collapse
Affiliation(s)
- Renata C Alves
- Department of Drugs and Medicines, School of Pharmaceutical Sciences of São Paulo State University (UNESP), Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville, 14800-903 Araraquara, São Paulo, Brazil
| | - Zachary M Schulte
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 1560, United States
| | - Marcela T Luiz
- Department of Pharmaceutical Sciences, School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo (USP), Avenida do Café, s/n - Campus da USP, 14040-903 Ribeirão Preto, Sao Paulo, Brazil
| | - Patrícia Bento da Silva
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia (UnB), Campus Universitario Darcy Ribeiro - Asa Norte, 70910-900 Brasilia, Federal District, Brazil
| | - Regina C G Frem
- Institute of Chemistry, São Paulo State University (UNESP), Prof. Francisco Degni 55, PO Box 355, 14800-970 Araraquara, São Paulo, Brazil
| | - Nathaniel L Rosi
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 1560, United States
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences of São Paulo State University (UNESP), Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville, 14800-903 Araraquara, São Paulo, Brazil
| |
Collapse
|
55
|
Ai X, Wang S, Duan Y, Zhang Q, Chen M, Gao W, Zhang L. Emerging Approaches to Functionalizing Cell Membrane-Coated Nanoparticles. Biochemistry 2021; 60:941-955. [PMID: 32452667 PMCID: PMC8507422 DOI: 10.1021/acs.biochem.0c00343] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
There has been significant interest in developing cell membrane-coated nanoparticles due to their unique abilities of biomimicry and biointerfacing. As the technology progresses, it becomes clear that the application of these nanoparticles can be drastically broadened if additional functions beyond those derived from the natural cell membranes can be integrated. Herein, we summarize the most recent advances in the functionalization of cell membrane-coated nanoparticles. In particular, we focus on emerging methods, including (1) lipid insertion, (2) membrane hybridization, (3) metabolic engineering, and (4) genetic modification. These approaches contribute diverse functions in a nondisruptive fashion while preserving the natural function of the cell membranes. They also improve on the multifunctional and multitasking ability of cell membrane-coated nanoparticles, making them more adaptive to the complexity of biological systems. We hope that these approaches will serve as inspiration for more strategies and innovations to advance cell membrane coating technology.
Collapse
Affiliation(s)
- Xiangzhao Ai
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Shuyan Wang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Yaou Duan
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Qiangzhe Zhang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Maggie Chen
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Weiwei Gao
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Liangfang Zhang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
56
|
Choi JS, Park JW, Seu YB, Doh KO. Enhanced efficacy of folate-incorporated cholesteryl doxorubicin liposome in folate receptor abundant cancer cell. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
57
|
Targeted nanoformulation of C1 inhibits the growth of KB spheroids and cancer stem cell-enriched MCF-7 mammospheres. Colloids Surf B Biointerfaces 2021; 202:111702. [PMID: 33780906 DOI: 10.1016/j.colsurfb.2021.111702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
C1, a synthetic analog of curcumin, has been reported to show potent antiproliferative effects against a variety of cancer cells. Here, we report a strong anticancer activity of the folate receptor-targeted lipid nanoparticle formulation of C1 against cancer cells and cancer stem cells both in 2D culture and 3D spheroids. The size of the C1 encapsulated folic acid functionalized nanoliposomes (Lipos-C1) was determined to be 83 ± 17 nm. Lipos-C1 nanoparticles displayed sustained C1 release kinetics at both pH 7.4 and 5.5. The folate receptor (FR) targeted nanoliposomes were internalized into FR-positive KB cells via the folate receptor-mediated endocytosis process. Lipos-C1 killed KB cells much more efficiently than C1. Lipos-C1 depolymerized microtubules, generated ROS, caused DNA damage, and induced apoptosis in KB cells. Importantly, Lipos-C1 strongly inhibited the growth of the 3D KB spheroids than C1. Interestingly, Lipos-C1 also suppressed cancer stem cells (CSCs) enriched MCF-7 mammosphere growth by impeding breast cancer stem cells (BCSCs) enrichment, growth, and proliferation. The results suggested that Lipos-C1 could be a promising nanoformulation for cancer chemotherapy.
