Abstract

Small molecule, peptide, and protein-based drugs have been developed over decades to treat various diseases. The importance of gene therapy as an alternative to traditional drugs has increased after the discovery of gene-based drugs such as Gendicine for cancer and Neovasculgen for peripheral artery disease. Since then, the pharma sector is focusing on developing gene-based drugs for various diseases. After the discovery of the RNA interference (RNAi) mechanism, the development of siRNA-based gene therapy has been accelerated immensely. siRNA-based treatment for hereditary transthyretin-mediated amyloidosis (hATTR) using Onpattro and acute hepatic porphyria (AHP) by Givlaari and three more FDA-approved siRNA drugs has set up a milestone and further improved the confidence for the development of gene therapeutics for a spectrum of diseases. siRNA-based gene drugs have more advantages over other gene therapies and are under study to treat different types of diseases such as viral infections, cardiovascular diseases, cancer, and many more. However, there are a few bottlenecks to realizing the full potential of siRNA-based gene therapy. They include chemical instability, nontargeted biodistribution, undesirable innate immune responses, and off-target effects. This review provides a comprehensive view of siRNA-based gene drugs: challenges associated with siRNA delivery, their potential, and future prospects.
1. Introduction
Gene therapy is one of the most advanced biotechnologies in medicine.1 Gene therapy is defined as the correction of defective/disease-causing gene/s by transferring genetic medicine such as DNA/RNA to a patient’s cells to deactivate, replace, or repair.2,3 The first gene manipulation was attempted as early as 1989 when human DNA is modified by direct insertion of DNA into the nuclear genome.4 Later on, several scientific groups developed various types of gene-based drugs.5 The nucleic acid-based gene drugs broadly can be categorized into DNA, RNA, and artificial nucleotide (XNA)-based drugs. DNA-based drugs include plasmids, antisense oligonucleotides based on DNA, DNA aptamers, and DNAzymes.6−8 Likewise, RNA-based drugs can be further categorized as RNAi (viz., siRNA and miRNA), shRNA, mRNA, RNA-based antisense oligonucleotides, ribozyme, and aptamer-based drugs.7,9,10 XNA-based gene drugs mainly fall under antisense oligonucleotide-based drugs.11 CRISPR/Cas9 system, chimeric antigen receptor T (CAR T) cell therapy for cancer, and stem cell therapy also come under gene therapy.12 Gene therapy mainly works by three methods: (i) Introducing a healthy copy of a gene in place of a disease-causing gene, (ii) inactivation of a nonfunctioning disease-causing gene, and (iii) treating disease by introducing a new copy of a modified gene into the body.13 The nucleic acid-based gene drugs are studied to treat several diseases including cancer, viral infections, heart diseases, and rare genetic diseases.14−16 By 2021, more than 2600 gene therapy trials are going on worldwide.17,18 For example, Luxturna is a DNA-based gene drug to treat inherited retinal disease by targeting retinal pigment epithelium-specific 65 (RPE65),19 Spinraza is an antisense oligonucleotide to treat spinal muscular atrophy by correcting the Survival of motor neuron 2 (SMN2) gene,20 Patisiran is a siRNA-based gene-drug to treat familial amyloidotic polyneuropathy by targeting Transthyretin (TTR) gene,21 Givosiran is also a siRNA-based gene-drug to cure acute hepatic porphyria by targeting δ-aminolevulinate synthase 1 (ALAS1),22 and Golodirsen is an antisense oligonucleotide-based gene-drug to treat Duchenne muscular dystrophy (DMD) by targeting the mutated dystrophin gene.23 The single-stranded antisense oligos silences a gene (mRNA) by following an RNaseH-dependent mRNA cleavage mechanism.24 Kynamro is an another antisense oligonucleotide-based gene-drug to treat homozygous familial hypercholesterolemia by deactivating the apolipoprotein B-100 (apoB-100) gene translation.25 Besides, cell-based therapies are also considered as gene therapy, for example, Yescarta is a CAR-T-based gene therapy where T-cells are genetically engineered and are used to treat B-cell cancer.26 These are a few examples of approved gene drugs. There are almost 39 gene drugs that have been approved by 2022 and many more are in clinical trials at different levels.27 However, siRNA-based gene therapy got more attention due to the ease of siRNA production and characterization. siRNAs are smaller in size, hence easy to modify chemically to improve their nuclease stability, and can be produced in large quantities by synthetic means.28 Besides, siRNA-based gene drugs are widely studied for various diseases; moreover, five siRNA-based drugs have already been approved by FDA for the treatment of various diseases like hereditary transthyretin amyloidosis (hATTR), acute hepatic porphyria (AHP), primary hyperoxaluria type 1 (PH1), primary hypercholesterolaemia, and amyloid transthyretin-mediated (ATTR) amyloidosis.29−33 Thus, this review mainly focuses on siRNA-based gene therapy and its potential, challenges, and prospects.
2. RNAi Mechanism
The discovery of the RNAi mechanism won the Noble Prize in Physiology in the year 2006. Most eukaryotic cells use the regulatory RNAi mechanism to defend themselves against viruses and transposons by developing small RNAs such as small interfering RNAs (siRNAs) and micro RNAs (miRNAs) naturally.7 The siRNAs and miRNAs are central to the RNAi mechanism. Particularly, the siRNAs are produced from endonuclease cleavage of the double-stranded noncoding small hairpin (shRNAs) by Dicer (an RNase III endonuclease). Dicer is an important enzyme and is responsible for the processing and handover of the siRNAs to Argonaute-2 (Ago-2). Ago-2 unwinds and removes the “passenger” strand while leaving the “guide” strand on Ago-2 to form an activated RISC as shown in Figure 1, following the natural siRNA mechanism.34 The breathing ends of the siRNAs are responsible for the selective loading of them into RISC. The less thermodynamically stable end of the passenger strand binds to the PIWI domain of Ago-2 and is cleaved by the Ago-2.35 The activated RISC with a guide strand specifically binds to the target mRNA through Watson–Crick complementarity. The guide strand of siRNA binding to mRNAs results in mRNA cleavage by the RISC complex, thus deactivation of translation. Therefore, siRNA is a widely used research tool both in vitro and in vivo as the siRNAs introduced to cells can suppress the gene of interest with high selectivity and efficacy. It has become evident from various studies that siRNA-based therapy is a precise, efficient, and stable gene therapy to treat various diseases.36 However, siRNA-based therapy has its challenges to translate as an entirely acceptable gene therapeutic.37
Figure 1.
Schematic representation of the natural siRNA mechanism and target mRNA degradation.
3. Physiological and Intracellular Challenges in siRNA Delivery
There are various physiological and intracellular barriers to overcome to realize the full potential of siRNA-based gene therapeutics. After systemic administration of the siRNA formulation, it must reach the target site by avoiding renal clearance, phagocyte uptake, interaction with serum proteins, and nuclease degradation. Various siRNA modifications are studied to overcome the problems.38 Though, the siRNA modification whether they are naked siRNAs, nonviral vectors, or viral vectors exhibits high efficiency in vitro but face a chain of obstacles in vivo before they reach the target cells. The siRNAs can be administered topically or intravenously depending upon access to the site of disease.
3.1. Route of Administration
Topical administration: The siRNA-based drugs are initially administrated topically to treat ocular pain and dry eye disease. However, topical administration is possible only for external tissues such as the eye and epidermis.39,40
Intravenous administration: Administration of siRNA through an intravenous (IV) is one of the choices when topical administration is not possible. For example, a vast majority of tumors and diseases in humans are not accessible for topical administration where IV administration is only preferable.41 siRNAs can be administered through the subcutaneous,42 intrathecal (IT),43 and intracerebroventricular (ICV) routes as well.43
3.2. Physiological Barriers
Most of the drugs need to overcome and cross all physiological barriers before reaching the site of action. There are several types of physiological barriers such as the dermal barrier, nasal barrier, and intestinal barriers, but when it comes to siRNA delivery, the main physiological barriers are nuclease stability in serum, renal clearance, and reticuloendothelial system (RES) as the siRNA administration is mostly intravenous.
3.2.1. Nuclease Cleavage
Unmodified siRNAs are unstable in biological fluids due to their degradation by exonucleases such as RNases. Nuclease degradation is one of the biological barriers the siRNA encounters in plasma and tissues. Exonuclease is one of the major nucleases present in plasma. The half-life of the naked siRNAs is about 5–10 min.44
3.2.2. Renal Clearance
Renal clearance is another problem that naked siRNAs face. Systemic delivery of siRNA of the naked siRNAs are very susceptible to renal clearance due to their smaller size, which reduces their half-life in the blood. Various biodistribution studies in animals showed that the majority of the naked siRNAs accumulated in the kidneys.45
3.2.3. Reticuloendothelial System (RES)
Besides the aforementioned physiological barriers, the uptake of siRNA by RES is another major barrier to overcome for efficient siRNA delivery in vivo. The RES is made-up of phagocytic cells. The macrophages which are part of phagocytic cells are highly abundant in the liver and spleen and result in a high accumulation of siRNA in these organs.46
3.3. Intracellular Barriers
Even after crossing the physiological barriers, the siRNA also must overcome the intracellular barriers to reach the cytoplasm of the cells, thus to exert the RNAi mechanism and the knockdown of the target mRNA. The intracellular barriers are endosomal trapping, immune stimulation, and off-target effects.38
3.3.1. Endosomal Trapping
Naked siRNA is anionic and hydrophilic that cannot be readily taken up by cells due to repulsion by the anionic cell surface.47 Though delivery vehicles such as nanoparticles are employed to deliver siRNAs to target cells, the nanoparticles remain trapped inside endosomes, leading to siRNA degradation. Therefore, nanoparticles must be accompanied by an endosomal escape strategy to deliver siRNA into the cell cytoplasm.38
3.3.2. Immune Stimulation
Immune stimulation is another challenge to address in order to realize the full potential of siRNA therapy. Innate immune stimulation is the body’s first response against germs and foreign substances entering the body.48 Therefore, innate immune stimulation is possible if too much siRNA is used, and it might be activated through the double-stranded RNA (dsRNA) sensor. GC-rich sequences in siRNA might activate nuclear factor kappa B (NF-κB), interferon regulatory factors, and toll-like receptors (TLR7, TLR8, and TLR9).49
3.3.3. Off-target Effects
As the siRNA sequences are short, there is a possibility for off-target binding. For example, a gene with shared strong homology to the target gene would potentially be knocked down and can lead to severe unintended side effects.50
4. Methods of siRNA Delivery
The naked siRNA as such cannot be used as a drug candidate for clinical applications due to their physiological and intracellular barriers. Various groups worked on addressing the challenges and could overcome some of the hurdles associated with siRNA delivery. Different methods are developed for safe siRNA delivery for clinical use; they mainly include (i) lipid nanoparticle (LNP), (ii) siRNA-ligand conjugates, (iii) polymer-based nanoparticles, (iv) viral vectors, (v) cell vesicles, and (vi) nucleic acid-based nanoparticles.
4.1. Lipid Nanoparticles (LNP)-Based siRNA Delivery
Lipid nanoparticles are spherical in shape and are composed of a lipid bilayer with an aqueous core, and the liposomal membrane is either positively charged, negatively charged, or neutral. The commonly used lipids to synthesize the lipid nanoparticles are (i) N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), (ii) 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), (iii) dioleoylphosphatidylethanolamine (DOPE), (iv) 1,2-dioleyl-sn-glycero-3-phosphocholine (DOPC), (v) 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DSPE), (vi) dimethyldioctadecylammonium bromide (DDAB), and (vii) cholesterol (Chol).51 On the basis of the charge of the liposome surface, they are categorized into cationic, anionic, and neutral liposomes.52 The lipid nanoparticles are used as a carrier to treat various diseases, including TTR-mediated amyloidosis, human immunodeficiency virus (HIV)-associated Kaposi’s sarcoma, neoplastic meningitis, and various types of tumors.53
4.1.1. Cationic Liposomes
Cationic lipids usually stabilize the siRNAs through electrostatic interactions, thus facilitating siRNA delivery. Cationic liposomes are lipid-bilayered structures that carry an overall positive charge and can be used in the delivery of hydrophobic cargo.54 Cationic liposomes are synthesized by utilizing positively charged lipids such as DOTAP, DSPE, and DDAB, along with neutral lipids like DOPE, DOPC, and PEG. Several research groups have explored the use of cationic liposomes for siRNA delivery both in vitro and in vivo to treat various cancers and diseases.55,56 Sheng Yu and colleagues developed and utilized the cationic nanoparticles functionalized with AS1411 antinucleolin aptamer for targeted delivery of siRNA against the Polo-like kinase 1 (siPLK-1) gene and Paclitaxel (chemotherapeutic) and could inhibit the breast cancer progression due to the synergistic effect between siPLK-1 with Paclitaxel.57 Another group tested the cationic liposomes to deliver a combination therapy of siRNA (siMcl-1) targeting myeloid cell leukemia 1 (MCL1) and gemcitabine (chemotherapeutic) against pancreatic cancer. The cationic liposomes carrying the combination therapy (siMcl-1 + Gemcitabine) could inhibit tumor progression in vivo with higher antitumor efficiency than liposomes delivering individual drugs (siMcl-1 or Gem).58 Though the success rate for siRNA delivery using the cationic liposomes is high in vitro, it is too less in vivo due to their toxicity and pulmonary inflammation due to the positive charge present on the liposomes.59 To avoid the toxicity arising from the cationic nature of the cationic lipids, the (polyethylene glycol) PEG polymer is usually mixed with the cationic lipids to synthesize hybrid PEG/lipid-based liposomes.55 For example, Guan et al., developed a cationic liposome formulation with an outer layer composed of DSPE-PEG2000 (DSPE-(polyethylene glycol)-2000]) for siRNA delivery to overcome the toxicity arising from the cationic liposomes. The DSPE-PEG2000 coating on the liposome surface improved the circulation time and reduced the toxicity. The PEG-coated liposomes are loaded with a combination therapy of siRNA targeting the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene and paclitaxel. The nanoparticle formulation reduced the tumor burden significantly in a mouse model (Hela) upon IV injection of the combinational therapy.60 The cationic liposomes derived from the blending of PEG, positively charged DC-cholesterol, and DOPE (PEG/DC-Chol/DOPE) which are also used as carriers to deliver siRNA targeting kinesin spindle protein (KSP) to inhibit ovarian cancer growth in PDX-resistant mouse model.61 Alnylam Pharmaceuticals used the lipid nanoparticle formulation to test two siRNAs (ALN-TTR01 and ALN-TTR02) against transthyretin (TTR) mRNA to treat TTR-mediated amyloidosis. Out of the two siRNA-based drugs, ALN-TTR02 (Patisiran) exhibited better prospects compared to ALN-TTR01 with fewer adverse reactions with more effectiveness and is approved by FDA.62 LNP-formulated siRNA accumulates in the liver, and the siRNA degrades the TTR mRNA using the RNAi pathway and reduces the TTR protein production.21,63−65 Though the PEG covering on the liposomal surface helped reduce the toxicity, the PEG limits the cellular uptake of lipid nanoparticles, thus lowering the transfection efficiency. Besides, PEG interferes with endosomal escape and leads to siRNA degradation.
