1
|
Choudry MW, Riaz R, Raza MH, Nawaz P, Ahmad B, Jahan N, Rafique S, Afza S, Amin I, Shahid M. Development of Non-Viral Targeted RNA Delivery Vehicles - A Key Factor in Success of Therapeutic RNA. J Drug Target 2024:1-24. [PMID: 39392510 DOI: 10.1080/1061186x.2024.2416241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/23/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
Decade-long efforts in medicinal biotechnology have enabled large-scale in-vitro production of optimized therapeutic RNA constructs for stable in-vivo delivery and modify the expression of disease-related genes. The success of lipid nanoparticle-formulated mRNA vaccines against Severe acute respiratory syndrome Coronavirus-2 (SARS-Cov2) has opened a new era of RNA therapeutics and non-viral drug delivery systems. The major limiting factor in the clinical translation of RNA-based drugs is the availability of suitable delivery vehicles that can protect RNA payloads from degradation, offer controlled release, and pose minimal inherent toxicity. Unwanted immune response, payload size constraints, genome integration, and non-specific tissue targeting limit the application of conventional viral drug-delivery vehicles. This review summarizes current research on nano-sized drug carriers, including lipid nanoparticles, polymer-based formulations, cationic nanoemulsion, and cell-penetrating peptides, for targeted therapeutic RNA delivery. Further, this paper highlights the biomimetic approaches (i.e., mimicking naturally occurring bio-compositions, molecular designs, and systems), including virus-like particles (VLPs), exosomes, and selective endogenous eNcapsidation (SEND) technology being explored as safer and more efficient alternatives.
Collapse
Affiliation(s)
- Muhammad Waqas Choudry
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Rabia Riaz
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Hassan Raza
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Pashma Nawaz
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Bilal Ahmad
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Neelam Jahan
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Shazia Rafique
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Samia Afza
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Iram Amin
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Shahid
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
2
|
Salman DM, Mohammad TAM. siRNA-based therapy for gastric adenocarcinoma: what's next step? Pathol Res Pract 2024; 258:155328. [PMID: 38744002 DOI: 10.1016/j.prp.2024.155328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Gastric cancer continues to have a high death rate despite advancements in their diagnosis and treatment. Novel treatment techniques are thus desperately needed. This is where double-stranded RNA molecules known as small interfering RNA (siRNA), which may selectively target the mRNA of disease-causing genes, may find use in medicine. For siRNAs to function properly in the human body, they must be shielded from deterioration. Furthermore, in order to maintain organ function, they must only target the tumor and spare normal tissue. siRNAs have been designed using clever delivery mechanisms including polymers and lipids to achieve these objectives. Although siRNA protection is not hard to acquire, it is still challenging to target cancer cells with them. Here, we first discuss the basic characteristics of gastric cancer before describing the properties of siRNA and typical delivery methods created specifically for gastric tumors. Lastly, we provide a succinct overview of research using siRNAs to treat gastric tumors.
Collapse
Affiliation(s)
- Dyar Mudhafar Salman
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Pharmacy department, School of Medicine, University of Kurdistan Hewlêr (UKH), Erbil, Kurdistan Region, Iraq.
| |
Collapse
|
3
|
Zhang W, Shen J, Liang J, Ge C, Zhou Y, Yin L, Ji Y. Pulmonary RNA interference against acute lung injury mediated by mucus- and cell-penetrating nanocomplexes. Acta Biomater 2024; 177:332-346. [PMID: 38290689 DOI: 10.1016/j.actbio.2024.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/03/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
Trans-mucosal delivery of anti-inflammatory siRNA into alveolar macrophages represents a promising modality for the treatment of acute lung injury (ALI). However, its therapeutic efficacy is often hurdled by the lack of effective carriers that can simultaneously overcome the mucosal barrier and cell membrane barrier. Herein, we developed mucus/cell membrane dual-penetrating, macrophage-targeting polyplexes which enabled efficient intratracheal delivery of TNF-α siRNA (siTNF-α) to attenuate pulmonary inflammation against lipopolysaccharide (LPS)-induced ALI. P-G@Zn, a cationic helical polypeptide bearing both guanidine and zinc dipicolylamine (Zn-DPA) side charged groups, was designed to condense siTNF-α and promote macrophage internalization due to its helicity-dependent membrane activity. Coating of the polyplexes with charge-neutralizing carboxylated mannan (Man-COOH) greatly enhanced the mucus penetration potency due to shielding of the electrostatic adhesive interactions with the mucus, and it cooperatively enabled active targeting to alveolar macrophages to potentiate the intracellular delivery efficiency of siTNF-α. As such, intratracheally administered Man-COOH/P-G@Zn/siTNF-α polyplexes provoked notable TNF-α silencing by ∼75 % in inflamed lung tissues at 500 μg siRNA/kg, and demonstrated potent anti-inflammatory performance to treat ALI. This study provides an effective tool for the synchronized trans-mucosal delivery of siRNA into macrophages, and the unique properties of the polyplexes render remarkable potentials for anti-inflammatory therapy against ALI. STATEMENT OF SIGNIFICANCE: siRNA-mediated anti-inflammatory management of acute lung injury (ALI) is greatly challenged by the insufficient delivery across the mucus layer and cell membrane. To address such critical issue, mucus/cell membrane dual-penetrating, macrophage-targeting polyplexes are herein developed, which are comprised of an outer shell of carboxylated mannan (Man-COOH) and an inner nanocore formed by TNF-α siRNA (siTNF-α) and a cationic helical polypeptide P-G@Zn. Man-COOH coating endowed the polyplexes with high mucus-penetrating capability and macrophage-targeting ability, while P-G@Zn bearing both guanidine and zinc dipicolylamine afforded potent siTNF-α condensation capacity and high intracellular delivery efficiency with reduced cytotoxicity. Intratracheally administered polyplexes solicit pronounced TNF-α silencing and anti-inflammatory efficiencies in ALI mice. This study renders an effective example for overcoming the multiple barriers against trans-mucosal delivery of siRNA into macrophages, and holds profound potentials for gene therapy against ALI.
Collapse
Affiliation(s)
- Wenxin Zhang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Jingrui Shen
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Jialong Liang
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Chenglong Ge
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Yang Zhou
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Yong Ji
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China.
| |
Collapse
|
4
|
Ranasinghe P, Addison ML, Dear JW, Webb DJ. Small interfering RNA: Discovery, pharmacology and clinical development-An introductory review. Br J Pharmacol 2023; 180:2697-2720. [PMID: 36250252 DOI: 10.1111/bph.15972] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/28/2022] Open
Abstract
Post-transcriptional gene silencing targets and degrades mRNA transcripts, silencing the expression of specific genes. RNA interference technology, using synthetic structurally well-defined short double-stranded RNA (small interfering RNA [siRNA]), has advanced rapidly in recent years. This introductory review describes the utility of siRNA, by exploring the underpinning biology, pharmacology, recent advances and clinical developments, alongside potential limitations and ongoing challenges. Mediated by the RNA-induced silencing complex, siRNAs bind to specific complementary mRNAs, which are subsequently degraded. siRNA therapy offers advantages over other therapeutic approaches, including ability of specifically designed siRNAs to potentially target any mRNA and improved patient adherence through infrequent administration associated with a very long duration of action. Key pharmacokinetic and pharmacodynamic challenges include targeted administration, poor tissue penetration, nuclease inactivation, rapid renal elimination, immune activation and off-target effects. These have been overcome by chemical modification of siRNA and/or by utilising a range of delivery systems, increasing bioavailability and stability to allow successful clinical translation. Patisiran (hereditary transthyretin-mediated amyloidosis) was the first licensed siRNA, followed by givosiran (acute hepatic porphyria), lumasiran (primary hyperoxaluria type 1) and inclisiran (familial hypercholesterolaemia), which all use N-acetylgalactosamine (GalNAc) linkage for effective liver-directed delivery. Others are currently under development for indications varying from rare genetic diseases to common chronic non-communicable diseases (hypertension, cancer). Technological advances are paving the way for broader clinical use. Ongoing challenges remain in targeting organs beyond the liver and reaching special sites (e.g., brain). By overcoming these barriers, siRNA therapy has the potential to substantially widen its therapeutic impact.
Collapse
Affiliation(s)
- Priyanga Ranasinghe
- Department of Pharmacology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Melisande L Addison
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - James W Dear
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - David J Webb
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Zeng Y, Shen M, Singhal A, Sevink GJA, Crone N, Boyle AL, Kros A. Enhanced Liposomal Drug Delivery Via Membrane Fusion Triggered by Dimeric Coiled-Coil Peptides. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301133. [PMID: 37199140 DOI: 10.1002/smll.202301133] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/06/2023] [Indexed: 05/19/2023]
Abstract
An ideal nanomedicine system improves the therapeutic efficacy of drugs. However, most nanomedicines enter cells via endosomal/lysosomal pathways and only a small fraction of the cargo enters the cytosol inducing therapeutic effects. To circumvent this inefficiency, alternative approaches are desired. Inspired by fusion machinery found in nature, synthetic lipidated peptide pair E4/K4 is used to induce membrane fusion previously. Peptide K4 interacts specifically with E4, and it has a lipid membrane affinity and resulting in membrane remodeling. To design efficient fusogens with multiple interactions, dimeric K4 variants are synthesized to improve fusion with E4-modified liposomes and cells. The secondary structure and self-assembly of dimers are studied; the parallel PK4 dimer forms temperature-dependent higher-order assemblies, while linear K4 dimers form tetramer-like homodimers. The structures and membrane interactions of PK4 are supported by molecular dynamics simulations. Upon addition of E4, PK4 induced the strongest coiled-coil interaction resulting in a higher liposomal delivery compared to linear dimers and monomer. Using a wide spectrum of endocytosis inhibitors, membrane fusion is found to be the main cellular uptake pathway. Doxorubicin delivery results in efficient cellular uptake and concomitant antitumor efficacy. These findings aid the development of efficient delivery systems of drugs into cells using liposome-cell fusion strategies.