Collapse
|
58
|
Jurczyk M, Jelonek K, Musiał-Kulik M, Beberok A, Wrześniok D, Kasperczyk J. Single- versus Dual-Targeted Nanoparticles with Folic Acid and Biotin for Anticancer Drug Delivery. Pharmaceutics 2021; 13:326. [PMID: 33802531 PMCID: PMC8001342 DOI: 10.3390/pharmaceutics13030326] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer is one of the major causes of death worldwide and its treatment remains very challenging. The effectiveness of cancer therapy significantly depends upon tumour-specific delivery of the drug. Nanoparticle drug delivery systems have been developed to avoid the side effects of the conventional chemotherapy. However, according to the most recent recommendations, future nanomedicine should be focused mainly on active targeting of nanocarriers based on ligand-receptor recognition, which may show better efficacy than passive targeting in human cancer therapy. Nevertheless, the efficacy of single-ligand nanomedicines is still limited due to the complexity of the tumour microenvironment. Thus, the NPs are improved toward an additional functionality, e.g., pH-sensitivity (advanced single-targeted NPs). Moreover, dual-targeted nanoparticles which contain two different types of targeting agents on the same drug delivery system are developed. The advanced single-targeted NPs and dual-targeted nanocarriers present superior properties related to cell selectivity, cellular uptake and cytotoxicity toward cancer cells than conventional drug, non-targeted systems and single-targeted systems without additional functionality. Folic acid and biotin are used as targeting ligands for cancer chemotherapy, since they are available, inexpensive, nontoxic, nonimmunogenic and easy to modify. These ligands are used in both, single- and dual-targeted systems although the latter are still a novel approach. This review presents the recent achievements in the development of single- or dual-targeted nanoparticles for anticancer drug delivery.
Collapse
Affiliation(s)
- Magdalena Jurczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Katarzyna Jelonek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
| | - Monika Musiał-Kulik
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Janusz Kasperczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland
| |
Collapse
|
59
|
McCord E, Pawar S, Koneru T, Tatiparti K, Sau S, Iyer AK. Folate Receptors' Expression in Gliomas May Possess Potential Nanoparticle-Based Drug Delivery Opportunities. ACS OMEGA 2021; 6:4111-4118. [PMID: 33623837 PMCID: PMC7893640 DOI: 10.1021/acsomega.0c05500] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/22/2021] [Indexed: 05/05/2023]
Abstract
Brain cancer effected around estimated 23 890 adults and 3540 children under the age of 15 in 2020. The chemotherapeutic agents that are already approved by the FDA for brain cancer are proving to be not highly effective because of the interference from the tumor microenvironment as well as their own toxicities. Added to this is the impedance presented by the extremely restrictive permeability of the blood brain barrier (BBB). Targeted nanoparticulate drug delivery systems offer a good opportunity to traverse the BBB and selectively target the tumor cells. Folate receptors are found to be one of the most useful targets for drug delivery to the brain. Hence, this Mini-Review discusses the folate receptors and their application in the treatment of brain cancers using targeted nanoparticles.