4.1.2. Stable Nucleic Acid-Lipid Particle (SNALP)
The SNALPs are composed of both cationic and neutral lipids. The cationic lipid enhances the cellular uptake while the neutral lipid promotes the endosomal escape of the siRNA drug.66 The lipid bilayer is further protected from rapid systemic clearance by modifying it with a PEG-lipid conjugate.67 Tekmira Pharmaceuticals first tried to utilize the SNALP to treat hypercholesterolemia by delivering TKM-ApoB siRNA, then the same company used siRNA-mediated treatment against the Ebola virus (TKM-100201).68 The company also used a SNALP/PLK1-siRNA formulation to treat neuroendocrine tumors and adrenocortical carcinoma by targeting PLK1 (Polo-like kinase 1).69 However, the trials do not yield good results though the formulation has good tolerance. The SNALP-siRNA nanoparticles are tested against the hepatitis-B virus (HBV), and they showed reduced serum HBV levels; however, the HBV levels reduction in serum is dose-dependent.70 Alnylam Pharmaceuticals also utilized the SNALP formulation to deliver two siRNAs; siRNA targeting vascular endothelial growth factor (VEGF), and siRNA targeting KSP (the formulation is named as ALN-VSP02) have shown increased therapeutic effect in treatment of liver tumors. The phase-I clinical trials of the ALN-VSP02 from the initial 28 patients were demonstrated to be safe even at the highest dose of 1.25 mg/kg.71
4.1.3. Anionics Liposomes
Anionic liposomes are also used as one of the vectors for the delivery of siRNA-formulated drugs.72 Similar to the cationic liposome, anionic liposomes is also a made of lipid bilayer and are negatively charged macromolecules that delivers DNA or RNA or oligonucleotides.73 Anionic lipid nanoparticles for siRNA delivery are often developed by blending anionic lipids such as dioleoylphosphatidylethanolamine (DOPE), phosphatidylserine (PS), 1,2-dioleoyl-sn-glycero-3-[phospho-rac-(1-glycerol)] (DOPG), phosphatidic acid (PA), phosphatidylglycerol (PG), phosphatidylcholine (PC), phosphatidylinositol (PI), and caproylamine (CAP) with other cationic or neutral lipids. Anionic liposomes that are composed of anionic lipids, Ca2+ ions, and siRNA have been demonstrated to be secure and effective in siRNA delivery.74−77 The anionic liposomes exhibit less immunogenicity, have more circulating time, and showed high tolerance in animal models.72 The anionic liposomes composed of both anionic and cationic lipids results in lipid nanoparticles with a pH-responsive zeta potential profile which indeed helps in releasing the siRNA from endosomes through the proton-sponge effect.78 Anionic liposomes have been explored for siRNA delivery to treat various diseases such as breast cancer, hepatocellular carcinoma, neuronal diseases, prion diseases, systemic disease, and protein misfolding diseases in CNS.
(72,74,79−82) A study states that siRNA shows improved resistance, and it can play an effective role in gene knockdown and also states that anionic liposomes prepared using DOPG, DOPE, and glycerol show safe and effective means of nucleic acid-based drug delivery.83 siRNA delivery through anionic liposomes (DOPG/DOPE/Ca2+/siRNA molar charge ratio) has been shown to be safe and efficient in breast cancer.79 Anionic liposomes composed of DOPG and DOPE are used as carriers to deliver siRNA against the GFP gene and could silence the GFP expression with about 70% efficiency.84 Though, the anionic liposomes show promising results in siRNA delivery; however, finding the right composition of anion lipids with other cationic or neutral lipids with just the right proportion is difficult. Moreover, the lipid nanoparticles formed are heterogeneous in size.
4.1.4. Neutral Liposomes
A lipid bilayer macromolecule that carries no charge can also be used for the delivery of siRNA. Neutral lipids such as DOPC and DOPE can encapsulate and deliver siRNA into tumor cells with high efficacy compared to naked siRNA.85,86 For example, DOPC-encapsulated siRNA liposomes are studied against EPH Receptor A2 (EphA2), focal adhesion kinase (FAK), and neuropilin-2 genes, and demonstrated significant inhibition of tumor growth inhibition in both ovarian carcinomas as well as colorectal cancer xenografts.87 Likewise, neutral liposomes loaded with mTOR siRNA inhibited the tumor growth of breast cancer and restored its morphological alterations. Furthermore, the same study found that the accumulation of siRNA in mammary cancer tissue has been enhanced to a great extent that promoted its distribution into the deep cytosolic tumor area, allowing apoptosis and aiding its antitumor capability.88 Neutral liposomes have been explored for siRNA delivery to treat various diseases such as ovarian carcinoma therapy, DMBA-induced mammary carcinogenesis, and lung cancer.88−91
4.2. siRNA-Ligand Conjugates
FDA-approved siRNA-based drugs were developed by conjugating them with cell-specific ligands to sense strands of the siRNA, thus facilitating siRNA delivery to diseased/cancer cells.38 The sense strand of the siRNA is chemically modified to stabilize the siRNA against nuclease degradation.149 And, the ligands that are conjugated to the siRNA could be small molecules, peptides, or aptamers. For example, Vutrisiran is developed by conjugation of hepatocyte cell-specific tri-GalNAc ligands to the sense strand of the siRNA-targeting TTR gene and was approved in the year 2022.33,92 Acute hepatic porphyrias, a hereditary disorder can be treated using Givosiran (ALN-AS1). Givosirna is a GalNAc-siRNA conjugate that targets hepatic 5-aminolevulinic acid synthase 1 (ALAS1).93−95 Another GalNAc-siRNA conjugate drug Inclisiran (Leqvio; Novartis) is developed to treat homozygous familial hypercholesterolemia and elevated low-density lipoprotein cholesterol (LDL-C).32,96,97 Recently, the FDA approved another siRNA base drug Lumasiran (ALN-GO1) and is also a GalNAc-siRNA conjugate used in the treatment of primary hyperoxaluria (PH) type 1 by targeting glycolate oxidase (GO).98,7,99,100 Likewise, arginylglycylaspartic acid (RGD) peptides and their derivatives such as cyclopentapeptide (RGDfK) and octreotide (cyclooctapeptide) are selective toward integrin receptors (αvβ3) and somatostatin subtype-2 receptor (SSTR2), highly expressed in various tumor cells.101 Aptamers are also widely used as targeting ligands that are specific to cell-surface receptors. Aptamers are specific to prostate-specific membrane antigen (PSMA) and are employed to home two siRNAs targeting EGFR and Survivin genes using a bivalent aptamer chimera to treat prostate cancer. The two siRNAs are sandwiched between two PSMA aptamers, and the resulting bivalent aptamer chimera could home the two siRNA to a prostate tumor and exhibit high tumor inhibition and angiogenesis with low immunogenicity and high tumor specificity.102 Nair and co-workers demonstrated that the GalNAc-siRNA conjugate targeting the TTR gene enters into hepatocytes through ASGP receptor-mediated endocytosis, and results showed robust TTR gene silencing (Figure 2).103 As mentioned earlier, the off-target effect is one of the biggest barriers for siRNA, and to mitigate the off-target effect, Schlegel et al. utilized the rotated nucleobase orientation ability of the glycol nucleic acids (GNAs) by placing them within the region of the guide strand of a siRNA and exhibited fewer off-target effects while retaining the siRNA activity both in vitro and in vivo.7,104 Strand selection between sense and antisense by the RISC complex is another crucial step in mitigating off-target effects. The antisense strand selection over the sense strand by the RISC can be achieved by blocking the phosphorylation of the 5′-end of the sense strand. Manoharan et al. could solve the problem by placing a morpholino nucleotide at the end of the sense strand which not only improved antisense loading into the RISC but also increased its siRNA activity.105
Figure 2.
TTR gene silencing by conjugate 21 in mice. (a) % mTTR mRNA expression and tissue levels of conjugate 21 in liver (light blue is primary ordinate and dark blue is secondary ordinate). (b) % TTR mRNA expression comparison with control; (light blue) single dose treatments of 25, 5, and 1 mg/kg and (dark blue) multidose treatments of 5 × 5, 5 × 1, and 5 × 0.2 mg/kg. Adapted from ref (103). Copyright 2014 American Chemical Society.
4.3. Polymer-Based Nanoparticles and siRNA Delivery
Polymer-based nanoparticles are widely tested as a vehicle for siRNA delivery due to their favorable properties such as biocompatibility and diversity in modifiable structures.106 The following are a few examples of the polymeric-nanoparticle studied for siRNA delivery, they include poly-l-lysine (PLL), polyethylenimine (PEI), PEG, cyclodextrin, chitosan, hyaluronic acid (HA), proteins, and dendrimers.
4.3.1. PEI-Derived Nanosystems
Polyethylenimine (PEI) polymers resemble PEG where the two oxygen atoms are replaced with two amine groups and are responsible for improved solubility.107 Though, the PEI nanoparticles are responsible for effective intracellular delivery of siRNA; however, PEI often causes cytotoxicity.108 Therefore, the PEI is usually blended with biocompatible polymers such as chitosan, cyclodextrin (CD), PEG, and hyaluronic acid (HA) to form nanoparticles with reduced toxicity and safe delivery of the siRNA to treat different cancer types and diseases.109 For example, the HA is used as a targeting ligand as it is selective for CD44 receptors, and the receptors are abundantly expressed on the cancer cell surface.109 Thus, the HA decorated HA-PEI/PEG nanoparticle formulation utilized to deliver two siRNAs: (i) siRNA targeting the Sjogren syndrome antigen B (SSB) gene (small RNA binding exonuclease protection factor La) and (ii) siRNA targeting PLK1 gene in mice model to treat lung cancer.110 Likewise, nanoparticles composed of PEI and cyclodextrin are utilized for the delivery of siRNA and hydrophobic drugs. The amphiphilic nanoparticles could enhance the pharmacodynamics (PD) properties of the hydrophobic drugs; they usually diminished PK properties.111 The cationic PEI binds to siRNA through electrostatic interactions, whereas hydrophobic drugs accumulated in the interior of the cyclodextrin making them ideal carriers for combinational therapeutics. Also, the CD-PEI-gold nanoparticles are used as a vehicle to deliver the hydrophobic chemotherapeutic drug docetaxel (DTX) alone with siRNA against the p65 protein. Inhibition of the p65 protein leads to an enhanced cytotoxic effect by DTX and thus inhibited tumor growth in vivo.112 Likewise, polyplexes of PEI and siRNA specific to human epidermal growth factor receptor 2 (HER-2) receptor have led to gene knockdown and induced antitumor effects in vivo.87 Lipid-linked PEI nanoparticles also improved the siRNA delivery; for example, a phase I trial (NCT00689065) of siRNA CALAA-01 against the ribonucleoside-diphosphate reductase subunit M2 (RRM2) resulted in the inhibition of tumor growth in humanized cancer-bearing xenograft of mice by downregulation of RRM2. Also, the siRNA is covered by adamantane PEG-modified cationic cyclodextrin nanoparticles and a human transferrin protein which serves as a targeting ligand to deliver the siRNA to cancer cells.113
4.3.2. PLL-Derived Nanosystems
Poly-l-lysine (PLL) is synthesized by polymerizing l-lysine and is studied for siRNA delivery by complexing with the siRNA through electrostatic interactions.114,115 However, the cationic PLL/siRNA or PLL/PEG/siRNA complexes bind to serum proteins and thus reduces their ability to reach the target site.116 Nevertheless, Zhao and his team created a nanoparticle formulation LENP-Gem-si-HIF1α by loading siRNA against hypoxia-inducible factor 1α (HIF1α) and Gemicidine into the lipid-polymer hybrid nanoparticles (LENPs) and evaluated them for their efficacy in both in vitro and in vivo, showing improved results (Figure 3).117 Likewise, blending of PLL with poly(ε-caprolactone), PEG, and siRNA resulted in micelle formation and could overcome the aforementioned problem to some extent.118 Likewise, the copolymerization of PLL-PEG-PCL carrying oxaliplatin, cisplatin, and the siBcl-2 (siRNA targeting B-cell lymphoma 2) gene showed Bcl-2 mRNA downregulation in vitro.119 On the other hand, melanin-conjugated PLL polymers are also used as a vehicle, particularly to overcome the problem of endosomal escape by generating heat after NIR irradiation.120 The melanin/PLL polymer carrying siRNA against survivin could inhibit 4T1 cell proliferation in vitro and could reduce tumor burden in vivo.120 A triblock copolymer developed by Sun et al. was evaluated for combinational delivery of DOX and siRNA against the Bcl-2 gene and could inhibit HepG2/adriamycin tumors in vivo.121 Wang et al. developed PLL-SS-PEG nanoparticles by incorporating a disulfide bond (-SS-) between PLL and PEG to improve endosomal escape of the siRNA encapsulated into the nanoparticles. The PLL-SS-PEG nanoparticles could carry and deliver a siRNA against VEGF gene (siVEGF) to HepG2 tumor which resulted in the tumor inhibition in a mice model.122 A study conducted by Kozielski et al. designed and studied a biodegradable poly(beta-amino ester) (PBAE) nanoparticle for its ability to deliver five different siRNAs (anti-GBM genes) to primary human GBM cells both in vitro and then in vivo. This study has shown effective knockdown of all five genes and reduced the cancer burden.123
Figure 3.
(a) Tumour growth curves of lipid nanoparticles formulation with siHIF1 and Gemicidine. (b) Tumour weights of the mice after the treatment. Adapted from ref (117). Copyright 2015 Elsevier.
4.3.3. Dendrimers for siRNA Delivery
Dendritic polymers with tunable size and terminal functional groups can encapsulate payload into a nanometer size.124 Polymeric dendrimers are being extensively used in drug delivery for the treatment of cancers and infectious diseases.125−127 Perez et al. tested the polyamidoamine (PAMAM) dendrimers for their ability to deliver siRNA against the Green Fluorescent Protein (siGFP) gene in both T98G-EGFP and J774-EGFP cells, and results showed that the GFP successfully downregulated.128 Surface-modified PAMAM-siRNA dendrimers were used for the delivery of siRNA targeting TWIST1 transcription factor in breast cancer cell metastasis. PAMAM-based delivery systems are also used for the delivery of siRNA against ApoB.129 Patil et al. improved siRNA (siRNA against BCL-2 gene) stability by formulating it with a triblock nanocarrier and resulting in the downregulation of the BCL-2 gene in cell uptake studies on the ovarian cancer cell line.130
4.3.4. siRNA-Polymer Bioconjugates
Biocompatible conjugation to the sense strand of the siRNA is one of the widely used methods to deliver the siRNA double strands to overcome their physiological as well as intracellular barriers, thus improving efficient cellular uptake.131 Polymers such as PEG and poly(lactic-co-glycolic acid) (PLGA) are often conjugated to sense the strand of the siRNA. The siRNA-PLGA conjugate usually self-assembles into nanoparticles, thus facilitating the delivery of siRNAs.132 Saltzman et al. used the PLGA-spermidine nanoparticles for siRNA delivery against endogenous gene silencing in the vaginal lumen and uterine in a mice model.133
4.3.5. Polymeric Complexes
Polymers of positively charged tertiary amines are also tested for siRNA delivery as they can self-assemble with siRNA through electrostatic interactions.134 Being positively charged the polymeric complex enables endosomal release due to proton sponge effects, thus improving the siRNA’s cellular uptake.135 In addition to positively charged functional groups that usually bind to siRNA, hydrophobic polymers are also efficacious for siRNA delivery.136,137 The hydrophobic molecules such as cholesterol conjugation to siRNA with an optimal balance of pKa between 6.0 and 6.5 are implicated in effective siRNA delivery.134 For example, PEG-b-PLA/BHEM-Chol nanoparticles are a combination of both cationic lipid (N,N-bis(2-hydroxyethyl)-N-methyl-N-(2-cholesteryloxycarbonyl aminoethyl) ammonium bromide, BHEM-Chol) and an amphiphilic polymer that could deliver siRNA against the PLK1 gene and inhibit tumor growth in mice.138 PLGA-based block copolymers are explored for siRNA delivery against prostate cancer mice models.139 A combination of siRNA-based drugs and chemotherapy is also one of the cancer therapeutic methods. Local Drug EluteR (LODER), a biodegradable implant, is developed to deliver a combination therapy of siG12D (siRNA against mutated KRAS) and chemotherapy (gemcitabine or FOLFIRINOX) to treat locally advanced pancreatic cancer (LAPC) and has completed phase I trials and is in phase II clinical trials.140,141
4.3.6. Biopolymers
Biocompatible biopolymers such as cyclodextrin, chitosan, and proteins have been utilized for the siRNA delivery to diseases like melanoma, lung metastasis, and breast cancer.142−144 CD47 immunoglobulin is highly expressed in many cancer models, as CD47 and signal-regulatory protein α (SIRPα) forms a signaling complex that helps in the escape of cancerous cells from macrophage-mediated phagocytosis. Thus, targeting the CD47 selectively can lead to the inhibition of tumor growth. Thus, silencing of CD47 using siRNA can inhibit tumor growth and is evident from various phase 1 clinical trials.145 In another example, cyclodextrin is used as a carrier to deliver siRRM2 (siRNA targeting RRM2) and reduced the RRM2 mRNA and protein levels drastically in humans as well as in mice models.146 Chitosan is another natural biopolymer often used for siRNA delivery.147 Chitosan with higher molecular weight provides better complexation and stability with siRNA targeting CD47 while lower molecular weight chitosan needs to be mixed with specific degrees of protamine-hyaluronic acid (LPH) to achieve better silencing effects of the CD47 in tumor tissues.86,143 Protein is a biocompatible material and is explored as a carrier for the safe delivery of siRNA. Particularly, the protamine (HIV-1 envelope antibody fusion protein) is explored to deliver siRNA to the cells which expresses the HIV-1 envelope.148,149 For example, HER2-ScFv-protamine fusion protein expressed on the cell surface of various cancer models is used for targeted delivery of siRNA by complexing the siRNA with the HER2-ScFv-protamine fusion protein and forms a siRNA-protein complex.150 Likewise, cell-penetrating peptides (CPPs) are another class of peptides proven to be effective in delivering siRNA to cells by passive cell membrane penetration.151
4.4. Viral Vectors based siRNA Delivery
The recombinant adeno-associated viruses (rAAV) are explored for siRNA delivery where the inverted terminal repeat (ITR) sequences needed for viral replication as well as packaging are retained while deleting all the viral protein genes.152,153 Therapeutic transgenes (siRNA) along with regulatory elements are inserted in between the ITR sequences. The transgene capacity of rAAV can be increased by deleting the viral protein genes which in turn reduces the viral toxicity.154 The rAAV vector also does not integrate into the host genome as it cannot express the Rep protein, thus reducing the immunogenicity and cytotoxicity.154 And the rAAV vector is used for siRNA delivery in the treatment of many diseases such as myotnic dystrophy, tumor, hepatocellular carcinoma, and Huntington’s disease.155−158 Likewise, Lentiviruses are also used for siRNA delivery, for example, doxycycline (Dox)-inducible Lentiviral-mediated siRNA delivery against the Spinal microglial Toll-like receptor 4 (TLR4) gene in rats with chronic constriction injury (CCI) and reduced TLR4 expression in the spinal cord of CCI rats that resulted in reduced neuropathic pain.159 Also, siRNA offers a great opportunity to treat untreatable neurodegenerative disorders. A recent study showed that siRNA delivery using rAAV against the mutant ataxin-1 gene rescues a phenotype in a spinocerebellar ataxia mouse model.160 Likewise, administration of lentiviral vectors expressing β-secretase siRNA (siRNA against BACE1) to a transgenic model of Alzheimer’s disease lowered the BACE1 levels and thus reduced amyloid production, indicating the potential therapeutic value of siRNAs in treating Alzheimer’s.161
4.5. Cell Vesicle-Based siRNA Delivery
Cell vesicle-based nanoparticles are also used as one type of siRNA-based drug delivery for different varieties of cancer treatment.162 Biomimetic drug delivery systems have been developed by modifying natural cell membranes of red blood cells (RBC), white blood cells (WBCs), platelets, cancer cells, macrophages, and mesenchymal cells into a cell-membrane-modified nanoparticle.162 A deep study is going on cell membranes to form biomimetic nanoparticles to achieve functions like targeted drug delivery, immune escape, and immune modulation that can interact within in vivo environments.163,162 Along with cell vesicle-based RNA delivery, there are also extracellular vesicles that are used for RNA delivery from cells to intercellular space to the target site.164 Using ultrasonic dispersion, Qiu et al have developed an endoplasmic reticulum membrane (EM) decorated hybrid nanoparticle (EhCv/siRNA NPs), which has an improved version of the intracellular fate of siRNA. These EhCv/siRNA NPs after being injected peritumorally escape from the endosome/lysosome pathway. The NP is taken up by the coat protein complex I or II (COPI or COPII) vesicles for directional transportation, which is induced by the several EM resident proteins present between the endoplasmic reticulum (ER) and Golgi complex. The EM-decorated EhCv/siRNA NPs have shown maximum tumor growth inhibition for EhCv/siEGFR formulation (Figure 4).162
Figure 4.