Collapse
Affiliation(s)
- Ye Zeng
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Mengjie Shen
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Ankush Singhal
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Geert Jan Agur Sevink
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Niek Crone
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Aimee L Boyle
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Alexander Kros
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| |
Collapse
|
6
|
Nanotechnology for DNA and RNA delivery. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
7
|
Zhang L, Jin GZ, Li D. Tat-hspb1 Suppresses Clear Cell Renal Cell Carcinoma (ccRCC) Growth via Lysosomal Membrane Permeabilization. Cancers (Basel) 2022; 14:cancers14225710. [PMID: 36428802 PMCID: PMC9688814 DOI: 10.3390/cancers14225710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney cancer, of which the incidence is increasing worldwide with a high mortality rate. Bioactive peptides are considered a significant class of natural medicines. We applied mass spectrometry-based peptidomic analysis to explore the peptide profile of human renal clear cell carcinoma and adjacent normal tissues. A total of 18,031 peptides were identified, of which 105 unique peptides were differentially expressed (44 were up-regulated and 61 were down-regulated in ccRCC tissues). Through bioinformatic analysis, we finally selected one peptide derived from the HSPB1 protein (amino acids 12-35 of the N-terminal region of HSPB1). Next, we fused this peptide to the HIV-Tat, generated a novel peptide named Tat-hspb1, and found that Tat-hspb1 inhibited ccRCC cells' viability while being less cytotoxic to normal epithelial cells. Furthermore, Tat-hspb1 induced apoptosis and inhibited the proliferation and migration of ccRCC cells. Furthermore, we demonstrated that Tat-hspb1 was predominantly localized in lysosomes after entering the ccRCC cell and induced lysosomal membrane permeabilization (LMP) and the release of cathepsin D from lysosomes. Taken together, Tat-hspb1 has the potential to serve as a new anticancer drug candidate.
Collapse
Affiliation(s)
- Lin Zhang
- Departments of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Guang-Zhi Jin
- Hongqiao International Institute of Medicine, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Correspondence: (G.-Z.J.); (D.L.)
| | - Dong Li
- Departments of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Correspondence: (G.-Z.J.); (D.L.)
| |
Collapse
|
8
|
Yang Y, Zhang X, Wu S, Zhang R, Zhou B, Zhang X, Tang L, Tian Y, Men K, Yang L. Enhanced nose-to-brain delivery of siRNA using hyaluronan-enveloped nanomicelles for glioma therapy. J Control Release 2021; 342:66-80. [PMID: 34973309 DOI: 10.1016/j.jconrel.2021.12.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 02/05/2023]
Abstract
Gliomas are the most malignant brain tumors, and their treatment is very challenging because of the presence of the blood-brain barrier (BBB). Intranasal administration has been considered a noninvasive strategy for glioma therapy in recent years, but our explorations of the intranasal delivery of siRNA-based therapies are still clearly inadequate. In this study, the cell-penetrating peptide DP7-C was enveloped with hyaluronic acid (HA) to develop the multifunctional core-shell structure nanomicelle HA/DP7-C. In vitro studies of HA/DP7-C revealed low cytotoxicity and a higher cell uptake efficiency, which was associated with the interaction between HA and CD44. In vivo experiments indicated that HA/DP7-C delivered the siRNA to the central nervous system through the trigeminal nerve pathway within hours after intranasal administration and that the interaction between HA and CD44 also increased its accumulation at the tumor site. Successful intracellular delivery of an antiglioma siRNA inhibited tumor growth and ultimately prolonged the survival time and decreased the tumor volume in GL261 tumor-bearing mice. In addition, toxicity tests on rats showed no adverse effects on the nasal mucosa and trigeminal nerves. In conclusion, HA/DP7-C is a potential intranasal delivery system for siRNAs in glioma therapy.
Collapse
Affiliation(s)
- YuLing Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - XueYan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - SiWen Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Rui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - BaiLing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - XiaoYu Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
9
|
Miliotou AN, Pappas IS, Spyroulias G, Vlachaki E, Tsiftsoglou AS, Vizirianakis IS, Papadopoulou LC. Development of a novel PTD-mediated IVT-mRNA delivery platform for potential protein replacement therapy of metabolic/genetic disorders. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:694-710. [PMID: 34703653 PMCID: PMC8517095 DOI: 10.1016/j.omtn.2021.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 09/09/2021] [Indexed: 12/22/2022]
Abstract
The potential clinical applications of the powerful in vitro-transcribed (IVT)-mRNAs, to restore defective protein functions, strongly depend on their successful intracellular delivery and transient translation through the development of safe and efficient delivery platforms. In this study, an innovative (international patent-pending) methodology was developed, combining the IVT-mRNAs with the protein transduction domain (PTD) technology, as an efficient delivery platform. Based on the PTD technology, which enables the intracellular delivery of various cargoes intracellularly, successful conjugation of a PTD to the IVT-mRNAs was achieved and evaluated by band-shift assay and NMR spectroscopy. In addition, the PTD-IVT-mRNAs were applied and evaluated in two protein-disease models, including the mitochondrial disorder fatal infantile cardioencephalomyopathy and cytochrome c oxidase (COX) deficiency (attributed to SCO2 gene mutations) and β-thalassemia. The PTD-IVT-mRNA of SCO2 was successfully transduced and translated to the corresponding Sco2 protein inside the primary fibroblasts of a SCO2/COX-deficient patient, whereas the PTD-IVT-mRNA of β-globin was transduced and translated in bone marrow cells, derived from three β-thalassemic patients. The transducibility and the structural stability of the PDT-IVT-mRNAs, in both cases, were confirmed at the RNA and protein levels. We propose that our novel delivery platform could be clinically applicable as a protein therapy for metabolic/genetic disorders.
Collapse
Affiliation(s)
- Androulla N Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 546 42 Macedonia, Greece
| | - Ioannis S Pappas
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Science, University of Thessaly, Karditsa, 431 00 Thessaly, Greece
| | | | - Efthimia Vlachaki
- Adult Thalassemia Unit, Hippokrateion General Hospital, Thessaloniki, 546 42 Macedonia, Greece
| | - Asterios S Tsiftsoglou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 546 42 Macedonia, Greece
| | - Ioannis S Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 546 42 Macedonia, Greece.,Department of Life and Health Sciences, University of Nicosia, 1700 Nicosia, Cyprus
| | - Lefkothea C Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 546 42 Macedonia, Greece
| |
Collapse
|
10
|
Messerschmidt VL, Chintapula U, Kuriakose AE, Laboy S, Truong TTD, Kydd LA, Jaworski J, Pan Z, Sadek H, Nguyen KT, Lee J. Notch Intracellular Domain Plasmid Delivery via Poly(Lactic-Co-Glycolic Acid) Nanoparticles to Upregulate Notch Pathway Molecules. Front Cardiovasc Med 2021; 8:707897. [PMID: 34651022 PMCID: PMC8507495 DOI: 10.3389/fcvm.2021.707897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
Notch signaling is a highly conserved signaling system that is required for embryonic development and regeneration of organs. When the signal is lost, maldevelopment occurs and leads to a lethal state. Delivering exogenous genetic materials encoding Notch into cells can reestablish downstream signaling and rescue cellular functions. In this study, we utilized the negatively charged and FDA approved polymer poly(lactic-co-glycolic acid) to encapsulate Notch Intracellular Domain-containing plasmid in nanoparticles. We show that primary human umbilical vein endothelial cells (HUVECs) readily uptake the nanoparticles with and without specific antibody targets. We demonstrated that our nanoparticles are non-toxic, stable over time, and compatible with blood. We further demonstrated that HUVECs could be successfully transfected with these nanoparticles in static and dynamic environments. Lastly, we elucidated that these nanoparticles could upregulate the downstream genes of Notch signaling, indicating that the payload was viable and successfully altered the genetic downstream effects.
Collapse
Affiliation(s)
- Victoria L Messerschmidt
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Aneetta E Kuriakose
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Samantha Laboy
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Thuy Thi Dang Truong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - LeNaiya A Kydd
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Justyn Jaworski
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Zui Pan
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, United States
| | - Hashem Sadek
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
11
|
Zhou L, Rubin LE, Liu C, Chen Y. Short interfering RNA (siRNA)-Based Therapeutics for Cartilage Diseases. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021; 7:283-290. [PMID: 34589570 DOI: 10.1007/s40883-020-00149-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Articular cartilage injury, as a hallmark of arthritic diseases, is difficult to repair and causes joint pain, stiffness, and loss of mobility. Over the years, the most significant problems for the drug-based treatment of arthritis have been related to drug administration and delivery. In recent years, much research has been devoted to developing new strategies for repairing or regenerating the damaged osteoarticular tissue. The RNA interference (RNAi) has been suggested to have the potential for implementation in targeted therapy in which the faulty gene can be edited by delivering its complementary Short Interfering RNA (siRNA) at the post-transcriptional stage. The successful editing of a specific gene by the delivered siRNA might slow or halt osteoarthritic diseases without side effects caused by chemical inhibitors. However, cartilage siRNA delivery remains a challenging objective because cartilage is an avascular and very dense tissue with very low permeability. Furthermore, RNA is prone to degradation by serum nucleases (such as RNase H and RNase A) due to an extra hydroxyl group in its phosphodiester backbone. Therefore, successful delivery is the first and most crucial requirement for efficient RNAi therapy. Nanomaterials have emerged as highly advantage tools for these studies, as they can be engineered to protect siRNA from degrading, address barriers in siRNA delivery to joints, and target specific cells. This review will discuss recent breakthroughs of different siRNA delivery technologies for cartilage diseases.
