Subscriber access provided by Northern Illinois University Review Cellular Delivery of RNA Nanoparticles Lorena Parlea, Anu Puri, Wojciech K. Kasprzak, Eckart Bindewald, Paul Zakrevsky, Emily Satterwhite, Kenya Joseph, Kirill A Afonin, and Bruce A Shapiro ACS Comb. Sci., Just Accepted Manuscript • DOI: 10.1021/acscombsci.6b00073 • Publication Date (Web): 10 Aug 2016 Downloaded from http://pubs.acs.org on August 21, 2016

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Combinatorial Science is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties. Page 1 of 81 ACS Combinatorial Science

1 2 3 Cellular Delivery of RNA Nanoparticles 4 5 6 7 8 Lorena Parlea a, Anu Puri a, Wojciech Kasprzak b, Eckart Bindewald b, Paul Zakrevsky a, Emily 9 10 c c c,d a* 11 Satterwhite , Kenya Joseph , Kirill A. Afonin and Bruce A. Shapiro 12 13 14 15 a Gene Regulation and Chromosome Biology Laboratory, National Cancer Institute, Frederick, MD, 16 17 18 21702, USA 19 20 b Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer 21 22 Research, Frederick, MD 21702, USA 23 24 c 25 Department of Chemistry, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, 26 27 USA 28 29 d Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte NC 28223, USA; The 30 31 32 Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte 33 34 NC 28223, USA. 35 36 37 38 39 * To whom correspondence should be addressed ( [email protected] ) 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 1 ACS Paragon Plus Environment ACS Combinatorial Science Page 2 of 81

1 2 3 Keywords : RNA, selfassembly, Nanoconstruct, RNA Nanoparticle, Therapeutics, Delivery 4 5 6 7 8 Abbreviations: 9 10 11 3D – three dimensional, kDA – kiloDalton, MD – molecular dynamics, NP – nanoparticles, nt – 12 13 nucleotide, OEL – Oxime ether lipids, polyethylenglycol – PEG, miRNA microRNA, piRNA – piwi 14 15 RNA, siRNA – small interfering RNA, RNAi – RNA interference, DS RNA – Dices substrate RNA, 16 17 18 RISC – RNAinduced Silencing Complex 19 20 21 22 Table of Contents 23 24 25 26 27 Keywords ...... 2 28 29 Abstract ...... 4 30 Introduction ...... 5 31 32 Functional ...... 5 33 34 Interfering RNAs ...... 5 35 Aptamers ...... 8 36 37 Splicing Modulators ...... 9 38 39 RNA Switches ...... 9 40 Ribozymes ...... 9 41 42 Exogenous mRNAs ...... 10 43 44 CRISPR RNAs ...... 10 45 46 RNA/DNA hybrids ...... 11 47 Rational RNA Nanostructure Design ...... 11 48 49 Manual design of RNA Nanostructures based on naturally occurring RNA 3D motifs ...... 13 50 51 Computational design of RNA Nanostructures using naturally occurring RNA 3D motifs ...... 14 52 3-D Motif-based nanostructures ...... 14 53 54 De-novo Design of RNA Scaffolds ...... 16 55 56 Functionalization of RNA Nanostructures ...... 17 57 Hybrid technology in RNA NPs ...... 18 58 59 60 2 ACS Paragon Plus Environment Page 3 of 81 ACS Combinatorial Science

1 2 3 Challenges in delivering RNAs ...... 20 4 5 Degradation ...... 21 6 7 Stability in blood ...... 22 8 Hepato-Renal Clearance ...... 22 9 10 Toxicity and Immunogenicity ...... 23 11 12 Cellular Uptake ...... 24 13 14 Endosomal Escape ...... 26 15 Off-target Effects ...... 26 16 17 Delivery methods for RNA therapeutics and RNA NPs ...... 26 18 19 “Naked” Delivery ...... 27 20 Mediated Delivery ...... 28 21 22 Polymers ...... 29 23 24 Hydrogels ...... 29 25 Peptides ...... 29 26 27 Dendrimers ...... 30 28 29 Conjugated Delivery ...... 30 30 31 Aptamers ...... 30 32 Inorganic nanoparticles ...... 32 33 34 Microsponges ...... 32 35 36 Encapsulated Delivery ...... 33 37 Cationic lipids ...... 33 38 39 Oxime Ether Lipids ...... 36 40 41 Bolaamphiphile surfactants ...... 36 42 Exosomes ...... 39 43 44 Viral Delivery ...... 39 45 46 Prospects in RNA Nanobiology ...... 39 47 48 Acknowledgements ...... 42 49 50 51 52 53 54 55 56 57 58 59 60 3 ACS Paragon Plus Environment ACS Combinatorial Science Page 4 of 81

1 2 3 Abstract 4 5 6 RNA nanostructures have capabilities to simultaneously incorporate different defined functions, 7 8 such as specific binding to a ligand, or programmable regulation of gene expression. Selfassembling 9 10 11 nanostructures containing functional RNAs provide further options for nanobiology applications. This 12 13 review is focused on four growing lists of importance to RNA nanotechnology: the types of RNAs of 14 15 particular interest for nanobiology, the assembly of RNA nanoconstructs, the challenges of cellular 16 17 18 delivery of RNAs in vivo , and the delivery carriers that aid in the matter. The available strategies for the 19 20 design of nucleic acid nanostructures, as well as for formulation of their carriers, make RNA 21 22 nanotechnology an important tool in both basic research and applied biomedical science. 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 4 ACS Paragon Plus Environment Page 5 of 81 ACS Combinatorial Science

1 2 3 Introduction 4 5 1, 2 6 While proteinbased nanotechnology is a wellexplored subject by now, nucleic acid 7 8 nanotechnology is a field still in its infancy. Both deoxyribo and ribonucleic acids (DNA and RNA, 9 10 311 11 respectively) have been used extensively to build various nanostructures some of which were 12 10, 12 13 functionalized towards therapeutic or diagnostic applications. The suitability of RNAs as a material 14 15 for use in nanotechnology and nanomedicine is inherent in their chemical, structural and functional 16 17 13 18 properties. Chemically, not only can RNA store genetic information, but it can also be easily 19 20 modified.14 Structurally, RNA can be modulated to form nanostructures of various shapes.15 RNA is 21 22 capable of selfassembling under physiological conditions 12, 16 through both intramolecular recognition 23 24 17 25 and long range intermolecular interactions. Functionally, RNA can perform catalytic, enzymelike 26 27 activities.1821 Furthermore, as a biological molecule, RNA is biocompatible and biodegradable, and thus 28 29 an excellent contender as a material for therapeutics and medicine. 30 31 32 33 34 Functional RNAs 35 36 RNAs are key molecules in a plethora of cellular processes, ranging from gene regulation to 37 38 39 protein synthesis. The central dogma of molecular biology states that DNA encodes for RNA and RNA 40 41 codes (in the form of mRNA) for proteins, but this is not the entire picture. Cellular noncoding RNAs 42 43 are involved in various other molecular processes such as splicing, intracellular transport and post 44 45 46 transcriptional regulation of gene expression. Several types of functional RNA or RNA elements have 47 48 particular interests for the RNA nanotechnology field, as seen below. 49 50 51 Interfering RNAs 52 53 RNA interference (RNAi) is an RNAmediated, posttranscriptional gene regulation process 54 55 through which the synthesis of targeted proteins is downregulated by interfering in the expression of the 56 57 22 58 genetically encoded message. The interference is performed by short noncoding RNAs that hybridize 59 60 5 ACS Paragon Plus Environment ACS Combinatorial Science Page 6 of 81

1 2 3 with complementary mRNA, resulting in translation inhibition or message degradation. The principal 4 5 6 RNAs involved in the RNAi mechanism are endogenous interfering micro RNAs (miRNAs), piwi RNAs 7 8 (piRNA), and exogenous small interfering RNAs (siRNAs). 9 10 23 11 More than 60% of the human genes are miRNAregulated posttranscriptionally, and 12 24 13 dysregulation of miRNAs levels have been associated with cancer. The same miRNA can regulate 14 15 various genes, and a gene can be regulated by several miRNAs. 25 Their biogenesis begins with their own 16 17 18 transcription, or in gene introns. Primary miRNA encoding transcripts (primiRNAs) are processed in 19 20 the nucleus into a single stranded, 6070 nts long stem loop precursor miRNAs (premiRNAs) by 21 22 Drosha, an RNase III nuclease. 26 miRNAs spliced from introns (mirtrons) bypass the Drosha and 23 24 25 Microprocessor complex (associated from Drosha and DGCR8, the nuclear protein that recognizes 26 27 double stranded RNA), and a lariatedebranching enzyme aids in generation of a premiRNA stem 28 29 loop. 27 These short hairpin RNAs (shRNAs) are transported by Exportin5 from nucleus to cytoplasm, 30 31 32 where Dicer, an RNase III type enzyme, cleaves the hairpin loop, leaving 22 nts long mature miRNAs 33 34 ready to enter the RNA interference pathway.28 miRNA is unwound by any of the Argonaut proteins 35 36 37 Ago 14, yet only Ago 2 has high dicing activity. The Argonaut protein Ago2, a component protein of the 38 39 RNAinduced Silencing Complex (RISC), is the protein which recognizes the short double stranded 40 41 RNA, uses one strand to guide the RISC complex to the mRNA, and usually degrades the other strand.29 42 43 44 The incorporated RNA strand helps guide RISC to the complementary sequence, generally on a 3’UTR 45 46 region of the mRNA. miRNA typically pairs imperfectly to the mRNA, with only a seed region 47 48 consisting of nucleotide positions 28 required to be complementary base paired to the mRNA. miRNA 49 50 51 RISC complexes tend to stall or repress message translation, by cleaving the polyA tail, destabilizing the 52 53 mRNA, or sometimes degrading the mRNA message.30 54 55 56 57 58 59 60 6 ACS Paragon Plus Environment Page 7 of 81 ACS Combinatorial Science

1 2 3 piRNAs are another type of endogenously generated interfering RNA, and are longer than 4 5 6 mature miRNAs, ranging from 2631 nts. piRNAs complex with piwi Argonaute proteins to silence the 7 8 expression of certain genetic elements and retrotransposons, and destabilize heterochromatin formation 9 10 31 11 in germline cells. 12 13 siRNAs are processed from cytoplasmic double stranded RNAs, regardless of their exogenous or 14 15 endogenous origin. 32 Cytoplasmic doublestranded RNAs are cleaved by Dicer, which cuts double 16 17 18 stranded RNA into 2123 nucleotides (nts) long siRNAs. These resulting duplexes are phosphorylated at 19 20 their 5’ end and hydroxylated at their 3’ end, with two nt 3’ overhangs. The thermodynamically favored 21 22 end is unwound by endonucleases, and the “guide” strand (or antisense strand) is loaded into RISC and 23 24 33 25 transported to the complementary region of an mRNA, while the passenger strand is degraded. The 26 27 RISC complex cleaves the mRNA in the middle of the guide siRNA recognition site. While only the 28 29 seed region of the miRNA is required to be complementary to its mRNA target, the guide strand of an 30 31 32 siRNA pairs perfectly to the mRNA. Whereas miRNARISC generally reduces the efficiency of 33 34 message translation, guide siRNARISC complexes cleave mRNA site specifically, resulting in a 35 36 34 37 repression of translation. 38 39 Because RNA interference is triggered by the presence of short doublestranded RNA, not only 40 41 do interfering RNAs function as a regulators of endogenous protein expression, but the RISC 42 43 44 mechanism also acts as an innate cellular defense mechanism against pathogenic doublestranded RNAs 45 46 of viral or transposon origin. Due to the ability of RNA interference to temporarily downregulate 47 48 virtually any target gene, rationally designed siRNAs makes RNAi a highly attractive tool for both basic 49 50 51 research as well as clinical applications. Indeed, more than 20 clinical trials utilizing RNAibased drugs 52 53 have been undertaken or are currently ongoing. 35 54 55 56 57 58 59 60 7 ACS Paragon Plus Environment ACS Combinatorial Science Page 8 of 81

1 2 3 Aptamers 4 5 6 Aptamers are relatively short oligonucleotides that bind selectively and robustly to their target. 7 8 These short nucleic acid segments can fold into particular secondary and tertiary structures to bind to 9 10 36 11 specific ligands, small molecules, or proteins. Naturally, they occur in riboswitches, and their 12 3739 13 structure and conformation change upon binding to a target ligand. Aptamers can also be selected in 14 15 vitro to bind small molecules and peptides with high affinity and specificity. 4042 In addition to 16 17 43, 44 18 recognition, aptamers can be used in drug therapies themselves. Their high selectivity and their 19 20 potential to be used as effectors make them an invaluable therapeutic tool. Aptamers that affect the 21 22 corresponding protein’s function have been employed as selective response modulators. Activators, or 23 24 45 25 agonistic aptamers, have been selected for insulin, for Gprotein coupled receptors agonistic 26 27 autoantibody in cardiomyopathy,46 for neurodegenerative disease’s TrkB neurotrophin receptor ,47 or for 28 29 CD40, CD28, CD8+ presenting immunocells.48 Antagonistic aptamers that bind and block their 30 31 32 proteins or receptors can be used for both diagnostic and therapeutic purposes, with several aptamers in 33 34 various stages of clinical trials, including phase III.49, 50 For example, aptamers binding and inhibiting 35 36 37 the human prothrombin protein have been selected. The inhibition of this bloodclotting factor prevents 38 51 39 the formation of thrombin, thus implementing an RNAbased anticoagulant. Other antagonist 40 41 aptamers have been proven effective against arthritis’ Interleukin6 receptor, 52 Eselectin expressing 42 43 53 54 44 vascular cells, metastatic gastric cancer’ receptor periostin, and immune checkpoints proteins on T 45 46 cell receptors TIM3.55 Furthermore, a particular advantage of nucleic acid based aptamer therapies can 47 48 be exploited by using a nucleic acid strand antisense to the designed functional aptamer that can bind 49 50 51 and deactivate the compound. In this way, it is straightforward to design an antidote for the initial 52 53 aptamer/drug. The existence of an antidote makes the use of higher dose therapies feasible, since the 54 55 aptamer/drug can be deactivated as soon as needed 51 56 57 58 59 60 8 ACS Paragon Plus Environment Page 9 of 81 ACS Combinatorial Science

1 2 3 Splicing Modulators 4 5 56, 6 Aberrant RNA splicing is associated with cancer as well as a variety of congenital conditions. 7 8 57 RNA splicing can be targeted using a variety of different approaches such as small molecules, 9 10 57 11 antisense oligonucleotides, modified small nuclear RNAs (snRNAs). Using synthetic antisense 12 13 oligonucleotides, it has been demonstrated that hearing can be restored in a mouse model of human 14 15 hereditary deafness. 58 16 17 18 19 20 21 22 RNA Switches 23 24 25 Some RNAs change their conformation upon binding to other molecules or changes in 26 27 temperature. This conformational switch can then be a signal for a subsequent molecular event. 28 29 Naturally occurring RNA switches, called riboswitches, are segments of mRNA capable of binding a 30 31 59 32 small molecule or ligand and subsequently altering the mRNA’s expression pattern. 33 34 This switching capability can be combined with the RNA functionalities mentioned thus far. For 35 36 37 example, the specific binding of a ligand to an RNA region upstream of a gene (akin to specific 38 39 aptamerligand binding) can, in some cases, modulate transcription or translation. RNA interference can 40 41 also be modulated with a switching capability: designed shRNAbased complexes that change their 42 43 44 conformation upon binding to a biomarker mRNA were reported recently. This capability can be utilized 45 46 to convey therapeutic functionality preferentially in diseased cells. 60 47 48 Ribozymes 49 50 51 A particular class of RNAs capable of catalyzing biological reactions in an enzyme-like manner was 52 53 discovered more than 30 years ago 19, 20 , yet their biological functions are still being uncovered. These RNA 54 55 enzymes (ribozymes 20 ) of unknown evolutionary origin are found in all phyla and are essential to life,61 56 57 supporting the “RNA world“ hypothesis.62, 63 The chemical reactions they catalyze are self-cleavage, nucleic 58 59 60 9 ACS Paragon Plus Environment ACS Combinatorial Science Page 10 of 81

1 2 3 acid ligation (both DNA and RNA) 64 and peptide bond formation. 65 Each ribozyme family has its own 4 5 6 structure, organized by junctions or pseudoknots, and its own distinct geometry of the active site. The most 7 8 common reaction they catalyze is self-scission through the acid-base mechanism 66 , promoted by increased 9 10 pH and mediated generally by divalent cation presence (Mg 2+ , Ca 2+ , Mn 2+ , Co 2+ , Pb 2+ ) or high 11 12 concentrations of monovalent cations (Na +). Some ribozymes bind metabolites or other organic molecules, 13 14 yet the 2’-OH nucleophilic attack on the 3’ phosphate consistently yields two RNAs, a 5’ RNA with a 15 16 67 17 3’monophosphate and a 3’ RNA with a 5’ –OH, with a 2’3’ cyclic monophosphate intermediate. Ribozymes 18 19 can cis -cleave themselves from transcripts, as in the case of self-splicing introns, or trans -cleave cellular 20 21 RNAs, such as tRNA and other ncRNAs, as in the case of RNase P- assisted RNA 68 or S-adenosylmethionine 22 23 (SAM) riboswitches.68 Recent studies uncovered nine naturally occurring self-cleaving motifs, and six 24 25 distinct classes of self-scissing ribozymes, catalyzing the transesterification reactions. The involvement of 26 27 28 ribozymes goes beyond self-cleaving, and bioinformatics and experimental studies suggest ribozymes 29 30 catalyze RNA splicing, transfer RNA biosynthesis, viral replication, as well as other molecular processes. 67 31 32 In vitro selection allows for retention of synthetic ribozymes with particular catalytic proprieties, such as 33 34 self-cleaving 69 , self-replicating 70 and self-cleaving riboswitches (aptozymes) 71 . Development of sequence- 35 36 72-75 37 specific self-scissing ribozyme, biosensing and therapeutic ribozymes is an area of ongoing research. 38 39 40 Exogenous mRNAs 41 42 The upregulation of a gene or the introduction of a novel gene can be accomplished by the 43 44 delivery of synthetic mRNA. In order to increase the stability and efficacy of the delivered mRNA a 45 46 47 variety of molecular properties can be tuned, such as the use of modified nucleotides (to increase 48 49 resistance against nuclease degradation), 5’ cap modifications (to increase eIF4E binding or increase 50 51 resistance against decapping), optimization of codon usage (to increase translation processivity) or poly 52 53 76 54 A tail length modifications (to modulate the stability of the mRNA). 55 56 CRISPR RNAs 57 58 59 60 10 ACS Paragon Plus Environment Page 11 of 81 ACS Combinatorial Science

1 2 3 Another approach that incorporates functional RNAs into a nanoconstruct utilizes CRISPR 4 5 6 (Clustered regularlyinterspaced short palindromic repeats), a system of acquired immunity against 7 8 phages found in bacteria and archaebacteria. 77 Repurposing this system for biomedical applications is 9 10 11 based on the capability of an exogenous Cas9 protein complexed with an RNA guide strand to bind a 12 13 targeted DNA region and induce DNA singlestrand breaks or double strand breaks, as well as DNA 14 15 methylation. The capability of inducing programmable genomic singlestrand or doublestrand breaks 16 17 18 can be utilized to excise or replace genomic regions. The functionality of the Cas protein to cause 19 20 targeted DNA methylation can be used for programmed downregulation of gene expression. 21 22 Furthermore, it has been shown that Cas proteins with deactivated nuclease activity retain their 23 24 25 capability to target a programmed DNA region. Such “dead” Cas proteins (dCas or dCas9) have been 26 27 used to transport molecular cargo to desired DNA regions. For example, the dCas9mediated transport 28 29 of transcription factors can be utilized for programmed gene activation. 78, 79 Designed CRISPR systems 30 31 80 32 utilizing RNA nanostructures such as RNA scaffolds have been reported. 33 34 RNA/DNA hybrids 35 36 37 Hybrid structures consisting of duplexed RNA and DNA strands have interesting properties. 38 39 Because most types of nucleases do not degrade RNA/DNA hybrid duplexes, such structures can have a 40 41 higher in vivo stability compared to RNAonly equivalents. Pairs of cognate RNA/DNA hybrid 42 43 44 structures with toeholds can be delivered separately leading to a reassociation resulting, for example, in 45 46 RNAi activation in cells. 16, 81, 82 47 48 49 50 51 Rational RNA Nanostructure Design 52 53 RNA structures can be thought of as being composed of structural modules, or “RNA motifs”. 54 55 Different motifs can have different functions: some have architectural roles (helices, branched junctions, 56 57 58 59 60 11 ACS Paragon Plus Environment ACS Combinatorial Science Page 12 of 81

