1
|
Dastgerdi NK, Dastgerdi NK, Bayraktutan H, Costabile G, Atyabi F, Dinarvand R, Longobardi G, Alexander C, Conte C. Enhancing siRNA cancer therapy: Multifaceted strategies with lipid and polymer-based carrier systems. Int J Pharm 2024; 663:124545. [PMID: 39098747 DOI: 10.1016/j.ijpharm.2024.124545] [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: 01/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Cancers are increasing in prevalence and many challenges remain for their treatment, such as chemoresistance and toxicity. In this context, siRNA-based therapeutics have many potential advantages for cancer therapies as a result of their ability to reduce or prevent expression of specific cancer-related genes. However, the direct delivery of naked siRNA is hindered by issues like enzymatic degradation, insufficient cellular uptake, and poor pharmacokinetics. Hence, the discovery of a safe and efficient delivery vehicle is essential. This review explores various lipid and polymer-based delivery systems for siRNA in cancer treatment. Both polymers and lipids have garnered considerable attention as carriers for siRNA delivery. While all of these systems protect siRNA and enhance transfection efficacy, each exhibits its unique strengths. Lipid-based delivery systems, for instance, demonstrate high entrapment efficacy and utilize cost-effective materials. Conversely, polymeric-based delivery systems offer advantages through chemical modifications. Nonetheless, certain drawbacks still limit their usage. To address these limitations, combining different materials in formulations (lipid, polymer, or targeting agent) could enhance pharmaceutical properties, boost transfection efficacy, and reduce side effects. Furthermore, co-delivery of siRNA with other therapeutic agents presents a promising strategy to overcome cancer resistance. Lipid-based delivery systems have been demonstrated to encapsulate many therapeutic agents and with high efficiency, but most are limited in terms of the functionalities they display. In contrast, polymeric-based delivery systems can be chemically modified by a wide variety of routes to include multiple components, such as release or targeting elements, from the same materials backbone. Accordingly, by incorporating multiple materials such as lipids, polymers, and/or targeting agents in RNA formulations it is possible to improve the pharmaceutical properties and therapeutic efficacy while reducing side effects. This review focuses on strategies to improve siRNA cancer treatments and discusses future prospects in this important field.
Collapse
Affiliation(s)
- Nazgol Karimi Dastgerdi
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Karimi Dastgerdi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran.
| | | | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy.
| |
Collapse
|
2
|
Espuche B, Moya SE, Calderón M. Nanogels: Smart tools to enlarge the therapeutic window of gene therapy. Int J Pharm 2024; 653:123864. [PMID: 38309484 DOI: 10.1016/j.ijpharm.2024.123864] [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: 10/21/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Gene therapy can potentially treat a great number of diseases, from cancer to rare genetic disorders. Very recently, the development and emergency approval of nucleic acid-based COVID-19 vaccines confirmed its strength and versatility. However, gene therapy encounters limitations due to the lack of suitable carriers to vectorize therapeutic genetic material inside target cells. Nanogels are highly hydrated nano-size crosslinked polymeric networks that have been used in many biomedical applications, from drug delivery to tissue engineering and diagnostics. Due to their easy production, tunability, and swelling properties they have called the attention as promising vectors for gene delivery. In this review, nanogels are discussed as vectors for nucleic acid delivery aiming to enlarge gene therapy's therapeutic window. Recent works highlighting the optimization of inherent transfection efficiency and biocompatibility are reviewed here. The importance of the monomer choice, along with the internal structure, surface decoration, and responsive features are outlined for the different transfection modalities. The possible sources of toxicological endpoints in nanogels are analyzed, and the strategies to limit them are compared. Finally, perspectives are discussed to identify the remining challenges for the nanogels before their translation to the market as transfection agents.
Collapse
Affiliation(s)
- Bruno Espuche
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain; POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain
| | - Sergio E Moya
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain.
| |
Collapse
|
3
|
Moazzam M, Zhang M, Hussain A, Yu X, Huang J, Huang Y. The landscape of nanoparticle-based siRNA delivery and therapeutic development. Mol Ther 2024; 32:284-312. [PMID: 38204162 PMCID: PMC10861989 DOI: 10.1016/j.ymthe.2024.01.005] [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: 05/23/2023] [Revised: 10/01/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
Five small interfering RNA (siRNA)-based therapeutics have been approved by the Food and Drug Administration (FDA), namely patisiran, givosiran, lumasiran, inclisiran, and vutrisiran. Besides, siRNA delivery to the target site without toxicity is a big challenge for researchers, and naked-siRNA delivery possesses several challenges, including membrane impermeability, enzymatic degradation, mononuclear phagocyte system (MPS) entrapment, fast renal excretion, endosomal escape, and off-target effects. The siRNA therapeutics can silence any disease-specific gene, but their intracellular and extracellular barriers limit their clinical applications. For this purpose, several modifications have been employed to siRNA for better transfection efficiency. Still, there is a quest for better delivery systems for siRNA delivery to the target site. In recent years, nanoparticles have shown promising results in siRNA delivery with minimum toxicity and off-target effects. Patisiran is a lipid nanoparticle (LNP)-based siRNA formulation for treating hereditary transthyretin-mediated amyloidosis that ultimately warrants the use of nanoparticles from different classes, especially lipid-based nanoparticles. These nanoparticles may belong to different categories, including lipid-based, polymer-based, and inorganic nanoparticles. This review briefly discusses the lipid, polymer, and inorganic nanoparticles and their sub-types for siRNA delivery. Finally, several clinical trials related to siRNA therapeutics are addressed, followed by the future prospects and conclusions.
Collapse
Affiliation(s)
- Muhammad Moazzam
- Faculty of Engineering and Science, University of Greenwich, Medway Campus, Chatham Maritime, Kent ME4 4TB, UK
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaotong Yu
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing 100191, China.
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China; Rigerna Therapeutics Co. Ltd., Suzhou 215127, China.
| |
Collapse
|
4
|
Van de Vyver T, Muntean C, Efimova I, Krysko DV, De Backer L, De Smedt SC, Raemdonck K. The alpha-adrenergic antagonist prazosin promotes cytosolic siRNA delivery from lysosomal compartments. J Control Release 2023; 364:142-158. [PMID: 37816483 DOI: 10.1016/j.jconrel.2023.10.014] [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: 04/12/2023] [Revised: 08/26/2023] [Accepted: 10/06/2023] [Indexed: 10/12/2023]
Abstract
The widespread use of small interfering RNA (siRNA) is limited by the multiple extra- and intracellular barriers upon in vivo administration. Hence, suitable delivery systems, based on siRNA encapsulation in nanoparticles or its conjugation to targeting ligands, have been developed. Nevertheless, at the intracellular level, these state-of-the-art delivery systems still suffer from a low endosomal escape efficiency. Consequently, the bulk of the endocytosed siRNA drug rapidly accumulates in the lysosomal compartment. We recently reported that a wide variety of cationic amphiphilic drugs (CADs) can promote small nucleic acid delivery from the endolysosomal compartment into the cytosol via transient induction of lysosomal membrane permeabilization. Here, we describe the identification of alternate siRNA delivery enhancers from the NIH Clinical Compound Collection that do not have the typical physicochemical properties of CADs. Additionally, we demonstrate improved endolysosomal escape of siRNA via a cholesterol conjugate and polymeric carriers with the α1-adrenergic antagonist prazosin, which was identified as the best performing delivery enhancer from the compound screen. A more detailed assessment of the mode-of-action of prazosin suggests that a different cellular phenotype compared to typical CAD adjuvants drives cytosolic siRNA delivery. As it has been described in the literature that prazosin also induces cancer cell apoptosis and promotes antigen cross-presentation in dendritic cells, the proof-of-concept data in this work provides opportunities for the repurposing of prazosin in an anti-cancer combination strategy with siRNA.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Cristina Muntean
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent, 9000 Ghent, Belgium.
| | - Iuliia Efimova
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent, 9000 Ghent, Belgium.
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent, 9000 Ghent, Belgium; Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia.
| | - Lynn De Backer
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent, 9000 Ghent, Belgium.
| |
Collapse
|
5
|
Liu Y, Chen M, Li G, Xu S, Liu H. Construction of Core-Cross-Linked Polymer Micelles with High Biocompatibility and Stability for pH/Reduction Controllable Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12671-12679. [PMID: 37647573 DOI: 10.1021/acs.langmuir.3c01341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Polymer micelles have been studied extensively in drug delivery systems (DDS), and their stability is well known to directly affect drug delivery. In this article, a series of amphiphilic copolymers LA-PDPAn-PVPm were synthesized to prepare core-cross-linked nanoparticles (CNP) applied to controllable and targeted anticancer drug delivery. The copolymers could self-assemble in aqueous solution and form homogeneous spherical micelles with particle sizes of between 100 and 150 nm. A comparison between un-cross-linked UCNP and CNP showed that the cross-linking of LA could significantly improve the stability and responsive ability of the nanoparticles. From the in vitro-simulated drug release experiments, CNP was found to have great drug blocking ability under normal physiological conditions and could achieve rapid and efficient drug release under acidic/reducing conditions. In addition, cell experiments showed that CNP had superior biocompatibility and could target tumor cells for drug release. In conclusion, a drug carrier based on copolymer LA-PDPA-PVP realized effective controlled drug release due to the cross-linking of LA. The results will provide guidance for the design strategy of polymer micelles for drug carriers.
