Synthesis of spherical nucleic acids using lipophilic moieties

Information

  • Patent Grant
  • 11696954
  • Patent Number
    11,696,954
  • Date Filed
    Friday, April 27, 2018
    6 years ago
  • Date Issued
    Tuesday, July 11, 2023
    11 months ago
Abstract
Spherical nucleic acids (SNA) carrying self-aggregating oligonucleotides are described herein. Compositions of the SNA include discrete nanostructures that are not aggregated. Related methods are also described.
Description
BACKGROUND OF INVENTION

Spherical Nucleic Acids (SNAs) are a novel class of therapeutic agents that consist of oligonucleotides densely packed and radially oriented around spherical liposomal nanoparticles. SNAs by virtue of their 3-dimensional architecture exhibit the ability to enter cells without the need for auxiliary delivery vehicles or transfection reagents, by engaging scavenger receptors and lipid rafts. Previously, hydrophobic mono-lipophilic moieties, such as cholesterol or tocopherol have been used, and conjugated to oligonucleotides for synthesizing SNAs.


When using single lipophilic moiety, especially cholesterol, oligonucleotides containing G-rich or self-complementary motifs pose a particular challenge. If an oligonucleotide sequence contains self-complementary or G-rich motifs, and is functionalized to the liposome surface using a single lipophilic moiety, such as cholesterol, the resulting SNAs form a cloudy solution, tend to self-aggregate, and eventually precipitate out of solution. These SNA formulations are difficult to filter because the aggregates clog the filter. Bulk synthesis and long term storage are also problematic because the precipitated SNAs may not have the same properties and aggregates may have poor activity or cause unexpected side effects.


SUMMARY OF INVENTION

Some aspects of the present disclosure include a nanostructure comprising a spherical nucleic acid (SNA) comprising a core, a lipid shell having an inner surface surrounding the core and an outer surface with a oligonucleotide functionalized to the outer surface of the nanostructure by a moiety comprised of two or more lipophilic groups. In some embodiments, the core is a hollow or a solid core. In other embodiments, the core is a liposomal core. In some embodiments, the lipid shell is comprised of one type of lipid. In another embodiment, the lipid is a phospholipid. In another embodiment, the phospholipid is 1,2-dioleoyl-sn-glycero-3-phophocholine (DOPC).


In some embodiments, the moiety comprised of two or more lipophilic groups is attached to the oligonucleotide through a linker. In another embodiment, the linker is a hexaethyleneglycol linker. In other embodiments, the oligonucleotide is a single stranded oligonucleotide. In another embodiment, the oligonucleotide is an immunostimulatory oligonucleotide. In another embodiment, the oligonucleotide contains a self-complementary motif. In another embodiment, the oligonucleotide contains a G-rich motif. In some embodiments, the immunostimulatory oligonucleotide stimulates a toll-like receptor (TLR). In another embodiment, the TLR is TLR9. In some embodiments, the oligonucleotide is an antisense oligonucleotide.


In some embodiments, the moiety comprised of the two or more lipophilic groups is a di-alkyl. In another embodiment, the moiety comprised of the two or more lipophilic groups is distearyl. In other embodiments, the moiety comprised of the two or more lipophilic groups is a tri-alkyl. In other embodiments, the moiety comprised of the two or more lipophilic groups is comprised by an alkyl chain. In another embodiment, the alkyl chain is comprised of at least 10 carbons. In another embodiment, the alkyl chain is comprised of at least 14 carbons.


In some embodiments, the nanostructure contains 26 to 7,000 oligonucleotides. In another embodiment, the nanostructure contains 26 to 500 oligonucleotides. In another embodiment, the nanostructure contains 26 to 80 oligonucleotides. In another embodiment, the nanostructure contains at least 40 oligonucleotides. In yet other embodiments, the nanostructure contains 26 to 5,000, 26 to 2,000, 26 to 1,000, 26 to 800, 25 to 500, 26 to 300, 26 to 200, 26 to 100, 50 to 5,000, 50 to 2,000, 50 to 1,000, 50 to 800, 50 to 500, 50 to 300, 50 to 200, 50 to 100, 100 to 5,000, 100 to 2,000, 100 to 1,000, 100 to 800, 100 to 500, 100 to 300, 100 to 200, or 100 to 150 oligonucleotides.


In some embodiments, the nanostructure moiety comprised of two or more lipophilic groups is more stable in solution than a nanostructure with a moiety comprised of one lipophilic group.


In some embodiments, the nanostructure has a diameter of about 10 nm to about 100 nm. In another embodiment, the nanostructure has a diameter of about 20 nm to about 50 nm. In another embodiment, the nanostructure has a diameter of about 27 nm to about 37 nm. In another embodiment, the nanostructure has a diameter of about 27 nm. In another embodiment, the nanostructure has a diameter of about 37 nm.


Some aspects of the present disclosure include a composition of discrete nanostructures, wherein each nanostructure comprises a spherical nucleic acid (SNA) comprising a core, a lipid shell having an inner surface surrounding the core and an outer surface with 26-7,000 oligonucleotides functionalized to the outer surface of the nanostructure, wherein the oligonucleotides contain a self-complementary motif. In some embodiments, each discrete nanostructure has a diameter of about 10 nm to about 100 nm. In another embodiment, each discrete nanostructure has a diameter of about 20 nm to about 50 nm. In yet other embodiments, the nanostructure contains 26 to 5,000, 26 to 2,000, 26 to 1,000, 26 to 800, 25 to 500, 26 to 300, 26 to 200, 26 to 100, 50 to 5,000, 50 to 2,000, 50 to 1,000, 50 to 800, 50 to 500, 50 to 300, 50 to 200, 50 to 100, 100 to 5,000, 100 to 2,000, 100 to 1,000, 100 to 800, 100 to 500, 100 to 300, 100 to 200, or 100 to 150 oligonucleotides.


In some embodiments, the oligonucleotides contain a G-rich motif. In another embodiment, the oligonucleotides are immunostimulatory oligonucleotides. In another embodiment, the immunostimulatory oligonucleotides stimulate a toll-like receptor 9 (TLR9). In other embodiments, the oligonucleotides are antisense oligonucleotides.


In some embodiments, the core is a hollow or a solid core. In some embodiments, the composition has a polydispersity (PDI) of 0.1-0.4. In some embodiments, each discrete nanostructure has a Z average diameter of 30-1,300.


A method for eliciting an immune response is provided according to other aspects of the invention. The method involves contacting a cell with the nanostructure described herein or a composition described herein. In some embodiments, the nanostructure induces cytokine secretion. In another embodiment, the nanostructure activates interferon alpha (IFNα). In some embodiments, the cell is a peripheral blood mononuclear cell.


A method for regulating gene expression is provided according to other aspects of the invention. The method involves contacting a cell with a nanostructure described herein to regulate gene expression.


A method for treating an immune disorder is provided according to other aspects of the invention. The method involves administering to a cell in a subject a nanostructure described herein to deliver an immunostimulatory oligonucleotide that promotes an immune response or to deliver an immunoinhibitory oligonucleotide that decreases or prevents an immune response to treat the immune disorder. In some embodiments, the subject is a mammal. In another embodiment, the subject is a human. In some embodiments, the nanostructure is in contact with the cell at a concentration of 1 nM to 100 μM. In another embodiment, the nanostructure is in contact with the cell at a concentration of 1 μM to 10 μM. In some embodiments, the nanostructure is in contact with the cell for 24 hours.


Kits comprising one or more sealed vials comprising an amount of any of the oligonucleotides and related nanostructure reagents of the present invention are also provided. The kit may optionally include instructions for generating and/or using nanostructures and compositions of the present invention in hard copy or computer readable form.


Each of the limitations of the invention can encompass various embodiments of the invention. It is, therefore, anticipated that each of the limitations of the invention involving any one element or combinations of elements can be included in each aspect of the invention. This invention is not limited in its application to the details of construction and the arrangement of components set forth in the following description or illustrated in the drawings. The invention is capable of other embodiments and of being practiced or of being carried out in various ways. Also, the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of “including,” “comprising,” or “having,” “containing”, “involving”, and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.





BRIEF DESCRIPTION OF DRAWINGS

The accompanying drawings are not intended to be drawn to scale. In the drawings, each identical or nearly identical component that is illustrated in various figures is represented by a like numeral. For purposes of clarity, not every component may be labeled in every drawing. In the drawings:



FIG. 1 agarose gel electrophoresis of SNA constructs and oligonucleotide-lipophilic moiety conjugates. Gel electrophoresis of SNA 1_30 (Lane 1), Oligo 1 (Lane 2), SNA 2 (Lane 3), and Oligo 2 (Lane 4), using 0.5% agarose stained with 0.5 μg/ml ethidium bromide is shown. SNA formation is observed for both SNA 1_30 and SNA 2, which have slower migration than their respective lipophilic moiety-conjugated oligonucleotides.



FIG. 2 depicts agarose gel electrophoresis of SNA constructs and oligonucleotide-lipophilic moiety conjugates. Gel electrophoresis of SNA4 (Lane 1), Oligo 5 (Lane 2), SNA 6 (Lane 3), Oligo 9 (Lane 4), SNA 5(Lane 5), Oligo 6 (Lane 6), SNA 10 (Lane 7), Oligo 14 (Lane 8), SNA 13 (Lane 9), Oligo 18 (Lane 10), SNA 8 (Lane 11), Oligo 12 (Lane 12), SNA 15 (Lane 13), Oligo 21 (Lane 14), SNA 16 (Lane 15), Oligo 22 (Lane 16), SNA 14 (Lane 17), Oligo 20 (Lane 18), Oligo 35 (Lane 19), using 0.5 agarose stained with 0.5 μg/ml ethidium bromide is shown. SNA formation is observed for SNA 10, SNA 13, SNA 8, SNA 15, SNA 16, and SNA 14, which have distinctive migrations compared to their respective constitutive lipophilic moiety-conjugated oligonucleotides.



FIG. 3 shows size exclusion chromatography (SEC) analysis of SNAs functionalized with oligonucleotides with various lipophilic moieties. SNA 1_25, SNA 11, SNA 10, SNA 9, SNA 7, and SNA 3 were analyzed by SEC with PBS running buffer. SNA 3 shows elution of the unbound oligonucleotide only (11-12 minutes) with no SNA peak (6-7 minutes) indicating no self-assembly and formation of SNAs with the corresponding Oligo 3. SNA 1_25, SNA 11, SNA 10, SNA 9, and SNA 7, all elute at 6-7 minutes indicating self-assembly and SNA formation, with no free unbound lipophilic moiety-conjugated oligo.



FIGS. 4A-4B show SEC analyses of oligonucleotide loading on SNA with various lipophilic moieties. In FIG. 4A, SNA 1_10, SNA 1_20, SNA 1_25, SNA 1_30, and SNA 1_50 were analyzed by SEC with PBS running buffer. SNA 1_50 shows elution of the unbound oligonucleotide (11-12 minutes) with SNA peak (6-7 minutes) indicating unbound oligonucleotide when SNAs are loaded with over 25 oligos per nanoparticle with Oligo 1. In FIG. 4B, SNA 18_50, SNA 18_60, SNA 18_70, SNA 18_80, SNA 18_90, and SNA 18_100 were analyzed by SEC with PBS running buffer. SNA 18_70 shows elution of the unbound oligonucleotide (9 minutes) with SNA peak (6-7 minutes) indicating unbound oligonucleotide when SNAs are loaded at over 60 oligos per nanoparticle with Oligo 32.





DETAILED DESCRIPTION

Spherical nucleic acids (SNAs) consist of densely packed, radially oriented nucleic acids. This architecture gives them unique properties, enabling cellular uptake of SNAs mediated via scavenger receptors. Cellular uptake of SNAs is fast and efficient and leads to endosomal accumulation.


Spherical nucleic acids (SNAs) are formed by organizing nucleic acids radially around a core. These structures exhibit the ability to enter cells without the need for auxiliary delivery vehicles or transfection reagents by engaging class A scavenger receptors (SR-A) and lipid rafts Once inside the cell, the nucleic acid components of traditional SNAs resist nuclease degradation, leading to longer intracellular lifetimes. Moreover, SNAs, due to their multi-functional chemical structures, have the ability to bind their targets in a multivalent fashion.


It has been discovered herein that SNA structures can be modified to significantly improve loading density with strategically designed lipophilic groups. SNAs have been developed according to the invention which have a densely packed oligonucleotide shell around a lipid structure. It was found that densely packing the oligonucleotides onto the surface can be achieved using a moiety comprised of two or more lipophilic groups, such as di-stearyl.


It has also been discovered herein that SNA formulation technology can be utilized to deliver self-aggregating oligonucleotides that have heretofore produced unacceptable aggregates that prevented their therapeutic use. SNAs composed of self-aggregating oligonucleotides have been developed according to the invention which incorporate oligonucleotides in a densely packed oligonucleotide shell. These unique molecules can be used to efficiently deliver any type of therapeutic or diagnostic self-aggregating oligonucleotide to a cell, and in particular to endosomes. A liposome or lipoplex can be functionalized into an SNA by inserting lipid-conjugated self-aggregating oligonucleotides onto the external surface. It has been discover that one method for densely packing the self-aggregating oligonucleotides onto the surface can be achieved using a moiety comprised of two or more lipophilic groups.


It is shown herein that when oligonucleotides containing self-complementary, e.g., G-rich motifs, are functionalized to liposome surface using two or more lipophilic groups, such as di-stearyl, the resulting SNAs do not aggregate or precipitate. This is particularly advantageous for large scale clinical and non-clinical preparations where SNAs need to be made in bulk, filter sterilized, and stored over extended periods. It is shown that the presence of multiple lipophilic groups enables higher density of oligonucleotides to be added to the liposome surface. The higher oligonucleotide density imparts stability to these SNAs, which are otherwise unstable, presumably by increasing electrostatic repulsion between SNAs, and promoting intra-SNA oligonucleotide interactions instead of inter-SNA oligonucleotide interactions. The resulting SNAs are active in immunostimulatory assays and can be used for other therapeutic indications such as gene regulation when functionalized with the appropriate antisense sequences.


Self-aggregating oligonucleotides in some embodiments have a self-complementary motif or are G-rich or GC rich. A self-complementary motif may be a region of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more nucleotides that base pair with a region of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more nucleotides in the same oligonucleotide or in other oligonucleotides within the SNAs. In some embodiments nucleotides are consecutive and in other embodiments there are 1 or more intervening nucleotides.


Compositions of SNA of the invention include discrete nanoparticles. The term “discrete” when used in the context of the nanoparticles refers to unaggregated nanoparticles. A composition of discrete nanoparticles includes at least 30% of the nanoparticles in the composition in an aggregated form. In some embodiments a composition of discrete nanoparticles includes at least 40, 45, 50, 55, 60, 65, 70, 75, 80, 85 90, 95, 96, 97, 98, or 99% or in some cases 100% of the nanoparticles in the composition in an unaggregated form.


A moiety comprised of two or more lipophilic groups as used herein is any compound having two lipophilic moieties capable of embedding in a lipid membrane. In some embodiments a moiety comprised of two or more lipophilic groups is a saturated or unsaturated, multi-alkyl chain lipophilic moiety, with carbon chains ranging from, for instance, C10 to C22, or di- and tri-alkyl chain lipophilic moiety. Di- and tri-alkyl chain lipophilic moiety-oligonucleotide conjugates can be synthesized by using symmetrical branching (doubler) and trebler reagents, respectively. As shown in the Examples below, various lipophilic moiety-oligonucleotide conjugates were studied for their ability to form SNAs with 20 nm DOPC liposomes. In contrast to the mono-alkyl lipophilic moiety-oligonucleotide conjugates that did not form SNAs, both di- and tri-alkyl chain lipophilic moiety-oligonucleotide conjugates formed SNAs.


Thus, the nanostructures of the invention are typically composed of lipid nanoparticles having a shell of oligonucleotides, which is formed by arranging oligonucleotides such that they point radially outwards from the core in a densely packed manner. A hydrophobic (e.g. lipophilic moiety) anchor group attached to either the 5′- or 3′-end of the oligonucleotide, depending on whether the oligonucleotides are arranged with the 5′- or 3′-end facing outward from the core preferably is used to embed the oligonucleotides in the lipid nanoparticle. The anchor acts to drive insertion into the lipid nanoparticle and to anchor the oligonucleotides to the lipids.


The density of self-aggregating oligonucleotides on the surface of the SNA of the invention is greater than the density of oligonucleotides positioned on the surface of traditional SNA which have oligonucleotides held on the surface using mono-lipophilic moieties such as cholesterol. Quite surprisingly, the improved density was shown to be associated with less inter-SNA aggregation. Compositions of unaggregated SNA are more stable. The density of the oligonucleotides can be described as a number of oligonucleotides per surface area.