1 2 3 kinkturns, etc.), and can be interchanged between different molecular contexts, being used as structural 4 5 8 6 units to engineer novel constructs with new properties. Some RNA motifs can interact with other RNA 7 8 motifs, giving RNA selfassembling properties, or with other molecules, giving RNA molecular 9 10 11 recognition functions. RNA motifs have been compiled and cataloged in various databases. Some 12 13 databases are motifspecific: RNAJunction is a compilation of kissing loops, internal loops, bulges and 14 15 multiway junctions 83 while the kturn database provides a detailed collection and analysis of kink 16 17 84 85 86 87 18 turns. Other RNA tertiary motif databases include SCOR, FRABASE and Motif Atlas. RNA 19 20 STRAND 88 and RNA CoSSMos 89 are databases geared towards secondary structure analysis and 21 22 secondary structure searches. They can be used as tools to identify motifs with specific geometries and 23 24 9 25 functions, and potential candidates for interchanging structural modules in a given structure. All these 26 27 databases can be used as motif pools from which to assemble RNA nanoconstructs, and thus they can 28 29 serve as a starting point for the RNA nanostructure designer. A unique database of insilico assembled 30 31 32 RNA nanostructures, the Ring Catalog, compiles ringlike RNA nanostructures that are computationally 33 34 predicted by combinatorial assembly of helices, multiway junctions, single nucleotide internal loops 35 36 90 37 (bulges), general internal loops, and kissing loops found in the RNAJunction database. 38 39 RNA nanoconstructs can be used as scaffolds for further functionalization, and the 40 41 functionalized constructs are generally called RNA Nanoparticles (NPs). 91 There are several advantages 42 43 44 to using RNA nanoconstructs as scaffolds for nanoparticles. Naturally, macromolecular complexes such 45 46 as ribosomes, ribozymes, RNase P, spliceosomes, etc., are RNAbased machines. Hence, RNA rather 47 48 than DNA is a component of choice for all these “natural nanoparticles”. 92 Artificially, RNA NPs can 49 50 51 be engineered to present precise sizes, shapes and stoichiometries. Their synthesis is simple and 52 53 straightforward, both chemically and enzymatically. RNA scaffolds can be functionalized with a variety 54 55 of functional moieties, depending on the desired applications. A polyvalent RNA NP can incorporate not 56 57 58 59 60 12 ACS Paragon Plus Environment Page 13 of 81 ACS Combinatorial Science

1 2 3 only multiple siRNAs for increased potency, but several functional moieties, such as distinct siRNAs for 4 5 6 a synergistic effect, aptamers, fluorescent tags, peptides, etc., for multipurpose effects, such as direct 7 8 targeting, diagnostics and therapeutics. Thus, due to their chemical, structural and enzymatic properties, 9 10 11 RNA is a biologically active material that can be utilized in nanotechnology and biomedical 12 13 applications. 14 15 Manual design of RNA Nanostructures based on naturally occurring RNA 3D motifs 16 17 18 Naturally occurring RNAs and RNA motifs can be employed towards generating RNA NPs, as 19 20 in the case of pRNA. Symmetric, multimeric structures are frequently associated with proteins and 21 22 rarely encountered in RNA molecules. Yet, the phi29 has a symmetrical structure 23 24 25 assembled by long range tertiary WatsonCrick base pairing between repeated molecular units. This 26 27 packaging RNA (pRNA) assembles thus through looploop interaction in a hexameric ring and 28 29 translocates the DNA in the bacteriophage phi29 (Figure 1.a.i).93, 94 By manipulating the sequence at the 30 31 32 interface between the repeating monomers, various nanoconstructs assembling via “handinhand”, 33 34 “foottofoot” and “armonarm” orientations were produced. 95 Addition of four nucleotides in the 35 36 37 interacting hand loops insured specific complementarity and allowed for nine predicted interfaces. 38 39 Seven of the designed loops mediated dimerization in vitro and were used to design a pRNA dimer, 40 41 trimer, tetramer, pentamer, hexamer, and heptamer (Figure 1.a.ii). Addition of short, palindromic 42 43 44 sequences to the end of the pRNA sequence allows for “foottofoot” dimerization, and combined design 45 46 allows for assembling polymers up to 14 units in length (Figure 1.a.iii).96 The core obtained after 47 48 removal of the hairpin loops (pRNA3WJ),97 and the core engineered by “ligating” two 3WJ in an X 49 50 98 51 shaped core, were utilized to create branched hexameric constructs (Figure 1.a.iv). Among these two 52 53 core structures, 3WJpRNA and XpRNA, armonarm branched hexamer, and foottofoot hexamer are 54 55 the notable designs. All these scaffolds have been used for functional attachments. pRNAs have been 56 57 58 59 60 13 ACS Paragon Plus Environment ACS Combinatorial Science Page 14 of 81

1 2 3 successfully incorporated in RNA NPs and used to deliver therapeutic and diagnostic molecules to 4 5 6 diseased cells like cancer and viralinfected cells, see section ‘Functionalization of RNA 7 8 Nanostructures”. 96, 97, 99 9 10 11 Computational design of RNA Nanostructures using naturally occurring RNA 3D motifs 12 13 14 3-D Motif-based nanostructures 15 16 Another approach to intermolecular assembly of RNA nanostructures involves the use and 17 18 development of computational tools for exploring and modeling feasible 3D conformations, and for 19 20 100, 101 21 designing strand sequences capable of selfassembling into these designed structures. Such 22 23 computational methods speed up the entire design of the RNAbased nanoparticles and go hand in hand 24 25 26 with experimental procedures. Due to the plethora of solved RNA tertiary structures available, detailed 27 28 bioinformatics analysis has resulted in the identification of recurrent RNA structural motifs. With 29 30 knowledge of RNA structure and folding characteristics, these structural RNA motifs can be 31 32 33 implemented as modular building blocks for designing RNA based nanoconstructs. 34 35 The process can start with a collection of nway junctions and kissingloops from the 36 37 RNAJunction database and assembled by the program NanoTiler.83 Modeling programs, such as 38 39 101 40 NanoTiler and RNA2D3D can use such building blocks as “corner stones” in the 3D models. Initially 41 42 the selected building blocks are handled as rigid objects and are roughly fit together. However, as is the 43 44 case in natural RNAs where larger structural contexts may exert local distortions, deformations can be 45 46 47 applied to the elements of the models in order to achieve full structure backbone connectivity. NanoTiler 48 49 applies them to idealized helices connecting the junctions, and gives the user a measure of the 50 51 82 52 deformation as feedback. A collection of nanoringlike structures can be found in the Ring Catalog. 53 54 RNA2D3D facilitates interactive distortions and assists in the relaxation process of the selected 55 56 fragments via short energy minimizations and/or MD runs, leaving the expert decision making to the 57 58 59 60 14 ACS Paragon Plus Environment Page 15 of 81 ACS Combinatorial Science

1 2 3 user. Alternative conformations of comprising motifs based on experimental data (Xray 4 5 6 crystallography, NMR, CryoEM), available in RNAJunction or in any other source for the same basic 7 8 motif type, can also be used to explore the extent of structure variability and potentially be used “as is” 9 10 11 in the modeling process. 12 13 RNA nanorings were designed to assemble as hexagonshaped supramolecular structures through 14 15 formation of kissing complexes between six “dumbbell” shaped monomer units, i.e. helices capped by 16 17 11 18 kissing loops at each end (Figure 1.b.i). In this particular design, the kissing loops are sequence 19 20 variants of the ColE1 kissing complex, a unique assembly motif that exhibits a 120degree angle 21 22 between adjacent helices.102 The use of six distinct kissing complexes to assemble the nanoring scaffold 23 24 25 provides the means to have complete control over the functional composition of each heterogeneous 26 27 monomer unit.91 The 5’ and 3’ ends of each monomer strand are located at the midpoint of the helix. 28 29 While other supramolecular RNA architectures have been generated that make use of kissing loops for 30 31 93, 103105 32 assembly, the nanoring is distinct in that the particle’s geometry is defined by the intermolecular 33 34 assembly interaction, rather than intricate structure motifs embedded within the core of each monomer. 35 36 37 A consequence of this monomer design is robust intramolecular folding and intermolecular assembly. 38 39 Fully assembled nanorings can be generated from isothermal, singlepot cotranscription reactions with 40 41 multiple DNA templates that are required to encode the RNA strands for nanoring formation.12 Co 42 43 44 transcriptional assembly was proven for the nanocube constructs as well, see section “Denovo design of 45 46 RNA Scaffolds”. 10 The capability to cotranscriptionally generate functional nanoring and nanocube 47 48 assemblies eliminates many of the typical steps associated with RNA nanoparticle synthesis, purification 49 50 51 and assembly, greatly reducing the time required to prepare particles for delivery. This co 52 53 transcriptional assembly can also be made compatible with use of 2’modified NTPs. Incorporation of 54 55 56 57 58 59 60 15 ACS Paragon Plus Environment ACS Combinatorial Science Page 16 of 81

1 2 3 2’FdUTP during cotranscriptional nanoring (or nanocube) assembly greatly increases its resistance to 4 5 12 6 nuclease degradation in blood serum, a desirable trait for applications associated with in vivo delivery. 7 8 De-novo Design of RNA Scaffolds 9 10 11 For some RNA scaffolds of particular shapes and properties, no naturally occurring motifs may 12 13 be able to be identified. As in the case of the in silico designed RNA cube, the RNAjunction database 14 15 does not contain any 3way junction that corresponds approximately to a cube corner.10, 83 In this case, 16 17 18 several 3D design strategies present themselves: one approach is to design a 3D structure with the proper 19 20 3D motifs (in this case a 3way junction with 90° interhelix angles) de novo (Figure 1.c.i). 21 22 Alternatively, instead of focusing on the 3D motifs, one may optimize the position of the involved 23 24 25 helices and then place bridging RNA strands de novo. Using the latter method, an RNA nanocube was 26 27 designed in this manner and assembled experimentally. 106 28 29 Atomiclevel molecular dynamics simulations may aid in improving the 3D structural model of 30 31 32 the nucleic acid nanostructure and may aid in predicting some of its properties. Alternative 33 34 conformations of the desired building blocks can also be obtained by subjecting only the building blocks 35 36 107 37 to molecular dynamics simulations. This approach can be used when, because of its size, detailed 38 108 39 simulations are not feasible for an entire nanostructure model. Computationally less costly coarse 40 41 grained methods, such as those based on the elastic network models, 109112 can also provide valuable 42 43 44 information about the most biologically relevant low frequency motions (normal modes) of the building 45 46 blocks, as has been shown for nucleic acids and nucleic acids/protein complexes. 11, 113117 Through these 47 48 methods it is feasible to evaluate the entire nanostructure that atomic resolution methods may still be 49 50 51 unable to handle. 52 53 A coarsegrained Anisotropic Network Model (ANM) was applied to the characterization of the 54 55 RNA cubic nanostructures. 106 The cubes have helical edges and singlestranded corner linkers, and the 56 57 58 59 60 16 ACS Paragon Plus Environment Page 17 of 81 ACS Combinatorial Science

1 2 3 models were created for several cube variants, differing from each other by the length of the single 4 5 6 stranded bridges between the helical edges (03 nts). The resulting models predicted different 7 8 efficiencies of assembly for the considered variants due to steric constraints in the cube corners. These 9 10 11 predictions were confirmed experimentally in vitro . However, the sizes of the nanocubes measured in 12 13 solvent appeared to be larger than the initial models would suggest. ANM simulations were used to 14 15 demonstrate that the dynamics or distortions of the cubes increase their radii of gyration. This was 16 17 18 needed to be considered in order to reconcile the initial differences between the modeling results and the 19 20 experimental data. In addition, the ANM simulation results deepened our understanding of the reasons 21 22 for the differences in the efficiency of assembly and in the melting temperatures of the cube variants. 23 24 25 Different assembly strategies were tested as well: nanostructures assembling from 6 and 10 RNA 26 27 molecules were tested.106, 118 28 29 30 Functionalization of RNA Nanostructures 31 32 33 Regions of complementary base pairing in RNAbased nanostructures can be used to easily 34 35 append functional RNA motifs to the scaffold core, without the need for separate covalent linkages and 36 37 complex chemistries. 91 RNA nanostructures are versatile scaffolds that can be repurposed to harbor 38 39 40 various functional RNA elements. Individual monomer molecules of nanoconstructs can be 41 42 functionalized with additional RNA moieties by extension of either the 5’ or 3’ ends to encode 43 44 functional elements for specific applications. 12, 91, 119 120 In fact, design strategies can be devised so the 45 46 47 functional elements, such as siRNA, are encoded within the sequence of a scaffolding unit, if so desired. 48 49 RNA nanorings present a robust and versatile method for the delivery of siRNA and/or other 50 51 52 functional RNA entities to cells. The RNA nanoring was originally envisioned as a delivery vehicle for 53 121 54 Dicer substrate RNA duplexes (DSRNAs), and indeed DSRNA functionalized nanorings are 55 56 processed by Dicer to liberate siRNA duplexes from the ring scaffold (Figure 1.b.ii).91 More recent 57 58 59 60 17 ACS Paragon Plus Environment ACS Combinatorial Science Page 18 of 81

1 2 3 work has shown the nanoring can be effectively delivered to cells in both tissue culture and xenograft 4 5 14 6 tumor models. 7 8 Nanorings functionalized with 6 different Dicersubstrate RNAs (that are processed by Dicer to 9 10 11 siRNA) against 6 different targets of HIV1 show significant reduction in viral protein expression in 12 14 13 transfected cells. Incorporation of singlestranded toeholds extending from each monomer unit further 14 15 increases the ease with which functional entities can be attached for given applications by simply 16 17 18 incubating the scaffold with new functional elements that harbor complementary toeholds to specific 19 20 monomers of the ring. 21 22 Building on the ease with which siRNA could be conjugated to an RNA scaffold, additional 23 24 25 functional RNA moieties including, miRNA, aptamers, ribozymes, fluorescent tags, proteins, and other 26 27 elements can be added to increase the functional diversity of the resulting RNA nanoparticle. Nanorings 28 29 functionalized with the J18 aptamer against human epidermal growth factor receptor (EGFR) bind to 30 31 32 cells which express high levels of EGFR on their surface, while dye labeled nanorings can be used to 33 34 locate the scaffolds associated with the cells (Figure 1.b.iii). 14 35 36 37 The same functionalization strategies outlined for the nanoring scaffold can be applied to the 38 39 nanocube scaffold, as well as other related RNA nanoconstructs that have been extensively tested and 40 41 characterized (Figure 1.c.ii).10,106 The key difference is the assembly process relying on interstrand 42 43 44 interactions of multiple RNA chains. 45 46 Hybrid technology in RNA NPs 47 48 In a DS RNA duplex, one RNA strand can be substituted by a corresponding complementary 49 50 51 DNA strand, to confer nuclease resistance to the construct, resulting in an RNA/DNA hybrid. Another 52 53 complementary construct, a DNA/RNA hybrid, is required to assemble with the first, in order to confer 54 55 the functionality to the recombined construct (Figure 2.a.i). The extension of the hybrid strands, either 56 57 58 59 60 18 ACS Paragon Plus Environment Page 19 of 81 ACS Combinatorial Science

1 2 3 RNA or DNA, with single stranded nnucleotide toeholds, offers anchorage for initiation of hybrid 4 5 6 strand exchange (Figure 2.a.ii). The reassociation resulting in the original DS RNA duplex, with a 7 8 double stranded DNA side product (Figure 2.a.iii), can be initiated through recognition of 9 10 11 complementary toeholds, composed of either RNA or DNA, due to thermodynamic differences between 12 13 the RNA and the DNA duplexes. Such constructs have the unique property of being conditionally 14 15 activated only when both complementary hybrids are present. Complementary hybrid constructs can be 16 17 81 18 designed for DS RNAs, and as well as other constructs incorporating multiple functionalities. 19 20 Extending the hybrid concept to RNA NPs, the functionalized arms of the nanoparticles can be 21 22 split into hybrids with either DNA 14 or RNA toeholds.81 Recent advances show that the functional 23 24 25 elements associated with the ring scaffold can be designed for conditional activation in response to a 26 27 supplemental hybrid construct. For instance, nanorings have been functionalized with RNA/DNA 28 29 hybrid arms which are able to produce Dicersubstrate RNA duplexes upon interaction with a separate 30 31 14, 122 32 hybrid entity (Figure 2.b.i). This conditional activation can be extended to other functional entities 33 34 such as FRET pairs and aptamers,16 producing even greater control over the degree of functionalization 35 36 123 37 of the constructs. Cubes with hybrid RNA/DNA arms reassociated with their cognate DNA/RNA 38 39 hybrids to produce conditionally activated functional nanoparticles with DS RNAs, capable of silencing 40 41 GFP (Figure 2.b.ii). 118 42 43 44 One advantage of this novel technique is theoretically many functional RNAs (silencing, 45 46 activators, aptamers, etc.) can be split with the use of complementary into the nonfunctional 47 48 RNA/DNA hybrids, and the initial RNA function can be restored only through the hybrid reassociation. 49 50 51 Besides therapeutic functionalizations, additional incorporation of functional elements into the RNA 52 53 nanoparticles allows for multiple simultaneous applications, such as molecular imaging and 54 55 biosensing.124 In some cases, RNA can be substituted by DNA to provide a DNA scaffold for the 56 57 58 59 60 19 ACS Paragon Plus Environment ACS Combinatorial Science Page 20 of 81

1 2 3 conditionally activated hybrid RNA/DNA arms. In the nanocube’s case (Figure 2.b.iii) , c omparative 4 5 6 characterization studies indicated significantly lower interferon and proinflammatory cytokine 7 8 responses for a DNAbased scaffold. These are important characteristics for the intended therapeutic 9 10 118 11 use. 12 13 14 15 16 17 18 19

20 21 22 Challenges in delivering RNAs 23 24 There are several issues that need to be addressed when delivering drugs and/or nanoparticles to 25 26 cancer and diseased cells. The therapeutic agents need to get in the vicinity of the targeted cells, and 27 28 29 they have to enter these cells to be fully effective. The Enhanced Permeability and Retention (EPR) 30 31 effect in many cases increases drugs or particle accumulation in the vicinity of a tumor environment, 32 33 normally due to the leaky vasculature surrounding the tumor. 125 This can usually be accomplished 34 35 36 without targeting agents. However, it is felt that the addition of appropriate targeting moieties can 37 38 facilitate the delivery of the therapeutic agents.126 Such targeting moieties can be designed to 39 40 127 128,44 41 specifically target tissues, whole cells, or particular proteins or membrane receptors. Various 42 43 kinetic and thermodynamic hindrances can also impede the RNA NPs cellular delivery to their target. 44 45 These challenges exist at various levels of the delivery process, and include not only cellular uptake, but 46 47 48 extracellular barriers, i.e. RNA’s stability and degradation in blood serum, its organ uptake and 49 50 biodistribution, and the hepatorenal clearance, as well as immunogenic response, and intracellular 51 52 barriers, including endosomal escape and offtarget effects. These challenges are discussed more in 53 54 55 detail below. 56 57 58 59 60 20 ACS Paragon Plus Environment Page 21 of 81 ACS Combinatorial Science

1 2 3 Degradation 4 5 6 RNA’s halflife at physiological conditions is fairly short, ranging from a few minutes to an 7 8 hour, leaving little time for the systemically administered RNA to reach an intracellular target. 129 9 10 11 Several chemical modifications have been explored in an attempt to increase the resistance of siRNAs to 12 13 nuclease digestion. The chemical versatility of RNA allows for easy modifications of the ribose 2’ 14 15 position, as well as the 3’ position at the oligonucleotide terminus.129, 130 At the strand’s 3’ end, the 16 17 131 18 phosphate group can be replaced with phosphothioate or boranophosphate. Although the latter 19 20 modification prolongs siRNA’s halflife, it might result in decreased interference activity and toxicity 21 22 caused by metabolites. As the 2’OH within a siRNA is not required for silencing, 2’ ribose 23 24 25 modifications have become a common method to increase siRNA stability without sacrificing potency. 26 27 Several 2’ modifications can be used to protect RNA from nuclease degradation, such as 2’ fluorination, 28 29 2’ oxymethilation, and 2’amination of pyrimidines. 12, 132134 In some cases, the substitutions can be made 30 31 135 32 without negatively affecting siRNA efficiency. In fact, in some instances, 2’ modified siRNAs have 33 34 been observed to display enhanced potencies, such as in vitro increased target silencing, reduced off 35 36 136 37 target effects, and decreased innate immune response, as compared to unmodified siRNAs . Locked 38 39 nucleic acids (LNAs) with methyl linkages between the ribose’s 2’ and 4’ positions, are another 40 41 methodology used to increase RNA nuclease resistance. 136 This modification does not affect its 42 43 44 compatibility with the RNAi machinery, preserves or increases hybridization affinity with mRNA and 45 46 can decrease offtarget effects, sometimes with improved efficiency of the siLNA. 136, 137 47 48 Previously, the synthesis of RNA molecules containing 2’ modified nucleotides required the use 49 50 51 of chemical synthesis or in vitro transcription using a mutant T7 polymerase to achieve significant RNA 52 53 yields. However, recent work has revealed that the addition of Mn 2+ during in vitro transcription allows 54 55 56 57 58 59 60 21 ACS Paragon Plus Environment ACS Combinatorial Science Page 22 of 81