Collapse
Affiliation(s)
- Yehong Liu
- Key Laboratory for Advanced Materials and School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Miaoxin Chen
- Key Laboratory for Advanced Materials and School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Gaoyang Li
- Key Laboratory for Advanced Materials and School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Shouhong Xu
- Key Laboratory for Advanced Materials and School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Honglai Liu
- Key Laboratory for Advanced Materials and School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
6
|
Fazal T, Murtaza BN, Shah M, Iqbal S, Rehman MU, Jaber F, Dera AA, Awwad NS, Ibrahium HA. Recent developments in natural biopolymer based drug delivery systems. RSC Adv 2023; 13:23087-23121. [PMID: 37529365 PMCID: PMC10388836 DOI: 10.1039/d3ra03369d] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023] Open
Abstract
Targeted delivery of drug molecules to diseased sites is a great challenge in pharmaceutical and biomedical sciences. Fabrication of drug delivery systems (DDS) to target and/or diagnose sick cells is an effective means to achieve good therapeutic results along with a minimal toxicological impact on healthy cells. Biopolymers are becoming an important class of materials owing to their biodegradability, good compatibility, non-toxicity, non-immunogenicity, and long blood circulation time and high drug loading ratio for both macros as well as micro-sized drug molecules. This review summarizes the recent trends in biopolymer-based DDS, forecasting their broad future clinical applications. Cellulose chitosan, starch, silk fibroins, collagen, albumin, gelatin, alginate, agar, proteins and peptides have shown potential applications in DDS. A range of synthetic techniques have been reported to design the DDS and are discussed in the current study which is being successfully employed in ocular, dental, transdermal and intranasal delivery systems. Different formulations of DDS are also overviewed in this review article along with synthesis techniques employed for designing the DDS. The possibility of these biopolymer applications points to a new route for creating unique DDS with enhanced therapeutic qualities for scaling up creative formulations up to the clinical level.
Collapse
Affiliation(s)
- Tanzeela Fazal
- Department of Chemistry, Abbottabad University of Science and Technology Pakistan
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology Pakistan
| | - Mazloom Shah
- Department of Chemistry, Faculty of Science, Grand Asian University Sialkot Pakistan
| | - Shahid Iqbal
- Department of Chemistry, School of Natural Sciences (SNS), National University of Science and Technology (NUST) H-12 Islamabad 46000 Pakistan
| | - Mujaddad-Ur Rehman
- Department of Microbiology, Abbottabad University of Science & Technology Pakistan
| | - Fadi Jaber
- Department of Biomedical Engineering, Ajman University Ajman UAE
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University Ajman UAE
| | - Ayed A Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University Abha Saudi Arabia
| | - Nasser S Awwad
- Chemistry Department, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| | - Hala A Ibrahium
- Biology Department, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| |
Collapse
|
7
|
Altuntaş E, Özkan B, Güngör S, Özsoy Y. Biopolymer-Based Nanogel Approach in Drug Delivery: Basic Concept and Current Developments. Pharmaceutics 2023; 15:1644. [PMID: 37376092 DOI: 10.3390/pharmaceutics15061644] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Due to their increased surface area, extent of swelling and active substance-loading capacity and flexibility, nanogels made from natural and synthetic polymers have gained significant interest in scientific and industrial areas. In particular, the customized design and implementation of nontoxic, biocompatible, and biodegradable micro/nano carriers makes their usage very feasible for a range of biomedical applications, including drug delivery, tissue engineering, and bioimaging. The design and application methodologies of nanogels are outlined in this review. Additionally, the most recent advancements in nanogel biomedical applications are discussed, with particular emphasis on applications for the delivery of drugs and biomolecules.
Collapse
Affiliation(s)
- Ebru Altuntaş
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| | - Burcu Özkan
- Graduate School of Natural and Applied Science, Yildiz Technical University, 34220 Istanbul, Türkiye
| | - Sevgi Güngör
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| | - Yıldız Özsoy
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| |
Collapse
|
8
|
Lei H, Fan D. A Combination Therapy Using Electrical Stimulation and Adaptive, Conductive Hydrogels Loaded with Self-Assembled Nanogels Incorporating Short Interfering RNA Promotes the Repair of Diabetic Chronic Wounds. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201425. [PMID: 36064844 PMCID: PMC9596839 DOI: 10.1002/advs.202201425] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/24/2022] [Indexed: 05/08/2023]
Abstract
In addition to oxidative stress and impaired angiogenesis, the overexpression of metalloproteinases (MMPs) and proinflammatory cytokines, which are promoted by hyperglycemia, causes chronic inflammation in diabetic wounds. Herein, TA-siRNA nanogels are prepared for the first time on the basis of the self-assembling interaction between tannic acid (TA) and short interfering RNA (siRNA). The efficient, biodegradable nanogels are cross-linked with poly(vinyl alcohol) (PVA), human-like collagen (HLC), TA, and borax to prepare adaptive, conductive PHTB (TA-siRNA) hydrogels. In response to high levels of reactive oxygen species (ROS), the ROS-responsive borate ester bonds in the hydrogels are oxidized and broken, and TA-siRNA nanogels are released into cells to reduce the expression of the MMP-9. Moreover, the TA and HLC promote collagen expression, reduce inflammation, and ROS level. It is found that electrical stimulation (ES) promotes the in vivo release of TA-siRNA nanogels from PHTB (TA-siRNA) hydrogels and endocytosis of the nanogels. The combination therapy using ES and PHTB (TA-siRNA) hydrogels accelerates the healing of diabetic wounds by reducing the levels of ROS and MMP-9 and promoting the polarization of macrophages, production of collagen, and angiogenesis. This study provides insights on the design of functional gene-delivery and efficient therapeutic strategies to promote the repair of diabetic chronic wounds.
Collapse
Affiliation(s)
- Huan Lei
- Shaanxi Key Laboratory of Degradable Biomedical MaterialsShaanxi R&D Center of Biomaterials and Fermentation EngineeringBiotech. & Biomed. Research InstituteNorthwest UniversityTaibai North Road 229Xi'anShaanxi710069China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical MaterialsShaanxi R&D Center of Biomaterials and Fermentation EngineeringBiotech. & Biomed. Research InstituteNorthwest UniversityTaibai North Road 229Xi'anShaanxi710069China
| |
Collapse
|
9
|
Reddy Shetty P, Batchu UR, Buddana SK, Sambasiva Rao K, Penna S. A comprehensive review on α-D-Glucans: Structural and functional diversity, derivatization and bioapplications. Carbohydr Res 2021; 503:108297. [PMID: 33813321 DOI: 10.1016/j.carres.2021.108297] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Glucans are the most abundant natural polysaccharides across the living kingdom with tremendous biological activities. Now a days, α-D-glucans are gaining importance as a prebiotics, nutraceuticals, immunostimulants, antiproliferative agents and biodegradable polymers in pharmaceutical and cosmetic sectors. A wide variety of bioresources including bacteria, fungi, lichens, algae, plants and animals produce α-D-glucans either as an exopolysaccharide (EPS) or a cell wall component or an energy storage polymer. The α-D-glucans exhibit great structural and functional diversity as the type of linkage and percentage of branching dictate the functional properties of glucans. Among the different linkages, bioactivities are greatly confined to the α-D-(1 → 3) linkages whereas starch and other polymers consisting of α-D-(1 → 4) (1 → 6) linkages are specific for food and pharmaceutical applications. However, the bioactivities of the α-D-(1 → 3) glucans in native form is limited mainly due to their hydrophobic nature. Hence several derivatization techniques have been developed to improve the bioavailability as well as bioactive features such as antiviral, antimicrobial, anti-inflammatory, antioxidant, immunomodulatory and antitumor properties. Though, several reports have presented about α-D-glucans, still there is an ambiguity in terms of their structure among different natural sources and moreover no comprehensive information was available on their derivatization techniques and application potential. Therefore, the present review summarizes distinct description on diverse sources, type of linkages, derivatization techniques as well as the application potential of the native and modified α-D-glucans.