The absolute number of oligonucleotides on the surface of a particle will depend on the size of the particle. For instance, on a 20 nm liposome an ideal number of oligonucleotides on the surface may be about 25-80, 26-80, 25-100, 26-100, 25-60, 26-60, 25-50, 26-50, 30-100, 30-80, 30-70, 30-60, 30-50, 30-40, 40-50, 40-60, or 50-60. Alternatively, on a 100 nm liposome core an ideal number of oligonucleotides on the surface may be about 5,000-6,000, 4,000-6,000, 4,500-6,000, or 5,500-6,000. In other embodiments the surface density of the multi-lipophilic moiety-self-aggregating oligonucleotide-SNAs of the invention is at least 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, or 10% greater than the density of oligonucleotides positioned on the surface of traditional SNA which have oligonucleotides held on the surface using mono-lipophilic moieties such as cholesterol.


A surface density adequate to make the nanoparticles stable and not aggregate and the conditions necessary to obtain it for a desired combination of nanoparticles and oligonucleotides can be determined empirically.


In some aspects the SNAs may be used to deliver a therapeutic oligonucleotide to any tissue in which it is desirable to present the nucleic acid. For instance, it may be desirable to deliver the therapeutic oligonucleotide to the skin, a mucosal membrane, or an internal organ. The stable SNAs described herein are useful for delivering therapeutic oligonucleotides to these tissues for the treatment of disease or for diagnostic purposes.


The invention in some aspects relates to the delivery of an active agent that is a therapeutic nucleic acid. Therapeutic nucleic acids include inhibitory oligonucleotides and oligonucleotides that upregulate expression. In some embodiments the therapeutic nucleic acids specifically downregulate or upregulate the expression of a protein which is useful for being upregulated or downregulated in the eye and in particular in the cornea or retina or other related tissue. In other embodiments the therapeutic nucleic acids specifically downregulate or upregulate the expression of a protein which is useful for being upregulated or downregulated in other tissues. In some embodiments the nucleic acids are selected from the group consisting of a ribozyme, an interfering RNA (RNAi) molecule, a small inhibitory RNA (siRNA) molecule, a triple helix forming molecule, DNA, RNA, plasmids, antisense oligonucleotides, immunostimulatory oligonucleotides, immunoinhibitory oligonucleotides, mRNA, long ncRNA, and miRNA.


The terms “nucleic acid” and “oligonucleotide” are used interchangeably to mean multiple nucleotides (i.e., molecules comprising a sugar (e.g., ribose or deoxyribose) linked to a phosphate group and to an exchangeable organic base, which is either a substituted pyrimidine (e.g., cytosine (C), thymine (T) or uracil (U)) or a substituted purine (e.g., adenine (A) or guanine (G)). As used herein, the terms “nucleic acid” and “oligonucleotide” refer to oligoribonucleotides as well as oligodeoxyribonucleotides. The terms “nucleic acid” and “oligonucleotide” shall also include polynucleosides (i.e., a polynucleotide minus the phosphate) and any other organic base containing polymer. Nucleic acid molecules are preferably synthetic (e.g., produced by nucleic acid synthesis). The oligonucleotides may be any size useful for producing antisense effects. In some embodiments they are 18-23 nucleotides in length. In other embodiments the antisense oligonucleotide is 18 nucleotides in length.


The terms “nucleic acid” and “oligonucleotide” may also encompass nucleic acids or oligonucleotides with substitutions or modifications, such as in the bases and/or sugars. For example, they include nucleic acids having backbone sugars that are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 2′ position and other than a phosphate group or hydroxy group at the 5′ position. Thus modified nucleic acids may include a 2′-O-alkylated ribose group. In addition, modified nucleic acids may include sugars such as arabinose or 2′-fluoroarabinose instead of ribose. Thus the nucleic acids may be heterogeneous in backbone composition thereby containing any possible combination of polymer units linked together such as peptide-nucleic acids (which have an amino acid backbone with nucleic acid bases). Other examples are described in more detail below.


The oligonucleotides may be DNA, RNA, PNA, LNA, ENA or hybrids including any chemical or natural modification thereof. Chemical and natural modifications are well known in the art. Such modifications include, for example, modifications designed to increase binding to a target strand (i.e., increase their melting temperatures), to assist in identification of the oligonucleotide or an oligonucleotide-target complex, to increase cell penetration, to stabilize against nucleases and other enzymes that degrade or interfere with the structure or activity of the oligonucleotides, to provide a mode of disruption (a terminating event) once sequence-specifically bound to a target, and to improve the pharmacokinetic properties of the oligonucleotide.


Modifications include, but are not limited to, for example, (a) end modifications, e.g., 5′ end modifications (phosphorylation dephosphorylation, conjugation, inverted linkages, etc.), 3′ end modifications (conjugation, DNA nucleotides, inverted linkages, etc.), (b) base modifications, e.g., replacement with modified bases, stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners, or conjugated bases, (c) sugar modifications (e.g., at the 2′ position or 4′ position) or replacement of the sugar, as well as (d) internucleoside linkage modifications, including modification or replacement of the phosphodiester linkages. To the extent that such modifications interfere with translation (i.e., results in a reduction of 50%, 60%, 70%, 80%, or 90% or more in translation relative to the lack of the modification—e.g., in an in vitro translation assay), the modification may not be optimal for the methods and compositions described herein.


Non-limiting examples of modified internucleoside linkages include phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3′-5′ to 5′-3′ or 2′-5′ to 5′-2′. Various salts, mixed salts and free acid forms are also included.


Modified internucleoside linkages that do not include a phosphorus atom therein have internucleoside linkages that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.


Substituted sugar moieties include, but are not limited to one of the following at the 2′ position: H (deoxyribose); OH (ribose); F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl can be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl.


A chemically or naturally modified oligonucleotide may include, for example, at least one nucleotide modified at the 2′ position of the sugar, most preferably a 2′-O-alkyl, 2′-O-alkyl-O-alkyl or 2′-fluoro-modified nucleotide or an end cap. In other embodiments, RNA modifications include 2′-fluoro, 2′-amino and 2′ O-methyl modifications on the ribose of pyrimidines, abasic residues or an inverted base at the 3′ end of the RNA.


The oligonucleotides useful according to the invention may include a single modified nucleoside. In other embodiments the oligonucleotide may include at least two modified nucleosides, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20 or more nucleosides, up to the entire length of the oligonucleotide.


Nucleosides or nucleobases include the natural purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleosides include other synthetic and natural nucleobases such as inosine, xanthine, hypoxanthine, nubularine, isoguanisine, tubercidine, 2-(halo)adenine, 2-(alkyl)adenine, 2-(propyl)adenine, 2 (amino)adenine, 2-(aminoalkyll)adenine, 2 (aminopropyl)adenine, 2 (methylthio) N6 (isopentenyl)adenine, 6 (alkyl)adenine, 6 (methyl)adenine, 7 (deaza)adenine, 8 (alkenyl)adenine, 8-(alkyl)adenine, 8 (alkynyl)adenine, 8 (amino)adenine, 8-(halo)adenine, 8-(hydroxyl)adenine, 8 (thioalkyl) adenine, 8-(thiol)adenine, N6-(isopentyl)adenine, N6 (methyl)adenine, N6, N6 (dimethyl)adenine, 2-(alkyl)guanine, 2 (propyl)guanine, 6-(alkyl)guanine, 6 (methyl)guanine, 7 (alkyl)guanine, 7 (methyl)guanine, 7 (deaza)guanine, 8 (alkyl)guanine, 8-(alkenyl)guanine, 8 (alkynyl)guanine, 8-(amino)guanine, 8 (halo)guanine, 8-(hydroxyl)guanine, 8 (thioalkyl)guanine, 8-(thiol)guanine, N (methyl)guanine, 2-(thio)cytosine, 3 (deaza) 5 (aza)cytosine, 3-(alkyl)cytosine, 3 (methyl)cytosine, 5-(alkyl)cytosine, 5-(alkynyl)cytosine, 5 (halo)cytosine, 5 (methyl)cytosine, 5 (propynyl)cytosine, 5 (propynyl)cytosine, 5 (trifluoromethyl)cytosine, 6-(azo)cytosine, N4 (acetyl)cytosine, 3 (3 amino-3 carboxypropyl)uracil, 2-(thio)uracil, 5 (methyl) 2 (thio)uracil, 5 (methylaminomethyl)-2 (thio)uracil, 4-(thio)uracil, 5 (methyl) 4 (thio)uracil, 5 (methylaminomethyl)-4 (thio)uracil, 5 (methyl) 2,4 (dithio)uracil, 5 (methylaminomethyl)-2,4 (dithio)uracil, 5 (2-aminopropyl)uracil, 5-(alkyl)uracil, 5-(alkynyl)uracil, 5-(allylamino)uracil, 5 (aminoallyl)uracil, 5 (aminoalkyl)uracil, 5 (guanidiniumalkyl)uracil, 5 (1,3-diazole-1-alkyl)uracil, 5-(cyanoalkyl)uracil, 5-(dialkylaminoalkyl)uracil, 5 (dimethylaminoalkyl)uracil, 5-(halo)uracil, 5-(methoxy)uracil, uracil-5 oxyacetic acid, 5 (methoxycarbonylmethyl)-2-(thio)uracil, 5 (methoxycarbonyl-methyl)uracil, 5 (propynyl)uracil, 5 (propynyl)uracil, 5 (trifluoromethyl)uracil, 6 (azo)uracil, dihydrouracil, N3 (methyl)uracil, 5-uracil (i.e., pseudouracil), 2 (thio)pseudouracil, 4 (thio)pseudouracil, 2,4-(dithio)psuedouracil, 5-(alkyl)pseudouracil, 5-(methyl)pseudouracil, 5-(alkyl)-2-(thio)pseudouracil, 5-(methyl)-2-(thio)pseudouracil, 5-(alkyl)-4 (thio)pseudouracil, 5-(methyl)-4 (thio)pseudouracil, 5-(alkyl)-2,4 (dithio)pseudouracil, 5-(methyl)-2,4 (dithio)pseudouracil, 1 substituted pseudouracil, 1 substituted 2(thio)-pseudouracil, 1 substituted 4 (thio)pseudouracil, 1 substituted 2,4-(dithio)pseudouracil, 1 (aminocarbonylethylenyl)-pseudouracil, 1 (aminocarbonylethylenyl)-2(thio)-pseudouracil, 1 (aminocarbonylethylenyl)-4 (thio)pseudouracil, 1 aminocarbonylethylenyl)-2,4-(dithio)pseudouracil, 1 (arninoalkylarninocarbonylethylenyl)-pseudouracil, 1 (arninoalkylarnino-carbonylethylenyl)-2(thio)-pseudouracil, 1(arninoalkylarninocarbonylethylenyl)-4 (thio)pseudouracil, 1 (arninoalkylarninocarbonylethylenyl)-2,4-(dithio)pseudouracil, 1,3-(diaza)-2-(oxo)-phenoxazin-1-yl, 1-(aza)-2-(thio)-3-(aza)-phenoxazin-1-yl, 1,3-(diaza)-2-(oxo)-phenthiazin-1-yl, 1-(aza)-2-(thio)-3-(aza)-phenthiazin-1-yl, 7-substituted 1,3-(diaza)-2-(oxo)-phenoxazin-1-yl, 7-substituted 1-(aza)-2-(thio)-3-(aza)-phenoxazin-1-yl, 7-substituted 1,3-(diaza)-2-(oxo)-phenthiazin-1-yl, 7-substituted 1-(aza)-2-(thio)-3-(aza)-phenthiazin-1-yl, 7-(arninoalkylhydroxy)-1,3-(diaza)-2-(oxo)-phenoxazin-1-yl, 7-(arninoalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenoxazin-1-yl, 7-(aminoalkylhydroxy)-1,3-(diaza)-2-(oxo)-phenthiazin-1-yl, 7-(arninoalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenthiazin-1-yl, 7-(guanidiniumalkylhydroxy)-1,3-(diaza)-2-(oxo)-phenoxazin-1-yl, 7-(guanidiniumalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenoxazin-1-yl, 7-(guanidiniumalkyl-hydroxy)-1,3-(diaza)-2-(oxo)-phenthiazin-1-yl, 7-(guanidiniumalkylhydroxy)-1-(aza)-2-(thio)-3-(aza)-phenthiazin-1-yl, 1,3,5-(triaza)-2,6-(dioxa)-naphthalene, inosine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, inosinyl, 2-aza-inosinyl, 7-deaza-inosinyl, nitroimidazolyl, nitropyrazolyl, nitrobenzimidazolyl, nitroindazolyl, aminoindolyl, pyrrolopyrimidinyl, 3-(methyl)isocarbostyrilyl, 5-(methyl)isocarbostyrilyl, 3-(methyl)-7-(propynyl)isocarbostyrilyl, 7-(aza)indolyl, 6-(methyl)-7-(aza)indolyl, imidizopyridinyl, 9-(methyl)-imidizopyridinyl, pyrrolopyrizinyl, isocarbostyrilyl, 7-(propynyl)isocarbostyrilyl, propynyl-7-(aza)indolyl, 2,4,5-(trimethyl)phenyl, 4-(methyl)indolyl, 4,6-(dimethyl)indolyl, phenyl, napthalenyl, anthracenyl, phenanthracenyl, pyrenyl, stilbenyl, tetracenyl, pentacenyl, diiluorotolyl, 4-(iluoro)-6-(methyl)benzimidazole, 4-(methyl)benzimidazole, 6-(azo)thymine, 2-pyridinone, 5 nitroindole, 3 nitropyrrole, 6-(aza)pyrimidine, 2 (amino)purine, 2,6-(diamino) purine, 5 substituted pyrimidines, N2-substituted purines, N6-substituted purines, 06-substituted purines, substituted 1,2,4-triazoles, pyrrolo-pyrimidin-2-on-3-yl, 6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, para-substituted-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, ortho-substituted-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, bis-ortho-substituted-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, para-(aminoalkylhydroxy)-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, ortho-(aminoalkylhydroxy)-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, bis-ortho-(aminoalkylhydroxy)-6-phenyl-pyrrolo-pyrimidin-2-on-3-yl, pyridopyrimidin-3-yl, 2-oxo-7-amino-pyridopyrimidin-3-yl, 2-oxo-pyridopyrimidine-3-yl, or any O-alkylated or N-alkylated derivatives thereof.


The oligonucleotides of the invention may be chimeric oligonucleotides. Chimeric compounds of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleotides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. In particular a gapmer is an oligonucleotide that has at least three discrete portions, two of which are similar i.e. include one or more backbone modifications, and surround a region that is distinct, i.e., does not include backbone modifications.


In some embodiments, the backbone of the oligonucleotide is modified. In some embodiments, the backbone of the oligonucleotide has a phosphorothioate modification. The backbone of the oligonucleotide may have other modifications apparent to one of ordinary skill in the art.


Aspects of the invention relate to delivery of SNAs to a subject for therapeutic and/or diagnostic use. The SNAs may be administered alone or in any appropriate pharmaceutical carrier, such as a liquid, for example saline, or a powder, for administration in vivo. They can also be co-delivered with larger carrier particles or within administration devices. The SNAs may be formulated. The formulations of the invention can be administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients. In some embodiments, SNAs associated with the invention are mixed with a substance such as a lotion (for example, aquaphor) and are administered to the skin of a subject, whereby the SNAs are delivered through the skin of the subject. It should be appreciated that any method of delivery of nanoparticles known in the art may be compatible with aspects of the invention.


For use in therapy, an effective amount of the SNAs can be administered to a subject by any mode that delivers the SNAs to the desired cell. Administering pharmaceutical compositions may be accomplished by any means known to the skilled artisan. Routes of administration include but are not limited to oral, parenteral, intramuscular, intravenous, subcutaneous, mucosal, intranasal, sublingual, intratracheal, inhalation, ocular, vaginal, dermal, rectal, and by direct injection.


In some embodiments the oligonucleotide is a G-rich oligonucleotide. The G-rich nucleic acids may have a sequence that includes at least 50% G's. In some embodiment the G-rich nucleic acids have a sequence that includes at least 60%, 70%, 80% or 90% G's. The G-rich nucleic acids may also have one or multiple G repeats. For instance, a G-rich nucleic acid may have a stretch of at least 4 G's. In other embodiments the G-rich nucleic acid may have one or more stretches of 3 G's. In yet other embodiments the G-rich nucleic acid may have multiple G dimers (e.g., 2, 3, 4, 5, or 6 dimers) separated by one or more other nucleotides.


The oligonuceltide of the SNA in some embodiments is comprised of densely packed, radially oriented nucleic acids which stimulate an immune response, and in particular stimulate the toll-like receptors (TLR) such as TLR9. In some embodiments the SNA is an agonist of a TLR (TLR agonist). A TLR agonist, as used herein is a nucleic acid molecule that interacts with and stimulates the activity of a TLR. The SNA, in some embodiments, is a TLR-9 targeted Immunostimulatory Sperical Nucleic Acid.