(a) In vivo tumor inhibition effects of siRNA-loaded nanoparticles (NPs). Tumour growth curves of nude mice after treated by siRNA-loaded NPs at a dose of 0.5 mg kg–1. (b) Tumours harvested from mice after the treatment. (c) Western blot analysis after the treatment for EGFR gene regulation. Adapted from ref (162). Copyright 2019 Nature Portfolio.
Exosomes (Exos) are naturally derived biocompatible nanocarriers with a size range of 30–160 nm.165 They are harvested from various cell types and biological fluids such as milk and blood. Exos are natural carriers and are explored to develop exosome-based drug delivery systems.166 As the exosomes are biocompatible, they are versatile carriers of small RNAs, mRNAs, and proteins. Besides, the surface proteins of exosomes can be engineered to append cell-specific targeting ligands on the surface of exosomes for targeted siRNA delivery.167 Also, the targeting ligands can be imparted on the exosome surface using RNA nanoparticles. For example, Guo et al. decorated the exosomes with folate using the engineered 3WJ RNA nanoparticles for targeted delivery of siRNA to various cancer types expressing folate receptors on the cell surface.168 Plant-based ginger-derived exosome-like nanovesicles (GDENs) are also explored for siRNA delivery. The surface of GDENs are displayed with folic acid as a ligand using RNA nanotechnology and delivered the survivin siRNA to folate receptor expressing KB cancer xenograft models, and the results showed significant inhibition of tumor growth.169 Non-small-cell lung cancer (NSCLC) poses considerable health risks and exosomes decorated with RNA aptamer-targeting EGFR (epidermal growth factor receptor) as a ligand could deliver survivin siRNA for NSCLC regression by silencing the antiapoptotic factor survivin in animal trials. And tumor regression is observed at an IC50 value of 20 nmol/kg siRNA in vivo.170
4.6. Nucleotide Nanoparticles-Based siRNA Delivery
Materials composed of DNA and RNA are extensively used for smart siRNA delivery. For example, DNA nanodevices are used for the targeted delivery combination therapy of siRNAs against Bcl-2 and P-glycoprotein genes and chemotherapeutic drug Doxorubicin to human breast adenocarcinoma (MCF-7R), while the transactivator of transcription (TAT) peptide was used as a targeting ligand. Both Bcl2 and P-glycoprotein genes are suppressed in vitro as well as in vivo.171 Likewise, a 3D DNA nanogel is employed for siRNA delivery. The nanogel is assembled along with siRNA against EGFP having overhang RNA strands. The siRNA then evaluated the silencing effect on the EGFP gene, and the EGFP gene is suppressed significantly.172 The naturally derived pRNA-3-way junction (pRNA-3WJ) derived from bacteriophage nanoparticles is explored and studied as a vehicle for the delivery of various small RNA to various cancer models. For example, a 3-WJ pRNA-HER2apt-siMED1 nanoparticle bearing human epidermal growth factor receptor 2 (HER2)-RNA aptamer as a targeting ligand and siRNA against MED1 gene is employed to target HER2-overexpressing human breast cancer. The RNA nanoparticles harboring siMED1 inhibit cancer proliferation and metastasis, by sensitizing the breast cancer cells to the chemotherapeutic drug tamoxifen.173 Pang et al. created a combination of epithelial molecule (EpCAM) aptamer and Delta-5-Desaturase (D5D) siRNA into a 3WJ-EpCAM-D5D siRNA nanoparticle which specifically targets and accumulates in lung cancer cells and knockdown D5D gene. By targeting the D5D gene, the 3WJ-EpCAM-D5D siRNA nanoparticle could inhibit lung cancer growth (Figure 5).174 The 3/4WJ RNA nanoparticles are explored to treat various cancer types and include ovarian cancer, breast cancer, and glioblastoma.175−178
Figure 5.
(a) Tumor Curves of mice treated with nanoparticles carrying D5D siRNA. (b) Images of tumors at the end of the treatment. (c) COX-2 and D5D expression in tumor tissues after treatment with nanoformulation carrying D5D siRNA. Adapted from ref (174). Copyright 2020 Elsevier.
5. Treatable Diseases by siRNA
siRNA has been studied to treat various diseases, and some of them are already approved for treatment and many are in clinical trials at different stages. Thus, siRNA gene therapy can be used for the treatment of cardiovascular diseases, diabetes, cancer, viral infections, rare genetic diseases, and many more.
5.1. Hereditary Genetic Diseases
A disease or disorder that is inheritable from one generation to another is called hereditary genetic disease. Huntington’s disease, arthritis, cystic fibrosis, and multiple sclerosis are a few examples of hereditary genetic diseases. siRNA therapeutics are being developed to treat several hereditary genetic diseases such as hereditary transthyretin-mediated (hATTR) amyloidosis, Huntington’s disease, hemophilia A or B, Primary hyperoxaluria (PH), and Duchenne muscular dystrophy.36,179−181
5.2. Cardiovascular Disease
Increased levels of low-density lipoproteins (LDL) and cholesterol in the blood are causative in atherosclerotic cardiovascular diseases.182 Alnylam Pharmaceuticals are developing siRNA-based drugs for the treatment of cardiovascular diseases (CVD) that are in phase 3 clinical trials.183 The SNALP system is also used to deliver siRNA to target the ApoB gene which is responsible for recurrent cardiac events. Suppression of the ApoB gene resulted in a reduction in ApoB and LDL levels.184 Alnylam pharmaceuticals developed a GalNAc-siRNA conjugate (Inclisiran, ALB-PPCSSC) targeting the PCSK9 gene.185
5.3. Diabetes
Diabetes is a metabolic disorder caused by genetic or environmental factors.186,187 Apart from conventional methods, diabetes also would be treated using siRNA, and the ongoing research is in clinical trials. Unregulated gluconeogenesis is common in Type 2 diabetes and causes fasting and postprandial hyperglycaemia. The downregulation of the phosphoenolpyruvate carboxykinase-1 (PCK-1) by siRNA against PCK1 improved glucose homeostasis.188 Galectin-1 (Gal-1) is a highly expressed protein in diabetic mice kidneys due to Hyperglycaemia and causes renal fibrosis. And research showed that the inhibition of Gal-1 by siRNA-Gal-1 prevents the accumulation of Gal-1 in kidneys and would be a novel therapeutic method to treat renal fibrosis in diabetes.189
5.4. Cancer
Research on cancer treatment using siRNA is actively going on both in vitro and in vivo to treat a variety of cancer types such as liver cancer, pancreatic cancer, lung cancer, colorectal cancer, breast cancer, ovarian cancer, lymphoma, glioblastoma, and many more.190−197
5.5. Viral Infection
The demand for antiviral vaccines and drugs using RNA is rapidly growing nowadays. For example, the siRNA gene therapy is extended to treat various viral infections by targeting the viral RNA or DNA, and they include influenza-A virus, coxsackievirus, SARS-CoV, food-and-mouth-disease virus (FMDV), human papillomavirus (HPV), hepatitis B virus (HBV), and Zaire ebolavirus (ZEBOV).198,199
5.6. Other Diseases
siRNA-based therapy is involved in a variety of disease treatments and is studied to treat various other diseases including acute lung injury/respiratory distress syndrome, spinal cord injury, neurological diseases, ocular diseases, inflammatory diseases, rheumatic diseases, and skin diseases.43,200−207
6. Future Prospectives
The discovery of the RNAi mechanism leads to the advancement in biotechnology and medicine. The development of gene-based drugs using small RNA such as siRNA, miRNA, shRNA, Gapmers, and antisense oligos has gained immense attention due to the ease of their synthesis with modifications to improve their stability and specificity toward target genes.10,237,238 Among them, siRNA-based gene therapy has provided treatments for various untreatable diseases and it shows great potential for the treatment of a spectrum of diseases as discussed in this review. The small RNA are easy to synthesize as they are usually about 21–27 nucleotides.35 The siRNAs can perfectly search and bind to their target mRNA through perfect Watson–Crick complementarity. Moreover, the modification of the sense strand of the siRNA improves its stability, half-life, specificity, and efficacy.239 The FDA approved and ongoing clinical trials show great promise for the future of this exciting expanding field (Table 1).213 Nevertheless, the siRNAs can be excreted out through urine due to their small size.240 And lack of targeting ability to the naked siRNAs often leads to off-target effects.241 Therefore, various materials have been explored to home the siRNA to the site of action to overcome the aforementioned physiological as well as intracellular barriers. However, biocompatibility and endosomal escape of the nanomaterials are the two major concerns to address for safe siRNA delivery.242 pH-sensitive polymers of arginine, lysine, aspartic acids, and glutamic acids can be conjugated to nanoparticles to induce the endosomal release of the siRNA through proton sponge effects. Endosomolytic peptides and fusogenic lipids also can be employed to improve endosomal escape.243 Nevertheless, adding these pH-sensitive materials to the nanoparticles further complicates their pharmacodynamic and pharmacokinetic properties.244 Thus, choosing the right nanoparticles for the delivery of siRNA is important while avoiding most of the aforementioned problems. The use of biocompatible nanoparticles such as RNA nanoparticles and exosomes might be used as carriers and would eliminate the need for extra material for endosomal escape as the nanoparticles themselves are pH-sensitive as they can be protonated at low pH and escape from the endosomal entrapment.243 Targeted delivery is an important strategy to deliver nanoparticles to the disease site. RNA nanoparticles are perfectly suited for this purpose as they can be conjugated targeting ligands conveniently.169 However, RNA strands with high GC content usually exert immune responses, therefore it is important to tailor RNA nanoparticles carefully to minimize innate immune activation.49 On the other hand, exosomes are highly biocompatible and can release the payload through the back-fusion mechanism.168 However, exosome production from various cell sources is expensive. Nevertheless, exosomes extracted from biological fluids such as bovine colostrum would be cheaper than other sources.245 Besides, surface modulation of exosomes can also impart targeting ability to the exosomes.92 Thus, the engineered exosomes derived from cheap biological fluids would be another option for siRNA delivery. Besides choosing the right carrier for the siRNA delivery, targeting multiple disease-causing genes would be ideal compared to a single gene suppression. Thus, the RNA nanoparticles and engineered exosomes might serve as carriers to deliver combinational siRNA-based gene drugs to treat various cancer or diseases.
Table 1. Approved and Clinical Trials of siRNA-Based Drugs for Different Diseases.
| disease | target | vehicle | status | ref |
|---|---|---|---|---|
| hereditary transthyretin-mediated amyloidosis | transthyretin | siRNA-lipid nano particle (Patisiran) | approved | (65, 192, and 208) |
| acute hepatic porphyria | targets amino levulinate synthase I (ALAS-1) | siRNA-GalNAc conjugate (Givosiran) | approved | (94, 95, and 209) |
| primary hyperoxaluria type 1 (PH1) | hydroxy acid oxidase I | chemically stabilized siRNA (Lumasiran) | approved | (210−212) |
| transthyretin-mediated amyloidosis | TTR | siRNA-GalNAc conjugate (Vutrisiran) | approved | (33 and 213) |
| heterozygous familial hypercholesterolemia | PCSK9 | siRNA-GalNAc conjugate (Inclisiran) | approved | (97 and 213) |
| primary hyperoxaluria | PHI | siRNA-GalNAc conjugate (Nedosiran) | phase III | (211 and 214) |
| hemophilia A and B | antithrombin (AT) | siRNA-GalNAc conjugate (fitusiran) | phase III | (215 and 216) |
| acute kidney injury | p53 | chemically stabilized-siRNA (teprasian) | phase III | (217) |
| nonarteritic anterior ischemic optic neurotherapy and primary angle glaucoma | CASP2 | naked siRNA (cosdorian) | phase III | (211) |
| ocular pain and dry eye disease | TRPV1 | naked siRNA (tivanisiran) | phase III | (211 and 218) |
| multiple cancers | PLK1 | lipid nanoparticle | phases II and III | (192 and 219) |
| solid tumor | KSP and VEGF | lipid nanoparticle | phases II and III | (192 and 220) |
| AMD/CNV | VEGFR | naked siRNA | phase II | (192 and 221) |
| AMD/DME | RTP801 | naked siRNA | phase II | (192 and 222) |
| RSV virus infection | RSV nucleocapsid | naked siRNA | phase II | (192 and 223) |
| hypercholesterolemia | PCSK9 | lipid nanoparticle | phase II | (192 and 224) |
| diabetic AMD | VEGF | naked siRNA | phase II | (192 and 222) |
| advanced solid tumors | PKN3 | lipid nanoparticle | phase II | (192 and 225) |
| HCC, multiple melanoma | c-Myc | lipid nanoparticle | phase II | (192 and 226) |
| Ebola virus infection | VP24, VP35, Zaire Ebola Lpolymerase | lipid nanoparticle | phase II | (192 and 227) |
| glaucoma ocular hypertension | ADRB2 | naked siRNA | phases I and II | (192) |
| delayed graft function kidney transplant | P53 | naked siRNA | phases I and II | (228) |
| advanced cancers | EphA2 | lipid nanoparticle | phase I | (192 and 229) |
| chronic optic nerve atrophy | caspase 2 | naked siRNA | phase I | (192 and 230) |
| metastatic melanoma | LMP2, LMP7, MECL1 | LODER polymer | phase I | (140 and 192) |
| prostate cancer treatment | polo-like kinase gene | siRNA-peptide based | clinical trials | (231) |
| targeted stem cell therapy | OCT4 transcript | dendrimer | pre-clinical trials | (232 and 233) |
| idiopathic pulmonary fibrosis (IPF) | Interleukin-11 (IL-11) | polymeric nanoparticle | clinical trials | (234) |
| chronic myeloid leukemia (CML) | BCR-ABL fusion oncogene | lipid nanoparticle | preclinical studies | (235 and 236) |
Acknowledgments
The PI S.E. acknowledges the DST-SERB for the Ramanujan fellowship (RJF/2021/000067). India. S.E. also acknowledges SRM University-AP for supporting research scholars and providing infrastructure (SRMAP/URG/E&PP/2022-23/007).