1 2 3 wildtype T7 RNA polymerase to incorporate 2’fluoro dNTPs and produces transcription yields 4 5 12 6 comparable to a standard transcription reaction using unmodified NTPs. 7 8 Stability in blood 9 10 11 Interactions with proteins in the blood stream are a concern, as retention and stability in blood 12 13 are necessary to achieve cellular uptake following systemic delivery. The ribosephosphate RNA 14 15 backbone of RNA is susceptible to enzymatic hydrolysis. Nanoparticle surface modifications with 16 17 18 hydrophilic polymers can increase the circulation time and delay nanoparticle clearance, allowing the 19 20 nanoparticles to reach their targeted tissue. 138, 139 However, characterization of nanoparticle interactions 21 22 with protein components of the blood is essential, as nanocarriers have unique properties and 23 24 25 mechanisms of interaction. Interaction between nanoparticles and coagulation factors could result in 26 27 deficient, prolonged, and/or unwanted coagulation. Formation of larger aggregates in the bloodstream 28 29 can lead to thrombosis and presents a major concern when considering nanoparticle injection. Even in 30 31 32 cases in which the same basic material and overarching methodology are used to create nanoparticles 33 34 that may differ only slightly from one another (e.g. differing liposome, dendrimers, quantum dots, etc.), 35 36 37 these nanoparticles may result in very different interactions with various components found in the 38 140 39 blood. 40 41 Hepato-Renal Clearance 42 43 44 Clearance via kidneys impacts very small particles (below approximately 8 nm or about 40 kDa), 45 46 while very large nanoparticles (greater than 200 nm) can be cleared by the spleen or get trapped in the 47 48 lungs. 35, 141 “Naked” siRNAs, for example, are subject to renal clearance minutes after systemic 49 50 142 51 administration. siRNAs with chemicallymodified nucleotides or conjugated with cholesterol 52 53 withstand the renal clearance up to half hour. Furthermore, siLNAs (Locked Nucleic Acids) and siRNAs 54 55 complexated with lipidbased formulations, such as Chitosan, liposomes, or JetPEI, impede the renal 56 57 58 59 60 22 ACS Paragon Plus Environment Page 23 of 81 ACS Combinatorial Science

1 2 3 clearance. Biodistribution studies show accumulation of these formulations in various organs after 24h. 4 5 6 Conversely, the potential problem of rapid clearance of small molecules presents an opportunity for 7 8 nanotechnology: increasing the particle size above 40 kDa may increase the halflife for renal 9 10 35 11 clearance. 12 13 Toxicity and Immunogenicity 14 15 Exogenous RNA delivered to cells can activate an immune response through the recognition of 16 17 18 the delivered RNA by Pattern Recognition Receptors (PRRs) via Tolllikereceptor (TLR)dependent 19 20 pathways and TLRindependent pathways. 143 Tolllike receptors are membrane receptors that are 21 22 activated via different structural properties of RNAs. Tolllike receptors TLR7 and TLR8 are bound to 23 24 25 membranes of intracellular vesicles and recognize singlestranded RNA. TLR3 is activated by binding 26 27 doublestranded RNA. 144 Recognition of doublestranded RNA by TLR3 seems to be due to at least two 28 29 different binding conformations (a stable conformation attainable by RNA duplexes with at least 46 base 30 31 145 32 pairs and a less stable conformation for RNA duplexes with lengths between 21 and 30 base pairs). 33 34 In contrast, some Pattern Recognition Receptors are not membranebound but cytoplasmic, and 35 36 37 are activators of TLRindependent pathways. Examples are Protein Kinase R (PKR) as well as RIGI 38 39 (retinoic acidinducible gene 1). It was shown that RNA duplexes with lengths of at least 33 base pairs 40 41 lead to PKR activation and that the response is maximal for duplexes that are 80 base pairs in length.146 42 43 44 Another pathway that is activated by doublestranded RNA is the 2’, 5’AS/RNase L pathway. Double 45 46 stranded RNA of at least 70 base pairs activates 2’, 5’oligoadenylate synthetase (2’, 5’AS) that then 47 48 leads to the activation of RNase L. 147 RNase L is a cytoplasmic endoribonuclease, whose activation can 49 50 51 trigger the degradation of cellular and viral RNA (thus leading to the reduction of protein synthesis). 52 53 Lastly, the protein RIGI mediates an immune response by recognizing uncapped 5’phosphorylated 54 55 RNAs – another molecular pattern that is indicative of RNA originating from an invading pathogen. 148 56 57 58 59 60 23 ACS Paragon Plus Environment ACS Combinatorial Science Page 24 of 81

1 2 3 Activation of these pattern recognition receptors can lead to an activation of an immune response 4 5 6 in a variety of ways. For example, activation of interferons can lead to the expression of interferon 7 8 induced proteins (the mentioned pattern recognition receptors like PKR and RNase L are themselves 9 10 147 11 interferoninducible). Induced interferons can be secreted by a cell and trigger inflammatory 12 13 responses in neighboring cells. Another response is the activation of eIF2α (a translation initiation 14 15 factor) that in turn leads to inhibition of translation. 147 Such responses of the innate immune system can 16 17 18 have a cumulative effect of increasing the prevalence of apoptosis. The working of the innate immune 19 20 system was summarized by the following statement: “Similar to virologists, the innate immune system 21 22 may therefore have learned to classify by their genomes.” 148 23 24 25 Importantly, utilizing modified nucleotides can reduce the activation of innate immune 26 27 response. 149 For example, exchanging the 2′hydroxyl of uridines with 2′fluoro, 2′deoxy, or 2′O 28 29 methyl dramatically reduces the recognition of exogenous RNA by Tolllike receptors. 150 30 31 32 It should, however, also be remembered that the majority of delivered material in nucleicacid 33 34 based therapeutics can be attributed to the delivery agent or to the formulation as opposed to the active 35 36 37 molecules. In other words, the toxicity and immunogenicity of the combined system depends a 38 39 substantial amount on the properties of the delivery agent. Because there is such a wide variety of 40 41 delivery agents with vastly different properties, reviewing corresponding cellular responses is beyond 42 43 44 the scope of this review. The reader is encouraged to consult the literature for ascertaining the properties 45 46 of the considered delivery agents. 47 48 Cellular Uptake 49 50 51 Cells have developed distinct pathways for cellular uptake of materials including endocytosis, 52 53 pinocytosis and macropinocytosis. These processes can be dependent on active energy and cytoskeletal 54 55 elements. Cellular uptake can be viewed as molecules mastering the challenge of passing a formidable 56 57 58 59 60 24 ACS Paragon Plus Environment Page 25 of 81 ACS Combinatorial Science

1 2 3 barrier in the form of the lipid bilayer cellular membrane. Cellular uptake is mediated by a variety of 4 5 6 different mechanisms: single molecules can diffuse through the cellular membrane, provided that their 7 8 physicochemical properties are within certain ranges. The “Lipinski Rule of 5” summarizes these 9 10 11 constraints by stating that molecules have “druglike” properties if they have limited watersolubility, 12 13 are “small” (molecular weight less than 500 Da) and contain less than 5 hydrogen bond donor sites and 14 15 less than 10 hydrogen bond acceptors. 151 Lipinski himself noticed exceptions to these observations and 16 17 18 attributed this to molecular transport mechanisms that go beyond diffusion across the membrane. 19 20 21 Efficient cellular uptake is also possible for larger complexes and nanoparticles: In a recent 22 23 report, several classes of antisense oligonucleotides (ASOs) were analyzed for uptake mechanism and 24 25 152 26 uptake efficiency. It was found, that scavenger receptors (SCARAs) contributed to receptormediated 27 28 cellular uptake of the ASOs. Moreover, their incorporation into nanoparticles was correlated with 29 30 improved cellular uptake, resulting in a “second window” of efficient uptake for particle sizes ranging 31 32 33 from 22 to 60 nanometers. Scavenger receptors are involved in the cellular uptake not only of ASOs, but 34 35 of a wide range of molecules such as cell penetrating peptides (CPPs).153, 154 Combined with the fact that 36 37 some CPPs selfassemble into nanoparticles, 155 one can view this as emerging evidence for a general 38 39 152 40 preference for receptormediated cellular uptake for selfassembling nanoparticles. 41 42 Due to their high molecular weight, size and charge, RNAs cannot readily diffuse 43 44 through cellular membranes. Diffusion of charged molecules, such as RNA which is negatively charged, 45 46 47 is thermodynamically unfavorable. Depending on whether RNAs are delivered “naked” or assisted by 48 49 carriers, and on the resulting size of RNA/carrier complexes, RNAs will enter the cell through different 50 51 52 pathways. Particles of 200 nm or less seem to be internalized by clathrinmediated endocytosis, with 53 54 more than half passing through the membrane in the first half of hour. Particles bigger than 200 nm 55 56 57 58 59 60 25 ACS Paragon Plus Environment ACS Combinatorial Science Page 26 of 81

1 2 3 internalize seemingly by caveolaemediated endocytosis, and they take hours to accumulate. No uptake 4 5 156 6 has been detected for particles 1000 nm in size. 7 8 9 Endosomal Escape 10 11 Since endocytosis is the main entry route for delivered RNAs, endosomal escape is necessary 12 13 14 crucial for their functioning. One can distinguish “early” and “late” stages of endosomes. These stages 15 16 are characterized by increasing acidity of the endosomal compartment. 35 The most common endosomal 17 18 escape mechanisms are membrane destabilization 157 or rupture due to the “protonsponge effect”.158 19 20 21 New delivery systems exploit the change in the environment’s pH when the nucleic aciddelivery agent 22 23 complex enters the endosome. pHdependent polymers, dendrimers, and peptides, fusogenic lipids, or 24 25 159 26 chemical agents have been used to aid in the particle’s release from the endosome. The efficiency of 27 28 therapeutic interfering RNAs can be evaluated through biodistribution, cellular and subcellular 29 30 localization studies, endosomal escape efficiency and RISCloading efficacy studies. 160 31 32 33 Off-target Effects 34 35 The efficacy of the siRNAs silencing can vary as a function of the targeted mRNA sequence. Of 36 37 concern are offtarget effects, where siRNA binds in a miRNAlike manner to the message, with only 38 39 40 partial complementarity to mRNA, thus repressing the message of nontargeted genes and possibly 41 42 leading to undesired effects. Sequence searches ensuring the uniqueness of the siRNA target site can 43 44 alleviate offtarget effects to some extent. Offtarget effects can occur in an analogous manner for other 45 46 47 types of RNA therapeutics that rely on sequencecomplementarity to the transcript or genomic regions. 48 49 50 51 52 Delivery methods for RNA therapeutics and RNA NPs 53 54 55 56 57 58 59 60 26 ACS Paragon Plus Environment Page 27 of 81 ACS Combinatorial Science

1 2 3 In the remainder of this review we present a variety of strategies for cellular delivery of RNA 4 5 6 nanostructures. It should be understood, that there may be a considerable amount of variability and 7 8 uncertainty in the precise structural composition of the shown assemblies. 9 10 11 “Naked” Delivery 12 13 Nucleic acid delivery methods can be roughly classified into physical or chemical methods. Non 14 15 modified naked RNAs can be delivered directly to the target site through a variety of physical means, 16 17 18 frequently aided by high concentration or a mechanical, magnetic or electric force. Nuclear or 19 20 cytoplasmic microinjections (mechanical force) 161, 162 , electroporation,163 electrical mediation,164 cell 21 22 squeezing (physical force) ,165 sonoporation (ultrasound) ,166, 167 and optical (lasers) ,168170 23 24 25 deliver nucleic acids directly to the targeted site with limited inhibiting effects due to poor uptake and 26 27 degradation. 171 Nonetheless, there is currently a variety of clinical trials of siRNAbased therapeutics 28 29 that are based on naked delivery.35 30 31 32 In certain cases, “hybrid” methods employing a combination of physical and chemical means are 33 34 used to deliver nucleic acids to cells. For example, nucleic acids, such as siRNA, can be chemically 35 36 37 conjugated with nanoparticles, and the nanoparticles delivered to cells through physical means. The 38 39 nanoparticles also present specific physical and chemical properties that assist nucleic acids in entering 40 41 cells. Such hybrid delivery methods include the gene gun (Inert solid (gold) NP +NA),172 42 43 173 174 44 magnetofection (Magnetic NP + NA), impalefection (Nanostructure NP + NA), and bombardment 45 46 (microprojectiles + NA).175, 176 Hybrid methods also include nucleofection, where electroporation is used 47 48 in conjunction with cell specific reagents that aid in membrane disruption.177 Physical methods offer the 49 50 51 advantage of direct therapeutic delivery to the targeted site or tissue, increased availability due to 52 53 proximity and avoidance of systemically induced side effects, such as immune system stimulation. 178 54 55 56 57 58 59 60 27 ACS Paragon Plus Environment ACS Combinatorial Science Page 28 of 81

1 2 3 The biggest drawback is that not all types of cancers and diseases are topical or localized, and thus 4 5 6 amenable to physical siRNA delivery methods. 7 8 Another form of delivery is the escape of the RNA cargo from a repository placed in proximity 9 10 11 to the target site. In a recent study a siRNAreleasing polymer pellet (called LODER) was surgically 12 13 implanted in close proximity to an otherwise difficult to treat pancreatic cancer. The synthetic siRNAs 14 15 were programmed to target the mRNA of the oncogenic KRAS gene. The authors demonstrate favorable 16 17 179 18 delivery properties of their approach and a clinical benefit of their RNAibased cancer therapy. The 19 20 biggest disadvantage of the physical delivery methods is nonspecific targeting, limiting their use to 21 22 topical or local tissues.180182 23 24 25 A more general approach to evade nuclease degradation is incorporation of modified nucleotides. 26 27 pRNA NPs with fluorinated Us and Cs (2′ fluorinedeoxyCTP and 2′ fluorinedeoxyUTP) have a 28 29 much longer halflife and are more resistant to nucleases. 183 Such pRNA NPs, incorporating modified 30 31 32 pyrimidines into the RNA strands, were functionalized with siRNAs. The tetramer pRNA construct 33 34 augmented with distinct siRNAs targeted successfully firefly luciferase. 184 A 3WJbased pRNA NP 35 36 37 functionalized with Folic Acid for epithelial cancer targeting and decorated with fluorescent dies was 38 185 39 used for imaging in vitro . Systemically delivered Xcore pRNA incorporating folic acid targeting 40 41 moieties and Alexa 647 displayed preferential accumulation in targeted tissues in vivo . Other pRNA NPs 42 43 44 incorporating aptamers, such as Malachite Green, STVbinding, Prostate cancer aptamer, Endothelial 45 46 Growth Factor aptamer, or other functional elements such as fluorescent image markers, the HBV 47 48 ribozyme, and various siRNAs were studied and characterized in vitro and in vivo .97, 98, 186190 49 50 51 Mediated Delivery 52 53 Mediated delivery refers to cases where RNAs cross biological barriers while forming non 54 55 covalent complexes with other molecules that act as delivery agents. 56 57 58 59 60 28 ACS Paragon Plus Environment Page 29 of 81 ACS Combinatorial Science

1 2 3 Polymers 4 5 6 RNAs can form complexes with various cationic polymers through electrostatic interactions 7 8 potentially leading to efficient delivery in vitro and in vivo . Potential advantages of current polymer 9 10 11 based carriers include tunable chemical properties, chemical stability and biocompatibility. The 12 13 properties of RNApolymer complexes depend on the type of chosen polymer. For example, delivery 14 15 agents complexated with the cancer drug paclitaxel (Taxol) can decrease or increase its toxicity. 179 16 17 18 Polysaccharide polymers, such as chitosan, have received attention as nanocarriers because of their high 19 20 biocompatibility and low toxicity. 191 Several current synthetic polymers are used for delivery of nucleic 21 22 acids, include polylactic acid, N(2hydroxypropyl)methacrylamide copolymer, and dendrimers. 23 24 25 Delivery of nucleic acids in vivo has been achieved as well with a commercially available linear 26 27 polymer, “ in vivo JetPEI TM ”.192, 193 28 29 Hydrogels 30 31 32 Nanohydrogels are yet another promising type of nanocarrier for siRNA delivery, synthesized by 33 34 polymerization of pentafluorophenyl methacrylate and tri(ethyleneglycol)methylether methacrylate.194 35 36 37 They are polymerbased cationic carriers which can complexate with and deliver siRNA in vivo . Both 38 39 the hydrogels and the siRNA delivered separately aggregate with the proteins in blood, but the hydrogel 40 41 siRNA complex does not aggregate. Complexing siRNA with nanohydrogel particles shows strong 42 43 195 44 stability while minimizing aggregation, providing an effective method for in vivo siRNA delivery. 45 46 Peptides 47 48 An alternative crossmembrane delivery mechanism can be achieved with cell penetrating 49 50 196 51 peptides (CPPs) that are capable of crossing the cellular membrane directly into the cytoplasm. The 52 53 mechanism of entry may depend on the type and composition of peptide and is a subject of ongoing 54 55 56 57 58 59 60 29 ACS Paragon Plus Environment ACS Combinatorial Science Page 30 of 81

1 2 3 research. The possible entry mechanisms include endocytosis, poreformation or receptor mediated 4 5 6 uptake. 7 8 9 10 11 12 13 Dendrimers 14 15 Cellular delivery of functional RNAs has been accomplished using small molecule dendrimers 16 17 197 18 (i.e. molecules that selfassemble in a treelike fashion). For example, positively charged ammonium 19 20 terminated carbosilane dendrimers have been used for the cellular delivery of siRNAs that target HIV 21 22 genes. 198 Poly(amidoamine) (PAMAM) and polypropylenimine (PPI) dendrimers are frequently utilized 23 24 25 for drug delivery. The delivered RNAs form a complex with the dendrimer (called dendriplex) based on 26 27 electrostatic interactions. Advantages of dendrimerbased delivery systems are their favorable protection 28 29 of the RNA cargo from degradation, their ability to deliver a wide variety of compounds and their 30 31 32 biocompatibility. Potential disadvantages are nonspecific cytotoxicity and liver accumulation due to the 33 34 high charge density as well as rapid clearance and limited control over drug release. 197 35 36 37 Conjugated Delivery 38 39 RNAs can be covalently conjugated to various carrier platforms to aid their intracellular delivery. 40 41 Possible RNAcarrier conjugate systems range from the relatively small and “simple”, such as aptamers 42 43 44 or small drug molecules, to lipids, peptides and proteins, polymers, and composite nanoparticles. 45 46 Aptamers 47 48 Nucleic acids aptamers can be evolved through SELEX and SELEXadjacent methods against 49 50 51 particular cell types based on the differential expression of certain membrane proteins in the normal 52 53 versus pathogenic cells. Consequently, aptamers binding membrane receptors have been selected against 54 55 whole cells. Thus, aptamers can be used to facilitate targeted delivery of therapeutic molecules to a 56 57 58 59 60 30 ACS Paragon Plus Environment Page 31 of 81 ACS Combinatorial Science

1 2 3 specific cell type. Note that aptamers were mentioned earlier in the section on “Functional RNAs”. This 4 5 6 quality of aptamers reflects their dual role: aptamers can be used as therapeutic agents or as delivery 7 8 agents. In the first case it is sufficient for aptamers to bind and block or activate a protein; in the later it 9 10 11 is necessary for the aptamers to be internalized inside the cell along with their cargo. Thus, aptamers 12 13 can facilitate targeted delivery, as well as have the role of an RNA nanoparticle cargo that carries out a 14 15 molecular function related to, for example, protein inhibition or imaging.199 16 17 18 There are several well established aptamers targeting oncogenic or pathogenic cells, such as 19 20 lymphoma or thrombosis, to name a few. Thus, aptamers can be used for cell specific targeting, such as 21 22 tumor cell recognition. 200 Wholecell aptamers have been selected against bacteria, parasites, viruses, 23 24 201203 199 25 and cells, and some were proven to internalize into the cell. Moreover, aptamers have been 26 27 shown to have low, if any, toxicity or immunogenicity. 200, 204 Aptamers have been used both as direct 28 29 carriers for drugs or interfering RNAs, or incorporated in nanoparticles containing therapeutic 30 31 32 molecules. Aptamers conjugated with drugs were used to target various types of cancers, such as breast, 33 34 colorectal, prostate, pancreatic, liver, and lymphoma, and certain inflammatory diseases, as for example 35 36 37 Crohn’s, rheumatoid arthritis, psoriasis, and systemic sclerosis, agerelated macular degeneration, and 38 205 39 HIVtargeting therapies. Aptamersinterfering RNA Chimeras were designed to deliver si, mi, 40 41 antimiR, sh RNAs for tissue specific 206, 207 or HIV 208 targeting and therapeutics. Aptamers have also 42 43 44 been coupled with inorganic nanomaterials, such as gold nanoparticles, through modified chemistry on 45 46 the nanoparticle surface. 209 A DNA aptamer, Sgc8, selected against Tcell acute lymphoblastic leukemia 47 48 cells (CCRFCEM cell line),210 was covalently coupled with the drug Doxorubicin and used in targeted 49 50 209 51 chemical therapies. The same aptamer has been attached to a gold nanoparticle incorporating 52 53 Doxorubicin to target specifically the CCRFCEM cell line.211 A prostatespecific membrane antigen 54 55 56 57 58 59 60 31 ACS Paragon Plus Environment ACS Combinatorial Science Page 32 of 81