Toll-like receptors (TLRs) are a family of highly conserved polypeptides that play a critical role in innate immunity in mammals. At least ten family members, designated TLR1-TLR10, have been identified. The cytoplasmic domains of the various TLRs are characterized by a Toll-interleukin 1 (IL-1) receptor (TIR) domain. Medzhitov R et al. (1998) Mol Cell 2:253-8. Recognition of microbial invasion by TLRs triggers activation of a signaling cascade that is evolutionarily conserved in Drosophila and mammals. The TIR domain-containing adaptor protein MyD88 has been reported to associate with TLRs and to recruit IL-1 receptor-associated kinase (IRAK) and tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) to the TLRs. The MyD88-dependent signaling pathway is believed to lead to activation of NF-κB transcription factors and c-Jun NH2 terminal kinase (Jnk) mitogen-activated protein kinases (MAPKs), critical steps in immune activation and production of inflammatory cytokines. For a review, see Aderem A et al. (2000) Nature 406:782-87.


TLRs are believed to be differentially expressed in various tissues and on various types of immune cells. For example, human TLR7 has been reported to be expressed in placenta, lung, spleen, lymph nodes, tonsil and on plasmacytoid precursor dendritic cells (pDCs). Chuang T-H et al. (2000) Eur Cytokine Netw 11:372-8); Kadowaki N et al. (2001) J Exp Med 194:863-9. Human TLR8 has been reported to be expressed in lung, peripheral blood leukocytes (PBL), placenta, spleen, lymph nodes, and on monocytes. Kadowaki N et al. (2001) J Exp Med 194:863-9; Chuang T-H et al. (2000) Eur Cytokine Netw 11:372-8. Human TLR9 is reportedly expressed in spleen, lymph nodes, bone marrow, PBL, and on pDCs, and B cells. Kadowaki N et al. (2001) J Exp Med 194:863-9; Bauer S et al. (2001) Proc Natl Acad Sci USA 98:9237-42; Chuang T-H et al. (2000) Eur Cytokine Netw 11:372-8.


Nucleotide and amino acid sequences of human and murine TLR9 are known. See, for example, GenBank Accession Nos. NM 017442, AF259262, AB045180, AF245704, AB045181, AF348140, AF314224, NM 031178; and NP 059138, AAF72189, BAB19259, AAF78037, BAB19260, AAK29625, AAK28488, and NP 112455, the contents of all of which are incorporated herein by reference. Human TLR9 is reported to exist in at least two isoforms, one 1032 amino acids long and the other 1055 amino acids. Murine TLR9 is 1032 amino acids long. TLR9 polypeptides include an extracellular domain having a leucine-rich repeat region, a transmembrane domain, and an intracellular domain that includes a TIR domain.


As used herein, the term “TLR9 signaling” refers to any aspect of intracellular signaling associated with signaling through a TLR9. As used herein, the term “TLR9-mediated immune response” refers to the immune response that is associated with TLR9 signaling. A TLR9-mediated immune response is a response associated with TLR9 signaling. This response is further characterized at least by the production/secretion of IFN-γ and IL-12, albeit at levels lower than are achieved via a TLR8-mediated immune response.


The term “TLR9 agonist” refers to any agent that is capable of increasing TLR9 signaling (i.e., an agonist of TLR9). TLR9 agonists specifically include, without limitation, immunostimulatory oligonucleotides, and in particular CpG immunostimulatory oligonucleotides.


An “immunostimulatory oligonucleotide” as used herein is any nucleic acid (DNA or RNA) containing an immunostimulatory motif or backbone that is capable of inducing an immune response. An induction of an immune response refers to any increase in number or activity of an immune cell, or an increase in expression or absolute levels of an immune factor, such as a cytokine. Immune cells include, but are not limited to, NK cells, CD4+T lymphocytes, CD8+T lymphocytes, B cells, dendritic cells, macrophage and other antigen-presenting cells.


As used herein, the term “CpG oligonucleotides,” “immunostimulatory CpG nucleic acids” or “immunostimulatory CpG oligonucleotides” refers to any CpG-containing oligonucleotide that is capable of activating an immune cell. At least the C of the CpG dinucleotide is typically unmethylated. Immunostimulatory CpG oligonucleotides are described in a number of issued patents and published patent applications, including U.S. Pat. Nos. 6,194,388; 6,207,646; 6,218,371; 6,239,116; 6,339,068; 6,406,705; and 6,429,199.


In some embodiments, the CpG oligonucleotides are 4-100 nucleotides in length. In other embodiments, the CpG oligonucleotides are 4-90, 4-80, 4-70, 4-60, 4-50, 4-40, 4-30, 4-20, or 4-10 nucleotides in length.


In some embodiments the immunostimulatory oligonucleotides have a modified backbone such as a phosphorothioate (PS) backbone. In other embodiments the immunostimulatory oligonucleotides have a phosphodiester (PO) backbone. In yet other embodiments immunostimulatory oligonucleotides have a mixed PO and PS backbone. The CpG oligonucleotides may be A-class oligonucleotides, B-class oligonucleotides, or C-class oligonucleotides. “A-class” CpG immunostimulatory oligonucleotides have been described in published PCT application WO 01/22990. These oligonucleotides are characterized by the ability to induce high levels of interferon-alpha while having minimal effects on B cell activation. The A class CpG immunostimulatory nucleic acid may contain a hexamer palindrome GACGTC, AGCGCT, or AACGTT described by Yamamoto and colleagues. Yamamoto S et al. J Immunol 148:4072-6 (1992). Traditional A-class oligonucleotides have poly-G rich 5′ and 3′ ends and a palindromic center region. Typically the nucleotides at the 5′ and 3′ ends have stabilized internucleotide linkages and the center palindromic region has phosphodiester linkages (chimeric).


B class CpG immunostimulatory nucleic acids strongly activate human B cells but have minimal effects inducing interferon-α without further modification. Traditionally, the B-class oligonucleotides include the sequence 5′ TCN1TX1X2CGX3X4 3′, wherein X1 is G or A; X2 is T, G, or A; X3 is T or C and X4 is T or C; and N is any nucleotide, and N1 and N2 are nucleic acid sequences of about 0-25 N's each. B-class CpG oligonucleotides that are typically fully stabilized and include an unmethylated CpG dinucleotide within certain preferred base contexts are potent at activating B cells but are relatively weak in inducing IFN-α and NK cell activation. See, e.g., U.S. Pat. Nos. 6,194,388; 6,207,646; 6,214,806; 6,218,371; 6,239,116; and 6,339,068.


In one embodiment a B class CpG oligonucleotide is represented by at least the formula:

5′X1X2CGX3X43′

wherein X1, X2, X3, and X4 are nucleotides. In one embodiment X2 is adenine, guanine, or thymine. In another embodiment X3 is cytosine, adenine, or thymine.


In another embodiment the invention provides an isolated B class CpG oligonucleotide represented by at least the formula:

5′N1X1X2CGX3X4N23′

wherein X1, X2, X3, and X4 are nucleotides and N is any nucleotide and N1 and N2 are nucleic acid sequences composed of from about 0-25 N's each. In one embodiment X1X2 is a dinucleotide selected from the group consisting of: GpT, GpG, GpA, ApA, ApT, ApG, CpT, CpA, CpG, TpA, TpT, and TpG; and X3X4 is a dinucleotide selected from the group consisting of: TpT, ApT, TpG, ApG, CpG, TpC, ApC, CpC, TpA, ApA, and CpA. Preferably X1X2 is GpA or GpT and X3X4 is TpT. In other embodiments X1 or X2 or both are purines and X3 or X4 or both are pyrimidines or X1X2 is GpA and X3 or X4 or both are pyrimidines. In another preferred embodiment X1X2 is a dinucleotide selected from the group consisting of: TpA, ApA, ApC, ApG, and GpG. In yet another embodiment X3X4 is a dinucleotide selected from the group consisting of: TpT, TpA, TpG, ApA, ApG, GpA, and CpA. X1X2 in another embodiment is a dinucleotide selected from the group consisting of: TpT, TpG, ApT, GpC, CpC, CpT, TpC, GpT and CpG; X3 is a nucleotide selected from the group consisting of A and T and X4 is a nucleotide, but wherein when X1X2 is TpC, GpT, or CpG, X3X4 is not TpC, ApT or ApC.


In another preferred embodiment the CpG oligonucleotide has the sequence 5′ TCN1TX1X2CGX3X4 3′. The CpG oligonucleotides of the invention in some embodiments include X1X2 selected from the group consisting of GpT, GpG, GpA and ApA and X3X4 is selected from the group consisting of TpT, CpT and TpC.


The C class immunostimulatory nucleic acids contain at least two distinct motifs have unique and desirable stimulatory effects on cells of the immune system. Some of these ODN have both a traditional “stimulatory” CpG sequence and a “GC-rich” or “B-cell neutralizing” motif. These combination motif nucleic acids have immune stimulating effects that fall somewhere between those effects associated with traditional “class B” CpG ODN, which are strong inducers of B cell activation and dendritic cell (DC) activation, and those effects associated A-class CpG ODN which are strong inducers of IFN-α and natural killer (NK) cell activation but relatively poor inducers of B-cell and DC activation. Krieg A M et al. (1995) Nature 374:546-9; Ballas Z K et al. (1996) J Immunol 157:1840-5; Yamamoto S et al. (1992) J Immunol 148:4072-6. While preferred class B CpG ODN often have phosphorothioate backbones and preferred class A CpG ODN have mixed or chimeric backbones, the C class of combination motif immune stimulatory nucleic acids may have either stabilized, e.g., phosphorothioate, chimeric, or phosphodiester backbones, and in some preferred embodiments, they have semi-soft backbones.


The stimulatory domain or motif is defined by a formula: 5′ X1DCGHX2 3′. D is a nucleotide other than C. C is cytosine. G is guanine. H is a nucleotide other than G.


X1 and X2 are any nucleic acid sequence 0 to 10 nucleotides long. X1 may include a CG, in which case there is preferably a T immediately preceding this CG. In some embodiments DCG is TCG. X1 is preferably from 0 to 6 nucleotides in length. In some embodiments X2 does not contain any poly G or poly A motifs. In other embodiments the immunostimulatory nucleic acid has a poly-T sequence at the 5′ end or at the 3′ end. As used herein, “poly-A” or “poly-T” shall refer to a stretch of four or more consecutive A's or T's respectively, e.g., 5′ AAAA 3′ or 5′ TTTT 3′.


As used herein, “poly-G end” shall refer to a stretch of four or more consecutive G's, e.g., 5′ GGGG 3′, occurring at the 5′ end or the 3′ end of a nucleic acid. As used herein, “poly-G nucleic acid” shall refer to a nucleic acid having the formula 5′ X1X2GGGX3X4 3′ wherein X1, X2, X3, and X4 are nucleotides and preferably at least one of X3 and X4 is a G.


Some preferred designs for the B cell stimulatory domain under this formula comprise TTTTTCG, TCG, TTCG, TTTCG, TTTTCG, TCGT, TTCGT, TTTCGT, TCGTCGT.


The second motif of the nucleic acid is referred to as either P or N and is positioned immediately 5′ to X1 or immediately 3′ to X2.


N is a B-cell neutralizing sequence that begins with a CGG trinucleotide and is at least 10 nucleotides long. A B-cell neutralizing motif includes at least one CpG sequence in which the CG is preceded by a C or followed by a G (Krieg A M et al. (1998) Proc Natl Acad Sci USA 95:12631-12636) or is a CG containing DNA sequence in which the C of the CG is methylated. As used herein, “CpG” shall refer to a 5′ cytosine (C) followed by a 3′ guanine (G) and linked by a phosphate bond. At least the C of the 5′ CG 3′ must be unmethylated. Neutralizing motifs are motifs which has some degree of immunostimulatory capability when present in an otherwise non-stimulatory motif, but, which when present in the context of other immunostimulatory motifs serve to reduce the immunostimulatory potential of the other motifs.


P is a GC-rich palindrome containing sequence at least 10 nucleotides long. As used herein, “palindrome” and, equivalently, “palindromic sequence” shall refer to an inverted repeat, i.e., a sequence such as ABCDEE′D′C′B′A′ in which A and A′, B and B′, etc., are bases capable of forming the usual Watson-Crick base pairs.


As used herein, “GC-rich palindrome” shall refer to a palindrome having a base composition of at least two-thirds G's and C's. In some embodiments the GC-rich domain is preferably 3′ to the “B cell stimulatory domain”. In the case of a 10-base long GC-rich palindrome, the palindrome thus contains at least 8 G's and C's. In the case of a 12-base long GC-rich palindrome, the palindrome also contains at least 8 G's and C's. In the case of a 14-mer GC-rich palindrome, at least ten bases of the palindrome are G's and C's. In some embodiments the GC-rich palindrome is made up exclusively of G's and C's.


In some embodiments the GC-rich palindrome has a base composition of at least 81% G's and C's. In the case of such a 10-base long GC-rich palindrome, the palindrome thus is made exclusively of G's and C's. In the case of such a 12-base long GC-rich palindrome, it is preferred that at least ten bases (83%) of the palindrome are G's and C's. In some preferred embodiments, a 12-base long GC-rich palindrome is made exclusively of G's and C's. In the case of a 14-mer GC-rich palindrome, at least twelve bases (86%) of the palindrome are G's and C's. In some preferred embodiments, a 14-base long GC-rich palindrome is made exclusively of G's and C's. The C's of a GC-rich palindrome can be unmethylated or they can be methylated.


In general this domain has at least 3 Cs and Gs, more preferably 4 of each, and most preferably 5 or more of each. The number of Cs and Gs in this domain need not be identical. It is preferred that the Cs and Gs are arranged so that they are able to form a self-complementary duplex, or palindrome, such as CCGCGCGG. This may be interrupted by As or Ts, but it is preferred that the self-complementarity is at least partially preserved as for example in the motifs CGACGTTCGTCG (SEQ ID NO: 4) or CGGCGCCGTGCCG (SEQ ID NO: 5). When complementarity is not preserved, it is preferred that the non-complementary base pairs be TG. In a preferred embodiment there are no more than 3 consecutive bases that are not part of the palindrome, preferably no more than 2, and most preferably only 1. In some embodiments the GC-rich palindrome includes at least one CGG trimer, at least one CCG trimer, or at least one CGCG tetramer.


In other embodiments the oligonucleotide is an inhibitory nucleic acid. The oligonucleotide that is an inhibitory nucleic acid may be, for instance, an siRNA or an antisense molecule that inhibits expression of a protein that will have a therapeutic effect. The inhibitory nucleic acids may be designed using routine methods in the art.


An inhibitory nucleic acid typically causes specific gene knockdown, while avoiding off-target effects. Various strategies for gene knockdown known in the art can be used to inhibit gene expression. For example, gene knockdown strategies may be used that make use of RNA interference (RNAi) and/or microRNA (miRNA) pathways including small interfering RNA (siRNA), short hairpin RNA (shRNA), double-stranded RNA (dsRNA), miRNAs, and other small interfering nucleic acid-based molecules known in the art. In one embodiment, vector-based RNAi modalities (e.g., shRNA expression constructs) are used to reduce expression of a gene in a cell. In some embodiments, therapeutic compositions of the invention comprise an isolated plasmid vector (e.g., any isolated plasmid vector known in the art or disclosed herein) that expresses a small interfering nucleic acid such as an shRNA. The isolated plasmid may comprise a specific promoter operably linked to a gene encoding the small interfering nucleic acid. In some cases, the isolated plasmid vector is packaged in a virus capable of infecting the individual. Exemplary viruses include adenovirus, retrovirus, lentivirus, adeno-associated virus, and others that are known in the art and disclosed herein.


A broad range of RNAi-based modalities could be employed to inhibit expression of a gene in a cell, such as siRNA-based oligonucleotides and/or altered siRNA-based oligonucleotides. Altered siRNA based oligonucleotides are those modified to alter potency, target affinity, safety profile and/or stability, for example, to render them resistant or partially resistant to intracellular degradation. Modifications, such as phosphorothioates, for example, can be made to oligonucleotides to increase resistance to nuclease degradation, binding affinity and/or uptake. In addition, hydrophobization and bioconjugation enhances siRNA delivery and targeting


Other molecules that can be used to inhibit expression of a gene include anti sense nucleic acids (single or double stranded), ribozymes, peptides, DNAzymes, peptide nucleic acids (PNAs), triple helix forming oligonucleotides, antibodies, and aptamers and modified form(s) thereof directed to sequences in gene(s), RNA transcripts, or proteins. Antisense and ribozyme suppression strategies have led to the reversal of a tumor phenotype by reducing expression of a gene product or by cleaving a mutant transcript at the site of the mutation. Ribozymes have also been proposed as a means of both inhibiting gene expression of a mutant gene and of correcting the mutant by targeted trans-splicing.


Triple helix approaches have also been investigated for sequence-specific gene suppression. Triple helix forming oligonucleotides have been found in some cases to bind in a sequence-specific manner. Similarly, peptide nucleic acids have been shown to inhibit gene expression. Minor-groove binding polyamides can bind in a sequence-specific manner to DNA targets and hence may represent useful small molecules for suppression at the DNA level.