Author Contributions
H.K. and S.V. contributed equally.
The authors declare no competing financial interest.
References
- Cojocaru E.; Ghitman J.; Stan R. Electrospun-Fibrous-Architecture-Mediated Non-Viral Gene Therapy Drug Delivery in Regenerative Medicine. Polymers (Basel) 2022, 14, 2647. 10.3390/polym14132647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang R.; Xu Z. Gene therapy: a double-edged sword with great powers. Mol. Cell. Biochem. 2020, 474, 73–81. 10.1007/s11010-020-03834-3. [DOI] [PubMed] [Google Scholar]
- Wahane A.; Waghmode A.; Kapphahn A.; Dhuri K.; Gupta A.; Bahal R. Role of Lipid-Based and Polymer-Based Non-Viral Vectors in Nucleic Acid Delivery for Next-Generation Gene Therapy. Molecules 2020, 25, 2866. 10.3390/molecules25122866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roberts L. Human gene transfer test approved. Science 1989, 243, 473. 10.1126/science.2911753. [DOI] [PubMed] [Google Scholar]
- Coles D. J.; Toth I. Dendritic peptide-based carriers for gene delivery. Curr. Drug Deliv 2009, 6, 338–342. 10.2174/156720109789000465. [DOI] [PubMed] [Google Scholar]
- Lu Y.; Liu J. Functional DNA nanotechnology: emerging applications of DNAzymes and aptamers. Curr. Opin Biotechnol 2006, 17, 580–588. 10.1016/j.copbio.2006.10.004. [DOI] [PubMed] [Google Scholar]
- Egli M.; Manoharan M. Chemistry, structure and function of approved oligonucleotide therapeutics. Nucleic Acids Res. 2023, 51, 2529–2573. 10.1093/nar/gkad067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fan Y.; Marioli M.; Zhang K. Analytical characterization of liposomes and other lipid nanoparticles for drug delivery. J. Pharm. Biomed Anal 2021, 192, 113642. 10.1016/j.jpba.2020.113642. [DOI] [PubMed] [Google Scholar]
- Kokkinos J.; Ignacio R. M. C.; Sharbeen G.; Boyer C.; Gonzales-Aloy E.; Goldstein D.; Australian Pancreatic Cancer Genome Initiative APGI; McCarroll J. A.; Phillips P. A. Targeting the undruggable in pancreatic cancer using nano-based gene silencing drugs. Biomaterials 2020, 240, 119742. 10.1016/j.biomaterials.2019.119742. [DOI] [PubMed] [Google Scholar]
- Bajan S.; Hutvagner G. RNA-Based Therapeutics: From Antisense Oligonucleotides to miRNAs. Cells 2020, 9, 137. 10.3390/cells9010137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chi X.; Gatti P.; Papoian T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov Today 2017, 22, 823–833. 10.1016/j.drudis.2017.01.013. [DOI] [PubMed] [Google Scholar]
- Liu J.; Zhou G.; Zhang L.; Zhao Q. Building Potent Chimeric Antigen Receptor T Cells With CRISPR Genome Editing. Front Immunol 2019, 10, 456. 10.3389/fimmu.2019.00456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goncalves G. A. R.; Paiva R. M. A. Gene therapy: advances, challenges and perspectives. Einstein (Sao Paulo) 2017, 15, 369–375. 10.1590/s1679-45082017rb4024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anthony K. RNA-based therapeutics for neurological diseases. RNA Biol. 2022, 19, 176–190. 10.1080/15476286.2021.2021650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finotti A.; Fabbri E.; Lampronti I.; Gasparello J.; Borgatti M.; Gambari R. MicroRNAs and Long Non-coding RNAs in Genetic Diseases. Mol. Diagn Ther 2019, 23, 155–171. 10.1007/s40291-018-0380-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hattori Y.; Hattori K.; Suzuki T.; Palikhe S.; Matsuda N. Nucleic-acid based gene therapy approaches for sepsis. Eur. J. Pharmacol. 2018, 833, 403–410. 10.1016/j.ejphar.2018.06.031. [DOI] [PubMed] [Google Scholar]
- Belete T. M. The Current Status of Gene Therapy for the Treatment of Cancer. Biologics 2021, 15, 67–77. 10.2147/BTT.S302095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sung Y. K.; Kim S. W. Recent advances in the development of gene delivery systems. Biomater Res. 2019, 23, 8. 10.1186/s40824-019-0156-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padhy S. K.; Takkar B.; Narayanan R.; Venkatesh P.; Jalali S. Voretigene Neparvovec and Gene Therapy for Leber’s Congenital Amaurosis: Review of Evidence to Date. Appl. Clin Genet 2020, 13, 179–208. 10.2147/TACG.S230720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neil E. E.; Bisaccia E. K. Nusinersen: A Novel Antisense Oligonucleotide for the Treatment of Spinal Muscular Atrophy. J. Pediatr Pharmacol Ther 2019, 24, 194–203. 10.5863/1551-6776-24.3.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Urits I.; Swanson D.; Swett M. C.; Patel A.; Berardino K.; Amgalan A.; Berger A. A.; Kassem H.; Kaye A. D.; Viswanath O. A Review of Patisiran (ONPATTRO) for the Treatment of Polyneuropathy in People with Hereditary Transthyretin Amyloidosis. Neurol Ther 2020, 9, 301–315. 10.1007/s40120-020-00208-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ricci A.; Ventura P. Givosiran for the treatment of acute hepatic porphyria. Expert Rev. Clin Pharmacol 2022, 15, 383–393. 10.1080/17512433.2022.2075848. [DOI] [PubMed] [Google Scholar]
- Roberts T. C.; Langer R.; Wood M. J. A. Advances in oligonucleotide drug delivery. Nat. Rev. Drug Discov 2020, 19, 673–694. 10.1038/s41573-020-0075-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H.; Su Y.; Chen D.; Li Q.; Shi S.; Huang X.; Fang M.; Yang M. Advances in the mechanisms and applications of inhibitory oligodeoxynucleotides against immune-mediated inflammatory diseases. Front Pharmacol 2023, 14, 1119431. 10.3389/fphar.2023.1119431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong E.; Goldberg T. Mipomersen (kynamro): a novel antisense oligonucleotide inhibitor for the management of homozygous familial hypercholesterolemia. P t 2014, 39, 119–122. [PMC free article] [PubMed] [Google Scholar]
- Shahryari A.; Saghaeian Jazi M.; Mohammadi S.; Razavi Nikoo H.; Nazari Z.; Hosseini E. S.; Burtscher I.; Mowla S. J.; Lickert H. Development and Clinical Translation of Approved Gene Therapy Products for Genetic Disorders. Front Genet 2019, 10, 868. 10.3389/fgene.2019.00868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arabi F.; Mansouri V.; Ahmadbeigi N. Gene therapy clinical trials, where do we go? An overview. Biomed Pharmacother 2022, 153, 113324. 10.1016/j.biopha.2022.113324. [DOI] [PubMed] [Google Scholar]
- Hu B.; Zhong L.; Weng Y.; Peng L.; Huang Y.; Zhao Y.; Liang X. J. Therapeutic siRNA: state of the art. Signal Transduct Target Ther 2020, 5, 101. 10.1038/s41392-020-0207-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoy S. M. Patisiran: First Global Approval. Drugs 2018, 78, 1625–1631. 10.1007/s40265-018-0983-6. [DOI] [PubMed] [Google Scholar]
- Syed Y. Y. Givosiran: A Review in Acute Hepatic Porphyria. Drugs 2021, 81, 841–848. 10.1007/s40265-021-01511-3. [DOI] [PubMed] [Google Scholar]
- D’Ambrosio V.; Ferraro P. M. Lumasiran in the Management of Patients with Primary Hyperoxaluria Type 1: From Bench to Bedside. Int. J. Nephrol Renovasc Dis 2022, 15, 197–206. 10.2147/IJNRD.S293682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lamb Y. N. Inclisiran: First Approval. Drugs 2021, 81, 389–395. 10.1007/s40265-021-01473-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keam S. J. Vutrisiran: First Approval. Drugs 2022, 82, 1419–1425. 10.1007/s40265-022-01765-5. [DOI] [PubMed] [Google Scholar]
- Betakova T.; Svancarova P. Role and application of RNA interference in replication of influenza viruses. Acta Virol 2013, 57, 97–104. 10.4149/av_2013_02_97. [DOI] [PubMed] [Google Scholar]
- Carthew R. W.; Sontheimer E. J. Origins and Mechanisms of miRNAs and siRNAs. Cell 2009, 136, 642–655. 10.1016/j.cell.2009.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alshaer W.; Zureigat H.; Al Karaki A.; Al-Kadash A.; Gharaibeh L.; Hatmal M. M.; Aljabali A. A. A.; Awidi A. siRNA: Mechanism of action, challenges, and therapeutic approaches. Eur. J. Pharmacol. 2021, 905, 174178. 10.1016/j.ejphar.2021.174178. [DOI] [PubMed] [Google Scholar]
- Bogaert B.; Sauvage F.; Guagliardo R.; Muntean C.; Nguyen V. P.; Pottie E.; Wels M.; Minnaert A. K.; De Rycke R.; Yang Q.; Peer D.; Sanders N.; Remaut K.; Paulus Y. M.; Stove C.; De Smedt S. C.; Raemdonck K. A lipid nanoparticle platform for mRNA delivery through repurposing of cationic amphiphilic drugs. J. Controlled Release 2022, 350, 256–270. 10.1016/j.jconrel.2022.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J.; Lu Z.; Wientjes M. G.; Au J. L. Delivery of siRNA therapeutics: barriers and carriers. Aaps j 2010, 12, 492–503. 10.1208/s12248-010-9210-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guzman-Aranguez A.; Loma P.; Pintor J. Small-interfering RNAs (siRNAs) as a promising tool for ocular therapy. Br. J. Pharmacol. 2013, 170, 730–747. 10.1111/bph.12330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohigashi H.; Hashimoto D.; Hayase E.; Takahashi S.; Ara T.; Yamakawa T.; Sugita J.; Onozawa M.; Nakagawa M.; Teshima T. Ocular instillation of vitamin A-coupled liposomes containing HSP47 siRNA ameliorates dry eye syndrome in chronic GVHD. Blood Adv. 2019, 3, 1003–1010. 10.1182/bloodadvances.2018028431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vhora I.; Patil S.; Amrutiya J.; Misra A. Liposomes and Lipid Envelope-Type Systems for Systemic siRNA Delivery. Curr. Pharm. Des 2015, 21, 4541–4555. 10.2174/138161282131151013185850. [DOI] [PubMed] [Google Scholar]
- Jeon J. Y.; Ayyar V. S.; Mitra A. Pharmacokinetic and Pharmacodynamic Modeling of siRNA Therapeutics - a Minireview. Pharm. Res. 2022, 39, 1749–1759. 10.1007/s11095-022-03333-8. [DOI] [PubMed] [Google Scholar]
- Holm A.; Hansen S. N.; Klitgaard H.; Kauppinen S. Clinical advances of RNA therapeutics for treatment of neurological and neuromuscular diseases. RNA Biol. 2022, 19, 594–608. 10.1080/15476286.2022.2066334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sajid M. I.; Moazzam M.; Kato S.; Yeseom Cho K.; Tiwari R. K. Overcoming Barriers for siRNA Therapeutics: From Bench to Bedside. Pharmaceuticals (Basel) 2020, 13, 294. 10.3390/ph13100294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akhtar S.; Benter I. F. Nonviral delivery of synthetic siRNAs in vivo. J. Clin Invest 2007, 117, 3623–3632. 10.1172/JCI33494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurahashi K.; Sawa T.; Ota M.; Kajikawa O.; Hong K.; Martin T. R.; Wiener-Kronish J. P. Depletion of phagocytes in the reticuloendothelial system causes increased inflammation and mortality in rabbits with Pseudomonas aeruginosa pneumonia. Am. J. Physiol Lung Cell Mol. Physiol 2009, 296, L198–209. 10.1152/ajplung.90472.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao S.; Lin C.; Li X.; Liang Y.; Saw P. E. TME-Responsive Multistage Nanoplatform for siRNA Delivery and Effective Cancer Therapy. Int. J. Nanomedicine 2021, 16, 5909–5921. 10.2147/IJN.S322901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrarca C.; Clemente E.; Amato V.; Pedata P.; Sabbioni E.; Bernardini G.; Iavicoli I.; Cortese S.; Niu Q.; Otsuki T.; Paganelli R.; Di Gioacchino M. Engineered metal based nanoparticles and innate immunity. Clin Mol. Allergy 2015, 13, 13. 10.1186/s12948-015-0020-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng Z.; Lu M. RNA Interference-Induced Innate Immunity, Off-Target Effect, or Immune Adjuvant?. Front Immunol 2017, 8, 331. 10.3389/fimmu.2017.00331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Semizarov D.; Frost L.; Sarthy A.; Kroeger P.; Halbert D. N.; Fesik S. W. Specificity of short interfering RNA determined through gene expression signatures. Proc. Natl. Acad. Sci. U. S. A. 2003, 100, 6347–6352. 10.1073/pnas.1131959100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aldosari B. N.; Alfagih I. M.; Almurshedi A. S. Lipid Nanoparticles as Delivery Systems for RNA-Based Vaccines. Pharmaceutics 2021, 13, 206. 10.3390/pharmaceutics13020206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y. Liposome as a delivery system for the treatment of biofilm-mediated infections. J. Appl. Microbiol. 2021, 131, 2626–2639. 10.1111/jam.15053. [DOI] [PubMed] [Google Scholar]
- Nisini R.; Poerio N.; Mariotti S.; De Santis F.; Fraziano M. The Multirole of Liposomes in Therapy and Prevention of Infectious Diseases. Front Immunol 2018, 9, 155. 10.3389/fimmu.2018.00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suarez D. L.; Schultz-Cherry S. The effect of eukaryotic expression vectors and adjuvants on DNA vaccines in chickens using an avian influenza model. Avian Dis 2000, 44, 861–868. 10.2307/1593058. [DOI] [PubMed] [Google Scholar]
- Schroeder A.; Levins C. G.; Cortez C.; Langer R.; Anderson D. G. Lipid-based nanotherapeutics for siRNA delivery. J. Intern Med. 2010, 267, 9–21. 10.1111/j.1365-2796.2009.02189.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lechanteur A.; Sanna V.; Duchemin A.; Evrard B.; Mottet D.; Piel G. Cationic Liposomes Carrying siRNA: Impact of Lipid Composition on Physicochemical Properties, Cytotoxicity and Endosomal Escape. Nanomaterials (Basel) 2018, 8, 270. 10.3390/nano8050270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu S.; Bi X.; Yang L.; Wu S.; Yu Y.; Jiang B.; Zhang A.; Lan K.; Duan S. Co-Delivery of Paclitaxel and PLK1-Targeted siRNA Using Aptamer-Functionalized Cationic Liposome for Synergistic Anti-Breast Cancer Effects In Vivo. J. Biomed Nanotechnol 2019, 15, 1135–1148. 10.1166/jbn.2019.2751. [DOI] [PubMed] [Google Scholar]
- Wang Y.; Gao F.; Jiang X.; Zhao X.; Wang Y.; Kuai Q.; Nie G.; He M.; Pan Y.; Shi W.; Ren S.; Yu Q. Co-Delivery of Gemcitabine and Mcl-1 SiRNA via Cationic Liposome-Based System Enhances the Efficacy of Chemotherapy in Pancreatic Cancer. J. Biomed Nanotechnol 2019, 15, 966–978. 10.1166/jbn.2019.2762. [DOI] [PubMed] [Google Scholar]