Collapse
Affiliation(s)
- Libo Zhou
- Department of Biomedical Engineering, University of Connecticut
| | - Lee E Rubin
- Department of Orthopaedics & Rehabilitation, Yale University School of Medicine
| | - Chuanju Liu
- Department of Orthopaedic Surgery and Cell Biology, New York University School of Medicine
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut
| |
Collapse
|
12
|
Sreedurgalakshmi K, Srikar R, Harikrishnan K, Srinivasan L, Rajkumari R. Cetuximab-siRNA Conjugate Linked Through Cationized Gelatin Knocks Down KRAS G12C Mutation in NSCLC Sensitizing the Cells Toward Gefitinib. Technol Cancer Res Treat 2021; 20:15330338211041453. [PMID: 34542333 PMCID: PMC8461128 DOI: 10.1177/15330338211041453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Delivery of small-interfering RNA (siRNA) has been of great interest in the past decade for effective gene silencing. To overcome synthetic and regulatory challenges posed by nanoparticle-mediated siRNA delivery, antibody–siRNA conjugate (ARC) platform is emerging as a potential siRNA delivery system suitable for clinical translation. Herein, we have developed a delivery technology based on the ARC platform for stable delivery of siRNA called as Gelatin-Antibody Delivery System (GADS). In GADS, positively charged gelatin acts as a linker between antibody–siRNA and enables the endosomal escape of siRNA for gene silencing postcellular internalization. For proof of concept, we synthesized a scalable GADS conjugate comprising of Cetuximab (CTB), cationized gelatin (cGel) and NSCLC KRASG12C-specific siRNA. CTB was chemically conjugated to cGel through an amide link to form the CTB–cGel complex. Thereafter, siRNA was chemically conjugated to the cGel moiety of the complex through the thioether link to form CTB–cGel–siRNA conjugate. RP-HPLC analysis was used to monitor the reaction while gel retardation assay was used to determine siRNA loading capacity. SPR analysis showed the preservation of ligand binding affinity of antibody conjugates with KD of ∼0.3 nM. Furthermore, cellular internalization study using florescent microscopy revealed receptor-mediated endocytosis. The conjugate targeted EGFR receptor of KRAS mutant NSCLC to specifically knockdown G12C mutation. The oncogene knockdown sensitized the cells toward small molecule inhibitor—Gefitinib causing ∼70% loss in cell viability. Western blot analysis revealed significant downregulation for various RAS downstream proteins postoncogene knockdown. Comparison of the efficiency of GADS vis-à-vis positive siRNA control and CRISPR–Cas9-based knockout of KRAS Exon 2 in the NCI-H23 NSCLC cell line suggests GADS as a potential technology for clinical translation of gene therapy.
Collapse
Affiliation(s)
- K. Sreedurgalakshmi
- Vellore Institute of Technology, Vellore, Tamil Nadu, India
- R&D, Levim Biotech LLP, Chennai, Tamil Nadu, India
| | - R. Srikar
- R&D, Levim Biotech LLP, Chennai, Tamil Nadu, India
- R. Srikar, Division of Biosimilars and Gene Therapy, R&D,
Levim Biotech LLP, Chennai, Tamil Nadu, India.
Reena Rajkumari, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.
| | | | | | | |
Collapse
|
13
|
Yu Z, Zhang X, Pei X, Cao W, Ye J, Wang J, Sun L, Yu F, Wang J, Li N, Lee K, Barth S, Yang VC, He H. Antibody-siRNA conjugates (ARCs) using multifunctional peptide as a tumor enzyme cleavable linker mediated effective intracellular delivery of siRNA. Int J Pharm 2021; 606:120940. [PMID: 34310959 DOI: 10.1016/j.ijpharm.2021.120940] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
The tissue-specific targeted delivery and efficient cellular uptake of siRNAs are the main obstacles to their clinical application. Antibody-siRNA-conjugates (ARCs) can deliver siRNA by exploiting the targeting property of antibodies like antibody-drug conjugates (ADCs). However, the effective conjugation of antibodies and siRNAs and the release of siRNAs specifically at target sites have posed challenges to the development of ARCs. In this study, the successful conjugation of antibodies and siRNAs was achieved using a multifunctional peptide as a linker, composed of a cell-penetrating peptide (CPP) and a substrate peptide (SP), which is highly expressed in solid tumors. The resulting antibody-multifunctional peptide (SP-CPP)-siRNA system delivered the siRNA to target tumor cells by the specific binding of the antibody. Once the enzymes on the tumor cell surface hydrolyzed the substrate peptide linker, siRNA-CPP was released from ARCs. The released siRNA-CPP entered the targeted cells via the cellular penetrating ability of CPP, resulting in improved siRNA-mediated gene silencing efficiency, verified both in vitro and in vivo. After intravenous administration, the designed ARCs achieved approximately 66.7% EGFP (Enhanced Green Fluorescent Protein) downregulation efficiency in nude mice xenografted with the HCT116-EGFP tumor model. The proposed system provides a prospective choice for ARC production and the safe and efficient delivery of siRNAs.
Collapse
Affiliation(s)
- Zhili Yu
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaojuan Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xing Pei
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Weiran Cao
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Junxiao Ye
- College of Pharmacy, Tsinghua University, Beijing 100084, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Lu Sun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Fei Yu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Kyuri Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52828, Republic of Korea
| | - Stefan Barth
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Victor C Yang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109-1065, USA
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
14
|
Tian Z, Liang G, Cui K, Liang Y, Wang Q, Lv S, Cheng X, Zhang L. Insight Into the Prospects for RNAi Therapy of Cancer. Front Pharmacol 2021; 12:644718. [PMID: 33796026 PMCID: PMC8007863 DOI: 10.3389/fphar.2021.644718] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
RNA interference (RNAi), also known as gene silencing, is a biological process that prevents gene expression in certain diseases such as cancer. It can be used to improve the accuracy, efficiency, and stability of treatments, particularly genetic therapies. However, challenges such as delivery of oligonucleotide drug to less accessible parts of the body and the high incidence of toxic side effects are encountered. It is therefore imperative to improve their delivery to target sites and reduce their harmful effects on noncancerous cells to harness their full potential. In this study, the role of RNAi in the treatment of COVID-19, the novel coronavirus disease plaguing many countries, has been discussed. This review aims to ascertain the mechanism and application of RNAi and explore the current challenges of RNAi therapy by identifying some of the cancer delivery systems and providing drug information for their improvement. It is worth mentioning that delivery systems such as lipid-based delivery systems and exosomes have revolutionized RNAi therapy by reducing their immunogenicity and improving their cellular affinity. A deeper understanding of the mechanism and challenges associated with RNAi in cancer therapy can provide new insights into RNAi drug development.
Collapse
Affiliation(s)
- Zhili Tian
- Institute of Molecular Medicine, Henan University, Kaifeng, China.,School of Clinical Medical Sciences, Henan University, Kaifeng, China
| | - Guohui Liang
- Institute of Molecular Medicine, Henan University, Kaifeng, China.,School of Clinical Medical Sciences, Henan University, Kaifeng, China
| | - Kunli Cui
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yayu Liang
- Institute of Molecular Medicine, Henan University, Kaifeng, China.,School of Stomatology, Henan University, Kaifeng, China
| | - Qun Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuangyu Lv
- Institute of Molecular Medicine, Henan University, Kaifeng, China
| | - Xiaoxia Cheng
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
15
|
A Novel 89Zr-labeled DDS Device Utilizing Human IgG Variant (scFv): "Lactosome" Nanoparticle-Based Theranostics for PET Imaging and Targeted Therapy. Life (Basel) 2021; 11:life11020158. [PMID: 33670777 PMCID: PMC7923095 DOI: 10.3390/life11020158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/22/2022] Open
Abstract
“Theranostics,” a new concept of medical advances featuring a fusion of therapeutic and diagnostic systems, provides promising prospects in personalized medicine, especially cancer. The theranostics system comprises a novel 89Zr-labeled drug delivery system (DDS), derived from the novel biodegradable polymeric micelle, “Lactosome” nanoparticles conjugated with specific shortened IgG variant, and aims to successfully deliver therapeutically effective molecules, such as the apoptosis-inducing small interfering RNA (siRNA) intracellularly while offering simultaneous tumor visualization via PET imaging. A 27 kDa-human single chain variable fragment (scFv) of IgG to establish clinically applicable PET imaging and theranostics in cancer medicine was fabricated to target mesothelin (MSLN), a 40 kDa-differentiation-related cell surface glycoprotein antigen, which is frequently and highly expressed by malignant tumors. This system coupled with the cell penetrating peptide (CPP)-modified and photosensitizer (e.g., 5, 10, 15, 20-tetrakis (4-aminophenyl) porphyrin (TPP))-loaded Lactosome particles for photochemical internalized (PCI) driven intracellular siRNA delivery and the combination of 5-aminolevulinic acid (ALA) photodynamic therapy (PDT) offers a promising nano-theranostic-based cancer therapy via its targeted apoptosis-inducing feature. This review focuses on the combined advances in nanotechnology and material sciences utilizing the “89Zr-labeled CPP and TPP-loaded Lactosome particles” and future directions based on important milestones and recent developments in this platform.
Collapse
|
16
|
Xie X, Zheng T, Li W. Recent Progress in Ionic Coassembly of Cationic Peptides and Anionic Species. Macromol Rapid Commun 2020; 41:e2000534. [PMID: 33225490 DOI: 10.1002/marc.202000534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/10/2020] [Indexed: 12/25/2022]
Abstract
Peptide assembly has been extensively exploited as a promising platform for the creation of hierarchical nanostructures and tailor-made bioactive materials. Ionic coassembly of cationic peptides and anionic species is paving the way to provide particularly important contribution to this topic. In this review, the recent progress of ionic coassembly soft materials derived from the electrostatic coupling between cationic peptides and anionic species in aqueous solution is systematically summarized. The presentation of this review starts from a brief background on the general importance and advantages of peptide-based ionic coassembly. After that, diverse combinations of cationic peptides with small anions, macro- and/or oligo-anions, anionic polymers, and inorganic polyoxometalates are described. Emphasis is placed on the hierarchical structures, value-added properties, and applications. The molecular design of cationic peptides and the general principles behind the ionic coassembled structures are discussed. It is summarized that the combination of interesting and unique characteristics that arise both from the chemical diversity of peptides and the wide range of anionic species may contribute in a variety of output, including drug delivery, tissue engineering, gene transfection, and antibacterial activity. The emergent new phenomena and findings are illustrated. Finally, the outlook for the peptide-based ionic coassembly systems is also presented.
Collapse
Affiliation(s)
- Xiaoming Xie
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjing Avenue 2699, Changchun, 130012, China.,Department of Chemistry, Xinzhou Teachers' University, Xinzhou, Shanxi, 034000, China
| | - Tingting Zheng
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjing Avenue 2699, Changchun, 130012, China
| | - Wen Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjing Avenue 2699, Changchun, 130012, China
| |
Collapse
|
17
|
Nanocarriers in effective pulmonary delivery of siRNA: current approaches and challenges. Ther Deliv 2020; 10:311-332. [PMID: 31116099 DOI: 10.4155/tde-2019-0012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Research on siRNA is increasing due to its wide applicability as a therapeutic agent in irreversible medical conditions. siRNA inhibits expression of the specific gene after its delivery from formulation to cytosol region of a cell. RNAi (RNA interference) is a mechanism by which siRNA is silencing gene expression for a particular disease. Numerous studies revealed that naked siRNA delivery is not preferred due to instability and poor pharmacokinetic performance. Nanocarriers based delivery of siRNA has the advantage to overcome physiological barriers and protect the integrity of siRNA from degradation by RNAase. Various diseases like lung cancer, cystic fibrosis, asthma, etc can be treated effectively by local lung delivery. The selective targeted therapeutic action in diseased organ and least off targeted cytotoxicity are the key benefits of pulmonary delivery. The current review highlights recent developments in pulmonary delivery of siRNA with novel nanosized formulation approach with the proven in vitro/in vivo applications.