Collapse
Affiliation(s)
- Prakasham Reddy Shetty
- Medicinal Chemistry and Biotechnology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, Telangana, India.
| | - Uma Rajeswari Batchu
- Medicinal Chemistry and Biotechnology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, Telangana, India.
| | - Sudheer Kumar Buddana
- Medicinal Chemistry and Biotechnology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology, Ghaziabad, 201001, New Delhi, India.
| | - Krs Sambasiva Rao
- Department of Biotechnology, Acharya Nagarjuna University, Guntur, 522510, Andhra Pradesh, India.
| | - Suprasanna Penna
- Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre (BARC), Mumbai, 400085, Maharashtra, India.
| |
Collapse
|
10
|
Contributions of exopolysaccharides from lactic acid bacteria as biotechnological tools in food, pharmaceutical, and medical applications. Int J Biol Macromol 2021; 173:79-89. [PMID: 33482209 DOI: 10.1016/j.ijbiomac.2021.01.110] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/10/2020] [Accepted: 01/16/2021] [Indexed: 01/08/2023]
Abstract
Exopolysaccharides (EPS) are important bioproducts produced by some genera of lactic acid bacteria. EPS are famous for their shelf-life improving properties, techno-functional enhancing abilities in food and dairy industries, besides their beneficial health effects. Furthermore, exopolysaccharides have many prospective and well-established contributions in the field of drugs and diagnostic industry. In this review, classification of EPS produced by LAB was presented. Moreover, current and potential applications of EPS in food, dairy, baking industries, cereal-based, and functional products were described. Also, some clinical and pharmaceutical applications of EPS such as intelligent drug delivery systems (microsystems and nanosystems for sustained delivery), interpenetrating polymer networks (IPNs), anticancer drug-targeting, recombinant macromolecular biopharmaceuticals, gene delivery, tissue engineering, and role of EPS in diagnostics were highlighted. Finally, future prospects concerning enhancing EPS production, minimizing costs of their production, and exploring their contribution in further applications were discussed.
Collapse
|
11
|
Torres J, Dhas N, Longhi M, García MC. Overcoming Biological Barriers With Block Copolymers-Based Self-Assembled Nanocarriers. Recent Advances in Delivery of Anticancer Therapeutics. Front Pharmacol 2020; 11:593197. [PMID: 33329001 PMCID: PMC7734332 DOI: 10.3389/fphar.2020.593197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/15/2020] [Indexed: 11/21/2022] Open
Abstract
Cancer is one of the most common life-threatening illness and it is the world's second largest cause of death. Chemotherapeutic anticancer drugs have many disadvantages, which led to the need to develop novel strategies to overcome these shortcomings. Moreover, tumors are heterogenous in nature and there are various biological barriers that assist in treatment reisistance. In this sense, nanotechnology has provided new strategies for delivery of anticancer therapeutics. Recently, delivery platforms for overcoming biological barriers raised by tumor cells and tumor-bearing hosts have been reported. Among them, amphiphilic block copolymers (ABC)-based self-assembled nanocarriers have attracted researchers worldwide owing to their unique properties. In this work, we addressed different biological barriers for effective cancer treatment along with several strategies to overcome them by using ABC-based self-assembled nanostructures, with special emphasis in those that have the ability to act as responsive nanocarriers to internal or external environmental clues to trigger release of the payload. These nanocarriers have shown promising properties to revolutionize cancer treatment and diagnosis, but there are still challenges for their successful translation to clinical applications.
Collapse
Affiliation(s)
- Jazmin Torres
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Namdev Dhas
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Marcela Longhi
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Mónica C. García
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
12
|
Shah S, Rangaraj N, Laxmikeshav K, Sampathi S. “Nanogels as drug carriers – Introduction, chemical aspects, release mechanisms and potential applications”. Int J Pharm 2020; 581:119268. [DOI: 10.1016/j.ijpharm.2020.119268] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/12/2020] [Accepted: 03/24/2020] [Indexed: 12/28/2022]
|
13
|
Van de Vyver T, Bogaert B, De Backer L, Joris F, Guagliardo R, Van Hoeck J, Merckx P, Van Calenbergh S, Ramishetti S, Peer D, Remaut K, De Smedt SC, Raemdonck K. Cationic Amphiphilic Drugs Boost the Lysosomal Escape of Small Nucleic Acid Therapeutics in a Nanocarrier-Dependent Manner. ACS NANO 2020; 14:4774-4791. [PMID: 32250113 DOI: 10.1021/acsnano.0c00666] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Small nucleic acid (NA) therapeutics, such as small interfering RNA (siRNA), are generally formulated in nanoparticles (NPs) to overcome the multiple extra- and intracellular barriers upon in vivo administration. Interaction with target cells typically triggers endocytosis and sequesters the NPs in endosomes, thus hampering the pharmacological activity of the encapsulated siRNAs that occurs in the cytosol. Unfortunately, for most state-of-the-art NPs, endosomal escape is largely inefficient. As a result, the bulk of the endocytosed NA drug is rapidly trafficked toward the degradative lysosomes that are considered as a dead end for siRNA nanomedicines. In contrast to this paradigm, we recently reported that cationic amphiphilic drugs (CADs) could strongly promote functional siRNA delivery from the endolysosomal compartment via transient induction of lysosomal membrane permeabilization. However, many questions still remain regarding the broader applicability of such a CAD adjuvant effect on NA delivery. Here, we report a drug repurposing screen (National Institutes of Health Clinical Collection) that allowed identification of 56 CAD adjuvants. We furthermore demonstrate that the CAD adjuvant effect is dependent on the type of nanocarrier, with NPs that generate an appropriate pool of decomplexed siRNA in the endolysosomal compartment being most susceptible to CAD-promoted gene silencing. Finally, the CAD adjuvant effect was verified on human ovarian cancer cells and for antisense oligonucleotides. In conclusion, this study strongly expands our current knowledge on how CADs increase the cytosolic release of small NAs, providing relevant insights to more rationally combine CAD adjuvants with NA-loaded NPs for future therapeutic applications.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Bram Bogaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Lynn De Backer
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Freya Joris
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Roberta Guagliardo
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Jelter Van Hoeck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Pieterjan Merckx
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | | | | | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
14
|
Delcassian D, Luzhansky I, Spanoudaki V, Bochenek M, McGladrigan C, Nguyen A, Norcross S, Zhu Y, Shan CS, Hausser R, Shakesheff KM, Langer R, Anderson DG. Magnetic Retrieval of Encapsulated Beta Cell Transplants from Diabetic Mice Using Dual-Function MRI Visible and Retrievable Microcapsules. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904502. [PMID: 32134138 DOI: 10.1002/adma.201904502] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 12/03/2019] [Indexed: 05/18/2023]
Abstract
Encapsulated beta cell transplantation offers a potential cure for a subset of diabetic patients. Once transplanted, beta cell grafts can help to restore glycemic control; however, locating and retrieving cells in the event of graft failure may pose a surgical challenge. Here, a dual-function nanoparticle-loaded hydrogel microcapsule is developed that enables graft retrieval under an applied magnetic field. Additionally, this system facilitates graft localization via magnetic resonance imaging (MRI), and graft isolation from the immune system. Iron oxide nanoparticles encapsulated within alginate hydrogel capsules containing viable islets are transplanted and the in vitro and in vivo retrieval of capsules containing nanoparticles functionalized with various ligands are compared. Capsules containing islets co-encapsulated with COOH-coated nanoparticles restore normal glycemia in immunocompetent diabetic mice for at least 6 weeks, can be visualized using MRI, and are retrievable in a magnetic field. Application of a magnetic field for 90 s via a magnetically assisted retrieval device facilitates rapid retrieval of up to 94% (±3.1%) of the transplant volume 24 h after surgical implantation. This strategy aids monitoring of cell-capsule locations in vivo, facilitates graft removal at the end of the transplant lifetime, and may be applicable to many encapsulated cell transplant systems.