- Dokka S.; Toledo D.; Shi X.; Castranova V.; Rojanasakul Y. Oxygen radical-mediated pulmonary toxicity induced by some cationic liposomes. Pharm. Res. 2000, 17, 521–525. 10.1023/A:1007504613351. [DOI] [PubMed] [Google Scholar]
- Guan J.; Sun J.; Sun F.; Lou B.; Zhang D.; Mashayekhi V.; Sadeghi N.; Storm G.; Mastrobattista E.; He Z. Hypoxia-induced tumor cell resistance is overcome by synergistic GAPDH-siRNA and chemotherapy co-delivered by long-circulating and cationic-interior liposomes. Nanoscale 2017, 9, 9190–9201. 10.1039/C7NR02663C. [DOI] [PubMed] [Google Scholar]
- Lee J.; Ahn H. J. PEGylated DC-Chol/DOPE cationic liposomes containing KSP siRNA as a systemic siRNA delivery Carrier for ovarian cancer therapy. Biochem. Biophys. Res. Commun. 2018, 503, 1716–1722. 10.1016/j.bbrc.2018.07.104. [DOI] [PubMed] [Google Scholar]
- Kulkarni J. A.; Witzigmann D.; Chen S.; Cullis P. R.; van der Meel R. Lipid Nanoparticle Technology for Clinical Translation of siRNA Therapeutics. Acc. Chem. Res. 2019, 52, 2435–2444. 10.1021/acs.accounts.9b00368. [DOI] [PubMed] [Google Scholar]
- Suzuki Y.; Ishihara H. Difference in the lipid nanoparticle technology employed in three approved siRNA (Patisiran) and mRNA (COVID-19 vaccine) drugs. Drug Metab Pharmacokinet 2021, 41, 100424. 10.1016/j.dmpk.2021.100424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coelho T.; Adams D.; Silva A.; Lozeron P.; Hawkins P. N.; Mant T.; Perez J.; Chiesa J.; Warrington S.; Tranter E.; Munisamy M.; Falzone R.; Harrop J.; Cehelsky J.; Bettencourt B. R.; Geissler M.; Butler J. S.; Sehgal A.; Meyers R. E.; Chen Q.; Borland T.; Hutabarat R. M.; Clausen V. A.; Alvarez R.; Fitzgerald K.; Gamba-Vitalo C.; Nochur S. V.; Vaishnaw A. K.; Sah D. W.; Gollob J. A.; Suhr O. B. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J. Med. 2013, 369, 819–829. 10.1056/NEJMoa1208760. [DOI] [PubMed] [Google Scholar]
- Akinc A.; Maier M. A.; Manoharan M.; Fitzgerald K.; Jayaraman M.; Barros S.; Ansell S.; Du X.; Hope M. J.; Madden T. D.; Mui B. L.; Semple S. C.; Tam Y. K.; Ciufolini M.; Witzigmann D.; Kulkarni J. A.; van der Meel R.; Cullis P. R. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat. Nanotechnol 2019, 14, 1084–1087. 10.1038/s41565-019-0591-y. [DOI] [PubMed] [Google Scholar]
- Xia Y.; Tian J.; Chen X. Effect of surface properties on liposomal siRNA delivery. Biomaterials 2016, 79, 56–68. 10.1016/j.biomaterials.2015.11.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge X.; Chen L.; Zhao B.; Yuan W. Rationale and Application of PEGylated Lipid-Based System for Advanced Target Delivery of siRNA. Front Pharmacol 2021, 11, 598175. 10.3389/fphar.2020.598175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarisozen C.; Salzano G.; Torchilin V. P. Recent advances in siRNA delivery. Biomol Concepts 2015, 6, 321–341. 10.1515/bmc-2015-0019. [DOI] [PubMed] [Google Scholar]
- Zhao Y.; Huang L. Lipid nanoparticles for gene delivery. Adv. Genet 2014, 88, 13–36. 10.1016/B978-0-12-800148-6.00002-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van den Berg F.; Limani S. W.; Mnyandu N.; Maepa M. B.; Ely A.; Arbuthnot P. Advances with RNAi-Based Therapy for Hepatitis B Virus Infection. Viruses 2020, 12, 851. 10.3390/v12080851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen H.; Sun T.; Ferrari M. Nanovector delivery of siRNA for cancer therapy. Cancer Gene Ther. 2012, 19, 367–373. 10.1038/cgt.2012.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bender H. R.; Kane S.; Zabel M. D. Delivery of Therapeutic siRNA to the CNS Using Cationic and Anionic Liposomes. J. Vis Exp 2016, 113, e54106 10.3791/54106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nsairat H.; Khater D.; Sayed U.; Odeh F.; Al Bawab A.; Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022, 8, e09394. 10.1016/j.heliyon.2022.e09394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu Q.; Zhang B.; Zhou Y.; Ge Q.; Chang J.; Chen Y.; Zhang K.; Peng D.; Chen W. Co-delivery of gambogenic acid and VEGF-siRNA with anionic liposome and polyethylenimine complexes to HepG2 cells. J. Liposome Res. 2019, 29, 322–331. 10.1080/08982104.2018.1473423. [DOI] [PubMed] [Google Scholar]
- daCosta C. J.; Wagg I. D.; McKay M. E.; Baenziger J. E. Phosphatidic acid and phosphatidylserine have distinct structural and functional interactions with the nicotinic acetylcholine receptor. J. Biol. Chem. 2004, 279, 14967–14974. 10.1074/jbc.M310037200. [DOI] [PubMed] [Google Scholar]
- Marković V.; Jaillais Y. Phosphatidylinositol 4-phosphate: a key determinant of plasma membrane identity and function in plants. New Phytol 2022, 235, 867–874. 10.1111/nph.18258. [DOI] [PubMed] [Google Scholar]
- Frentzen M. Phosphatidylglycerol and sulfoquinovosyldiacylglycerol: anionic membrane lipids and phosphate regulation. Curr. Opin Plant Biol. 2004, 7, 270–276. 10.1016/j.pbi.2004.03.001. [DOI] [PubMed] [Google Scholar]
- Zhang R.; El-Mayta R.; Murdoch T. J.; Warzecha C. C.; Billingsley M. M.; Shepherd S. J.; Gong N.; Wang L.; Wilson J. M.; Lee D.; Mitchell M. J. Helper lipid structure influences protein adsorption and delivery of lipid nanoparticles to spleen and liver. Biomater Sci. 2021, 9, 1449–1463. 10.1039/D0BM01609H. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kapoor M.; Burgess D. J. Physicochemical characterization of anionic lipid-based ternary siRNA complexes. Biochim. Biophys. Acta 2012, 1818, 1603–1612. 10.1016/j.bbamem.2012.03.013. [DOI] [PubMed] [Google Scholar]
- Tagalakis A. D.; Lee D. H.; Bienemann A. S.; Zhou H.; Munye M. M.; Saraiva L.; McCarthy D.; Du Z.; Vink C. A.; Maeshima R.; White E. A.; Gustafsson K.; Hart S. L. Multifunctional, self-assembling anionic peptide-lipid nanocomplexes for targeted siRNA delivery. Biomaterials 2014, 35, 8406–8415. 10.1016/j.biomaterials.2014.06.003. [DOI] [PubMed] [Google Scholar]
- Pattipeiluhu R.; Arias-Alpizar G.; Basha G.; Chan K. Y. T.; Bussmann J.; Sharp T. H.; Moradi M. A.; Sommerdijk N.; Harris E. N.; Cullis P. R.; Kros A.; Witzigmann D.; Campbell F. Anionic Lipid Nanoparticles Preferentially Deliver mRNA to the Hepatic Reticuloendothelial System. Adv. Mater. 2022, 34, 2201095. 10.1002/adma.202201095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zabel M. D.; Mollnow L.; Bender H. siRNA Therapeutics for Protein Misfolding Diseases of the Central Nervous System. Methods Mol. Biol. 2021, 2282, 377–394. 10.1007/978-1-0716-1298-9_20. [DOI] [PubMed] [Google Scholar]
- Han X.; Lu Y.; Xu Z.; Chu Y.; Ma X.; Wu H.; Zou B.; Zhou G. Anionic liposomes prepared without organic solvents for effective siRNA delivery. IET Nanobiotechnol 2023, 17, 269. 10.1049/nbt2.12117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kapoor M.; Burgess D. J. Efficient and safe delivery of siRNA using anionic lipids: Formulation optimization studies. Int. J. Pharm. 2012, 432, 80–90. 10.1016/j.ijpharm.2012.04.058. [DOI] [PubMed] [Google Scholar]
- Ozpolat B.; Sood A. K.; Lopez-Berestein G. Liposomal siRNA nanocarriers for cancer therapy. Adv. Drug Deliv Rev. 2014, 66, 110–116. 10.1016/j.addr.2013.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Babu A.; Muralidharan R.; Amreddy N.; Mehta M.; Munshi A.; Ramesh R. Nanoparticles for siRNA-Based Gene Silencing in Tumor Therapy. IEEE Trans Nanobioscience 2016, 15, 849–863. 10.1109/TNB.2016.2621730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh A.; Trivedi P.; Jain N. K. Advances in siRNA delivery in cancer therapy. Artif Cells Nanomed Biotechnol 2018, 46, 274–283. 10.1080/21691401.2017.1307210. [DOI] [PubMed] [Google Scholar]
- Sahu R.; Jha S.; Pattanayak S. P. Therapeutic silencing of mTOR by systemically administered siRNA-loaded neutral liposomal nanoparticles inhibits DMBA-induced mammary carcinogenesis. Br. J. Cancer 2022, 127, 2207–2219. 10.1038/s41416-022-02011-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halder J.; Kamat A. A.; Landen C. N. Jr.; Han L. Y.; Lutgendorf S. K.; Lin Y. G.; Merritt W. M.; Jennings N. B.; Chavez-Reyes A.; Coleman R. L.; Gershenson D. M.; Schmandt R.; Cole S. W.; Lopez-Berestein G.; Sood A. K. Focal adhesion kinase targeting using in vivo short interfering RNA delivery in neutral liposomes for ovarian carcinoma therapy. Clin. Cancer Res. 2006, 12, 4916–4924. 10.1158/1078-0432.CCR-06-0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Landen C. N. Jr.; Chavez-Reyes A.; Bucana C.; Schmandt R.; Deavers M. T.; Lopez-Berestein G.; Sood A. K. Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res. 2005, 65, 6910–6918. 10.1158/0008-5472.CAN-05-0530. [DOI] [PubMed] [Google Scholar]
- Garbuzenko O. B.; Saad M.; Betigeri S.; Zhang M.; Vetcher A. A.; Soldatenkov V. A.; Reimer D. C.; Pozharov V. P.; Minko T. Intratracheal versus intravenous liposomal delivery of siRNA, antisense oligonucleotides and anticancer drug. Pharm. Res. 2009, 26, 382–394. 10.1007/s11095-008-9755-4. [DOI] [PubMed] [Google Scholar]
- Debacker A. J.; Voutila J.; Catley M.; Blakey D.; Habib N. Delivery of Oligonucleotides to the Liver with GalNAc: From Research to Registered Therapeutic Drug. Mol. Ther 2020, 28, 1759–1771. 10.1016/j.ymthe.2020.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yasuda M.; Gan L.; Chen B.; Kadirvel S.; Yu C.; Phillips J. D.; New M. I.; Liebow A.; Fitzgerald K.; Querbes W.; Desnick R. J. RNAi-mediated silencing of hepatic Alas1 effectively prevents and treats the induced acute attacks in acute intermittent porphyria mice. Proc. Natl. Acad. Sci. U. S. A. 2014, 111, 7777–7782. 10.1073/pnas.1406228111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scott L. J. Givosiran: First Approval. Drugs 2020, 80, 335–339. 10.1007/s40265-020-01269-0. [DOI] [PubMed] [Google Scholar]
- Li J.; Liu J.; Zhang X.; Clausen V.; Tran C.; Arciprete M.; Wang Q.; Rocca C.; Guan L. H.; Zhang G.; Najarian D.; Xu Y.; Smith P.; Wu J. T.; Chong S. Nonclinical Pharmacokinetics and Absorption, Distribution, Metabolism, and Excretion of Givosiran, the First Approved N-Acetylgalactosamine-Conjugated RNA Interference Therapeutic. Drug Metab. Dispos. 2021, 49, 572–580. 10.1124/dmd.121.000381. [DOI] [PubMed] [Google Scholar]
- Ray K. K.; Wright R. S.; Kallend D.; Koenig W.; Leiter L. A.; Raal F. J.; Bisch J. A.; Richardson T.; Jaros M.; Wijngaard P. L.J.; Kastelein J. J.P. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N Engl J. Med. 2020, 382, 1507–1519. 10.1056/NEJMoa1912387. [DOI] [PubMed] [Google Scholar]
- Padda I. S.; Mahtani A. U.; Parmar M.. Small Interfering RNA (siRNA) Based Therapy. StatPearls; StatPearls Publishing LLC: Treasure Island, FL, 2023. [PubMed] [Google Scholar]
- Liebow A.; Li X.; Racie T.; Hettinger J.; Bettencourt B. R.; Najafian N.; Haslett P.; Fitzgerald K.; Holmes R. P.; Erbe D.; Querbes W.; Knight J. An Investigational RNAi Therapeutic Targeting Glycolate Oxidase Reduces Oxalate Production in Models of Primary Hyperoxaluria. J. Am. Soc. Nephrol 2017, 28, 494–503. 10.1681/ASN.2016030338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Springer A. D.; Dowdy S. F. GalNAc-siRNA Conjugates: Leading the Way for Delivery of RNAi Therapeutics. Nucleic Acid Ther 2018, 28, 109–118. 10.1089/nat.2018.0736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garrelfs S. F.; Metry E. L.; van Harskamp D.; Vaz F. M.; van den Akker C. H. P.; Schierbeek H.; Groothoff J. W.; Oosterveld M. J. S. Glycolate oxidase inhibition by lumasiran varies between patients with primary hyperoxaluria type 1. Kidney Int. 2023, 103, 990–993. 10.1016/j.kint.2023.01.029. [DOI] [PubMed] [Google Scholar]
- Liu S. Radiolabeled cyclic RGD peptides as integrin alpha(v)beta(3)-targeted radiotracers: maximizing binding affinity via bivalency. Bioconjug Chem. 2009, 20, 2199–2213. 10.1021/bc900167c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H. Y.; Yu X.; Liu H.; Wu D.; She J. X. Co-targeting EGFR and survivin with a bivalent aptamer-dual siRNA chimera effectively suppresses prostate cancer. Sci. Rep 2016, 6, 30346. 10.1038/srep30346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair J. K.; Willoughby J. L.; Chan A.; Charisse K.; Alam M. R.; Wang Q.; Hoekstra M.; Kandasamy P.; Kel’in A. V.; Milstein S.; Taneja N.; O’Shea J.; Shaikh S.; Zhang L.; van der Sluis R. J.; Jung M. E.; Akinc A.; Hutabarat R.; Kuchimanchi S.; Fitzgerald K.; Zimmermann T.; van Berkel T. J.; Maier M. A.; Rajeev K. G.; Manoharan M. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J. Am. Chem. Soc. 2014, 136, 16958–16961. 10.1021/ja505986a. [DOI] [PubMed] [Google Scholar]
- Schlegel M. K.; Matsuda S.; Brown C. R.; Harp J. M.; Barry J. D.; Berman D.; Castoreno A.; Schofield S.; Szeto J.; Manoharan M.; Charissé K.; Egli M.; Maier M. A. Overcoming GNA/RNA base-pairing limitations using isonucleotides improves the pharmacodynamic activity of ESC+ GalNAc-siRNAs. Nucleic Acids Res. 2021, 49, 10851–10867. 10.1093/nar/gkab916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar P.; Parmar R. G.; Brown C. R.; Willoughby J. L. S.; Foster D. J.; Babu I. R.; Schofield S.; Jadhav V.; Charisse K.; Nair J. K.; Rajeev K. G.; Maier M. A.; Egli M.; Manoharan M. 5′-Morpholino modification of the sense strand of an siRNA makes it a more effective passenger. Chem. Commun. (Camb) 2019, 55, 5139–5142. 10.1039/C9CC00977A. [DOI] [PubMed] [Google Scholar]
