ABSTRACT
In November 2019 givosiran became the second small interfering RNA (siRNA)-based drug to receive US Food and Drug Administration (FDA) approval, it has been developed for the treatment of acute intermittent porphyria (AIP), a disorder characterized by life-threatening acute neurovisceral attacks. The porphyrias are a group of disorders in which enzymatic deficiencies in heme production lead to toxic accumulation of delta-aminolevulinic acid (ALA) and porphobilinogen (PBG), which are involved in the neurovisceral attacks. Givosiran acts as a conventional siRNA to trigger RNA interference (RNAi)-mediated gene silencing on delta-ALA synthase 1 (ALAS1), thus returning ALA and PBG metabolites to the physiological level to attenuate further neurotoxicity. Givosiran makes use of a new hepatic-delivery system that conjugates three GalNac (N-acetylgalactosamine) molecules to the siRNA passenger strand. GalNac binds to the liver asialoglycoprotein receptor, favoring the internalization of these GalNac-conjugated siRNAs into the hepatic cells. In a phase I study, subcutaneous monthly administration of givosiran 2.5 mg/kg reduced > 90% of ALA and PBG content. This siRNA is being analyzed in ENVISION (NCT03338816), a phase III, multicenter, placebo-controlled randomized controlled trial. In preliminary results, givosiran achieved clinical endpoints for AIP, reducing urinary ALA levels, and presented a safety profile that enabled further drug development. The clinical performance of givosiran revealed that suppression of ALAS1 by GalNac-decorated siRNAs represents an additional approach for the treatment of patients with AIP that manifests recurrent acute neurovisceral attacks.
Subject(s)
Acetylgalactosamine/analogs & derivatives , Gene Silencing , Genetic Therapy , Nitric Oxide Synthase Type I/genetics , Pyrrolidines/therapeutic use , RNA Interference , RNA, Small Interfering/therapeutic use , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/adverse effects , Acetylgalactosamine/pharmacokinetics , Acetylgalactosamine/therapeutic use , Clinical Trials as Topic , Drug Development , Genetic Therapy/adverse effects , Genetic Therapy/methods , Heme/biosynthesis , Humans , Porphyria, Acute Intermittent , Pyrrolidines/administration & dosage , Pyrrolidines/adverse effects , Pyrrolidines/pharmacokinetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/adverse effects , RNA, Small Interfering/pharmacokinetics , Treatment OutcomeABSTRACT
In 2018, patisiran was the first-ever RNA interference (RNAi)-based drug approved by the US Food and Drug Administration. Now pharmacology textbooks may include a new drug class that results in the effect first described by Fire and Mello 2 decades ago: post-transcriptional gene silencing by a small-interfering RNA (siRNA). Patients with hereditary transthyretin-mediated amyloidosis (hATTR amyloidosis) present with mutations in the transthyretin (TTR) gene that lead to the formation of amyloid deposits in peripheral nerves and heart. The disease may also affect the eye and central nervous system. The formulation of patisiran comprises the RNAi drug encapsulated into a nanoparticle especially developed to deliver the anti-TTR siRNA into the main TTR producer: the liver. Hepatic cells contain apolipoprotein E receptors that recognize ApoE proteins opsonized in the lipid carrier and internalize the drug by endocytosis. Lipid vesicles are disrupted in the cell cytoplasm, and siRNAs are free to trigger the RNAi-based TTR gene silencing. The silencing process involves the binding of siRNA guide strand to 3'-untranslated region sequence of both mutant and wild-type TTR messenger RNA, which culminates in the TTR mRNA cleavage by the RNA-induced silencing complex (RISC) as the first biochemical drug effect. Patisiran 0.3 mg/kg is administered intravenously every 3 weeks. Patients require premedication with anti-inflammatory drugs and antagonists of histamine H1 and H2 receptors to prevent infusion-related reactions and may require vitamin A supplementation. Following patisiran treatment, TTR knockdown remained stable for at least 2 years. Adverse effects were mild to moderate with unchanged hematological, renal, or hepatic parameters. No drug-related severe adverse effects occurred in a 24-month follow-up phase II open-label extension study. At the recommended dosage of patisiran, Cmax and AUC values (mean ± standard deviation) were 7.15 ± 2.14 µg/mL and 184 ± 159 µg·h/mL, respectively. The drug showed stability in circulation with > 95% encapsulated in lipid particles. Metabolization occurred by ribonuclease enzymes, with less than 1% excreted unchanged in the urine. Patisiran ameliorated neuropathy impairment according to the modified Neuropathy Impairment Score + 7 analysis of the phase III study. The Norfolk Quality of Life-Diabetic Neuropathy score and gait speed improved in 51% of the patisiran-treated group in 18 months. Additionally, the modified body mass index showed positive outcomes. Altogether, the data across phase I-III clinical trials points to patisiran as an effective and safe drug for the treatment of hATTR amyloidosis. It is hoped that real-world data from a larger number of patients treated with patisiran will confirm the effectiveness of this first-approved siRNA-based drug.