Collapse
Affiliation(s)
- Derfogail Delcassian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Igor Luzhansky
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Virginia Spanoudaki
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Matthew Bochenek
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Collin McGladrigan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Amy Nguyen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Samuel Norcross
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Yuhan Zhu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Crystal Shuo Shan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Reed Hausser
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Kevin M Shakesheff
- Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
15
|
Leber N, Zentel R. Improved SiRNA Loading of Cationic Nanohydrogel Particles by Variation of Crosslinking Density. MACROMOL CHEM PHYS 2019. [DOI: 10.1002/macp.201900298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Nadine Leber
- Institute of Organic ChemistryJohannes Gutenberg‐University of Mainz Duesbergweg 10‐14 55128 Mainz Germany
| | - Rudolf Zentel
- Institute of Organic ChemistryJohannes Gutenberg‐University of Mainz Duesbergweg 10‐14 55128 Mainz Germany
| |
Collapse
|
16
|
Kordalivand N, Tondini E, Lau CYJ, Vermonden T, Mastrobattista E, Hennink WE, Ossendorp F, Nostrum CFV. Cationic synthetic long peptides-loaded nanogels: An efficient therapeutic vaccine formulation for induction of T-cell responses. J Control Release 2019; 315:114-125. [PMID: 31672626 DOI: 10.1016/j.jconrel.2019.10.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/23/2019] [Accepted: 10/26/2019] [Indexed: 12/18/2022]
Abstract
Recent studies have shown a high potency of protein-based vaccines for cell-mediated cancer immunotherapy. However, due to their poor cellular uptake, efficient immune responses with soluble protein antigens are often not observed. As a result of superior cellular uptake, nanogels loaded with antigenic peptides were investigated in this study as carrier systems for cancer immunotherapy. Different synthetic long peptides (SLPs) containing the CTL and CD4+ T-helper (Help) epitopes were synthesized and covalently conjugated via disulfide bonds to the polymeric network of cationic dextran nanogels. Cationic nanogels with a size of 210 nm, positive zeta potential (+24 mV) and high peptide loading content (15%) showed triggered release of the loaded peptides under reducing conditions. An in vitro study demonstrated the capability of cationic nanogels to maturate dendritic cells (DCs). Importantly, covalently SLP-loaded nanogels adjuvanted with poly(I:C) showed superior CD8+ T cell responses compared to soluble peptides and nanogel formulations with physically loaded peptides both in vitro and in vivo. In conclusion, covalently SLPs-loaded cationic nanogels are a promising system to provoke immune responses for therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Neda Kordalivand
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Elena Tondini
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Chun Yin Jerry Lau
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
17
|
Nanocarrier Lipid Composition Modulates the Impact of Pulmonary Surfactant Protein B (SP-B) on Cellular Delivery of siRNA. Pharmaceutics 2019; 11:pharmaceutics11090431. [PMID: 31450805 PMCID: PMC6781292 DOI: 10.3390/pharmaceutics11090431] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
Two decades since the discovery of the RNA interference (RNAi) pathway, we are now witnessing the approval of the first RNAi-based treatments with small interfering RNA (siRNA) drugs. Nevertheless, the widespread use of siRNA is limited by various extra- and intracellular barriers, requiring its encapsulation in a suitable (nanosized) delivery system. On the intracellular level, the endosomal membrane is a major barrier following endocytosis of siRNA-loaded nanoparticles in target cells and innovative materials to promote cytosolic siRNA delivery are highly sought after. We previously identified the endogenous lung surfactant protein B (SP-B) as siRNA delivery enhancer when reconstituted in (proteo) lipid-coated nanogels. It is known that the surface-active function of SP-B in the lung is influenced by the lipid composition of the lung surfactant. Here, we investigated the role of the lipid component on the siRNA delivery-promoting activity of SP-B proteolipid-coated nanogels in more detail. Our results clearly indicate that SP-B prefers fluid membranes with cholesterol not exceeding physiological levels. In addition, SP-B retains its activity in the presence of different classes of anionic lipids. In contrast, comparable fractions of SP-B did not promote the siRNA delivery potential of DOTAP:DOPE cationic liposomes. Finally, we demonstrate that the beneficial effect of lung surfactant on siRNA delivery is not limited to lung-related cell types, providing broader therapeutic opportunities in other tissues as well.
Collapse
|
18
|
Merckx P, De Backer L, Van Hoecke L, Guagliardo R, Echaide M, Baatsen P, Olmeda B, Saelens X, Pérez-Gil J, De Smedt SC, Raemdonck K. Surfactant protein B (SP-B) enhances the cellular siRNA delivery of proteolipid coated nanogels for inhalation therapy. Acta Biomater 2018; 78:236-246. [PMID: 30118853 DOI: 10.1016/j.actbio.2018.08.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022]
Abstract
Despite the many advantages of small interfering RNA (siRNA) inhalation therapy and a growing prevalence of respiratory pathologies, its clinical translation is severely hampered by inefficient intracellular delivery. To this end, we previously developed hybrid nanoparticles consisting of an siRNA-loaded nanosized hydrogel core (nanogel) coated with Curosurf®, a clinically used pulmonary surfactant (PS). Interestingly, the PS shell was shown to markedly improve particle stability as well as intracellular siRNA delivery in vitro and in vivo. The major aim of this work was to identify the key molecular components of PS responsible for the enhanced siRNA delivery and evaluate how the complexity of the PS coat could be reduced. We identified surfactant protein B (SP-B) as a potent siRNA delivery enhancer when reconstituted in proteolipid coated hydrogel nanocomposites. Improved cytosolic siRNA delivery was achieved by inserting SP-B into a simplified phospholipid mixture prior to nanogel coating. This effect was observed both in vitro (lung epithelial cell line) and in vivo (murine acute lung injury model), albeit that distinct phospholipids were required to achieve these results. Importantly, the developed nanocomposites have a low in vivo toxicity and are efficiently taken up by resident alveolar macrophages, a main target cell type for treatment of inflammatory pulmonary pathologies. Our results demonstrate the potential of the endogenous protein SP-B as an intracellular siRNA delivery enhancer, paving the way for future design of nanoformulations for siRNA inhalation therapy. STATEMENT OF SIGNIFICANCE Despite the therapeutic potential of small interfering RNA (siRNA) and a growing prevalence of lung diseases for which innovative therapies are needed, a safe and effective siRNA inhalation therapy remains non-existing due to a lack of suitable formulations. We identified surfactant protein B (SP-B) as a potent enhancer of siRNA delivery by proteolipid coated nanogel formulations in vitro in a lung epithelial cell line. The developed nanocomposites have a low in vivo toxicity and show a high uptake by alveolar macrophages, a main target cell type for treatment of inflammatory pulmonary pathologies. Importantly, in vivo SP-B is also critical for the developed formulation to obtain a significant silencing of TNFα in a murine LPS-induced acute lung injury model.
Collapse
|
19
|
Villarreal-Gómez LJ, Serrano-Medina A, José Torres-Martínez E, Lizeth Perez-González G, Manuel Cornejo-Bravo J. Polymeric advanced delivery systems for antineoplasic drugs: doxorubicin and 5-fluorouracil. E-POLYMERS 2018. [DOI: 10.1515/epoly-2017-0202] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AbstractConventional pharmaceuticals generally display the inability to transport active ingredients directly to specific regions of the body, amongst some of their main limitations. The distribution of the drugs in the circulatory system may lead to undesired toxicity, and therefore, adverse reactions. To address this situation, a selective transport of drugs is required, that is, releasing drugs specifically to the site of action in appropriate concentrations and in the right time. To achieve this goal, it is necessary to develop delivery systems that respond to several features, such as low toxicity, optimum properties for the transport and release of the drug, as well as a long half-life in the body. This feature paper critically provides an overview of different strategies of controlled drug release for two model antineoplasic drugs, i.e. doxorubicin (DOX) and 5-fluorouracil (5-FU). Any of the presented strategies for drug release possess advantages and disadvantages, and the selection of the strategy used will depend on the targeted tissue and nature of the drug.
Collapse
Affiliation(s)
- Luis Jesús Villarreal-Gómez
- Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana, Baja California C.P. 22390, México
- Escuela de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Unidad Valle de las Palmas, Tijuana, Baja California, México
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Unidad Otay, Tijuana, Baja California, México
| | - Aracely Serrano-Medina
- Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana, Baja California C.P. 22390, México
- Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Unidad Otay, Tijuana, Baja California, México
| | - Erick José Torres-Martínez
- Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana, Baja California C.P. 22390, México
- Escuela de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Unidad Valle de las Palmas, Tijuana, Baja California, México
| | - Graciela Lizeth Perez-González
- Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana, Baja California C.P. 22390, México
- Escuela de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Unidad Valle de las Palmas, Tijuana, Baja California, México
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Unidad Otay, Tijuana, Baja California, México
| | - José Manuel Cornejo-Bravo
- Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana, Baja California C.P. 22390, México
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Unidad Otay, Tijuana, Baja California, México
| |
Collapse
|
20
|
Interaction of drugs amlodipine and paroxetine with the metabolizing enzyme CYP2B4: a molecular dynamics simulation study. J Mol Model 2018; 24:67. [DOI: 10.1007/s00894-018-3617-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/08/2018] [Indexed: 12/26/2022]
|
21
|
Abstract
Over the last century, there has been a dramatic change in the nature of therapeutic, biologically active molecules available to treat disease. Therapies have evolved from extracted natural products towards rationally designed biomolecules, including small molecules, engineered proteins and nucleic acids. The use of potent drugs which target specific organs, cells or biochemical pathways, necessitates new tools which can enable controlled delivery and dosing of these therapeutics to their biological targets. Here, we review the miniaturisation of drug delivery systems from the macro to nano-scale, focussing on controlled dosing and controlled targeting as two key parameters in drug delivery device design. We describe how the miniaturisation of these devices enables the move from repeated, systemic dosing, to on-demand, targeted delivery of therapeutic drugs and highlight areas of focus for the future.
Collapse
Affiliation(s)
- Derfogail Delcassian
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,b Department of Anaesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , MA , USA.,c Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Asha K Patel
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,d Division of Cancer and Stem Cells, School of Medicine, and Division of Advanced Materials and Healthcare Technologies, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Abel B Cortinas
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,e Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Robert Langer
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,e Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA.,f Institute for Medical Engineering and Science , Massachusetts Institute of Technology , Cambridge , MA , USA.,g Media Lab , Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
22
|
Hong SJ, Ahn MH, Sangshetti J, Choung PH, Arote RB. Sugar-based gene delivery systems: Current knowledge and new perspectives. Carbohydr Polym 2018; 181:1180-1193. [DOI: 10.1016/j.carbpol.2017.11.105] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/26/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022]
|
23
|
Dilnawaz F, Acharya S, Sahoo SK. Recent trends of nanomedicinal approaches in clinics. Int J Pharm 2018; 538:263-278. [PMID: 29339248 DOI: 10.1016/j.ijpharm.2018.01.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 12/20/2022]
Abstract
Nanotechnology has become the indispensable cutting edge science providing solutions to many problems associated with human being. The application of nanotechnology associated to human health "nanomedicine" has revolutionized the drug delivery system by providing improved pharmacological and therapeutic properties of drugs. These advantageous effects of drug loaded nanocarrier systems are embraced by the pharmaceutical industries for the development of different effective nanocarriers. Currently, several drug loaded nanoformulations are approved by the Food and Drug Administration (FDA), and some of them are undergoing clinical trials for the human use. In this review, we have discussed the progress achieved so far for various drug loaded nanoformulations along with few emerging nanoformulations that are about to enter into clinical trials.
