Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(11): e2219523120, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36893269

RESUMEN

The continuous evolution of SARS-CoV-2 variants complicates efforts to combat the ongoing pandemic, underscoring the need for a dynamic platform for the rapid development of pan-viral variant therapeutics. Oligonucleotide therapeutics are enhancing the treatment of numerous diseases with unprecedented potency, duration of effect, and safety. Through the systematic screening of hundreds of oligonucleotide sequences, we identified fully chemically stabilized siRNAs and ASOs that target regions of the SARS-CoV-2 genome conserved in all variants of concern, including delta and omicron. We successively evaluated candidates in cellular reporter assays, followed by viral inhibition in cell culture, with eventual testing of leads for in vivo antiviral activity in the lung. Previous attempts to deliver therapeutic oligonucleotides to the lung have met with only modest success. Here, we report the development of a platform for identifying and generating potent, chemically modified multimeric siRNAs bioavailable in the lung after local intranasal and intratracheal delivery. The optimized divalent siRNAs showed robust antiviral activity in human cells and mouse models of SARS-CoV-2 infection and represent a new paradigm for antiviral therapeutic development for current and future pandemics.


Asunto(s)
COVID-19 , Humanos , Animales , Ratones , ARN Interferente Pequeño/genética , COVID-19/terapia , SARS-CoV-2/genética , Antivirales/farmacología , Antivirales/uso terapéutico , Oligonucleótidos , Pulmón
2.
Mol Ther ; 30(8): 2709-2721, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35477658

RESUMEN

Aberrant activation of interferon (IFN)-γ signaling plays a key role in several autoimmune skin diseases, including lupus erythematosus, alopecia areata, vitiligo, and lichen planus. Here, we identify fully chemically modified small interfering RNAs (siRNAs) that silence the ligand binding chain of the IFN-γ receptor (IFNGR1), for the modulation of IFN-γ signaling. Conjugating these siRNAs to docosanoic acid (DCA) enables productive delivery to all major skin cell types local to the injection site, with a single dose of injection supporting effective IFNGR1 protein reduction for at least 1 month in mice. In an ex vivo model of IFN-γ signaling, DCA-siRNA efficiently inhibits the induction of IFN-γ-inducible chemokines, CXCL9 and CXCL10, in skin biopsies from the injection site. Our data demonstrate that DCA-siRNAs can be engineered for functional gene silencing in skin and establish a path toward siRNA treatment of autoimmune skin diseases.


Asunto(s)
Quimiocina CXCL10 , Enfermedades de la Piel , Animales , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Interferón gamma/metabolismo , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética
3.
RNA Biol ; 19(1): 313-332, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35188077

RESUMEN

RNA-based therapeutics are emerging as a powerful platform for the treatment of multiple diseases. Currently, the two main categories of nucleic acid therapeutics, antisense oligonucleotides and small interfering RNAs (siRNAs), achieve their therapeutic effect through either gene silencing, splicing modulation or microRNA binding, giving rise to versatile options to target pathogenic gene expression patterns. Moreover, ongoing research seeks to expand the scope of RNA-based drugs to include more complex nucleic acid templates, such as messenger RNA, as exemplified by the first approved mRNA-based vaccine in 2020. The increasing number of approved sequences and ongoing clinical trials has attracted considerable interest in the chemical development of oligonucleotides and nucleic acids as drugs, especially since the FDA approval of the first siRNA drug in 2018. As a result, a variety of innovative approaches is emerging, highlighting the potential of RNA as one of the most prominent therapeutic tools in the drug design and development pipeline. This review seeks to provide a comprehensive summary of current efforts in academia and industry aimed at fully realizing the potential of RNA-based therapeutics. Towards this, we introduce established and emerging RNA-based technologies, with a focus on their potential as biosensors and therapeutics. We then describe their mechanisms of action and their application in different disease contexts, along with the strengths and limitations of each strategy. Since the nucleic acid toolbox is rapidly expanding, we also introduce RNA minimal architectures, RNA/protein cleavers and viral RNA as promising modalities for new therapeutics and discuss future directions for the field.