Subject(s)
Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/therapy , Genetic Therapy , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Drug Administration Routes , Drug Evaluation, Preclinical , Gene Silencing , Humans , Oligonucleotides/administration & dosage , Prealbumin/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/adverse effects , RNA, Small Interfering/pharmacokinetics , Treatment OutcomeABSTRACT
BACKGROUND: The molecular mechanism of silencing genes using small interference RNA is as particular and innovative phenomenon as the proposed delivery systems to release them. Recent advances in RNAi have resulted in the development of multiple siRNA candidates that are currently being evaluated in preclinical / clinical instances. SNALP®, Atuplex® and Rondel® technologies stand out; they are mainly based on polymers, cyclodextrins or lipids. METHOD: The objective of this work is to review the main features that Gene Therapy Medicinal Product under current clinical evaluation present from a pharmaceutical technology point of view; it tries to bring up theoretical concepts that give scientific support to the interpretation of data obtained during pharmaceutical development process. It is basically focused on improving the translation from bench/theoretical concepts to bedside of non viral vectors carrying siRNA. RESULTS: The extensive presence of lipid-based nanoparticle non-viral systems in clinical stages is due to the advantages of their formulations. These include: safety, low immunogenicity, high degree of material properties control, function tuning and ability to impact pharmacokinetics and in vivo biodistribution. This work presents a pharmaceutical approach so as to improve the potential of success in siRNA delivery using liposomal systems. CONCLUSION: Formulation design should be increasingly addressed with industrial criteria; it should be based on quality by design and on the estimation of critical attributes that affect product performance, and supported by a range of characterization techniques and appropriate analytical methods.
Subject(s)
Lipids/chemistry , Nanoparticles , RNA, Small Interfering/administration & dosage , Animals , Gene Silencing , Gene Transfer Techniques , Humans , RNA Interference , RNA, Small Interfering/pharmacokinetics , Tissue Distribution , Translational Research, BiomedicalABSTRACT
BACKGROUND: RNA interference is a promising therapeutic tool for the treatment of a variety of diseases, with great potential for cancer therapy. Small interfering RNA (siRNA), however, presents several drawbacks that hamper its therapeutic application. Lipid nanoparticles, including liposomes, are delivery systems with great potential for siRNA delivery, protecting it from degradation, enhancing its cell uptake with the ability of controlled release. However, non-specific delivery and side effects could potentially limit the in vivo application. Therefore, targeting lipid nanoparticles to overexpressed receptors of cancer cells represents a strategy for better therapeutic outcome, with improved efficacy and reduced toxicity. For this purpose, lipid nanoparticles could be functionalized with several moieties that can be recognized by cancer cells more than by normal cells. These ligands include folate, transferrin, peptides, oligosaccharides, monoclonal antibodies and aptamers. METHODS: In this paper, we reviewed functionalization strategies and addressed the major in vitro and in vivo findings in the field of cancer treatment with siRNA. RESULTS: Many papers showed enhanced siRNA delivery by targeted liposomes, resulting in enhanced drug uptake and better cytotoxicity, with consequent better tumor growth control in xenograft studies. CONCLUSION: siRNA delivery mediated by functionalized liposomes is promising, but clinical trials need to be conducted.
Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Neoplasms/genetics , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , RNAi Therapeutics , Animals , Antineoplastic Agents/pharmacokinetics , Humans , LiposomesABSTRACT
The past decades have witnessed the successful transition of several nanotechnology platforms into the clinical trials. However, specific delivery of therapeutics to tumors is hindered by several barriers including cancer recognition and tissue penetration, particle heterogeneity and aggregation, and unfavorable pharmacokinetic profiles such as fast clearance and organ accumulation. With the advent of RNA nanotechnology, a series of RNA nanoparticles have been successfully constructed to overcome many of the aforementioned challenges for in vivo cancer targeting with favorable biodistribution profiles. Compared to other nanodelivery platforms, the physiochemical properties of RNA nanoparticles can be tuned with relative ease for investigating the in vivo behavior of nanoparticles upon systemic injection. The size, shape, and surface chemistry, especially hydrophobic modifications, exert significant impacts on the in vivo fate of RNA nanoparticles. Rationally designed RNA nanoparticles with defined stoichiometry and high homogeneity have been demonstrated to specifically target tumor cells while avoiding accumulation in healthy vital organs after systemic injection. RNA nanoparticles were proven to deliver therapeutics such as siRNA and anti-miRNA to block tumor growth in several animal models. Although the release of anti-miRNA from the RNA nanoparticles has achieved high efficiency of tumor regression in multiple animal models, the efficiency of endosomal escape for siRNA delivery needs further improvement. This review focuses on the advances and perspectives of this promising RNA nanotechnology platform for cancer targeting and therapy.