1 2 3 targeting aptamer was conjugated to siRNA with streptavidinbiotin interactions and shown to uptake 4 5 212 6 even without the aid of delivery agents. 7 8 As with other RNA entities, incorporation of modified nucleotides can be used to avoid the 9 10 11 degradation of aptamers by nucleases. To this end, SELEX methods can be performed using 2’ modified 12 13 nucleotides to produce active, high affinity aptamers with increased resistance to nucleases. 14 15 Inorganic nanoparticles 16 17 18 Gold nanoscale spheres and rods have a variety of applications in biomedicine. For example, 19 20 Gold nanorods have been utilized for the cellular delivery of RNA/DNA nanoparticles. 213 Gold is a 21 22 frequently used moiety for the in vivo delivery of compounds because of its ease of conjugation, high 23 24 25 stability and biocompatibility. Gold nanoparticles have electronic and optical properties tunable through 26 27 their shape and size. Flow cytometry and atomic absorption spectroscopy showed selective targeting for 28 29 cancer cells and the effective killing of the targeted cells. Gold nanorods with the AS1411 aptamer were 30 31 214 32 used to construct a particle with a dimeric Gquadruplex. A gold nanoparticle functionalized with the 33 34 Sgc8c aptamer was used to deliver doxorubicin. 211 35 36 37 Microsponges 38 39 RNA’s instability in vitro and in vivo limits the amount of siRNA that can reach its intracellular 40 41 target. Traditionally, this is mitigated through incorporation of modified nucleotides in the RNA or the 42 43 44 use of additional carriers that may result in increased cytotoxicity or immunogenicity. A novel approach 45 46 to therapeutics that allows for delivery of more than half million siRNA copies in a particle presents 47 48 itself in RNAimicrosponges. DNA encoding for both sense and antisense siRNA strands and the T7 49 50 51 promoter sequence were nicked in a circular DNA. The rolling circle transcription principle was used to 52 53 produce long RNA strands containing tandem copies of hairpined siRNAs. The resulting RNA folds into 54 55 secondary structures and selfassembles into a compact tertiary structure. The final structure is 56 57 58 59 60 32 ACS Paragon Plus Environment Page 33 of 81 ACS Combinatorial Science

1 2 3 comprised of repetitive copies of Dicercleavable RNA strands that selfassemble into pleated sheets and 4 5 6 ultimately form spongelike microspheres. The microsponges’ structure was visualized by scanning 7 8 electron microscopy. 215, 216 9 10 11 Microsponges circumvent several cellular barriers by encompassing the properties of both a 12 13 carrier with its cargo. Additional complexation with cationic polymers like polyethylenimine (PEI) 14 15 generates a net positive surface for enhanced uptake without altering the siRNA conformation. The 16 17 18 microsponge’s design principles allows for incorporation of multiple siRNA species intended for 19 20 combination therapies. The efficiency of RNAimicrosponges for delivery and carrier volume provides 21 22 an effective new tool in RNA nanoparticle therapeutics. 217 The disadvantage of this technology is that 23 24 25 the RNAi machinery can be overpowered by the presence of a high dosage of small interfering RNA 26 27 molecules, at any given step, from exporting5 to Ago /RISC overloading. 218, 219 28 29 Encapsulated Delivery 30 31 32 Some drug delivery vehicles function by encapsulating the molecular cargo in a particle, so the 33 34 delivery vehicle rather than cargo predominantly determines the delivery properties. A particle that 35 36 37 consists of a lipidphase membrane and an aqueous phase core is called a vesicle; a particle consisting 38 39 only of a hydrophobic phase without an aqueous phase is called a micelle. These different molecular 40 41 entities can lead to encapsulation of molecular cargo. 42 43 44 Cationic lipids 45 46 Some lipids can form micelles or vesicles, thus being potential nanocarriers for drugs and 47 48 pharmaceuticals. 220222 Several features of lipids make them an attractive candidate for drug delivery, 49 50 51 such as their biocompatibility, ability to assemble into discrete structures to accommodate a payload of 52 53 drugs,223 ability to manipulate the particle size distribution 224226 and modification of the nanoparticle 54 55 56 57 58 59 60 33 ACS Paragon Plus Environment ACS Combinatorial Science Page 34 of 81

1 2 3 surface for ligand attachment. 222, 227 These basic traits of lipid molecules have been exploited further for 4 5 228, 229 6 delivery of nucleic acids including DNA and RNA. 7 8 An important consideration for using lipids for nucleic acid delivery relies on the presence of 9 10 11 one or more positive charges in the molecules of cationic lipids that can electrostatically interact with 12 13 negatively charged nucleic acids. Needless to say, the choice of lipid with desired hydrophobic, 14 15 hydrophilic and linker domains will dictate the resulting interactions with nucleic acids and overall 16 17 18 particle design. In this review we will mention the lipoplexes (liposomes), in the context of siRNA 19 20 delivery.230233 21 22 Historically, in the field of nucleic acid (including siRNA) delivery, the cationic lipids DOTAP 23 24 25 and DOTMA have been extensively utilized and studied for cell culturebased systems as well as for 26 27 small animal studies (Figure 3.a). As expected, electrostatic interactions between the positive charges 28 29 associated with the head groups of the lipid molecules and the negative charges on the siRNA are 30 31 32 critical for the formation and stability of the resulting lipoplexes. It may be noted that the 33 34 stereochemistry of the cationic lipids as well as their selfassembly characteristics also contribute to the 35 36 234 37 overall efficiency of siRNA transfection . 38 39 Noncationic lipids such as cholesterol or DOPE are often included to provide stability of the 40 41 selfassembled particles that contain the cationic lipids. These lipids are known to facilitate the 42 43 235 44 complexation with RNA, as well as improve transfection efficiency. Therefore, such lipids are dubbed 45 46 as “helper lipids”. A lipid mixture containing DOTAP and DOPE is a frequently used formulation for 47 48 RNA delivery and additional lipid molecules such as DSPC and cholesterol have also been included in 49 50 230 51 the DOTAP/DOPE mixture to enhance the efficiency of intracellular delivery of siRNA. 52 53 Lipofectamine 2000 and RNAimax are commercially available cationic lipids frequently utilized 54 55 for transfection of nucleic acids (including siRNAs or RNA nanostructures).236 The electrostatic 56 57 58 59 60 34 ACS Paragon Plus Environment Page 35 of 81 ACS Combinatorial Science

1 2 3 interactions between the positively charged lipid and the negatively charged nucleic acids lead to the 4 5 35 6 formation of lipoplexes. Nanorings functionalized with DSRNAs and complexed with the 7 8 commercially available transfection agent Lipofectamine 2000 (Invitrogen) display silencing of eGFP in 9 10 12 11 human breast cancer cells (MDA MB231) that stably express the fluorescent protein. 12 237 13 Sood and colleagues reported studies using neutral liposomes for siRNA delivery. A neutral 14 15 phospholipid, DOPC, was utilized to encapsulate a fluorescent siRNA targeted to downregulate an 16 17 18 oncoprotein EphA2, overexpressed in bladder cancer. DOPC formulations also contain low levels of a 19 20 detergent Tween20 that presumably assists in maintaining the liposome integrity during the 21 22 reconstitution step. 23 24 25 Stable nucleic acidlipid particles (SNALPs) are composed of cationic and fusogenic lipids and 26 27 contain a polyethylenglycol (PEG)lipid. 238 The combination of these lipids in SNALPs renders them 28 29 amenable to efficient intracellular uptake with an endosomal release potential. As expected the PEG 30 31 32 lipid modulates the surface charge from positive to neutral, while contributing to stability in circulation. 33 34 The SNALP design also relies on uncoating of the PEGlipid near diseased tissue resulting in the 35 36 37 presentation of the positively charged siRNAlipoplexes for intracellular uptake. PLK1 SNALP (TKM 38 39 PLK1, TKM080301) is a formulation that contains small interfering RNA (siRNA) directed against the 40 41 serine/threonine kinase PLK1 (an enzyme involved in cell cycle progression) and is currently 42 43 44 undergoing clinical trials (NCT01262235). 45 46 Lipidbased nanoparticles (LNP) have been employed to deliver both in vitro and in vivo yet 47 48 another class of therapeutic RNAs: mRNAs for protein supplementing or protein replacement 49 50 76 51 therapies. Moreover, mRNAbased vaccines successfully induce immune response with prophylactic 52 53 resolutions. These vaccines are delivered with smart designed lipid nanoparticles, which incorporate 54 55 targeting moieties on their surface and endosomal escape adjuvants in their composition, and are in 56 57 58 59 60 35 ACS Paragon Plus Environment ACS Combinatorial Science Page 36 of 81

1 2 3 various stages of clinical trials. 239 LNP formulation for mRNA delivery can be specifically optimized for 4 5 6 structure and stability, protection from aggregation and nonspecific endocytosis, and enhanced 7 8 endosomal escape. 240 9 10 11 Oxime Ether Lipids 12 13 Oxime ether lipids (OELs) are a relatively new class of cationic agents explored as efficient 14 15 transfection tools (Figure 3.a).241, 242 In contrast to commonly used transfection lipids, OELs contain 16 17 18 oxime ether bonds and can be synthesized by a simple and efficient click chemistry approach. Oxime 19 20 ether linkages are relatively stable at neutral pH, but can be cleaved at low pH values, thus providing a 21 22 builtin nucleic acid release mechanism. By synthesizing a variety of oxime ether lipids and examining 23 24 25 their siRNA delivery potential, it was demonstrated that an interplay between the hydrophobicity, degree 26 27 of unsaturation of the fatty acyl chains and head group polarities play a role in their electrostatic 28 29 interaction with RNA, protection from nucleases, their uptake and gene silencing in a cell culture 30 31 243 32 system. 33 34 Bolaamphiphile surfactants 35 36 37 Amphiphilic molecules contain both hydrophilic as well as hydrophobic regions. Aggregates of 38 39 amphiphilic molecules can lead to the situation that regions with mostly hydrophilic groups try to 40 41 maximize their contact with water, while the hydrophobic groups try to minimize their contact with 42 43 44 water. Bolaamphiphiles consist of a hydrophobic aliphatic chain that is terminated by a hydrophilic head 45 46 group or groups at each of the two ends (Figure 3.b). They are able to form either micelles or vesicles. 47 48 The name originates from “bola”, a type of weapon consisting of a cord with weights at each end.244 49 50 51 Bolas have been shown to be capable not only of crossing the , but also of 52 53 delivering cargo across the bloodbrain barrier.193 The types of cargos tested with bola carriers include 54 55 RNAs, plasmid DNAs, proteins and peptides.245247 In one of the studies aimed at characterization of the 56 57 58 59 60 36 ACS Paragon Plus Environment Page 37 of 81 ACS Combinatorial Science

1 2 3 biophysical properties of different bola variants it was demonstrated in vitro and in vivo that siRNAs 4 5 6 formulated with bolas GLH19 and GLH20 (see Figure 3.b) can be delivered efficiently and with low 7 8 toxicity.248 These two bola variants have a hydrophobic core derived from vernonia oil and 9 10 11 acetylcholinebased head groups (AChHG). In the GLH20 variant, AChHG can be hydrolyzed by 12 13 acetylcholine esterase, thus helping to free the cargo from the carrier in the brain. Molecular dynamics 14 15 (MD) simulations of the GLH19 and GLH20 micelle variants predicted higher binding affinity to RNA 16 17 18 and thus better protection against degradation for GLH19. In vitro results agreed with these 19 20 predictions, showing nearly no degradation by nucleases and a slightly better cellular uptake of the RNA 21 22 with the GLH19 carrier, while the silencing of the green fluorescent protein (GFP) expression in human 23 24 25 breast cancer cells (MDAMB231) by siRNAs formulated with GLH19 and GLH 20 was equally 26 27 effective. In vivo biodistribution studies in athymic nude mice with MDAMB231based xenograft 28 29 tumor demonstrated differentially higher uptake of the siRNA formulated with GLH19 into the tumor 30 31 248 32 following a systemic tailvein injection. Finally, independently of the previous result, hexameric ring 33 34 nanoparticles with siRNA Dicersubstrate arms, using bolas GLH19 and 20 as carriers and delivered by 35 36 14 37 intratumoral injections demonstrated silencing of the GFP expression. The computational and in vitro 38 39 characterization of bolas GLH19 and 20 illustrate the balance between the binding affinity affecting 40 41 transfection efficiency and the ease of RNA release inside the cell that effectively may compensate for 42 43 44 the lower transfection rate. Similar effects were observed in a more recent study in which two other bola 45 46 micelle variants GLH58 and GLH60 were characterized both computationally and experimentally ( in 47 48 vitro ).243 GLH58 and GLH60 have hydrophobic cores derived from jojoba oil, and two (GLH58) or 49 50 51 four (GLH60) GLH20like ACh head groups. 3D structure modeling and MD simulations (Figure 4), 52 53 combined with experimental results indicated that the design differences between GLH58 and GLH60 54 55 influence the balance between the RNA protection against degradation, efficiency of delivery and ease 56 57 58 59 60 37 ACS Paragon Plus Environment ACS Combinatorial Science Page 38 of 81

1 2 3 of RNA release. In MD simulations the GLH58 bolas formed more stable nanosize micelles, while the 4 5 6 GLH60sbased micelle models tended to fall apart quickly, indicating the repulsive forces of these four 7 8 headed bolas as disruptive to their stability and aggregation. MD results showed similar amounts of 9 10 11 solventaccessible surface areas of RNA helices complexed with both bolas, implying similar levels of 12 13 direct contact with solvent (and nucleases). These results are consistent with the experimental data 14 15 showing comparable protection against nucleases in DNA helices formulated with GLH58 and GLH 16 17 18 60. The fourheaded GLH60 showed a tendency to bind more strongly by bringing more head groups in 19 20 the proximity of RNA, but it reached saturation levels at lower concentration, with one layer of the bolas 21 22 repulsing additional bola molecules present in solvent. By contrast, the twoheaded GLH58 can coat 23 24 25 RNA in larger numbers and potentially multiple layers, yet its binding, measured by the number of head 26 27 groups in the proximity of RNA, is weaker, thus enabling easier release of its cargo (see Figure 3.b). All 28 29 these characteristics combined are consistent with the experimentally observed better silencing of the 30 31 32 GFP gene in human breast cancer cells by siRNAs using GLH58 as carrier, relative to the GLH60. All 33 34 of the computational, in vitro and in vivo results consistently indicate that bolas can be practical nucleic 35 36 37 acid carriers with adjustable protection and release qualities. 38 39 RNA NPs can be complexated with delivery agents that assist their uptake into cells. The 40 41 presumed mechanism of cellular entry for such formulations is endosomal uptake, regardless of the type 42 43 44 of interaction between the delivery agent and the RNA NPs (conjugation, mediation or encapsulation). 45 46 Upon endosomal release, the NP is available to perform its intended role. Such a role may simply be the 47 48 delivery of a payload to the cytoplasm, where it can be acted upon by cellular components, and 49 50 51 ultimately result in a predetermined response. Delivery of DS RNA by RNA nanoparticles allows for 52 53 the generation of siRNA once processed by Dicer, resulting in decreased levels of target protein 54 55 expression due the resulting RNAi response (Figure 5). 56 57 58 59 60 38 ACS Paragon Plus Environment Page 39 of 81 ACS Combinatorial Science

1 2 3 Exosomes 4 5 6 Exosomes are vesicles of cellular origin. The vesicles do not possess cellular organelles but 7 8 contain cellular molecular content like proteins, RNAs and even DNA fragments. These vesicles can be 9 10 11 repurposed for drugdelivery by loading them with the desired cargo. Cellular siRNAs delivery using 12 249 13 exosomes has been accomplished. 14 15 Viral Delivery 16 17 18 Cellular delivery of RNA or DNA can be accomplished with viral delivery, where viral vectors 19 20 transfect the exogenous cargo. Cellular delivery of exogenous RNA has been accomplished with 21 22 attenuated Adenovirus, Adenoassociated , Lentivirus, Baculovirus and others. 250 Most applications 23 24 25 are based on replicationdefective viral vectors. A potential use of replicationcompetent viral vectors is 26 27 to deliver drugs to otherwise difficulttoreach solid tumortissue.250 An advantage of viral vectors are 28 29 their efficient transfection of molecular cargo. A potential disadvantage is immunogenicity due to innate 30 31 35 32 immune system response. 33 34 35 36 37 Prospects in RNA Nanobiology 38 39 Nucleic acids are a perfectly suitable nanotechnological material for in vivo applications: their 40 41 biological origin provides natural solutions to toxicity, biocompatibility and biodegradability concerns. 42 43 44 Their inexpensive, simple and accurate synthesis, both chemically and enzymatically, is an additional 45 46 advantage for utilizing them in nanomedical applications. RNAbased nanoparticles are fully 47 48 programmable, with precise control over the dimension, geometry, and composition. Concurrent self 49 50 51 assembly with cotranscriptional synthesis allows for their in vivo production, another great advantage 52 53 over other materials. The ease of designing and fabricating an “antidote” based on sequence 54 55 complementarity confers RNA NPs rapid reversal of drug activity. Various RNA nanoconstructs have 56 57 58 59 60 39 ACS Paragon Plus Environment ACS Combinatorial Science Page 40 of 81

1 2 3 been assembled, yet only few have been functionalized and tested in cell culture and in vivo . Efficacy 4 5 6 and transfectability of the RNA NPs are directly correlated with their size and shape. Thus, first and 7 8 foremost, an optimization of functional construct dimension and form would be imperative for RNA 9 10 11 therapeutic applications. Other challenges left in RNA nanotechnology pertain to the scaling up of 12 13 production and the purity of the nanoconstructs. 14 15 Systemic, intravenous delivery of therapeutic RNA NPs requires consideration to be given to all 16 17 18 cellular barriers. The enzymatic stability of RNA in the blood stream is one of the primary concerns. In 19 20 order to increase their resistance to nucleases, one option is to chemically modify (at least) the most 21 22 susceptible nucleotides in the single strandedelements of the nanoparticles. Moreover, chemical 23 24 25 modifications provide a way to tune the efficiency, enzymatic and thermal stability, biodistribution, 26 27 uptake and toxicity of RNAs, especially siRNAs. Silencing lasts a few days to a week, and re 28 29 administration of siRNAs might be necessary for prolonging their effect. An optimized sequence design 30 31 32 can aid in the matter as well, further minimizing siRNA offtarget effects. Therapeutic usage of RNA 33 34 NPs with modified nucleotides incorporated permits functionalization for direct targeting and minimal 35 36 37 immune response, allowing for repeated treatment required by cancer and chronic diseases. 38 39 An alternative protective method is to couple the RNA NPs with carrier platforms that can aid in 40 41 their cellular delivery. Such compounds can act as transfection agents and give the RNA NPs additional 42 43 44 protection against aggregation with serum proteins, which may interfere with the designed NP’s 45 46 function. Both the RNA NP and the carrier have to avoid undesirable protein interactions and 47 48 stimulation of an innate immune response. Current viral delivery platforms are used to deliver plasmid 49 50 51 DNA and permit higher nucleic acid packing capacity, but present side effects, such as insertional 52 53 mutagenesis, and intrinsic immunogenicity. Nonviral delivery carriers are generally less immunogenic 54 55 and are easily appended with other functional moieties for multivalent purposes or direct targeting 56 57 58 59 60 40 ACS Paragon Plus Environment Page 41 of 81 ACS Combinatorial Science

1 2 3 capabilities, but have less packing ability. In the case of using a delivery vehicle to supply the RNA NPs 4 5 6 to the cells, the overall size of the complex should be 1060 nm in order to avoid renal clearance and yet 7 8 be able to be internalized by the cells. 9 10 11 Addition of cell specific targeting elements to the RNA NPs themselves or to the carriers can 12 13 further enhance the strength and the selectivity of the initial contact between the NPs and cells. Ideally, 14 15 the distribution of the particles should be specific to and uniform within the tumor, with little or no 16 17 18 accumulation in organs, and no immune reaction. 19 20 Since endocytosis is the typical mechanism responsible for the uptake of nanoparticles into cells, 21 22 late endosomal escape is a crucial aspect to be considered in the delivery platform design. Furthermore, 23 24 25 the release of the functional components of the nanoparticle in the cellular environment has to be 26 27 achieved in order to activate the designed functionalities. Latest intelligent designs of nucleic acid 28 29 delivery platforms avoid their endosomal entrapment and lysosomal degradation. Incorporating proton 30 31 32 absorbing elements in cationic Polyethylenimine (PEI) polymers and Poly(amidoamine) (PAMAM) 33 34 dendrimers, or hemolytic, pHdependent elements in anionic polymers enhances endosomal escape. 35 36 37 Acidificationdependent, membrane disruptive peptide or protein make use as well of the change in the 38 39 environment’s pH, ensuring the release of their cargo. Natural or natureimitating poreforming toxins 40 41 have been employed to aid in phagosomes or phagolysosomes escape. Lipoplexes resulting from 42 43 44 fusogenic lipid incorporated liposomes, such as 1,2dioleoylsn glycero3phosphoethanolamine 45 46 (DOPE), also promote endosomal escape. Likewise, chemical compounds, such as Amphotericin B, 47 48 chloroquine or chloroquine analogs with a lower cytotoxicity have been successfully used as 49 50 51 endosomedisruptive agents. 52 53 Plasmid DNA has to reach the nucleus, while miRNAs and siRNAs have to reach the cytoplasm. 54 55 Several delivery platforms are suitable for in vitro use, yet they prove to be toxic in vivo . The fine 56 57 58 59 60 41 ACS Paragon Plus Environment ACS Combinatorial Science Page 42 of 81