- Begines B.; Ortiz T.; Perez-Aranda M.; Martinez G.; Merinero M.; Arguelles-Arias F.; Alcudia A. Polymeric Nanoparticles for Drug Delivery: Recent Developments and Future Prospects. Nanomaterials (Basel) 2020, 10, 1403. 10.3390/nano10071403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao L.; Zhu J.; Gong J.; Song N.; Wu S.; Qiao W.; Yang J.; Zhu M.; Zhao J. Polyethylenimine-based theranostic nanoplatform for glioma-targeting single-photon emission computed tomography imaging and anticancer drug delivery. J. Nanobiotechnology 2020, 18, 143. 10.1186/s12951-020-00705-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Helmschrodt C.; Hobel S.; Schoniger S.; Bauer A.; Bonicelli J.; Gringmuth M.; Fietz S. A.; Aigner A.; Richter A.; Richter F. Polyethylenimine Nanoparticle-Mediated siRNA Delivery to Reduce alpha-Synuclein Expression in a Model of Parkinson’s Disease. Mol. Ther Nucleic Acids 2017, 9, 57–68. 10.1016/j.omtn.2017.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong K. H.; Lu A.; Chen X.; Yang Z. Natural Ingredient-Based Polymeric Nanoparticles for Cancer Treatment. Molecules 2020, 25, 3620. 10.3390/molecules25163620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganesh S.; Iyer A. K.; Morrissey D. V.; Amiji M. M. Hyaluronic acid based self-assembling nanosystems for CD44 target mediated siRNA delivery to solid tumors. Biomaterials 2013, 34, 3489–3502. 10.1016/j.biomaterials.2013.01.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J.; Ma P. X. Cyclodextrin-based supramolecular systems for drug delivery: recent progress and future perspective. Adv. Drug Deliv Rev. 2013, 65, 1215–1233. 10.1016/j.addr.2013.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang D.; Wang T.; Xu Z.; Yu H.; Feng B.; Zhang J.; Guo C.; Yin Q.; Zhang Z.; Li Y. Cooperative Treatment of Metastatic Breast Cancer Using Host-Guest Nanoplatform Coloaded with Docetaxel and siRNA. Small 2016, 12, 488–498. 10.1002/smll.201502913. [DOI] [PubMed] [Google Scholar]
- Zuckerman J. E.; Gritli I.; Tolcher A.; Heidel J. D.; Lim D.; Morgan R.; Chmielowski B.; Ribas A.; Davis M. E.; Yen Y. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc. Natl. Acad. Sci. U. S. A. 2014, 111, 11449–11454. 10.1073/pnas.1411393111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng M.; Pan M.; Zhang W.; Lin H.; Wu S.; Lu C.; Tang S.; Liu D.; Cai J. Poly(α-l-lysine)-based nanomaterials for versatile biomedical applications: Current advances and perspectives. Bioact Mater. 2021, 6, 1878–1909. 10.1016/j.bioactmat.2020.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng M.; Pan M.; Zhang W.; Lin H.; Wu S.; Lu C.; Tang S.; Liu D.; Cai J. Poly(alpha-l-lysine)-based nanomaterials for versatile biomedical applications: Current advances and perspectives. Bioact Mater. 2021, 6, 1878–1909. 10.1016/j.bioactmat.2020.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mainini F.; Eccles M. R. Lipid and Polymer-Based Nanoparticle siRNA Delivery Systems for Cancer Therapy. Molecules 2020, 25, 2692. 10.3390/molecules25112692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao X.; Li F.; Li Y.; Wang H.; Ren H.; Chen J.; Nie G.; Hao J. Co-delivery of HIF1alpha siRNA and gemcitabine via biocompatible lipid-polymer hybrid nanoparticles for effective treatment of pancreatic cancer. Biomaterials 2015, 46, 13–25. 10.1016/j.biomaterials.2014.12.028. [DOI] [PubMed] [Google Scholar]
- Li J.; Du Y.; Su H.; Cheng S.; Zhou Y.; Jin Y.; Qi X. R. Interfacial properties and micellization of triblock poly(ethylene glycol)-poly(epsilon-caprolactone)-polyethyleneimine copolymers. Acta Pharm. Sin B 2020, 10, 1122–1133. 10.1016/j.apsb.2020.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beh C. W.; Seow W. Y.; Wang Y.; Zhang Y.; Ong Z. Y.; Ee P. L.; Yang Y. Y. Efficient delivery of Bcl-2-targeted siRNA using cationic polymer nanoparticles: downregulating mRNA expression level and sensitizing cancer cells to anticancer drug. Biomacromolecules 2009, 10, 41–48. 10.1021/bm801109g. [DOI] [PubMed] [Google Scholar]
- Yang X.; Fan B.; Gao W.; Li L.; Li T.; Sun J.; Peng X.; Li X.; Wang Z.; Wang B.; Zhang R.; Xie J. Enhanced endosomal escape by photothermal activation for improved small interfering RNA delivery and antitumor effect. Int. J. Nanomedicine 2018, 13, 4333–4344. 10.2147/IJN.S161908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun W.; Chen X.; Xie C.; Wang Y.; Lin L.; Zhu K.; Shuai X. Co-Delivery of Doxorubicin and Anti-BCL-2 siRNA by pH-Responsive Polymeric Vector to Overcome Drug Resistance in In Vitro and In Vivo HepG2 Hepatoma Model. Biomacromolecules 2018, 19, 2248–2256. 10.1021/acs.biomac.8b00272. [DOI] [PubMed] [Google Scholar]
- Wang G.; Gao X.; Gu G.; Shao Z.; Li M.; Wang P.; Yang J.; Cai X.; Li Y. Polyethylene glycol-poly(epsilon-benzyloxycarbonyl-l-lysine)-conjugated VEGF siRNA for antiangiogenic gene therapy in hepatocellular carcinoma. Int. J. Nanomedicine 2017, 12, 3591–3603. 10.2147/IJN.S131078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kozielski K. L.; Ruiz-Valls A.; Tzeng S. Y.; Guerrero-Cazares H.; Rui Y.; Li Y.; Vaughan H. J.; Gionet-Gonzales M.; Vantucci C.; Kim J.; Schiapparelli P.; Al-Kharboosh R.; Quinones-Hinojosa A.; Green J. J. Cancer-selective nanoparticles for combinatorial siRNA delivery to primary human GBM in vitro and in vivo. Biomaterials 2019, 209, 79–87. 10.1016/j.biomaterials.2019.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mittal P.; Saharan A.; Verma R.; Altalbawy F. M. A.; Alfaidi M. A.; Batiha G. E.; Akter W.; Gautam R. K.; Uddin M. S.; Rahman M. S. Dendrimers: A New Race of Pharmaceutical Nanocarriers. Biomed Res. Int. 2021, 2021, 8844030. 10.1155/2021/8844030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mhlwatika Z.; Aderibigbe B. A. Application of Dendrimers for the Treatment of Infectious Diseases. Molecules 2018, 23, 2205. 10.3390/molecules23092205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baker J. R. Jr. Dendrimer-based nanoparticles for cancer therapy. Hematology Am. Soc. Hematol Educ Program 2009, 2009, 708–719. 10.1182/asheducation-2009.1.708. [DOI] [PubMed] [Google Scholar]
- Wang J.; Li B.; Qiu L.; Qiao X.; Yang H. Dendrimer-based drug delivery systems: history, challenges, and latest developments. J. Biol. Eng. 2022, 16, 18. 10.1186/s13036-022-00298-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez A. P.; Romero E. L.; Morilla M. J. Ethylendiamine core PAMAM dendrimers/siRNA complexes as in vitro silencing agents. Int. J. Pharm. 2009, 380, 189–200. 10.1016/j.ijpharm.2009.06.035. [DOI] [PubMed] [Google Scholar]
- Finlay J.; Roberts C. M.; Lowe G.; Loeza J.; Rossi J. J.; Glackin C. A. RNA-based TWIST1 inhibition via dendrimer complex to reduce breast cancer cell metastasis. Biomed Res. Int. 2015, 2015, 382745. 10.1155/2015/382745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patil M. L.; Zhang M.; Minko T. Multifunctional triblock Nanocarrier (PAMAM-PEG-PLL) for the efficient intracellular siRNA delivery and gene silencing. ACS Nano 2011, 5, 1877–1887. 10.1021/nn102711d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X.; Mangala L. S.; Rodriguez-Aguayo C.; Kong X.; Lopez-Berestein G.; Sood A. K. RNA interference-based therapy and its delivery systems. Cancer Metastasis Rev. 2018, 37, 107–124. 10.1007/s10555-017-9717-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singha K.; Namgung R.; Kim W. J. Polymers in small-interfering RNA delivery. Nucleic Acid Ther 2011, 21, 133–147. 10.1089/nat.2011.0293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woodrow K. A.; Cu Y.; Booth C. J.; Saucier-Sawyer J. K.; Wood M. J.; Saltzman W. M. Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA. Nat. Mater. 2009, 8, 526–533. 10.1038/nmat2444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong Y.; Siegwart D. J.; Anderson D. G. Strategies, design, and chemistry in siRNA delivery systems. Adv. Drug Deliv Rev. 2019, 144, 133–147. 10.1016/j.addr.2019.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Draz M. S.; Fang B. A.; Zhang P.; Hu Z.; Gu S.; Weng K. C.; Gray J. W.; Chen F. F. Nanoparticle-mediated systemic delivery of siRNA for treatment of cancers and viral infections. Theranostics 2014, 4, 872–892. 10.7150/thno.9404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solomun J. I.; Cinar G.; Mapfumo P.; Richter F.; Moek E.; Hausig F.; Martin L.; Hoeppener S.; Nischang I.; Traeger A. Solely aqueous formulation of hydrophobic cationic polymers for efficient gene delivery. Int. J. Pharm. 2021, 593, 120080. 10.1016/j.ijpharm.2020.120080. [DOI] [PubMed] [Google Scholar]
- Tai W.; Gao X. Functional peptides for siRNA delivery. Adv. Drug Deliv Rev. 2017, 110–111, 157–168. 10.1016/j.addr.2016.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang X. Z.; Dou S.; Wang Y. C.; Long H. Y.; Xiong M. H.; Mao C. Q.; Yao Y. D.; Wang J. Single-step assembly of cationic lipid-polymer hybrid nanoparticles for systemic delivery of siRNA. ACS Nano 2012, 6, 4955–4965. 10.1021/nn300500u. [DOI] [PubMed] [Google Scholar]
- Cheng J.; Teply B. A.; Sherifi I.; Sung J.; Luther G.; Gu F. X.; Levy-Nissenbaum E.; Radovic-Moreno A. F.; Langer R.; Farokhzad O. C. Formulation of functionalized PLGA-PEG nanoparticles for in vivo targeted drug delivery. Biomaterials 2007, 28, 869–876. 10.1016/j.biomaterials.2006.09.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golan T.; Khvalevsky E. Z.; Hubert A.; Gabai R. M.; Hen N.; Segal A.; Domb A.; Harari G.; David E. B.; Raskin S.; Goldes Y.; Goldin E.; Eliakim R.; Lahav M.; Kopleman Y.; Dancour A.; Shemi A.; Galun E. RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget 2015, 6, 24560–24570. 10.18632/oncotarget.4183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hattab D.; Gazzali A. M.; Bakhtiar A. Clinical Advances of siRNA-Based Nanotherapeutics for Cancer Treatment. Pharmaceutics 2021, 13, 1009. 10.3390/pharmaceutics13071009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia W.; Tao Z.; Zhu B.; Zhang W.; Liu C.; Chen S.; Song M. Targeted Delivery of Drugs and Genes Using Polymer Nanocarriers for Cancer Therapy. Int. J. Mol. Sci. 2021, 22, 9118. 10.3390/ijms22179118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y.; Xu Z.; Guo S.; Zhang L.; Sharma A.; Robertson G. P.; Huang L. Intravenous delivery of siRNA targeting CD47 effectively inhibits melanoma tumor growth and lung metastasis. Mol. Ther 2013, 21, 1919–1929. 10.1038/mt.2013.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Subhan M. A.; Torchilin V. P. Biopolymer-Based Nanosystems for siRNA Drug Delivery to Solid Tumors including Breast Cancer. Pharmaceutics 2023, 15, 153. 10.3390/pharmaceutics15010153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W.; Huang Q.; Xiao W.; Zhao Y.; Pi J.; Xu H.; Zhao H.; Xu J.; Evans C. E.; Jin H. Advances in Anti-Tumor Treatments Targeting the CD47/SIRPalpha Axis. Front Immunol 2020, 11, 18. 10.3389/fimmu.2020.00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahman M. A.; Amin A. R.; Wang X.; Zuckerman J. E.; Choi C. H.; Zhou B.; Wang D.; Nannapaneni S.; Koenig L.; Chen Z.; Chen Z. G.; Yen Y.; Davis M. E.; Shin D. M. Systemic delivery of siRNA nanoparticles targeting RRM2 suppresses head and neck tumor growth. J. Controlled Release 2012, 159, 384–392. 10.1016/j.jconrel.2012.01.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiménez-Gómez C. P.; Cecilia J. A. Chitosan: A Natural Biopolymer with a Wide and Varied Range of Applications. Molecules 2020, 25, 3981. 10.3390/molecules25173981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song E.; Zhu P.; Lee S. K.; Chowdhury D.; Kussman S.; Dykxhoorn D. M.; Feng Y.; Palliser D.; Weiner D. B.; Shankar P.; Marasco W. A.; Lieberman J. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat. Biotechnol. 2005, 23, 709–717. 10.1038/nbt1101. [DOI] [PubMed] [Google Scholar]
- Bobbin M. L.; Burnett J. C.; Rossi J. J. RNA interference approaches for treatment of HIV-1 infection. Genome Med. 2015, 7, 50. 10.1186/s13073-015-0174-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao Y. D.; Sun T. M.; Huang S. Y.; Dou S.; Lin L.; Chen J. N.; Ruan J. B.; Mao C. Q.; Yu F. Y.; Zeng M. S.; Zang J. Y.; Liu Q.; Su F. X.; Zhang P.; Lieberman J.; Wang J.; Song E. Targeted delivery of PLK1-siRNA by ScFv suppresses Her2+ breast cancer growth and metastasis. Sci. Transl Med. 2012, 4, 130ra148. 10.1126/scitranslmed.3003601. [DOI] [PubMed] [Google Scholar]
- Borrelli A.; Tornesello A. L.; Tornesello M. L.; Buonaguro F. M. Cell Penetrating Peptides as Molecular Carriers for Anti-Cancer Agents. Molecules 2018, 23, 295. 10.3390/molecules23020295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei J.; Zhang M.; Bu B.; Zhang S.; Xu J. Packaging and functional identification of recombinant adeno-associated virus encoding cdc2-siRNA. J. Huazhong Univ Sci. Technolog Med. Sci. 2008, 28, 626–629. 10.1007/s11596-008-0602-y. [DOI] [PubMed] [Google Scholar]
- Naso M. F.; Tomkowicz B.; Perry W. L. 3rd; Strohl W. R. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs 2017, 31, 317–334. 10.1007/s40259-017-0234-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang D.; Tai P. W. L.; Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat. Rev. Drug Discov 2019, 18, 358–378. 10.1038/s41573-019-0012-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bisset D. R.; Stepniak-Konieczna E. A.; Zavaljevski M.; Wei J.; Carter G. T.; Weiss M. D.; Chamberlain J. R. Therapeutic impact of systemic AAV-mediated RNA interference in a mouse model of myotonic dystrophy. Hum. Mol. Genet. 2015, 24, 4971–4983. 10.1093/hmg/ddv219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H.; Wang Y.; Bai Y.; Shao Y.; Bai J.; Ma Z.; Liu Q.; Wu S. Recombinant adeno-associated virus expressing a p53-derived apoptotic peptide (37AA) inhibits HCC cells growth in vitro and in vivo. Oncotarget 2017, 8, 16801–16810. 10.18632/oncotarget.15160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cong M.; Liu T.; Wang P.; Xu Y.; Tang S.; Wang B.; Jia J.; Liu Y.; Hermonat P. L.; You H. Suppression of tissue inhibitor of metalloproteinase-1 by recombinant adeno-associated viruses carrying siRNAs in hepatic stellate cells. Int. J. Mol. Med. 2009, 24, 685–692. 10.3892/ijmm_00000280. [DOI] [PubMed] [Google Scholar]
