101
|
Garland KM, Sheehy TL, Wilson JT. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem Rev 2022; 122:5977-6039. [PMID: 35107989 PMCID: PMC8994686 DOI: 10.1021/acs.chemrev.1c00750] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The stimulator of interferon genes (STING) cellular signaling pathway is a promising target for cancer immunotherapy. Activation of the intracellular STING protein triggers the production of a multifaceted array of immunostimulatory molecules, which, in the proper context, can drive dendritic cell maturation, antitumor macrophage polarization, T cell priming and activation, natural killer cell activation, vascular reprogramming, and/or cancer cell death, resulting in immune-mediated tumor elimination and generation of antitumor immune memory. Accordingly, there is a significant amount of ongoing preclinical and clinical research toward further understanding the role of the STING pathway in cancer immune surveillance as well as the development of modulators of the pathway as a strategy to stimulate antitumor immunity. Yet, the efficacy of STING pathway agonists is limited by many drug delivery and pharmacological challenges. Depending on the class of STING agonist and the desired administration route, these may include poor drug stability, immunocellular toxicity, immune-related adverse events, limited tumor or lymph node targeting and/or retention, low cellular uptake and intracellular delivery, and a complex dependence on the magnitude and kinetics of STING signaling. This review provides a concise summary of the STING pathway, highlighting recent biological developments, immunological consequences, and implications for drug delivery. This review also offers a critical analysis of an expanding arsenal of chemical strategies that are being employed to enhance the efficacy, safety, and/or clinical utility of STING pathway agonists and lastly draws attention to several opportunities for therapeutic advancements.
Collapse
Affiliation(s)
- Kyle M Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - Taylor L Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
| |
Collapse
|
102
|
Thambi T, Lee J, Yoon AR, Kasala D, Yun CO. A pH- and Bioreducible Cationic Copolymer with Amino Acids and Piperazines for Adenovirus Delivery. Pharmaceutics 2022; 14:pharmaceutics14030597. [PMID: 35335972 PMCID: PMC8950541 DOI: 10.3390/pharmaceutics14030597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022] Open
Abstract
Adenoviruses (Ads) are attractive nonviral vectors and show great potential in cancer gene therapy. However, inherent properties of Ads, including immunogenicity, nonspecific toxicity, and coxsackie and adenovirus receptor (CAR)-dependent cell uptake, limit their clinical use. To surmount these issues, we developed a pH- and glutathione-responsive poly(ethylene glycol)-poly(ꞵ-aminoester)-polyethyleneimine (PPA) for conjugation with Ad. The pH sensitivity of the PPA copolymer was elegantly tuned by substitution with different amino acids (arginine, histidine, and tryptophan), piperazines (Pip1, Pip2, and Pip3), and guanidine residues in the backbone of the PPA conjugate. PPA copolymer was further functionalized with short-chain cross-linker succinimidyl 3-(2-pyridyldithio)propionate) (SPDP) to obtain PPA-SPDP for facile conjugation with Ad. The PPA-conjugated Ad (PPA-Ad) conjugate was obtained by reacting PPA-SPDP conjugate with thiolated Ad (Ad-SH). Ad-SH was prepared by reacting Ad with 2-iminothiolane. The size distribution and zeta potential results of PPA-Ad conjugate showed an increasing trend with an increase in copolymer dose. From in vitro test, it was found that the transduction efficiency of PPA-Ad conjugate in CAR-positive cells (A549 and H460 cells) was remarkably increased at the acidic pH condition (pH 6.2) when compared with PPA-Ad conjugate incubated under the physiological condition (pH 7.4). Interestingly, the increase in transduction efficiency was evidenced in CAR-negative cells (MDA-MB-231 and T24 cells). These results demonstrated that biocompatible and biodegradable PPA copolymers can efficiently cover the surface of Ad and can increase the transduction efficiency, and hence PPA copolymers can be a useful nanomaterial for viral vector delivery in cancer therapy.
Collapse
Affiliation(s)
- Thavasyappan Thambi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea; (T.T.); (J.L.); (A.-R.Y.); (D.K.)
| | - Jeongmin Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea; (T.T.); (J.L.); (A.-R.Y.); (D.K.)
- GeneMedicine Co., Ltd., 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea; (T.T.); (J.L.); (A.-R.Y.); (D.K.)
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, Korea
| | - Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea; (T.T.); (J.L.); (A.-R.Y.); (D.K.)
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea; (T.T.); (J.L.); (A.-R.Y.); (D.K.)