Subject(s)
Endosomes/metabolism , Nanoparticles/administration & dosage , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , RNA/administration & dosage , Animals , Humans , Mice, Nude , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/metabolism , RNA/genetics , RNA/pharmacokinetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays/methodsABSTRACT
PURPOSE: To investigate whether intracavernosal injection of short hairpin RNA for IGFBP-3 could improve erectile function in streptozotocin-induced diabetic rats. MATERIALS AND METHODS: After 12 weeks of IGFBP-3 short hairpin RNA injection treatment, intracavernous pressure responses to electrical stimulation of cavernous nerves were evaluated. The expression of IGFBP-3 and IGF-1 at mRNA and protein levels were detected by quantitative real-time PCR analysis and Western blot, respectively. The concentration of cavernous cyclic guanosine monophosphate was detected by enzyme-linked immunosorbent assay. RESULTS: At 12 weeks after intracavernous administration of IGFBP-3 shRNA, the cavernosal pressure was significantly increased in response to the cavernous nerves stimulation compared to the diabetic group (P<0.05). Cavernous IGFBP-3 expression at both mRNA and protein levels was significantly inhibited. At the same time, cavernous IGF-1 expression was significantly increased in the IGFBP-3 shRNA treatment group compared to the diabetic group (P<0.01). Cavernous cyclic guanosine monophosphate concentration was significantly increased in the IGFBP-3 shRNA treatment group compared to the diabetic group (P<0.01). CONCLUSIONS: Gene transfer of IGFBP-3 shRNA could improve erectile function via the restoration of cavernous IGF-1 bioavailability and an increase of cavernous cGMP concentration in the pathogenesis of erectile dysfunction in streptozotocin-induced diabetic rats.
Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Erectile Dysfunction/drug therapy , Erectile Dysfunction/physiopathology , Insulin-Like Growth Factor Binding Protein 3/pharmacokinetics , Insulin-Like Growth Factor I/drug effects , Penis/drug effects , RNA, Small Interfering/pharmacokinetics , Animals , Biological Availability , Blotting, Western , Diabetes Mellitus, Experimental/complications , Enzyme-Linked Immunosorbent Assay , Erectile Dysfunction/etiology , Injections , Insulin-Like Growth Factor I/analysis , Male , Random Allocation , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reproducibility of Results , StreptozocinABSTRACT
ABSTRACT Purpose To investigate whether intracavernosal injection of short hairpin RNA for IGFBP-3 could improve erectile function in streptozotocin-induced diabetic rats. Materials and methods After 12 weeks of IGFBP-3 short hairpin RNA injection treatment, intracavernous pressure responses to electrical stimulation of cavernous nerves were evaluated. The expression of IGFBP-3 and IGF-1 at mRNA and protein levels were detected by quantitative real-time PCR analysis and Western blot, respectively. The concentration of cavernous cyclic guanosine monophosphate was detected by enzyme-linked immunosorbent assay. Results At 12 weeks after intracavernous administration of IGFBP-3 shRNA, the cavernosal pressure was significantly increased in response to the cavernous nerves stimulation compared to the diabetic group (P<0.05). Cavernous IGFBP-3 expression at both mRNA and protein levels was significantly inhibited. At the same time, cavernous IGF-1 expression was significantly increased in the IGFBP-3 shRNA treatment group compared to the diabetic group (P<0.01). Cavernous cyclic guanosine monophosphate concentration was significantly increased in the IGFBP-3 shRNA treatment group compared to the diabetic group (P<0.01). Conclusions Gene transfer of IGFBP-3 shRNA could improve erectile function via the restoration of cavernous IGF-1 bioavailability and an increase of cavernous cGMP concentration in the pathogenesis of erectile dysfunction in streptozotocin-induced diabetic rats.