Other inhibitor molecules that can be used include antisense nucleic acids (single or double stranded). Antisense nucleic acids include modified or unmodified RNA, DNA, or mixed polymer nucleic acids, and primarily function by specifically binding to matching sequences resulting in modulation of peptide synthesis. Antisense nucleic acid binds to target RNA by Watson Crick base-pairing and blocks gene expression by preventing ribosomal translation of the bound sequences either by steric blocking or by activating RNase H enzyme. Antisense molecules may also alter protein synthesis by interfering with RNA processing or transport from the nucleus into the cytoplasm.


As used herein, the term “antisense nucleic acid” describes a nucleic acid that is an oligoribonucleotide, oligodeoxyribonucleotide, modified oligoribonucleotide, or modified oligodeoxyribonucleotide which hybridizes under physiological conditions to DNA comprising a particular gene or to an mRNA transcript of that gene and, thereby, inhibits the transcription of that gene and/or the translation of that mRNA. The antisense molecules are designed so as to interfere with transcription or translation of a target gene upon hybridization with the target gene or transcript. Those skilled in the art will recognize that the exact length of the antisense oligonucleotide and its degree of complementarity with its target will depend upon the specific target selected, including the sequence of the target and the particular bases which comprise that sequence.


An inhibitory nucleic acid useful in the invention will generally be designed to have partial or complete complementarity with one or more target genes. The target gene may be a gene derived from the cell, an endogenous gene, a transgene, or a gene of a pathogen which is present in the cell after infection thereof. Depending on the particular target gene, the nature of the inhibitory nucleic acid and the level of expression of inhibitory nucleic acid (e.g. depending on copy number, promoter strength) the procedure may provide partial or complete loss of function for the target gene. Quantitation of gene expression in a cell may show similar amounts of inhibition at the level of accumulation of target mRNA or translation of target protein. “Inhibition of gene expression” refers to the absence or observable decrease in the level of protein and/or mRNA product from a target gene. “Specificity” refers to the ability to inhibit the target gene without manifest effects on other genes of the cell. The consequences of inhibition can be confirmed by examination of the outward properties of the cell or organism or by biochemical techniques such as RNA solution hybridization, nuclease protection, Northern hybridization, reverse transcription, gene expression monitoring with a microarray, antibody binding, enzyme linked immunosorbent assay (ELISA), Western blotting, radioimmunoassay (RIA), other immunoassays, and fluorescence activated cell analysis (FACS). For RNA-mediated inhibition in a cell line or whole organism, gene expression is conveniently assayed by use of a reporter or drug resistance gene whose protein product is easily assayed. Such reporter genes include acetohydroxyacid synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucoronidase (GUS), chloramphenicol acetyltransferase (CAT), green fluorescent protein (GFP), horseradish peroxidase (HRP), luciferase (Luc), nopaline synthase (NOS), octopine synthase (OCS), and derivatives thereof. Multiple selectable markers are available that confer resistance to ampicillin, bleomycin, chloramphenicol, gentamycin, hygromycin, kanamycin, lincomycin, methotrexate, phosphinothricin, puromycin, and tetracyclin. Depending on the assay, quantitation of the amount of gene expression allows one to determine a degree of inhibition which is greater than 10%, 33%, 50%, 90%, 95% or 99% as compared to a cell not treated according to the present invention. As an example, the efficiency of inhibition may be determined by assessing the amount of gene product in the cell: mRNA may be detected with a hybridization probe having a nucleotide sequence outside the region used for the inhibitory nucleic acid, or translated polypeptide may be detected with an antibody raised against the polypeptide sequence of that region.


An expression enhancing oligonucleotide as used herein is a synthetic oligonucleotide that encodes a protein. The synthetic oligonucleotide may be delivered to a cell such that it is used by a cells machinery to produce a protein based on the sequence of the synthetic oligonucleotide. The synthetic oligonucleotide may be, for instance, synthetic DNA or synthetic RNA. “Synthetic RNA” refers to a RNA produced through an in vitro transcription reaction or through artificial (non-natural) chemical synthesis. In some embodiments, a synthetic RNA is an RNA transcript. In some embodiments, a synthetic RNA encodes a protein. In some embodiments, the synthetic RNA is a functional RNA. In some embodiments, a synthetic RNA comprises one or more modified nucleotides. In some embodiments, a synthetic RNA is up to 0.5 kilobases (kb), 1 kb, 1.5 kb, 2 kb, 2.5 kb, 3 kb, 4 kb, 5 kb, 6 kb, 7 kb, 8 kb, 9 kb, 10 kb, 15 kb, 20 kb, 25 kb, 30 kb or more in length. In some embodiments, a synthetic RNA is in a range of 0.1 kb to 1 kb, 0.5 kb to 2 kb, 0.5 kb to 10 kb, 1 kb to 5 kb, 2 kb to 5 kb, 1 kb to 10 kb, 3 kb to 10 kb, 5 kb to 15 kb, or 1 kb to 30 kb in length.


A diagnostic oligonucleotide is an oligonucleotide that interacts with a cellular marker to identify the presence of the marker in a cell or subject. Diagnostic oligonucleotides are well known in the art and typically include a label or are otherwise detectable.


This invention is not limited in its application to the details of construction and the arrangement of components set forth in the following description or illustrated in the drawings. The invention is capable of other embodiments and of being practiced or of being carried out in various ways. Also, the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of “including,” “comprising,” or “having,” “containing,” “involving,” and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.


Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.


All references, including patent documents, disclosed herein are incorporated by reference in their entirety.


EXAMPLES

Materials and Methods


Synthesis of Oligonucleotide-Lipophilic Moiety Conjugates


Lipophilic moiety conjugated oligonucleotides were synthesized in 5′- to 3′-direction using β-cyanoethyl phosphoramidite chemistry on appropriate solid supports. Syntheses were carried out on Mermade12 DNA/RNA synthesizer. After the synthesis, oligonucleotides were cleaved from the solid support and deprotected by standard protocols using ammonia solution, and purified by RP-HPLC. Oligonucleotide-lipophilic moiety conjugate concentrations were measured using UV absorbance at 260 nm. All the oligonucleotide conjugates synthesized were characterized by MALDI-TOF mass spectrometry for molecular mass.


Liposome Synthesis


Liposomes were synthesized by homogenization of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) hydrated in phosphate buffered saline solution (PBS) (137 mM NaCl, 10 M phosphate, 2.7 mM KCl, pH 7.4, hyclone) using a homogenizer (Avestin). Liposome diameters (˜20 nm) were measured using dynamic light scattering using a Malvern Zetasizer Nano (Malvern Instruments). Lipid concentration was determined using a phospholipid assay kit (Sigma).


SNA Synthesis and Characterization


Oligonucleotide-lipophilic moiety conjugates (see Table 1) were used to synthesize SNAs at various loadings (see Table 2). SNAs were formulated by mixing a molar excess of lipophilic moiety-oligonucleotide conjugate to a liposome in PBS and storing them overnight at 4° C. SNAs were analyzed using 0.5% agarose gel electrophoresis and staining with 0.5 μg/ml ethidium bromide and size-exclusion chromatography (SEC) on a SEC-4000 column (Phenomenex). Light transmission of SNAs was measured at 700 nm using a Cary100Bio UV/VIS Spectrophotometer (Agilent). SNA diameters were measured using dynamic light scattering using a Malvern Zetasizer Nano.









TABLE 1







Oligonucleotide sequences and modifications









Molecular weight











Oligo #
Seq ID No
Sequence and modifications
Calculated
Observed














1
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R1
9143
9143


2
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R2
9086
9084


3
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R4
9035
9058


4
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R7
8804
8782


5
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R8
8719
8704


6
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R9
8746
8740


7
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R10
8774
8782


8
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R11
8733
8713


9
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R12
8756
8742


10
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R3)2
8981
8985


11
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R5)2
9063
9078


12
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R6)2
9119
9123


13
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R7)2
9349
9359


14
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R8)2
9205
9183


15
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R9)2
9261
9250


16
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R10)2
9287
9302


17
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R11)2
9201
9197


18
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R12)2
9297
9272


19
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}Y(R3)3
9419
9419


20
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}Y(R7)3
9966
9992


21
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}Y(R8)3
9755
9742


22
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}Y(R12)3
9894
9866


23
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R1
6234
6235


24
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R8
5814
5832


25
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}AR9
5841
5851


26
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}AR10
5869
5871


27
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R3)2
6049
6083


28
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R5)2
6157
6176


29
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R6)2
6205
6233


30
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R7)2
6411
6469


31
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R8)2
6269
6290


32
2
T*C*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R9)2
6325
9342


33
2
T*G*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R10)2
6381
6413


34
2
T*G*G*T*T*C*G*T*C*G*A*C*G*A*A{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R11)2
6295
6287


35
1
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T
7698
7699


36
3
mUmGmGmGmAmGT*A*G*A*C*A*mAmGmGmUmAmC{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}R1
7485
7487


37
3
mUmGmGmGmAmGT*A*G*A*C*A*mAmGmGmUmAmC{circumflex over ( )}SP18{circumflex over ( )}SP18{circumflex over ( )}X(R9)2
7608
7543









Abbreviations



embedded image


embedded image


Spherical nucleic acid (SNA) using the oligonucleotides described herein were generated and assigned a SNA number (also referred to by a letter designation in the Figures and Table). The composition of each SNA compound is listed below in Table 3, identifying the oligo number and number of oligo's loaded on the surface.









TABLE 2







Spherical nucleic acid (SNA) constructs













Approximate



Letter
Oligo
Oligo Loading


SNA#
designation
sequence #
per Particle













SNA1_10
A
Oligo 1
10


SNA1_20
B
Oligo 1
20


SNA1_25
C
Oligo 1
25


SNA1_30
D
Oligo 1
25


SNA1_50
E
Oligo 1
25


SNA 2
F
Oligo 2
25


SNA 3
G
Oligo 3
0


SNA 4
H
Oligo 5
0


SNA 5
I
Oligo 6
0


SNA 6
J
Oligo 9
0


SNA 7
K
Oligo 11
25


SNA 8
L
Oligo 12
25


SNA 9
M
Oligo 13
25


SNA 10
N
Oligo 14
25


SNA 11
O
Oligo 15
25


SNA 12
P
Oligo 16
25


SNA 13
Q
Oligo 18
25


SNA 14
R
Oligo 20
25


SNA 15
S
Oligo 21
25


SNA 16
T
Oligo 22
25


SNA 17
U
Oligo 23
30


SNA 18_30
V
Oligo 32
30


SNA 18_50
W
Oligo 32
50


SNA 18_60
X
Oligo 32
60


SNA 18_70
Y
Oligo 32
60


SNA 18_80
Z
Oligo 32
60


SNA 18_90
AA
Oligo 32
60


SNA 18_100
AB
Oligo 32
60


SNA 19_30
AC
Oligo 36
25


SNA 20_30
AD
Oligo 37
30


SNA 20_60
AE
Oligo 37
60










Human PBMC Cultures


Fresh human peripheral blood mononuclear cells (PBMCs) from five different donors (Zenbio) were cultured in RPMI supplemented with 10% fetal bovine serum, 2 mM L-glutamine, 50 U/mL penicillin, and 50 mg/mL streptomycin. Cells were maintained at 37° C. in a 5% CO2 humidified incubator. SNAs were applied to PBMCs in 96-well tissue culture plates for 24 hours at concentrations listed in Table 4 and Table 5. After 24 hours of treatment, PBMC culture supernatants were collected for cytokine analysis.









TABLE 3







Characterization of SNAs with various oligonucleotide loading densities













Oligo Loading
Number Mean
Polydispersity
Z-Average
@700 nm %


SNA#
per Particle
Diameter
(PDI)
Diameter
Transmission















SNA17
25
831.5
0.404
1345.0
0.76


SNA18_30
30
544.8
0.221
1260.3
2.58


SNA18_60
60
27.6
0.223
54.2
97.3


SNA19_30
25
2462.7
0.21
2358.3
1.5


SNA20_30
30
26.6
0.42
121.1
41.5


SNA20_60
60
36.6
0.36
104.6
72.8
















TABLE 4





In vitro cytokine secretion in human PBMC cultures







Donor 1













IL-6
IL-12p70
TNFα



Compounda
(pg/ml)
(pg/mL)
(pg/mL)







PBS
28.6
7.4
30.2



SNA 1_30
1128.9
12.2
129.1



SNA 2
558.0
12.1
59.7



SNA 9
875.8
13.1
95.4



SNA 10
1039.7
8.0
182.7



SNA 13
782.7
12.9
55.7



SNA 14
684.4
7.7
175.9



SNA 16
474.2
11.1
71.8


















IL-6
IL-12p70
TNFα
IFNγ



Compounda
(pg/ml)
(pg/mL)
(pg/mL)
(pg/mL)











Donor 2













PBS
21.2
25.5
36.6
138.4



SNA 1_30
997.3
75.9
221.6
245.3



SNA 2
709.5
49.3
123.8
276.6



SNA 9
955.0
69.2
267.4
236.5



SNA 10
1336.8
27.2
411.4
271.3



SNA 13
1395.4
25.6
122.2
268.6



SNA 14
1057.9
25.5
283.5
356.7



SNA 16
1236.5
53.6
318.8
264.1







Donor 3













PBS
5
5
5
7



SNA 1_30
367
249
65
23



SNA 8
538
186
57
45



SNA 9
382
232
57
24



SNA 10
431
237
62
55



SNA 11
503
167
71
69



SNA 12
791
81
167
32








aAt 2.5 μM oligonucleotide concentration














TABLE 5







In vitro cytokine secretion in human


PBMC cultures with SNA 18_60












IL-6
IL-12p40
TNFα
IFNα


Compound
(pg/ml) a
(pg/mL) a
(pg/mL) a
(pg/mL) b










Donor 1











PBS
28.3
6.82
16.69
6.38


SNA 18_60
709.79
33.80
173.14
4642.07







Donor 2











PBS
12.94
12.78
30.2
0.3


SNA 18_60
529.58
176.67
376.0
3885.0






a At 3.33 μM oligonucleotide concentration




b At 0.123 μM oligonucleotide concentration




a At 5.00 μM oligonucleotide concentration




b At 0.312 μM oligonucleotide concentration








Cytokine Induction in Mouse Serum


Female, 6-week old C57BL6 mice were administered 7.5 mg/kg SNA subcutaneously. At 10 hours following SNA administration, serum was collected for cytokine analysis.


Cytokine Analysis Using Q-Plex Array


The cytokine levels in human PBMC culture supernatants and mouse seum were measured using a Q-Plex chemiluminescent arrays (Quansys) following the manufacturer's instructions. The plates were imaged using a Bio-Rad ChemiDoc XRS+ imager and the data were analyzed using the Q-view software (Quansys).


Results


Nature and Number of Lipophilic Moieties Conjugated to Oligonucleotide Determine SNA Assembly


To determine the effect of the lipophilic moiety on SNA formation, SNAs were synthesized using oligonucleotide-lipophilic moiety conjugates listed in Table 1. Agarose gel electrophoresis was used to identify the formation of SNAs. Oligonucleotides conjugated to lipophilic moieties alone were run alongside the SNAs that were self-assembled with the same oligonucleotide. SNA formation results in a characteristic band which migrates between 1000-1500 bases which can be differentiated from the lipophilic moiety-conjugated oligonucleotide. It was observed that oligonucleotide conjugates of mono-alkyl lipophilic moieties did not form SNAs, but di- and tri-alkyl lipophilic moieties (≥C14) formed SNAs on DOPC liposomes (see FIG. 1 and FIG. 2). Further, size-exclusion chromatography (SEC) was used to characterize SNA formation from non-functionalized oligonucleotide-lipophilic moiety conjugates and observed that a mono-alkyl lipid moiety did not form SNA (SNA 3 through SNA 6) as demonstrated by late retention of the oligonucleotide-mono alkyl lipophilic moiety conjugate (11-12 mins). Mono-cholesterol- and di-alkyl lipophilic moiety-oligonucleotide conjugates formed SNAs as demonstrated by elution of the SNAs in the void volume (6-8 mins) (see FIG. 3).


Oligonucleotide-Stearyl Lipophilic Moiety Conjugate Results in Higher SNA Loading


Based on evidence of SNA formation determined using gel electrophoresis (FIGS. 1 and 2) it was sought to determine the loading capacities of oligonucleotide-lipophilic moieties per particle using SEC method. Oligonucleotide conjugates 1, 2, 11-16 and 18-22 with mono-cholesterol, mono-tocopherol, or different alkyl chain lipophilic moieties resulted in up to 25 oligonucleotides per particle (see FIG. 4A; Table 2). In contrast to other alkyl lipophilic moieties, cholesterol, and tocopherol, oligonucleotide-di-stearyl conjugates had the capability of loading up to 60-oligonucleotides per nanoparticle (see FIG. 4B).