- GeneMedicine Co., Ltd., 222 Wangsimni-ro Seongdong-gu, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, Korea
- Institute of Nano Science and Technology (INST), Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
- Correspondence: ; Tel.: +82-2-2220-0491; Fax: +82-2-2220-4850
| |
Collapse
|
103
|
Haegebaert RM, Kempers M, Ceelen W, Lentacker I, Remaut K. Nanoparticle mediated targeting of toll-like receptors to treat colorectal cancer. Eur J Pharm Biopharm 2022; 172:16-30. [PMID: 35074555 DOI: 10.1016/j.ejpb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
104
|
Nanoparticle-based delivery strategies of multifaceted immunomodulatory RNA for cancer immunotherapy. J Control Release 2022; 343:564-583. [DOI: 10.1016/j.jconrel.2022.01.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022]
|
105
|
Li M, Li S, Li Y, Li X, Yang G, Li M, Xie Y, Su W, Wu J, Jia L, Li S, Ma W, Li H, Guo N, Yu P. Cationic liposomes co-deliver chemotherapeutics and siRNA for the treatment of breast cancer. Eur J Med Chem 2022; 233:114198. [DOI: 10.1016/j.ejmech.2022.114198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/26/2022]
|
106
|
A Comprehensive Review on Novel Liposomal Methodologies, Commercial Formulations, Clinical Trials and Patents. BIONANOSCIENCE 2022; 12:274-291. [PMID: 35096502 PMCID: PMC8790012 DOI: 10.1007/s12668-022-00941-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Liposomes are well-recognized and essential nano-sized drug delivery systems. Liposomes are phospholipid vesicles comprised of cell membrane components and have been employed as artificial cell models to mimic structure and functions of cells and are of immense use in various biological analyses. Liposomes acquire great advantages and provide wide range of applications as useful drug carriers in pre-clinical and clinical trials. This review summarizes exclusively on scalable techniques for liposome preparation and focuses on the strengths and limitations with respect to industrial applicability. Also, this review discusses the updated recent advancements in biomedical applications with a mention of key highlights of commercially available formulations, clinical trials and patents in recent past. Furthermore, this review also provides brief information of the classification, composition and characterization of liposomes.
Collapse
|
107
|
Zorko M, Jones S, Langel Ü. Cell-penetrating peptides in protein mimicry and cancer therapeutics. Adv Drug Deliv Rev 2022; 180:114044. [PMID: 34774552 DOI: 10.1016/j.addr.2021.114044] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022]
Abstract
Extensive research has been undertaken in the pursuit of anticancer therapeutics. Many anticancer drugs require specificity of delivery to cancer cells, whilst sparing healthy tissue. Cell-penetrating peptides (CPPs), now well established as facilitators of intracellular delivery, have in recent years advanced to incorporate target specificity and thus possess great potential for the targeted delivery of anticancer cargoes. Though none have yet been approved for clinical use, this novel technology has already entered clinical trials. In this review we present CPPs, discuss their classification, mechanisms of cargo internalization and highlight strategies for conjugation to anticancer moieties including their incorporation into therapeutic proteins. As the mainstay of this review, strategies to build specificity into tumor targeting CPP constructs through exploitation of the tumor microenvironment and the use of tumor homing peptides are discussed, whilst acknowledging the extensive contribution made by CPP constructs to target specific protein-protein interactions integral to intracellular signaling pathways associated with tumor cell survival and progression. Finally, antibody/antigen CPP conjugates and their potential roles in cancer immunotherapy and diagnostics are considered. In summary, this review aims to harness the potential of CPP-aided drug delivery for future cancer therapies and diagnostics whilst highlighting some of the most recent achievements in selective delivery of anticancer drugs, including cytostatic drugs, to a range of tumor cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Matjaž Zorko
- University of Ljubljana, Medical Faculty, Institute of Biochemistry and Molecular Genetics, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Sarah Jones
- University of Wolverhampton, School of Pharmacy, Faculty of Science & Engineering, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Ülo Langel
- University of Stockholm, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16, 106 91 Stockholm, Sweden; Institute of Technology, University of Tartu, Nooruse 1, Tartu, Estonia 50411, Estonia.
| |
Collapse
|
108
|
Singh P, Muhammad I, Nelson NE, Tran KTM, Vinikoor T, Chorsi MT, D’Orio E, Nguyen TD. Transdermal delivery for gene therapy. Drug Deliv Transl Res 2022; 12:2613-2633. [PMID: 35538189 PMCID: PMC9089295 DOI: 10.1007/s13346-022-01138-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
Gene therapy is a critical constituent of treatment approaches for genetic diseases and has gained tremendous attention. Treating and preventing diseases at the genetic level using genetic materials such as DNA or RNAs could be a new avenue in medicine. However, delivering genes is always a challenge as these molecules are sensitive to various enzymes inside the body, often produce systemic toxicity, and suffer from off-targeting problems. In this regard, transdermal delivery has emerged as an appealing approach to enable a high efficiency and low toxicity of genetic medicines. This review systematically summarizes outstanding transdermal gene delivery methods for applications in skin cancer treatment, vaccination, wound healing, and other therapies.