- Denovan-Wright E. M.; Rodriguez-Lebron E.; Lewin A. S.; Mandel R. J. Unexpected off-targeting effects of anti-huntingtin ribozymes and siRNA in vivo. Neurobiol Dis 2008, 29, 446–455. 10.1016/j.nbd.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan R.; Di H.; Zhang J.; Huang Z.; Sun Y.; Yu W.; Wu F. Inducible Lentivirus-Mediated siRNA against TLR4 Reduces Nociception in a Rat Model of Bone Cancer Pain. Mediators Inflamm 2015, 2015, 523896. 10.1155/2015/523896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keiser M. S.; Monteys A. M.; Corbau R.; Gonzalez-Alegre P.; Davidson B. L. RNAi prevents and reverses phenotypes induced by mutant human ataxin-1. Ann. Neurol. 2016, 80, 754–765. 10.1002/ana.24789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singer O.; Marr R. A.; Rockenstein E.; Crews L.; Coufal N. G.; Gage F. H.; Verma I. M.; Masliah E. Targeting BACE1 with siRNAs ameliorates Alzheimer disease neuropathology in a transgenic model. Nat. Neurosci 2005, 8, 1343–1349. 10.1038/nn1531. [DOI] [PubMed] [Google Scholar]
- Qiu C.; Han H. H.; Sun J.; Zhang H. T.; Wei W.; Cui S. H.; Chen X.; Wang J. C.; Zhang Q. Regulating intracellular fate of siRNA by endoplasmic reticulum membrane-decorated hybrid nanoplexes. Nat. Commun. 2019, 10, 2702. 10.1038/s41467-019-10562-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu M.; Le W.; Mei T.; Wang Y.; Chen B.; Liu Z.; Xue C. Cell membrane camouflaged nanoparticles: a new biomimetic platform for cancer photothermal therapy. Int. J. Nanomedicine 2019, 14, 4431–4448. 10.2147/IJN.S200284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien K.; Breyne K.; Ughetto S.; Laurent L. C.; Breakefield X. O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. 10.1038/s41580-020-0251-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Y.; Liu P.; Tan H.; Chen X.; Wang Q.; Chen T. Exosomes as Smart Nanoplatforms for Diagnosis and Therapy of Cancer. Front Oncol 2021, 11, 743189. 10.3389/fonc.2021.743189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J.; Li S.; Zhang S.; Wang J. Plant-derived exosome-like nanoparticles and their therapeutic activities. Asian J. Pharm. Sci. 2022, 17, 53–69. 10.1016/j.ajps.2021.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samanta S.; Rajasingh S.; Drosos N.; Zhou Z.; Dawn B.; Rajasingh J. Exosomes: new molecular targets of diseases. Acta Pharmacol Sin 2018, 39, 501–513. 10.1038/aps.2017.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng Z.; Li Z.; Xu C.; Guo B.; Guo P. Folate-displaying exosome mediated cytosolic delivery of siRNA avoiding endosome trapping. J. Controlled Release 2019, 311–312, 43–49. 10.1016/j.jconrel.2019.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z.; Wang H.; Yin H.; Bennett C.; Zhang H. G.; Guo P. Arrowtail RNA for Ligand Display on Ginger Exosome-like Nanovesicles to Systemic Deliver siRNA for Cancer Suppression. Sci. Rep 2018, 8, 14644. 10.1038/s41598-018-32953-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z.; Yang L.; Wang H.; Binzel D. W.; Williams T. M.; Guo P. Non-Small-Cell Lung Cancer Regression by siRNA Delivered Through Exosomes That Display EGFR RNA Aptamer. Nucleic Acid Ther 2021, 31, 364–374. 10.1089/nat.2021.0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z.; Song L.; Liu Q.; Tian R.; Shang Y.; Liu F.; Liu S.; Zhao S.; Han Z.; Sun J.; Jiang Q.; Ding B. A Tubular DNA Nanodevice as a siRNA/Chemo-Drug Co-delivery Vehicle for Combined Cancer Therapy. Angew. Chem., Int. Ed. Engl. 2021, 60, 2594–2598. 10.1002/anie.202009842. [DOI] [PubMed] [Google Scholar]
- Ding F.; Mou Q.; Ma Y.; Pan G.; Guo Y.; Tong G.; Choi C. H. J.; Zhu X.; Zhang C. A Crosslinked Nucleic Acid Nanogel for Effective siRNA Delivery and Antitumor Therapy. Angew. Chem., Int. Ed. Engl. 2018, 57, 3064–3068. 10.1002/anie.201711242. [DOI] [PubMed] [Google Scholar]
- Zhang Y.; Leonard M.; Shu Y.; Yang Y.; Shu D.; Guo P.; Zhang X. Overcoming Tamoxifen Resistance of Human Breast Cancer by Targeted Gene Silencing Using Multifunctional pRNA Nanoparticles. ACS Nano 2017, 11, 335–346. 10.1021/acsnano.6b05910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pang L.; Shah H.; Wang H.; Shu D.; Qian S. Y.; Sathish V. EpCAM-Targeted 3WJ RNA Nanoparticle Harboring Delta-5-Desaturase siRNA Inhibited Lung Tumor Formation via DGLA Peroxidation. Mol. Ther Nucleic Acids 2020, 22, 222–235. 10.1016/j.omtn.2020.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binzel D. W.; Li X.; Burns N.; Khan E.; Lee W. J.; Chen L. C.; Ellipilli S.; Miles W.; Ho Y. S.; Guo P. Thermostability, Tunability, and Tenacity of RNA as Rubbery Anionic Polymeric Materials in Nanotechnology and Nanomedicine-Specific Cancer Targeting with Undetectable Toxicity. Chem. Rev. 2021, 121, 7398–7467. 10.1021/acs.chemrev.1c00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu C.; Li H.; Zhang K.; Binzel D. W.; Yin H.; Chiu W.; Guo P. Photo-controlled release of paclitaxel and model drugs from RNA pyramids. Nano Res. 2019, 12, 41–48. 10.1007/s12274-018-2174-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen S. A.; Day E. S.; Ko C. H.; Hurley L. A.; Luciano J. P.; Kouri F. M.; Merkel T. J.; Luthi A. J.; Patel P. C.; Cutler J. I.; Daniel W. L.; Scott A. W.; Rotz M. W.; Meade T. J.; Giljohann D. A.; Mirkin C. A.; Stegh A. H. Spherical nucleic acid nanoparticle conjugates as an RNAi-based therapy for glioblastoma. Sci. Transl Med. 2013, 5, 209ra152. 10.1126/scitranslmed.3006839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salzano G.; Navarro G.; Trivedi M. S.; De Rosa G.; Torchilin V. P. Multifunctional Polymeric Micelles Co-loaded with Anti-Survivin siRNA and Paclitaxel Overcome Drug Resistance in an Animal Model of Ovarian Cancer. Mol. Cancer Ther 2015, 14, 1075–1084. 10.1158/1535-7163.MCT-14-0556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L.; Wu T.; Shan Y.; Li G.; Ni X.; Chen X.; Hu X.; Lin L.; Li Y.; Guan Y.; Gao J.; Chen D.; Zhang Y.; Pei Z.; Chen X. Therapeutic reversal of Huntington’s disease by in vivo self-assembled siRNAs. Brain 2021, 144, 3421–3435. 10.1093/brain/awab354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pasi K. J.; Lissitchkov T.; Mamonov V.; Mant T.; Timofeeva M.; Bagot C.; Chowdary P.; Georgiev P.; Gercheva-Kyuchukova L.; Madigan K.; Van Nguyen H.; Yu Q.; Mei B.; Benson C. C.; Ragni M. V. Targeting of antithrombin in hemophilia A or B with investigational siRNA therapeutic fitusiran-Results of the phase 1 inhibitor cohort. J. Thromb Haemost 2021, 19, 1436–1446. 10.1111/jth.15270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engelbeen S.; Pasteuning-Vuhman S.; Boertje-van der Meulen J.; Parmar R.; Charisse K.; Sepp-Lorenzino L.; Manoharan M.; Aartsma-Rus A.; van Putten M. Efficient Downregulation of Alk4 in Skeletal Muscle After Systemic Treatment with Conjugated siRNAs in a Mouse Model for Duchenne Muscular Dystrophy. Nucleic Acid Ther 2023, 33, 26–34. 10.1089/nat.2022.0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ference B. A.; Ginsberg H. N.; Graham I.; Ray K. K.; Packard C. J.; Bruckert E.; Hegele R. A.; Krauss R. M.; Raal F. J.; Schunkert H.; Watts G. F.; Borén J.; Fazio S.; Horton J. D.; Masana L.; Nicholls S. J.; Nordestgaard B. G.; van de Sluis B.; Taskinen M. R.; Tokgözoglu L.; Landmesser U.; Laufs U.; Wiklund O.; Stock J. K.; Chapman M. J.; Catapano A. L. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. 10.1093/eurheartj/ehx144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kosmas C. E.; Muñoz Estrella A.; Sourlas A.; Silverio D.; Hilario E.; Montan P. D.; Guzman E. Inclisiran: A New Promising Agent in the Management of Hypercholesterolemia. Diseases 2018, 6, 63. 10.3390/diseases6030063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmermann T. S.; Lee A. C.; Akinc A.; Bramlage B.; Bumcrot D.; Fedoruk M. N.; Harborth J.; Heyes J. A.; Jeffs L. B.; John M.; Judge A. D.; Lam K.; McClintock K.; Nechev L. V.; Palmer L. R.; Racie T.; Röhl I.; Seiffert S.; Shanmugam S.; Sood V.; Soutschek J.; Toudjarska I.; Wheat A. J.; Yaworski E.; Zedalis W.; Koteliansky V.; Manoharan M.; Vornlocher H. P.; MacLachlan I. RNAi-mediated gene silencing in non-human primates. Nature 2006, 441, 111–114. 10.1038/nature04688. [DOI] [PubMed] [Google Scholar]
- Fitzgerald K.; White S.; Borodovsky A.; Bettencourt B. R.; Strahs A.; Clausen V.; Wijngaard P.; Horton J. D.; Taubel J.; Brooks A.; Fernando C.; Kauffman R. S.; Kallend D.; Vaishnaw A.; Simon A. A Highly Durable RNAi Therapeutic Inhibitor of PCSK9. N Engl J. Med. 2017, 376, 41–51. 10.1056/NEJMoa1609243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Surampudi P. N.; John-Kalarickal J.; Fonseca V. A. Emerging concepts in the pathophysiology of type 2 diabetes mellitus. Mt Sinai J. Med. 2009, 76, 216–226. 10.1002/msj.20113. [DOI] [PubMed] [Google Scholar]
- Galicia-Garcia U.; Benito-Vicente A.; Jebari S.; Larrea-Sebal A.; Siddiqi H.; Uribe K. B.; Ostolaza H.; Martin C. Pathophysiology of Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2020, 21, 6275. 10.3390/ijms21176275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh V.; Sagar P.; Kaul S.; Sandhir R.; Singhal N. K. Liver Phosphoenolpyruvate Carboxykinase-1 Downregulation via siRNA-Functionalized Graphene Oxide Nanosheets Restores Glucose Homeostasis in a Type 2 Diabetes Mellitus In Vivo Model. Bioconjug Chem. 2021, 32, 259–278. 10.1021/acs.bioconjchem.0c00645. [DOI] [PubMed] [Google Scholar]
- Al-Obaidi N.; Mohan S.; Liang S.; Zhao Z.; Nayak B. K.; Li B.; Sriramarao P.; Habib S. L. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes. FASEB J. 2019, 33, 373–387. 10.1096/fj.201800555RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fu Z.; Zhang X.; Zhou X.; Ur-Rehman U.; Yu M.; Liang H.; Guo H.; Guo X.; Kong Y.; Su Y.; Ye Y.; Hu X.; Cheng W.; Wu J.; Wang Y.; Gu Y.; Lu S. F.; Wu D.; Zen K.; Li J.; Yan C.; Zhang C. Y.; Chen X. In vivo self-assembled small RNAs as a new generation of RNAi therapeutics. Cell Res. 2021, 31, 631–648. 10.1038/s41422-021-00491-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang T.; Fogarty B.; LaForge B.; Aziz S.; Pham T.; Lai L.; Bai S. Delivery of Small Interfering RNA to Inhibit Vascular Endothelial Growth Factor in Zebrafish Using Natural Brain Endothelia Cell-Secreted Exosome Nanovesicles for the Treatment of Brain Cancer. AAPS J. 2017, 19, 475–486. 10.1208/s12248-016-0015-y. [DOI] [PubMed] [Google Scholar]
- Subhan M. A.; Torchilin V. P. Efficient nanocarriers of siRNA therapeutics for cancer treatment. Transl Res. 2019, 214, 62–91. 10.1016/j.trsl.2019.07.006. [DOI] [PubMed] [Google Scholar]
- Wang J.; Wang C.; Meng Q.; Li S.; Sun X.; Bo Y.; Yao W. siRNA targeting Notch-1 decreases glioma stem cell proliferation and tumor growth. Mol. Biol. Rep 2012, 39, 2497–2503. 10.1007/s11033-011-1001-1. [DOI] [PubMed] [Google Scholar]
- Mirzaei S.; Gholami M. H.; Ang H. L.; Hashemi F.; Zarrabi A.; Zabolian A.; Hushmandi K.; Delfi M.; Khan H.; Ashrafizadeh M.; Sethi G.; Kumar A. P. Pre-Clinical and Clinical Applications of Small Interfering RNAs (siRNA) and Co-Delivery Systems for Pancreatic Cancer Therapy. Cells 2021, 10, 3348. 10.3390/cells10123348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamran S.; Seyedrezazadeh E.; Shanehbandi D.; Asadi M.; Zafari V.; Shekari N.; Namvar L.; Zarredar H. Combination Therapy with KRAS and P38alpha siRNA Suppresses Colorectal Cancer Growth and Development in SW480 Cell Line. J. Gastrointest Cancer 2022, 53, 597–604. 10.1007/s12029-021-00667-1. [DOI] [PubMed] [Google Scholar]
- Wang Z.; Zhao S.; Shi J.; Meng F.; Yuan J.; Zhong Z. Folate-mediated targeted PLK1 inhibition therapy for ovarian cancer: A comparative study of molecular inhibitors and siRNA therapeutics. Acta Biomater 2022, 138, 443–452. 10.1016/j.actbio.2021.10.043. [DOI] [PubMed] [Google Scholar]
- Sahin B.; Fife J.; Parmar M. B.; Valencia-Serna J.; Gul-Uludag H.; Jiang X.; Weinfeld M.; Lavasanifar A.; Uludag H. siRNA therapy in cutaneous T-cell lymphoma cells using polymeric carriers. Biomaterials 2014, 35, 9382–9394. 10.1016/j.biomaterials.2014.07.029. [DOI] [PubMed] [Google Scholar]
- Thangamani L.; Balasubramanian B.; Easwaran M.; Natarajan J.; Pushparaj K.; Meyyazhagan A.; Piramanayagam S. GalNAc-siRNA conjugates: Prospective tools on the frontier of anti-viral therapeutics. Pharmacol. Res. 2021, 173, 105864. 10.1016/j.phrs.2021.105864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Idris A.; Davis A.; Supramaniam A.; Acharya D.; Kelly G.; Tayyar Y.; West N.; Zhang P.; McMillan C. L. D.; Soemardy C.; Ray R.; O’Meally D.; Scott T. A.; McMillan N. A. J.; Morris K. V. A SARS-CoV-2 targeted siRNA-nanoparticle therapy for COVID-19. Mol. Ther 2021, 29, 2219–2226. 10.1016/j.ymthe.2021.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zoulikha M.; Xiao Q.; Boafo G. F.; Sallam M. A.; Chen Z.; He W. Pulmonary delivery of siRNA against acute lung injury/acute respiratory distress syndrome. Acta Pharm. Sin B 2022, 12, 600–620. 10.1016/j.apsb.2021.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fujita Y.; Takeshita F.; Kuwano K.; Ochiya T. RNAi Therapeutic Platforms for Lung Diseases. Pharmaceuticals (Basel) 2013, 6, 223–250. 10.3390/ph6020223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi M.; Lee M.; Rhim T. Dexamethasone-conjugated polyethylenimine/MIF siRNA complex regulation of particulate matter-induced airway inflammation. Biomaterials 2013, 34, 7453–7461. 10.1016/j.biomaterials.2013.05.082. [DOI] [PubMed] [Google Scholar]
- Rosa J.; Suzuki I.; Kravicz M.; Caron A.; Pupo A. V.; Praca F. G.; Bentley M. Current Non-viral siRNA Delivery Systems as a Promising Treatment of Skin Diseases. Curr. Pharm. Des 2018, 24, 2644–2663. 10.2174/1381612824666180807120017. [DOI] [PubMed] [Google Scholar]