Collapse
|
18
|
Guo Y, Zhang J, Pan G, Choi CHJ, Wang P, Li Y, Zhu X, Zhang C. Grafting multi-maleimides on antisense oligonucleotide to enhance its cellular uptake and gene silencing capability. Chem Commun (Camb) 2020; 56:7439-7442. [PMID: 32494799 DOI: 10.1039/d0cc02548h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A multitude of maleimides are grafted onto the backbone of a phosphorothioate antisense oligonucleotide (ASO) to generate the construct of maleimide-grafted ASO (Mal-g-ASO). Through click conjugation with cell membrane thiols that triggers endocytosis-independent cellular internalization, Mal-g-ASO exhibited enhanced cellular uptake efficiency, resulting in a remarkable improvement of ASO-based gene silencing.
Collapse
Affiliation(s)
- Yuanyuan Guo
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Shu Z, Ota A, Takayama Y, Katsurada Y, Kusamori K, Abe N, Nakamoto K, Tomoike F, Tada S, Ito Y, Nishikawa M, Kimura Y, Abe H. Intracellular Delivery of Antisense DNA and siRNA with Amino Groups Masked with Disulfide Units. Chem Pharm Bull (Tokyo) 2020; 68:129-132. [DOI: 10.1248/cpb.c19-00811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Zhaoma Shu
- Department of Chemistry, Nagoya University
| | - Azumi Ota
- Department of Chemistry, Nagoya University
| | - Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Yuri Katsurada
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Naoko Abe
- Department of Chemistry, Nagoya University
| | | | | | - Seiichi Tada
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | | | - Hiroshi Abe
- Department of Chemistry, Nagoya University
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science
- CREST, Japan Science and Technology Agency
| |
Collapse
|
20
|
Zhou J, Shao Z, Liu J, Duan Q, Wang X, Li J, Yang H. From Endocytosis to Nonendocytosis: The Emerging Era of Gene Delivery. ACS APPLIED BIO MATERIALS 2020; 3:2686-2701. [DOI: 10.1021/acsabm.9b01131] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Zhentao Shao
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Jia Liu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Qiao Duan
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Xiang Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
| |
Collapse
|
21
|
Bae HD, Lee JS, Pyun H, Kim M, Lee K. Optimization of formulation for enhanced intranasal delivery of insulin with translationally controlled tumor protein-derived protein transduction domain. Drug Deliv 2019; 26:622-628. [PMID: 31210056 PMCID: PMC6586149 DOI: 10.1080/10717544.2019.1628119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intranasal delivery of insulin is an alternative approach to treat diabetes, as it enables higher patient compliance than conventional therapy with subcutaneously injected insulin. However, the use of intranasal delivery of insulin is limited for insulin’s hydrophilicity and vulnerability to enzymatic degradation. This limitation makes optimization of formulation intranasal insulin for commercial purpose indispensable. This study evaluated bioavailability (BA) of various formulations of insulin intranasally delivered with protein transduction domain (PTD) derived from translationally controlled tumor protein. The therapeutic efficacy of newly formulated intranasal insulin + PTD was compared in vivo studies with normal and alloxan-induced diabetic rats, to those of free insulin and subcutaneously injected insulin. BA of insulin in two new formulations was, respectively, 60.71% and 45.81% of subcutaneously injected insulin, while the BA of free insulin was only 3.34%. Histological analysis of tissues, lactate dehydrogenase activity in nasal fluid, and biochemical analysis of sera revealed no detectable topical or systemic toxicity in rats and mice. Furthermore, stability analysis of newly formulated insulin + PTD to determine the optimal conditions for storage revealed that when stored at 4 °C, the delivery capacity of insulin was maintained up to 7 d. These results suggest that the new formulations of intranasal insulin are suitable for use in diabetes therapy and are easier to administer.
Collapse
Affiliation(s)
- Hae-Duck Bae
- a Graduate School of Pharmaceutical Sciences, College of Pharmacy , Ewha Woman's University , Seoul , Korea
| | - Ji-Sun Lee
- a Graduate School of Pharmaceutical Sciences, College of Pharmacy , Ewha Woman's University , Seoul , Korea
| | - Haejun Pyun
- a Graduate School of Pharmaceutical Sciences, College of Pharmacy , Ewha Woman's University , Seoul , Korea
| | - Moonhee Kim
- a Graduate School of Pharmaceutical Sciences, College of Pharmacy , Ewha Woman's University , Seoul , Korea
| | - Kyunglim Lee
- a Graduate School of Pharmaceutical Sciences, College of Pharmacy , Ewha Woman's University , Seoul , Korea
| |
Collapse
|
22
|
Alsulays BB, Anwer MK, Soliman GA, Alshehri SM, Khafagy ES. Impact Of Penetratin Stereochemistry On The Oral Bioavailability Of Insulin-Loaded Solid Lipid Nanoparticles. Int J Nanomedicine 2019; 14:9127-9138. [PMID: 31819423 PMCID: PMC6883942 DOI: 10.2147/ijn.s225086] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/25/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose This study evaluated the stereoisomeric effect of L- and D-penetratin—cell-penetrating peptides (CPPs)—incorporated insulin-loaded solid lipid nanoparticles (INS-SLNs) on the bioavailability (BA) of oral insulin (INS). Methods Insulin-loaded solid nanoparticles, L-penetratin-INS-SLNs (LP-INS-SLNs), and D-penetratin-INS-SLNs (DP-INS-SLNs) were formulated by double emulsification. The developed SLNs were evaluated for particle size, zeta potential (ZP), and drug encapsulation and subjected to differential scanning calorimetry (DSC), Fourier transform infrared spectroscopy (FTIR), and evaluated for stability against enzymatic degradation in rat intestinal fluid. Finally, the SLNs were administered to rats to evaluate the BA of INS-SLNs that contained L- and D-penetratin. Results The mean particle size, PDI, and ZP values of INS-SLNs, LP-INS-SLNs, and DP-INS-SLNs ranged from 618.5 to 973.0 nm, 0.227 to 0.734, and −17.0 to −23.7 mV, respectively. The encapsulation efficiency (%EE) and drug loading (%DL) of INS-SLNs, LP-INS-SLNs, and DP-INS-SLNs ranged from 59.03% to 67.42% and from 1.62% to 1.82%, respectively. Differential scanning calorimetry and FTIR analyses indicated that INS was successfully encapsulated in SLNs. Enzymatic degradation of DP-INS-SLNs was slower in intestinal fluid, and the half-life (t1/2) was significantly prolonged, compared to all other SLNs. The pharmacological availability (PA) and BA of orally administered LP-INS-SLNs, which were the most effective SLNs, were 13.1% and 15.7% relative to s.c. administration, respectively. Conclusion Penetratin stereochemistry significantly impacted oral BA of INS-SLNs, which are promising carriers for oral INS administration.
Collapse
Affiliation(s)
- Bader B Alsulays
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Gamal A Soliman
- Department of Pharmacology, College of Veterinary Medicine, Cairo University, Cairo 12211, Egypt.,Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj 11942, Saudi Arabia
| | - Sultan M Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 415-22, Egypt
| |
Collapse
|
23
|
Barba-Bon A, Pan YC, Biedermann F, Guo DS, Nau WM, Hennig A. Fluorescence Monitoring of Peptide Transport Pathways into Large and Giant Vesicles by Supramolecular Host–Dye Reporter Pairs. J Am Chem Soc 2019; 141:20137-20145. [DOI: 10.1021/jacs.9b09563] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Andrea Barba-Bon
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring
1, 28759 Bremen, Germany
| | - Yu-Chen Pan
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Frank Biedermann
- Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Werner M. Nau
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring
1, 28759 Bremen, Germany
| | - Andreas Hennig
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring
1, 28759 Bremen, Germany
| |
Collapse
|
24
|
Cell-Penetrating Peptide and Transferrin Co-Modified Liposomes for Targeted Therapy of Glioma. Molecules 2019; 24:molecules24193540. [PMID: 31574945 PMCID: PMC6804123 DOI: 10.3390/molecules24193540] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/02/2022] Open
Abstract
Glioma is one of the most aggressive and common malignant brain tumors. Due to the presence of the blood-brain barrier (BBB), the effectiveness of therapeutics is greatly affected. In this work, to develop an efficient anti-glioma drug with targeting and which was able to cross the BBB, cell-penetrating peptides (R8) and transferrin co-modified doxorubicin (DOX)-loaded liposomes (Tf-LPs) were prepared. Tf-LPs possessed a spherical shape and uniform size with 128.64 nm and their ξ-potential was 6.81 mV. Tf-LPs were found to be stable in serum within 48 h. Uptake of Tf-LPs in both U87 and GL261 cells was analyzed by confocal laser scanning microscopy and by flow cytometry. Tf-LPs were efficiently taken up by both U87 and GL261 cells. Moreover, Tf-LPs exhibited sustained-release. The cumulative release of DOX from Tf-LPs reached ~50.0% and showed excellent anti-glioma efficacy. Histology of major organs, including brain, heart, liver, spleen, lungs and kidney, and the bodyweight of mice, all indicated low toxicity of Tf-LPs. In conclusion, Tf-LPs showed great promise as an anti-glioma therapeutic agent.