Subject(s)
Animals , Male , Penis/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacokinetics , RNA, Small Interfering/pharmacokinetics , Diabetes Mellitus, Experimental/physiopathology , Erectile Dysfunction/physiopathology , Erectile Dysfunction/drug therapy , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/drug effects , Enzyme-Linked Immunosorbent Assay , Biological Availability , Random Allocation , Blotting, Western , Reproducibility of Results , Rats, Wistar , Streptozocin , Diabetes Mellitus, Experimental/complications , Real-Time Polymerase Chain Reaction , Erectile Dysfunction/etiology , InjectionsABSTRACT
The ability of small interfering RNAs (siRNAs) to potently but reversibly silence genes in vivo has made them particularly well suited as a new class of drugs that interfere with disease-causing or disease-promoting genes. However, the largest remaining hurdle for the widespread use of this technology in skin is the lack of an effective delivery system. The aim of the present study was to evaluate nanodispersed systems in liquid crystalline phases that deliver siRNA into the skin. The proposed systems present important properties for the delivery of macromolecules in a biological medium, as they are formed by substances that have absorption-enhancing and fusogenic effects; additionally, they facilitate entrapment by cellular membranes due to their nano-scale structure. The cationic polymer polyethylenimine (PEI) or the cationic lipid oleylamine (OAM) were added to monoolein (MO)-based systems in different concentrations, and after dispersion in aqueous medium, liquid crystalline phase nanodispersions were obtained and characterized by their physicochemical properties. Then, in vitro penetration studies using diffusion cell and pig ear skin were carried out to evaluate the effect of the nanodispersions on the skin penetration of siRNA; based on these results, the nanodispersions containing MO/OA/PEI/aqueous phase (8:2:5:85, w/w/w/w) and MO/OA/OAM/aqueous phase (8:2:2:88, w/w/w/w) were selected. These systems were investigated in vivo for skin penetration, skin irritation, and the ability to knockdown glyceraldehyde 3-phosphate dehydrogenase (GAPDH) protein levels in animal models. The results showed that the studied nanodispersions may represent a promising new non-viral vehicle and can be considered highly advantageous in the treatment of skin disorders; they were effective in optimizing the skin penetration of siRNA and reducing the levels of the model protein GAPDH without causing skin irritation.
Subject(s)
Liquid Crystals , Nanoparticles , RNA, Small Interfering/administration & dosage , Skin Absorption , Amines/chemistry , Animals , Female , Gene Knockdown Techniques , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glycerides/chemistry , Male , Mice , Mice, Hairless , Polyethyleneimine/chemistry , RNA, Small Interfering/pharmacokinetics , Skin/metabolism , SwineABSTRACT
BACKGROUND: Molecules taken up by olfactory and trigeminal nerve neurons directly access the brain by the nose-to-brain pathway. In situ-forming mucoadhesive gels would increase the residence time of intranasal material, favoring the nose-to-brain delivery. In this first approach, brain radioactivity after intranasal administration of (32)P-small interference RNA (siRNA) complexed with poly(amidoamine) G7 dendrimers (siRNA dendriplexes) within in situ-forming mucoadhesive gels, was determined. MATERIALS: (32)P-siRNA dendriplexes were incorporated into in situ-forming mucoadhesive gels prepared by blending thermosensitive poloxamer (23% w/w) with mucoadhesive chitosan (1% w/w, PxChi) or carbopol (0.25% w/w, PxBCP). Rheological properties, radiolabel release profile, and local toxicity in rat nasal mucosa were determined. The best-suited formulation was intranasally administered to rats, and blood absorption and brain distribution of radioactivity were measured. RESULTS: The gelation temperature of both formulations was 23°C. The PxChi liquid showed non-Newtonian pseudoplastic behavior of high consistency and difficult manipulation, and the gel retained 100% of radiolabel after 150 minutes. The PxCBP liquid showed a Newtonian behavior of low viscosity and easy manipulation, while in the gel phase showed apparent viscosity similar to that of the mucus but higher than that of aqueous solution. The gel released 35% of radiolabel and the released material showed silencing activity in vitro. Three intranasal doses of dendriplexes in PxCBP gel did not damage the rat nasal mucosa. A combination of (32)P-siRNA complexation with dendrimers, incorporation of the dendriplexes into PxCBP gel, and administration of two intranasal doses was necessary to achieve higher brain radioactivity than that achieved by intravenous dendriplexes or intranasal naked siRNA. CONCLUSION: The increased radioactivity within the olfactory bulb suggested that the combination above mentioned favored the mediation of a direct brain delivery.