- Guo S.; Perets N.; Betzer O.; Ben-Shaul S.; Sheinin A.; Michaelevski I.; Popovtzer R.; Offen D.; Levenberg S. Intranasal Delivery of Mesenchymal Stem Cell Derived Exosomes Loaded with Phosphatase and Tensin Homolog siRNA Repairs Complete Spinal Cord Injury. ACS Nano 2019, 13, 10015–10028. 10.1021/acsnano.9b01892. [DOI] [PubMed] [Google Scholar]
- Moreno-Montanes J.; Bleau A. M.; Martinez T.; Vargas B.; Gonzalez M. V.; Jimenez A. I. siRNA Therapeutics in Ocular Diseases. Methods Mol. Biol. 2021, 2282, 417–442. 10.1007/978-1-0716-1298-9_23. [DOI] [PubMed] [Google Scholar]
- Ponnappa B. C. siRNA for inflammatory diseases. Curr. Opin Investig Drugs 2009, 10, 418–424. [PubMed] [Google Scholar]
- Apparailly F.; Jorgensen C. siRNA-based therapeutic approaches for rheumatic diseases. Nat. Rev. Rheumatol 2013, 9, 56–62. 10.1038/nrrheum.2012.176. [DOI] [PubMed] [Google Scholar]
- Hrkach J.; Langer R. From micro to nano: evolution and impact of drug delivery in treating disease. Drug Deliv Transl Res. 2020, 10, 567–570. 10.1007/s13346-020-00769-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fairman K.; Li M.; Ning B.; Lumen A. Physiologically based pharmacokinetic (PBPK) modeling of RNAi therapeutics: Opportunities and challenges. Biochem. Pharmacol. 2021, 189, 114468. 10.1016/j.bcp.2021.114468. [DOI] [PubMed] [Google Scholar]
- Scott L. J.; Keam S. J. Lumasiran: First Approval. Drugs 2021, 81, 277–282. 10.1007/s40265-020-01463-0. [DOI] [PubMed] [Google Scholar]
- Zhang M. M.; Bahal R.; Rasmussen T. P.; Manautou J. E.; Zhong X. B. The growth of siRNA-based therapeutics: Updated clinical studies. Biochem. Pharmacol. 2021, 189, 114432. 10.1016/j.bcp.2021.114432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kara G.; Calin G. A.; Ozpolat B. RNAi-based therapeutics and tumor targeted delivery in cancer. Adv. Drug Deliv Rev. 2022, 182, 114113. 10.1016/j.addr.2022.114113. [DOI] [PubMed] [Google Scholar]
- Friedrich M.; Aigner A. Therapeutic siRNA: State-of-the-Art and Future Perspectives. BioDrugs 2022, 36, 549–571. 10.1007/s40259-022-00549-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Setten R. L.; Rossi J. J.; Han S. P. The current state and future directions of RNAi-based therapeutics. Nat. Rev. Drug Discov 2019, 18, 421–446. 10.1038/s41573-019-0017-4. [DOI] [PubMed] [Google Scholar]
- Machin N.; Ragni M. V. An investigational RNAi therapeutic targeting antithrombin for the treatment of hemophilia A and B. J. Blood Med. 2018, 9, 135–140. 10.2147/JBM.S159297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srivastava A.; Rangarajan S.; Kavakli K.; Klamroth R.; Kenet G.; Khoo L.; You C. W.; Xu W.; Malan N.; Frenzel L.; Bagot C. N.; Stasyshyn O.; Chang C. Y.; Poloskey S.; Qiu Z.; Andersson S.; Mei B.; Pipe S. W. Fitusiran prophylaxis in people with severe haemophilia A or haemophilia B without inhibitors (ATLAS-A/B): a multicentre, open-label, randomised, phase 3 trial. Lancet Haematol 2023, 10, e322. 10.1016/S2352-3026(23)00037-6. [DOI] [PubMed] [Google Scholar]
- Thielmann M.; Corteville D.; Szabo G.; Swaminathan M.; Lamy A.; Lehner L. J.; Brown C. D.; Noiseux N.; Atta M. G.; Squiers E. C.; Erlich S.; Rothenstein D.; Molitoris B.; Mazer C. D. Teprasiran, a Small Interfering RNA, for the Prevention of Acute Kidney Injury in High-Risk Patients Undergoing Cardiac Surgery: A Randomized Clinical Study. Circulation 2021, 144, 1133–1144. 10.1161/CIRCULATIONAHA.120.053029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno-Montañés J.; Bleau A. M.; Jimenez A. I. Tivanisiran, a novel siRNA for the treatment of dry eye disease. Expert Opin Investig Drugs 2018, 27, 421–426. 10.1080/13543784.2018.1457647. [DOI] [PubMed] [Google Scholar]
- Li L.; Xue W.; Shen Z.; Liu J.; Hu M.; Cheng Z.; Wang Y.; Chen Y.; Chang H.; Liu Y.; Liu B.; Zhao J. A Cereblon Modulator CC-885 Induces CRBN- and p97-Dependent PLK1 Degradation and Synergizes with Volasertib to Suppress Lung Cancer. Mol. Ther Oncolytics 2020, 18, 215–225. 10.1016/j.omto.2020.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simonelli M.; Garralda E.; Eskens F.; Gil-Martin M.; Yen C. J.; Obermannova R.; Chao Y.; Lonardi S.; Melichar B.; Moreno V.; Yu M. L.; Bongiovanni A.; Calvo E.; Rottey S.; Machiels J. P.; Gonzalez-Martin A.; Paz-Ares L.; Chang C. L.; Mason W.; Lin C. C.; Reardon D. A.; Vieito M.; Santoro A.; Meng R.; Abbadessa G.; Menas F.; Lee H.; Liu Q.; Combeau C.; Ternes N.; Ziti-Ljajic S.; Massard C. Isatuximab plus atezolizumab in patients with advanced solid tumors: results from a phase I/II, open-label, multicenter study. ESMO Open 2022, 7, 100562. 10.1016/j.esmoop.2022.100562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X.; Ward K.; Xavier C.; Jann J.; Clark A. F.; Pang I. H.; Wu H. The novel triterpenoid RTA 408 protects human retinal pigment epithelial cells against H2O2-induced cell injury via NF-E2-related factor 2 (Nrf2) activation. Redox Biol. 2016, 8, 98–109. 10.1016/j.redox.2015.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen Q. D.; Schachar R. A.; Nduaka C. I.; Sperling M.; Basile A. S.; Klamerus K. J.; Chi-Burris K.; Yan E.; Paggiarino D. A.; Rosenblatt I.; Aitchison R.; Erlich S. S. Dose-ranging evaluation of intravitreal siRNA PF-04523655 for diabetic macular edema (the DEGAS study). Invest Ophthalmol Vis Sci. 2012, 53, 7666–7674. 10.1167/iovs.12-9961. [DOI] [PubMed] [Google Scholar]
- DeVincenzo J.; Lambkin-Williams R.; Wilkinson T.; Cehelsky J.; Nochur S.; Walsh E.; Meyers R.; Gollob J.; Vaishnaw A. A randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virus. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 8800–8805. 10.1073/pnas.0912186107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu M.; Zhu X.; Wu J.; Zhang Y.; Zhao D.; Wang X.; Ding Y.; Cao Y.; Li C.; Hu W.; Sheng J.; Luo Z.; Zheng Z.; Hu J.; Liu J.; Zhou X.; Shen A.; Ding X.; Zhang Y.; Zhao Y.; Li Y.; Zhong S.; An S.; Zou J.; Yan L. PCSK9 inhibitor recaticimab for hypercholesterolemia on stable statin dose: a randomized, double-blind, placebo-controlled phase 1b/2 study. BMC Med. 2022, 20, 13. 10.1186/s12916-021-02208-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schultheis B.; Strumberg D.; Kuhlmann J.; Wolf M.; Link K.; Seufferlein T.; Kaufmann J.; Feist M.; Gebhardt F.; Khan M.; Stintzing S.; Pelzer U. Safety, Efficacy and Pharcacokinetics of Targeted Therapy with The Liposomal RNA Interference Therapeutic Atu027 Combined with Gemcitabine in Patients with Pancreatic Adenocarcinoma. A Randomized Phase Ib/IIa Study. Cancers (Basel) 2020, 12, 3130. 10.3390/cancers12113130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Löffler M. W.; Gori S.; Izzo F.; Mayer-Mokler A.; Ascierto P. A.; Königsrainer A.; Ma Y. T.; Sangro B.; Francque S.; Vonghia L.; Inno A.; Avallone A.; Ludwig J.; Alcoba D. D.; Flohr C.; Aslan K.; Mendrzyk R.; Schuster H.; Borrelli M.; Valmori D.; Chaumette T.; Heidenreich R.; Gouttefangeas C.; Forlani G.; Tagliamonte M.; Fusco C.; Penta R.; Iñarrairaegui M.; Gnad-Vogt U.; Reinhardt C.; Weinschenk T.; Accolla R. S.; Singh-Jasuja H.; Rammensee H. G.; Buonaguro L. Phase I/II Multicenter Trial of a Novel Therapeutic Cancer Vaccine, HepaVac-101, for Hepatocellular Carcinoma. Clin. Cancer Res. 2022, 28, 2555–2566. 10.1158/1078-0432.CCR-21-4424. [DOI] [PubMed] [Google Scholar]
- Pollard A. J.; Launay O.; Lelievre J. D.; Lacabaratz C.; Grande S.; Goldstein N.; Robinson C.; Gaddah A.; Bockstal V.; Wiedemann A.; Leyssen M.; Luhn K.; Richert L.; Bétard C.; Gibani M. M.; Clutterbuck E. A.; Snape M. D.; Levy Y.; Douoguih M.; Thiebaut R. Safety and immunogenicity of a two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in adults in Europe (EBOVAC2): a randomised, observer-blind, participant-blind, placebo-controlled, phase 2 trial. Lancet Infect Dis 2021, 21, 493–506. 10.1016/S1473-3099(20)30476-X. [DOI] [PubMed] [Google Scholar]
- Chakraborty C.; Sharma A. R.; Sharma G.; Doss C. G. P.; Lee S. S. Therapeutic miRNA and siRNA: Moving from Bench to Clinic as Next Generation Medicine. Mol. Ther Nucleic Acids 2017, 8, 132–143. 10.1016/j.omtn.2017.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gan H. K.; Parakh S.; Lee F. T.; Tebbutt N. C.; Ameratunga M.; Lee S. T.; O’Keefe G. J.; Gong S. J.; Vanrenen C.; Caine J.; Giovannetti M.; Murone C.; Scott F. E.; Guo N.; Burvenich I. J. G.; Paine C.; Macri M. J.; Kotsuma M.; Senaldi G.; Venhaus R.; Scott A. M. A phase 1 safety and bioimaging trial of antibody DS-8895a against EphA2 in patients with advanced or metastatic EphA2 positive cancers. Invest New Drugs 2022, 40, 747–755. 10.1007/s10637-022-01237-3. [DOI] [PubMed] [Google Scholar]
- Lam B. L.; Feuer W. J.; Davis J. L.; Porciatti V.; Yu H.; Levy R. B.; Vanner E.; Guy J. Leber Hereditary Optic Neuropathy Gene Therapy: Adverse Events and Visual Acuity Results of All Patient Groups. Am. J. Ophthalmol 2022, 241, 262–271. 10.1016/j.ajo.2022.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang X.; Pallaoro A.; Braun G. B.; Morales D. P.; Ogunyankin M. O.; Zasadzinski J.; Reich N. O. Modular plasmonic nanocarriers for efficient and targeted delivery of cancer-therapeutic siRNA. Nano Lett. 2014, 14, 2046–2051. 10.1021/nl500214e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziraksaz Z.; Nomani A.; Soleimani M.; Bakhshandeh B.; Arefian E.; Haririan I.; Tabbakhian M. Evaluation of cationic dendrimer and lipid as transfection reagents of short RNAs for stem cell modification. Int. J. Pharm. 2013, 448, 231–238. 10.1016/j.ijpharm.2013.03.035. [DOI] [PubMed] [Google Scholar]
- Eljack S.; David S.; Faggad A.; Chourpa I.; Allard-Vannier E. Nanoparticles design considerations to co-deliver nucleic acids and anti-cancer drugs for chemoresistance reversal. Int. J. Pharm. X 2022, 4, 100126. 10.1016/j.ijpx.2022.100126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai X.; Zhao G.; Chen Q.; Li Z.; Gao M.; Ho W.; Xu X.; Zhang X. Q. Inhaled siRNA nanoparticles targeting IL11 inhibit lung fibrosis and improve pulmonary function post-bleomycin challenge. Sci. Adv. 2022, 8, eabn7162. 10.1126/sciadv.abn7162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jyotsana N.; Sharma A.; Chaturvedi A.; Budida R.; Scherr M.; Kuchenbauer F.; Lindner R.; Noyan F.; Suhs K. W.; Stangel M.; Grote-Koska D.; Brand K.; Vornlocher H. P.; Eder M.; Thol F.; Ganser A.; Humphries R. K.; Ramsay E.; Cullis P.; Heuser M. Lipid nanoparticle-mediated siRNA delivery for safe targeting of human CML in vivo. Ann. Hematol 2019, 98, 1905–1918. 10.1007/s00277-019-03713-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S. H.; Menon H.; Jootar S.; Saikia T.; Kwak J. Y.; Sohn S. K.; Park J. S.; Jeong S. H.; Kim H. J.; Kim Y. K.; Oh S. J.; Kim H.; Zang D. Y.; Chung J. S.; Shin H. J.; Do Y. R.; Kim J. A.; Kim D. Y.; Choi C. W.; Park S.; Park H. L.; Lee G. Y.; Cho D. J.; Shin J. S.; Kim D. W. Efficacy and safety of radotinib in chronic phase chronic myeloid leukemia patients with resistance or intolerance to BCR-ABL1 tyrosine kinase inhibitors. Haematologica 2014, 99, 1191–1196. 10.3324/haematol.2013.096776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marrosu E.; Ala P.; Muntoni F.; Zhou H. Gapmer Antisense Oligonucleotides Suppress the Mutant Allele of COL6A3 and Restore Functional Protein in Ullrich Muscular Dystrophy. Mol. Ther Nucleic Acids 2017, 8, 416–427. 10.1016/j.omtn.2017.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin Q.; Song X.; Yang P.; Yang W.; Li X.; Wang X.; Wang S. Incorporation of glycyrrhizic acid and polyene phosphatidylcholine in lipid nanoparticles ameliorates acute liver injury via delivering p65 siRNA. Nanomedicine 2023, 48, 102649. 10.1016/j.nano.2022.102649. [DOI] [PubMed] [Google Scholar]
- Chiu Y. L.; Rana T. M. siRNA function in RNAi: a chemical modification analysis. Rna 2003, 9, 1034–1048. 10.1261/rna.5103703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang Y.; Hong J.; Zheng S.; Ding Y.; Guo S.; Zhang H.; Zhang X.; Du Q.; Liang Z. Elimination pathways of systemically delivered siRNA. Mol. Ther 2011, 19, 381–385. 10.1038/mt.2010.266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pecot C. V.; Calin G. A.; Coleman R. L.; Lopez-Berestein G.; Sood A. K. RNA interference in the clinic: challenges and future directions. Nat. Rev. Cancer 2011, 11, 59–67. 10.1038/nrc2966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zare M.; Pemmada R.; Madhavan M.; Shailaja A.; Ramakrishna S.; Kandiyil S. P.; Donahue J. M.; Thomas V. Encapsulation of miRNA and siRNA into Nanomaterials for Cancer Therapeutics. Pharmaceutics 2022, 14, 1620. 10.3390/pharmaceutics14081620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erazo-Oliveras A.; Muthukrishnan N.; Baker R.; Wang T. Y.; Pellois J. P. Improving the endosomal escape of cell-penetrating peptides and their cargos: strategies and challenges. Pharmaceuticals (Basel) 2012, 5, 1177–1209. 10.3390/ph5111177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang K. N.; Zhang C. Q.; Wang W.; Wang P. C.; Zhou J. P.; Liang X. J. pH-responsive mesoporous silica nanoparticles employed in controlled drug delivery systems for cancer treatment. Cancer Biol. Med. 2014, 11, 34–43. 10.7497/j.issn.2095-3941.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samuel M.; Chisanga D.; Liem M.; Keerthikumar S.; Anand S.; Ang C. S.; Adda C. G.; Versteegen E.; Jois M.; Mathivanan S. Bovine milk-derived exosomes from colostrum are enriched with proteins implicated in immune response and growth. Sci. Rep 2017, 7, 5933. 10.1038/s41598-017-06288-8. [DOI] [PMC free article] [PubMed] [Google Scholar]