Collapse
Affiliation(s)
- Fahima Dilnawaz
- Laboratory of Nanomedicine, Institute of Life Sciences, Bhubaneswar 751023, Odisha, India
| | - Sarbari Acharya
- Laboratory of Nanomedicine, Institute of Life Sciences, Bhubaneswar 751023, Odisha, India
| | - Sanjeeb Kumar Sahoo
- Laboratory of Nanomedicine, Institute of Life Sciences, Bhubaneswar 751023, Odisha, India.
| |
Collapse
|
24
|
Stimuli-responsive nanocarriers for intracellular delivery. Biophys Rev 2017; 9:931-940. [PMID: 29178081 DOI: 10.1007/s12551-017-0341-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
The emergence of different nanoparticles (NPs) has made a significant revolution in the field of medicine. Different NPs in the form of metallic NPs, dendrimers, polymeric NPs, carbon quantum dots and liposomes have been functionalized and used as platforms for intracellular delivery of biomolecules, drugs, imaging agents and nucleic acids. These NPs are designed to improve the pharmacokinetic properties of the drug, improve their bioavailability and successfully surpass physiological or pathological obstacles in the biological system so that therapeutic efficacy is achieved. In this review I present some of the current approaches used in intracellular delivery systems, with a focus on various stimuli-responsive nanocarriers, including cell-penetrating peptides, to highlight their various biomedical applications.
Collapse
|
25
|
Leber N, Nuhn L, Zentel R. Cationic Nanohydrogel Particles for Therapeutic Oligonucleotide Delivery. Macromol Biosci 2017; 17. [PMID: 28605133 DOI: 10.1002/mabi.201700092] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 05/04/2017] [Indexed: 02/02/2023]
Abstract
Short pharmaceutical active oligonucleotides such as small interfering RNA (siRNA) or cytidine-phosphate-guanosine (CpG) are considered as powerful therapeutic alternatives, especially to medicate hard-to-treat diseases (e.g., liver fibrosis or cancer). Unfortunately, these molecules are equipped with poor pharmacokinetic properties that prevent them from translation. Well-defined nanosized carriers can provide opportunities to optimize their delivery and guide them to their site of action. Among several concepts, this Feature Article focuses on cationic nanohydrogel particles as a universal delivery system for small anionic molecules including siRNA and CpG. Cationic nanohydrogels are derived from preaggregated precursor block copolymers, which are further cross-linked to obtain well-defined nanoparticles of tunable sizes and with (degradable) cationic cores. Novel opportunities for oligonucleotide delivery in vitro and in vivo with respect to liver fibrosis therapies will be highlighted as well as perspectives toward modulating the immune system. In general, the approach of covalently stabilized cationic carrier systems can contribute to find advanced oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Nadine Leber
- Institute of Organic Chemistry, Johannes Gutenberg University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Lutz Nuhn
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Rudolf Zentel
- Institute of Organic Chemistry, Johannes Gutenberg University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|
26
|
Polysaccharide-based Nanoparticles for Gene Delivery. Top Curr Chem (Cham) 2017; 375:31. [DOI: 10.1007/s41061-017-0114-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/25/2017] [Indexed: 12/17/2022]
|
27
|
Nguyen MK, McMillan A, Huynh CT, Schapira DS, Alsberg E. Photocrosslinkable, biodegradable hydrogels with controlled cell adhesivity for prolonged siRNA delivery to hMSCs to enhance their osteogenic differentiation. J Mater Chem B 2017; 5:485-495. [PMID: 28652917 PMCID: PMC5482539 DOI: 10.1039/c6tb01739h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Photocrosslinked, biodegradable hydrogels have been extensively investigated for biomedical applications, including drug delivery and tissue engineering. Here, dextran (DEX) was chemically modified with mono(2-acryloyloxyethyl) succinate (MAES) via an esterification reaction, resulting in macromers that could be photocrosslinked to form hydrolytically degradable hydrogels. Hydrogel swelling ratio and degradation rate were controlled by varying the degree of MAES modification. Thiolated cell adhesion peptides (GRGDSPC) were conjugated to acrylated dextran via thiol-acrylate reaction to regulate the interactions of human mesenchymal stem cells (hMSCs) with the photocrosslinkable hydrogels. The hydrogels permitted sustained release of short interfering RNA (siRNA) over 7 weeks and were cytocompatible with hMSCs. Sustained presentation of siRNA from these photocrosslinked DEX hydrogels enhanced the osteogenic differentiation of encapsulated hMSCs. These DEX hydrogels with tunable siRNA delivery and cell adhesive properties may provide an excellent platform for bioactive molecule delivery and tissue regeneration applications.
Collapse
Affiliation(s)
- Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Alexandra McMillan
- Department of Pathology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Daniel S Schapira
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
- National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| |
Collapse
|
28
|
Li D, Sun F, Bourajjaj M, Chen Y, Pieters EH, Chen J, van den Dikkenberg JB, Lou B, Camps MGM, Ossendorp F, Hennink WE, Vermonden T, van Nostrum CF. Strong in vivo antitumor responses induced by an antigen immobilized in nanogels via reducible bonds. NANOSCALE 2016; 8:19592-19604. [PMID: 27748778 DOI: 10.1039/c6nr05583d] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cancer vaccines are at present mostly based on tumor associated protein antigens but fail to elicit strong cell-mediated immunity in their free form. For protein-based vaccines, the main challenges to overcome are the delivery of sufficient proteins into the cytosol of dendritic cells (DCs) and processing by, and presentation through, the MHC class I pathway. Recently, we developed a cationic dextran nanogel in which a model antigen (ovalbumin, OVA) is reversibly conjugated via disulfide bonds to the nanogel network to enable redox-sensitive intracellular release. In the present study, it is demonstrated that these nanogels, with the bound OVA, were efficiently internalized by DCs and were capable of maturating them. On the other hand, when the antigen was just physically entrapped in the nanogels, OVA was prematurely released before the particles were taken up by cells. When combined with an adjuvant (polyinosinic-polycytidylic acid, poly(I:C)), nanogels with conjugated OVA induced a strong protective and curative effect against melanoma in vivo. In a prophylactic vaccination setting, 90% of the mice vaccinated with nanogels with conjugated OVA + poly(I:C) did not develop a tumor. Moreover, in a therapeutic model, 40% of the mice showed clearance of established tumors and survived for the duration of the experiment (80 days) while the remaining mice showed substantial delay in tumor progression. In conclusion, our results demonstrate that conjugation of antigens to nanogels via reducible covalent bonds for intracellular delivery is a promising strategy to induce effective antigen-specific immune responses against cancer.
Collapse
Affiliation(s)
- Dandan Li
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Feilong Sun
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Meriem Bourajjaj
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Yinan Chen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Ebel H Pieters
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Jian Chen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Joep B van den Dikkenberg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Bo Lou
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Marcel G M Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584CG, The Netherlands.
| |
Collapse
|
29
|
Debele TA, Mekuria SL, Tsai HC. Polysaccharide based nanogels in the drug delivery system: Application as the carrier of pharmaceutical agents. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 68:964-981. [DOI: 10.1016/j.msec.2016.05.121] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/23/2016] [Accepted: 05/27/2016] [Indexed: 11/08/2022]
|
30
|
Soni KS, Desale SS, Bronich TK. Nanogels: An overview of properties, biomedical applications and obstacles to clinical translation. J Control Release 2016; 240:109-126. [PMID: 26571000 PMCID: PMC4862943 DOI: 10.1016/j.jconrel.2015.11.009] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/01/2015] [Accepted: 11/09/2015] [Indexed: 01/09/2023]
Abstract
Nanogels have emerged as a versatile hydrophilic platform for encapsulation of guest molecules with a capability to respond to external stimuli that can be used for a multitude of applications. These are soft materials capable of holding small molecular therapeutics, biomacromolecules, and inorganic nanoparticles within their crosslinked networks, which allows them to find applications for therapy as well as imaging of a variety of disease conditions. Their stimuli-responsive behavior can be easily controlled by selection of constituent polymer and crosslinker components to achieve a desired response at the site of action, which imparts nanogels the ability to participate actively in the intended function of the carrier system rather than being passive carriers of their cargo. These properties not only enhance the functionality of the carrier system but also help in overcoming many of the challenges associated with the delivery of cargo molecules, and this review aims to highlight the distinct and unique capabilities of nanogels as carrier systems for the delivery of an array of cargo molecules over other nanomaterials. Despite their obvious usefulness, nanogels are still not a commonplace occurrence in clinical practice. We have also made an attempt to highlight some of the major challenges that need to be overcome to advance nanogels further in the field of biomedical applications.