1 2 3 balance between their transfection efficiency and their cellular toxicity is still a work in progress. 4 5 6 Accordingly, the process of designing the RNA NPs and their delivery platforms has to balance 7 8 protection, cellular delivery and release characteristics. 9 10 11 12 13 Acknowledgements 14 15 This work has been funded in whole or in part with Federal funds from the Frederick National 16 17 18 Laboratory for Cancer Research, National Institutes of Health, under Contract No. 19 20 HHSN261200800001E. This research was supported [in part] by the Intramural Research Program of 21 22 the NIH, National Cancer Institute, Center for Cancer Research. The content of this publication does not 23 24 25 necessarily reflect the views or policies of the Department of Health and Human Services, nor does 26 27 mention of trade names, commercial products, or organizations imply endorsement by the U.S. 28 29 Government. 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 42 ACS Paragon Plus Environment Page 43 of 81 ACS Combinatorial Science

1 2 3 References 4 5 6 7 8 1. Gerrard, J. A. Protein nanotechnology: what is it? Methods Mol Biol . 2013 , 996 , 115. 9 10 2. Patil, S.; Vhora, I.; Amrutiya, J.; Lalani, R.; Misra, A. Role of Nanotechnology in Delivery of 11 12 13 Protein and Peptide Drugs . Curr Pharm Des . 2015 , 21(29) , 415573. 14 15 3. Jones, M. R.; Seeman, N. C.; Mirkin, C. A. Nanomaterials. Programmable materials and the 16 17 nature of the DNA bond . Science . 2015 , 347(6224) , 1260901. 18 19 20 4. Seeman, N. C. DNA nanotechnology: novel DNA constructions . Annu Rev Biophys Biomol 21 22 Struct . 1998 , 27 , 22548. 23 24 5. Seeman, N. C. Nanomaterials based on DNA . Annu Rev Biochem . 2010 , 79 , 6587. 25 26 27 6. Saaem, I.; LaBean, T. H. Overview of DNA origami for molecular selfassembly . Wiley 28 29 Interdiscip Rev Nanomed Nanobiotechnol . 2013 , 5(2) , 15062. 30 31 32 7. Sacca, B.; Niemeyer, C. M. DNA origami: the art of folding DNA . Angew Chem Int Ed Engl . 33 34 2012 , 51(1) , 5866. 35 36 8. Jaeger, L.; Westhof, E.; Leontis, N. B. TectoRNA: modular assembly units for the construction 37 38 39 of RNA nanoobjects . Nucleic Acids Res . 2001 , 29(2) , 45563. 40 41 9. Nasalean, L.; Stombaugh, J.; Zirbel, C. L.; Leontis, N. B., RNA 3D structural motifs: definition, 42 43 identification, annotation, and database searching., In Non-protein coding RNAs . 2009, Springer. 44 45 46 pp. 126. 47 48 10. Afonin, K. A.; Bindewald, E.; Yaghoubian, A. J.; Voss, N.; Jacovetty, E.; Shapiro, B. A.; Jaeger, 49 50 L. In vitro assembly of cubic RNAbased scaffolds designed in silico . Nature Nanotech . 2010 , 51 52 53 5(9) , 676682. 54 55 11. Yingling, Y. G.; Shapiro, B. A. Computational Design of an RNA Hexagonal Nanoring and an 56 57 58 RNA Nanotube . Nano Letters . 2007 , 7(8) , 23282334. 59 60 43 ACS Paragon Plus Environment ACS Combinatorial Science Page 44 of 81

1 2 3 12. Afonin, K. A.; Kireeva, M.; Grabow, W. W.; Kashlev, M.; Jaeger, L.; Shapiro, B. A. Co 4 5 6 transcriptional Assembly of Chemically Modified RNA Nanoparticles Functionalized with 7 8 siRNAs . Nano Letters . 2012 , 12(10) , 51925195. 9 10 11 13. Diener, T. O.; Raymer, W. B. Potato spindle tuber virus: a plant virus with properties of a free 12 13 nucleic acid . Science . 1967 , 158(3799) , 37881. 14 15 14. Afonin, K. A.; Viard, M.; Koyfman, A. Y.; Martins, A. N.; Kasprzak, W. K.; Panigaj, M.; Desai, 16 17 18 R.; Santhanam, A.; Grabow, W. W.; Jaeger, L.; Heldman, E.; Reiser, J.; Chiu, W.; Freed, E. O.; 19 20 Shapiro, B. A. Multifunctional RNA Nanoparticles . Nano Letters . 2014 , 14(10) , 56625671. 21 22 15. Ishikawa, J.; Furuta, H.; Ikawa, Y. RNA tectonics (tectoRNA) for RNA nanostructure design and 23 24 25 its application in synthetic biology . Wiley Interdiscip Rev RNA . 2013 , 4(6) , 65164. 26 27 16. Afonin, K. A.; Desai, R.; Viard, M.; Kireeva, M. L.; Bindewald, E.; Case, C. L.; Maciag, A. E.; 28 29 Kasprzak, W. K.; Kim, T.; Sappe, A.; Stepler, M.; KewalRamani, V. N.; Kashlev, M.; 30 31 32 Blumenthal, R.; Shapiro, B. A. Cotranscriptional production of RNADNA hybrids for 33 34 simultaneous release of multiple split functionalities . Nucleic Acids Research . 2013 , 42(3) , 2085 35 36 37 2097. 38 39 17. Grabow, W. W.; Jaeger, L. RNA selfassembly and RNA nanotechnology . Acc Chem Res . 2014 , 40 41 47(6) , 187180. 42 43 44 18. Cech, T. R.; Zaug, A. J.; Grabowski, P. J. In vitro splicing of the ribosomal RNA precursor of 45 46 Tetrahymena: involvement of a guanosine nucleotide in the excision of the intervening sequence . 47 48 Cell . 1981 , 27(3 Pt 2) , 48796. 49 50 51 19. GuerrierTakada, C.; Gardiner, K.; Marsh, T.; Pace, N.; Altman, S. The RNA moiety of 52 53 ribonuclease P is the catalytic subunit of the enzyme . Cell . 1983 , 35(3 Pt 2) , 84957. 54 55 56 57 58 59 60 44 ACS Paragon Plus Environment Page 45 of 81 ACS Combinatorial Science

1 2 3 20. Kruger, K.; Grabowski, P. J.; Zaug, A. J.; Sands, J.; Gottschling, D. E.; Cech, T. R. Selfsplicing 4 5 6 RNA: autoexcision and autocyclization of the ribosomal RNA intervening sequence of 7 8 Tetrahymena . Cell . 1982 , 31(1) , 14757. 9 10 11 21. Ramakrishnan, V. The ribosome emerges from a black box . Cell . 2014 , 159(5) , 97984. 12 13 22. Fire, A.; Xu, S.; Montgomery, M. K.; Kostas, S. A.; Driver, S. E.; Mello, C. C. Potent and 14 15 specific genetic interference by doublestranded RNA in Caenorhabditis elegans . Nature . 1998 , 16 17 18 391(6669) , 80611. 19 20 23. Friedman, R. C.; Farh, K. K.; Burge, C. B.; Bartel, D. P. Most mammalian mRNAs are 21 22 conserved targets of microRNAs . Genome Res . 2009 , 19(1) , 92105. 23 24 25 24. Lin, S.; Gregory, R. I. MicroRNA biogenesis pathways in cancer . Nat Rev Cancer . 2015 , 15(6) , 26 27 32133. 28 29 25. Selbach, M.; Schwanhausser, B.; Thierfelder, N.; Fang, Z.; Khanin, R.; Rajewsky, N. 30 31 32 Widespread changes in protein synthesis induced by microRNAs . Nature . 2008 , 455(7209) , 58 33 34 63. 35 36 37 26. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Radmark, O.; Kim, S.; 38 39 Kim, V. N. The nuclear RNase III Drosha initiates microRNA processing . Nature . 2003 , 40 41 425(6956) , 4159. 42 43 44 27. Okamura, K.; Hagen, J. W.; Duan, H.; Tyler, D. M.; Lai, E. C. The mirtron pathway generates 45 46 microRNAclass regulatory RNAs in Drosophila . Cell . 2007 , 130(1) , 89100. 47 48 28. Lund, E.; Dahlberg, J. E. Substrate selectivity of exportin 5 and Dicer in the biogenesis of 49 50 51 microRNAs . Cold Spring Harb Symp Quant Biol . 2006 , 71 , 5966. 52 53 29. Pratt, A. J.; MacRae, I. J. The RNAinduced silencing complex: a versatile genesilencing 54 55 machine . J Biol Chem . 2009 , 284(27) , 17897901. 56 57 58 59 60 45 ACS Paragon Plus Environment ACS Combinatorial Science Page 46 of 81

1 2 3 30. Davis, B. N.; Hata, A. Regulation of MicroRNA Biogenesis: A miRiad of mechanisms . Cell 4 5 6 Commun Signal . 2009 , 7, 18. 7 8 31. Hirakata, S.; Siomi, M. C. piRNA biogenesis in the germline: From transcription of piRNA 9 10 11 genomic sources to piRNA maturation . Biochim Biophys Acta . 2016 , 1859(1) , 8292. 12 13 32. Nykanen, A.; Haley, B.; Zamore, P. D. ATP requirements and small interfering RNA structure in 14 15 the RNA interference pathway . Cell . 2001 , 107(3) , 30921. 16 17 18 33. Dillin, A. The specifics of small interfering RNA specificity . Proc Natl Acad Sci U S A . 2003 , 19 20 100(11) , 628991. 21 22 34. Carthew, R. W.; Sontheimer, E. J. Origins and Mechanisms of miRNAs and siRNAs . Cell . 2009 , 23 24 25 136(4) , 64255. 26 27 35. Xu, C.; Wang, J. Delivery systems for siRNA drug development in cancer therapy . Asian 28 29 Journal of Pharmaceutical Sciences . 2015 , 10(1) , 112. 30 31 32 36. Nahvi, A.; Sudarsan, N.; Ebert, M. S.; Zou, X.; Brown, K. L.; Breaker, R. R. Genetic control by 33 34 a metabolite binding mRNA . Chem Biol . 2002 , 9(9) , 1043. 35 36 37 37. Gilbert, S. D.; Stoddard, C. D.; Wise, S. J.; Batey, R. T. Thermodynamic and kinetic 38 39 characterization of ligand binding to the purine riboswitch aptamer domain . J Mol Biol . 2006 , 40 41 359(3) , 75468. 42 43 44 38. Lemay, J. F.; Lafontaine, D. A. Core requirements of the adenine riboswitch aptamer for ligand 45 46 binding . RNA . 2007 , 13(3) , 33950. 47 48 39. Noeske, J.; Buck, J.; Furtig, B.; Nasiri, H. R.; Schwalbe, H.; Wohnert, J. Interplay of 'induced fit' 49 50 51 and preorganization in the ligand induced folding of the aptamer domain of the guanine binding 52 53 riboswitch . Nucleic Acids Res . 2007 , 35(2) , 57283. 54 55 56 57 58 59 60 46 ACS Paragon Plus Environment Page 47 of 81 ACS Combinatorial Science

1 2 3 40. Ellington, A. D.; Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands . 4 5 6 Nature . 1990 , 346(6287) , 81822. 7 8 41. Gold, L. SELEX: How It Happened and Where It will Go . J Mol Evol . 2015 , 81(5-6) , 1403. 9 10 11 42. Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to 12 13 bacteriophage T4 DNA polymerase . Science . 1990 , 249(4968) , 50510. 14 15 43. Zhu, H.; Li, J.; Zhang, X. B.; Ye, M.; Tan, W. Nucleic acid aptamermediated drug delivery for 16 17 18 targeted cancer therapy . ChemMedChem . 2015 , 10(1) , 3945. 19 20 44. Xiang, D.; Shigdar, S.; Qiao, G.; Wang, T.; Kouzani, A. Z.; Zhou, S. F.; Kong, L.; Li, Y.; Pu, C.; 21 22 Duan, W. Nucleic acid aptamerguided cancer therapeutics and diagnostics: the next generation 23 24 25 of cancer medicine . Theranostics . 2015 , 5(1) , 2342. 26 27 45. Yunn, N. O.; Koh, A.; Han, S.; Lim, J. H.; Park, S.; Lee, J.; Kim, E.; Jang, S. K.; Berggren, P. 28 29 O.; Ryu, S. H. Agonistic aptamer to the insulin receptor leads to biased signaling and functional 30 31 32 selectivity through allosteric modulation . Nucleic Acids Res . 2015 , 43(16) , 7688701. 33 34 46. Wallukat, G.; Muller, J.; Haberland, A.; Berg, S.; Schulz, A.; Freyse, E. J.; Vetter, R.; Salzsieder, 35 36 37 E.; Kreutz, R.; Schimke, I. Aptamer BC007 for neutralization of pathogenic autoantibodies 38 39 directed against Gprotein coupled receptors: A vision of future treatment of patients with 40 41 cardiomyopathies and positivity for those autoantibodies . Atherosclerosis . 2016 , 244 , 447. 42 43 44 47. Huang, Y. Z.; Hernandez, F. J.; Gu, B.; Stockdale, K. R.; Nanapaneni, K.; Scheetz, T. E.; 45 46 Behlke, M. A.; Peek, A. S.; Bair, T.; Giangrande, P. H.; McNamara, J. O., 2nd RNA aptamer 47 48 based functional ligands of the neurotrophin receptor, TrkB . Mol Pharmacol . 2012 , 82(4) , 623 49 50 51 35. 52 53 48. Khedri, M.; Rafatpanah, H.; Abnous, K.; Ramezani, P.; Ramezani, M. Cancer immunotherapy 54 55 via nucleic acid aptamers . Int Immunopharmacol . 2015 , 29(2) , 92636. 56 57 58 59 60 47 ACS Paragon Plus Environment ACS Combinatorial Science Page 48 of 81

1 2 3 49. Jo, H.; Ban, C. Aptamernanoparticle complexes as powerful diagnostic and therapeutic tools . 4 5 6 Exp Mol Med . 2016 , 48 , e230. 7 8 50. Maier, K. E.; Levy, M. From selection hits to clinical leads: progress in aptamer discovery . Mol 9 10 11 Ther Methods Clin Dev . 2016 , 5, 16014. 12 13 51. Bompiani, K. M.; Monroe, D. M.; Church, F. C.; Sullenger, B. A. A high affinity, antidote 14 15 controllable prothrombin and thrombinbinding RNA aptamer inhibits thrombin generation and 16 17 18 thrombin activity . J Thromb Haemost . 2012 , 10(5) , 87080. 19 20 52. Hirota, M.; Murakami, I.; Ishikawa, Y.; Suzuki, T.; Sumida, S.; Ibaragi, S.; Kasai, H.; Horai, N.; 21 22 Drolet, D. W.; Gupta, S.; Janjic, N.; Schneider, D. J. Chemically Modified Interleukin6 23 24 25 Aptamer Inhibits Development of CollagenInduced Arthritis in Cynomolgus Monkeys . Nucleic 26 27 Acid Ther . 2016 , 26(1) , 109. 28 29 53. Kang, S. A.; Hasan, N.; Mann, A. P.; Zheng, W.; Zhao, L.; Morris, L.; Zhu, W.; Zhao, Y. D.; 30 31 32 Suh, K. S.; Dooley, W. C.; Volk, D.; Gorenstein, D. G.; Cristofanilli, M.; Rui, H.; Tanaka, T. 33 34 Blocking the adhesion cascade at the premetastatic niche for prevention of breast cancer 35 36 37 metastasis . Mol Ther . 2015 , 23(6) , 104454. 38 39 54. Liu, G. X.; Xi, H. Q.; Sun, X. Y.; Wei, B. Role of periostin and its antagonist PNDA3 in gastric 40 41 cancer metastasis . World J Gastroenterol . 2015 , 21(9) , 260513. 42 43 44 55. HervasStubbs, S.; Soldevilla, M. M.; Villanueva, H.; Mancheno, U.; Bendandi, M.; Pastor, F. 45 46 Identification of TIM3 2'fluoro oligonucleotide aptamer by HTSELEX for cancer 47 48 immunotherapy . Oncotarget . 2016 , 7(4) , 452230. 49 50 51 56. Douglas, A. G.; Wood, M. J. RNA splicing: disease and therapy . Brief Funct Genomics . 2011 , 52 53 10(3) , 15164. 54 55 56 57 58 59 60 48 ACS Paragon Plus Environment Page 49 of 81 ACS Combinatorial Science

1 2 3 57. Havens, M. A.; Duelli, D. M.; Hastings, M. L. Targeting RNA splicing for disease therapy . 4 5 6 Wiley Interdiscip Rev RNA . 2013 , 4(3) , 24766. 7 8 58. Lentz, J. J.; Jodelka, F. M.; Hinrich, A. J.; McCaffrey, K. E.; Farris, H. E.; Spalitta, M. J.; Bazan, 9 10 11 N. G.; Duelli, D. M.; Rigo, F.; Hastings, M. L. Rescue of hearing and vestibular function by 12 13 antisense oligonucleotides in a mouse model of human deafness . Nat Med . 2013 , 19(3) , 34550. 14 15 59. Serganov, A.; Nudler, E. A decade of riboswitches . Cell . 2013 , 152(1-2) , 1724. 16 17 18 60. Bindewald, E.; Afonin, K. A.; Viard, M.; Zakrevsky, P.; Kim, T.; Shapiro, B. A. Multistrand 19 20 Structure Prediction of Nucleic Acid Assemblies and Design of RNA Switches . Nano Lett . 2016 , 21 22 16(3) , 172635. 23 24 25 61. FerreD'Amare, A. R.; Scott, W. G. Small selfcleaving ribozymes . Cold Spring Harb Perspect 26 27 Biol . 2010 , 2(10) , a003574. 28 29 62. Woese, C. R.; Dugre, D. H.; Saxinger, W. C.; Dugre, S. A. The molecular basis for the genetic 30 31 32 code . Proc Natl Acad Sci U S A . 1966 , 55(4) , 96674. 33 34 63. Gilbert, W. Origin of life: The RNA world . Nature . 1986 , 319(6055) , 618618. 35 36 37 64. Gwiazda, S.; Salomon, K.; Appel, B.; Muller, S. RNA selfligation: from oligonucleotides to full 38 39 length ribozymes . Biochimie . 2012 , 94(7) , 145763. 40 41 65. Fedor, M. J.; Williamson, J. R. The catalytic diversity of RNAs . Nat Rev Mol Cell Biol . 2005 , 42 43 44 6(5) , 399412. 45 46 66. Burke, C. R.; Lupták, A., Catalytic RNA, In eLS . 2001, John Wiley & Sons, Ltd. 47 48 67. Jimenez, R. M.; Polanco, J. A.; Luptak, A. Chemistry and Biology of SelfCleaving Ribozymes . 49 50 51 Trends Biochem Sci . 2015 , 40(11) , 64861. 52 53 54 55 56 57 58 59 60 49 ACS Paragon Plus Environment ACS Combinatorial Science Page 50 of 81

1 2 3 68. Loh, E.; Dussurget, O.; Gripenland, J.; Vaitkevicius, K.; Tiensuu, T.; Mandin, P.; Repoila, F.; 4 5 6 Buchrieser, C.; Cossart, P.; Johansson, J. A transacting riboswitch controls expression of the 7 8 virulence regulator PrfA in Listeria monocytogenes . Cell . 2009 , 139(4) , 7709. 9 10 11 69. Tang, J.; Breaker, R. R. Structural diversity of selfcleaving ribozymes . Proc Natl Acad Sci U S 12 13 A. 2000 , 97(11) , 57849. 14 15 70. Lincoln, T. A.; Joyce, G. F. Selfsustained replication of an RNA enzyme . Science . 2009 , 16 17 18 323(5918) , 122932. 19 20 71. Winkler, W. C.; Nahvi, A.; Roth, A.; Collins, J. A.; Breaker, R. R. Control of gene expression by 21 22 a natural metaboliteresponsive ribozyme . Nature . 2004 , 428(6980) , 2816. 23 24 25 72. Ge, L.; Resnick, N. M.; Ernst, L. K.; Salvucci, L. A.; Asman, D. C.; Cooper, D. L. Gene 26 27 therapeutic approaches to primary and metastatic brain tumors: II. ribozymemediated 28 29 suppression of CD44 expression . J Neurooncol . 1995 , 26(3) , 2517. 30 31 32 73. Kuriyama, M.; Kondo, Y.; Tanaka, Y. Pseudoknot interactionmediated activation of type I 33 34 hammerhead ribozyme: a new class of genetherapeutic agents . Nucleosides Nucleotides Nucleic 35 36 37 Acids . 2014 , 33(7) , 46680. 38 39 74. Leavitt, M. C.; Yu, M.; Yamada, O.; Kraus, G.; Looney, D.; Poeschla, E.; WongStaal, F. 40 41 Transfer of an antiHIV1 ribozyme gene into primary human lymphocytes . Hum Gene Ther . 42 43 44 1994 , 5(9) , 111520. 45 46 75. Shen, S.; Rodrigo, G.; Prakash, S.; Majer, E.; Landrain, T. E.; Kirov, B.; Daros, J. A.; Jaramillo, 47 48 A. Dynamic signal processing by ribozymemediated RNA circuits to control gene expression . 49 50 51 Nucleic Acids Res . 2015 , 43(10) , 515870. 52 53 76. Sahin, U.; Kariko, K.; Tureci, O. mRNAbased therapeuticsdeveloping a new class of drugs . 54 55 Nat Rev Drug Discov . 2014 , 13(10) , 75980. 56 57 58 59 60 50 ACS Paragon Plus Environment Page 51 of 81 ACS Combinatorial Science