Subject(s)
Brain Chemistry/drug effects , Gels/pharmacology , Nasal Mucosa/metabolism , Phosphorus Isotopes/pharmacokinetics , RNA, Small Interfering/pharmacokinetics , Administration, Intranasal/methods , Animals , Brain/metabolism , Chitosan/chemistry , Dendrimers/chemistry , Elastic Modulus , Gels/administration & dosage , Gels/adverse effects , Gels/chemistry , Histocytochemistry , Male , Nasal Mucosa/chemistry , Nasal Mucosa/drug effects , Phosphorus Isotopes/administration & dosage , Phosphorus Isotopes/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Rats , Rats, Sprague-Dawley , Temperature , ViscosityABSTRACT
BACKGROUND: Gene silencing using small interfering RNA (siRNA) is a promising new therapeutic approach for glioblastoma. The endocytic uptake and delivery of siRNA to intracellular compartments could be enhanced by complexation with polyamidoamine dendrimers. In the present work, the uptake mechanisms and intracellular traffic of siRNA/generation 7 dendrimer complexes (siRNA dendriplexes) were screened in T98G glioblastoma and J774 macrophages. METHODS: The effect of a set of chemical inhibitors of endocytosis on the uptake and silencing capacity of dendriplexes was determined by flow cytometry. Colocalization of fluorescent dendriplexes with endocytic markers and occurrence of intracellular dissociation were assessed by confocal laser scanning microscopy. RESULTS: Uptake of siRNA dendriplexes by T98G cells was reduced by methyl-ß-cyclodextrin, and genistein, and cytochalasine D, silencing activity was reduced by genistein; dendriplexes colocalized with cholera toxin subunit B. Therefore, caveolin-dependent endocytosis was involved both in the uptake and silencing activity of siRNA dendriplexes. On the other hand, uptake of siRNA dendriplexes by J774 cells was reduced by methyl-ß-cyclodextrin, genistein, chlorpromazine, chloroquine, cytochalasine D, and nocodazole, the silencing activity was not affected by chlorpromazine, genistein or chloroquine, and dendriplexes colocalized with transferrin and cholera toxin subunit B. Thus, both clathrin-dependent and caveolin-dependent endocytosis mediated the uptake and silencing activity of the siRNA dendriplexes. SiRNA dendriplexes were internalized at higher rates by T98G but induced lower silencing than in J774 cells. SiRNA dendriplexes showed relatively slow dissociation kinetics, and their escape towards the cytosol was not mediated by acidification independently of the uptake pathway. CONCLUSION: The extent of cellular uptake of siRNA dendriplexes was inversely related to their silencing activity. The higher silencing activity of siRNA dendriplexes in J774 cells could be ascribed to the contribution of clathrin-dependent and caveolin-dependent endocytosis vs only caveolin-dependent endocytosis in T98G cells.
Subject(s)
Dendrimers/pharmacokinetics , Endocytosis/drug effects , Gene Silencing , Glioblastoma/metabolism , Macrophages/metabolism , RNA, Small Interfering/pharmacokinetics , Analysis of Variance , Animals , Biomarkers/metabolism , Caveolins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Clathrin/metabolism , Dendrimers/administration & dosage , Endosomes/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Intracellular Space/drug effects , Macrophages/drug effects , Mice , RNA, Small Interfering/administration & dosageABSTRACT
We have screened the formation of complexes between ethylendiamine (EDA) core polyamidoamine (PAMAM) dendrimers (D) and a short interfering RNA (siRNA) as a function of three variables: the ionic strength of the medium (lacking or containing 150 mM NaCl), the D generation (G4, G5, G6 and G7) and the N/P ratio (nitrogen amines in D/phosphate in siRNA). It was observed that all D formed complexes with siRNA, being the size of the complexes strictly dependent on the ionic strength of the media. The strong electrostatic interactions occurring in NaCl lacking medium made siRNA-D complexes (siRNA-D) smaller than those obtained in NaCl containing medium (30-130 nm, +25 mV zeta potential vs. several microm-800 nm, 0 zeta potential, respectively). Not surprisingly, both the uptake and inhibition of EGFP expression in cell culture, resulted dependent on siRNA-D size. siRNA-D prepared in NaCl containing medium were poorly captured and presented a basal activity on phagocytic (J-774-EGFP) cells, being inactive on non-phagocytic cells (T98G-EGFP). However, the smaller siRNA-D prepared in NaCl lacking medium were massively captured, exhibiting the highest inhibition of EGFP expression at 50 nM siRNA (non-cytotoxic concentration). Remarkably, siRNA-G7 produced the highest inhibition of EGFP expression both in T98G-EGFP (35%) and J-774-EGFP (45%) cells, in spite of inducing a lower protection of siRNA against RNase A degradation. Taken together, our results showed that modifying the chemical structure of D is not the only way of achieving siRNA-D suitable for silencing activity. The simple use of a low ionic strength preparation media has been critical to get small siRNA-D that could be captured by cells and in particular, siRNA-G7 but not those formed by lower generation D, possessed structural constraints other than size that could favor its silencing activity.