Collapse
Affiliation(s)
- Eva McCord
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Grosse
Pointe South High School, Grosse
Pointe Farms, Michigan 48236, United States
| | - Shreya Pawar
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Troy
High School, Troy, Michigan 48098, United States
| | - Tejaswi Koneru
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Walled
Lake Central High School, Commerce Charter Township, Walled Lake, Michigan 48390, United States
| | - Katyayani Tatiparti
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
| | - Samaresh Sau
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
| | - Arun K. Iyer
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Molecular
Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| |
Collapse
|
60
|
Gangopadhyay S, Nikam RR, Gore KR. Folate Receptor-Mediated siRNA Delivery: Recent Developments and Future Directions for RNAi Therapeutics. Nucleic Acid Ther 2021; 31:245-270. [PMID: 33595381 DOI: 10.1089/nat.2020.0882] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RNA interference (RNAi), a gene regulatory process mediated by small interfering RNAs (siRNAs), has made remarkable progress as a potential therapeutic agent against various diseases. However, RNAi is associated with fundamental challenges such as poor systemic delivery and susceptibility to the nucleases. Targeting ligand-bound delivery vehicles has improved the accumulation of drug at the target site, which has resulted in high transfection efficiency and enhanced gene silencing. Recently, folate receptor (FR)-mediated targeted delivery of siRNAs has garnered attention due to their enhanced cellular uptake and high transfection efficiency toward tumor cells. Folic acid (FA), due to its small size, low immunogenicity, high in vivo stability, and high binding affinity toward FRs, has attracted much attention for targeted siRNA delivery. FRs are overexpressed in a large number of tumors, including ovarian, breast, kidney, and lung cancer cells. In this review, we discuss recent advances in FA-mediated siRNA delivery to treat cancers and inflammatory diseases. This review summarizes various FA-conjugated nanoparticle systems reported so far in the literature, including liposome, silica, metal, graphene, dendrimers, chitosan, organic copolymers, and RNA nanoparticles. This review will help in the design and development of potential delivery vehicles for siRNA drug targeting to tumor cells using an FR-mediated approach.
Collapse
Affiliation(s)
- Sumit Gangopadhyay
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rahul R Nikam
- Department of Chemistry, University of Mumbai, Mumbai, India
| | - Kiran R Gore
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
61
|
Quantum dots as targeted doxorubicin drug delivery nanosystems in human lung cancer cells. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00077-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract
Background
Lung cancer is one of the most frequently diagnosed cancers all over the world and is also one of the leading causes of cancer-related mortality. The main treatment option for small cell lung cancer, conventional chemotherapy, is characterized by a lack of specificity, resulting in severe adverse effects. Therefore, this study aimed at developing a new targeted drug delivery (TDD) system based on Ag–In–Zn–S quantum dots (QDs). For this purpose, the QD nanocrystals were modified with 11-mercaptoundecanoic acid (MUA), L-cysteine, and lipoic acid decorated with folic acid (FA) and used as a novel TDD system for targeting doxorubicin (DOX) to folate receptors (FARs) on adenocarcinomic human alveolar basal epithelial cells (A549). NIH/3T3 cells were used as FAR-negative controls. Comprehensive physicochemical, cytotoxicity, and genotoxicity studies were performed to characterize the developed novel TDDs.
Results
Fourier transformation infrared spectroscopy, dynamic light scattering, and fluorescence quenching confirmed the successful attachment of FA to the QD nanocrystals and of DOX to the QD–FA nanocarriers. UV–Vis analysis helped in determining the amount of FA and DOX covalently anchored to the surface of the QD nanocrystals. Biological screening revealed that the QD–FA–DOX nanoconjugates had higher cytotoxicity in comparison to the other forms of synthesized QD samples, suggesting the cytotoxic effect of DOX liberated from the QD constructs. Contrary to the QD–MUA–FA–DOX nanoconjugates which occurred to be the most cytotoxic against A549 cells among others, no such effect was observed for NIH/3T3 cells, confirming FARs as molecular targets. In vitro scratch assay also revealed significant inhibition of A549 cell migration after treatment with QD–MUA–FA–DOX. The performed studies evidenced that at IC50 all the nanoconjugates induced significantly more DNA breaks than that observed in nontreated cells. Overall, the QD–MUA–FA–DOX nanoconjugates showed the greatest cytotoxicity and genotoxicity, while significantly inhibiting the migratory potential of A549 cells.
Conclusion
QD–MUA–FA–DOX nanoconjugates can thus be considered as a potential drug delivery system for the effective treatment of adenocarcinomic human alveolar basal epithelial cells.