Collapse
Affiliation(s)
- Parbeen Singh
- Department of Mechanical Engineering, University of Connecticut, Storrs, USA
| | - I’jaaz Muhammad
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Nicole E. Nelson
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Khanh T. M. Tran
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Tra Vinikoor
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Meysam T. Chorsi
- Department of Mechanical Engineering, University of Connecticut, Storrs, USA ,Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Ethan D’Orio
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA ,Department of Biomedical Engineering and Department of Advanced Manufacturing for Energy Systems, Storrs, USA
| | - Thanh D. Nguyen
- Department of Mechanical Engineering, University of Connecticut, Storrs, USA ,Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| |
Collapse
|
109
|
Fobian SF, Cheng Z, ten Hagen TLM. Smart Lipid-Based Nanosystems for Therapeutic Immune Induction against Cancers: Perspectives and Outlooks. Pharmaceutics 2021; 14:26. [PMID: 35056922 PMCID: PMC8779430 DOI: 10.3390/pharmaceutics14010026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, a promising and widely applied mode of oncotherapy, makes use of immune stimulants and modulators to overcome the immune dysregulation present in cancer, and leverage the host's immune capacity to eliminate tumors. Although some success has been seen in this field, toxicity and weak immune induction remain challenges. Liposomal nanosystems, previously used as targeting agents, are increasingly functioning as immunotherapeutic vehicles, with potential for delivery of contents, immune induction, and synergistic drug packaging. These systems are tailorable, multifunctional, and smart. Liposomes may deliver various immune reagents including cytokines, specific T-cell receptors, antibody fragments, and immune checkpoint inhibitors, and also present a promising platform upon which personalized medicine approaches can be built, especially with preclinical and clinical potentials of liposomes often being frustrated by inter- and intrapatient variation. In this review, we show the potential of liposomes in cancer immunotherapy, as well as the methods for synthesis and in vivo progression thereof. Both preclinical and clinical studies are included to comprehensively illuminate prospects and challenges for future research and application.
Collapse
Affiliation(s)
| | | | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology (LEO), Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (S.-F.F.); (Z.C.)
| |
Collapse
|
110
|
Chavda VP, Pandya R, Apostolopoulos V. DNA vaccines for SARS-CoV-2: toward third-generation vaccination era. Expert Rev Vaccines 2021; 20:1549-1560. [PMID: 34582298 PMCID: PMC8567274 DOI: 10.1080/14760584.2021.1987223] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/24/2021] [Indexed: 12/19/2022]
Abstract
Introduction: Coronavirus outbreak 2019 (COVID-19) has affected all the corners of the globe and created chaos to human life. In order to put some control on the pandemic, vaccines are urgently required that are safe, cost effective, easy to produce, and most importantly induce appropriate immune responses and protection against viral infection. DNA vaccines possess all these features and are promising candidates for providing protection against SARS-CoV-2.Area covered: Current understanding and advances in DNA vaccines toward COVID-19, especially those under various stages of clinical trials.Expert opinion: Through DNA vaccines, host cells are momentarily transformed into factories that produce proteins of the SARS-CoV-2. The host immune system detects these proteins to develop antibodies that neutralize and prevent the infection. This vaccine platform has additional benefits compared to traditional vaccination strategies like strong cellular immune response, higher safety margin, a simple production process as per cGMP norms, lack of any infectious agent, and a robust platform for large-scale production.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, Gujarat, India
| | - Radhika Pandya
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, Gujarat, India
| | - Vasso Apostolopoulos
- Department of Immunology, Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| |
Collapse
|
111
|
Magnetofection In Vivo by Nanomagnetic Carriers Systemically Administered into the Bloodstream. Pharmaceutics 2021; 13:pharmaceutics13111927. [PMID: 34834342 PMCID: PMC8619128 DOI: 10.3390/pharmaceutics13111927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Nanoparticle-based technologies are rapidly expanding into many areas of biomedicine and molecular science. The unique ability of magnetic nanoparticles to respond to the magnetic field makes them especially attractive for a number of in vivo applications including magnetofection. The magnetofection principle consists of the accumulation and retention of magnetic nanoparticles carrying nucleic acids in the area of magnetic field application. The method is highly promising as a clinically efficient tool for gene delivery in vivo. However, the data on in vivo magnetofection are often only descriptive or poorly studied, insufficiently systematized, and sometimes even contradictory. Therefore, the aim of the review was to systematize and analyze the data that influence the in vivo magnetofection processes after the systemic injection of magnetic nanostructures. The main emphasis is placed on the structure and coating of the nanomagnetic vectors. The present problems and future trends of the method development are also considered.