Collapse
Affiliation(s)
- Kruti S Soni
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Swapnil S Desale
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| |
Collapse
|
31
|
Aguirre G, Ramos J, Forcada J. Advanced design of t and pH dual-responsive PDEAEMA-PVCL core-shell nanogels for siRNA delivery. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/pola.28207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Garbiñe Aguirre
- POLYMAT, Bionanoparticles Group, Department of Applied Chemistry, UFI 11/56, Faculty of Chemistry; University of the Basque Country UPV/EHU; Apdo. 1072 Donostia-San Sebastián 20080 Spain
| | - Jose Ramos
- POLYMAT, Bionanoparticles Group, Department of Applied Chemistry, UFI 11/56, Faculty of Chemistry; University of the Basque Country UPV/EHU; Apdo. 1072 Donostia-San Sebastián 20080 Spain
| | - Jacqueline Forcada
- POLYMAT, Bionanoparticles Group, Department of Applied Chemistry, UFI 11/56, Faculty of Chemistry; University of the Basque Country UPV/EHU; Apdo. 1072 Donostia-San Sebastián 20080 Spain
| |
Collapse
|
32
|
Soni G, Yadav KS. Nanogels as potential nanomedicine carrier for treatment of cancer: A mini review of the state of the art. Saudi Pharm J 2016; 24:133-9. [PMID: 27013905 PMCID: PMC4792897 DOI: 10.1016/j.jsps.2014.04.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 04/02/2014] [Indexed: 12/15/2022] Open
Abstract
Nanogels are being explored as drug delivery agents for targeting cancer due to their easy tailoring properties and ability to efficiently encapsulate therapeutics of diverse nature through simple mechanisms. Nanogels are proficiently internalized by the target cells, avoid accumulating in nontarget tissues thereby lower the therapeutic dosage and minimize harmful side effects. However, there is an urgent need for relevant clinical data from nanogels so as to allow translation of the nanogel concept into a viable therapeutic application for the treatment of cancer. This review highlights some of the recent progress in nanogels as a carrier in the field of nanomedicine for the treatment of cancer. The present review critically analyzes the use of extracellular pH targeting for nanogels, siRNA delivery, PEGylated nanogels, multi-responsive nanogels and intracellular delivery of nanogels for improved therapy of cancer.
Collapse
Affiliation(s)
| | - Khushwant S. Yadav
- Department of Pharmaceutics, Rajeev Gandhi College of Pharmacy, Salaiya, Kolar Road, Bhopal 462042, MP, India
| |
Collapse
|
33
|
Kaps L, Nuhn L, Aslam M, Brose A, Foerster F, Rosigkeit S, Renz P, Heck R, Kim YO, Lieberwirth I, Schuppan D, Zentel R. In Vivo Gene-Silencing in Fibrotic Liver by siRNA-Loaded Cationic Nanohydrogel Particles. Adv Healthc Mater 2015; 4:2809-15. [PMID: 26627192 DOI: 10.1002/adhm.201500826] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Indexed: 12/20/2022]
Abstract
Cationic nanohydrogel particles loaded with anti-Col1α1 siRNA suppress collagen synthesis and deposition in fibrotic mice: Systemically administered 40 nm sized nanogel particles accumulate in collagen-expressing cells in the liver. Their siRNA payload induces a sequence specific in vivo gene knockdown affording an efficient antifibrotic effect in mice with liver fibrosis.
Collapse
Affiliation(s)
- Leonard Kaps
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Lutz Nuhn
- Institute of Organic Chemistry; Johannes Gutenberg-University Mainz; Duesbergweg 10-14 55128 Mainz Germany
- Department of Pharmaceutics; Ghent University; Ottergemsesteenweg 460 9000 Ghent Belgium
| | - Misbah Aslam
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Alexander Brose
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Friedrich Foerster
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
- Department of Medicine I (Gastroenterology Hepatology, and Nephrology); University Medical Center of Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Sebastian Rosigkeit
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Patricia Renz
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | - Rosario Heck
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy (FZI); University Medical Center of the Johannes Gutenberg-University Mainz; Langenbeckstraße 1 55101 Mainz Germany
- Division of Gastroenterology; Beth Israel Deaconess Medical Center; Harvard Medical School; 330 Brookline Avenue Boston MA 02215 USA
| | - Rudolf Zentel
- Institute of Organic Chemistry; Johannes Gutenberg-University Mainz; Duesbergweg 10-14 55128 Mainz Germany
| |
Collapse
|
34
|
Zulliger R, Conley SM, Naash MI. Non-viral therapeutic approaches to ocular diseases: An overview and future directions. J Control Release 2015; 219:471-487. [PMID: 26439665 PMCID: PMC4699668 DOI: 10.1016/j.jconrel.2015.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 12/31/2022]
Abstract
Currently there are no viable treatment options for patients with debilitating inherited retinal degeneration. The vast variability in disease-inducing mutations and resulting phenotypes has hampered the development of therapeutic interventions. Gene therapy is a logical approach, and recent work has focused on ways to optimize vector design and packaging to promote optimized expression and phenotypic rescue after intraocular delivery. In this review, we discuss ongoing ocular clinical trials, which currently use viral gene delivery, but focus primarily on new advancements in optimizing the efficacy of non-viral gene delivery for ocular diseases. Non-viral delivery systems are highly customizable, allowing functionalization to improve cellular and nuclear uptake, bypassing cellular degradative machinery, and improving gene expression in the nucleus. Non-viral vectors often yield transgene expression levels lower than viral counterparts, however their favorable safety/immune profiles and large DNA capacity (critical for the delivery of large ocular disease genes) make their further development a research priority. Recent work on particle coating and vector engineering presents exciting ways to overcome limitations of transient/low gene expression levels, but also highlights the fact that further refinements are needed before use in the clinic.
Collapse
Affiliation(s)
- Rahel Zulliger
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States.
| |
Collapse
|
35
|
Oliveira ACN, Raemdonck K, Martens T, Rombouts K, Simón-Vázquez R, Botelho C, Lopes I, Lúcio M, González-Fernández Á, Real Oliveira MECD, Gomes AC, Braeckmans K. Stealth monoolein-based nanocarriers for delivery of siRNA to cancer cells. Acta Biomater 2015. [PMID: 26225736 DOI: 10.1016/j.actbio.2015.07.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
While the delivery of small interfering RNAs (siRNAs) is an attractive strategy to treat several clinical conditions, siRNA-nanocarriers' stability after intravenous administration is still a major obstacle for the development of RNA-interference based therapies. But, although the need for stability is well recognized, the notion that strong stabilization can decrease nanocarriers' efficiency is sometimes neglected. In this work we evaluated two stealth functionalization strategies to stabilize the previously validated dioctadecyldimethylammonium bromide (DODAB):monoolein (MO) siRNA-lipoplexes. The nanocarriers were pre- and post-pegylated, forming vectors with different stabilities in biological fluids. The stealth nanocarriers' behavior was tested under biological mimetic conditions, as the production of stable siRNA-lipoplexes is determinant to achieve efficient intravenous siRNA delivery to cancer cells. Upon incubation in human serum for 2h, by fluorescence Single Particle Tracking microscopy, PEG-coated lipoplexes were found to have better colloidal stability as they could maintain a relatively stable size. In addition, using fluorescence fluctuation spectroscopy, post-pegylation also proved to avoid siRNA dissociation from the nanocarriers in human serum. Concomitantly it was found that PEG-coated lipoplexes improved cellular uptake and transfection efficiency in H1299 cells, and had the ability to silence BCR-ABL, affecting the survival of K562 cells. Based on an efficient cellular internalization, good silencing effect, good siRNA retention and good colloidal stability in human serum, DODAB:MO (2:1) siRNA-lipoplexes coated with PEG-Cer are considered promising nanocarriers for further in vivo validation. STATEMENT OF SIGNIFICANCE This work describes two stealth functionalization strategies for the stabilization of the previously validated dioctadecyldimethylammonium bromide (DODAB):monoolein (MO) siRNA-lipoplexes. These nanocarriers are capable of efficiently incorporating and delivering siRNA molecules to cells in order to silence genes whose expression is implicated in a pathological condition. The main objective was to functionalize these nanocarriers with a coating conferring protection to siRNA in blood without compromising its efficient delivery to cancer cells, validating the potential of DODAB:MO (2:1) siRNA-lipoplexes as therapeutic vectors. We show that the stealth strategy is determinant to achieve a stable and efficient nanocarrier, and that DODAB:MO mixtures have a very promising potential for systemic siRNA delivery to leukemic cells.