Collapse
|
25
|
Zou Y, Pan R, Liu Y, Liu X, Chen X, Wang J, Wan Z, Guo J, Yang X. Effects of γ-zein peptides on lipid membrane organization: Quartz crystal microbalance with dissipation and Langmuir monolayer studies. Colloids Surf A Physicochem Eng Asp 2019. [DOI: 10.1016/j.colsurfa.2019.04.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Covalent conjugates of granulin-epithelial precursor-siRNA with arginine-rich peptide for improved stability and intracellular delivery in hepatoma cells. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0028-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Jiang H, Hu X, Mosel S, Knauer SK, Hirschhäuser C, Schmuck C. A Branched Tripeptide with an Anion‐Binding Motif as a New Delivery Carrier for Efficient Gene Transfection. Chembiochem 2019; 20:1410-1416. [DOI: 10.1002/cbic.201800728] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Hao Jiang
- Key Laboratory of Materials Chemistry for Energy Conversion and StorageSchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST) Wuhan 430074 P.R. China
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
| | - Xiao‐Yu Hu
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
- Applied Chemistry DepartmentSchool of Material Science and EngineeringNanjing University of Aeronautics and Astronautics Nanjing 210016 P.R. China
| | - Stefanie Mosel
- Institute for BiologyUniversity of Duisburg–Essen Universitätsstrasse 5 45141 Essen Germany
| | - Shirley K. Knauer
- Institute for BiologyUniversity of Duisburg–Essen Universitätsstrasse 5 45141 Essen Germany
| | - Christoph Hirschhäuser
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
| | - Carsten Schmuck
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
| |
Collapse
|
28
|
Shu Z, Tanaka I, Ota A, Fushihara D, Abe N, Kawaguchi S, Nakamoto K, Tomoike F, Tada S, Ito Y, Kimura Y, Abe H. Disulfide‐Unit Conjugation Enables Ultrafast Cytosolic Internalization of Antisense DNA and siRNA. Angew Chem Int Ed Engl 2019; 58:6611-6615. [DOI: 10.1002/anie.201900993] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/27/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Zhaoma Shu
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Iku Tanaka
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Azumi Ota
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Daichi Fushihara
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Naoko Abe
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Saki Kawaguchi
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Kosuke Nakamoto
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Fumiaki Tomoike
- Research Center for Materials Science Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Seiichi Tada
- Emergent Bioengineering Materials Research Team RIKEN Center for Emergent Matter Science 2-1, Hirosawa Wako-Shi Saitama 351-0198 Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team RIKEN Center for Emergent Matter Science 2-1, Hirosawa Wako-Shi Saitama 351-0198 Japan
| | - Yasuaki Kimura
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Hiroshi Abe
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
- Emergent Bioengineering Materials Research Team RIKEN Center for Emergent Matter Science 2-1, Hirosawa Wako-Shi Saitama 351-0198 Japan
- CREST (Japan) Science and Technology Agency Tokyo 102-0076 Japan
| |
Collapse
|
29
|
Shu Z, Tanaka I, Ota A, Fushihara D, Abe N, Kawaguchi S, Nakamoto K, Tomoike F, Tada S, Ito Y, Kimura Y, Abe H. Disulfide‐Unit Conjugation Enables Ultrafast Cytosolic Internalization of Antisense DNA and siRNA. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201900993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zhaoma Shu
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Iku Tanaka
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Azumi Ota
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Daichi Fushihara
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Naoko Abe
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Saki Kawaguchi
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Kosuke Nakamoto
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Fumiaki Tomoike
- Research Center for Materials Science Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Seiichi Tada
- Emergent Bioengineering Materials Research Team RIKEN Center for Emergent Matter Science 2-1, Hirosawa Wako-Shi Saitama 351-0198 Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team RIKEN Center for Emergent Matter Science 2-1, Hirosawa Wako-Shi Saitama 351-0198 Japan
| | - Yasuaki Kimura
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
| | - Hiroshi Abe
- Chemistry Department Nagoya University, Furo-cho, Chikusa-Ku Nagoya Aichi 464-8602 Japan
- Emergent Bioengineering Materials Research Team RIKEN Center for Emergent Matter Science 2-1, Hirosawa Wako-Shi Saitama 351-0198 Japan
- CREST (Japan) Science and Technology Agency Tokyo 102-0076 Japan
| |
Collapse
|
30
|
Benizri S, Gissot A, Martin A, Vialet B, Grinstaff MW, Barthélémy P. Bioconjugated Oligonucleotides: Recent Developments and Therapeutic Applications. Bioconjug Chem 2019; 30:366-383. [PMID: 30608140 PMCID: PMC6766081 DOI: 10.1021/acs.bioconjchem.8b00761] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oligonucleotide-based agents have the potential to treat or cure almost any disease, and are one of the key therapeutic drug classes of the future. Bioconjugated oligonucleotides, a subset of this class, are emerging from basic research and being successfully translated to the clinic. In this Review, we first briefly describe two approaches for inhibiting specific genes using oligonucleotides-antisense DNA (ASO) and RNA interference (RNAi)-followed by a discussion on delivery to cells. We then summarize and analyze recent developments in bioconjugated oligonucleotides including those possessing GalNAc, cell penetrating peptides, α-tocopherol, aptamers, antibodies, cholesterol, squalene, fatty acids, or nucleolipids. These novel conjugates provide a means to enhance tissue targeting, cell internalization, endosomal escape, target binding specificity, resistance to nucleases, and more. We next describe those bioconjugated oligonucleotides approved for patient use or in clinical trials. Finally, we summarize the state of the field, describe current limitations, and discuss future prospects. Bioconjugation chemistry is at the centerpiece of this therapeutic oligonucleotide revolution, and significant opportunities exist for development of new modification chemistries, for mechanistic studies at the chemical-biology interface, and for translating such agents to the clinic.
Collapse
Affiliation(s)
- Sebastien Benizri
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| | - Arnaud Gissot
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| | - Andrew Martin
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, Massachusetts 02215, United States
| | - Brune Vialet
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, Massachusetts 02215, United States
| | - Philippe Barthélémy
- Inserm U1212, F-33076 Bordeaux, France
- CNRS 5320, F-33076 Bordeaux, France
- Universitéde Bordeaux, 146 rue Léo Saignat, F-33076 Bordeaux Cedex, France
| |
Collapse
|
31
|
Chandela A, Ueno Y. Systemic Delivery of Small Interfering RNA Therapeutics: Obstacles and Advances. ACTA ACUST UNITED AC 2019. [DOI: 10.7831/ras.7.10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Akash Chandela
- United Graduate School of Agricultural Science, Gifu University
| | - Yoshihito Ueno
- United Graduate School of Agricultural Science, Gifu University
- Course of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| |
Collapse
|
32
|
Chen W, Luan J, Wei G, Zhang X, Fan J, Zai W, Wang S, Wang Y, Liang Y, Nan Y, Yin C, Li Y, Liu ML, Ju D. In vivo hepatocellular expression of interleukin-22 using penetratin-based hybrid nanoparticles as potential anti-hepatitis therapeutics. Biomaterials 2018; 187:66-80. [PMID: 30296739 DOI: 10.1016/j.biomaterials.2018.09.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/29/2018] [Indexed: 12/11/2022]
Abstract
Hepatocellular injury is the pathological hallmark of hepatitis and a crucial driver for the progression of liver diseases, while the treatment options are commonly restricted. Interleukin-22 (IL-22) has attracted special attention as a potent survival factor for hepatocytes that both prevents and repairs the injury of hepatocytes through activation of STAT3 signaling pathway. We hypothesized that the ability to generate potent expression of IL-22 locally for the treatment of severe hepatocellular injury in hepatitis was a promising strategy to enhance efficacy and overcome off-target effects. Accordingly, we developed a polypeptide penetratin-based hybrid nanoparticle system (PDPIA) carrying IL-22 gene by a self-assembly process. This nanocomplex modified with penetratin featured direct translocation across the cellular or endosomal membrane but mild zeta-potential to facilitate the high cellular internalization and endosomal escape of the gene cargos as well as scarcely Kupffer cells uptake. More importantly, PDPIA afforded preferential liver accumulation and predominant hepatocytes internalization following systemic administration, which showed pharmacologically suitable organ and sub-organ-selective properties. Subsequent studies confirmed a considerable protective role of PDPIA in a model of severe hepatitis induced by concanavalin A, evidenced by reduced hepatocellular injury and evaded immune response. The locally expressed IL-22 by PDPIA activated STAT3/Erk signal transduction, and thus promoted hepatocyte regeneration, inhibited reactive oxygen species (ROS) accumulation as well as prevented the dysfunction of mitochondrial. In addition, this system did not manifest side effects or systemic toxicity in mice. Collectively, the high versatility of PDPIA rendered its promising applications might be an effective agent to treat various hepatic disorders.
Collapse
Affiliation(s)
- Wei Chen
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Jingyun Luan
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Gang Wei
- Department of Pharmaceutics & The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xuyao Zhang
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Jiajun Fan
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Wenjing Zai
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Shaofei Wang
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Yichen Wang
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Yanxu Liang
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Yanyang Nan
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China
| | - Chuzhen Yin
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Yubin Li
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Ming-Lin Liu
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Dianwen Ju
- Department of Microbiological and Biochemical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, PR China.
| |
Collapse
|
33
|
Catuogno S, Esposito CL, Condorelli G, de Franciscis V. Nucleic acids delivering nucleic acids. Adv Drug Deliv Rev 2018; 134:79-93. [PMID: 29630917 DOI: 10.1016/j.addr.2018.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/20/2018] [Accepted: 04/03/2018] [Indexed: 01/07/2023]
Abstract
Nucleic acid therapeutics, including siRNAs, miRNAs/antimiRs, gRNAs and ASO, represent innovative and highly promising molecules for the safe treatment of a wide range of pathologies. The efficiency of systemic treatments is impeded by 1) the need to overcome physical and functional barriers in the organism, and 2) to accumulate in the intracellular active site at therapeutic concentrations. Although oligonucleotides either as modified naked molecules or complexed with delivery carriers have revealed to be effectively delivered to the affected target cells, this is restricted to topic treatments or to a few highly vascularized tissues. Therefore, the development of effective strategies for therapeutic nucleic acid selective delivery to target tissues is of primary importance in order to reduce the occurrence of undesired effects on non-target healthy tissues and to permit their translation to clinic. Due to their high affinity for specific ligands, high tissue penetration and chemical flexibility, short single-stranded nucleic acid aptamers are emerging as very attractive carriers for various therapeutic oligonucleotides. Yet, different aptamer-based bioconjugates, able to provide accumulation into target tissues, as well as efficient processing of therapeutic oligonucleotides, have been developed. In this respect, nucleic acid aptamer-mediated delivery strategies represent a powerful approach able to increase the therapeutic efficacy also highly reducing the overall toxicity. In this review, we will summarize recent progress in the field and discuss achieved objectives and optimization of aptamers as delivery carriers of short oligonucleotides.