Asunto(s)
Terapia Genética , ARN/genética , ARN/uso terapéutico , Investigación , Animales , Biotecnología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Terapia Genética/tendencias , Humanos , Nanotecnología , Oligonucleótidos Antisentido , ARN/química , ARN Mensajero , ARN Interferente Pequeño , Investigación/tendencias
4.
Mol Ther ; 29(4): 1382-1394, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33348054

RESUMEN

Oligonucleotide therapeutics hold promise for the treatment of muscle- and heart-related diseases. However, oligonucleotide delivery across the continuous endothelium of muscle tissue is challenging. Here, we demonstrate that docosanoic acid (DCA) conjugation of small interfering RNAs (siRNAs) enables efficient (~5% of injected dose), sustainable (>1 month), and non-toxic (no cytokine induction at 100 mg/kg) gene silencing in both skeletal and cardiac muscles after systemic injection. When designed to target myostatin (muscle growth regulation gene), siRNAs induced ~55% silencing in various muscle tissues and 80% silencing in heart, translating into a ~50% increase in muscle volume within 1 week. Our study identifies compounds for RNAi-based modulation of gene expression in skeletal and cardiac muscles, paving the way for both functional genomics studies and therapeutic gene modulation in muscle and heart.


Asunto(s)
Ácidos Grasos/farmacología , Técnicas de Transferencia de Gen , Miostatina/genética , Oligonucleótidos/farmacología , ARN Interferente Pequeño/farmacología , Animales , Modelos Animales de Enfermedad , Ácidos Grasos/química , Corazón/efectos de los fármacos , Corazón/fisiopatología , Cardiopatías/genética , Cardiopatías/patología , Cardiopatías/terapia , Humanos , Ratones , Músculo Esquelético/efectos de los fármacos , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Miocardio/patología , Miostatina/antagonistas & inhibidores , Oligonucleótidos/química , Oligonucleótidos/genética , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética
5.
Nucleic Acids Res ; 48(14): 7665-7680, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32672813

RESUMEN

Small interfering RNAs (siRNAs) have revolutionized the treatment of liver diseases. However, robust siRNA delivery to other tissues represents a major technological need. Conjugating lipids (e.g. docosanoic acid, DCA) to siRNA supports extrahepatic delivery, but tissue accumulation and gene silencing efficacy are lower than that achieved in liver by clinical-stage compounds. The chemical structure of conjugated siRNA may significantly impact invivo efficacy, particularly in tissues with lower compound accumulation. Here, we report the first systematic evaluation of the impact of siRNA scaffold-i.e. structure, phosphorothioate (PS) content, linker composition-on DCA-conjugated siRNA delivery and efficacy in vivo. We found that structural asymmetry (e.g. 5- or 2-nt overhang) has no impact on accumulation, but is a principal factor for enhancing activity in extrahepatic tissues. Similarly, linker chemistry (cleavable versus stable) altered activity, but not accumulation. In contrast, increasing PS content enhanced accumulation of asymmetric compounds, but negatively impacted efficacy. Our findings suggest that siRNA tissue accumulation does not fully define efficacy, and that the impact of siRNA chemical structure on activity is driven by intracellular re-distribution and endosomal escape. Fine-tuning siRNA chemical structure for optimal extrahepatic efficacy is a critical next step for the progression of therapeutic RNAi applications beyond liver.