Increased Loading Density Allows SNAs without Aggregation for Secondary Structure-Forming Oligonucleotides


The oligonucleotide sequences 23-34 and 36-37 (see Table 1) have a self-complementary or a G-rich nucleotide sequence, respectively. Synthesis of SNAs with these oligonucleotide-cholesterol conjugates (25-oligonucleotides per particle) leads to aggregation of SNA to a suspension as determined by enlarged particle diameters and low light transmission (SNA 17 and SNA 19_30, see Table 3). This aggregation is inherent to SNAs that are synthesized with oligonucleotides that contain a self-complementary or a G-rich sequence that can potentially form secondary structures such as duplexes or G-quadruplexes, respectively, due to inter-particle interactions of oligonucleotides. When the self-complementary oligonucleotide was conjugated with a di-stearyl lipophilic moiety and synthesized SNA at 60-oligonucleotides per nanoparticle, the resulting SNAs did not aggregate as demonstrated by the small 27 nm particle diameter and high light transmission (SNA 18_60, see Table 3). However, loading of only 30-self-complementary oligonucleotide-di-stearyl moiety conjugates on liposomes resulted in SNA aggregation as in the case of cholesterol-oligonucleotide SNAs with enlarged particle diameters and low light transmission (SNA 18_30, see Table 3). Similarly, SNAs synthesized with G-rich oligonucleotide-di-stearyl moiety conjugates also at 60-oligonucleotides per nanoparticle abrogated aggregation significantly as demonstrated by the small 36.6 nm particle diameter and high light transmission (SNA 20_60, see Table 3). SNAs synthesized with a loading of only 30-oligonucleotides per nanoparticle of G-rich oligonucleotide-di-stearyl conjugates showed more aggregation than SNA 20_60, but less than that of cholesterol-oligonucleotide SNA 19_30 as demonstrated by decreased light transmission (SNA 20_30, see Table 3).


Immunostimulatory Activity of SNAs with Various Oligonucleotide-Lipophilic Moiety Conjugates


Immunostimulatory activity of SNAs synthesized with a TLR9 stimulating oligonucleotide with various lipophilic moieties that formed SNAs was also evaluated. Human PBMCs from three different donors were treated with SNAs synthesized to characterize their cytokine release profiles. In general, SNAs exhibited similar cytokine induction profiles characteristic of TLR9 activation regardless of the type of lipophilic moiety conjugated to the oligonucleotide to synthesize the SNA (see Table 4). Self-complementary oligonucleotide sequence in SNA 18_60 is designed to elicit a strong IFNα response. As expected, SNA 18_60 stimulated IFNα induction in PBMCs from two different healthy human donors (see Table 5).


Cytokine induction in mouse serum following SNA administration was used to assess the in vivo immunostimulatory activity of SNAs with a TLR9 stimulating oligonucleotide and two different lipophilic moieties. SNAs with both lipophilic moieties induced similar cytokine profiles in mouse serum (see Table 6).









TABLE 6







In vivo cytokine induction in mouse serum











Compound
IL-6
IL-12p40
MCP-1
RANTES





PBS
128 ± 0 
73 ± 0
63 ± 1
68 ± 1


SNA 1_25
1343 ± 292
159 ± 44
4389 ± 294
 748 ± 118


SNA 2
530 ± 53
270 ± 36
4300 ± 137
579 ± 77





Mean serum cytokine level (pg/mL) ± SEM of n = 4 mice per group






Together, these results demonstrate that multiple lipophilic moieties, including di- or tri-alkyl lipophilic moieties, conjugated to oligonucleotides permit synthesis of SNAs with desired characteristics without significant effect on their biological activity.


EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.


All references, including patent documents, disclosed herein are incorporated by reference in their entirety.

Claims
  • 1. A nanostructure comprising: a spherical nucleic acid (SNA) comprising a core, a lipid shell having an inner surface surrounding the core and an outer surface, and an oligonucleotide linked to a lipophilic moiety wherein the oligonucleotide is functionalized to the lipid shell through interaction between the lipid shell and at least a portion of the lipophilic moiety and the oligonucleotide is oriented radially outwards, wherein the lipophilic moiety is comprised of two or more lipophilic groups and has the following structure: -L1-(Spacer)n-L2-(Spacer)n1-L3-X-(alkyl group)m,wherein L1, L2, and L3 are each linkers and independent of one another are a phosphodiester or phosphorothioate bond or short chain linkage, wherein the spacer is an oligoethylene glycol spacer, wherein n and n1 are independently 0-3, wherein at least one of n and n1 is not 0, wherein X is a doubler or trebler, the alkyl group is a C6-C30 saturated or unsaturated alkyl group and m is 2-3, wherein the doubler and trebler have the following structures, respectively:
  • 2. The nanostructure of claim 1, wherein the oligoethylene glycol spacer is a HEG spacer (hexaethylene glycol).
  • 3. The nanostructure of claim 1, wherein L1, L2, and L3 are phosphodiester linkages.
  • 4. The nanostructure of claim 1, wherein the lipid shell comprises 1,2-dioleoyl-sn-glycero-3-phophocholine (DOPC).
  • 5. The nanostructure of claim 1, wherein the oligonucleotide is a single stranded oligonucleotide and/or an immunostimulatory oligonucleotide.
  • 6. The nanostructure of claim 1, wherein the oligonucleotide is a self-aggregating oligonucleotide containing a self-complementary motif.
  • 7. A composition of the nanostructures of claim 1.
  • 8. The nanostructure of claim 1, wherein the oligonucleotide contains a G-rich motif.
  • 9. The nanostructure of claim 1, wherein the oligonucleotide is an immunostimulatory oligonucleotide and/or an antisense oligonucleotide.
  • 10. The nanostructure of claim 9, wherein the immunostimulatory oligonucleotide stimulates a toll-like receptor 9 (TLR9).
  • 11. The nanostructure of claim 1, wherein the core is a hollow core or a solid core.
  • 12. The composition of claim 7, wherein the composition has a polydispersity index (PDI) of 0.1-0.4.
  • 13. The composition of claim 7, wherein the nanostructures have a Z average diameter of 30-1,300 nm.
RELATED APPLICATIONS