Collapse
|
112
|
Rahiman N, Zamani P, Badiee A, Arabi L, Alavizadeh SH, Jaafari MR. An insight into the role of liposomal therapeutics in the reversion of Multiple Sclerosis. Expert Opin Drug Deliv 2021; 18:1795-1813. [PMID: 34747298 DOI: 10.1080/17425247.2021.2003327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Multiple Sclerosis (MS), as an autoimmune disease, has complicated immunopathology, which makes its management relevant to various factors. Novel pharmaceutical vehicles, especially liposomes, can support efficacious handling of this disease both in early detection and prognosis and also in a therapeutic manner. The most well-known trigger of MS onset is the predominance of cellular to humoral immunity and enhancement of inflammatory cytokines level. The installation of liposomes as nanoparticles to control this disease holds great promise up to now. AREAS COVERED Various types of liposomes with different properties and purposes have been formulated and targeted immune cells with their surface manipulations. They may be encapsulated with anti-inflammatory, MS-related therapeutics, or immunodominant myelin-specific peptides for attaining a higher therapeutic efficacy of the drugs or tolerance induction. Cationic liposomes are also highly applicable for gene delivery of the anti-inflammatory cytokines or silencing the inflammatory cytokines. Liposomes have also been used as biotools for comprehending MS pathomechanisms or as diagnostic agents. EXPERT OPINION The efforts to manage MS through nanomedicine, especially liposomal therapeutics, pave a new avenue to a high-throughput medication of this autoimmune disease and their translation to the clinic in the future for overcoming the challenges that MS patients confront.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
113
|
Maiti B, Bhattacharya S. Liposomal nanoparticles based on steroids and isoprenoids for nonviral gene delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1759. [PMID: 34729941 DOI: 10.1002/wnan.1759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/24/2021] [Accepted: 08/10/2021] [Indexed: 11/11/2022]
Abstract
Natural lipid molecules are an essential part of life as they constitute the membrane of cells and organelle. In most of these cases, the hydrophobicity of natural lipids is contributed by alkyl chains. Although natural lipids with a nonfatty acid hydrophobic backbone are quite rare, steroids and isoprenoids have been strong candidates as part of a lipid. Over the years, these natural molecules (steroid and isoprenoids) have been used to make either lipid-based nanoparticle or functionalize in such a way that it could form nano assembly alone for therapeutic delivery. Here we mainly focus on the synthetic functionalized version of these natural molecules which forms cationic liposomal nanoparticles (LipoNPs). These cationic LipoNPs were further used to deliver various negatively charged genetic materials in the form of pDNA, siRNA, mRNA (nucleic acids), and so on. This article is categorized under: Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Bappa Maiti
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata, India
| | - Santanu Bhattacharya
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata, India.,School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, India.,Department of Organic Chemistry, Indian Institute of Science, Bangalore, India
| |
Collapse
|
114
|
Abstract
INTRODUCTION Vaccination is so far the most effective way of eradicating infections. Rapidly emerging drug resistance against infectious diseases and chemotherapy-related toxicities in cancer warrant immediate vaccine development to save mankind. Subunit vaccines alone, however, fail to elicit sufficiently strong and long-lasting protective immunity against deadly pathogens. Nanoparticle (NP)-based delivery vehicles like microemulsions, liposomes, virosomes, nanogels, micelles and dendrimers offer promising strategies to overcome limitations of traditional vaccine adjuvants. Nanovaccines can improve targeted delivery, antigen presentation, stimulation of body's innate immunity, strong T cell response combined with safety to combat infectious diseases and cancers. Further, nanovaccines can be highly beneficial to generate effective immutherapeutic formulations against cancer. AREAS COVERED This review summarizes the emerging nanoparticle strategies highlighting their success and challenges in preclinical and clinical trials in infectious diseases and cancer. It provides a concise overview of current nanoparticle-based vaccines, their adjuvant potential and their cellular delivery mechanisms. EXPERT OPINION The nanovaccines (50-250 nm in size) are most efficient in terms of tissue targeting, prolonged circulation and preferential uptake by the professional APCs chiefly due to their small size. More rational designing, improved antigen loading, extensive functionalization and targeted delivery are some of the future goals of ideal nanovaccines.