1 2 3 77. Wiedenheft, B.; Sternberg, S. H.; Doudna, J. A. RNAguided genetic silencing systems in 4 5 6 bacteria and archaea . Nature . 2012 , 482(7385) , 3318. 7 8 78. PerezPinera, P.; Kocak, D. D.; Vockley, C. M.; Adler, A. F.; Kabadi, A. M.; Polstein, L. R.; 9 10 11 Thakore, P. I.; Glass, K. A.; Ousterout, D. G.; Leong, K. W.; Guilak, F.; Crawford, G. E.; Reddy, 12 13 T. E.; Gersbach, C. A. RNAguided gene activation by CRISPRCas9based transcription 14 15 factors . Nat Methods. 2013 , 10(10) , 9736. 16 17 18 79. Maeder, M. L.; Linder, S. J.; Cascio, V. M.; Fu, Y.; Ho, Q. H.; Joung, J. K. CRISPR RNA 19 20 guided activation of endogenous human genes . Nat Methods . 2013 , 10(10) , 9779. 21 22 80. Zalatan, J. G.; Lee, M. E.; Almeida, R.; Gilbert, L. A.; Whitehead, E. H.; La Russa, M.; Tsai, J. 23 24 25 C.; Weissman, J. S.; Dueber, J. E.; Qi, L. S.; Lim, W. A. Engineering complex synthetic 26 27 transcriptional programs with CRISPR RNA scaffolds . Cell . 2015 , 160(1-2) , 33950. 28 29 81. Afonin, K. A.; Bindewald, E.; Kireeva, M.; Shapiro, B. A. Computational and Experimental 30 31 32 Studies of Reassociating RNA/DNA Hybrids Containing Split Functionalities , in Methods in 33 34 Enzymology. 2015 , Elsevier BV. p. 313334. 35 36 37 82. Afonin, K. A.; Viard, M.; Martins, A. N.; Lockett, S. J.; Maciag, A. E.; Freed, E. O.; Heldman, 38 39 E.; Jaeger, L.; Blumenthal, R.; Shapiro, B. A. Activation of different split functionalities on re 40 41 association of RNA–DNA hybrids . Nature Nanotech . 2013 , 8(4) , 296304. 42 43 44 83. Bindewald, E.; Hayes, R.; Yingling, Y. G.; Kasprzak, W.; Shapiro, B. A. RNAJunction: a 45 46 database of RNA junctions and kissing loops for threedimensional structural analysis and 47 48 nanodesign . Nucleic Acids Research . 2007 , 36(Database) , D392D397. 49 50 51 84. Schroeder, K. T.; McPhee, S. A.; Ouellet, J.; Lilley, D. M. A structural database for kturn motifs 52 53 in RNA . RNA . 2010 , 16(8) , 14638. 54 55 56 57 58 59 60 51 ACS Paragon Plus Environment ACS Combinatorial Science Page 52 of 81

1 2 3 85. Tamura, M.; Hendrix, D. K.; Klosterman, P. S.; Schimmelman, N. R.; Brenner, S. E.; Holbrook, 4 5 6 S. R. SCOR: Structural Classification of RNA, version 2.0 . Nucleic Acids Res . 2004 , 7 8 32(Database issue) , D1824. 9 10 11 86. Popenda, M.; Szachniuk, M.; Blazewicz, M.; Wasik, S.; Burke, E. K.; Blazewicz, J.; Adamiak, 12 13 R. W. RNA FRABASE 2.0: an advanced webaccessible database with the capacity to search the 14 15 threedimensional fragments within RNA structures . BMC Bioinformatics . 2010 , 11 , 231. 16 17 18 87. Petrov, A. I.; Zirbel, C. L.; Leontis, N. B. Automated classification of RNA 3D motifs and the 19 20 RNA 3D Motif Atlas . RNA . 2013 , 19(10) , 132740. 21 22 88. Andronescu, M.; Bereg, V.; Hoos, H. H.; Condon, A. RNA STRAND: the RNA secondary 23 24 25 structure and statistical analysis database . BMC Bioinformatics . 2008 , 9, 340. 26 27 89. Vanegas, P. L.; Hudson, G. A.; Davis, A. R.; Kelly, S. C.; Kirkpatrick, C. C.; Znosko, B. M. 28 29 RNA CoSSMos: Characterization of Secondary Structure Motifsa searchable database of 30 31 32 secondary structure motifs in RNA threedimensional structures . Nucleic Acids Res . 2012 , 33 34 40(Database issue) , D43944. 35 36 37 90. Parlea, L.; Bindewald, E.; Sharan, R.; Bartlett, N.; Moriarty, D.; Oliver, J.; Afonin, K. A.; 38 39 Shapiro, B. A. Ring Catalog: A resource for designing selfassembling RNA nanostructures . 40 41 Methods . 2016 , 103 , 12837. 42 43 44 91. Grabow, W. W.; Zakrevsky, P.; Afonin, K. A.; Chworos, A.; Shapiro, B. A.; Jaeger, L. Self 45 46 Assembling RNA Nanorings Based on RNAI/II Inverse Kissing Complexes . Nano Letters . 2011 , 47 48 11(2) , 878887. 49 50 51 92. Lin, C.; Rinker, S.; Wang, X.; Liu, Y.; Seeman, N. C.; Yan, H. In vivo of artificial DNA 52 53 nanostructures . Proc Natl Acad Sci U S A . 2008 , 105(46) , 1762631. 54 55 56 57 58 59 60 52 ACS Paragon Plus Environment Page 53 of 81 ACS Combinatorial Science

1 2 3 93. Guo, P.; Zhang, C.; Chen, C.; Garver, K.; Trottier, M. InterRNA Interaction of Phage φ29 4 5 6 pRNA to Form a Hexameric Complex for Viral DNA Transportation . Molecular Cell . 1998 , 7 8 2(1) , 149155. 9 10 11 94. Hoeprich, S.; Guo, P. Computer modeling of threedimensional structure of DNApackaging 12 13 RNA (pRNA) monomer, dimer, and hexamer of Phi29 DNA packaging motor . J Biol Chem . 14 15 2002 , 277(23) , 20794803. 16 17 18 95. Shu, Y.; Pi, F.; Sharma, A.; Rajabi, M.; Haque, F.; Shu, D.; Leggas, M.; Evers, B. M.; Guo, P. 19 20 Stable RNA nanoparticles as potential new generation drugs for cancer therapy . Adv Drug Deliv 21 22 Rev . 2014 , 66 , 7489. 23 24 25 96. Shu, Y.; Haque, F.; Shu, D.; Li, W.; Zhu, Z.; Kotb, M.; Lyubchenko, Y.; Guo, P. Fabrication of 26 27 14 different RNA nanoparticles for specific tumor targeting without accumulation in normal 28 29 organs . RNA . 2013 , 19(6) , 767777. 30 31 32 97. Shu, D.; Shu, Y.; Haque, F.; Abdelmawla, S.; Guo, P. Thermodynamically stable RNA three 33 34 way junction for constructing multifunctional nanoparticles for delivery of therapeutics . Nat 35 36 37 Nanotechnol . 2011 , 6(10) , 65867. 38 39 98. Haque, F.; Shu, D.; Shu, Y.; Shlyakhtenko, L. S.; Rychahou, P. G.; Evers, B. M.; Guo, P. 40 41 Ultrastable synergistic tetravalent RNA nanoparticles for targeting to cancers . Nano Today . 42 43 44 2012 , 7(4) , 245257. 45 46 99. Shu, Y.; Shu, D.; Haque, F.; Guo, P. Fabrication of pRNA nanoparticles to deliver therapeutic 47 48 RNAs and bioactive compounds into tumor cells . Nat Protoc . 2013 , 8(9) , 163559. 49 50 51 100. Bindewald, E.; Grunewald, C.; Boyle, B.; O’Connor, M.; Shapiro, B. A. Computational 52 53 strategies for the automated design of RNA nanoscale structures from building blocks using 54 55 NanoTiler . Journal of Molecular Graphics and Modelling . 2008 , 27(3) , 299308. 56 57 58 59 60 53 ACS Paragon Plus Environment ACS Combinatorial Science Page 54 of 81

1 2 3 101. Martinez, H. M.; Maizel, J. V.; Shapiro, B. A. RNA2D3D: A program for Generating, Viewing, 4 5 6 and Comparing 3Dimensional Models of RNA. Journal of Biomolecular Structure and 7 8 Dynamics . 2008 , 25(6) , 669683. 9 10 11 102. Lee, A. J.; Crothers, D. M. The solution structure of an RNA loop–loop complex: the ColE1 12 13 inverted loop sequence . Structure . 1998 , 6(8) , 9931007. 14 15 103. Chworos, A.; Severcan, I.; Koyfman, A. Y.; Weinkam, P.; Oroudjev, E.; Hansma, H. G.; Jaeger, 16 17 18 L. Building programmable jigsaw puzzles with RNA . Science . 2004 , 306(5704) , 206872. 19 20 104. Severcan, I.; Geary, C.; Chworos, A.; Voss, N.; Jacovetty, E.; Jaeger, L. A polyhedron made of 21 22 tRNAs . Nature Chemistry . 2010 , 2(9) , 772779. 23 24 25 105. Severcan, I.; Geary, C.; Verzemnieks, E.; Chworos, A.; Jaeger, L. SquareShaped RNA Particles 26 27 from Different RNA Folds . Nano Letters . 2009 , 9(3) , 12701277. 28 29 106. Afonin, K. A.; Kasprzak, W.; Bindewald, E.; Puppala, P. S.; Diehl, A. R.; Hall, K. T.; Kim, T. J.; 30 31 32 Zimmermann, M. T.; Jernigan, R. L.; Jaeger, L.; Shapiro, B. A. Computational and experimental 33 34 characterization of RNA cubic nanoscaffolds . Methods . 2014 , 67(2) , 256265. 35 36 37 107. Case, D. A.; Berryman, J. T.; Betz, R. M.; Cerutti, D. S.; Cheatham, I., T.E.; Darden, T. A.; 38 39 Duke, R. E.; Giese, T. J.; Gohlke, H.; Goetz, A. W.; Homeyer, N.; Izadi, S.; Janowski, P.; Kaus, 40 41 J.; Kovalenko, A.; Lee, T. S.; LeGrand, S.; Li, P.; Luchko, T.; Luo, R.; Madej, B.; Merz, K. M.; 42 43 44 Monard, G.; Needham, P.; Nguyen, H.; Nguyen, H. T.; Omelyan, I.; Onufriev, A.; Roe, D. R.; 45 46 Roitberg, A.; SalomonFerrer, R.; Simmerling, C. L.; Smith, W.; Swails, J.; Walker, R. C.; 47 48 Wang, J.; Wolf, R. M.; Wu, X.; York, D. M.; Kollman, P. A. AMBER 2015 . 2015 , University of 49 50 51 California: San Francisco. 52 53 108. Kasprzak, W.; Bindewald, E.; Kim, T.J.; Jaeger, L.; Shapiro, B. A. Use of RNA structure 54 55 flexibility data in nanostructure modeling . Methods . 2011 , 54(2) , 239250. 56 57 58 59 60 54 ACS Paragon Plus Environment Page 55 of 81 ACS Combinatorial Science

1 2 3 109. Bahar, I.; Atilgan, A. R.; Erman, B. Direct evaluation of thermal fluctuations in proteins using a 4 5 6 singleparameter harmonic potential . Folding and Design . 1997 , 2(3) , 173181. 7 8 110. Bahar, I.; Erman, B.; Haliloglu, T.; Jernigan, R. L. Efficient Characterization of Collective 9 10 11 Motions and Interresidue Correlations in Proteins by LowResolution Simulations † . 12 13 Biochemistry . 1997 , 36(44) , 1351213523. 14 15 111. Bahar, I.; Rader, A. J. Coarsegrained normal mode analysis in structural biology . Current 16 17 18 Opinion in Structural Biology . 2005 , 15(5) , 586592. 19 20 112. Sherwood, P.; Brooks, B. R.; Sansom, M. S. P. Multiscale methods for macromolecular 21 22 simulations . Current Opinion in Structural Biology . 2008 , 18(5) , 630640. 23 24 25 113. Wang, Y.; Jernigan, R. L. Comparison of tRNA Motions in the Free and Ribosomal Bound 26 27 Structures . Biophysical Journal . 2005 , 89(5) , 33993409. 28 29 114. Wang, Y.; Rader, A. J.; Bahar, I.; Jernigan, R. L. Global ribosome motions revealed with elastic 30 31 32 network model . Journal of Structural Biology . 2004 , 147(3) , 302314. 33 34 115. Yan, A.; Wang, Y.; Kloczkowski, A.; Jernigan, R. L. Effects of Protein Subunits Removal on the 35 36 37 Computed Motions of Partial 30S Structures of the Ribosome . J. Chem. Theory Comput. 2008 , 38 39 4(10) , 17571767. 40 41 116. Yang, L.; Song, G.; Jernigan, R. L. How Well Can We Understand LargeScale Protein Motions 42 43 44 Using Normal Modes of Elastic Network Models? Biophysical Journal . 2007 , 93(3) , 920929. 45 46 117. Zimmermann, M. T.; Jernigan, R. L. Elastic network models capture the motions apparent within 47 48 ensembles of RNA structures . RNA . 2014 , 20(6) , 792804. 49 50 51 118. Afonin, K. A.; Viard, M.; Kagiampakis, I.; Case, C. L.; Dobrovolskaia, M. A.; Hofmann, J.; 52 53 Vrzak, A.; Kireeva, M.; Kasprzak, W. K.; KewalRamani, V. N.; Shapiro, B. A. Triggering of 54 55 56 57 58 59 60 55 ACS Paragon Plus Environment ACS Combinatorial Science Page 56 of 81

1 2 3 RNA Interference with RNA–RNA, RNA–DNA, and DNA–RNA Nanoparticles . ACS Nano . 4 5 6 2015 , 9(1) , 251259. 7 8 119. Afonin, K. A.; Kasprzak, W. K.; Bindewald, E.; Kireeva, M.; Viard, M.; Kashlev, M.; Shapiro, 9 10 11 B. A. In Silico Design and Enzymatic Synthesis of Functional RNA Nanoparticles . Accounts of 12 13 Chemical Research . 2014 , 47(6) , 17311741. 14 15 120. Shukla, G. C.; Haque, F.; Tor, Y.; Wilhelmsson, L. M.; Toulme, J. J.; Isambert, H.; Guo, P.; 16 17 18 Rossi, J. J.; Tenenbaum, S. A.; Shapiro, B. A. A boost for the emerging field of RNA 19 20 nanotechnology . ACS Nano . 2011 , 5(5) , 340518. 21 22 121. Snead, N. M.; Wu, X.; Li, A.; Cui, Q.; Sakurai, K.; Burnett, J. C.; Rossi, J. J. Molecular basis for 23 24 25 improved gene silencing by Dicer substrate interfering RNA compared with other siRNA 26 27 variants . Nucleic Acids Res . 2013 , 41(12) , 620921. 28 29 122. Afonin, K. A.; Viard, M.; Tedbury, P.; Bindewald, E.; Parlea, L.; Howington, M.; Valdman, M.; 30 31 32 JohnsBoehme, A.; Brainerd, C.; Freed, E. O.; Shapiro, B. A. The Use of Minimal RNA 33 34 Toeholds to Trigger the Activation of Multiple Functionalities . Nano Lett . 2016 , 16(3) , 174653. 35 36 37 123. Dao, B. N.; Viard, M.; Martins, A. N.; Kasprzak, W. K.; Shapiro, B. A.; Afonin, K. A. 38 39 Triggering RNAi with multifunctional RNA nanoparticles and their delivery . DNA and RNA 40 41 Nanotechnology . 2015 , 1(1) , 2738. 42 43 44 124. Afonin, K. A.; Danilov, E. O.; Novikova, I. V.; Leontis, N. B. TokenRNA: A New Type of 45 46 SequenceSpecific, LabelFree Fluorescent Biosensor for Folded RNA Molecules . 47 48 ChemBioChem . 2008 , 9(12) , 19021905. 49 50 51 125. Cerqueira, B. B.; Lasham, A.; Shelling, A. N.; AlKassas, R. Nanoparticle therapeutics: 52 53 Technologies and methods for overcoming cancer . Eur J Pharm Biopharm . 2015 , 97(Pt A) , 140 54 55 51. 56 57 58 59 60 56 ACS Paragon Plus Environment Page 57 of 81 ACS Combinatorial Science

1 2 3 126. Phillips, M. A.; Gran, M. L.; Peppas, N. A. Targeted Nanodelivery of Drugs and Diagnostics . 4 5 6 Nano Today . 2010 , 5(2) , 143159. 7 8 127. Lyu, Y.; Chen, G.; Shangguan, D.; Zhang, L.; Wan, S.; Wu, Y.; Zhang, H.; Duan, L.; Liu, C.; 9 10 11 You, M.; Wang, J.; Tan, W. Generating Cell Targeting Aptamers for Nanotheranostics Using 12 13 CellSELEX . Theranostics . 2016 , 6(9) , 144052. 14 15 128. Zhang, H.; Zhou, L.; Zhu, Z.; Yang, C. Recent Progress in AptamerBased Functional Probes for 16 17 18 Bioanalysis and Biomedicine . Chemistry . 2016 , 22(29) , 9886900. 19 20 129. Layzer, J. M.; McCaffrey, A. P.; Tanner, A. K.; Huang, Z.; Kay, M. A.; Sullenger, B. A. In vivo 21 22 activity of nucleaseresistant siRNAs . RNA . 2004 , 10(5) , 76671. 23 24 25 130. Braasch, D. A.; Paroo, Z.; Constantinescu, A.; Ren, G.; Oz, O. K.; Mason, R. P.; Corey, D. R. 26 27 Biodistribution of phosphodiester and phosphorothioate siRNA . Bioorg Med Chem Lett . 2004 , 28 29 14(5) , 113943. 30 31 32 131. Hall, A. H.; Wan, J.; Shaughnessy, E. E.; Ramsay Shaw, B.; Alexander, K. A. RNA interference 33 34 using boranophosphate siRNAs: structureactivity relationships . Nucleic Acids Res . 2004 , 35 36 37 32(20) , 59916000. 38 39 132. Guo, P. The emerging field of RNA nanotechnology . Nature Nanotech . 2010 , 5(12) , 833842. 40 41 133. Shu, Y.; Shu, D.; Haque, F.; Guo, P. Fabrication of pRNA nanoparticles to deliver therapeutic 42 43 44 RNAs and bioactive compounds into tumor cells . Nat Protoc . 2013 , 8(9) , 16351659. 45 46 134. Burmeister, P. E.; Lewis, S. D.; Silva, R. F.; Preiss, J. R.; Horwitz, L. R.; Pendergrast, P. S.; 47 48 McCauley, T. G.; Kurz, J. C.; Epstein, D. M.; Wilson, C.; Keefe, A. D. Direct in vitro selection 49 50 51 of a 2'Omethyl aptamer to VEGF . Chem Biol . 2005 , 12(1) , 2533. 52 53 135. Soutschek, J.; Akinc, A.; Bramlage, B.; Charisse, K.; Constien, R.; Donoghue, M.; Elbashir, S.; 54 55 Geick, A.; Hadwiger, P.; Harborth, J.; John, M.; Kesavan, V.; Lavine, G.; Pandey, R. K.; Racie, 56 57 58 59 60 57 ACS Paragon Plus Environment ACS Combinatorial Science Page 58 of 81

1 2 3 T.; Rajeev, K. G.; Rohl, I.; Toudjarska, I.; Wang, G.; Wuschko, S.; Bumcrot, D.; Koteliansky, 4 5 6 V.; Limmer, S.; Manoharan, M.; Vornlocher, H. P. Therapeutic silencing of an endogenous gene 7 8 by systemic administration of modified siRNAs . Nature . 2004 , 432(7014) , 1738. 9 10 11 136. Elmen, J.; Thonberg, H.; Ljungberg, K.; Frieden, M.; Westergaard, M.; Xu, Y.; Wahren, B.; 12 13 Liang, Z.; Orum, H.; Koch, T.; Wahlestedt, C. Locked nucleic acid (LNA) mediated 14 15 improvements in siRNA stability and functionality . Nucleic Acids Res . 2005 , 33(1) , 43947. 16 17 18 137. Kauppinen, S.; Vester, B.; Wengel, J. Locked nucleic acid (LNA): High affinity targeting of 19 20 RNA for diagnostics and therapeutics . Drug Discov Today Technol . 2005 , 2(3) , 28790. 21 22 138. Amin, M. L.; Joo, J. Y.; Yi, D. K.; An, S. S. A. Surface modification and local orientations of 23 24 25 surface molecules in nanotherapeutics . Journal of Controlled Release . 2015 , 207 , 131142. 26 27 139. Ku, S. H.; Jo, S. D.; Lee, Y. K.; Kim, K.; Kim, S. H. Chemical and structural modifications of 28 29 RNAi therapeutics . Adv Drug Deliv Rev . 2016 , 104 , 1628. 30 31 32 140. Xue, H. Y.; Liu, S.; Wong, H. L. Nanotoxicity: a key obstacle to clinical translation of siRNA 33 34 based nanomedicine . Nanomedicine (Lond) . 2014 , 9(2) , 295312. 35 36 37 141. Stern, S. T.; Hall, J. B.; Yu, L. L.; Wood, L. J.; Paciotti, G. F.; Tamarkin, L.; Long, S. E.; 38 39 McNeil, S. E. Translational considerations for cancer nanomedicine . J Control Release . 2010 , 40 41 146(2) , 16474. 42 43 44 142. Gao, S.; DagnaesHansen, F.; Nielsen, E. J.; Wengel, J.; Besenbacher, F.; Howard, K. A.; Kjems, 45 46 J. The effect of chemical modification and nanoparticle formulation on stability and 47 48 biodistribution of siRNA in mice . Mol Ther . 2009 , 17(7) , 122533. 49 50 51 143. Kanasty, R. L.; Whitehead, K. A.; Vegas, A. J.; Anderson, D. G. Action and reaction: the 52 53 biological response to siRNA and its delivery vehicles . Mol Ther . 2012 , 20(3) , 51324. 54 55 56 57 58 59 60 58 ACS Paragon Plus Environment Page 59 of 81 ACS Combinatorial Science