Collapse
|
62
|
Omar MM, Hasan OA, Zaki RM, Eleraky NE. Externally Triggered Novel Rapid-Release Sonosensitive Folate-Modified Liposomes for Gemcitabine: Development and Characteristics. Int J Nanomedicine 2021; 16:683-700. [PMID: 33536754 PMCID: PMC7850458 DOI: 10.2147/ijn.s266676] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/10/2020] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To develop an externally triggered rapid-release targeted system for treating ovarian cancer, gemcitabine (GMC) was entrapped into sonosensitive (SoS) folate (Fo)-modified liposomes (LPs). METHODS GMC-loaded LPs (GMC LPs), GMC-loaded Fo-targeted LPs (GMC-Fo LPs), and GMC-loaded Fo-targeted SoS LPs (GMC-SoS Fo LPs) were prepared utilizing a film-hydration technique and evaluated based on particle size, ζ-potential, and percentage entrapped drug. Cellular uptake of the fluorescent delivery systems in Fo-expressing ovarian cancer cells was quantified using flow cytometry. Finally, tumor-targeting ability, in vivo evaluation, and pharmacokinetic studies were performed. RESULTS GMC LPs, GMC-Fo LPs, and GMC-SoS Fo LPs were successfully prepared, with sizes of <120.3±2.4 nm, 39.7 mV ζ-potential, and 86.3%±1.84% entrapped drug. Cellular uptake of GMC-SoS Fo LPs improved 6.51-fold over GMC LPs (under ultrasonic irradiation - p<0.05). However, cellular uptake of GMC-Fo LPs improved just 1.24-fold over GMC LPs (p>0.05). Biodistribution study showed that of GMC concentration in tumors treated with GMC-SoS-Fo LPs (with ultrasound) improved 2.89-fold that of free GMC (p<0.05). In vivo, GMC-SoS Fo LPs showed the highest antiproliferative and antitumor action on ovarian cancer. CONCLUSION These findings showed that externally triggered rapid-release SoS Fo-modified LPs are a promising system for delivering rapid-release drugs into tumors.
Collapse
Affiliation(s)
- Mahmoud M Omar
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Deraya University, Minia, 61768, Egypt
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy,Sohag University, Sohag, 82524, Egypt
| | - Omiya Ali Hasan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Deraya University, Minia, 61768, Egypt
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy,Sohag University, Sohag, 82524, Egypt
| | - Randa Mohammed Zaki
- Department of Pharmaceutics, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Nermin E Eleraky
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
63
|
Wang H, Ding T, Guan J, Liu X, Wang J, Jin P, Hou S, Lu W, Qian J, Wang W, Zhan C. Interrogation of Folic Acid-Functionalized Nanomedicines: The Regulatory Roles of Plasma Proteins Reexamined. ACS NANO 2020; 14:14779-14789. [PMID: 33084315 DOI: 10.1021/acsnano.0c02821] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Folic acid (FA) has been extensively exploited to facilitate targeted delivery of nanomedicines by recognizing the folate receptor-α (FR-α) overexpressed in many human cancers. Unfortunately, none have been approved for clinical use yet. Here we reveal that FA functionalization induces heavy natural IgM absorption on the liposomal surface, depriving FA of receptor recognition and accelerating complement activation in vivo. FA functionalization does not enhance distribution of liposomes in FR-α-overexpressed tumors in comparison to plain liposomes (without FA), but leads to aggravated capture of liposomes by macrophages in the tumor, liver, and spleen. In addition, FA-functionalized polymeric nanoparticles are also vulnerable to natural IgM absorption. This work highlights the pivotal roles of natural IgM in regulating in vivo delivery of FA-functionalized nanomedicines. Due to the prevalent association of immune disorders and varying levels of immunoglobulins with cancer patients, extraordinary cautiousness is urged for clinical translation of FA-enabled targeted delivery systems.