Collapse
Affiliation(s)
- Amrita Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
115
|
Liu X, Dong S, Dong M, Li Y, Sun Z, Zhang X, Wang Y, Teng L, Wang D. Transferrin-conjugated liposomes loaded with carnosic acid inhibit liver cancer growth by inducing mitochondria-mediated apoptosis. Int J Pharm 2021; 607:121034. [PMID: 34425193 DOI: 10.1016/j.ijpharm.2021.121034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/27/2022]
Abstract
Our previous studies have proven that carnosic acid (CA) induces apoptosis of liver cancer cells. However, the poor chemical properties of CA limit its in vivo anti-cancer effects. In this study, CA was loaded into liposomes (LP-CA), and LP-CA was further conjugated with transferrin (Tf-LP-CA) to overcome the shortcomings of poor solubility and absorption at the lesion site. In HepG2 and SMMC-7721 cells, compared with CA and LP-CA, more Tf-LP-CA was absorbed by liver cancer cells, which induced higher levels of apoptosis and reduced the mitochondrial membrane potential more effectively. In HepG2- and SMMC-7721-xenotransplanted mice, Tf-LP-CA inhibited tumor growth with no cytotoxicity to the liver, spleen, or kidney. Furthermore, compared with CA and LP-CA, Tf-LP-CA targeted the tumor site more effectively, enhanced the expressions of cleaved poly(ADP-ribose) polymerase, and Caspase-3 and -9, and regulated the expression levels of B-cell lymphoma 2 (Bcl2) family members in the tumor tissues. Tf-LP-CA was taken up by tumor cells and targeted at tumor tissues, ensuring the precise delivery of CA, which further promoted mitochondria-mediated intrinsic apoptosis in the liver cancer cells. These results provide evidence for the clinical application of the Tf-LP-based CA drug delivery system for liver cancer.
Collapse
Affiliation(s)
- Xin Liu
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Shiyan Dong
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Mingyuan Dong
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yuan Li
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Zhen Sun
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Xinrui Zhang
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yingwu Wang
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lesheng Teng
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| | - Di Wang
- Shcool of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
116
|
Sattari S, Adeli M, Beyranvand S, Nemati M. Functionalized Graphene Platforms for Anticancer Drug Delivery. Int J Nanomedicine 2021; 16:5955-5980. [PMID: 34511900 PMCID: PMC8416335 DOI: 10.2147/ijn.s249712] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022] Open
Abstract
Two-dimensional nanomaterials are emerging as promising candidates for a wide range of biomedical applications including tissue engineering, biosensing, pathogen incapacitation, wound healing, and gene and drug delivery. Graphene, due to its high surface area, photothermal property, high loading capacity, and efficient cellular uptake, is at the forefront of these materials and plays a key role in this multidisciplinary research field. Poor water dispersibility and low functionality of graphene, however, hamper its hybridization into new nanostructures for future nanomedicine. Functionalization of graphene, either by covalent or non-covalent methods, is the most useful strategy to improve its dispersion in water and functionality as well as processability into new materials and devices. In this review, recent advances in functionalization of graphene derivatives by different (macro)molecules for future biomedical applications are reported and explained. In particular, hydrophilic functionalization of graphene and graphene oxide (GO) to improve their water dispersibility and physicochemical properties is discussed. We have focused on the anticancer drug delivery of polyfunctional graphene sheets.
Collapse
Affiliation(s)
- Shabnam Sattari
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| | - Mohsen Adeli
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| | - Siamak Beyranvand
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| | - Mohammad Nemati
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| |
Collapse
|
117
|
Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems. Pharmaceuticals (Basel) 2021; 14:ph14090835. [PMID: 34577537 PMCID: PMC8466509 DOI: 10.3390/ph14090835] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
According to a 2020 World Health Organization report (Globocan 2020), cancer was a leading cause of death worldwide, accounting for nearly 10 million deaths in 2020. The aim of anticancer therapy is to specifically inhibit the growth of cancer cells while sparing normal dividing cells. Conventional chemotherapy, radiotherapy and surgical treatments have often been plagued by the frequency and severity of side effects as well as severe patient discomfort. Cancer targeting by drug delivery systems, owing to their selective targeting, efficacy, biocompatibility and high drug payload, provides an attractive alternative treatment; however, there are technical, therapeutic, manufacturing and clinical barriers that limit their use. This article provides a brief review of the challenges of conventional anticancer therapies and anticancer drug targeting with a special focus on liposomal drug delivery systems.
Collapse
|
118
|
Berkowitz SA, Laue T. Boundary convection during velocity sedimentation in the Optima analytical ultracentrifuge. Anal Biochem 2021; 631:114306. [PMID: 34274312 DOI: 10.1016/j.ab.2021.114306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022]
Abstract
Analytical ultracentrifugation (AUC) provides the most widely applicable, precise, and accurate means for characterizing solution hydrodynamic and thermodynamic properties. While generally useful, boundary sedimentation velocity AUC (SV-AUC) analysis has become particularly important in assessing protein aggregation, fragmentation and conformational variants in the same solvents used during drug development and production. In early 2017 the only manufacturer of the analytical ultracentrifuge released its newest analytical ultracentrifuge, the Optima, to replace the aging second-generation XLA/I series ultracentrifuges. However, SV-AUC data from four Optima units used in the characterization of adeno-associated virus (AAV) have shown evidence of sample convection. Further investigation reveals this problem arises from the design of the temperature control system, which makes it prone to producing destabilizing temperature-induced density gradients that can lead to density inversions. The problem is intermittent and variable in severity within a given Optima unit and between Optima units. This convection appears to be associated mainly with low rotor speeds and dilute concentration of solvent components, i.e., AAV analysis conditions. Data features diagnostic for this problem and strategies for its elimination or minimization are provided.