Collapse
Affiliation(s)
- Ana C N Oliveira
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; Centre of Physics, Department of Physics, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Thomas Martens
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium; Center for Nano- and Biophotonics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Koen Rombouts
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Rosana Simón-Vázquez
- Immunology, Biomedical Research Center (CINBIO) and Institute of Biomedical Research of Vigo (IBIV), University of Vigo, Campus Lagoas Marcosende, 36310 Vigo, Pontevedra, Spain
| | - Cláudia Botelho
- Centre of Biological Engineering (CEB), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Ivo Lopes
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; Centre of Physics, Department of Physics, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Marlene Lúcio
- Centre of Physics, Department of Physics, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - África González-Fernández
- Immunology, Biomedical Research Center (CINBIO) and Institute of Biomedical Research of Vigo (IBIV), University of Vigo, Campus Lagoas Marcosende, 36310 Vigo, Pontevedra, Spain
| | | | - Andreia C Gomes
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium; Center for Nano- and Biophotonics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| |
Collapse
|
36
|
Moscovici M. Present and future medical applications of microbial exopolysaccharides. Front Microbiol 2015; 6:1012. [PMID: 26483763 PMCID: PMC4586455 DOI: 10.3389/fmicb.2015.01012] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 09/07/2015] [Indexed: 11/13/2022] Open
Abstract
Microbial exopolysaccharides (EPS) have found outstanding medical applications since the mid-20th century, with the first clinical trials on dextran solutions as plasma expanders. Other EPS entered medicine firstly as conventional pharmaceutical excipients (e.g., xanthan - as suspension stabilizer, or pullulan - in capsules and oral care products). Polysaccharides, initially obtained from plant or animal sources, became easily available for a wide range of applications, especially when they were commercially produced by microbial fermentation. Alginates are used as anti-reflux, dental impressions, or as matrix for tablets. Hyaluronic acid and derivatives are used in surgery, arthritis treatment, or wound healing. Bacterial cellulose is applied in wound dressings or scaffolds for tissue engineering. The development of drug controlled-release systems and of micro- and nanoparticulated ones, has opened a new era of medical applications for biopolymers. EPS and their derivatives are well-suited potentially non-toxic, biodegradable drug carriers. Such systems concern rating and targeting of controlled release. Their large area of applications is explained by the available manifold series of derivatives, whose useful properties can be thereby controlled. From matrix inclusion to conjugates, different systems have been designed to solubilize, and to assure stable transport in the body, target accumulation and variable rate-release of a drug substance. From controlled drug delivery, EPS potential applications expanded to vaccine adjuvants and diagnostic imaging systems. Other potential applications are related to the bioactive (immunomodulator, antitumor, antiviral) characteristics of EPS. The numerous potential applications still wait to be developed into commercial pharmaceuticals and medical devices. Based on previous and recent results in important medical-pharmaceutical domains, one can undoubtedly state that EPS medical applications have a broad future ahead.
Collapse
Affiliation(s)
- Misu Moscovici
- National Institute for Chemical Pharmaceutical Research and Development, BucharestRomania
| |
Collapse
|
37
|
De Backer L, Cerrada A, Pérez-Gil J, De Smedt SC, Raemdonck K. Bio-inspired materials in drug delivery: Exploring the role of pulmonary surfactant in siRNA inhalation therapy. J Control Release 2015; 220:642-50. [PMID: 26363301 DOI: 10.1016/j.jconrel.2015.09.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 01/13/2023]
Abstract
Many pathologies of the respiratory tract are inadequately treated with existing small molecule-based therapies. The emergence of RNA interference (RNAi) enables the post-transcriptional silencing of key molecular disease factors that cannot readily be targeted with conventional small molecule drugs. Pulmonary administration of RNAi effectors, such as small interfering RNA (siRNA), allows direct delivery into the lung tissue, hence reducing systemic exposure. Unfortunately, the clinical translation of RNAi is severely hampered by inefficient delivery of siRNA therapeutics towards the cytoplasm of the target cells. In order to have a better control of the siRNA delivery process, both extra- and intracellular, siRNAs are typically formulated in nanosized delivery vehicles (nanoparticles, NPs). In the lower airways, which are the targeted sites of action for multiple pulmonary disorders, these siRNA-loaded NPs will encounter the pulmonary surfactant (PS) layer, covering the entire alveolar surface. The interaction between the instilled siRNA-loaded NPs and the PS at this nano-bio interface results in the adsorption of PS components onto the surface of the NPs. The formation of this so-called biomolecular corona conceals the original NP surface and will therefore profoundly determine the biological efficacy of the NP. Though this interplay has initially been regarded as a barrier towards efficient siRNA delivery to the respiratory target cell, recent reports have illustrated that the interaction with PS might also be beneficial for local pulmonary siRNA delivery.
Collapse
Affiliation(s)
- Lynn De Backer
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Alejandro Cerrada
- Departamento de Bioquimica y Biologia Molecular, Facultad de Biologia, and Research Institute Hospital 12 Octubre, Universidad Complutense, José Antonio Novais 2, 28040 Madrid, Spain.
| | - Jesús Pérez-Gil
- Departamento de Bioquimica y Biologia Molecular, Facultad de Biologia, and Research Institute Hospital 12 Octubre, Universidad Complutense, José Antonio Novais 2, 28040 Madrid, Spain.
| | - Stefaan C De Smedt
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
38
|
Li Y, Maciel D, Rodrigues J, Shi X, Tomás H. Biodegradable Polymer Nanogels for Drug/Nucleic Acid Delivery. Chem Rev 2015; 115:8564-608. [PMID: 26259712 DOI: 10.1021/cr500131f] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yulin Li
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, East China University of Science and Technology , Shanghai 200237, People's Republic of China
| | - Dina Maciel
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| | - João Rodrigues
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| | - Xiangyang Shi
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University , Shanghai 201620, People's Republic of China
| | - Helena Tomás
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| |
Collapse
|
39
|
Abstract
Among the several delivery materials available so far, polysaccharides represent very attractive molecules as they can undergo a wide range of chemical modifications, are biocompatible, biodegradable, and have low immunogenic properties. Thus, polysaccharides can contribute to significantly overcome the limitation in the use of many types of drugs, including anti-cancer drugs. The use of conventional anti-cancer drugs is hampered by their high toxicity, mostly depending on the indiscriminate targeting of both cancer and normal cells. Additionally, for nucleic acid based drugs (NABDs), an emerging class of drugs with potential anti-cancer value, the practical use is problematic. This mostly depends on their fast degradation in biological fluids and the difficulties to cross cell membranes. Thus, for both classes of drugs, the development of optimal delivery materials is crucial. Here we discuss the possibility of using different kinds of polysaccharides, such as chitosan, hyaluronic acid, dextran, and pullulan, as smart drug delivery materials. We first describe the main features of polysaccharides, then a general overview about the aspects ruling drug release mechanisms and the pharmacokinetic are reported. Finally, notable examples of polysaccharide-based delivery of conventional anti-cancer drugs and NABDs are reported. Whereas additional research is required, the promising results obtained so far, fully justify further efforts, both in terms of economic support and investigations in the field of polysaccharides as drug delivery materials.
Collapse
|
40
|
Hill MC, Nguyen MK, Jeon O, Alsberg E. Spatial control of cell gene expression by siRNA gradients in biodegradable hydrogels. Adv Healthc Mater 2015; 4:714-22. [PMID: 25530099 PMCID: PMC4406766 DOI: 10.1002/adhm.201400458] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/22/2014] [Indexed: 11/10/2022]
Abstract
The extracellular environment exposes cells to numerous biochemical and physical signals that regulate their behavior. Strategies for generating continuous gradients of signals in biomaterials may allow for spatial control and patterning of cell behavior, and ultimately aid in the engineering of complex tissues. Short interfering RNA (siRNA) can regulate gene expression by silencing specific mRNA molecules post-transcriptionally, which may be valuable when presented in a continuous gradient for regenerative or therapeutic applications. Here, a biodegradable hydrogel system containing a gradient of siRNA is presented, and its capacity to regulate protein expression of encapsulated cells in a spatially continuous manner is demonstrated. Photocross-linkable dextran hydrogels containing a gradient of siRNA have been successfully fabricated using a dual-programmable syringe pump system, and differential gene silencing in incorporated cells that is sustained over time has been shown using green fluorescent protein as a reporter. This platform technology may be applied in tissue engineering to spatially control biologically relevant cellular processes.
Collapse
Affiliation(s)
- Michael C. Hill
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| | - Minh K. Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| | - Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA. Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA. National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| |
Collapse
|
41
|
Nuhn L, Tomcin S, Miyata K, Mailänder V, Landfester K, Kataoka K, Zentel R. Size-Dependent Knockdown Potential of siRNA-Loaded Cationic Nanohydrogel Particles. Biomacromolecules 2014; 15:4111-21. [DOI: 10.1021/bm501148y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lutz Nuhn
- Institute
of Organic Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, D-55099 Mainz, Germany
| | - Stephanie Tomcin
- Max-Planck-Institute
for Polymer Research, Ackermannweg
10, D-55128 Mainz, Germany
| | | | - Volker Mailänder
- Max-Planck-Institute
for Polymer Research, Ackermannweg
10, D-55128 Mainz, Germany
- III.