1 2 3 144. Alexopoulou, L.; Holt, A. C.; Medzhitov, R.; Flavell, R. A. Recognition of doublestranded 4 5 6 RNA and activation of NFkappaB by Tolllike receptor 3 . Nature . 2001 , 413(6857) , 7328. 7 8 145. Pirher, N.; Ivicak, K.; Pohar, J.; Bencina, M.; Jerala, R. A second binding site for double 9 10 11 stranded RNA in TLR3 and consequences for interferon activation . Nat Struct Mol Biol . 2008 , 12 13 15(7) , 7613. 14 15 146. Manche, L.; Green, S. R.; Schmedt, C.; Mathews, M. B. Interactions between doublestranded 16 17 18 RNA regulators and the protein kinase DAI . Mol Cell Biol . 1992 , 12(11) , 523848. 19 20 147. Wang Q; GG, C. Effects of length and location on the cellular response to doublestranded RNA . 21 22 Microbiol Mol Biol Rev. 2004 , 68(3) , 43252. 23 24 25 148. Pichlmair, A.; Schulz, O.; Tan, C. P.; Naslund, T. I.; Liljestrom, P.; Weber, F.; Reis e Sousa, C. 26 27 RIGImediated antiviral responses to singlestranded RNA bearing 5'phosphates . Science . 28 29 2006 , 314(5801) , 9971001. 30 31 32 149. Sioud, M. RNA interference and innate immunity. Adv Drug Deliv Rev . 2007 , 59(2-3) , 15363. 33 34 150. Sioud, M. Singlestranded small interfering RNA are more immunostimulatory than their 35 36 37 doublestranded counterparts: a central role for 2'hydroxyl uridines in immune responses . Eur J 38 39 Immunol . 2006 , 36(5) , 122230. 40 41 151. Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational 42 43 44 approaches to estimate solubility and permeability in drug discovery and development settings . 45 46 Adv Drug Deliv Rev . 2001 , 46(1-3) , 326. 47 48 152. Ezzat, K.; Aoki, Y.; Koo, T.; McClorey, G.; Benner, L.; CoenenStass, A.; O'Donovan, L.; 49 50 51 Lehto, T.; GarciaGuerra, A.; Nordin, J.; Saleh, A. F.; Behlke, M.; Morris, J.; Goyenvalle, A.; 52 53 Dugovic, B.; Leumann, C.; Gordon, S.; Gait, M. J.; ElAndaloussi, S.; Wood, M. J. Self 54 55 56 57 58 59 60 59 ACS Paragon Plus Environment ACS Combinatorial Science Page 60 of 81

1 2 3 Assembly into Nanoparticles Is Essential for Receptor Mediated Uptake of Therapeutic 4 5 6 Antisense Oligonucleotides . Nano Lett . 2015 , 15(7) , 436473. 7 8 153. Kohno, M.; Horibe, T.; Ohara, K.; Ito, S.; Kawakami, K. The MembraneLytic Peptides K8L9 9 10 11 and Melittin Enter Cancer Cells via Receptor Endocytosis following Subcytotoxic Exposure . 12 13 Chemistry & Biology . 2014 , 21(11) , 15221532. 14 15 154. Kondo, E.; Saito, K.; Tashiro, Y.; Kamide, K.; Uno, S.; Furuya, T.; Mashita, M.; Nakajima, K.; 16 17 18 Tsumuraya, T.; Kobayashi, N.; Nishibori, M.; Tanimoto, M.; Matsushita, M. Tumour lineage 19 20 homing cellpenetrating peptides as anticancer molecular delivery systems . Nature 21 22 Communications . 2012 , 3, 951. 23 24 25 155. Pujals, S.; FernándezCarneado, J.; LópezIglesias, C.; Kogan, M. J.; Giralt, E. Mechanistic 26 27 aspects of CPPmediated intracellular drug delivery: Relevance of CPP selfassembly . 28 29 Biochimica et Biophysica Acta (BBA) - Biomembranes . 2006 , 1758(3) , 264279. 30 31 32 156. Rejman, J.; Oberle, V.; Zuhorn, I. S.; Hoekstra, D. Sizedependent internalization of particles via 33 34 the pathways of clathrin and caveolaemediated endocytosis . Biochem J . 2004 , 377(Pt 1) , 159 35 36 37 69. 38 39 157. Zelphati, O.; Szoka, F. C., Jr. Mechanism of oligonucleotide release from cationic liposomes . 40 41 Proc Natl Acad Sci U S A . 1996 , 93(21) , 114938. 42 43 44 158. Boussif, O.; Lezoualc'h, F.; Zanta, M. A.; Mergny, M. D.; Scherman, D.; Demeneix, B.; Behr, J. 45 46 P. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: 47 48 polyethylenimine . Proc Natl Acad Sci U S A . 1995 , 92(16) , 7297301. 49 50 51 159. Wan, Y.; Moyle, P. M.; Toth, I. Endosome Escape Strategies for Improving the Efficacy of 52 53 Oligonucleotide Delivery Systems . Curr Med Chem . 2015 , 22(29) , 332646. 54 55 56 57 58 59 60 60 ACS Paragon Plus Environment Page 61 of 81 ACS Combinatorial Science

1 2 3 160. Shi, B.; Abrams, M. Technologies for investigating the physiological barriers to efficient lipid 4 5 6 nanoparticlesiRNA delivery . J Histochem Cytochem . 2013 , 61(6) , 40720. 7 8 161. Boutla, A.; Delidakis, C.; Livadaras, I.; Tabler, M. Variations of the 3' protruding ends in 9 10 11 synthetic short interfering RNA (siRNA) tested by microinjection in Drosophila embryos . 12 13 Oligonucleotides . 2003 , 13(5) , 295301. 14 15 162. Amanai, M.; Brahmajosyula, M.; Perry, A. C. A restricted role for spermborne microRNAs in 16 17 18 mammalian fertilization . Biol Reprod . 2006 , 75(6) , 87784. 19 20 163. Bartlett, D. W.; Davis, M. E. Effect of siRNA nuclease stability on the in vitro and in vivo 21 22 kinetics of siRNAmediated gene silencing . Biotechnol Bioeng . 2007 , 97(4) , 90921. 23 24 25 164. Golzio, M.; Mazzolini, L.; Ledoux, A.; Paganin, A.; Izard, M.; Hellaudais, L.; Bieth, A.; Pillaire, 26 27 M. J.; Cazaux, C.; Hoffmann, J. S.; Couderc, B.; Teissie, J. In vivo gene silencing in solid tumors 28 29 by targeted electrically mediated siRNA delivery . Gene Ther . 2007 , 14(9) , 7529. 30 31 32 165. Sharei, A.; Cho, N.; Mao, S.; Jackson, E.; Poceviciute, R.; Adamo, A.; Zoldan, J.; Langer, R.; 33 34 Jensen, K. F. Cell squeezing as a robust, microfluidic intracellular delivery platform . J Vis Exp . 35 36 37 2013 (81) , e50980. 38 39 166. Ishida, R.; Kami, D.; Kusaba, T.; Kirita, Y.; Kishida, T.; Mazda, O.; Adachi, T.; Gojo, S. 40 41 Kidneyspecific Sonoporationmediated Gene Transfer. Mol Ther . 2016 , 24(1) , 12534. 42 43 44 167. Tsunoda, S.; Mazda, O.; Oda, Y.; Iida, Y.; Akabame, S.; Kishida, T.; ShinYa, M.; Asada, H.; 45 46 Gojo, S.; Imanishi, J.; Matsubara, H.; Yoshikawa, T. Sonoporation using microbubble BR14 47 48 promotes pDNA/siRNA to murine heart . Biochem Biophys Res Commun . 2005 , 49 50 51 336(1) , 11827. 52 53 54 55 56 57 58 59 60 61 ACS Paragon Plus Environment ACS Combinatorial Science Page 62 of 81

1 2 3 168. Feng, L.; Yang, X.; Shi, X.; Tan, X.; Peng, R.; Wang, J.; Liu, Z. Polyethylene glycol and 4 5 6 polyethylenimine dualfunctionalized nanographene oxide for photothermally enhanced gene 7 8 delivery . Small . 2013 , 9(11) , 198997. 9 10 11 169. Stevenson, D. J.; GunnMoore, F. J.; Campbell, P.; Dholakia, K. Single cell optical transfection . 12 13 J R Soc Interface . 2010 , 7(47) , 86371. 14 15 170. Terakawa, M.; Tanaka, Y. Dielectric microsphere mediated transfection using a femtosecond 16 17 18 laser . Opt Lett . 2011 , 36(15) , 28779. 19 20 171. Behlke, M. A. Progress towards in vivo use of siRNAs . Mol Ther . 2006 , 13(4) , 64470. 21 22 172. Jafari, M.; Soltani, M.; Naahidi, S.; Karunaratne, D. N.; Chen, P. Nonviral approach for targeted 23 24 25 nucleic acid delivery . Curr Med Chem . 2012 , 19(2) , 197208. 26 27 173. Huang, W.; Liu, Z.; Zhou, G.; Ling, J.; Tian, A.; Sun, N. Silencing Bag1 gene via magnetic gold 28 29 nanoparticledelivered siRNA plasmid for colorectal cancer therapy in vivo and in vitro . Tumour 30 31 32 Biol . 2016; DOI: 10.1007/s1327701649260. 33 34 174. Mann, D. G.; McKnight, T. E.; McPherson, J. T.; Hoyt, P. R.; Melechko, A. V.; Simpson, M. L.; 35 36 37 Sayler, G. S. Inducible RNA interferencemediated gene silencing using nanostructured gene 38 39 delivery arrays . ACS Nano . 2008 , 2(1) , 6976. 40 41 175. Chen, Y.; Li, D.; Zhang, Z.; Takushige, N.; Kong, B. H.; Wang, G. Y. Effect of siRNA against 42 43 44 betaNGF on nerve fibers of a rat model with endometriosis . Reprod Sci . 2014 , 21(3) , 32939. 45 46 176. Sikorskaite, S.; Vuorinen, A. L.; Rajamaki, M. L.; Nieminen, A.; Gaba, V.; Valkonen, J. P. 47 48 HandyGun: An improved customdesigned, nonvacuum gene gun suitable for virus inoculation . 49 50 51 J Virol Methods . 2010 , 165(2) , 3204. 52 53 54 55 56 57 58 59 60 62 ACS Paragon Plus Environment Page 63 of 81 ACS Combinatorial Science

1 2 3 177. Maasho, K.; Marusina, A.; Reynolds, N. M.; Coligan, J. E.; Borrego, F. Efficient gene transfer 4 5 6 into the human natural killer cell line, NKL, using the Amaxa nucleofection system . J Immunol 7 8 Methods . 2004 , 284(1-2) , 13340. 9 10 11 178. Mellott, A. J.; Forrest, M. L.; Detamore, M. S. Physical nonviral methods for 12 13 tissue engineering . Ann Biomed Eng . 2013 , 41(3) , 44668. 14 15 179. Zorde Khvalevsky, E.; Gabai, R.; Rachmut, I. H.; Horwitz, E.; Brunschwig, Z.; Orbach, A.; 16 17 18 Shemi, A.; Golan, T.; Domb, A. J.; Yavin, E.; Giladi, H.; Rivkin, L.; Simerzin, A.; Eliakim, R.; 19 20 Khalaileh, A.; Hubert, A.; Lahav, M.; Kopelman, Y.; Goldin, E.; Dancour, A.; Hants, Y.; Arbel 21 22 Alon, S.; Abramovitch, R.; Shemi, A.; Galun, E. Mutant KRAS is a druggable target for 23 24 25 pancreatic cancer . Proc Natl Acad Sci U S A . 2013 , 110(51) , 207238. 26 27 180. Ferber, D. Gene therapy. Safer and virusfree? Science . 2001 , 294(5547) , 163842. 28 29 181. Wells, D. J. Gene therapy progress and prospects: electroporation and other physical methods . 30 31 32 Gene Ther . 2004 , 11(18) , 13639. 33 34 182. Wells, S. A.; Nevins, J. R. Evolving strategies for targeted cancer therapypast, present, and 35 36 37 future . J Natl Cancer Inst . 2004 , 96(13) , 9801. 38 39 183. Liu, J.; Guo, S.; Cinier, M.; Shlyakhtenko, L. S.; Shu, Y.; Chen, C.; Shen, G.; Guo, P. 40 41 Fabrication of stable and RNaseresistant RNA nanoparticles active in gearing the nanomotors 42 43 44 for viral DNA packaging . ACS Nano . 2011 , 5(1) , 23746. 45 46 184. Shu, Y.; Cinier, M.; Fox, S. R.; BenJohnathan, N.; Guo, P. Assembly of therapeutic pRNA 47 48 siRNA nanoparticles using bipartite approach . Mol Ther . 2011 , 19(7) , 130411. 49 50 51 185. Cui, D.; Zhang, C.; Liu, B.; Shu, Y.; Du, T.; Shu, D.; Wang, K.; Dai, F.; Liu, Y.; Li, C.; Pan, F.; 52 53 Yang, Y.; Ni, J.; Li, H.; BrandSaberi, B.; Guo, P. Regression of Gastric Cancer by Systemic 54 55 Injection of RNA Nanoparticles Carrying both Ligand and siRNA . Sci Rep . 2015 , 5, 10726. 56 57 58 59 60 63 ACS Paragon Plus Environment ACS Combinatorial Science Page 64 of 81

1 2 3 186. Binzel, D. W.; Shu, Y.; Li, H.; Sun, M.; Zhang, Q.; Shu, D.; Guo, B.; Guo, P. Specific Delivery 4 5 6 of MiRNA for High Efficient Inhibition of Prostate Cancer by RNA Nanotechnology . Mol Ther . 7 8 2016; DOI: 10.1038/mt.2016.85. 9 10 11 187. Shu, D.; Li, H.; Shu, Y.; Xiong, G.; Carson, W. E., 3rd; Haque, F.; Xu, R.; Guo, P. Systemic 12 13 Delivery of AntimiRNA for Suppression of Triple Negative Breast Cancer Utilizing RNA 14 15 Nanotechnology . ACS Nano . 2015 , 9(10) , 973140. 16 17 18 188. Rychahou, P.; Haque, F.; Shu, Y.; Zaytseva, Y.; Weiss, H. L.; Lee, E. Y.; Mustain, W.; 19 20 Valentino, J.; Guo, P.; Evers, B. M. Delivery of RNA nanoparticles into colorectal cancer 21 22 metastases following systemic administration . ACS Nano . 2015 , 9(2) , 110816. 23 24 25 189. Lee, T. J.; Haque, F.; Shu, D.; Yoo, J. Y.; Li, H.; Yokel, R. A.; Horbinski, C.; Kim, T. H.; Kim, 26 27 S. H.; Kwon, C. H.; Nakano, I.; Kaur, B.; Guo, P.; Croce, C. M. RNA nanoparticle as a vector 28 29 for targeted siRNA delivery into glioblastoma mouse model . Oncotarget . 2015 , 6(17) , 1476676. 30 31 32 190. Feng, L.; Li, S. K.; Liu, H.; Liu, C. Y.; LaSance, K.; Haque, F.; Shu, D.; Guo, P. Ocular delivery 33 34 of pRNA nanoparticles: distribution and clearance after subconjunctival injection . Pharm Res . 35 36 37 2014 , 31(4) , 104658. 38 39 191. Denkbas, E. B. Perspectives on: Chitosan Drug Delivery Systems Based on their Geometries . 40 41 Journal of Bioactive and Compatible Polymers . 2006 , 21(4) , 351368. 42 43 44 192. Kim, J. Y.; Park, K. J.; Kim, G. H.; Jeong, E. A.; Lee, D. Y.; Lee, S. S.; Kim, D. J.; Roh, G. S.; 45 46 Song, J.; Ki, S. H.; Kim, W. H. In vivo activating transcription factor 3 silencing ameliorates the 47 48 AMPK compensatory effects for ER stressmediated betacell dysfunction during the progression 49 50 51 of type2 diabetes . Cell Signal . 2013 , 25(12) , 234861. 52 53 54 55 56 57 58 59 60 64 ACS Paragon Plus Environment Page 65 of 81 ACS Combinatorial Science

1 2 3 193. Dakwar, G. R.; Hammad, I. A.; Popov, M.; Linder, C.; Grinberg, S.; Heldman, E.; Stepensky, D. 4 5 6 Delivery of proteins to the brain by bolaamphiphilic nanosized vesicles . Journal of Controlled 7 8 Release . 2012 , 160(2) , 315321. 9 10 11 194. Smith, M. H.; Lyon, L. A. Multifunctional nanogels for siRNA delivery . Acc Chem Res . 2012 , 12 13 45(7) , 98593. 14 15 195. Nuhn, L.; Gietzen, S.; Mohr, K.; Fischer, K.; Toh, K.; Miyata, K.; Matsumoto, Y.; Kataoka, K.; 16 17 18 Schmidt, M.; Zentel, R. Aggregation Behavior of Cationic Nanohydrogel Particles in Human 19 20 Blood Serum . Biomacromolecules . 2014 , 15(4) , 15261533. 21 22 196. Zorko, M.; Langel, U. Cellpenetrating peptides: mechanism and kinetics of cargo delivery . 23 24 25 Advanced Drug Delivery Reviews . 2005 , 57(4) , 529545. 26 27 197. Adesina, S. K.; Akala, E. O. Nanotechnology Approaches for the Delivery of Exogenous siRNA 28 29 for HIV Therapy . Mol Pharm . 2015 , 12(12) , 417587. 30 31 32 198. Weber, N.; Ortega, P.; Clemente, M. I.; Shcharbin, D.; Bryszewska, M.; de la Mata, F. J.; 33 34 Gomez, R.; MunozFernandez, M. A. Characterization of carbosilane dendrimers as effective 35 36 37 carriers of siRNA to HIVinfected lymphocytes . J Control Release . 2008 , 132(1) , 5564. 38 39 199. Jiang, F.; Liu, B.; Lu, J.; Li, F.; Li, D.; Liang, C.; Dang, L.; Liu, J.; He, B.; Badshah, S. A.; Lu, 40 41 C.; He, X.; Guo, B.; Zhang, X. B.; Tan, W.; Lu, A.; Zhang, G. Progress and Challenges in 42 43 44 Developing AptamerFunctionalized Targeted Drug Delivery Systems . Int J Mol Sci . 2015 , 45 46 16(10) , 23784822. 47 48 200. Jarosch, F.; Buchner, K.; Klussmann, S. In vitro selection using a dual RNA library that allows 49 50 51 primerless selection . Nucleic Acids Res . 2006 , 34(12) , e86. 52 53 201. Cibiel, A.; Dupont, D. M.; Ducongé, F. Methods To Identify Aptamers against Cell Surface 54 55 Biomarkers . Pharmaceuticals . 2011 , 4(9) , 12161235. 56 57 58 59 60 65 ACS Paragon Plus Environment ACS Combinatorial Science Page 66 of 81