Collapse
Affiliation(s)
- Huan Wang
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
| | - Tianhao Ding
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
| | - Juan Guan
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
| | - Xia Liu
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Pengpeng Jin
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| | - Weiyue Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
| | - Jun Qian
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
| | - Weiping Wang
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| |
Collapse
|
64
|
Kumar P, Behl G, Kaur S, Yadav N, Liu B, Chhikara A. Tumor microenvironment responsive nanogels as a smart triggered release platform for enhanced intracellular delivery of doxorubicin. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:385-404. [PMID: 33054642 DOI: 10.1080/09205063.2020.1837504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The fabrication of novel and intelligent delivery systems that can effectively deliver therapeutics to the targeted site and release payload in enhanced/controlled manner is highly desired to overcome the multiple challenges in chemotherapy. The present article demonstrates the potential application of dual stimuli responsive nanogels as tumor microenvironment targeted drug delivery carrier. Disulfide cross-linked pH and redox responsive PEG-PDMAEMA nanogels were synthesized by atom transfer radical polymerization (ATRP). The nanogels were characterized by nuclear magnetic resonance (NMR) spectroscopy, Fourier transform infrared spectroscopy (FTIR), and X-ray diffraction (XRD). The PEG-PDMAEMA nanogels exhibited dual stimuli-responsive release of the encapsulated model anticancer drug (doxorubicin, DOX) due to the acidic pH-response of dimethyl amine group in PDMAEMA and reductive cleavage of the disulfide linkages. A relatively higher release of DOX was observed from the nanogels at pH 5.0 than at pH 7.4. DOX release was further accelerated in tumor simulated environment of pH 5.0 and 10 mM glutathione (GSH). Confocal microscopy images revealed that DOX-loaded PEG-PDMAEMA nanogels can rapidly internalize and effectively deliver the drug into the cells. The nanogels exhibited higher cytotoxicity in GSH-OEt pretreated HeLa cells than untreated cells. The dual stimuli responsive nanogels synthesized in this study exhibited many favorable traits, such as pH and redox dependent controlled release of drug, biodegradability, biocompatibility, and enhanced cytotoxicity, which endow them as a promising candidate for anticancer drug delivery.
Collapse
Affiliation(s)
- Parveen Kumar
- Laboratory of Functional Molecules and Materials, School of Physics and Optoelectronic Engineering, Shandong University of Technology, Zibo, China.,Department of Chemistry, Dyal Singh College, University of Delhi, New Delhi, India
| | - Gautam Behl
- Department of Chemistry, Dyal Singh College, University of Delhi, New Delhi, India.,Pharmaceutical and Molecular Biotechnology Research Centre, Department of Science, Waterford Institute of Technology, Waterford, Ireland
| | - Sumeet Kaur
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Nalini Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Bo Liu
- Laboratory of Functional Molecules and Materials, School of Physics and Optoelectronic Engineering, Shandong University of Technology, Zibo, China
| | - Aruna Chhikara
- Department of Chemistry, Dyal Singh College, University of Delhi, New Delhi, India
| |
Collapse
|
65
|
Baranyai Z, Soria‐Carrera H, Alleva M, Millán‐Placer AC, Lucía A, Martín‐Rapún R, Aínsa JA, la Fuente JM. Nanotechnology‐Based Targeted Drug Delivery: An Emerging Tool to Overcome Tuberculosis. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000113] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Zsuzsa Baranyai
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Héctor Soria‐Carrera
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - Maria Alleva
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Ana C. Millán‐Placer
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
| | - Ainhoa Lucía
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Rafael Martín‐Rapún