Collapse
Affiliation(s)
- Silvia Catuogno
- Istituto di Endocrinologia ed Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Carla Lucia Esposito
- Istituto di Endocrinologia ed Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Gerolama Condorelli
- Istituto di Endocrinologia ed Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Vittorio de Franciscis
- Istituto di Endocrinologia ed Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy.
| |
Collapse
|
34
|
Sivakumar P, Kim S, Kang HC, Shim MS. Targeted siRNA delivery using aptamer-siRNA chimeras and aptamer-conjugated nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1543. [PMID: 30070426 DOI: 10.1002/wnan.1543] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/09/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022]
Abstract
The sequence-specific gene-silencing ability of small interfering RNA (siRNA) has been exploited as a new therapeutic approach for the treatment of a variety of diseases. However, efficient and safe delivery of siRNA into target cells is still a challenge in the clinical development of siRNA-based therapeutics. Recently, nucleic acid-based aptamers that target cell surface proteins have emerged as a new class of targeting moieties due to their high specificity and avidity. To date, various aptamer-mediated siRNA delivery systems have been developed to enhance the RNA interference (RNAi) efficacy of siRNA via targeted delivery. In this review, we summarize recent advances in developing aptamer-mediated siRNA delivery systems for RNAi therapeutics, mainly aptamer-siRNA chimeras and aptamer-functionalized nanocarriers incorporating siRNA, with a focus on their molecular designs and formulations. In addition, the challenges and engineering strategies of aptamer-mediated siRNA delivery systems for clinical translation are discussed. This article is categorized under: Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Padmanaban Sivakumar
- Division of Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Sumin Kim
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Han Chang Kang
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, Republic of Korea
| |
Collapse
|
35
|
Oh JH, Chong S, Nam S, Hyun S, Choi S, Gye H, Jang S, Jang J, Hwang SW, Yu J, Lee Y. Multimeric Amphipathic α-Helical Sequences for Rapid and Efficient Intracellular Protein Transport at Nanomolar Concentrations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800240. [PMID: 30128238 PMCID: PMC6096998 DOI: 10.1002/advs.201800240] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/04/2018] [Indexed: 06/08/2023]
Abstract
An amphipathic leucine (L) and lysine (K)-rich α-helical peptide is multimerized based on helix-loop-helix structures to maximize the penetrating activities. The multimeric LK-based cell penetrating peptides (LK-CPPs) can penetrate cells as protein-fused forms at 100-1000-fold lower concentrations than Tat peptide. The enhanced penetrating activity is increased through multimerization by degrees up to the tetramer level. The multimeric LK-CPPs show rapid cell penetration through macropinocytosis at low nanomolar concentrations, unlike the monomeric LK, which have slower penetrating kinetics at much higher concentrations. The heparan sulfate proteoglycan (HSPG) receptors are highly involved in the rapid internalization of multimeric LK-CPPs. As a proof of concept of biomedical applications, an adipogenic transcription factor, peroxisome proliferator-activated receptor gamma 2 (PPAR-γ 2), is delivered into preadipocytes, and highly enhanced expression of adipogenic genes at nanomolar concentrations is induced. The multimeric CPPs can be a useful platform for the intracellular delivery of bio-macromolecular reagents that have difficulty with penetration in order to control biological reactions in cells at feasible concentrations for biomedical purposes.
Collapse
Affiliation(s)
- Jae Hoon Oh
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Seung‐Eun Chong
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Sohee Nam
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Soonsil Hyun
- Department of Chemistry and EducationSeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Sejong Choi
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Hyojun Gye
- Department of Nano Science and Mechatronics EngineeringKonkuk University268 Chung Won Dae RoChungju‐CityChungbuk380‐701Republic of Korea
| | - Sangmok Jang
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Joomyung Jang
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Sung Won Hwang
- Department of Nano Science and Mechatronics EngineeringKonkuk University268 Chung Won Dae RoChungju‐CityChungbuk380‐701Republic of Korea
| | - Jaehoon Yu
- Department of Chemistry and EducationSeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| | - Yan Lee
- Department of ChemistrySeoul National University1 Gwanak‐roGwanak‐guSeoul08826Republic of Korea
| |
Collapse
|
36
|
Ye J, Pei X, Cui H, Yu Z, Lee H, Wang J, Wang X, Sun L, He H, Yang VC. Cellular uptake mechanism and comparative in vitro cytotoxicity studies of monomeric LMWP-siRNA conjugate. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2018.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Wang HX, Song Z, Lao YH, Xu X, Gong J, Cheng D, Chakraborty S, Park JS, Li M, Huang D, Yin L, Cheng J, Leong KW. Nonviral gene editing via CRISPR/Cas9 delivery by membrane-disruptive and endosomolytic helical polypeptide. Proc Natl Acad Sci U S A 2018; 115:4903-4908. [PMID: 29686087 PMCID: PMC5948953 DOI: 10.1073/pnas.1712963115] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Effective and safe delivery of the CRISPR/Cas9 gene-editing elements remains a challenge. Here we report the development of PEGylated nanoparticles (named P-HNPs) based on the cationic α-helical polypeptide poly(γ-4-((2-(piperidin-1-yl)ethyl)aminomethyl)benzyl-l-glutamate) for the delivery of Cas9 expression plasmid and sgRNA to various cell types and gene-editing scenarios. The cell-penetrating α-helical polypeptide enhanced cellular uptake and promoted escape of pCas9 and/or sgRNA from the endosome and transport into the nucleus. The colloidally stable P-HNPs achieved a Cas9 transfection efficiency up to 60% and sgRNA uptake efficiency of 67.4%, representing an improvement over existing polycation-based gene delivery systems. After performing single or multiplex gene editing with an efficiency up to 47.3% in vitro, we demonstrated that P-HNPs delivering Cas9 plasmid/sgRNA targeting the polo-like kinase 1 (Plk1) gene achieved 35% gene deletion in HeLa tumor tissue to reduce the Plk1 protein level by 66.7%, thereby suppressing the tumor growth by >71% and prolonging the animal survival rate to 60% within 60 days. Capable of delivering Cas9 plasmids to various cell types to achieve multiplex gene knock-out, gene knock-in, and gene activation in vitro and in vivo, the P-HNP system offers a versatile gene-editing platform for biological research and therapeutic applications.
Collapse
Affiliation(s)
- Hong-Xia Wang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Champaign, IL 61801
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Xin Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
- Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jing Gong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Du Cheng
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Syandan Chakraborty
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Ji Sun Park
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Mingqiang Li
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Dantong Huang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou 215123, China;
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
- Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Champaign, IL 61801;
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027;
| |
Collapse
|
38
|
Anderson AJ, Peters EB, Neumann A, Wagner J, Fairbanks B, Bryant SJ, Bowman CN. Cytocompatibility and Cellular Internalization of PEGylated "Clickable" Nucleic Acid Oligomers. Biomacromolecules 2018; 19:2535-2541. [PMID: 29698604 DOI: 10.1021/acs.biomac.8b00162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The recently developed synthetic oligonucleotides referred to as "click" nucleic acids (CNAs) are promising due to their relatively simple synthesis based on thiol-X reactions with numerous potential applications in biotechnology, biodetection, gene silencing, and drug delivery. Here, the cytocompatibility and cellular uptake of rhodamine tagged, PEGylated CNA copolymers (PEG-CNA-RHO) were evaluated. NIH 3T3 fibroblast cells treated for 1 h with 1, 10, or 100 μg/mL PEG-CNA-RHO maintained an average cell viability of 86%, which was not significantly different from the untreated control. Cellular uptake of PEG-CNA-RHO was detected within 30 s, and the amount internalized increased over the course of 1 h. Moreover, these copolymers were internalized within cells to a higher degree than controls consisting of either rhodamine tagged PEG or the rhodamine alone. Uptake was not affected by temperature (i.e., 4 or 37 °C), suggesting a passive uptake mechanism. Subcellular colocalization analysis failed to indicate significant correlations between the internalized PEG-CNA-RHO and the organelles examined (mitochondria, endoplasmic reticulum, endosomes and lysosomes). These results indicate that CNA copolymers are cytocompatible and are readily internalized by cells, supporting the idea that CNAs are a promising alternative to DNA in antisense therapy applications.
Collapse
|
39
|
Chen B, Yoo K, Xu W, Pan R, Han XX, Chen P. Characterization and evaluation of a peptide-based siRNA delivery system in vitro. Drug Deliv Transl Res 2018; 7:507-515. [PMID: 28349343 DOI: 10.1007/s13346-017-0371-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Since its inception more than a decade ago, gene silencing mediated by double-stranded small interfering RNA (siRNA) has been widely investigated as a potential therapeutic approach for a variety of diseases. However, the use of siRNA is hampered by its rapid degradation and poor cellular uptake in vitro and in vivo. Recently, peptide-based carriers have been applied to siRNA delivery, as an alternative to the traditional delivery systems. Here, a histidine-containing amphipathic amino acid pairing peptide, C6M3, which can form complexes with siRNA, was used as a new siRNA delivery system. This peptide exhibited a high affinity for siRNA and ability to efficiently deliver siRNA into the cells. The interaction of C6M3 with siRNA was investigated to determine the loading capacity of C6M3 at different peptide/siRNA molar ratios. At C6M3/siRNA molar ratio of 10/1, siRNA molecules were entirely associated with C6M3 as indicated by a gel electrophoretic assay and further confirmed by zeta potential analysis. The particle size distribution of the C6M3-siRNA complexes was studied using dynamic light scattering, which showed an intensity-based size distribution peaked approximately at 100 nm in RNase-free water and 220 nm in the Opti-MEM medium. C6M3 adopted a helical secondary structure in RNase-free water and became more so after forming complexes with siRNA. The interaction of siRNA with C6M3 is an entropy-driven spontaneous process, as determined by isothermal titration calorimetry (ITC) study. The efficiency of cellular uptake of the siRNA complexes at different C6M3/siRNA molar ratios was evaluated, and the results showed that C6M3 promoted efficient cellular uptake of siRNA into cells. Furthermore, a significant level of GAPDH gene silencing efficiency (69%) was achieved in CHO-K1 cells, with minimal cytotoxicity.