Asunto(s)
Oligonucleótidos Fosforotioatos/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/farmacocinética , Animales , Femenino , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Interferencia de ARN , Distribución Tisular
6.
Nucleic Acids Res ; 47(3): 1082-1096, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30544191

RESUMEN

Small interfering RNA (siRNA)-based therapies are proving to be efficient for treating liver-associated disorders. However, extra-hepatic delivery remains challenging, limiting therapeutic siRNA utility. We synthesized a panel of fifteen lipid-conjugated siRNAs and systematically evaluated the impact of conjugate on siRNA tissue distribution and efficacy. Generally, conjugate hydrophobicity defines the degree of clearance and the liver-to-kidney distribution profile. In addition to primary clearance tissues, several conjugates achieve significant siRNA accumulation in muscle, lung, heart, adrenal glands and fat. Oligonucleotide distribution to extra-hepatic tissues with some conjugates was significantly higher than with cholesterol, a well studied conjugate, suggesting that altering conjugate structure can enhance extra-hepatic delivery. These conjugated siRNAs enable functional gene silencing in lung, muscle, fat, heart and adrenal gland. Required levels for productive silencing vary (5-200 µg/g) per tissue, suggesting that the chemical nature of conjugates impacts tissue-dependent cellular/intracellular trafficking mechanisms. The collection of conjugated siRNA described here enables functional gene modulation in vivo in several extra-hepatic tissues opening these tissues for gene expression modulation. A systemic evaluation of a panel of conjugated siRNA, as reported here, has not previously been investigated and shows that chemical engineering of lipid siRNAs is essential to advance the RNA therapeutic field.


Asunto(s)
Lípidos/química , ARN Interferente Pequeño/farmacocinética , Animales , Carbocianinas , Colesterol , Ácidos Grasos , Femenino , Colorantes Fluorescentes , Riñón/metabolismo , Hígado/metabolismo , Ratones , Fosforilcolina , Interferencia de ARN , ARN Interferente Pequeño/síntesis química , Distribución Tisular
7.
Nucleic Acids Res ; 47(3): 1070-1081, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30535404

RESUMEN

Efficient delivery of therapeutic RNA beyond the liver is the fundamental obstacle preventing its clinical utility. Lipid conjugation increases plasma half-life and enhances tissue accumulation and cellular uptake of small interfering RNAs (siRNAs). However, the mechanism relating lipid hydrophobicity, structure, and siRNA pharmacokinetics is unclear. Here, using a diverse panel of biologically occurring lipids, we show that lipid conjugation directly modulates siRNA hydrophobicity. When administered in vivo, highly hydrophobic lipid-siRNAs preferentially and spontaneously associate with circulating low-density lipoprotein (LDL), while less lipophilic lipid-siRNAs bind to high-density lipoprotein (HDL). Lipid-siRNAs are targeted to lipoprotein receptor-enriched tissues, eliciting significant mRNA silencing in liver (65%), adrenal gland (37%), ovary (35%), and kidney (78%). Interestingly, siRNA internalization may not be completely driven by lipoprotein endocytosis, but the extent of siRNA phosphorothioate modifications may also be a factor. Although biomimetic lipoprotein nanoparticles have been explored for the enhancement of siRNA delivery, our findings suggest that hydrophobic modifications can be leveraged to incorporate therapeutic siRNA into endogenous lipid transport pathways without the requirement for synthetic formulation.


Asunto(s)
Lípidos/química , ARN Interferente Pequeño/farmacocinética , Animales , Proteínas Sanguíneas/metabolismo , Femenino , Células HeLa , Hepatocitos/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Riñón/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Interferencia de ARN , ARN Interferente Pequeño/síntesis química , ARN Interferente Pequeño/química , Receptores de LDL/metabolismo , Distribución Tisular
8.
Mol Ther ; 26(6): 1520-1528, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29699940

RESUMEN

Small extracellular vesicles (sEVs) show promise as natural nano-devices for delivery of therapeutic RNA, but efficient loading of therapeutic RNA remains a challenge. We have recently shown that the attachment of cholesterol to small interfering RNAs (siRNAs) enables efficient and productive loading into sEVs. Here, we systematically explore the ability of lipid conjugates-fatty acids, sterols, and vitamins-to load siRNAs into sEVs and support gene silencing in primary neurons. Hydrophobicity of the conjugated siRNAs defined loading efficiency and the silencing activity of siRNA-sEVs complexes. Vitamin-E-conjugated siRNA supported the best loading into sEVs and productive RNA delivery to neurons.