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Departamento de Química Orgánica Facultad de Ciencias Universidad de Zaragoza Zaragoza 50009 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - José A. Aínsa
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Jesús M. la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| |
Collapse
|
66
|
Sharma S, Pukale SS, Sahel DK, Agarwal DS, Dalela M, Mohanty S, Sakhuja R, Mittal A, Chitkara D. Folate-Targeted Cholesterol-Grafted Lipo-Polymeric Nanoparticles for Chemotherapeutic Agent Delivery. AAPS PharmSciTech 2020; 21:280. [PMID: 33037506 DOI: 10.1208/s12249-020-01812-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022] Open
Abstract
Docetaxel (DTX), an FDA approved chemotherapeutic agent, is used as a first-line treatment for triple-negative breast cancer (TNBC). Its poor aqueous solubility, rapid metabolism, short half-life, and effective targeting to the cancer cells limits its optimal therapeutic use. Herein, we report folate targeted amphiphilic lipopolymer grafted with cholesterol conjugated carbonate and DL-lactide prepared by microwave assisted ring opening polymerization, for the efficient actively targeted delivery of DTX. The DTX-loaded folate-targeted lipopolymeric nanoparticles (F-DTX-LPNs) prepared by the emulsion solvent evaporation method exhibited a smaller size of ∼115.17 nm with a PDI of 0.205 and encapsulation efficiency of >80%. Further, these lipopolymeric nanoparticles (F-DTX-LPNs) showed a good on-bench stability and sustained DTX release for 7 days. Cell-based assays in MDA-MB-231 cells revealed a significant enhancement in the intracellular uptake of folate-targeted lipopolymeric nanoparticles compared to non-targeted nanoparticles. Further, methyl beta-cyclodextrin (Mβ-CD) completely inhibited the uptake of these nanoparticles in the cells, indicating a lipid raft-mediated uptake mechanism. The developed F-DTX-LPNs showed improved cytotoxicity, apoptosis, and significant fold-change in expression levels of Bcl-2, BAX and Ki-67 as compared to non-targeted DTX-LPNs and free DTX. Further, F-DTX-LPNs showed an improved in vivo pharmacokinetic profile in Sprague Dawley rats as compared to the free DTX. The bio-imaging of ex vivo tissues demonstrated that the DiR loaded folate targeted LPNs exhibited intense signals after 24 h because of slow release of DiR dye from the nanoparticles.
Collapse
|
67
|
Das RP, Chakravarti S, Patel SS, Lakhamje P, Gurjar M, Gota V, Singh BG, Kunwar A. Tuning the pharmacokinetics and efficacy of irinotecan (IRI) loaded gelatin nanoparticles through folate conjugation. Int J Pharm 2020; 586:119522. [PMID: 32534159 DOI: 10.1016/j.ijpharm.2020.119522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 02/07/2023]
Abstract
Gelatin based nanocarriers have major limitation of shorter circulation half-life (t1/2). Present study addressed this issue by conjugating gelatin with folate followed by nanoprecipitation in presence of polysorbate 80 to form folate attached gelatin nanoparticles (GNP-F). The folic acid was conjugated with gelatin through the formation of amide linkage with a maximum conjugation yield of ~69%. Cryo-SEM analysis indicated that unconjugated gelatin nanoparticles (GNP) and GNP-F were spherical of nearly identical size of ~200 nm. The irinotecan (IRI)-loading efficiency estimated for IRI-GNP and IRI-GNP-F was 6.6 ± 0.42% and 11.2 ± 0.73% respectively and both formulations showed faster release of IRI at acidic pH (~5) than at physiological pH (~7). Further IRI-GNP-F demonstrated significantly higher cytotoxicity in folate receptor (FR)-positive HeLa cells than the unconjugated IRI-GNP nanoparticles confirming active targeting. Subsequently the antitumor activity of above formulations in FR-positive fibrosarcoma (syngeneic) tumor-bearing mice followed the order of IRI-GNP-F > IRI-GNP > free IRI. The pharmacokinetic evaluation of IRI-GNP and IRI-GNP-F revealed that encapsulation of IRI within GNP without folate improved its plasma maximum concentration (Cmax). However, folate conjugation of GNP remarkably improved the t1/2 of IRI. Taken together, folate as a targeting ligand modulates the pharmacokinetic property of IRI loaded GNP to favor active verses passive targeting.
