key: cord-332006-if46jycd authors: Whitehead, Kathryn A.; Langer, Robert; Anderson, Daniel G. title: Knocking down barriers: advances in siRNA delivery date: 2009 journal: Nat Rev Drug Discov DOI: 10.1038/nrd2742 sha: doc_id: 332006 cord_uid: if46jycd In the 10 years that have passed since the Nobel prize-winning discovery of RNA interference (RNAi), billions of dollars have been invested in the therapeutic application of gene silencing in humans. Today, there are promising data from ongoing clinical trials for the treatment of age-related macular degeneration and respiratory syncytial virus. Despite these early successes, however, the widespread use of RNAi therapeutics for disease prevention and treatment requires the development of clinically suitable, safe and effective drug delivery vehicles. Here, we provide an update on the progress of RNAi therapeutics and highlight novel synthetic materials for the encapsulation and intracellular delivery of nucleic acids. RNA interference (RNAi) gained international attention in 1998 when Fire, Mello and colleagues discovered the ability of double-stranded RNA to silence gene expression in the nematode worm Caenorhabditis elegans 1 . Three years later, Tuschl and co-workers published their celebrated proof-of-principle experiment demonstrating that synthetic small interfering RNA (siRNA) could achieve sequence-specific gene knockdown in a mammalian cell line 2 . The first successful use of siRNA for gene silencing in mice was achieved for a hepatitis C target shortly thereafter 3 . Since that time, the biotechnology sector has made considerable efforts in the advancement of siRNA therapeutics for the treatment of various disease targets, including viral infections 4,5 and cancer [6] [7] [8] . RNAi is a fundamental pathway in eukaryotic cells by which sequence-specific siRNA is able to target and cleave complementary mRNA 2 . RNAi is triggered by the presence of long pieces of double-stranded RNA, which are cleaved into the fragments known as siRNA (21) (22) (23) nucleotides long) by the enzyme Dicer 9 . In practice, siRNA can be synthetically produced and then directly introduced into the cell, thus circumventing Dicer mechanics (FIG. 1) . This shortcut reduces the potential for an innate immune interferon response and the shutdown of cellular protein expression that can occur following the interaction of long pieces (>30 nucleotides) of doublestranded RNA with intracellular RNA receptors 10 . Once siRNA is present in the cytoplasm of the cell, it is incorporated into a protein complex called the RNAinduced silencing complex (RISC) 11 . Argonaute 2, a multifunctional protein contained within RISC, unwinds the siRNA, after which the sense strand (or passenger strand) of the siRNA is cleaved 12 . The activated RISC, which contains the antisense strand (or guide strand) of the siRNA, selectively seeks out and degrades mRNA that is complementary to the antisense strand 13 (FIG. 1) . The cleavage of mRNA occurs at a position between nucleo tides 10 and 11 on the complementary antisense strand, relative to the 5′-end 14 . The activated RISC complex can then move on to destroy additional mRNA targets, which further propagates gene silencing 15 . This extra potency ensures a therapeutic effect for 3-7 days in rapidly dividing cells, and for several weeks in non-dividing cells 16 . Eventually, siRNAs are diluted below a certain therapeutic threshold or degraded within the cell, and so repeated administration is necessary to achieve a persistent effect. Theoretically, when using appropriately designed siRNA, the RNAi machinery can be exploited to silence nearly any gene in the body, giving it a broader therapeutic potential than typical small-molecule drugs. Indeed, it has already been reported that synthetic siRNAs are capable of knocking down targets in various diseases in vivo, including hypercholesterolaemia 17 , liver cirr hosis 18 , hepatitis B virus (HBV) 4, 19 , human papillomavirus 20 , ovarian cancer 21 and bone cancer 22 . In order for these advances to be implemented in a clinical setting, safe and effective delivery systems must be developed. While 'naked' , chemically modified siRNA has shown efficacy in certain physiological settings such as the brain 23 and the lung 24 , there are many tissues in the body that require an additional delivery system to facilitate transfection. This is because naked siRNA is subject to degradation by endogenous enzymes, and is too large (~13 kDa) and too negatively charged to cross cellular membranes. The strand of the siRNA molecule that is complementary to the target mRNA, which activates RISC and has an important role in target mRNA identification and destruction. The process of delivering nucleic acid material into the cell. The issue of effective and non-toxic delivery is a key challenge and serves as the most significant barrier between siRNA technology and its therapeutic application. The ease of siRNA delivery is partly dependent on the accessibility of the target organ or tissue within the body. localized siRNA delivery -that is, application of siRNA therapy directly to the target tissue -offers several benefits, including the potential for both higher bioavailability given the proximity to the target tissue, and reduced adverse effects typically associated with systemic administration. By contrast, systemic delivery, meaning the intravenous injection of delivery particles that then travel throughout the body to the target organ or tissue, requires that particles have the ability to avoid uptake and clearance by non-target tissues (FIG. 2) . There are several tissues that are amenable to topical or localized therapy, including the eye, skin, mucus membranes, and local tumours [25] [26] [27] [28] (TABLE 1) . local siRNA delivery is particularly well-suited for the treatment of lung diseases and infections. The direct instillation of siRNA into the lung through intranasal or intratracheal routes enables direct contact with lung epithelial cells. These cells play a part in a myriad of lung conditions and infections, including cystic fibrosis, asthma, influenza and the common cold 24 . It has been reported that respiratory syncytial virus (RSV) replication can be inhibited by nasally administered siRNA formulated with or without transfection agents in mice 29, 30 . Progress in the treatment of RSV continues with