1 2 3 202. Ohuchi, S. CellSELEX Technology . BioResearch Open Access . 2012 , 1(6) , 265272. 4 5 6 203. Zimbres, F.; #xe1; M., v.; #xe1; rnok, A.; Ulrich, H.; Wrenger, C. Aptamers: Novel Molecules as 7 8 Diagnostic Markers in Bacterial and Viral Infections? BioMed Research International . 2013 , 9 10 11 2013 , 7. 12 13 204. Liang, H.; Zhang, X. B.; Lv, Y.; Gong, L.; Wang, R.; Zhu, X.; Yang, R.; Tan, W. Functional 14 15 DNAcontaining nanomaterials: cellular applications in biosensing, imaging, and targeted 16 17 18 therapy . Acc Chem Res . 2014 , 47(6) , 1891901. 19 20 205. Zhu, G.; Niu, G.; Chen, X. AptamerDrug Conjugates . Bioconjug Chem . 2015 , 26(11) , 218697. 21 22 206. Aaldering, L. J.; Tayeb, H.; Krishnan, S.; Fletcher, S.; Wilton, S. D.; Veedu, R. N. Smart 23 24 25 functional nucleic acid chimeras: enabling tissue specific RNA targeting therapy . RNA Biol . 26 27 2015 , 12(4) , 41225. 28 29 207. Dassie, J. P.; Giangrande, P. H. Current progress on aptamertargeted oligonucleotide 30 31 32 therapeutics . Ther Deliv . 2013 , 4(12) , 152746. 33 34 208. Takahashi, M.; Burnett, J. C.; Rossi, J. J. AptamersiRNA chimeras for HIV . Adv Exp Med Biol . 35 36 37 2015 , 848 , 21134. 38 39 209. Huang, Y. F.; Shangguan, D.; Liu, H.; Phillips, J. A.; Zhang, X.; Chen, Y.; Tan, W. Molecular 40 41 assembly of an aptamerdrug conjugate for targeted drug delivery to tumor cells . Chembiochem . 42 43 44 2009 , 10(5) , 8628. 45 46 210. Shangguan, D.; Cao, Z.; Meng, L.; Mallikaratchy, P.; Sefah, K.; Wang, H.; Li, Y.; Tan, W. Cell 47 48 Specific Aptamer Probes for Membrane Protein Elucidation in Cancer Cells . Journal of 49 50 51 proteome research . 2008 , 7(5) , 21332139. 52 53 211. Luo, Y. L.; Shiao, Y. S.; Huang, Y. F. Release of photoactivatable drugs from plasmonic 54 55 nanoparticles for targeted cancer therapy . ACS Nano . 2011 , 5(10) , 7796804. 56 57 58 59 60 66 ACS Paragon Plus Environment Page 67 of 81 ACS Combinatorial Science

1 2 3 212. Chu, T. C.; Twu, K. Y.; Ellington, A. D.; Levy, M. Aptamer mediated siRNA delivery . Nucleic 4 5 6 Acids Res . 2006 , 34(10) , e73. 7 8 213. Zhang, P.; Wang, C.; Zhao, J.; Xiao, A.; Shen, Q.; Li, L.; Li, J.; Zhang, J.; Min, Q.; Chen, J.; 9 10 11 Chen, H. Y.; Zhu, J. J. Near InfraredGuided Smart Nanocarriers for MicroRNAControlled 12 13 Release of Doxorubicin/siRNA with Intracellular ATP as Fuel . ACS Nano . 2016 , 10(3) , 363747. 14 15 214. Bates, P. J.; Laber, D. A.; Miller, D. M.; Thomas, S. D.; Trent, J. O. Discovery and development 16 17 18 of the Grich oligonucleotide AS1411 as a novel treatment for cancer . Exp Mol Pathol . 2009 , 19 20 86(3) , 15164. 21 22 215. Lee, J. B.; Hong, J.; Bonner, D. K.; Poon, Z.; Hammond, P. T. Selfassembled RNA interference 23 24 25 microsponges for efficient siRNA delivery . Nat Mater . 2012 , 11(4) , 31622. 26 27 216. Shopsowitz, K. E.; Roh, Y. H.; Deng, Z. J.; Morton, S. W.; Hammond, P. T. RNAi 28 29 microsponges form through selfassembly of the organic and inorganic products of transcription . 30 31 32 Small . 2014 , 10(8) , 162333. 33 34 217. Roh, Y. H.; Deng, J. Z.; Dreaden, E. C.; Park, J. H.; Yun, D. S.; Shopsowitz, K. E.; Hammond, 35 36 37 P. T. A MultiRNAi Microsponge Platform for Simultaneous Controlled Delivery of Multiple 38 39 Small Interfering RNAs . Angew Chem Int Ed Engl . 2016 , 55(10) , 334751. 40 41 218. Grimm, D. The dose can make the poison: lessons learned from adverse in vivo toxicities caused 42 43 44 by RNAi overexpression . Silence . 2011 , 2, 8. 45 46 219. Grimm, D.; Wang, L.; Lee, J. S.; Schurmann, N.; Gu, S.; Borner, K.; Storm, T. A.; Kay, M. A. 47 48 Argonaute proteins are key determinants of RNAi efficacy, toxicity, and persistence in the adult 49 50 51 mouse liver . J Clin Invest . 2010 , 120(9) , 310619. 52 53 54 55 56 57 58 59 60 67 ACS Paragon Plus Environment ACS Combinatorial Science Page 68 of 81

1 2 3 220. Puri, A.; Loomis, K.; Smith, B.; Lee, J.H.; Yavlovich, A.; Heldman, E.; Blumenthal, R. Lipid 4 5 6 Based Nanoparticles as Pharmaceutical Drug Carriers: From Concepts to Clinic . Crit Rev Ther 7 8 Drug Carrier Syst . 2009 , 26(6) , 523580. 9 10 11 221. Kohli, A. G.; Kierstead, P. H.; Venditto, V. J.; Walsh, C. L.; Szoka, F. C. Designer lipids for 12 13 drug delivery: From heads to tails . Journal of Controlled Release . 2014 , 190 , 274287. 14 15 222. Allen, T. M. Drug Delivery Systems: Entering the Mainstream . Science . 2004 , 303(5665) , 1818 16 17 18 1822. 19 20 223. Haran, G.; Cohen, R.; Bar, L. K.; Barenholz, Y. Transmembrane ammonium sulfate gradients in 21 22 liposomes produce efficient and stable entrapment of amphipathic weak bases . Biochimica et 23 24 25 Biophysica Acta (BBA) - Biomembranes . 1993 , 1151(2) , 201215. 26 27 224. Ishida, O.; Maruyama, K.; Sasaki, K.; Iwatsuru, M. Sizedependent extravasation and interstitial 28 29 localization of polyethyleneglycol liposomes in solid tumorbearing mice . International Journal 30 31 32 of Pharmaceutics . 1999 , 190(1) , 4956. 33 34 225. Akbarzadeh, A.; RezaeiSadabady, R.; Davaran, S.; Joo, S. W.; Zarghami, N.; Hanifehpour, Y.; 35 36 37 Samiei, M.; Kouhi, M.; NejatiKoshki, K. Liposome: classification, preparation, and 38 39 applications . Nanoscale Res Lett . 2013 , 8(1) , 102. 40 41 226. Banerjee, K.; Banerjee, S.; Mandal, M. Liposomes as a Drug Delivery System , in Biological and 42 43 44 Pharmaceutical Applications of Nanomaterials. 2015 , Informa UK Limited. p. 53100. 45 46 227. Torchilin, V. P. Multifunctional, stimulisensitive nanoparticulate systems for drug delivery . 47 48 Nature Reviews Drug Discovery . 2014 , 13(11) , 813827. 49 50 51 228. Hassani, Z.; Lemkine, G. F.; Erbacher, P.; Palmier, K.; Alfama, G.; Giovannangeli, C.; Behr, J. 52 53 P.; Demeneix, B. A. Lipidmediated siRNA delivery downregulates exogenous gene expression 54 55 in the mouse brain at picomolar levels . The Journal of Gene Medicine . 2005 , 7(2) , 198207. 56 57 58 59 60 68 ACS Paragon Plus Environment Page 69 of 81 ACS Combinatorial Science

1 2 3 229. Lee, J.M.; Yoon, T.J.; Cho, Y.S. Recent Developments in NanoparticleBased siRNA 4 5 6 Delivery for Cancer Therapy . BioMed Research International . 2013 , 2013 , 110. 7 8 230. Khatri, N.; Baradia, D.; Vhora, I.; Rathi, M.; Misra, A. Development and Characterization of 9 10 11 siRNA Lipoplexes: Effect of Different Lipids, In Vitro Evaluation in Cancerous Cell Lines and 12 13 In Vivo Toxicity Study . AAPS PharmSciTech . 2014 , 15(6) , 16301643. 14 15 231. Ozpolat, B.; Sood, A. K.; LopezBerestein, G. Liposomal siRNA nanocarriers for cancer therapy . 16 17 18 Advanced Drug Delivery Reviews . 2014 , 66 , 110116. 19 20 232. Foged, C. siRNA Delivery with Lipidbased Systems: Promises and Pitfalls . CTMC . 2012 , 12(2) , 21 22 97107. 23 24 25 233. Sparks, J.; Slobodkin, G.; Matar, M.; Congo, R.; Ulkoski, D.; ReaRamsey, A.; Pence, C.; Rice, 26 27 J.; McClure, D.; Polach, K. J.; Brunhoeber, E.; Wilkinson, L.; Wallace, K.; Anwer, K.; Fewell, J. 28 29 G. Versatile cationic lipids for siRNA delivery . Journal of Controlled Release . 2012 , 158(2) , 30 31 32 269276. 33 34 234. Terp, M. C.; Bauer, F.; Sugimoto, Y.; Yu, B.; Brueggemeier, R. W.; Lee, L. J.; Lee, R. J. 35 36 37 Differential efficacy of DOTAP enantiomers for siRNA delivery in vitro . International Journal 38 39 of Pharmaceutics . 2012 , 430(1-2) , 328334. 40 41 235. Crook, K.; Stevenson, B. J.; Dubouchet, M.; Porteous, D. J. Inclusion of cholesterol in DOTAP 42 43 44 transfection complexes increases the delivery of DNA to cells in vitro in the presence of serum . 45 46 Gene Therapy . 1998 , 5(1) , 137143. 47 48 236. Santel, A.; Aleku, M.; Keil, O.; Endruschat, J.; Esche, V.; Fisch, G.; Dames, S.; Loffler, K.; 49 50 51 Fechtner, M.; Arnold, W.; Giese, K.; Klippel, A.; Kaufmann, J. A novel siRNAlipoplex 52 53 technology for RNA interference in the mouse vascular endothelium . Gene Ther . 2006 , 13(16) , 54 55 122234. 56 57 58 59 60 69 ACS Paragon Plus Environment ACS Combinatorial Science Page 70 of 81

1 2 3 237. Landen, C. N. Therapeutic EphA2 Gene Targeting In vivo Using Neutral Liposomal Small 4 5 6 Interfering RNA Delivery . Cancer Research . 2005 , 65(15) , 69106918. 7 8 238. GomesdaSilva, L. C.; Fonseca, N. A.; Moura, V.; Pedroso de Lima, M. C.; Simoes, S.; 9 10 11 Moreira, J. N. Lipidbased nanoparticles for siRNA delivery in cancer therapy: paradigms and 12 13 challenges . Acc Chem Res . 2012 , 45(7) , 116371. 14 15 239. Reichmuth, A. M.; Oberli, M. A.; Jeklenec, A.; Langer, R.; Blankschtein, D. mRNA vaccine 16 17 18 delivery using lipid nanoparticles . Ther Deliv . 2016 , 7(5) , 31934. 19 20 240. Kauffman, K. J.; Dorkin, J. R.; Yang, J. H.; Heartlein, M. W.; DeRosa, F.; Mir, F. F.; Fenton, O. 21 22 S.; Anderson, D. G. Optimization of Lipid Nanoparticle Formulations for mRNA Delivery in 23 24 25 Vivo with Fractional Factorial and Definitive Screening Designs . Nano Lett . 2015 , 15(11) , 7300 26 27 6. 28 29 241. Gupta, K.; Mattingly, S. J.; Knipp, R. J.; Afonin, K. A.; Viard, M.; Bergman, J. T.; Stepler, M.; 30 31 32 Nantz, M. H.; Puri, A.; Shapiro, B. A. Oxime ether lipids containing hydroxylated head groups 33 34 are more superior siRNA delivery agents than their nonhydroxylated counterparts . 35 36 37 Nanomedicine . 2015 , 10(18) , 28052818. 38 39 242. Biswas, S.; Knipp, R. J.; Gordon, L. E.; Nandula, S. R.; Gorr, S. U.; Clark, G. J.; Nantz, M. H. 40 41 Hydrophobic oxime ethers: a versatile class of pDNA and siRNA transfection lipids . 42 43 44 ChemMedChem . 2011 , 6(11) , 20639. 45 46 243. Gupta, K.; Afonin, K. A.; Viard, M.; Herrero, V.; Kasprzak, W.; Kagiampakis, I.; Kim, T.; 47 48 Koyfman, A. Y.; Puri, A.; Stepler, M.; Sappe, A.; KewalRamani, V. N.; Grinberg, S.; Linder, C.; 49 50 51 Heldman, E.; Blumenthal, R.; Shapiro, B. A. Bolaamphiphiles as carriers for siRNA delivery: 52 53 From chemical syntheses to practical applications . Journal of Controlled Release . 2015 , 213 , 54 55 142151. 56 57 58 59 60 70 ACS Paragon Plus Environment Page 71 of 81 ACS Combinatorial Science

1 2 3 244. Fariya, M.; Jain, A.; Dhawan, V.; Shah, S.; Nagarsenker, M. S. Bolaamphiphiles: a 4 5 6 pharmaceutical review . Adv Pharm Bull . 2014 , 4(Suppl 2) , 48391. 7 8 245. Grinberg, S.; Kolot, V.; Linder, C.; Shaubi, E.; Kas’yanov, V.; Deckelbaum, R. J.; Heldman, E. 9 10 11 Synthesis of novel cationic bolaamphiphiles from vernonia oil and their aggregated structures . 12 13 Chemistry and Physics of Lipids . 2008 , 153(2) , 8597. 14 15 246. Grinberg, S.; Linder, C.; Heldman, E. Progress in LipidBased Nanoparticles for Cancer 16 17 18 Therapy . Crit Rev Oncog . 2014 , 19(3-4) , 247260. 19 20 247. Grinberg, S.; Linder, C.; Kolot, V.; Waner, T.; Wiesman, Z.; Shaubi, E.; Heldman, E. Novel 21 22 Cationic Amphiphilic Derivatives from Vernonia Oil: Synthesis and SelfAggregation into 23 24 25 Bilayer Vesicles, Nanoparticles, and DNA Complexants . Langmuir . 2005 , 21(17) , 76387645. 26 27 248. Kim, T.; Afonin, K. A.; Viard, M.; Koyfman, A. Y.; Sparks, S.; Heldman, E.; Grinberg, S.; 28 29 Linder, C.; Blumenthal, R. P.; Shapiro, B. A. In Silico, In Vitro, and In Vivo Studies Indicate the 30 31 32 Potential Use of Bolaamphiphiles for Therapeutic siRNAs Delivery . Molecular Therapy — 33 34 Nucleic Acids . 2013 , 2(3) , e80. 35 36 37 249. Bryniarski, K.; Ptak, W.; Jayakumar, A.; Pullmann, K.; Caplan, M. J.; Chairoungdua, A.; Lu, J.; 38 39 Adams, B. D.; Sikora, E.; Nazimek, K.; Marquez, S.; Kleinstein, S. H.; Sangwung, P.; Iwakiri, 40 41 Y.; Delgato, E.; Redegeld, F.; Blokhuis, B. R.; Wojcikowski, J.; Daniel, A. W.; Groot 42 43 44 Kormelink, T.; Askenase, P. W. Antigenspecific, antibodycoated, exosomelike nanovesicles 45 46 deliver suppressor Tcell microRNA150 to effector T cells to inhibit contact sensitivity . J 47 48 Allergy Clin Immunol . 2013 , 132(1) , 17081. 49 50 51 250. Sliva, K.; Schnierle, B. S. Selective gene silencing by viral delivery of short hairpin RNA . Virol 52 53 J. 2010 , 7, 248. 54 55 56 57 58 59 60 71 ACS Paragon Plus Environment ACS Combinatorial Science Page 72 of 81

1 2 3 4 5 6 7 8 9 10 11 12 13 14 Graphical abstract. 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 72 ACS Paragon Plus Environment Page 73 of 81 ACS Combinatorial Science

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 Figure 1. RNA nanoconstructs and Functionalized RNA Nanoparticles. a. Bacteriophage phi29 46 packaging RNA (pRNA). i.3D computer model of the hexameric pRNA, modeled from PDB ID 1L4O 47 (ref 94) colored by monomeric units; ii. Schematic representations of monomer and handtohand 48 assemblies of dimer and heptamer pRNAs. Monomers can also assemble in handin hand trimers, 49 tetramers, pentamers and hexamers, not represented in this figure; iii. Schematic representations of foot 50 51 tofoot assemblies of dimers and heptamers. Foottofoot assemblies include trimers, tetramers, 52 pentamers and hexamers, not represented here; iv. Threeway junction core, xshape junction core and 53 branchextended trimer and hexamers; Adapted with permission from ref 97; b. RNA Nanoring: i. 3D 54 model of the hexameric nanoring, colored by monomer units; ii. CryoEM map superimposed on the 3D 55 model of the nanoring functionalized with six DS RNAs; iii. Nanoring functionalized with six DS RNAs 56 57 five of which are capped with human epidermal growth factor receptor aptamers (for direct cellular 58 targeting) and one with the red pigment phycoerythrin (for in vivo visualization), reprinted with 59 60 73 ACS Paragon Plus Environment ACS Combinatorial Science Page 74 of 81

1 2 3 permission from ref 14; c. RNA Nanocube: The nancube was in-silico designed to assemble through 4 5 intermolecular interactions between six strands. i. 3D model of the hexameric nancube, colored by 6 monomer units; ii. Nanocube functionalized with six DS RNAs; Adapted with permission from ref 118. 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 74 ACS Paragon Plus Environment Page 75 of 81 ACS Combinatorial Science

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 75 ACS Paragon Plus Environment ACS Combinatorial Science Page 76 of 81

1 2 3 Figure 2. DNA/RNA hybrid nanoconstructs: a. DNA/RNA hybrids: i: Two DNA/RNA hybrids, with 4 5 12 nucleotide toeholds, containing split functionality for a siRNA; ii. Strand exchange between hybrid 6 duplexes is initiated through base pairing of the complementary toeholds; iii. The DNA duplex and the 7 DS RNA resulting from reassociation of DNA/RNA hybrids; Adapted with permission from ref 16; b. 8 RNA Nanoparticles functionalized with RNA/DNA hybrid arms. To restore its functionality, a 9 nanoparticle needs to reassociate with six complementary DNA/RNA hybrids in order to produce 10 11 functional nanoparticle and six DNA duplexes. i. RNA nanoring with six hybrid RNA/DNA arms and 12 Alexa 546 dyes (for FRET studies), reprinted with permission from ref 14; ii. RNA cube with six hybrid 13 RNA/DNA arms, with DNA toeholds, ), adapted with permission from ref 118; iii. DNA cube with 14 DNA/RNA arms, with DNA toeholds, adapted with permission from ref 118. 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 76 ACS Paragon Plus Environment Page 77 of 81 ACS Combinatorial Science

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 Figure 3. Chemical structure of relevant lipids used as RNA delivery agents. a. Single-headed 29 30 lipids: DOTAP, DOTMA, and Oxime Ether Lipid. DOTAP and DOTMA are some of the most 31 commonly used lipids to deliver siRNAs. Both have hydrophilic heads and fatty acyl chains. Oxime 32 either lipids incorporate in their hydrophilic head oxime ether bonds. b. Double-headed lipids: Bola- 33 34 amphiphiles (Bolas). Bolas with two single charge heads are derived from vernonia oil (Bola 19 and 35 Bola 20) and Bolas with four single charge heads are derived from jojoba oil (Bola 58 and Bola 60). 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 77 ACS Paragon Plus Environment ACS Combinatorial Science Page 78 of 81

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Figure 4. Molecular Dynamics (MD) simulation snapshots of complexes formed by 22 23 bolaamphiphiles GLH-58 (green, left panel) and GLH-60 (blue, right panel) with RNA. Bola 24 hydrophobic chains are colored green and blue, and the central Nitrogen atoms in their head groups are 25 26 shown as spheres (two in each GLH58 and four in each bola GLH60). RNA is colored tan with its 27 backbone P atoms shown in purple. MD simulations start with bolas placed randomly around the RNA 28 fragments. At the end of simulations 19 GLH58s form a twolayered micelle, bringing 17 head groups 29 30 within 5Å of the RNA (red spheres), while only 11 GLH60s form a singlelayered micelle around the 31 RNA, with 24 head groups within 5Å of the RNA (red). The predicted RNA exposure to solvent (and 32 potential nucleases) is the same for both, but the electrostatic forces (and hydrogen bonds) are stronger 33 34 in the GL60/RNA complex. These predictions are consistent with the experimental results that showed 35 equal protection against RNA digestion offered by both bolas, but diminished release of siRNA from the 36 GLH60 micelles inside the cell (and less efficient silencing of the target gene). 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 78 ACS Paragon Plus Environment Page 79 of 81 ACS Combinatorial Science

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 Figure 5. Schematic representation of RNA Nanoparticle delivery, uptake and release of 40 functional RNAs. An RNA Nanoparticle, such as the RNA Nanoring chosen here to present the 41 concept, can be encapsulated or complexated with a delivery agent of choice in order to be delivered to 42 43 cells. The presumed cellular uptake mechanism is endocytosis. Once the endosomal escape occurs, the 44 RNA Nanoparticle is exposed to cellular components, such as the Dicer enzyme (colored in purple). 45 Following dicing of the DS RNAs into siRNAs, the guide RNA strand (colored in red) is loaded into the 46 47 RISC (represented schematically by the blue complex) and guided to the mRNA. The mRNA is cleaved 48 in the middle of the guide RNAmRNA complementary paired region, and the protein synthesis is 49 repressed, resulting in downregulation of the protein level of expression. 50 51 52 53 54 55 56 57 58 59 60 79 ACS Paragon Plus Environment ACS Combinatorial Science Page 80 of 81

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 80 ACS Paragon Plus Environment Page 81 of 81 ACS Combinatorial Science

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 Graphical abstract. 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 ACS Paragon Plus Environment