Collapse
Affiliation(s)
| | - Thomas Laue
- Emeritus, University of New Hampshire, 10 Kelsey Road, Lee, NH, 03861, USA
| |
Collapse
|
119
|
Berger M, Lechanteur A, Evrard B, Piel G. Innovative lipoplexes formulations with enhanced siRNA efficacy for cancer treatment: Where are we now? Int J Pharm 2021; 605:120851. [PMID: 34217823 DOI: 10.1016/j.ijpharm.2021.120851] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
Over the past two decades, RNA interference has become an extensively studied mechanism to silence gene and treat diseases including cancer. siRNA appears as a promising strategy that could avoid some side effects related to traditional chemotherapy. Considering the weak stability of naked siRNA in blood, vectors like cationic liposomes or Lipid Nanoparticles (LNPs) are widely used to carry and protect siRNA until it reaches the tumor targeted. Despite extensive research, only three RNAi drugs are currently approved by the Food and Drug Administration, including only one LNP formulation of siRNA to treat hereditary ATTR amyloidosis. This shows the difficulty of lipoplexes clinical translation, in particular in cancer therapy. To overcome the lipoplexes limitations, searches are made on innovative lipoplexes formulations with enhanced siRNA efficacy. The present review is focusing on the recent use of pH-sensitive lipids, peptides and cell-penetrating peptides or polymers. The incorporation of some of these components in the lipoplex formulation induces a fusogenic property or an enhanced endosomal escape, an enhanced cellular uptake, an enhanced tumor targeting, an improved stability in the blood stream …These innovations appear critical to obtain an efficient siRNA accumulation in tumor cells with effective antitumor effect considering the complex tumor environment.
Collapse
Affiliation(s)
- Manon Berger
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Belgium.
| | - Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Belgium.
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Belgium.
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liege, Belgium.
| |
Collapse
|
120
|
Kashapov R, Ibragimova A, Pavlov R, Gabdrakhmanov D, Kashapova N, Burilova E, Zakharova L, Sinyashin O. Nanocarriers for Biomedicine: From Lipid Formulations to Inorganic and Hybrid Nanoparticles. Int J Mol Sci 2021; 22:7055. [PMID: 34209023 PMCID: PMC8269010 DOI: 10.3390/ijms22137055] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Encapsulation of cargoes in nanocontainers is widely used in different fields to solve the problems of their solubility, homogeneity, stability, protection from unwanted chemical and biological destructive effects, and functional activity improvement. This approach is of special importance in biomedicine, since this makes it possible to reduce the limitations of drug delivery related to the toxicity and side effects of therapeutics, their low bioavailability and biocompatibility. This review highlights current progress in the use of lipid systems to deliver active substances to the human body. Various lipid compositions modified with amphiphilic open-chain and macrocyclic compounds, peptide molecules and alternative target ligands are discussed. Liposome modification also evolves by creating new hybrid structures consisting of organic and inorganic parts. Such nanohybrid platforms include cerasomes, which are considered as alternative nanocarriers allowing to reduce inherent limitations of lipid nanoparticles. Compositions based on mesoporous silica are beginning to acquire no less relevance due to their unique features, such as advanced porous properties, well-proven drug delivery efficiency and their versatility for creating highly efficient nanomaterials. The types of silica nanoparticles, their efficacy in biomedical applications and hybrid inorganic-polymer platforms are the subject of discussion in this review, with current challenges emphasized.
Collapse
Affiliation(s)
- Ruslan Kashapov
- A.E. Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Street 8, 420088 Kazan, Russia; (A.I.); (R.P.); (D.G.); (N.K.); (E.B.); (L.Z.); (O.S.)