Collapse
Affiliation(s)
- Baoling Chen
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Kimoon Yoo
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Wen Xu
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Ran Pan
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Xiao Xia Han
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - P Chen
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada. .,Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.
| |
Collapse
|
40
|
Tomassi S, Ieranò C, Mercurio ME, Nigro E, Daniele A, Russo R, Chambery A, Baglivo I, Pedone PV, Rea G, Napolitano M, Scala S, Cosconati S, Marinelli L, Novellino E, Messere A, Di Maro S. Cationic nucleopeptides as novel non-covalent carriers for the delivery of peptide nucleic acid (PNA) and RNA oligomers. Bioorg Med Chem 2018; 26:2539-2550. [PMID: 29656988 DOI: 10.1016/j.bmc.2018.04.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
Cationic nucleopeptides belong to a family of synthetic oligomers composed by amino acids and nucleobases. Their capability to recognize nucleic acid targets and to cross cellular membranes provided the basis for considering them as novel non-covalent delivery agents for nucleic acid pharmaceuticals. Herein, starting from a 12-mer nucleopeptide model, the number of cationic residues was modulated in order to obtain new nucleopeptides endowed with high solubility in acqueous medium, acceptable bio-stability, low cytotoxicity and good capability to bind nucleic acid. Two candidates were selected to further investigate their potential as nucleic acid carriers, showing higher efficiency to deliver PNA in comparison with RNA. Noteworthy, this study encourages the development of nucleopeptides as new carriers to extend the known strategies for those nucleic acid analogues, especially PNA, that still remain difficult to drive into the cells.
Collapse
Affiliation(s)
- Stefano Tomassi
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Caterina Ieranò
- Molecular Immunology and Immunoregulation, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS-Napoli, 80131 Naples, Italy
| | - Maria Emilia Mercurio
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ersilia Nigro
- Department of Cardiothoracic and Respiratory Sciences, University of Campania "Luigi Vanvitelli", Via Leonardo Bianchi c/o Ospedale Monaldi, 80131 Naples, Italy
| | - Aurora Daniele
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy; Ceinge-Biotecnologie Avanzate S.c.a r.l., Via G. Salvatore 486, 80145 Napoli, Italy
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ilaria Baglivo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Paolo Vincenzo Pedone
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Giuseppina Rea
- Molecular Immunology and Immunoregulation, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS-Napoli, 80131 Naples, Italy
| | - Maria Napolitano
- Molecular Immunology and Immunoregulation, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS-Napoli, 80131 Naples, Italy
| | - Stefania Scala
- Molecular Immunology and Immunoregulation, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS-Napoli, 80131 Naples, Italy
| | - Sandro Cosconati
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Luciana Marinelli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Anna Messere
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy.
| | - Salvatore Di Maro
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy.
| |
Collapse
|
41
|
Grasso G, Muscat S, Rebella M, Morbiducci U, Audenino A, Danani A, Deriu MA. Cell penetrating peptide modulation of membrane biomechanics by Molecular dynamics. J Biomech 2018; 73:137-144. [PMID: 29631749 DOI: 10.1016/j.jbiomech.2018.03.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 11/17/2022]
Abstract
The efficacy of a pharmaceutical treatment is often countered by the inadequate membrane permeability, that prevents drugs from reaching their specific intracellular targets. Cell penetrating peptides (CPPs) are able to route across cells' membrane various types of cargo, including drugs and nanoparticles. However, CPPs internalization mechanisms are not yet fully understood and depend on a wide variety of aspects. In this contest, the entry of a CPP into the lipid bilayer might induce molecular conformational changes, including marked variations on membrane's mechanical properties. Understanding how the CPP does influence the mechanical properties of cells membrane is crucial to design, engineer and improve new and existing penetrating peptides. Here, all atom Molecular Dynamics (MD) simulations were used to investigate the interaction between different types of CPPs embedded in a lipid bilayer of dioleoyl phosphatidylcholine (DOPC). In a greater detail, we systematically highlighted how CPP properties are responsible for modulating the membrane bending modulus. Our findings highlighted the CPP hydropathy strongly correlated with penetration of water molecules in the lipid bilayer, thus supporting the hypothesis that the amount of water each CPP can route inside the membrane is modulated by the hydrophobic and hydrophilic character of the peptide. Water penetration promoted by CPPs leads to a local decrease of the lipid order, which emerges macroscopically as a reduction of the membrane bending modulus.
Collapse
Affiliation(s)
- Gianvito Grasso
- Istituto Dalle Molle di Studi sull'Intelligenza Artificiale (IDSIA), Scuola universitaria professionale della Svizzera italiana (SUPSI), Università della Svizzera Italiana (USI), Centro Galleria 2, Manno CH-6928, Switzerland
| | - Stefano Muscat
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, IT-10128 Torino, Italy
| | - Martina Rebella
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, IT-10128 Torino, Italy
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, IT-10128 Torino, Italy
| | - Alberto Audenino
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, IT-10128 Torino, Italy
| | - Andrea Danani
- Istituto Dalle Molle di Studi sull'Intelligenza Artificiale (IDSIA), Scuola universitaria professionale della Svizzera italiana (SUPSI), Università della Svizzera Italiana (USI), Centro Galleria 2, Manno CH-6928, Switzerland
| | - Marco A Deriu
- Istituto Dalle Molle di Studi sull'Intelligenza Artificiale (IDSIA), Scuola universitaria professionale della Svizzera italiana (SUPSI), Università della Svizzera Italiana (USI), Centro Galleria 2, Manno CH-6928, Switzerland.
| |
Collapse
|
42
|
Perepelyuk M, Sacko K, Thangavel K, Shoyele SA. Evaluation of MUC1-Aptamer Functionalized Hybrid Nanoparticles for Targeted Delivery of miRNA-29b to Nonsmall Cell Lung Cancer. Mol Pharm 2018; 15:985-993. [PMID: 29432024 DOI: 10.1021/acs.molpharmaceut.7b00900] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The objective of this study was to evaluate the therapeutic efficacy and pharmacokinetic study of mucin1-aptamer functionalized miRNA-29b-loaded hybrid nanoparticles (MAFMILHNs) in lung tumor-bearing SCID mice. MAFMILHNs were manufactured using an isoelectric point based nanotechnology. They were then fully characterized for particle size, loading capacity, zeta potential, and encapsulation efficiency. The ability of MAFMILHNs to downregulate oncoprotein DNMT3B both at the cellular level and in vivo was monitored using Western blot, while the effect of the downregulation of DNMT3B on tumor growth was assessed using bioluminescence. Results indicate that the presence of MUC1-aptamer conjugated to the surface of the nanoparticles enhanced the selective delivery of miRNA-29b to tumor cells and tissues. Further, the downregulation of DNMT3B by MAFMILHNs resulted in the inhibition of tumor growth in mouse models.
Collapse
Affiliation(s)
- Maryna Perepelyuk
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| | - Koita Sacko
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| | - Karthik Thangavel
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| | - Sunday A Shoyele
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| |
Collapse
|
43
|
Gagat M, Zielińska W, Grzanka A. Cell-penetrating peptides and their utility in genome function modifications (Review). Int J Mol Med 2017; 40:1615-1623. [PMID: 29039455 PMCID: PMC5716439 DOI: 10.3892/ijmm.2017.3172] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/26/2017] [Indexed: 01/02/2023] Open
Abstract
For almost 30 years, studies have confirmed the effectiveness of cell-penetrating peptides (CPPs) in the facilitation of the intracellular delivery of various cargo molecules, including RNA, DNA, plasmids, proteins or nanoparticles, under in vitro and in vivo conditions. The cellular uptake of CPPs occurs via energy-dependent, as well as -independent mechanisms. In this relatively new direction of research, studies have attempted to introduce genome modification systems into cells by CPPs. Cellular uptake of CPPs carrying either covalently bound or electrostatically conjugated cargo, has several advantages over viral delivery systems, as it does not lead to any significant cytotoxicity or immunogenicity, and simultaneously it is more efficient than other non-viral systems. So far, CPPs have been successfully used to introduce Cre recombinase, zinc finger nucleases, transcription activator-like effector nucleases and clustered regularly interspaced short palindromic repeats systems into cells. The present article systematically reviewed the information obtained from studies on CPPs and assessed their utility with regard to their effectiveness and safety of use.
Collapse
Affiliation(s)
- Maciej Gagat
- Department of Histology and Embryology, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Pl-85-092 Bydgoszcz, Poland
| | - Wioletta Zielińska
- Department of Histology and Embryology, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Pl-85-092 Bydgoszcz, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Pl-85-092 Bydgoszcz, Poland
| |
Collapse
|
44
|
Abstract
The advent of RNA interference (RNAi) technology has profoundly impacted molecular biology research and medicine but has also advanced the field of skin care. Both effector molecules of RNAi, short-interfering RNA molecules and microRNAs (miRNAs), have been explored for their relative impact and utility for treating a variety of skin conditions. These post-transcriptional RNA regulatory molecules down-modulate protein expression through targeting of the 3' untranslated regions of messenger RNAs, leading to their degradation or repression through sequestration. As researchers hunt for genetic linkages to skin diseases, miRNA regulators have emerged as key players in the biology of keratinocytes, fibroblasts, melanocytes, and other cells of the skin. Herein, we attempt to coalesce the current efforts to combat various skin disorders and diseases through the development of miRNA-based technologies.
Collapse
Affiliation(s)
- Paul Lawrence
- Biocogent, LLC, 25 Health Sciences Drive, Stony Brook, NY 11790 USA
| | - Joseph Ceccoli
- Biocogent, LLC, 25 Health Sciences Drive, Stony Brook, NY 11790 USA
| |
Collapse
|
45
|
Tian Y, Zeng X, Li J, Jiang Y, Zhao H, Wang D, Huang X, Li Z. Achieving enhanced cell penetration of short conformationally constrained peptides through amphiphilicity tuning. Chem Sci 2017; 8:7576-7581. [PMID: 29568420 PMCID: PMC5848794 DOI: 10.1039/c7sc03614k] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/12/2017] [Indexed: 12/28/2022] Open
Abstract
We synthesized a panel of conformationally constrained peptides with either α-helix or β-hairpin conformations. We tuned the amphiphilicity of these constrained peptides with different distributions of charged or hydrophobic residues and compared their cellular uptake efficiencies in different cell lines.
Due to their enhanced stability and cell permeability, cyclic cell-penetrating peptides have been widely used as delivery vectors for transporting cell-impermeable cargos into cells. In this study, we synthesized a panel of conformationally constrained peptides with either α-helix or β-hairpin conformations. We tuned the amphiphilicity of these constrained peptides with different distributions of charged or hydrophobic residues and compared their cellular uptake efficiencies in different cell lines. We found that the amphipathicity of these conformationally constrained peptides correlates well with their cellular uptake efficiency. We proposed that peptides with larger hydrophobic moments (HMs) have stronger binding affinities with the cell membrane which further accelerates the endocytosis process. This finding should provide an approach towards the design of more potent conformationally constrained cell-penetrating peptides for biomedical applications.