Medical Clinic (Hematology, Oncology and Pneumology), University Medical Center of the Johannes-Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Katharina Landfester
- Max-Planck-Institute
for Polymer Research, Ackermannweg
10, D-55128 Mainz, Germany
| | | | - Rudolf Zentel
- Institute
of Organic Chemistry, Johannes Gutenberg-University, Duesbergweg 10-14, D-55099 Mainz, Germany
| |
Collapse
|
42
|
PEG — A versatile conjugating ligand for drugs and drug delivery systems. J Control Release 2014; 192:67-81. [DOI: 10.1016/j.jconrel.2014.06.046] [Citation(s) in RCA: 415] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/23/2014] [Accepted: 06/24/2014] [Indexed: 01/07/2023]
|
43
|
Oliveira ACN, Martens TF, Raemdonck K, Adati RD, Feitosa E, Botelho C, Gomes AC, Braeckmans K, Real Oliveira MECD. Dioctadecyldimethylammonium:monoolein nanocarriers for efficient in vitro gene silencing. ACS APPLIED MATERIALS & INTERFACES 2014; 6:6977-6989. [PMID: 24712543 DOI: 10.1021/am500793y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
This study describes a novel liposomal formulation for siRNA delivery, based on the mixture of the neutral lipid monoolein (MO) and cationic lipids of the dioctadecyldimethylammonium (DODA) family. The cationic lipids dioctadecyldimethylammonium bromide (DODAB) and chloride (DODAC) were compared in order to identify which one will most efficiently induce gene silencing. MO has a fluidizing effect on DODAC and DODAB liposomes, although it was more homogeneously distributed in DODAC bilayers. All MO-based liposomal formulations were able to efficiently encapsulate siRNA. Stable lipoplexes of small size (100-160 nm) with a positive surface charge (>+45 mV) were formed. A more uniform MO incorporation in DODAC:MO may explain an increase of the fusogenic potential of these liposomes. The siRNA-lipoplexes were readily internalized by human nonsmall cell lung carcinoma (H1299) cells, in an energy dependent process. DODAB:MO nanocarriers showed a higher internalization efficiency in comparison to DODAC:MO lipoplexes, and were also more efficient in promoting gene silencing. MO had a similar gene silencing ability as the commonly used helper lipid 1,2-dioleyl-3-phosphatidylethanolamine (DOPE), but with much lower cytotoxicity. Taking in consideration all the results presented, DODAB:MO liposomes are the most promising tested formulation for systemic siRNA delivery.
Collapse
Affiliation(s)
- Ana Cristina Norberto Oliveira
- CBMA (Center of Molecular and Environmental Biology), Department of Biology and ‡CFUM (Center of Physics), Department of Physics, University of Minho , Campus of Gualtar, 4710-057 Braga, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sagnella SM, Duong H, MacMillan A, Boyer C, Whan R, McCarroll JA, Davis TP, Kavallaris M. Dextran-Based Doxorubicin Nanocarriers with Improved Tumor Penetration. Biomacromolecules 2013; 15:262-75. [DOI: 10.1021/bm401526d] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Sharon M. Sagnella
- Children’s
Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, P.O. Box 81, Randwick, Australia
| | | | | | | | | | - Joshua A. McCarroll
- Children’s
Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, P.O. Box 81, Randwick, Australia
| | - Thomas P. Davis
- Monash
Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, Australia
- Department
of Chemistry, University of Warwick, Coventry, United Kingdom
| | - Maria Kavallaris
- Children’s
Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, P.O. Box 81, Randwick, Australia
| |
Collapse
|
45
|
Raemdonck K, Martens TF, Braeckmans K, Demeester J, De Smedt SC. Polysaccharide-based nucleic acid nanoformulations. Adv Drug Deliv Rev 2013; 65:1123-47. [PMID: 23680381 DOI: 10.1016/j.addr.2013.05.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 04/24/2013] [Accepted: 05/03/2013] [Indexed: 12/24/2022]
Abstract
Therapeutic application of nucleic acids requires their encapsulation in nanosized carriers that enable safe and efficient intracellular delivery. Before the desired site of action is reached, drug-loaded nanoparticles (nanomedicines) encounter numerous extra- and intracellular barriers. Judicious nanocarrier design is highly needed to stimulate nucleic acid delivery across these barriers and maximize the therapeutic benefit. Natural polysaccharides are widely used for biomedical and pharmaceutical applications due to their inherent biocompatibility. At present, there is a growing interest in applying these biopolymers for the development of nanomedicines. This review highlights various polysaccharides and their derivatives, currently employed in the design of nucleic acid nanocarriers. In particular, recent progress made in polysaccharide-assisted nucleic acid delivery is summarized and the specific benefits that polysaccharides might offer to improve the delivery process are critically discussed.
Collapse
|
46
|
Nishimura Y, Mieda H, Ishii J, Ogino C, Fujiwara T, Kondo A. Targeting cancer cell-specific RNA interference by siRNA delivery using a complex carrier of affibody-displaying bio-nanocapsules and liposomes. J Nanobiotechnology 2013; 11:19. [PMID: 23800313 PMCID: PMC3699380 DOI: 10.1186/1477-3155-11-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/18/2013] [Indexed: 12/12/2022] Open
Abstract
Background Small interfering RNA (siRNA) has attracted attention in the field of nucleic acid medicine as a RNA interference (RNAi) application that leads to gene silencing due to specific messenger RNA (mRNA) destruction. However, since siRNA is unstable in blood and unable to cross the cell membrane, encapsulation of siRNA into a carrier is required. Results In this study, we used a carrier that combined ZHER2-displaying bio-nanocapsule (derived from hepatitis B virus surface antigen) and liposomes in a complex in order to investigate the feasibility of effective and target-cell-specific RNAi applications. As a result, by observing RNAi only in HER2-expressing breast cancer cells, using our proposed methodology, we successfully demonstrated target-cell-specific delivery and effective function expression of siRNA. Conclusions These findings show that, in the field of nucleic acid medicine, ZHER2-BNC/LP can be a useful carrier for siRNA delivery, and could also become a useful tool for gene silencing and to accomplish protein knock-down.
Collapse
Affiliation(s)
- Yuya Nishimura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- Smriti Singh
- DWI an der RWTH Aachen e.V. Functional and Interactive Polymers and Institute for Technical and Macromolecular Chemistry, RWTH Aachen University; 52056 Aachen Germany
| | - Martin Möller
- DWI an der RWTH Aachen e.V. Functional and Interactive Polymers and Institute for Technical and Macromolecular Chemistry, RWTH Aachen University; 52056 Aachen Germany
| | - Andrij Pich
- DWI an der RWTH Aachen e.V. Functional and Interactive Polymers and Institute for Technical and Macromolecular Chemistry, RWTH Aachen University; 52056 Aachen Germany
| |
Collapse
|
48
|
Ahmed M, Wattanaarsakit P, Narain R. Cationic glyco-nanogels for epidermal growth factor receptor (EGFR) specific siRNA delivery in ovarian cancer cells. Polym Chem 2013. [DOI: 10.1039/c3py00425b] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
49
|
Fonseca AC, Ferreira P, Cordeiro RA, Mendonça PV, Góis JR, Gil MH, Coelho JFJ. Drug Delivery Systems for Predictive Medicine: Polymers as Tools for Advanced Applications. NEW STRATEGIES TO ADVANCE PRE/DIABETES CARE: INTEGRATIVE APPROACH BY PPPM 2013. [DOI: 10.1007/978-94-007-5971-8_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
50
|
Naeye B, Deschout H, Caveliers V, Descamps B, Braeckmans K, Vanhove C, Demeester J, Lahoutte T, De Smedt SC, Raemdonck K. In vivo disassembly of IV administered siRNA matrix nanoparticles at the renal filtration barrier. Biomaterials 2012; 34:2350-8. [PMID: 23261216 DOI: 10.1016/j.biomaterials.2012.11.058] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 11/29/2012] [Indexed: 01/24/2023]
Abstract
Intravenous administration of siRNA nanocarriers may provide unique therapeutic opportunities for tissue-specific gene silencing. Although often engineered to overcome the numerous barriers that exist in the systemic circulation, many nanocarriers fail in extending the circulation time of the siRNA. A more detailed assessment of the different clearance mechanisms that are in play after intravenous injection could therefore be of value to improve siRNA nanocarrier design. In this report, the biodistribution in mice of siRNA loaded dextran nanogels was investigated in detail. Both single photon emission computed tomography (SPECT) imaging and fluorescence single particle tracking (fSPT) indicate that the particles are rapidly cleared from the circulation. PEGylation of the nanogels was not able to increase the half-life in the bloodstream. Carrier disassembly in the systemic circulation and phagocytic clearance are known to facilitate the elimination of siRNA nanoparticles. Additionally, it is demonstrated for dextran nanogels that also the kidneys play an important role in their elimination from the bloodstream. SPECT imaging revealed an accumulation of the siRNA loaded dextran nanogels in the kidneys shortly after intravenous injection and a significantly delayed transition of siRNA from kidney to bladder, as opposed to the injection of free siRNA. These data indicate that components of the glomerular filtration barrier may contribute to the dissociation of siRNA from its carrier, as was recently suggested for cationic cyclodextrin siRNA polyplexes. This clearance mechanism should therefore be taken into account when designing siRNA nanocarriers for intravenous administration.
Collapse
Affiliation(s)
- Broes Naeye
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|