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Viñas JL, Spence M, Porter CJ, Douvris A, Gutsol A, Zimpelmann JA, Campbell PA, Burns KD. micro-RNA-486-5p protects against kidney ischemic injury and modifies the apoptotic transcriptome in proximal tubules. Kidney Int 2021; 100:597-612. [PMID: 34181969 DOI: 10.1016/j.kint.2021.05.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Acute kidney injury (AKI) carries high morbidity and mortality, and effective treatments are lacking. Preclinical models support involvement of micro-RNAs (miRs) in AKI pathogenesis, although effects on the kidney transcriptome are unclear. We previously showed that injection of cord blood endothelial colony forming cell-derived exosomes, enriched in miR-486-5p, prevented ischemic AKI in mice. To further define this, we studied direct effects of miR-486-5p in mice with kidney ischemia-reperfusion injury. RNA-Seq was used to compare the impact of miR-486-5p and exosomes on the transcriptome of proximal tubules and kidney endothelial cells 24 hours after ischemia-reperfusion. In mice with AKI, injection of miR-486-5p mimic increased its levels in proximal tubules and endothelial cells, and improved plasma creatinine, histological injury, neutrophil infiltration, and apoptosis. Additionally, miR-486-5p inhibited expression of its target phosphatase and tensin homolog, and activated protein kinase B. In proximal tubules, miR-486-5p or exosomes reduced expression of genes associated with ischemic injury and the tumor necrosis factor (TNF) pathway, and altered distinct apoptotic genes. In endothelial cells, genes associated with metabolic processes were altered by miR-486-5p or exosomes, although TNF pathway genes were not affected. Thus, our results suggest that miR-486-5p may have therapeutic potential in AKI.
Collapse
Affiliation(s)
- Jose L Viñas
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Matthew Spence
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Christopher J Porter
- Ottawa Bioinformatics Core Facility, the Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Adrianna Douvris
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Alex Gutsol
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Joseph A Zimpelmann
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Pearl A Campbell
- Regenerative Medicine Program, the Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Kevin D Burns
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
122
|
Zu H, Gao D. Non-viral Vectors in Gene Therapy: Recent Development, Challenges, and Prospects. AAPS JOURNAL 2021; 23:78. [PMID: 34076797 PMCID: PMC8171234 DOI: 10.1208/s12248-021-00608-7] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/07/2021] [Indexed: 12/16/2022]
Abstract
Gene therapy has been experiencing a breakthrough in recent years, targeting various specific cell groups in numerous therapeutic areas. However, most recent clinical studies maintain the use of traditional viral vector systems, which are challenging to manufacture cost-effectively at a commercial scale. Non-viral vectors have been a fast-paced research topic in gene delivery, such as polymers, lipids, inorganic particles, and combinations of different types. Although non-viral vectors are low in their cytotoxicity, immunogenicity, and mutagenesis, attracting more and more researchers to explore the promising delivery system, they do not carry ideal characteristics and have faced critical challenges, including gene transfer efficiency, specificity, gene expression duration, and safety. This review covers the recent advancement in non-viral vectors research and formulation aspects, the challenges, and future perspectives.
Collapse
Affiliation(s)
- Hui Zu
- Abbvie Inc., 1 N. Waukegan Rd, North Chicago, Illinois, 60064, USA
| | - Danchen Gao
- Abbvie Inc., 1 N. Waukegan Rd, North Chicago, Illinois, 60064, USA.
| |
Collapse
|
123
|
Della Pelle G, Kostevšek N. Nucleic Acid Delivery with Red-Blood-Cell-Based Carriers. Int J Mol Sci 2021; 22:5264. [PMID: 34067699 PMCID: PMC8156122 DOI: 10.3390/ijms22105264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has the potential to become a staple of 21st-century medicine. However, to overcome the limitations of existing gene-delivery therapies, that is, poor stability and inefficient and delivery and accumulation of nucleic acids (NAs), safe drug-delivery systems (DDSs) allowing the prolonged circulation and expression of the administered genes in vivo are needed. In this review article, the development of DDSs over the past 70 years is briefly described. Since synthetic DDSs can be recognized and eliminated as foreign substances by the immune system, new approaches must be found. Using the body's own cells as DDSs is a unique and exciting strategy and can be used in a completely new way to overcome the critical limitations of existing drug-delivery approaches. Among the different circulatory cells, red blood cells (RBCs) are the most abundant and thus can be isolated in sufficiently large quantities to decrease the complexity and cost of the treatment compared to other cell-based carriers. Therefore, in the second part, this article describes 70 years of research on the development of RBCs as DDSs, covering the most important RBC properties and loading methods. In the third part, it focuses on RBCs as the NA delivery system with advantages and drawbacks discussed to decide whether they are suitable for NA delivery in vivo.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, Jamova Cesta 39, 1000 Ljubljana, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
| |
Collapse
|
124
|
Li D, Gao C, Kuang M, Xu M, Wang B, Luo Y, Teng L, Xie J. Nanoparticles as Drug Delivery Systems of RNAi in Cancer Therapy. Molecules 2021; 26:2380. [PMID: 33921892 PMCID: PMC8073355 DOI: 10.3390/molecules26082380] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/26/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
RNA interference (RNAi) can mediate gene-silencing by knocking down the expression of a target gene via cellular machinery with much higher efficiency in contrast to other antisense-based approaches which represents an emerging therapeutic strategy for combating cancer. Distinct characters of nanoparticles, such as distinctive size, are fundamental for the efficient delivery of RNAi therapeutics, allowing for higher targeting and safety. In this review, we present the mechanism of RNAi and briefly describe the hurdles and concerns of RNAi as a cancer treatment approach in systemic delivery. Furthermore, the current nanovectors for effective tumor delivery of RNAi therapeutics are classified, and the characteristics of different nanocarriers are summarized.