Collapse
Affiliation(s)
- Yuan Tian
- School of Chemical Biology and Biotechnology , Shenzhen Graduate School of Peking University , Shenzhen , 518055 , China . .,School of Life Science and Engineering , Southwest Jiaotong University , Chengdu , 610031 , China
| | - Xiangze Zeng
- Department of Chemistry , Center of Systems Biology and Human Health , School of Science and Institute for Advance Study , The Hong Kong University of Science and Technology , Clear Water Bay , Kowloon , Hong Kong .
| | - Jingxu Li
- School of Chemical Biology and Biotechnology , Shenzhen Graduate School of Peking University , Shenzhen , 518055 , China .
| | - Yanhong Jiang
- School of Chemical Biology and Biotechnology , Shenzhen Graduate School of Peking University , Shenzhen , 518055 , China .
| | - Hui Zhao
- School of Chemical Biology and Biotechnology , Shenzhen Graduate School of Peking University , Shenzhen , 518055 , China .
| | - Dongyuan Wang
- School of Chemical Biology and Biotechnology , Shenzhen Graduate School of Peking University , Shenzhen , 518055 , China .
| | - Xuhui Huang
- Department of Chemistry , Center of Systems Biology and Human Health , School of Science and Institute for Advance Study , The Hong Kong University of Science and Technology , Clear Water Bay , Kowloon , Hong Kong .
| | - Zigang Li
- School of Chemical Biology and Biotechnology , Shenzhen Graduate School of Peking University , Shenzhen , 518055 , China .
| |
Collapse
|
46
|
Chung JY, Ul Ain Q, Lee HL, Kim SM, Kim YH. Enhanced Systemic Anti-Angiogenic siVEGF Delivery Using PEGylated Oligo-d-arginine. Mol Pharm 2017; 14:3059-3068. [DOI: 10.1021/acs.molpharmaceut.7b00282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jee Young Chung
- Department
of Bioengineering, Institute for Bioengineering and Biopharmaceutical
Research, and ‡BK 21 Plus Future Biopharmaceutical Human Resources Training and
Research Team, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - Qurrat Ul Ain
- Department
of Bioengineering, Institute for Bioengineering and Biopharmaceutical
Research, and ‡BK 21 Plus Future Biopharmaceutical Human Resources Training and
Research Team, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - Hyun Lin Lee
- Department
of Bioengineering, Institute for Bioengineering and Biopharmaceutical
Research, and ‡BK 21 Plus Future Biopharmaceutical Human Resources Training and
Research Team, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - So-Mi Kim
- Department
of Bioengineering, Institute for Bioengineering and Biopharmaceutical
Research, and ‡BK 21 Plus Future Biopharmaceutical Human Resources Training and
Research Team, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - Yong-Hee Kim
- Department
of Bioengineering, Institute for Bioengineering and Biopharmaceutical
Research, and ‡BK 21 Plus Future Biopharmaceutical Human Resources Training and
Research Team, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| |
Collapse
|
47
|
Nanoparticles and targeted drug delivery in cancer therapy. Immunol Lett 2017; 190:64-83. [PMID: 28760499 DOI: 10.1016/j.imlet.2017.07.015] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022]
Abstract
Surgery, chemotherapy, radiotherapy, and hormone therapy are the main common anti-tumor therapeutic approaches. However, the non-specific targeting of cancer cells has made these approaches non-effective in the significant number of patients. Non-specific targeting of malignant cells also makes indispensable the application of the higher doses of drugs to reach the tumor region. Therefore, there are two main barriers in the way to reach the tumor area with maximum efficacy. The first, inhibition of drug delivery to healthy non-cancer cells and the second, the direct conduction of drugs into tumor site. Nanoparticles (NPs) are the new identified tools by which we can deliver drugs into tumor cells with minimum drug leakage into normal cells. Conjugation of NPs with ligands of cancer specific tumor biomarkers is a potent therapeutic approach to treat cancer diseases with the high efficacy. It has been shown that conjugation of nanocarriers with molecules such as antibodies and their variable fragments, peptides, nucleic aptamers, vitamins, and carbohydrates can lead to effective targeted drug delivery to cancer cells and thereby cancer attenuation. In this review, we will discuss on the efficacy of the different targeting approaches used for targeted drug delivery to malignant cells by NPs.
Collapse
|
48
|
Zhao L, Wu X, Wang X, Duan C, Wang H, Punjabi A, Zhao Y, Zhang Y, Xu Z, Gao H, Han G. Development of Excipient-Free Freeze-Dryable Unimolecular Hyperstar Polymers for Efficient siRNA Silencing. ACS Macro Lett 2017; 6:700-704. [PMID: 35650873 DOI: 10.1021/acsmacrolett.7b00242] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We designed a unimolecular hyperstar polymer for efficient small interfering RNA (siRNA) delivery that can be processed under repeated lyophilization and reconstitution without the need of any excipient. The hyperstar polymer contains a biodegradable hyperbranched core and is bound to siRNA through its thousands of cationic arms that radiate from its core. The siRNA/hyperstar complexes showed siRNA transfection efficiency that was superior to that of Lipofectamine2000 in regard to the gene for human Cu, Zn superoxide dismutase 1 (SOD1), whose mutation causes familial amyotrophic lateral sclerosis. More importantly, hyperstar polymers as unimolecular containers minimized the multipolymer cross-interaction during lyophilization, and this maintained the uniquely high transfection efficiency of the siRNA/hyperstar complexes after repeated freeze-drying and reconstitution without the conventional need for excipient.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States.,State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116012, People's Republic of China
| | - Xiang Wu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Xiaofeng Wang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Chunying Duan
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116012, People's Republic of China
| | - Hongyan Wang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Amol Punjabi
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Yang Zhao
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Yuanwei Zhang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Zuoshang Xu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Haifeng Gao
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Gang Han
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
49
|
Ye J, Liu E, Gong J, Wang J, Huang Y, He H, Yang VC. High-Yield Synthesis of Monomeric LMWP(CPP)-siRNA Covalent Conjugate for Effective Cytosolic Delivery of siRNA. Am J Cancer Res 2017; 7:2495-2508. [PMID: 28744330 PMCID: PMC5525752 DOI: 10.7150/thno.19863] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/17/2017] [Indexed: 12/22/2022] Open
Abstract
Because of the unparalleled efficiency and universal utility in treating a variety of disease types, siRNA agents have evolved as the future drug-of-choice. Yet, the inability of the polyanionic siRNA macromolecules to cross the cell membrane remains as the bottleneck of possible clinical applications. With the cell penetrating peptides (CPP) being discovered lately, the most effective tactic to achieve the highest intracellular siRNA delivery deems to be by covalently conjugating the drug to a CPP; for instance, the arginine-rich Tat or low molecular weight protamine (LMWP) peptides. However, construction of such a chemical conjugate has been referred by scientists in this field as the “Holy Grail” challenge due to self-assembly of the cationic CPP and anionic siRNA into insoluble aggregates that are deprived of the biological functions of both compounds. Based on the dynamic motion of PEG, we present herein a concise coupling strategy that is capable of permitting a high-yield synthesis of the cell-permeable, cytosol-dissociable LMWP-siRNA covalent conjugates. Cell culture assessment demonstrates that this chemical conjugate yields by far the most effective intracellular siRNA delivery and its corresponded gene-silencing activities. This work may offer a breakthrough advance towards realizing the clinical potential of all siRNA therapeutics and, presumably, most anionic macromolecular drugs such as anti-sense oligonucleotides, gene compounds, DNA vectors and proteins where conjugation with the CPP encounters with problems of aggregation and precipitation. To this end, the impact of this coupling technique is significant, far-reaching and wide-spread.
Collapse
|
50
|
Tambunan USF, Alkaff AH, Nasution MAF, Parikesit AA, Kerami D. Screening of commercial cyclic peptide conjugated to HIV-1 Tat peptide as inhibitor of N-terminal heptad repeat glycoprotein-2 ectodomain Ebola virus through in silico analysis. J Mol Graph Model 2017; 74:366-378. [PMID: 28482272 DOI: 10.1016/j.jmgm.2017.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/04/2017] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
Abstract
Ebola Hemorrhagic Fever (EHF) is a disease caused by viruses from genus Ebolavirus. Zaire ebolavirus (EBOV) is the deadliest species which has 76% case fatality rate. Up until now, there is no U.S. Food and Drug Administration (FDA) approved drugs to treat EHF. Antiviral drug based on EBOV N-terminal heptad repeat glycoprotein-2 (NHR GP2) Ectodomain inhibitor is one kind of treatment that has not well developed. NHR GP2 Ectodomain has an important role in the process of EBOV entry into the cell through endocytosis mechanism. In this study, we used in silico methods to investigate the activity of commercial cyclic peptide conjugated to Human Immunodeficiency Virus type 1 Trans-activator of the transcription (HIV-1 Tat) peptide as ligands which act as an inhibitor of EBOV NHR GP2 Ectodomain. The commercial cyclic peptides which we used in this study were obtained from the selected chemical companies. Conjugation of the commercial cyclic peptides to HIV-1 Tat peptide was done in order to accumulate it inside the endosome. The ligands which had the best inhibition properties were screened using molecular docking and molecular dynamics simulation. Prediction of pharmacological properties of the peptides was done to choose the best drug candidate. The result of screening processes shows that Ligand 023 has the highest potency as the drug lead. The ligand needs to undergo further analysis through in vitro, in vivo, and a clinical trial to ensure that this ligand has a therapeutic ability as an antiviral drug for Ebola virus infection.
Collapse
Affiliation(s)
- Usman Sumo Friend Tambunan
- Bioinformatics Research Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Indonesia, Depok 16424, Indonesia.
| | - Ahmad Husein Alkaff
- Bioinformatics Research Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Indonesia, Depok 16424, Indonesia
| | - Mochammad Arfin Fardiansyah Nasution
- Bioinformatics Research Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Indonesia, Depok 16424, Indonesia
| | - Arli Aditya Parikesit
- Bioinformatics Research Group, Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Indonesia, Depok 16424, Indonesia
| | - Djati Kerami
- Mathematics Computation Research Group, Department of Mathematics, Faculty of Mathematics and Natural Science, Universitas Indonesia, Depok 1624, Indonesia
| |
Collapse
|