Asunto(s)
Vesículas Extracelulares/química , Lípidos/química , ARN Interferente Pequeño/química , Células Cultivadas , Silenciador del Gen/fisiología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Interferencia de ARN
9.
Mol Ther ; 26(8): 1973-1982, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29937418

RESUMEN

Extracellular vesicles are promising delivery vesicles for therapeutic RNAs. Small interfering RNA (siRNA) conjugation to cholesterol enables efficient and reproducible loading of extracellular vesicles with the therapeutic cargo. siRNAs are typically chemically modified to fit an application. However, siRNA chemical modification pattern has not been specifically optimized for extracellular vesicle-mediated delivery. Here we used cholesterol-conjugated, hydrophobically modified asymmetric siRNAs (hsiRNAs) to evaluate the effect of backbone, 5'-phosphate, and linker chemical modifications on productive hsiRNA loading onto extracellular vesicles. hsiRNAs with a combination of 5'-(E)-vinylphosphonate and alternating 2'-fluoro and 2'-O-methyl backbone modifications outperformed previously used partially modified siRNAs in extracellular vesicle-mediated Huntingtin silencing in neurons. Between two commercially available linkers (triethyl glycol [TEG] and 2-aminobutyl-1-3-propanediol [C7]) widely used to attach cholesterol to siRNAs, TEG is preferred compared to C7 for productive exosomal loading. Destabilization of the linker completely abolished silencing activity of loaded extracellular vesicles. The loading of cholesterol-conjugated siRNAs was saturated at ∼3,000 siRNA copies per extracellular vesicle. Overloading impaired the silencing activity of extracellular vesicles. The data reported here provide an optimization scheme for the successful use of hydrophobic modification as a strategy for productive loading of RNA cargo onto extracellular vesicles.


Asunto(s)
Colesterol/química , Vesículas Extracelulares/química , Proteína Huntingtina/genética , ARN Interferente Pequeño/química , Animales , Células Cultivadas , Humanos , Ratones , Mutación , Glicoles de Propileno/química
10.
Mol Ther ; 26(11): 2580-2591, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30143435

RESUMEN

Effective transvascular delivery of therapeutic oligonucleotides to the brain presents a major hurdle to the development of gene silencing technologies for treatment of genetically defined neurological disorders. Distribution to the brain after systemic administrations is hampered by the low permeability of the blood-brain barrier (BBB) and the rapid clearance kinetics of these drugs from the blood. Here we show that transient osmotic disruption of the BBB enables transvascular delivery of hydrophobically modified small interfering RNA (hsiRNA) to the rat brain. Intracarotid administration of 25% mannitol and hsiRNA conjugated to phosphocholine-docosahexanoic acid (PC-DHA) resulted in broad ipsilateral distribution of PC-DHA-hsiRNAs in the brain. PC-DHA conjugation enables hsiRNA retention in the parenchyma proximal to the brain vasculature and enabled active internalization by neurons and astrocytes. Moreover, transvascular delivery of PC-DHA-hsiRNAs effected Htt mRNA silencing in the striatum (55%), hippocampus (51%), somatosensory cortex (52%), motor cortex (37%), and thalamus (33%) 1 week after administration. Aside from mild gliosis induced by osmotic disruption of the BBB, transvascular delivery of PC-DHA-hsiRNAs was not associated with neurotoxicity. Together, these findings provide proof-of-concept that temporary disruption of the BBB is an effective strategy for the delivery of therapeutic oligonucleotides to the brain.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Proteína Huntingtina/genética , Neuronas/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Arterias Carótidas/fisiología , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/química , Silenciador del Gen , Terapia Genética/métodos , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Interacciones Hidrofóbicas e Hidrofílicas , Manitol/administración & dosificación , Neuronas/patología , Fosforilcolina/administración & dosificación , Fosforilcolina/química , ARN Interferente Pequeño/química , Ratas
11.
Org Biomol Chem ; 16(17): 3181-3188, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29645048