This application is a national stage filing under 35 U.S.C. § 371 of international application number PCT/US2018/030021, filed Apr. 27, 2018, which was published under PCT Article 21(2) in English and claims the benefit under 35 U.S.C. § 119(e) of U.S. provisional application Ser. No. 62/492,062, filed Apr. 28, 2017, each of which is herein incorporated by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/030021 4/27/2018 WO
Publishing Document Publishing Date Country Kind
WO2018/201090 11/1/2018 WO A
US Referenced Citations (289)
Number Name Date Kind
4904582 Tullis Feb 1990 A
5264618 Felgner et al. Nov 1993 A
5472881 Beebe et al. Dec 1995 A
5512439 Hornes et al. Apr 1996 A
5578718 Cook et al. Nov 1996 A
5780448 Davis Jul 1998 A
5869286 Yao et al. Feb 1999 A
5955589 Cook et al. Sep 1999 A
6051698 Janjic et al. Apr 2000 A
6072033 Yao et al. Jun 2000 A
6072037 Yao et al. Jun 2000 A
6096305 Yao et al. Aug 2000 A
6100235 Yao et al. Aug 2000 A
6191104 Spriggs et al. Feb 2001 B1
6194388 Krieg et al. Feb 2001 B1
6197525 Yao et al. Mar 2001 B1
6207646 Krieg et al. Mar 2001 B1
6214806 Krieg et al. Apr 2001 B1
6218371 Krieg et al. Apr 2001 B1
6239116 Krieg et al. May 2001 B1
6271209 Smith et al. Aug 2001 B1
6339068 Krieg et al. Jan 2002 B1
6361944 Mirkin et al. Mar 2002 B1
6406705 Davis et al. Jun 2002 B1
6429199 Krieg et al. Aug 2002 B1
6482923 Shi et al. Nov 2002 B1
6506564 Mirkin et al. Jan 2003 B1
6534062 Raz et al. Mar 2003 B2
6562578 Gorman et al. May 2003 B1
6569419 Moore et al. May 2003 B2
6569645 Chen et al. May 2003 B2
6579520 Chen et al. Jun 2003 B2
6610308 Haensler Aug 2003 B1
6635443 Shi et al. Oct 2003 B1
6849719 Shi et al. Feb 2005 B2
6902735 Jacobs et al. Jun 2005 B1
7094566 Medlock et al. Aug 2006 B2
7094886 Shaughnessy Aug 2006 B2
7115398 Chen et al. Oct 2006 B2
7129222 Van Nest et al. Oct 2006 B2
7217412 Chen et al. May 2007 B2
7223741 Krieg May 2007 B2
7250403 Van Nest et al. Jul 2007 B2
7255868 Fearon et al. Aug 2007 B2
7256264 Goddard et al. Aug 2007 B2
7276489 Agrawal et al. Oct 2007 B2
7332586 Franzen et al. Feb 2008 B2
7404969 Chen et al. Jul 2008 B2
7427405 Agrawal et al. Sep 2008 B2
7470674 Agrawal et al. Dec 2008 B2
7473763 Goddard et al. Jan 2009 B2
7514099 Chen et al. Apr 2009 B2
7544482 Goddard et al. Jun 2009 B2
7563618 Gryaznov et al. Jul 2009 B2
7611857 Medlock et al. Nov 2009 B2
7615539 Uhlmann et al. Nov 2009 B2
7628990 Tuck et al. Dec 2009 B2
7638603 Shi et al. Dec 2009 B2
7655761 Gorman et al. Feb 2010 B2
7666674 Klinman et al. Feb 2010 B2
7696150 Shaughnessy Apr 2010 B2
7713535 Agrawal et al. May 2010 B2
7718397 Goddard et al. May 2010 B2
7771719 Filvaroff et al. Aug 2010 B1
7833992 Vargeese et al. Nov 2010 B2
7879980 Golstein et al. Feb 2011 B2
7943738 Medlock et al. May 2011 B2
7956176 McSwiggen et al. Jun 2011 B2
7964578 Vargeese et al. Jun 2011 B2
7993659 Noelle et al. Aug 2011 B2
8008267 Kandimalla et al. Aug 2011 B2
8058249 Krieg et al. Nov 2011 B2
8088388 Sokoll Jan 2012 B2
8124590 Van Nest et al. Feb 2012 B2
8133734 Shi et al. Mar 2012 B2
8158768 Dina et al. Apr 2012 B2
8188254 Uhlmann et al. May 2012 B2
8202979 McSwiggen et al. Jun 2012 B2
8273866 McSwiggen et al. Sep 2012 B2
8309527 Krieg et al. Nov 2012 B2
8323686 Mirkin et al. Dec 2012 B2
8333980 Van Nest et al. Dec 2012 B2
8338132 Chen et al. Dec 2012 B2
8431544 Agrawal et al. Apr 2013 B1
8455217 Goddard et al. Jun 2013 B2
8470342 Klinman Jun 2013 B2
8871732 Dina et al. Oct 2014 B2
8889181 Kwon Nov 2014 B2
8933046 Machuy et al. Jan 2015 B2
8945590 Fairman et al. Feb 2015 B2
8968746 Baumhof et al. Mar 2015 B2
9066978 Ilyinskii et al. Jun 2015 B2
9139827 Mirkin et al. Sep 2015 B2
9216155 Thaxton et al. Dec 2015 B2
9389236 Fandl et al. Jul 2016 B2
9506056 Mirkin et al. Nov 2016 B2
9532948 Mirkin et al. Jan 2017 B2
9549901 Shi et al. Jan 2017 B2
9617541 Mirkin et al. Apr 2017 B2
9617547 Gemba Apr 2017 B2
9693957 Lin et al. Jul 2017 B2
9764031 Ilyinskii et al. Sep 2017 B2
9844562 Mirkin et al. Dec 2017 B2
9868955 Guiducci et al. Jan 2018 B2
9889209 Mirkin et al. Feb 2018 B2
9901616 Dhar et al. Feb 2018 B2
9907845 Reed et al. Mar 2018 B2
9999673 Rajeev et al. Jun 2018 B2
10029016 Irvine et al. Jul 2018 B2
10078092 Mutharasan et al. Sep 2018 B2
10098958 Mirkin et al. Oct 2018 B2
10144933 Gemba et al. Dec 2018 B2
10182988 Mirkin et al. Jan 2019 B2
10196643 Dina et al. Feb 2019 B2
10208310 Mader et al. Feb 2019 B2
10328026 Thaxton et al. Jun 2019 B2
10370656 Mirkin et al. Aug 2019 B2
10391116 Mirkin et al. Aug 2019 B2
10398784 Mirkin et al. Sep 2019 B2
10413565 Plebanek et al. Sep 2019 B2
10434064 Radovic-Moreno et al. Oct 2019 B2
10517927 Braddock et al. Dec 2019 B2
10568898 Thaxton et al. Feb 2020 B2
10704043 Daniel et al. Jul 2020 B2
10760080 Mader et al. Sep 2020 B2
10792251 Mirkin et al. Oct 2020 B2
10837018 Radovic-Moreno et al. Nov 2020 B2
10894963 Radovic-Moreno et al. Jan 2021 B2
11123294 Radovic-Moreno et al. Sep 2021 B2
20020039568 Moore et al. Apr 2002 A1
20020156033 Bratzler et al. Oct 2002 A1
20020172711 Martin et al. Nov 2002 A1
20020172953 Mirkin et al. Nov 2002 A1
20020197269 Lingnau et al. Dec 2002 A1
20030026782 Krieg Feb 2003 A1
20030050261 Krieg et al. Mar 2003 A1
20030091599 Davis et al. May 2003 A1
20030104044 Semple et al. Jun 2003 A1
20030129251 Van Nest et al. Jul 2003 A1
20030133988 Fearon et al. Jul 2003 A1
20030147966 Franzen et al. Aug 2003 A1
20030170162 Nayfeh et al. Sep 2003 A1
20040014956 Woolf et al. Jan 2004 A1
20040023335 Jing et al. Feb 2004 A1
20040023382 Dean et al. Feb 2004 A1
20040053384 Sligar et al. Mar 2004 A1
20040087534 Krieg et al. May 2004 A1
20040092468 Schwartz May 2004 A1
20040143112 Krieg et al. Jul 2004 A1
20040158051 Ozkan et al. Aug 2004 A1
20040170560 Fossheim et al. Sep 2004 A1
20040171109 Haudenschild et al. Sep 2004 A1
20040234500 Moore et al. Nov 2004 A1
20040247680 Farokhzad et al. Dec 2004 A1
20040266719 McCluskie et al. Dec 2004 A1
20050009773 Kandimalla et al. Jan 2005 A1
20050112578 Matsuura May 2005 A1
20050130911 Uhlmann et al. Jun 2005 A1
20050169888 Hartmann et al. Aug 2005 A1
20050208572 Shaughnessy Sep 2005 A1
20060003962 Ahluwalia et al. Jan 2006 A1
20060014191 Lao et al. Jan 2006 A1
20060014713 Agrawal et al. Jan 2006 A1
20060019916 Krieg et al. Jan 2006 A1
20060073572 Huang et al. Apr 2006 A1
20060083713 Glasebrook et al. Apr 2006 A1
20060083781 Shastri et al. Apr 2006 A1
20060100151 Troutt May 2006 A1
20060251623 Bachmann et al. Nov 2006 A1
20060292174 de los Rios et al. Dec 2006 A1
20070065868 Jing Mar 2007 A1
20070066554 Krieg et al. Mar 2007 A1
20070184068 Renner et al. Aug 2007 A1
20070243196 Bruck et al. Oct 2007 A1
20070280936 Moore et al. Dec 2007 A1
20080181928 Hakimi-Mehr et al. Jul 2008 A1
20080194463 Weller et al. Aug 2008 A1
20080206265 Kandimalla et al. Aug 2008 A1
20080213177 Rademacher et al. Sep 2008 A1
20080274454 Mirkin et al. Nov 2008 A1
20080306016 Mirkin et al. Dec 2008 A1
20080311182 Ferrari et al. Dec 2008 A1
20090018028 Lindsay et al. Jan 2009 A1
20090053169 Castillo et al. Feb 2009 A1
20090068737 Yao et al. Mar 2009 A1
20090075884 Jacobs et al. Mar 2009 A1
20090081157 Kornbluth et al. Mar 2009 A1
20090191188 Krieg et al. Jul 2009 A1
20090209629 Mirkin et al. Aug 2009 A1
20090275735 Golstein et al. Nov 2009 A1
20090286853 Gryaznov et al. Nov 2009 A1
20090299045 Richards et al. Dec 2009 A1
20090317802 Bhatia et al. Dec 2009 A1
20090322327 Gao Dec 2009 A1
20090324706 Mirkin et al. Dec 2009 A1
20100003287 Mills et al. Jan 2010 A1
20100136682 Mirkin et al. Jun 2010 A1
20100143246 Shi et al. Jun 2010 A1
20100144848 Vogel et al. Jun 2010 A1
20100166842 Lu et al. Jul 2010 A1
20100183634 Luo et al. Jul 2010 A1
20100184844 Mirkin et al. Jul 2010 A1
20100233270 Mirkin et al. Sep 2010 A1
20100278840 Mohler Nov 2010 A1
20110052697 Farokhzad et al. Mar 2011 A1
20110111974 Mirkin et al. May 2011 A1
20110201672 Krieg et al. Aug 2011 A1
20110293700 Bratzler et al. Dec 2011 A1
20110293701 Bratzler et al. Dec 2011 A1
20110293723 Bratzler et al. Dec 2011 A1
20110305734 Edelson et al. Dec 2011 A1
20120082616 Trawick et al. Apr 2012 A1
20120149843 Chien et al. Jun 2012 A1
20120244230 Mirkin et al. Sep 2012 A1
20120258126 Scholler et al. Oct 2012 A1
20120301499 Bachmann et al. Nov 2012 A1
20130028857 Gao et al. Jan 2013 A1
20130034599 Thaxton et al. Feb 2013 A1
20130071403 Rolland et al. Mar 2013 A1
20130089614 Zhang et al. Apr 2013 A1
20130095039 Lu et al. Apr 2013 A1
20130101512 Mirkin et al. Apr 2013 A1
20130123333 Mirkin et al. May 2013 A1
20130149374 Lee et al. Jun 2013 A1
20130178610 Mirkin et al. Jul 2013 A1
20130252852 Pfeiffer et al. Sep 2013 A1
20130315831 Shi et al. Nov 2013 A1
20140005258 Mirkin et al. Jan 2014 A1
20140037635 Medlock et al. Feb 2014 A1
20140065425 Bogdanov Mar 2014 A1
20140179770 Zhang Jun 2014 A1
20140199379 Tartour et al. Jul 2014 A1
20140294927 Thaxton et al. Oct 2014 A1
20150086985 Giljohann et al. Mar 2015 A1
20150118264 Baumhof et al. Apr 2015 A1
20160101128 Wang et al. Apr 2016 A1
20160108123 Freeman et al. Apr 2016 A1
20160123964 Tumeh et al. May 2016 A1
20160159905 Abdiche et al. Jun 2016 A1
20160186178 Radovic-Moreno et al. Jun 2016 A1
20160194642 Gryaznov et al. Jul 2016 A1
20160310425 Mirkin et al. Oct 2016 A1
20160339090 Hacohen et al. Nov 2016 A1
20160375115 Binder et al. Dec 2016 A1
20170044544 Mirkin et al. Feb 2017 A1
20170157048 Radovic-Moreno et al. Jun 2017 A1
20170175121 Gryaznov Jun 2017 A1
20170232109 Mirkin et al. Aug 2017 A1
20170240960 Giljohann et al. Aug 2017 A1
20170274004 Wang et al. Sep 2017 A1
20170306038 Brogdon et al. Oct 2017 A1
20170306331 Mader et al. Oct 2017 A1
20170326232 Guiducci et al. Nov 2017 A1
20180000851 Krieg Jan 2018 A1
20180042848 Gryaznov et al. Feb 2018 A1
20180043023 Ilyinskii et al. Feb 2018 A1
20180085350 Avigan et al. Mar 2018 A1
20180085398 Avigan et al. Mar 2018 A1
20180140691 Takasu et al. May 2018 A1
20180214376 Giljohann Aug 2018 A1
20180222982 Dranoff et al. Aug 2018 A1
20180311176 Ozsolak et al. Nov 2018 A1
20180318365 Yeung et al. Nov 2018 A1
20180320184 Radovic-Moreno et al. Nov 2018 A1
20180327741 Daniel et al. Nov 2018 A1
20180344873 Mirkin et al. Dec 2018 A1
20190030185 Mirkin et al. Jan 2019 A1
20190083626 Goldberg et al. Mar 2019 A1
20190142739 Patel et al. May 2019 A1
20190153098 Goldberg et al. May 2019 A1
20190211338 Mader et al. Jul 2019 A1
20190225968 Anderson et al. Jul 2019 A1
20190275166 Mirkin et al. Sep 2019 A1
20200022913 Mirkin et al. Jan 2020 A1
20200069587 Radovic-Moreno et al. Mar 2020 A1
20200101156 Mirkin et al. Apr 2020 A1
20200188521 Kang et al. Jun 2020 A1
20200248183 Nallagatla et al. Aug 2020 A1
20200255837 Anderson et al. Aug 2020 A9
20200291394 Mirkin et al. Sep 2020 A1
20200297867 Kang et al. Sep 2020 A1
20200308579 Kang Oct 2020 A1
20200339989 Daniel et al. Oct 2020 A1
20200339996 Mader et al. Oct 2020 A1
20200384104 Mirkin et al. Dec 2020 A1
20210002640 Kang et al. Jan 2021 A1
20210052497 Mirkin et al. Feb 2021 A1
20210102211 Radovic-Moreno et al. Apr 2021 A1
20210269806 Anderson Sep 2021 A1
Foreign Referenced Citations (144)
Number Date Country
1335884 Feb 2002 CN
1341660 Mar 2002 CN
1357042 Jul 2002 CN
102036652 Apr 2011 CN
102165061 Aug 2011 CN
1 251 872 Oct 2002 EP
1 255 837 Nov 2002 EP
1 266 002 Dec 2002 EP
1 326 974 Dec 2006 EP
1 889 911 Feb 2008 EP
0 959 897 Apr 2009 EP
0 817 847 Sep 2009 EP
1 015 488 Sep 2009 EP
2162117 Mar 2010 EP
2 366 406 Sep 2011 EP
2399608 Dec 2011 EP
1807094 Jan 2012 EP
2451974 May 2012 EP
2656858 Oct 2013 EP
1 294 765 Nov 2013 EP
3 112 468 Jan 2017 EP
WO 1992021330 Dec 1992 WO
WO 1993021528 Oct 1993 WO
WO 1994001550 Jan 1994 WO
WO-9401550 Jan 1994 WO
WO 1996029408 Sep 1996 WO
WO 1998004740 Feb 1998 WO
WO 1998018810 May 1998 WO
WO 1998023284 Jun 1998 WO
WO 1999027086 Jun 1999 WO
WO 1999035267 Jul 1999 WO
WO 2000013024 Mar 2000 WO
WO 2000015759 Mar 2000 WO
WO 2000020593 Apr 2000 WO
WO 2000020645 Apr 2000 WO
WO 2000029567 May 2000 WO
WO 2000042188 Jul 2000 WO
WO 2000055204 Sep 2000 WO
WO 2000069463 Nov 2000 WO
WO 2001000876 Jan 2001 WO
WO 2001022990 Apr 2001 WO
WO 2001057202 Aug 2001 WO
WO 2001059120 Aug 2001 WO
WO 2001064240 Sep 2001 WO
WO 2001068859 Sep 2001 WO
WO 2002058717 Aug 2002 WO
WO 2003008539 Jan 2003 WO
WO 2002038764 Jul 2003 WO
WO 2003055980 Jul 2003 WO
WO 2002102411 Sep 2003 WO
WO 2002064739 Jun 2004 WO
WO 2004047870 Jun 2004 WO
WO 2005063201 Jul 2005 WO
WO 2005063288 Jul 2005 WO
WO 2005116226 Dec 2005 WO
WO 2006015560 Feb 2006 WO
WO 2006080946 Aug 2006 WO
WO 2006088833 Aug 2006 WO
WO 2006110350 Oct 2006 WO
WO 2006138145 Dec 2006 WO
WO 2007047455 Apr 2007 WO
WO 2007055682 May 2007 WO
WO 2007055704 May 2007 WO
WO 2007060406 May 2007 WO
WO 2007064857 Jun 2007 WO
WO 2007089607 Aug 2007 WO
WO 2007106683 Sep 2007 WO
WO 2007122405 Nov 2007 WO
WO 2008014979 Feb 2008 WO
WO 2008042156 Apr 2008 WO
WO 2008097328 Aug 2008 WO
WO 2008127789 Oct 2008 WO
WO 2008137762 Nov 2008 WO
WO 2008141289 Nov 2008 WO
WO 2009012786 Jan 2009 WO
WO 2009026412 Feb 2009 WO
WO 2009051451 Apr 2009 WO
WO 2009061515 May 2009 WO
WO 2009073984 Jun 2009 WO
WO 2009105260 Aug 2009 WO
WO 2009120887 Oct 2009 WO
WO 2009131704 Oct 2009 WO
WO 2010017152 Feb 2010 WO
WO 2010017154 Feb 2010 WO
WO 2010085959 Aug 2010 WO
WO 2010091293 Aug 2010 WO
WO 2010120420 Oct 2010 WO
WO 2010148249 Dec 2010 WO
WO 2011017456 Feb 2011 WO
WO 2011017690 Feb 2011 WO
WO 2010147387 May 2011 WO
WO 2011053940 May 2011 WO
WO 2011072133 Jun 2011 WO
WO 2011079290 Jun 2011 WO
WO 2011091065 Jul 2011 WO
WO 2011113054 Sep 2011 WO
WO 2011143608 Nov 2011 WO
WO 2012055933 May 2012 WO
WO 2013011368 Jan 2013 WO
WO 2013012628 Jan 2013 WO
WO 2013151771 Oct 2013 WO
WO 2013177419 Nov 2013 WO
WO 2014025795 Feb 2014 WO
WO 2014123935 Aug 2014 WO
WO 2014169264 Oct 2014 WO
WO 2014175836 Oct 2014 WO
WO 2015013673 Jan 2015 WO
WO 2015013675 Jan 2015 WO
WO 2015126502 Aug 2015 WO
WO 2015153975 Oct 2015 WO
WO 2015187966 Dec 2015 WO
WO 2015195628 Dec 2015 WO
WO-2015187966 Dec 2015 WO
WO 2016004168 Jan 2016 WO
WO 2016057549 Apr 2016 WO
WO-2016057549 Apr 2016 WO
WO 2016115320 Jul 2016 WO
WO 2016134104 Aug 2016 WO
WO 2016149323 Sep 2016 WO
WO 2016149323 Sep 2016 WO
WO 2017011662 Jan 2017 WO
WO 2017035278 Mar 2017 WO
WO 2017136467 Aug 2017 WO
WO 2017161032 Sep 2017 WO
WO 2017173334 Oct 2017 WO
WO 2017181128 Oct 2017 WO
WO 2017184427 Oct 2017 WO
WO 2017193081 Nov 2017 WO
WO 2017193084 Nov 2017 WO
WO 2017193087 Nov 2017 WO
WO 2018067302 Apr 2018 WO
WO 2018152327 Aug 2018 WO
WO 2018201090 Nov 2018 WO
WO 2018209270 Nov 2018 WO
WO 2018213585 Nov 2018 WO
WO 2019118883 Jun 2019 WO
WO 2019168558 Sep 2019 WO
WO 2019169203 Sep 2019 WO
WO 2019169328 Sep 2019 WO
WO 2019246409 Dec 2019 WO
WO 2020168005 Aug 2020 WO
WO 2020219985 Oct 2020 WO
WO 2021046254 Mar 2021 WO
WO 2021202557 Oct 2021 WO
Non-Patent Literature Citations (194)
Entry
Resham J. Banga, Natalia Chernyak, Suguna P. Narayan, SonBinh T. Nguyen, and Chad A. Mirkin. “Liposomal Spherical Nucleic Acids.” Journal of the American Chemical Society, vol. 136, 2014, pp. 9866-9869. (Year: 2014).
Supporting Information for Resham J. Banga, Natalia Chernyak, Suguna P. Narayan, SonBinh T. Nguyen, and Chad A. Mirkin. “Liposomal Spherical Nucleic Acids.” Journal of the American Chemical Society, vol. 136, 2014, pp. S1-S13. (Year: 2014).
Efstathios Karathanasis, Cissy M. Geigerman, Charles A. Parkos, Leslie Chan, Ravi V. Bellamkonda, and David L. Jaye. “Selective Targeting of Nanocarriers to Neutrophils and Monocytes.” Annals of Biomedical Engineering, vol. 37, No. 10, Oct. 2009, pp. 1984-1992. (Year: 2009).
Kazunori Matsuura, Kouzo Masumoto, Yuuko Igami, Tatsuro Fujioka, and Nobuo Kimizuka. “In Situ Observation of Spherical DNA Assembly in Water and the Controlled Release of Bound Dyes.” Biomacromolecules, vol. 8, 2007, pp. 2726-2732. (Year: 2007).