Phase II clinical trials using an aerosolized siRNA delivery system 31 . Intratracheal administration of siRNA has also been reported to offer prophylactic and therapeutic effects in the treatment of severe acute respiratory syndrome 32 . Another example of local delivery is direct intratumoral injection of siRNA delivery complexes into various mouse xenograft models. siRNA complexed with the delivery agent polyethyleneimine (PEI) was shown to inhibit tumour growth upon intratumoral injection in mice bearing glioblastoma xenographs 28 . Niu and co-workers have also reported naked siRNA efficacy up on direct injection into a subcutaneous cervical cancer model in mice 20 . In contrast to the direct accessibility of localized targets, many tissues can only be reached through the systemic administration of delivery agents in the bloodstream. siRNA formulations for systemic application face a series of hurdles in vivo before reaching the cytoplasm of the target cell (FIG. 2) . Post-injection, the siRNA complex must navigate the circulatory system of the body while avoiding kidney filtration, uptake by phagocytes, aggregation with serum proteins, and enzymatic degradation by endogenous nucleases 33 . Phagocytosis serves as a significant immunological barrier, not only in the bloodstream but also in the extracellular matrix of tissues. Phagocytic cells such as macrophages and monocytes remove foreign material from the body to protect against infection by viruses, bacteria and fungi. unfortunately, phagocytes are also highly efficient at removing certain therapeutic nanocomplexes and macromolecules from the body, and steps must be taken to avoid opsonization when designing drug delivery vehicles 33 . Egress from the bloodstream and across the vascular endothelial barrier poses a significant challenge for delivery of siRNA to many tissues within the body. In general, molecules larger than 5 nm in diameter do not readily cross the capillary endothelium, and therefore will remain in the circulation until they are cleared from the body. There are certain tissues, however, that allow the entry of larger molecules, including the liver, spleen, and some tumours. These organs allow the passage of molecules up to 200 nm in diameter, which can accommodate a typical drug delivery nanocarrier 34 . is introduced into the cytoplasm, where it is cleaved into small interfering RNA (siRNA) by the enzyme Dicer. Alternatively, siRNA can be introduced directly into the cell. The siRNA is then incorporated into the RNA-induced silencing complex (RISC), resulting in the cleavage of the sense strand of RNA by argonaute 2 (AGO2). The activated RISC-siRNA complex seeks out, binds to and degrades complementary mRNA, which leads to the silencing of the target gene. The activated RISC-siRNA complex can then be recycled for the destruction of identical mRNA targets. After an siRNA complex leaves the bloodstream, it must diffuse through the extracellular matrix, which is a dense network of polysaccharides and fibrous proteins that can create resistance to the transport of macromolecules and nanoparticles 35 . This can slow or even halt the drug delivery process and create an additional opportunity for nanoparticles to be taken up by resident macrophages. Having been taken up by the target cell, particles must then escape the endosome to reach the cytoplasm 36 . If the siRNA nanocomplex is unable to exit the endosome, it will be trafficked through endomembrane compartments of decreasing pH and be subject to degradative conditions in the lysosome 37 . Finally, if formulated with delivery agents, siRNA must be released from the carrier to the cellular machinery. Modified siRNA for improved delivery Humans have evolved a number of host-defence mechanisms against siRNA, as it is a feature of certain viral infections. However, chemical modifications can be introduced into the siRNA molecule to evade immune defences in vivo. For example, many non-modified siRNAs can induce nonspecific activation of the immune system through the Toll-like receptor 7 (TlR7) pathway 38, 39 . This effect can be reduced by the incorporation of 2′-O-methyl modifications into the sugar structure of selected nucleotides within both the sense and antisense strands 38, 40 (FIG. 3a) . 2′-O-methyl modifications have also been shown to confer resistance to endo nuclease activity 41 and to abrogate off-target effects when incorporated into the seed region, which corresponds to nucleotides 2-8 on the antisense strand 42 . Other common modification approaches to mitigate enzymatic degradation include the introduction of phosphorothioate backbone linkages at the 3′-end of the RNA strands to reduce susceptibility to exonucleases. It is also possible to incorporate alternative 2′ sugar modifications (for example, a fluorine substitution) to increase resistance to endonucleases 43 . Another strategy to improve the therapeutic efficacy of siRNA involves the conjugation of small molecules or peptides to the sense strand of the siRNA. Several small molecules have been reported to increase target-gene knockdown in vitro, including membrane-permeant peptides 44 and polyethylene glycol (PEG) 45 . Of particular note are cholesterol-modified siRNAs, which have demonstrated increased binding to serum albumin, resulting in improved biodistribution to certain targets including the liver (FIG. 3b) . Cholesterol-modified siRNA were capable of silencing apolipoprotein B (ApoB) targets in mouse liver and jejunum, and of ultimately reducing total cholesterol levels 46 . Another study by DiFiglia and co-workers details the ability of a cholesterol-modified siRNA to knockdown a gene associated with Huntington's disease. A single intrastriatal injection was able to delay the abnormal behavioural phenotype observed in a rapid-onset mouse model of this disease 23 . Given the success of cholesterol-modified siRNA in vivo, Wolfrum and co-workers attempted to identify alternative lipid-like molecules to serve as RNA conjugates for improved delivery of siRNA 47 . Specifically, fatty acids and bile-salt derivatives were conjugated to siRNA and injected into mice and hamsters in order to elucidate how modified siRNA conjugates interact with the high-density lipoprotein (HDl) and low-density lipoprotein (lDl) receptors that enable delivery to the liver. It was found that shorter fatty-acid chain lengths (