RESUMEN

The synthesis and the impact of a disulfide bridge between 2'-O-positions of two adjacent nucleotides in an RNA duplex and in the loop of RNA hairpins are reported. The incorporation of this 2',2'-disulfide (S-S) bridge enabled thermal and enzymatic stabilization of the hairpin depending on its position in the loop. The influence of the disulfide bridge on RNA folding was studied at the HIV Dimerization Initiation Site (DIS) as an RNA sequence model. We have shown that this S-S bridge locked the hairpin form, whereas the extended duplex form was generated after the reduction of the disulfide bond in the presence of tris(2-carboxyethyl)phosphine or glutathione. Thus, the S-S bridge can be useful for understanding RNA folding; an RNA molecular beacon locked by an S-S bridge was also investigated as a sensor for the detection of glutathione.


Asunto(s)
Disulfuros/química , VIH-1/química , ARN Viral/química , Secuencia de Bases , Infecciones por VIH/virología , Humanos , Cinética , Conformación de Ácido Nucleico , Desnaturalización de Ácido Nucleico , Oligorribonucleótidos/química , Oxidación-Reducción , Pliegue del ARN , Estabilidad del ARN
12.
Bioconjug Chem ; 28(6): 1758-1766, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28462988

RESUMEN

Ligand-conjugated siRNAs have the potential to achieve targeted delivery and efficient silencing in neurons following local administration in the central nervous system (CNS). We recently described the activity and safety profile of a docosahexaenoic acid (DHA)-conjugated, hydrophobic siRNA (DHA-hsiRNA) targeting Huntingtin (Htt) mRNA in mouse brain. Here, we report the synthesis of an amide-modified, phosphocholine-containing DHA-hsiRNA conjugate (PC-DHA-hsiRNA), which closely resembles the endogenously esterified biological structure of DHA. We hypothesized that this modification may enhance neuronal delivery in vivo. We demonstrate that PC-DHA-hsiRNA silences Htt in mouse primary cortical neurons and astrocytes. After intrastriatal delivery, Htt-targeting PC-DHA-hsiRNA induces ∼80% mRNA silencing and 71% protein silencing after 1 week. However, PC-DHA-hsiRNA did not substantially outperform DHA-hsiRNA under the conditions tested. Moreover, at the highest locally administered dose (4 nmol, 50 µg), we observe evidence of PC-DHA-hsiRNA-mediated reactive astrogliosis. Lipophilic ligand conjugation enables siRNA delivery to neural tissues, but rational design of functional, nontoxic siRNA conjugates for CNS delivery remains challenging.


Asunto(s)
Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Tejido Parenquimatoso/metabolismo , ARN Interferente Pequeño/síntesis química , Animales , Encéfalo/patología , Ácidos Docosahexaenoicos/química , Estabilidad de Medicamentos , Silenciador del Gen , Proteína Huntingtina/genética , Ratones , Fosforilcolina/química , Interferencia de ARN , ARN Mensajero , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/uso terapéutico , Serina/química , Resultado del Tratamiento
13.
Chembiochem ; 17(21): 2054-2062, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27569765

RESUMEN

The in vivo application of siRNA depends on its cellular uptake and intracellular release, and this is an unsatisfactorily resolved technical hurdle in medicinal applications. Promising concepts directed towards providing efficient cellular and intracellular delivery include lipophilic chemical modification of siRNA. Here we describe chemistry for the production of modified siRNAs designed to display improved transmembrane transport into human cells while preserving the potency of the RNAi-based inhibitors. We report the synthesis and the biochemical and biophysical characteristics of 2'-O-phenylisobutyryloxymethyl (PiBuOM)-modified siRNAs and their impact on biological activity. In the case of spontaneous cellular uptake of naked PiBuOM-modified siRNA, we observed increased target suppression in human cells relative to unmodified or pivaloyloxymethyl (PivOM)-modified siRNA. We provide evidence of improved spontaneous cellular uptake of naked PiBuOM-modified siRNA and of substantial target suppression in human cells in serum-containing medium.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ésteres/química , Estabilidad del ARN , ARN Interferente Pequeño/síntesis química , ARN Interferente Pequeño/metabolismo , Ribonucleasas/metabolismo , Temperatura , Línea Celular Tumoral , Ésteres/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química
14.
Org Biomol Chem ; 14(29): 7010-7, 2016 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-27356960