Kazunori Matsuura, Taro Yamashita, Yuuko Igami and Nobuo Kimizuka. “‘Nucleo-nanocages’: designed ternary oligodeoxyribonucleotides spontaneously form nanosized DNA cages.” Chemical Communications, 2003, pp. 376-377. (Year: 2003).
Neeshma Dave and Juewen Liu. “Programmable Assembly of DNA Functionalized Liposomes by DNA.” ACS Nano, vol. 5, No. 2, 2011, pp. 1304-1312. (Year: 2011).
Qing Ge et al. “Effects of chemical modification on the potency, serum stability, and immunostimulatory properties of short shRNAs.” RNA, vol. 16, 2010, pp. 118-130. (Year: 2010).
J.N. Israelachvili, S. Marcelja, and R.G. Horn. “Physical principles of membrane organization.” Quarterly Reviews of Biophysics, vol. 13, No. 2, 1980, pp. 121-200. (Year: 1980).
Matthew P. Thompson, Miao-Ping Chien, Ti-Hsuan Ku, Anthony M. Rush, and Nathan C. Gianneschi. “Smart Lipids for Programmable Nanomaterials.” Nano Letters, vol. 10, 2010, pp. 2690-2693. (Year: 2010).
Xin Ming, Brian Laing. “Bioconjugates for targeted delivery of therapeutic oligonucleotides.” Advanced Drug Delivery Reviews, vol. 87, 2015, pp. 81-89. (Year: 2015).
Yee-Hung M. Chan, Bettina van Lengerich, and Steven G. Boxer. “Lipid-anchored DNA mediates vesicle fusion as observed by lipid and content mixing.” BioInterphases, vol. 3(2), Jun. 2008, pp. FA17-FA21. (Year: 2008).
International Search Report and Written Opinion dated Aug. 1, 2018 for PCT/US2018/030021.
International Preliminary Report on Patentability dated Nov. 7, 2019 for International Application No. PCT/US2018/030021.
Agbasi-Porter et al., Transcription inhibition using oligonucleotide-modified gold nanoparticles, Bioconjugate Chem., 17(5):1178-83 (2006).
Akhter et al., Gold nanoparticles in theranostic oncology: current state-of-the-art. Expert Opin Drug Deliv. Oct. 2012;9(10):1225-43. Epub Aug. 16, 2012.
Ali et al., Vaccines Combined with Immune Checkpoint Antibodies Promote Cytotoxic T-cell Activity and Tumor Eradication. Cancer Immunol Res. Feb. 2016;4(2):95-100. doi: 10.1158/2326-6066.CIR-14-0126. Epub Dec. 15, 2015.
Asthana et al., Mannosylated chitosan nanoparticles for delivery of antisense oligonucleotides for macrophage targeting. Biomed Res Int. 2014;2014:526391. doi: 10.1155/2014/526391. Epub Jun. 26, 2014.
Aurasense Therapeutics, NIH grant. Topically-delivered Target Gene Suppression of Immune Activation in Psoriasis. David Giljohann. Accessed on Aug. 2, 2017 from http://grantome.com/grant/NIH/R41-AR066438-01. Accessible online on Feb. 21, 2016 as verified through Wayback Machine.
Ballas et al., Induction of NK activity in murine and human cells by CpG motifs in oligodeoxynucleotides and bacterial DNA. J Immunol. Sep. 1, 1996;157(5):1840-5.
Banchelli, M. et al., “Phospholipid Membranes Decorated by Cholesterol-Based Oligonucleotides as Soft Hybrid Nanostructures,” J. Phys. Chem., 2008, 112 (35), 10942-10952.
Banga et al., Cross-linked miceller spherical nucleic acids from thermoresponsive templates, J. Am. Chem. Soc. 139:4278-81 (2017).
Banga et al., Liposomal spherical nucleic acids. J Am Chem Soc. Jul. 16, 2014;136(28):9866-9. doi: 10.1021/ja504845f. Epub Jul. 1, 2014.
Bhattarai et al., “Enhanced Gene and siRNA Delivery by Polycation-Modified Mesoporous Silica Nanoparticles Loaded with Chloroquine,” Pharm. Res., 2010, 27, 2556-2568.
Bitounis et al., Optimizing Druggability through Liposomal Formulations: New Approaches to an Old Concept. ISRN Pharm. 2012;2012:738432. doi: 10.5402/2012/738432. Epub Feb. 9, 2012.
Bode et al. CpG DNA as a vaccine adjuvant. Expert Rev Vaccines. Apr. 2011;10(4):499-511. doi: 10.1586/erv.10.174.
Bonoiu et al., Nanotechnology approach for drug addiction therapy: gene ; silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neurons. Proc Natl Acad Sci U S A. Apr. 7, 2009;106(14):5546-50. doi:; 10.1073/pnas.0901715106. Epub Mar. 23, 2009.
Bunge et al., Lipophilic oligonucleotides spontaneously insert into lipid membranes, bind complementary DNA strands, and sequester into lipid-disordered domains. Langmuir. Apr. 10, 2007;23(8):4455-64. Epub 2007 ar 17.
Chabaud et al., Enhancing effect of IL-17 on IL-1-induced IL-6 and leukemia inhibitory factor production by rheumatoid arthritis synoviocytes and its regulation by Th2 cytokines. J Immunol. Jul. 1, 1998;161(1):409-14.
Chen et al., Kinetics and thermodynamics of DNA hybridization on gold nanoparticles. Nucleic Acids Res. Jun. 2009;37(11):3756-65. doi: 10.1093/nar/gkp230. Epub Apr. 20, 2009.
Cheng et al., Interdigitated phospholipid/alkanethiol bilayers assembled on APTMS-supported gold colloid electrodes. Electroanalysis. 2004;16(1-2):127-31. doi:10.1002/elan.200302929.
Cheng et al., Tandem synthesis of core-shell brush copolymers and their transformation to peripherally cross-linked and hollowed nanostructures. J Am Chem Soc. May 31, 2006;128(21):6808-9. Published on web May 6, 2006.
Chinen et al., Spherical nucleic acid nanoparticle conjugates enhance G-quadruplex formation and increase serum protein interactions. Angew Chem Int Ed Engl. Jan. 7, 2015;54(2):527-31. doi: 10.1002/anie.201409211. Epub Nov. 13, 2014.
Cho et al., Targeted delivery of siRNA-generating DNA nanocassettes using multifunctional nanoparticles. Small. Jun. 10, 2013;9(11):1964-73. doi: 10.1002/smll.201201973. Epub Jan. 6, 2013.
Cho et al., Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res. Mar. 1, 2008;14(5):1310-6. doi: 10.1158/1078-0432.CCR-07-1441.
Choi et al., Mechanism for the endocytosis of spherical nucleic acid nanoparticle conjugates, Proc. Natl. Acad. Sci. U.S.A. 2013;110:7625-7630.
Cload et al., Polyether tethered oligonucleotide probes. J. Am. Chem. Soc. 1991;113(16): 6324-6.
Cormode et al., “Nanocrystal Core High-Density Lipoproteins: A Multimodality Contrast Agent Platform,” Nano Lett., 2008, 8 (11), 3715-3723.
Cutler et al., Polyvalent nucleic acid nanostructures. J Am Chem Soc. Jun. 22, 2011;133(24):9254-7. doi:10.1021/ja203375n. Epub Jun. 1, 2011.
Cutler et al., Polyvalent oligonucleotide iron oxide nanoparticle “click” conjugates. Nano Lett. Apr. 14, 2010;10(4):1477-80. doi:10.1021/n1100477m.
Cutler et al., Spherical nucleic acids. J Am Chem Soc. Jan. 25, 2012;134(3):1376-91. doi: 10.1021/ja209351u. Epub Jan. 9, 2012.
Daniel et al., Gold nanoparticles: assembly, supramolecular chemistry, quantum-size-related properties, and applications toward biology, catalysis, and nanotechnology. Chem Rev. Jan. 2004;104(1):293-346.
Dave et al., Programmable assembly of DNA-functionalized liposomes by DNA. ACS Nano. Feb. 22, 2011;5(2):1304-12. doi: 10.1021/nn1030093. Epub Jan. 4, 2011.
Demesmaeker et al., Antisense oligonucleotides. Acc. Chem. Res. 1995;28(9): 366-74.
Dhar et al., Polyvalent oligonucleotide gold nanoparticle conjugates as delivery vehicles for platinum (IV) warheads. J Am Chem Soc. Oct. 21, 2009; 131(41):14652-3. doi: 10.1021/ja907182.
Ding et al., A Crosslinked Nucleic Acid Nanogel for Effective siRNA Delivery and Antitumor Therapy. Angew Chem Int Ed Engl. Mar. 12, 2018;57(12):3064-3068. doi:10.1002/anie.201711242. Epub Feb. 22, 2018.
Ding et al., A statistical sampling algorithm for RNA secondary structure prediction. Nucleic Acids Res. Dec. 15, 2003;31(24):7280-301. doi: 10.1093/nar/gkg938.
Elbakry, A. et al., “Layer-by-Layer Assembled Gold Nanoparticles for siRNA Delivery,” Nano Lett., 2009, 9 (5), 2059-2064.
Fan, H. et al., “Self-Assembly of Ordered, Robust, Three-Dimensional Gold Nanocrystal/Silica Arrays,” Science, 2004, 403, 567-571.
Fattal et al., Biodegradable polyalkylcyanoacrylate nanoparticles for the delivery of oligonucleotides. J. Controlled Release. 1998;53:137-143.
Gao et al., Secondary structure effects on DNA hybridization kinetics: a solution versus surface comparison. Nucl. Acids Res. 2006;34: 3370-7.
Ghosh et al., Gold nanoparticles in delivery applications. Adv. Drug Deliv. Rev. 2008;60(11):1307-15.
Giljohann et al., Gene regulation with polyvalent siRNA-nanoparticle conjugates. J Am Chem Soc. Feb. 18, 2009; 131(6):2072-3.
Giljohann et al., Gold nanoparticles for biology and medicine. Angew Chem Int Ed Engl. Apr. 26, 2010;49(19):3280-94. doi: 10.1002/anie.200904359.
Giljohann et al., Oligonucleotide loading determines cellular uptake of DNA-modified gold nanoparticles. Nano Lett. Dec. 2007;7(12):3 818-21. Epub Nov. 13, 2007.
Gissot et al., Nucleoside, nucleotide and oligonucleotide based amphiphiles: a successful marriage of nucleic acids with lipids. Org. Biomol. Chem. 2008;6:1324-33.
Godard, G. et al., “Antisense Effects of Cholesterol-Oligodeoxynucleotide Conjugates Associated with Poly(alkylcyanoacrylate) Nanoparticles,” Eur. J. Biochem., 1995, 232 (2), 404-410.
Grijalvo et al., Oligonucleotide delivery: a patent review (2010-2013). Expert Opin Ther Pat. Jul. 2014;24(7):801-19. doi:10.1517/13543776.2014.915944. Epub May 5, 2014.
Gryaznov, Oligonucleotide n3′-->p5′ phosphoramidates and thio-phoshoramidates as potential therapeutic agents. Chem Biodivers. Mar. 2010;7(3):477-93. doi: 10.1002/cbdv.200900187. Review.
Hames et al., Gene Probes 1 A Practical Approach, IRL Press, New York (1995).
Han et al., Drug and gene delivery using gold nanoparticles. NanoBiotechnology. Mar. 2007;3(1):40-5.
Hashmi et al., Gold catalysis. Angew Chem Int Ed Engl. Dec. 4, 2006;45(47):7896-936.
Hashmi, Gold-catalyzed organic reactions. Chem Rev. Jul. 2007;107(7):3180-211. Epub Jun. 20, 2007.
Hayashi, Ultrafine particles. J. Vac. Sci. Technol. 1987;5(4):1375-1384.
He et al., Phospholipid-stabilized Au-nanoparticles. Biomacromolecules. May-Jun. 2005;6(3):1224-5.
Hellstrom et al., Epitaxial growth of DNA-assembled nanoparticle superlattices on patterned substrates. Nano Lett. 2013;13(12):6084-90. doi: 10.1021/n14033654. Epub Nov. 20, 2013.
Hong et al., Directed Assembly of Nucleic Acid-Based Polymeric Nanoparticles from Molecular Tetravalent Cores, J. Am. Chem. Soc. 137:8184-91 (2015).
Huang et al., Sequence Multiplicity within Spherical Nucleic Acids. ACS Nano. Jan. 28, 2020;14(1):1084-1092. doi: 10.1021/acsnano.9b08750. Epub Jan. 9, 2020.
Hurst, S. et al., “Maximizing DNA Loading on a Range of Gold Nanoparticle Sizes,” Anal. Chem., 2006, 78 (24), 8313-8318.
Jen et al., A nonviral transfection approach in vitro: the design of a gold nanoparticle vector joint with microelectromechanical systems. Langmuir, 2004;20(4): 1369-74.
Jensen et al., Spherical nucleic acid nanoparticle conjugates as an RNAi-based therapy for glioblastoma, Sci. Trans. Med., 5:209ra152 (2013).
Jin et al., What controls the melting properties of DNA-linked gold nanoparticle assemblies? J. Am. Chem. Soc. 2003;125: 1643.
Kachura et al., A CpG-Ficoll Nanoparticle Adjuvant for Anthrax Protective Antigen Enhances Immunogenicity and Provides Single-Immunization Protection against Inhaled Anthrax in Monkeys. J Immunol. Jan. 1, 2016;196(1):284-97. doi: 10.4049/jimmunol.1501903. Epub Nov. 25, 2015.
Karathanasis et al., Selective targeting of nanocarriers to neutrophils and monocytes. Ann Biomed Eng. Oct. 2009;37(10):1984-92. doi: 10.1007/s10439-009-9702-5. Epub Apr. 23, 2009.
Khmelinskaia et al., Effect of anchor positioning on binding and diffusion of elongated 3D DNA nanostructures on lipid membranes. J. Phys. D: Appl. Phys. Apr. 13, 2016;49(19):194001.
Kim et al., Cationic solid lipid nanoparticles reconstituted from low density lipoprotein components for delivery of siRNA. Mol Pharm. Jul.-Aug. 2008;5(4):622-31. doi: 10.1021/mp8000233. Epub May 8, 2008.
Kim, S. et al., “Systemic and Specific Delivery of Small Interfering RNAs to the Liver Mediated by Apolipoprotein A-I,” Mol. Ther., 2007, 15 (6), 1145-1152.
Kolarova et al., Preparation of magnetic oligo (dT) particles. Biotechniques. 1996;20: 196-8.
Kong et al., Cationic lipid-coated gold nanoparticles as efficient and non-cytotoxic intracellular siRNA delivery vehicles. Pharm Res. Feb. 2012;29(2):362-74. doi: 10.1007/s11095-011-0554-y. Epub Aug. 13, 2011.
Krieg et al., CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. Apr. 6, 1995;374(6522):546-9. doi: 10.1038/374546a0.
Krieg et al., Sequence motifs in adenoviral DNA block immune activation by stimulatory CpG motifs. Proc Natl Acad Sci U S A. Oct. 13, 1998;95(21):12631-6. doi: 10.1073/pnas.95.21.12631.
Kwoh et al., Stabilization of poly-L-lysine/DNA polyplexes for in vivo gene delivery to the liver. Biochim Biophys Acta. Feb. 16, 1999;1444(2):171-90.
Leander, D., “Mixed-Monolayer Gold Nanoparticles for Cancer Therapeutics,” Nanoscape, 2010, 7 (1), 11-14.
Lee et al., A DNA-Gold Nanoparticle-Based Colormetric Competition Assay for the Detection of Cysteine. Nano Letter. 2008;8(2):529-533.
Lee et al., All-in-one target-cell-specific magnetic nanoparticles for simultaneous molecular imaging and siRNA delivery. Angew Chem Int Ed Engl. 2009;48(23):4174-9. doi:10.1002/anie.200805998.
Lee et al., Chip-based scanometric detection of mercuric ion using DNA-functionalized gold nanoparticles. Anal. Chem. 2008;80(17):6805-8.
Lee et al., Imageable antigen-presenting gold nanoparticle vaccines for effective cancer immunotherapy in vivo. Angew Chem Int Ed Engl. Aug. 27, 2012;51(35):8800-5. doi:10.1002/anie.201203193.
Leleux et al., Biophysical Attributes of CpG Presentation Control TLR9 Signaling to Differentially Polarize Systemic Immune Responses. Cell Rep. Jan. 17, 2017;18(3):700-710. doi: 10.1016/j.celrep.2016.12.073.
Lenert et al., Inhibitory oligonucleotides block the induction of AP-1 transcription factor by stimulatory CpG oligonucleotides in B cells. Antisense Nucleic Acid Drug Dev. 2003;13(3):143-50.
Lennox et al., Characterization of modified antisense oligonucleotides in Xenopus laevis embryos. Oligonucleotides. 2006 Spring;16(1):26-42.
Lewandowski et al., Topically delivered spherical nucleic acid nanoconjugates targeting TNF improve the psoriatic phenotype. J Invest Dermatol. 2015 135:S71. Abstract 413.
Lewandowski et al., Topically Delivered Tumor Necrosis Factor-?-Targeted Gene Regulation for Psoriasis. J Invest Dermatol. 2017;137(9):2027-2030. doi:10.1016/j.jid.2017.04.027.
Li et al., “Molecular spherical nucleic acids,” PNAS pp. 1-5 (2018).
Li et al., Nanofabrication by DNA self-assembly. Materials Today. Elsevier Science. May 1, 2009;12(5)24-32.
Li et al., Nucleolin-targeting liposomes guided by aptamer AS1411 for the delivery of siRNA for the treatment of malignant melanomas. Biomaterials. Apr. 2014;35(12):3840-50. doi: 10.1016/j.biomaterials.2014.01.019. Epub Jan. 31, 2014.
Li et al., Reversible and Chemically Programmable Micelle Assembly with DNA Block-Copolymer Amphiphiles, Nano Lett. 2004;4:1055.
Li et al., Targeted delivery of antisense oligodeoxynucleotide and small interference RNA into lung cancer cells. Mol Pharm. Sep.-Oct. 2006;3(5):579-88. doi: 10.1021/mp060039w. Publication Date:Jul. 12, 2006.
Liu et al., “New poly(d-glucaramidoamine)s induce DNA nanoparticle formation and efficient gene delivery into mammalian cells,” J. Am. Chem. Soc. 126:7422-7423 (2004).
Liu et al., DNA-based micelles: synthesis, micellar properties and size-dependent cell permeability, Chemistrv. 2010;16:3791-7.
Liu et al., Membrane anchored immunostimulatory oligonucleotides for in vivo cell modification and localized immunotherapy. Angew Chem Int Ed Engl. Jul. 25, 2011;50(31):7052-5. doi: 10.1002/anie.201101266. Epub Jun. 17, 2011.
Liu et al., Structure-based programming of lymph-node targeting in molecular vaccines. Nature. Mar. 27, 2014;507(7493):519-22. doi: 10.1038/nature12978.
Liu, J. et al., Silica Nanoparticle Supported Lipid Bilayers for Gene Delivery, Chem. Commun., 2009, 5100-5102.
Ljubimova et al., Nanoconjugate based on polymalic acid for tumor targeting. Chem Biol Interact. Jan. 30, 2008;171(2):195-203. Epub Feb. 8, 2007.
Luo et al., Locally instilled tumor necrosis factor a antisense oligonucleotide contributes to inhibition of TH 2-driven pulmonary fibrosis via induced CD4+ CD25+ Foxp3+ regulatory T cells. J Gene Med. Nov.-Dec. 2013; 15(11-12):441-52. doi: 10.1002/jgm.2750.