Collapse
Affiliation(s)
- Ram P Das
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| | - Sarjak Chakravarti
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Snehal S Patel
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Pooja Lakhamje
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Murari Gurjar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Vikram Gota
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India; Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Beena G Singh
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| | - Amit Kunwar
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
68
|
Roma-Rodrigues C, Rivas-García L, Baptista PV, Fernandes AR. Gene Therapy in Cancer Treatment: Why Go Nano? Pharmaceutics 2020; 12:E233. [PMID: 32151052 PMCID: PMC7150812 DOI: 10.3390/pharmaceutics12030233] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023] Open
Abstract
The proposal of gene therapy to tackle cancer development has been instrumental for the development of novel approaches and strategies to fight this disease, but the efficacy of the proposed strategies has still fallen short of delivering the full potential of gene therapy in the clinic. Despite the plethora of gene modulation approaches, e.g., gene silencing, antisense therapy, RNA interference, gene and genome editing, finding a way to efficiently deliver these effectors to the desired cell and tissue has been a challenge. Nanomedicine has put forward several innovative platforms to overcome this obstacle. Most of these platforms rely on the application of nanoscale structures, with particular focus on nanoparticles. Herein, we review the current trends on the use of nanoparticles designed for cancer gene therapy, including inorganic, organic, or biological (e.g., exosomes) variants, in clinical development and their progress towards clinical applications.
Collapse
Affiliation(s)
- Catarina Roma-Rodrigues
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Campus de Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (L.R.-G.)
| | - Lorenzo Rivas-García
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Campus de Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (L.R.-G.)
- Biomedical Research Centre, Institute of Nutrition and Food Technology, Department of Physiology, Faculty of Pharmacy, University of Granada, Avda. del Conocimiento s/n. 18071 Armilla, Granada, Spain
| | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Campus de Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (L.R.-G.)
| | - Alexandra R. Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Campus de Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (L.R.-G.)
| |
Collapse
|
69
|
Lombardo D, Calandra P, Pasqua L, Magazù S. Self-assembly of Organic Nanomaterials and Biomaterials: The Bottom-Up Approach for Functional Nanostructures Formation and Advanced Applications. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E1048. [PMID: 32110877 PMCID: PMC7084717 DOI: 10.3390/ma13051048] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022]
Abstract
In this paper, we survey recent advances in the self-assembly processes of novel functional platforms for nanomaterials and biomaterials applications. We provide an organized overview, by analyzing the main factors that influence the formation of organic nanostructured systems, while putting into evidence the main challenges, limitations and emerging approaches in the various fields of nanotechology and biotechnology. We outline how the building blocks properties, the mutual and cooperative interactions, as well as the initial spatial configuration (and environment conditions) play a fundamental role in the construction of efficient nanostructured materials with desired functional properties. The insertion of functional endgroups (such as polymers, peptides or DNA) within the nanostructured units has enormously increased the complexity of morphologies and functions that can be designed in the fabrication of bio-inspired materials capable of mimicking biological activity. However, unwanted or uncontrollable effects originating from unexpected thermodynamic perturbations or complex cooperative interactions interfere at the molecular level with the designed assembly process. Correction and harmonization of unwanted processes is one of the major challenges of the next decades and requires a deeper knowledge and understanding of the key factors that drive the formation of nanomaterials. Self-assembly of nanomaterials still remains a central topic of current research located at the interface between material science and engineering, biotechnology and nanomedicine, and it will continue to stimulate the renewed interest of biologist, physicists and materials engineers by combining the principles of molecular self-assembly with the concept of supramolecular chemistry.
Collapse
Affiliation(s)
- Domenico Lombardo
- Consiglio Nazionale delle Ricerche, Istituto per i Processi Chimico-Fisici, 98158 Messina, Italy
| | - Pietro Calandra
- Consiglio Nazionale delle Ricerche, Istituto Studio Materiali Nanostrutturati, 00015 Roma, Italy;
| | - Luigi Pasqua
- Department of Environmental and Chemical Engineering, University of Calabria, 87036 Rende, Italy;
| | - Salvatore Magazù
- Dipartimento di Scienze Matematiche e Informatiche, Scienze Fisiche e Scienze della Terra, Università di Messina, 98166 Messina, Italy;
| |
Collapse
|