RESUMEN

An original post-synthetic method on a solid support was developed to introduce various disulfide bond containing groups at the 2'-OH of oligoribonucleotides (RNAs). It is based on a thiol disulfide exchange reaction between several readily accessible alkyldisulfanyl-pyridine derivatives and 2'-O-acetylthiomethyl RNA in the presence of butylamine. By this strategy, diverse 2'-O-alkyldithiomethyl RNAs were obtained. These modifications provided high nuclease resistance to RNA and were easily removed with glutathione treatment, thus featuring a potential use for siRNA prodrugs.


Asunto(s)
Profármacos/síntesis química , ARN Interferente Pequeño/síntesis química , ARN/química , Disulfuros/química , Estructura Molecular , Oxidación-Reducción , Profármacos/química , ARN Interferente Pequeño/química
15.
Bioorg Med Chem ; 23(17): 5360-8, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26260340

RESUMEN

We report on the synthesis and properties of oligonucleotides (ONs) with 2'-O-acetalester modifications containing cationic side chains in a prodrug-like approach. In the aim to improve cell penetration and nuclease resistance, various different amino- or guanidino-acetalester were grafted to 2'-OH of uridine and the corresponding phosphoramidites were incorporated into ONs. Introduction of 2'-O-(2-aminomethyl-2-ethyl)butyryloxymethyl (AMEBuOM) modification into 2'-OMe ONs leads to high resistance towards enzymatic degradation and to destabilization of duplexes with complementary RNA strand. Spontaneous uptake experiments of a twelve-mer containing ten 2'-O-AMEBuOM-U units into A673 cells showed moderate internalization of ON within the cells whereas substantial internalization of the corresponding lipophilic 2'-O-pivaloyloxymethyl ON was observed for the first time.


Asunto(s)
Oligonucleótidos/química , Oligonucleótidos/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Secuencia de Bases , Cationes/química , Cationes/metabolismo , Cationes/farmacocinética , Bovinos , Línea Celular , Esterificación , Humanos , Oligonucleótidos/farmacocinética , Compuestos Organofosforados/química , Serpientes , Uridina/química
16.
Chembiochem ; 15(18): 2674-9, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25346406

RESUMEN

An original and straightforward synthesis of partially modified 2'-O-pivaloyloxymethyl-substituted (PivOM-substituted) oligoribonucleotides has been achieved. The aim of this 2'-enzymolabile modification was to enhance nuclease stability of RNA and transmembrane transport. To make these modified RNAs easily available we developed a base-labile protecting group strategy with standard protections for nucleobases (acyl) and phosphates (cyanoethyl), a Q-linker and two different acetalester protection groups for 2'-OH: propionyloxymethyl (PrOM) and PivOM. Interestingly, orthogonal deprotection conditions based on anhydrous butylamine in THF were found to remove propionyloxymethyl groups selectively, while preserving PivOM groups. Duplex stability, circular dichroism studies and nuclease resistance, as well as the ability to inhibit gene expression of modified 2'-O-PivOM RNA, were evaluated.


Asunto(s)
Oligorribonucleótidos/química , ARN/química , Secuencia de Bases , Silenciador del Gen , Metilación , Oligorribonucleótidos/síntesis química , Profármacos/metabolismo , ARN/síntesis química
17.
Mol Ther Nucleic Acids ; 35(1): 102088, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38192611

RESUMEN

Inherited retinal dystrophies caused by dominant mutations in photoreceptor (PR) cell expressed genes are a major cause of irreversible vision loss. Oligonucleotide therapy has been of interest in diseases that conventional medicine cannot target. In the early days, small interfering RNAs (siRNAs) were explored in clinical trials for retinal disorders with limited success due to a lack of stability and efficient cellular delivery. Thus, an unmet need exists to identify siRNA chemistry that targets PR cell expressed genes. Here, we evaluated 12 different fully chemically modified siRNA configurations, where the valency and conjugate structure were systematically altered. The impact on retinal distribution following intravitreal delivery was examined. We found that the increase in valency (tetravalent siRNA) supports the best PR accumulation. A single intravitreal administration induces multimonths efficacy in rodent and porcine retinas while demonstrating a good safety profile. The data suggest that this configuration can treat retinal diseases caused by PR cell expressed genes with 1-2 intravitreal injections per year.