Lytton-Jean et al., A thermodynamic investigation into the binding properties of DNA functionalized gold nanoparticle probes and molecular fluorophore probes. J Am Chem Soc. Sep. 21, 2005;127(37):12754-5.
Major, M. et al., “Characterisation and Phase Behaviour of Phospholipid Bilayers Adsorbed on Spherical Polysaccharidic Nanoparticles,” Biochimica et Biophysica Acta, 1997, 1327, 32-40.
Marinakos et al., Template Synthesis of One-Dimensional Au, Au-Poly(pyrrole), and Poly(pyrrole) Nanoparticle Arrays. Chem. Mater. 1998;10:1214-19.
Massich et al., Regulating immune response using polyvalent nucleic acid-gold nanoparticle conjugates. Mol Pharm. Nov.-Dec. 2009;6(6):1934-40.
Matijevic et al., Fine Particles Part II: Formation Mechanisms and Applications. MRS Bulletin pp. 16-47 (1990).
Matsunaga, T. et al., “Biomagnetic Nanoparticle Formation and Application,” Supramolecular Science, 1998, 5 (3-4), 391-394.
Maye et al., A simple method for kinetic control of DNA-induced nanoparticle assembly. J. Am. Chem. Soc. 2006; 128: 14020-1.
McBain, S. et al., “Polyethyleneimine Functionalized Iron Oxide Nanoparticles as Agents for DNA Deliver and Transfection,” J. Mater. Chem., 2007, 17, 2561-2565.
McKay et al., Characterization of a potent and specific class of antisense oligonucleotide inhibitor of human protein kinase C-alpha expression. J Biol Chem. Jan. 15, 1999;274(3):1715-22.
McMahon et al., Biomimetic high density lipoprotein nanoparticles for nucleic acid delivery. Nano Lett. Mar. 9, 2011;11(3):1208-14. doi: 10.1021/n11041947. Epub Feb. 14, 2011.
Medintz et al., A reactive peptidic linker for self-assembling hybrid quantum dot-DNA bioconjugates. Nano Lett. Jun. 2007;7(6):1741-8. Epub May 26, 2007.
Miller et al., Antisense oligonucleotides: Strategies for delivery. PSTT. 1998;1(9): 377-86.
Ming et al., Albumin-based nanoconjugates for targeted delivery of ; therapeutic oligonucleotides. Biomaterials. Oct. 2013;34(32):7939-49. doi:; 10.1016/j.biomaterials.2013.06.066. Epub Jul. 19, 2013.
Miossec, Interleukin-17 in rheumatoid arthritis: if T cells were to contribute to inflammation and destruction through synergy. Arthritis Rheum. Mar. 2003;48(3):594-601. doi: 10.1002/art.10816. PMID: 12632409.
Mohamed et al., Effect of toll-like receptor 7 and 9 targeted therapy to prevent the development of hepatocellular carcinoma. Liver Int. Mar. 2015;35(3):1063-76. doi: 10.1111/liv.12626. Epub Jul. 30, 2014.
Monia et al., Nuclease resistance and antisense activity of modified oligonucleotides targeted to Ha-ras. J Biol Chem. Jun. 14, 1996;271(24):14533-40.
Moughton et al., Hollow nanostructures from self-assembled supramolecular metallo-triblock copolymers. Soft Matter. 2009;5(12):2361-70.
Mucic et al., Synthesis and characterization of DNA with ferrocenyl groups attached to their 5′-termini: electrochemical characterization of a redox-active nucleotide monolayer. Chem. Comm. 1996;555-7.
Nallagatla et al., Abstract 4706: Spherical nucleic acids targeting toll-like receptor 9 enhance antitumor activity in combination with anti-PD-1 antibody in mouse tumor models. Cancer Res. Jul. 1, 2017;22(13 Supplement):4706.
Nam et al., Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins. Science. Sep. 26, 2003;301(5641):1884-6.
Nemati et al., Using siRNA-based spherical nucleic acid nanoparticle conjugates for gene regulation in psoriasis. J Control Release. Dec. 28, 2017;268:259-268. doi: 10.1016/j.jconrel.2017.10.034. Epub Oct. 23, 2017.
Niemeyer, C. et al., “Bifunctional DNA-Gold Nanoparticle Conjugates as Building Blocks for the Self-Assembly of Cross-Linked Particle Layers,” Biochemical and Biophysical Research Communications, 2003, 311 (4), 995-999.
Nikolov et al., Bias-dependent admittance in hybrid bilayer membranes. Langmuir. Aug. 15, 2006;22(17):7156-8.
Nuzzo et al., Spontaneously organized molecular assemblies. 3. Preparation and properties of solution adsorbed monolayers of organic disulfides on gold surfaces. J. Am. Chem. Soc. 1987;109:2358-68.
Pan et al., Dendrimer-Modified Magnetic Nanoparticles Enhance Efficiency of Gene Delivery System. Cancer Res. 2007;67:8156-8163.
Park et al., DNA-programmable nanoparticle cystrallization. Nature. 2008;451: 553-6.
Patel et al., Peptide antisense nanoparticles. Proc Natl Acad Sci U S A. Nov. 11, 2008;105(45):17222-6. doi: 10.1073/pnas.0801609105.
Patel et al., Scavenger receptors mediate cellular uptake of polyvalent oligonucleotide-functionalized gold nanoparticles, Bioconj. Chem., 21:2250 (2010).
Patil et al., “Evidence for Novel Interdigitated Bilayer Formation of Fatty Acids During Three-Dimensional Self-Assembly on Silver Colloidal Particles,” J. Am. Chem. Soc., 1997, 119 (39), 9281-9282.
Patil et al., DNA-based therapeutics and DNA delivery systems: a comprehensive review. AAPS J., 2005;7(1): E61-77.
Patwa et al., Hybrid lipid oligonucleotide conjugates: synthesis, self-assemblies and biomedical applications. Chem Soc Rev. 2011;40:5844-54.
Paul, New Way to Kill Lymphoma without Chemotherapy uses Golden Nanoparticles. Feinberg School of Medicine: Northwestern University. Jan. 22, 2013. 4 pages. ww.feinberg.northwestern.edu/news/2013/01/lymphoma_nanoparticales.html.
Peter et al., Characterization of suppressive oligodeoxynucleotides that inhibit Toll-like receptor-9-mediated activation of innate immunity. Immunology. Jan. 2008;123(1):118-28. Epub Oct. 23, 2007.
Plant et al., Self-assembled phospholipid/alkanethiol biomimetic bilayers on gold. Langmuir. 1993;9:2764-7.
Pokholenko et al., Lipid oligonucleotide conjugates as responsive nanomaterials for drug delivery. J of Materials Chemistry B. 2013;5329-34.
Ponnappa et al., Inhibition of tumor necrosis factor alpha secretion and prevention of liver injury in ethanol-fed rats by antisense oligonucleotides. Biochem Pharmacol. Feb. 15, 2005;69(4):569-77. Epub Dec. 30, 2004.
Qin et al., Significantly improved analytical sensitivity of lateral flow immunoassays by using thermal contrast. Angew Chem Int Ed Engl. Apr. 27, 2012;51(18):4358-61. doi:10.1002/anie.201200997. Epub Mar. 23, 2012.
Radovic-Moreno et al., Immunomodulatory spherical nucleic acids. Proc Natl Acad Sci U S A. Mar. 31, 2015;112(13):3892-7. doi: 10.1073/pnas.1502850112. Epub Mar. 16, 2015.
Rana et al., Monolayer coated gold nanoparticles for delivery applications. Adv Drug Deliv Rev. Feb. 2012;64(2):200-16. doi: 10.1016/j.addr.2011.08.006. Epub Sep. 6, 2011.
Rojanasakul et al., Antisense inhibition of silica-induced tumor necrosis factor in alveolar macrophages. J Biol Chem. Feb. 14, 1997;272(7):3910-4.
Romanucci et al., Synthesis, biophysical characterization and anti-HIV activity of d(TG3AG) Quadruplexes bearing hydrophobic tails at the 5′-end. Bioorg Med Chem. Feb. 1, 2014;22(3):960-6. doi: 10.1016/j.bmc.2013.12.051. Epub Jan. 4, 2014.
Rosi et al., Oligonucleotide-modified gold nanoparticles for intracellular gene regulation. Science. May 19, 2006;312(5776):1027-30.
Rubenstein et al., Antisense oligonucleotide intralesional therapy for human PC-3 prostate tumors carried in athymic nude mice. J Surg Oncol. Jul. 1996;62(3):194-200. doi: 10.1002/(SICI)1096-9098(199607)62:3<194::AID-JSO9>3.0.CO;2-2.
Rush et al., Intracellular mRNA regulation with self-assembled locked nucleic acid polymer nanoparticles. J Am Chem Soc. May 28, 2014;136(21):7615-8. doi: 10.1021/ja503598z. Epub May 14, 2014.
Seferos et al., Polyvalent DNA nanoparticle conjugates stabilize nucleic acids. Nano Lett. Jan. 2009;9(1):308-11.
Shahzad et al., Targeted delivery of small interfering RNA using reconstituted high-density lipoprotein nanoparticles. Neoplasia. Apr. 2011;13(4):309-19.
Shukla et al., Development of streptavidin-based ; nanocomplex for siRNA delivery. Mol Pharm. Dec. 2, 2013;10(12):4534-45. doi:; 10.1021/mp400355q. Epub Oct. 25, 2013.
Sita et al., Dual bioluminescence and near-infrared fluorescence monitoring to evaluate spherical nucleic acid nanoconjugate activity in vivo. Proc Natl Acad Sci U S A. Apr. 18, 2017;114(16):4129-4134. doi: 10.1073/pnas.1702736114. Epub Apr. 3, 2017.
Song et al., Backbone-modified oligonucleotides for tuning the cellular uptake behaviour of spherical nucleic acids. Biomater Sci. Feb. 28, 2017;5(3):412-416. doi: 10.1039/c6bm00792a.
Storhoff et al., Sequence-Dependent Stability of DNA-Modified Gold Nanoparticles. Langmuir. 2002;18: 6666-6670.
Stunz et al., Inhibitory oligonucleotides specifically block effects of stimulatory CpG oligonucleotides in B cells. Eur J Immunol. May 2002;32(5):1212-22.
Tang et al., Probing hydroxyl radicals and their imaging in living cells by use of FAM-DNA-Au nanoparticles. Chemistry. Jan. 7, 2008;14(2):522-8.
Taton et al., Scanometric DNA array detection with nanoparticle probes. Science, 2000;289(5485):1757-60.
Thomas, “The Interaction of HgCl2 with Sodium Thymonucleate,” J. Am. Chem. Soc., 76:6032-6034 (1954).
Thompson et al., Smart lipids for programmable nanomaterials. Nano Lett. Jul. 14, 2010;10(7):2690-3. doi: 10.1021/n1101640k.
Tiwari et al., Functionalized gold nanoparticles and their biomedical applications. Nanomaterials. 2011;1:31-63. doi: 10.3390/nano1010031.
Tondelli et al., “Highly efficient cellular uptake of c-myb antisense oligonucleotides through specifically desianed polymeric nanospheres,” Nucl. Acids Res. 26:5425-5431 (1998).
Tripathy et al., High Density Lipoprotein Nanoparticles Deliver RNAi to Endothelial Cells to Inhibit Angiogenesis. Part Part Syst Charact. Nov. 1, 2014;31(11):1141-1150.
United States Securities and Exchange Commission Form 8-K Current Report, Date of Report (Date of earliest event reported): Sep. 26, 2017, Exicure, Inc. Dated: Oct. 2, 2017 by David Giljohann Accessed from the internet (Oct. 11, 2018) at https://www.sec.gov/Archives/edgar/data/1698530/000119312517301064/d461080d8k.htm.
Wagner et al., Gene inhibition using antisense oligodeoxynucleotides. Nature, 1994;372: 333-5.
Wei et al., Polyvalent immunostimulatory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticles. Angew Chem Int Ed Engl. Jan. 27, 2012;51(5):1202-6. doi:10.1002/anie.201105187. Epub Dec. 21, 2011.
Wilson et al., pH-Responsive nanoparticle vaccines for dual-delivery of antigens and immunostimulatory oligonucleotides. ACS Nano. May 28, 2013;7(5):3912-25. doi: 10.1021/nn305466z. Epub Apr. 30, 2013.
Wilton et al. Antisense oligonucleotide-induced exon skipping across the human dystrophin gene transcript. Mol Ther. Jul. 2007;15(7):1288-96. Epub Feb. 6, 2007.
Wolfrum et al., Mechanisms and optimization of in vivo delivery of lipophilic siRNAs. Nat Biotechnol. Oct. 2007;25(10):1149-57. Epub Sep. 16, 2007.
Wu et al., DNA aptamer-micelle as an efficient detection/delivery vehicle toward cancer cells. Proc Natl Acad Sci U S A. Jan. 5, 2010;107(1):5-10. doi: 10.1073/pnas.0909611107. Epub Dec. 22, 2009.
Wu et al., Intracellular fate of spherical nucleic acid nanoparticle conjugates. J Am Chem Soc. May 28, 2014;136(21):7726-33. doi: 10.1021/ja503010a. Epub May 19, 2014.
Xiao et al., Mannosylated bioreducible nanoparticle-mediated macrophage-specific Tnf-? RNA interference for IBD therapy. Biomaterials. Oct. 2013;34(30):7471-82. doi: 10.1016/j.biomaterials.2013.06.008. Epub Jun. 29, 2013.
Xu et al., A gold-nanoparticle-based real-time colorimetric screening method for endonuclease activity and inhibition. Angew. Chem. Int. Ed. Engl., 2007;46(19):3468-70.
Xu et al., Homogeneous detection of nucleic acids based upon the light scattering properties of silver-coated nanoparticle probes. Anal. Chem., 2007;79(17):6650-4.
Xu et al., Thermodynamics of DNA hybridization on gold nanoparticles. J. Am. Chem. Soc., 2005;127(38): 13227-31.
Yamamoto et al., Unique palindromic sequences in synthetic oligonucleotides are required to induce IFN [correction of INF] and augment IFN-mediated [correction of INF] natural killer activity. J Immunol. Jun. 15, 1992;148(12):4072-6.
Yamane et al., On the complexing of desoxyribonucleic acid (DNA) by mercuric ion. J. Am. Chem. Soc., 1961;83:2599-2607.
Yang et al., Inhibition of a C-rich oligodeoxynucleotide on activation of immune cells in vitro and enhancement of antibody response in mice. Immunology. Dec. 2010;131(4):501-12. doi: 10.1111/j.1365-2567.2010.03322.x.
Yao et al. Herpesvirus Saimiri encodes a new cytokine, IL-17, which binds to a novel cytokine receptor. Immunity. Dec. 1995;3(6):811-21.
Yin et al., Supramolecular self-assembled nanoparticles mediate oral delivery of therapeutic TNF-? siRNA against systemic inflammation. Angew Chem Int Ed Engl. May 27, 2013;52(22):5757-61. doi: 10.1002/anie.201209991. Epub Apr. 22, 2013.
Zhang et al., “A general strategy for the DNA-mediated self-assembly of functional nanoparticles into heterogeneous systems,” Nat Nanotechnol 8(11): 865-872 (2013).
Zhang et al., A general approach to DNA-programmable atom equivalents. Nat Mater. Aug. 2013;12(8):741-6. doi: 10.1038/nmat3647. Epub May 19, 2013.
Zhang et al., Informational liposomes: Complexes derived from cholesteryl-conjugated oligonucleotides and liposomes. Tetrahedron Letters. 1996. 37(35):6243-6.
Zhang et al., Nanopod formation through gold nanoparticle templated and catalyzed crosslinking of polymers bearing pendant propargyl ethers. J Am Chem Soc. Nov. 3, 2010;132(43):15151-3.
Zhang et al., Self-assembled monolayers of terminal alkynes on gold. J Am Chem Soc. Apr. 25, 2007;129(16):4876-7. Epub Mar. 31, 2007.
Zheng et al., A spherical nucleic acid platform based on self-assembled DNA biopolymer for high-performance cancer therapy. ACS Nano. Aug. 27, 2013;7(8):6545-54. doi: n402344v. Epub Jul. 23, 2013.
Zheng et al., Sterically controlled docking of gold nanoparticles on ferritin; surface by DNA hybridization. Nanotechnology. Jul. 8, 2011;22(27):275312. doi:; 10.1088/0957-4484/22/27/275312. Epub May 26, 2011.
Zheng et al., Topical delivery of siRNA-based spherical nucleic acid nanoparticle conjugates for gene regulation. Proc Natl Acad Sci U S A. Jul. 24, 2012;109(30):11975-80. doi: 10.1073/pnas.1118425109. Epub Jul. 6, 2012.
De Titta et al., Nanoparticle conjugation of CpG enhances adjuvancy for cellular immunity and memory recall at low dose. Proc Natl Acad Sci U S A. Dec. 3, 2013;110(49):19902-7. doi: 10.1073/pnas.1313152110. Epub Nov. 18, 2013.
Inoue, Oligonucleotide therapeutics: past, present and future. Drug Delivery System. Jan. 2016;31(1):10-23.
Radovic-Moreno et al., Immunomodulatory spherical nucleic acids. Proc Natl Acad Sci U S A PNAS. Mar. 31, 2015;112(13):3892-7. doi: 10.1073/pnas.1502850112. Epub Mar. 16, 2015.
Somiya et al., Potential of a non-cationic lioposomes-based delivery system for nucleic acid medicines. Drug Delivery System. Jan. 2016;31(1):35-43.
Zhang et al., Influence of anchoring ligands and particle size on the colloidal stability and in vivo biodistribution of polyethylene glycol-coated gold nanoparticles in tumor-xenografted mice. Biomaterials. Apr. 2009;30(10):1928-36. doi: 10.1016/j.biomaterials.2008.12.038. Epub Jan. 7, 2009. Author Manuscript, 22 pages.
Huang et al., CRISPR Spherical Nucleic Acids. J Am Chem Soc. Oct. 19, 2022;144(41):18756-18760. doi: 10.1021/jacs.2c07913. Epub Oct. 6, 2022.
Krieg et al., Induction of systemic TH1-like innate immunity in normal volunteers following subcutaneous but not intravenous administration of CPG 7909, a synthetic B-class CpG oligodeoxynucleotide TLR9 agonist. J Immunother. Nov.-Dec. 2004;27(6):460-71. doi: 10.1097/00002371-200411000-00006.
Kusmierz et al., Defining the Design Parameters for in Vivo Enzyme Delivery Through Protein Spherical Nucleic Acids. ACS Cent Sci. May 27, 2020;6(5):815-822. doi: 10.1021/acscentsci.0c00313. Epub Apr. 27, 2020.
Related Publications (1)
Number Date Country
20200188521 A1 Jun 2020 US
Provisional Applications (1)
Number Date Country
62492062 Apr 2017 US