Collapse
Affiliation(s)
- Diedie Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Chengzhi Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Meiyan Kuang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Minhao Xu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Ben Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Yi Luo
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Jing Xie
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| |
Collapse
|
125
|
Gagliardi M, Ashizawa AT. The Challenges and Strategies of Antisense Oligonucleotide Drug Delivery. Biomedicines 2021; 9:biomedicines9040433. [PMID: 33923688 PMCID: PMC8072990 DOI: 10.3390/biomedicines9040433] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/01/2021] [Accepted: 04/15/2021] [Indexed: 12/27/2022] Open
Abstract
Antisense oligonucleotides (ASOs) are used to selectively inhibit the translation of disease-associated genes via Ribonuclease H (RNaseH)-mediated cleavage or steric hindrance. They are being developed as a novel and promising class of drugs targeting a wide range of diseases. Despite the great potential and numerous ASO drugs in preclinical research and clinical trials, there are many limitations to this technology. In this review we will focus on the challenges of ASO delivery and the strategies adopted to improve their stability in the bloodstream, delivery to target sites, and cellular uptake. Focusing on liposomal delivery, we will specifically describe liposome-incorporated growth factor receptor-bound protein-2 (Grb2) antisense oligodeoxynucleotide BP1001. BP1001 is unique because it is uncharged and is essentially non-toxic, as demonstrated in preclinical and clinical studies. Additionally, its enhanced biodistribution makes it an attractive therapeutic modality for hematologic malignancies as well as solid tumors. A detailed understanding of the obstacles that ASOs face prior to reaching their targets and continued advances in methods to overcome them will allow us to harness ASOs’ full potential in precision medicine.
Collapse
|
126
|
Hall R, Alasmari A, Mozaffari S, Mahdipoor P, Parang K, Montazeri Aliabadi H. Peptide/Lipid-Associated Nucleic Acids (PLANAs) as a Multicomponent siRNA Delivery System. Mol Pharm 2021; 18:986-1002. [PMID: 33496597 DOI: 10.1021/acs.molpharmaceut.0c00969] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RNAi is a biological process that utilizes small interfering RNA (siRNA) to prevent the translation of mRNA to protein. This mechanism could be beneficial in preventing the overexpression of proteins in cancer. However, the cellular delivery of siRNA has proven to be challenging due to its inherent negative charge and relative instability. Here, we designed a multicomponent delivery system composed of a specifically designed peptide (linear or cyclic fatty acyl peptide conjugates and hybrid cyclic/linear peptides) and several lipids (DOTAP, DOPE, cholesterol, and phosphatidylcholine) to form a nanoparticle, which we have termed as peptide lipid-associated nucleic acids (PLANAs). Five formulations were prepared (a formulation with no peptide, which was named lipid-associated nucleic acid or LANA, and PLANA formulations A-D) using a mini extruder to form uniform nanoparticles around 100 nm in size with a slightly positive charge (less than +10 mv). Formulations were evaluated for peptide incorporation, siRNA encapsulation efficiency, release profile, toxicity, cellular uptake, and protein silencing. Our experiments showed effective encapsulation of siRNA (>95%), a controlled release profile, and negligible toxicity in formulations that did not contain a positively charged lipid. The results also revealed that PLANAs C and D exhibited optimum cellular uptake (with 80-90% siRNA-positive cells for most of the formulations). PLANA D formulation was selected to silence two model proteins (Src and RPS6KA5) in the triple-negative human breast cancer cell line MDA-MB-231, with promising silencing efficiency, which diminished the expression of RPS6KA5 and Src to approximately 29 and 38% compared to naïve cells, respectively. Many approaches have been investigated for safe and efficient delivery of nucleic acids in the last 20 years; however, many have failed due to the multifaceted challenges to overcome. Our results show a promising potential for a multicomponent design that incorporates different components for a variety of delivery tasks, which warrants further investigation of PLANAs in vivo.
Collapse
Affiliation(s)
- Ryley Hall
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Abdulaziz Alasmari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Saghar Mozaffari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Parvin Mahdipoor
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Hamidreza Montazeri Aliabadi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| |
Collapse
|
127
|
Synthesis of sulfanyl porphyrazines with bulky peripheral substituents – Evaluation of their photochemical properties and biological activity. J Photochem Photobiol A Chem 2021. [DOI: 10.1016/j.jphotochem.2020.112964] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|