18.
Cells ; 12(18)2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37759475

RESUMEN

The potential of oligonucleotide therapeutics is undeniable as more than 15 drugs have been approved to treat various diseases in the liver, central nervous system (CNS), and muscles. However, achieving effective delivery of oligonucleotide therapeutics to specific tissues still remains a major challenge, limiting their widespread use. Chemical modifications play a crucial role to overcome biological barriers to enable efficient oligonucleotide delivery to the tissues/cells of interest. They provide oligonucleotide metabolic stability and confer favourable pharmacokinetic/pharmacodynamic properties. This review focuses on the various chemical approaches implicated in mitigating the delivery problem of oligonucleotides and their limitations. It highlights the importance of linkers in designing oligonucleotide conjugates and discusses their potential role in escaping the endosomal barrier, a bottleneck in the development of oligonucleotide therapeutics.


Asunto(s)
Sistema Nervioso Central , Endosomas , Hígado , Músculos , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico
19.
bioRxiv ; 2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37790464

RESUMEN

Inherited retinal dystrophies caused by dominant mutations in photoreceptor-expressed genes, are a major cause of irreversible vision loss. Oligonucleotide therapy has been of interest in diseases that conventional medicine cannot target. In the early days, small interfering RNAs (siRNAs) were explored in clinical trials for retinal disorders with limited success due to a lack of stability and efficient cellular delivery. Thus, an unmet need exists to identify siRNA chemistry that targets photoreceptor-expressed genes. Here we evaluated 12 different fully chemically modified siRNA configurations, where the valency and conjugate structure were systematically altered. The impact on retinal distribution following intravitreal delivery was examined. We found that the increase in valency (tetravalent siRNA) supports the best photoreceptor accumulation. A single intravitreal administration induces multi-months efficacy in rodent and porcine retinas while showing a good safety profile. The data suggest that this configuration can treat retinal diseases caused by photoreceptor-expressed genes with 1-2 intravitreal injections per year.

20.
Nat Commun ; 14(1): 7099, 2023 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-37925520

RESUMEN

Inhibition of Janus kinase (JAK) family enzymes is a popular strategy for treating inflammatory and autoimmune skin diseases. In the clinic, small molecule JAK inhibitors show distinct efficacy and safety profiles, likely reflecting variable selectivity for JAK subtypes. Absolute JAK subtype selectivity has not yet been achieved. Here, we rationally design small interfering RNAs (siRNAs) that offer sequence-specific gene silencing of JAK1, narrowing the spectrum of action on JAK-dependent cytokine signaling to maintain efficacy and improve safety. Our fully chemically modified siRNA supports efficient silencing of JAK1 expression in human skin explant and modulation of JAK1-dependent inflammatory signaling. A single injection into mouse skin enables five weeks of duration of effect. In a mouse model of vitiligo, local administration of the JAK1 siRNA significantly reduces skin infiltration of autoreactive CD8+ T cells and prevents epidermal depigmentation. This work establishes a path toward siRNA treatments as a new class of therapeutic modality for inflammatory and autoimmune skin diseases.


Asunto(s)
Inhibidores de las Cinasas Janus , Vitíligo , Ratones , Animales , Humanos , ARN Interferente Pequeño/genética , Linfocitos T CD8-positivos/metabolismo , Autoinmunidad/genética , Vitíligo/tratamiento farmacológico , Vitíligo/genética , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , ARN Bicatenario
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA