Liposomal spherical nucleic acid (SNA) constructs presenting antisense oligonucleotides (ASO) for specific knockdown of interleukin 17 receptor mRNA

Information

  • Patent Grant
  • 11866700
  • Patent Number
    11,866,700
  • Date Filed
    Friday, May 5, 2017
    7 years ago
  • Date Issued
    Tuesday, January 9, 2024
    4 months ago
Abstract
Aspects of the invention relate to antisense oligonucleotides directed to the interleukin 17 receptor (IL-17R), and other targets. Spherical nucleic acid formulations or compositions of antisense oligonucleotides and related methods of treatment are also provided.
Description
BACKGROUND OF INVENTION

Inflammation, which can be classified as either acute or chronic, involves the activation of the immune system in response to harmful stimuli, such as, e.g., a pathogen, infection, irritant, or damage to cells. Acute inflammation is mediated by granulocytes, while chronic inflammation is mediated by mononuclear cells such as monocytes and lymphocytes.


The process of acute inflammation is initiated by cells such as macrophages, dendritic cells, histiocytes, Kupffer cells, mastocytes, vascular endothelial cells, and vascular smooth muscle cells. At the onset of a harmful stimulus, these cells undergo activation and release inflammatory mediating and sensitizing molecules, such as, e.g., pro-inflammatory cytokines, pro-inflammatory prostaglandins, leukotrienes, histamine, serotonin, neutral proteases, bradykinin and nitric oxide. These inflammatory molecules modulate a complex series of biological events involving cellular and acellular components of the local vascular system, the immune system, and the injured tissue site to propagate and mature the inflammatory response.


Severe or prolonged stimulation results in a chronic inflammatory response that leads to a progressive shift in the type of cells present at the site of tissue injury. Chronic inflammation may be characterized as the simultaneous destruction and healing of tissue from the inflammatory process, with the net result of provoking injury rather than mediating repair. As an inflammatory response can occur anywhere in the body, chronic inflammation has been implicated in the pathophysiology of a wide range of seemingly unrelated disorders which underlay a large and varied group of human diseases. For example, chronic inflammation is involved in diseases as diverse as psoriasis, cardiovascular diseases, cancers, allergies, obesity, diabetes, digestive system diseases, degenerative diseases, auto-immune disorders, and Alzheimer's disease.


SUMMARY OF INVENTION

The present disclosure, in some aspects, includes a single-stranded modified oligonucleotide consisting of 10-30 linked nucleosides and having: a gap segment consisting of two to eight linked deoxynucleosides; a 5′ wing segment consisting of linked nucleosides; and a 3′ wing segment consisting of linked nucleosides; wherein the gap segment is positioned between the 5′ wing segment and the 3′ wing segment; wherein at least some nucleosides of each wing segment comprises a modified nucleotide; wherein the internucleoside linkages within the gap segment and the linkages connecting the gap segment to the 3′ wing segments are all phosphorothioate linkages (*); and the internucleoside linkages connecting the rest of the nucleosides of both the 5′ and 3′ wing segments are phosphodiester linkages; and wherein the nucleobase sequence of the oligonucleotide consists of 10-30 contiguous nucleobases complementary to an equal length portion of SEQ ID NO: 302, or a pharmaceutically acceptable salt thereof.


In some embodiments the single-stranded modified oligonucleotide consists of 17-21 linked nucleosides. In other embodiments the gap segment consists of six linked deoxynucleosides. In yet other embodiments wherein each nucleoside of each wing segment comprises a modified nucleotide. In other embodiments the modified nucleotide is 2′O-methyl ribonucleoside (m). The oligonucleotide has 12 2′O-methyl ribonucleosides in other embodiments. In some embodiments the nucleobase sequence of the oligonucleotide consists of 17-21 contiguous nucleobases complementary to an equal length portion of SEQ ID NO: 302.


In one embodiment, the nucleobase sequence of the oligonucleotide is GCUUGGGCAGGTGGUGAA (SEQ ID NO: 225). In another embodiment, the nucleobase sequence of the oligonucleotide is CCCACAGGGGCATGUAGU (SEQ ID NO: 288). An additional embodiment includes an oligonucleotide with the nucleobase sequence of GUAGGGCGUGTGTGGGUC (SEQ ID NO: 291). In another embodiment, the nucleobase sequence of the oligonucleotide is mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA (SEQ ID NO: 225). In a further embodiment, the nucleobase sequence of the oligonucleotide is mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU (SEQ ID NO: 288). In yet another embodiment, the nucleobase sequence of the oligonucleotide is











(SEQ ID NO: 291)



mGmUmAmGmGmGmCmGmUG*T*G*T*G*G*mGmUmC.






In some embodiments, the compound is 20 nucleotides in length.


In some embodiments, the oligonucleotide further comprises a molecular species at one of the ends. In another embodiment, the compound further comprises a molecular species at both ends.


In some embodiments, the molecular species is selected from the group consisting of a spacer, a lipid, a sterol, cholesterol, NAcetylgalactosamine (GalNAc), modified GalNAc, derivatized or substituted GalNAc, stearyl, C16 alkyl chain, bile acids, cholic acid, taurocholic acid, deoxycholate, oleyl litocholic acid, oleoyl cholenic acid, glycolipids, phospholipids, sphingolipids, isoprenoids, such as steroids, vitamins, such as vitamin E, saturated fatty acids, unsaturated fatty acids, fatty acid esters, such as triglycerides, pyrenes, porphyrines, Texaphyrine, adamantane, acridines, biotin, coumarin, fluorescein, rhodamine, Texas-Red, digoxygenin, dimethoxytrityl, t-butyldimethylsilyl, t-butyldiphenylsilyl, cyanine dyes (e.g. Cy3 or Cy5), Hoechst 33258 dye, psoralen, and ibuprofen.


In other embodiments, the molecular species is selected from the group consisting of a lipophilic moiety; a folic acid radical; a steroid radical; a carbohydrate radical; a vitamin A radical; a vitamin E radical; or a vitamin K radical.


In some embodiments, the molecular species is connected directly to the compound through a linkage selected from the group consisting of phosphodiester, phosphorothioate, phosphorodithioate, methylphosphonate, and amide linkages.


In another embodiment, the molecular species is connected indirectly to the compound through a linker. In some embodiments, the linker is a non-nucleotidic linker selected from the group consisting of abasic residues (dSpacer), oligoethyleneglycol, such as triethyleneglycol (spacer 9) or hexaethylenegylcol (spacer 18), and alkane-diol, such as butanediol.


In some embodiments, the 3′ end of the oligonucleotide is connected to 2 consecutive linkers that are hexaethylenegylcol (spacer 18), the first hexaethylenegylcol connected to the 3′ end of the oligonucleotide, the second hexaethylenegylcol connected to the first hexaethylenegylcol and the second hexaethylenegylcol is connected to a cholesterol.


Another aspect of the present disclosure includes an oligonucleotide comprising mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA/isp18//isp18//3CholTEG/(SEQ ID NO: 225), wherein the oligonucleotide is 20 nucleotides in length, wherein m is a 2′O methyl, and wherein * is a phosphorothioate modification.


An additional aspect of the present disclosure includes an oligonucleotide comprising mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/.


(SEQ ID NO: 288), wherein the oligonucleotide is 20 nucleotides in length, wherein m is a 2′O methyl, and wherein * is a phosphorothioate modification.


A further aspect of the present disclosure includes an oligonucleotide comprising mGmUmAmGmGmGmCmGmUG*T*G*T*G*G*mGmUmC/isp18//isp18//3CholTEG/(SEQ ID NO: 291), wherein the oligonucleotide is 20 nucleotides in length, wherein m is a 2′O methyl, and wherein * is a phosphorothioate modification.


Another aspect of the present disclosure includes a stable self-assembling nanostructure, comprising a core having an oligonucleotide shell comprised of an antisense oligonucleotide 18 to 21 linked nucleosides in length targeted to Interleukin 17 receptor (IL-17R, IL-17RA) positioned on the exterior of the core. In some embodiments, the antisense oligonucleotide is 18 nucleotides in length. In other embodiments, IL-17RA has a sequence of SEQ ID NO: 302.


In another embodiment, less than all of the internucleoside linkages are phosphodiester. In some embodiments, the antisense oligonucleotide has phosphorothioate internucleoside linkages. In other embodiments, less than all of the internucleoside linkages are phosphorothioate. In another embodiment, the oligonucleotides have at least one internucleoside phosphorothioate linkage that is stereo-enriched. In another embodiment, the oligonucleotides have all the internucleoside phosphorothioate linkage that are stereo-enriched. The stereo-enriched phosphorothioate linkage may be Rp diastereomer, or Sp diastereomer.


In some embodiments, the antisense oligonucleotide has 2′O methyl modifications. In other embodiments, less than all of the nucleotides include a 2′O methyl modification.


In some embodiments, the antisense oligonucleotide has 2′O alkyl modifications. In other embodiments, less than all of the nucleotides include a 2′O alkyl modification.


In some embodiments, the antisense oligonucleotide has 17 internucleoside linkages and 6 central internucleoside linkages are phosphorothioate.


In some embodiments, the antisense oligonucleotide has a nucleobase sequence complementary to a sequence comprising at least 8 contiguous nucleobases of a sequence recited in SEQ ID NO: 302. In other embodiments, the antisense oligonucleotide is selected from the group consisting of

    • mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA/isp18//isp18//3CholTEG/(SEQ ID NO: 225);
    • mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288); and
    • mGmUmAmGmGmGmCmGmUG*T*G*T*G*G*mGmUmC/isp18//isp18//3CholTEG/(SEQ ID NO: 291) wherein—refers to a phosphodiester bond, * refers to a phosphorothioate bond, and m refers to a O methyl.


In some embodiments, the nanostructure includes 2-1,000 copies of the antisense oligonucleotide. In other embodiments, the nanostructure includes at least two different antisense oligonucleotides.


In some embodiments, the core is a solid or hollow core. In other embodiments, the core is a solid core and further comprising a lipid bilayer surrounding the core. In another embodiment, the solid core is comprised of noble metals, including gold and silver, transition metals including iron and cobalt, metal oxides including silica, polymers or combinations thereof. In some embodiments, the core is a polymeric core and wherein the polymeric core is comprised of amphiphilic block copolymers, hydrophobic polymers including polystyrene, poly(lactic acid), poly(lactic co-glycolic acid), poly(glycolic acid), poly(caprolactone) and other biocompatible polymers.


In other embodiments, the core is a liposomal core. In some embodiments, the liposomal core is comprised of one or more lipids selected from: 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dimyristoyl-sn-phosphatidylcholine (DMPC), 1-palmitoyl-2-oleoyl-sn-phosphatidylcholine (POPC), 1,2-distearoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (DSPG), 1,2-dioleoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (DOPG), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-di-(9Z-octadecenoyl)-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine (DPPE), sphingolipids such as sphingosine, sphingosine phosphate, methylated sphingosines and sphinganines, ceramides, ceramide phosphates, 1-0 acyl ceramides, dihydroceramides, 2-hydroxy ceramides, sphingomyelin, glycosylated sphingolipids, sulfatides, gangliosides, phosphosphingolipids, and phytosphingosines of various lengths and saturation states and their derivatives, phospholipids such as phosphatidylcholines, lysophosphatidylcholines, phosphatidic acids, lysophosphatidic acids, cyclic LPA, phosphatidylethanolamines, lysophosphatidylethanolamines, phosphatidylglycerols, lysophosphatidylglycerols, phosphatidylserines, lysophosphatidylserines, phosphatidylinositols, inositol phosphates, LPI, cardiolipins, lysocardiolipins, bis(monoacylglycero) phosphates, (diacylglycero) phosphates, ether lipids, diphytanyl ether lipids, and plasmalogens of various lengths, saturation states, and their derivatives, sterols such as cholesterol, desmosterol, stigmasterol, lanosterol, lathosterol, diosgenin, sitosterol, zymosterol, zymostenol, 14-demethyl-lanosterol, cholesterol sulfate, DHEA, DHEA sulfate, 14-demethyl-14-dehydrlanosterol, sitostanol, campesterol, ether anionic lipids, ether cationic lipids, lanthanide chelating lipids, A-ring substituted oxysterols, B-ring substituted oxysterols, D-ring substituted oxysterols, side-chain substituted oxysterols, double substituted oxysterols, cholestanoic acid derivatives, fluorinated sterols, fluorescent sterols, sulfonated sterols, phosphorylated sterols, and polyunsaturated sterols of different lengths, saturation states, and derivatives thereof.


The present disclosure, in some aspects, includes a multiplex antisense oligonucleotide spherical nucleic acid (mASO-SNA), comprising a core having an oligonucleotide shell comprised of an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a first gene and an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a second gene, wherein the core is a solid surrounded by a lipid bilayer or a liposome or lipoplex complex core and the oligonucleotide shell is positioned on the exterior of the core.


In some embodiments, the first gene and the second gene are associated with a disease. In an embodiment, the disease is an inflammatory disorder. In another embodiment, the disease is psoriasis.


In some embodiments, the first gene and the second gene are associated with a target pathway.


In another embodiment, the mASO-SNA further comprises an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a third gene. In some embodiments, the first gene is an Interleukin 17 receptor (IL-17RA). In other embodiments, the second gene is TNF. In some embodiments, the first gene and the second gene are present in an approximate equimolar amount in the oligonucleotide shell.


In another embodiment, the mASO-SNA comprises an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to four or more genes.


The present disclosure, in some aspects, provides a method for treating a disorder, comprising: administering to a subject having a disorder a multiplex antisense oligonucleotide spherical nucleic acid (mASO-SNA), comprising a core having an oligonucleotide shell comprised of an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a first gene and an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a second gene, wherein the core is a solid surrounded by a lipid bilayer or a liposome or lipoplex complex core and the oligonucleotide shell is positioned on the exterior of the core in an effective amount to treat the disorder.


In some embodiments, the disorder is an inflammatory disorder.


In other embodiments, the mASO-SNA produces simultaneous mRNA knock-down of the first and second gene. In another embodiment, the first gene and the second gene are associated with a target pathway. In some embodiments, the mASO-SNA produces additive knock-down of the target pathway.


In other embodiments, the disorder is psoriasis.


Another aspect of the present disclosure provides a method for treating an inflammatory disorder, including administering to a subject having an inflammatory disorder a composition comprising the oligonucleotide or the nanostructure described herein in an effective amount to treat the inflammatory disorder.


In some embodiments, the inflammatory disorder is selected from the group consisting of an autoimmune disease, an infectious disease, transplant rejection or graft-versus-host disease, malignancy, a pulmonary disorder, an intestinal disorder, a cardiac disorder, sepsis, a spondyloarthropathy, a metabolic disorder, anemia, pain, a hepatic disorder, a skin disorder, a nail disorder, rheumatoid arthritis, psoriasis, psoriasis in combination with psoriatic arthritis, ulcerative colitis, Crohn's disease, vasculitis, Behcet's disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disorder (COPD), idiopathic pulmonary fibrosis (IPF), restenosis, diabetes, anemia, pain, a Crohn's disease-related disorder, juvenile rheumatoid arthritis (JRA), a hepatitis C virus infection, psoriatic arthritis, and chronic plaque psoriasis.


In other embodiments, the inflammatory disorder is selected from the group consisting of rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis, and nephritic syndrome.


An additional aspect of the present disclosure includes a method for reducing expression levels of IL-17 receptor in vivo, including administering to a subject a composition comprising the oligonucleotide or the nanostructure described herein in an effective amount to reduce IL-17 receptor levels in vivo.


In some embodiments, the first gene, the second gene, and the third gene are associated with a target pathway. In some embodiments, the first gene and the second gene are associated with different target pathways. In some embodiments, the first gene, the second gene and the third gene are associated with different target pathways. In some embodiments, the first gene is an interleukin-4 receptor (IL-4R). In some embodiments, the second gene is interleukin-1 beta (IL-1β). In some embodiments, the third gene is connective tissue growth factor (CTGF).


In some embodiments, the first gene, the second gene, and the third gene are present in an approximate equimolar amount in the oligonucleotide shell.


According to another aspect, pharmaceutical compositions are provided herein. In some embodiments, the pharmaceutical composition includes a stable self-assembling nanostructure, wherein the self-assembling nanostructure comprises a core having an oligonucleotide shell comprised of an antisense oligonucleotide 18 to 21 linked nucleosides in length targeted to interleukin 17 receptor (IL-17R) positioned on the exterior of the core.


In some embodiments, the antisense oligonucleotide is 18 nucleotides in length. In some embodiments, the IL-17R has a sequence of SEQ ID NO: 302.


In some embodiments, less than all of the internucleoside linkages are phosphodiester. In some embodiments, the antisense oligonucleotide has phosphorothioate internucleoside linkages. In some embodiments, less than all of the internucleoside linkages are phosphorothioate.


In some embodiments, the antisense oligonucleotide has 2′O methyl modifications. In some embodiments, less than all of the nucleotides include a 2′O methyl modification.


In some embodiments, the antisense oligonucleotide has 17 internucleoside linkages and wherein 6 central internucleoside linkages are phosphorothioate. In some embodiments, the antisense oligonucleotide has a nucleobase sequence complementary to a sequence including at least 8 contiguous nucleobases of a sequence recited in SEQ ID NO: 302.


In some embodiments, the antisense oligonucleotide is selected from the group consisting of

    • mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA/isp18//isp18//3CholTEG/(SEQ ID NO: 225);
    • mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288); and
    • mGmUmAmGmGmGmCmGmUG*T*G*T*G*G*mGmUmC/isp18//isp18//3CholTEG/(SEQ ID NO: 291) wherein—refers to a phosphodiester bond, * refers to a phosphorothioate bond, and m refers to a O methyl.


In some embodiments, the pharmaceutical composition includes a gel vehicle. In some embodiments, the pharmaceutical composition includes one or more of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), diethylene glycol monoethyl ether (Transcutol P), glycerin, hydroxyethyl cellulose, methylparaben, propylparaben, disodium EDTA, sodium metabisulfite and water. In certain embodiments, the pharmaceutical composition includes at least 0.00007% mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288), at least 0.0003801% DOPC, at least 25% diethylene glycol monoethyl ether (Transcutol P), at least 5% glycerin, at least 1% hydroxyethyl cellulose, at least 0.15% methylparaben, at least 0.05% propylparaben, at least 0.1% disodium EDTA, at least 0.2% sodium metabisulfite, and at least 68.5% water.


In some embodiments, the pharmaceutical composition includes at least 0.007% mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288), at least 0.03801% DOPC, at least 25% diethylene glycol monoethyl ether (Transcutol P), at least 5% glycerin, at least 1% hydroxyethyl cellulose, at least 0.15% methylparaben, at least 0.05% propylparaben, at least 0.1% disodium EDTA, at least 0.2% sodium metabisulfite, and at least 68.45% water.


In some embodiments, the pharmaceutical composition comprises at least 0.01419% mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288), at least 0.2655% DOPC, at least 25% diethylene glycol monoethyl ether (Transcutol P), at least 5% glycerin, at least 1% hydroxyethyl cellulose, at least 0.15% methylparaben, at least 0.05% propylparaben, at least 0.1% disodium EDTA, at least 0.2% sodium metabisulfite, and at least 68.2% water.


In some embodiments, the pharmaceutical composition comprises at least 0.1419% mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288), at least 2.655% DOPC, at least 25% diethylene glycol monoethyl ether (Transcutol P), at least 5% glycerin, at least 1% hydroxyethyl cellulose, at least 0.15% methylparaben, at least 0.05% propylparaben, at least 0.1% disodium EDTA, at least 0.2% sodium metabisulfite, and at least 65.7% water.


In some embodiments, the pharmaceutical composition comprises at least 1.419% mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288), at least 26.55% DOPC, at least 25% diethylene glycol monoethyl ether (Transcutol P), at least 5% glycerin, at least 1% hydroxyethyl cellulose, at least 0.15% methylparaben, at least 0.05% propylparaben, at least 0.1% disodium EDTA, at least 0.2% sodium metabisulfite, and at least 40.5% water.


In some embodiments, the pharmaceutical composition includes an oligonucleotide described herein.


In some embodiments, the pharmaceutical composition further includes a gel vehicle. In some embodiments, the pharmaceutical composition includes one or more of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), diethylene glycol monoethyl ether (Transcutol P), glycerin, hydroxyethyl cellulose, methylparaben, propylparaben, disodium EDTA, sodium metabisulfite and water.


In some embodiments, the pharmaceutical composition includes a mASO-SNA described herein.


In some embodiments, the pharmaceutical composition further includes a gel vehicle. In some embodiments, the pharmaceutical composition comprises one or more of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), diethylene glycol monoethyl ether (Transcutol P), glycerin, hydroxyethyl cellulose, methylparaben, propylparaben, disodium EDTA, sodium metabisulfite and water.


According to another aspect, methods for treating a disorder using a pharmaceutical composition are also disclosed herein. In some embodiments, a method for treating a disorder comprises administering to a subject having a disorder a pharmaceutical composition including a multiplex antisense oligonucleotide spherical nucleic acid (mASO-SNA), including a core having an oligonucleotide shell comprised of an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a first gene and an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a second gene, wherein the core is a solid surrounded by a lipid bilayer or a liposome or lipoplex complex core and the oligonucleotide shell is positioned on the exterior of the core in an effective amount to treat the disorder.


In some embodiments, the pharmaceutical composition further comprises a gel vehicle. In some embodiments, the pharmaceutical composition comprises one or more of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), diethylene glycol monoethyl ether (Transcutol P), glycerin, hydroxyethyl cellulose, methylparaben, propylparaben, disodium EDTA, sodium metabisulfite and water.


In some embodiments, the disorder is an inflammatory disorder.


In some embodiments, the mASO-SNA produces simultaneous mRNA knock-down of the first and second gene. In some embodiments, the first gene and the second gene are associated with a target pathway. In some embodiments, the mASO-SNA produces additive knock-down of the target pathway.


In some embodiments, the disorder is psoriasis.


According to another aspect, methods for treating an inflammatory disorder using a pharmaceutical composition are also disclosed herein. In some embodiments, a method for treating an inflammatory disorder includes administering to a subject having an inflammatory disorder a pharmaceutical composition including an oligonucleotide, a nanostructure or a mASO-SNA described herein in an effective amount to treat the inflammatory disorder.


In some embodiments, the pharmaceutical composition further includes a gel vehicle. In some embodiments, the pharmaceutical composition includes one or more of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), diethylene glycol monoethyl ether (Transcutol P), glycerin, hydroxyethyl cellulose, methylparaben, propylparaben, disodium EDTA, sodium metabisulfite and water.


In some embodiments, the inflammatory disorder is selected from the group consisting of an autoimmune disease, an infectious disease, transplant rejection or graft-versus-host disease, malignancy, a pulmonary disorder, an intestinal disorder, a cardiac disorder, sepsis, a spondyloarthropathy, a metabolic disorder, anemia, pain, a hepatic disorder, a skin disorder, a nail disorder, rheumatoid arthritis, psoriasis, psoriasis in combination with psoriatic arthritis, ulcerative colitis, Crohn's disease, vasculitis, Behcet's disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disorder (COPD), idiopathic pulmonary fibrosis (IPF), restenosis, diabetes, anemia, pain, a Crohn's disease-related disorder, juvenile rheumatoid arthritis (JRA), a hepatitis C virus infection, psoriatic arthritis, and chronic plaque psoriasis.


In some embodiments, the inflammatory disorder is selected from the group consisting of rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis, and nephritic syndrome.


According to another aspect, methods for reducing expression of the IL-17 receptor in vivo are provided herein. In some embodiments, the method for reducing expression levels of IL-17 receptor in vivo includes administering to a subject a pharmaceutical composition including an oligonucleotide, a nanostructure, or a mASO-SNA described herein in an effective amount to reduce IL-17 receptor levels in vivo.


In some embodiments, the subject is a mammal. In some embodiments, the subject is human.


In some embodiments, the pharmaceutical composition is in contact with a cell in the subject for at least 24 hours.


In some embodiments, the pharmaceutical composition is a topical pharmaceutical composition.


According to another aspect, methods for reducing expression levels of IL-17 receptor (IL-17R) in vitro are provided herein.


In some embodiments, the method for reducing expression levels of IL-17R in vitro includes contacting a cell with an oligonucleotide, a nanostructure, or a mASO-SNA described herein in an effective amount to reduce IL-17R levels in vitro.


In some embodiments, the cell is a human foreskin keratinocyte (HFK).


In some embodiments, the cell is contacted with the oligonucleotide, the nanostructure, or the mASO-SNA at a concentration of 1 nM, 10 nM, 100 nM, or 1000 nM.


In some embodiments, the cell is contacted with the oligonucleotide, the nanostructure, or the mASO-SNA for 24 hours.


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.





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 depicts in vitro IL-17RA mRNA knockdown using targeted SNAs. IL-17RA mRNA expression is decreased in HFK cells treated with SNAs presenting an ASO sequence targeting IL-17RA. Cells treated with control SNAs do not demonstrate an appreciable decrease in IL-17RA mRNA expression.



FIG. 2 shows IL-17RA mRNA knockdown using topically applied targeted SNAs in the human skin equivalent, EpiDerm-FT™. IL-17RA mRNA expression is decreased in human skin equivalent tissues treated topically with SNAs presenting either ASO IL17RA_219 or ASO IL17RA_282 for 48 hours. Tissues treated with SNAs presenting the control ASO do not demonstrate an appreciable decrease in IL-17RA mRNA expression as measured by qRT-PCR. Data shown are representative of two independent experiments performed.



FIG. 3 shows ex vivo IL-17RA mRNA knockdown using topically applied targeted SNAs in human skin explants. IL-17RA mRNA expression is decreased in human skin explant biopsies treated topically with SNAs presenting either ASO IL17RA_219 or ASO IL17RA_282 for 96 hours. Biopsies treated with SNAs presenting the control ASO do not demonstrate an appreciable decrease in IL-17RA mRNA expression as measured by qRT-PCR.



FIG. 4 shows TNF mRNA knockdown using a mixture of SNAs. TNF mRNA expression is decreased in HFK cells treated with a mixture of anti-TNF-SNAs and anti-IL17RA-SNAs. Cells treated with Control-SNAs do not demonstrate an appreciable decrease in TNF mRNA levels. Data shown are representative of two independent experiments performed.



FIG. 5 shows IL17RA mRNA knockdown using a mixture of SNAs. IL17RA mRNA expression is decreased in HFK cells treated with a mixture of anti-TNF-SNAs and anti-IL17RA-SNAs. Cells treated with Control-SNAs do not demonstrate an appreciable decrease in TNF mRNA levels. Data shown are representative of two independent experiments performed.



FIGS. 6A-6D show configurations of multiplexed SNAs. Liposomes were surface-functionalized in the following configurations: (FIG. 6A) 33% TNF-ASO (A) and 66% control (D); (FIG. 6B) 33% IL17RA-ASO (B) and 66% control (D); (FIG. 6C) 33% TNF-ASO (A), 33% IL17RA-ASO (B), and 33% control (D); (FIG. 6D) 100% control (D).



FIG. 7 shows a comparison of TNF mRNA knockdown using multiplexed SNAs. TNF mRNA expression is decreased in HFK cells treated with anti-TNF-SNAs and multiplexed SNAs containing anti-TNF and anti-IL17RA ASOs. Cells treated with anti-IL17RA-SNAs do not demonstrate an appreciable decrease in TNF mRNA levels. Data shown are representative of two independent experiments performed.



FIG. 8 shows a comparison of IL17RA mRNA knockdown using multiplexed SNAs demonstrate additive knockdown effects. IL17RA mRNA expression is decreased in HFK cells treated with anti-IL17RA-SNAs and multiplexed SNAs containing anti-TNF and anti-IL17RA ASOs. Cells treated with anti-TNF-SNAs demonstrate a slight decrease potentially due to the related biological signaling pathways of TNF and IL17RA. Greater IL17RA mRNA knockdown is observed when cells are treated with anti-IL17RA-SNAs. Multiplexed SNAs targeting both TNF and IL17RA have an additive effect and show greater knockdown of IL17RA mRNA than when SNAs targeting only anti-TNF or anti-IL17RA are used alone. Data shown are representative of at least two independent experiments performed.



FIG. 9 depicts inhibition of IL-17RA mRNA expression. The graph shows IL-17RA mRNA expression in healthy human skin explants after 24 hours treatment, as measured by qRT-PCR (percent of untreated ±SEM). Each drug product treatment group, n=9 samples. Untreated group, n=8 samples. One-way ANOVA, 31 DF, p=0.0011 (0.007% versus untreated) and **** p<0.0001 versus 0% gel.



FIG. 10 depicts inhibition of IL-17RA protein expression. The graph shows IL-17RA protein expression in healthy human skin explants after 24 hours treatment with IL17RA_282 in formulation described in Table 3, as measured by western blot (percent of untreated). Pooled samples for each group.



FIG. 11 shows inhibition of IL-17RA mRNA expression. * p<0.05; ** p<0.01; **** p<0.0001 compared to vehicle. A one sample t-test was used.



FIGS. 12A-12D show configurations of multiplexed SNAs. Liposome were surface functionalized in the following configurations: monoplex SNA (FIG. 12A); multiplex SNA containing two targeted antisense oligonucleotides (FIG. 12B); multiplex SNA containing three targeted antisense oligonucleotides (FIG. 12C); non-targeted control SNA (FIG. 12D).



FIG. 13 shows inhibition of IL4R mRNA in cells treated with various multiplex SNA configurations. Liposome were surface functionalized various targeted antisense oligonucleotide(s) or control oligonucleotide. The expression of IL4R mRNA is reduced noticeably when cells are treated with SNAs that target IL4R. Control SNA and SNAs functionalized with non-targeting oligonucleotides do not reduce IL4R mRNA expression to the same extent.



FIG. 14 shows inhibition of IL1b mRNA in cells treated with various multiplex SNA configurations. Liposome were surface functionalized various targeted antisense oligonucleotide(s) or control oligonucleotide. The expression of IL1b mRNA is reduced noticeably when cells are treated with SNAs that target IL1b. Control SNA and SNAs functionalized with non-targeting oligonucleotides do not reduce IL1b mRNA expression to the same extent.



FIG. 15 shows inhibition of CTGF mRNA in cells treated with various multiplex SNA configurations. Liposome were surface functionalized with various targeted antisense oligonucleotide(s) or control oligonucleotide. The expression of CTGF mRNA is reduced noticeably when cells are treated with SNAs that target CTGF. Control SNA and SNAs functionalized with non-targeting oligonucleotides do not reduce CTGF mRNA expression to the same extent.





DETAILED DESCRIPTION

The invention in some aspects relates to antisense inhibitors of a cell signaling protein receptor involved in systemic inflammation, Interleukin 17 receptor (IL-17RA). IL-17 and its target receptor IL-17RA play an important role in psoriasis. IL-17 elicits its effects by binding IL-17RA, the latter of which is expressed almost exclusively in keratinocytes within the epidermis. Antisense technology is a useful means for reducing the expression of specific gene products by targeting a messenger RNA (mRNA) and preventing translation of the mRNA. However, the selection of specific therapeutically functional antisense oligonucleotides (ASOs) can be challenging. Further, ASO that are therapeutically active in a linear form do not necessarily retain activity when formulated as a nanoparticle or other type of three dimensional presentation format.


It has been discovered in aspects of the invention that Spherical Nucleic Acids (SNAs) surface functionalized with antisense oligonucleotides (ASOs) having appropriate structural properties can mediate highly effective gene knockdown of the intended target mRNA. For example, it is demonstrated herein that IL-17RA expression can be inhibited through the use of SNAs targeting human IL-17RA (anti-IL17RA-SNAs) (see for instance GeneBank accession number NM_014339.6) mRNA in primary human foreskin keratinocytes (HFK) with no associated toxicity or immune-stimulatory effects. Further, it is shown that anti-IL-17RA SNAs inhibit mRNA expression in a human skin equivalent model and human skin explants. In some embodiments, the response the anti-IL-17RA SNAs inhibit IL-17RA mRNA expression in a dose-dependent manner. As described herein, the oligonucleotides arranged in an SNA geometry exhibit enhanced penetration and increased cellular uptake.


It has also been discovered that multiplex SNAs having different ASOs directed at different therapeutic targets can produce significant target knockdown and therapeutic effects. The ability to load specific amounts of different ASOs on the surface of an SNA in a way such that they do not interfere with the activity of the other ASOs was unexpected. As shown in the Examples, when anti-IL-17RA-SNAs were mixed with anti-TNF-SNAs and co-administered to cells, simultaneous mRNA knockdown of each targeted gene was achieved. Additionally when SNAs presenting two or more ASO sequences each targeting different mRNAs on the same particle were administered to cells the simultaneous mRNA knockdown of each gene targeted using a single particle in the same cell was achieved. These SNAs are referred to as multiplexed ASO-SNAs (mASO-SNA). Furthermore, when mASO-SNAs were multiplexed with ASOs targeting two mRNAs of genes involved in a related biological pathway, additive knockdown effects were observed. Not only did the co-presentation of the two ASOs not interfere with the activity of the other co-presented ASO but when targeting a common pathway the therapeutic effect observed with the mASO-SNAs was greater than that observed with either ASO-SNA alone.


The invention in some aspects relates to compositions for reducing IL-17RA and methods for treating an inflammatory disorder using those compositions. Highly effective IL-17RA inhibitors have been identified according to aspects of the invention. The IL-17RA inhibitors are nucleic acid based antisense compositions. The term “IL-17RA” refers to a receptor for the cytokine IL-17.


An “IL-17RA inhibitor” as used herein refers to a nucleic acid based agent which interferes with IL-17RA activity. In particular, the IL-17RA antisense inhibitors or IL-17RA antisense oligonucleotides of the invention reduce the expression of the IL-17RA gene. By reducing expression of the IL-17RA gene the endogenous IL-17 is not able to promote cell signaling through its interaction with the receptor. When IL-17RA is available for binding to IL-17, IL17RA is activated leading to induction of expression of inflammatory chemokines and cytokines such as CXCL1, CXCL8/IL8 and IL6. These signaling cascades can be blocked using the ASO-SNA of the invention.


The IL17RA inhibitors of the invention are antisense nucleic acids. Antisense nucleic acids typically 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 acids bind to target RNA by Watson Crick base-pairing and block 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.


In some embodiments, the invention relates to compositions for reducing interleukin-4 receptor (IL-4R) (see e.g., GenBank: NM_000418 (SEQ ID NO: 306)) and methods for treating an inflammatory disorder using those compositions. Highly effective IL-4R inhibitors have been identified according to aspects of the invention. The IL-4R inhibitors are nucleic acid based antisense compositions. The term “IL-4R” refers to a receptor for the cytokine IL-4 or IL-13.


An “IL-4R inhibitor” as used herein refers to a nucleic acid based agent which interferes with IL-4R activity. In particular, the IL-4R antisense inhibitors or IL-4R antisense oligonucleotides of the invention reduce the expression of the IL-4R gene. In some embodiments, the antisense oligonucleotide is 18 nucleotides in length. In other embodiments, IL-17RA has a sequence of SEQ ID NO: 302. By reducing expression of the IL-4R gene the endogenous IL-4 or IL-13 is not able to promote cell signaling through its interaction with the receptor. When IL-4R is available for binding to IL-4 or IL-13, IL-4R is activated leading to coupling to the JAK1/2/3-STAT6 pathway. The IL4 response is involved in promoting Th2 differentiation. The IL-4/IL-13 responses are involved in regulating IgE production and, chemokine and mucus production at sites of allergic inflammation. In certain cell types, can signal through activation of insulin receptor substrates, IRS1/IRS2. Soluble IL4R (sIL4R) inhibits IL4-mediated cell proliferation and IL5 up-regulation by T-cells (Keegan et al., Cell (1994) 76:811-820). These signaling cascades can be blocked using the ASO-SNA of the invention.


The IL-4R inhibitors of the invention are antisense nucleic acids. Antisense nucleic acids typically 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 acids bind to target RNA by Watson Crick base-pairing and block 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.


In some embodiments, the invention relates to compositions for reducing interleukin-1 beta (IL-1β) (see e.g., GenBank: NM_000576 (SEQ ID NO: 307)) and methods for treating an inflammatory disorder using those compositions. Highly effective IL-1β inhibitors have been identified according to aspects of the invention. The IL-1β inhibitors are nucleic acid based antisense compositions. The term “IL-1β” refers to a proinflammatory cytokine that was initially discovered as the major endogenous pyrogen, induces prostaglandin synthesis, neutrophil influx and activation, T-cell activation and cytokine production, B-cell activation and antibody production, and fibroblast proliferation and collagen production. Promotes Th17 differentiation of T-cells.


An “IL-1β inhibitor” as used herein refers to a nucleic acid based agent which interferes with IL-1β activity. In particular, the IL-1β antisense inhibitors or IL-1β antisense oligonucleotides of the invention reduce the expression of the IL-1β gene. By reducing expression of the IL-1β gene, signaling is reduced or not carried out (Van Damme et al., Nature (1985) 314:266-268; Piccioli et al., Semin Immunol (2013) 25(6):425-9). The signaling cascades that involve IL-1 β can be blocked using the ASO-SNA of the invention.


The IL-1β inhibitors of the invention are antisense nucleic acids. Antisense nucleic acids typically 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 acids bind to target RNA by Watson Crick base-pairing and block 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.


In some embodiments, the invention relates to compositions for reducing connective tissue growth factor (CTGF) (see e.g., GenBank: NM_001901 (SEQ ID NO: 308)) and methods for treating a disorder (e.g., atherosclerosis, pulmonary and renal fibrotic disorders and cancer) using those compositions. Highly effective CTGF inhibitors have been identified according to aspects of the invention. The CTGF inhibitors are nucleic acid based antisense compositions. The term “CTGF” refers to a connective tissue mitoattractant secreted by cells (e.g., vascular endothelial cells) that, among other things, promotes proliferation and differentiation of chondrocytes; mediates heparin- and divalent cation-dependent cell adhesion in many cell types including fibroblasts, myofibroblasts, endothelial and epithelial cells; and enhances fibroblast growth factor-induced DNA synthesis.


An “CTGF inhibitor” as used herein refers to a nucleic acid based agent which interferes with CTGF activity. In particular, the CTGF antisense inhibitors or CTGF antisense oligonucleotides of the invention reduce the expression of the CTGF gene. By reducing expression of the CTGF gene, cell signaling is not promoted. For instance, CTGF interacts with growth factors, surface receptors and matrix components, with important roles in embryonic development and the maintenance of normal cell and connective tissue function, due to its widespread expression. CTGF is also important for tissue repair following injury, and has been implicated in common diseases including atherosclerosis, pulmonary and renal fibrotic disorders and cancer (de Winter et al., Growth Factors (2008) 26(2):80-91). The signaling cascades that CTGF is involved in can be blocked using the ASO-SNA of the invention.


The CTGF inhibitors of the invention are antisense nucleic acids. Antisense nucleic acids typically 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 acids bind to target RNA by Watson Crick base-pairing and block 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” or “antisense oligonucleotide” describes a nucleic acid that hybridizes under physiological conditions to DNA comprising a particular gene or to an mRNA transcript of that gene in this case IL17RA 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.


“Inhibition of gene expression” refers to the absence or observable decrease in the level of protein and/or mRNA product from a target gene, such as the IL17RA 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).


The antisense oligonucleotides of the invention inhibit IL17RA expression. Depending on the assay, quantitation of the amount of gene expression allows one to determine a degree of inhibition which is greater than 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 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.


The ASOs described herein include bioequivalent compounds, salts and prodrugs thereof. The term bioequivalent compounds, including pharmaceutically acceptable salts and prodrugs as used herein refers to antisense oligonucleotides having the same primary structure as the antisense oligonucleotide of interest, but including salt forms or structures which can be cleaved or modified to have the same type of biological effect as the antisense oligonucleotide of interest. This is intended to encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof.


“Pharmaceutically acceptable salts” are physiologically and pharmaceutically acceptable salts of the nucleic acids of the invention: i.e., salts that retain the desired biological activity of the compound of interest and do not impart undesired toxicological effects thereto. Pharmaceutically acceptable salts include but are not limited to (a) salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc.; (b) acid addition salts formed with inorganic acids, for example hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; (c) salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (d) salts formed from elemental anions such as chlorine, bromine, and iodine.


The compounds of the invention may also be prepared to be delivered in a “prodrug” form. A “prodrug” is a therapeutic agent that is prepared in an inactive form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions.


The antisense oligonucleotides of the invention are IL17RA antisense oligonucleotides. An antisense IL17RA oligonucleotide refers to a compound having a sequence of nucleotide bases and a subunit-to-subunit backbone that allows the antisense oligonucleotide to hybridize to a IL17RA target mRNA sequence typically by Watson-Crick base pairing, to form an RNA:oligomer heteroduplex within the target sequence.


The specific hybridization of an antisense oligonucleotide with its target nucleic acid, IL17RA mRNA, interferes with the normal function of the IL17RA mRNA. This modulation of function of a target nucleic acid by compounds which specifically hybridize to it is generally referred to as “antisense”. The functions of DNA to be interfered with include replication and transcription. The functions of RNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in or facilitated by the RNA. The overall effect of such interference with target nucleic acid function is modulation of the expression of IL17RA protein. In the context of the present invention, “modulation” means a decrease or inhibition in the expression of a gene.


An antisense oligonucleotide “specifically hybridizes” to a target polynucleotide if the oligonucleotide hybridizes to the IL17RA target under physiological conditions, with a thermal melting temperature (Tm) substantially greater than 37° C., preferably at least 45° C., and typically 50° C.-80° C. or higher. Such hybridization preferably corresponds to stringent hybridization conditions, selected to be about 10° C., and preferably about 50° C. lower than the Tm for the specific sequence at a defined ionic strength and pH. At a given ionic strength and pH, the Tm is the temperature at which 50% of a target sequence hybridizes to a complementary polynucleotide.


Polynucleotides are described as “complementary” to one another when hybridization occurs in an antiparallel configuration between two single-stranded polynucleotides. A double-stranded polynucleotide can be “complementary” to another polynucleotide, if hybridization can occur between one of the strands of the first polynucleotide and the second. Complementarity (the degree that one polynucleotide is complementary with another) is quantifiable in terms of the proportion of bases in opposing strands that are expected to form hydrogen bonds with each other, according to generally accepted base-pairing rules. An antisense compound may be complementary to a target region of a target transcript even if the two bases sequences are not 100% complementary, as long as the heteroduplex structure formed between the compound and transcript has the desired Tm stability.


Identifying an antisense oligonucleotide that targets a particular nucleic acid may be a multistep process. The process usually begins with the identification of a nucleic acid sequence whose function is to be modulated. This may be, for example, a cellular gene (or mRNA transcribed from the gene) whose expression is associated with a particular inflammatory disorder or disease state. The targeting process also includes determination of a site or sites within this IL17RA gene for the antisense interaction to occur such that the desired effect, e.g., detection or modulation of expression of the protein, will result.


In some embodiments, antisense oligonucleotides are designed to target human Interleukin 17 receptor (IL-17RA), for instance, the nucleotide sequence of SEQ ID NO: 302, corresponding to interleukin 17 receptor A (IL-17RA), set forth below. Human IL-17RA cDNA sequence has been disclosed in Genbank accession number NM_014339.










(SEQ ID NO: 302)










1
ctgggcccgg gctggaagcc ggaagcgagc aaagtggagc cgactcgaac tccaccgcgg






61
aaaagaaagc ctcagaacgt tcgttcgctg cgtccccagc cggggccgag ccctccgcga





121
cgccagccgg gccatggggg ccgcacgcag cccgccgtcc gctgtcccgg ggcccctgct





181
ggggctgctc ctgctgctcc tgggcgtgct ggccccgggt ggcgcctccc tgcgactcct





241
ggaccaccgg gcgctggtct gctcccagcc ggggctaaac tgcacggtca agaatagtac





301
ctgcctggat gacagctgga ttcaccctcg aaacctgacc ccctcctccc caaaggacct





361
gcagatccag ctgcactttg cccacaccca acaaggagac ctgttccccg tggctcacat





421
cgaatggaca ctgcagacag acgccagcat cctgtacctc gagggtgcag agttatctgt





481
cctgcagctg aacaccaatg aacgtttgtg cgtcaggttt gagtttctgt ccaaactgag





541
gcatcaccac aggcggtggc gttttacctt cagccacttt gtggttgacc ctgaccagga





601
atatgaggtg accgttcacc acctgcccaa gcccatccct gatggggacc caaaccacca





661
gtccaagaat ttccttgtgc ctgactgtga gcacgccagg atgaaggtaa ccacgccatg





721
catgagctca ggcagcctgt gggaccccaa catcaccgtg gagaccctgg aggcccacca





781
gctgcgtgtg agcttcaccc tgtggaacga atctacccat taccagatcc tgctgaccag





841
ttttccgcac atggagaacc acagttgctt tgagcacatg caccacatac ctgcgcccag





901
accagaagag ttccaccagc gatccaacgt cacactcact ctacgcaacc ttaaagggtg





961
ctgtcgccac caagtgcaga tccagccctt cttcagcagc tgcctcaatg actgcctcag





1021
acactccgcg actgtttcct gcccagaaat gccagacact ccagaaccaa ttccggacta





1081
catgcccctg tgggtgtact ggttcatcac gggcatctcc atcctgctgg tgggctccgt





1141
catcctgctc atcgtctgca tgacctggag gctagctggg cctggaagtg aaaaatacag





1201
tgatgacacc aaatacaccg atggcctgcc tgcggctgac ctgatccccc caccgctgaa





1261
gcccaggaag gtctggatca tctactcagc cgaccacccc ctctacgtgg acgtggtcct





1321
gaaattcgcc cagttcctgc tcaccgcctg cggcacggaa gtggccctgg acctgctgga





1381
agagcaggcc atctcggagg caggagtcat gacctgggtg ggccgtcaga agcaggagat





1441
ggtggagagc aactctaaga tcatcgtcct gtgctcccgc ggcacgcgcg ccaagtggca





1501
ggcgctcctg ggccgggggg cgcctgtgcg gctgcgctgc gaccacggaa agcccgtggg





1561
ggacctgttc actgcagcca tgaacatgat cctcccggac ttcaagaggc cagcctgctt





1621
cggcacctac gtagtctgct acttcagcga ggtcagctgt gacggcgacg tccccgacct





1681
gttcggcgcg gcgccgcggt acccgctcat ggacaggttc gaggaggtgt acttccgcat





1741
ccaggacctg gagatgttcc agccgggccg catgcaccgc gtaggggagc tgtcggggga





1801
caactacctg cggagcccgg gcggcaggca gctccgcgcc gccctggaca ggttccggga





1861
ctggcaggtc cgctgtcccg actggttcga atgtgagaac ctctactcag cagatgacca





1921
ggatgccccg tccctggacg aagaggtgtt tgaggagcca ctgctgcctc cgggaaccgg





1981
catcgtgaag cgggcgcccc tggtgcgcga gcctggctcc caggcctgcc tggccataga





2041
cccgctggtc ggggaggaag gaggagcagc agtggcaaag ctggaacctc acctgcagcc





2101
ccggggtcag ccagcgccgc agcccctcca caccctggtg ctcgccgcag aggagggggc





2161
cctggtggcc gcggtggagc ctgggcccct ggctgacggt gccgcagtcc ggctggcact





2221
ggcgggggag ggcgaggcct gcccgctgct gggcagcccg ggcgctgggc gaaatagcgt





2281
cctcttcctc cccgtggacc ccgaggactc gccccttggc agcagcaccc ccatggcgtc





2341
tcctgacctc cttccagagg acgtgaggga gcacctcgaa ggcttgatgc tctcgctctt





2401
cgagcagagt ctgagctgcc aggcccaggg gggctgcagt agacccgcca tggtcctcac





2461
agacccacac acgccctacg aggaggagca gcggcagtca gtgcagtctg accagggcta





2521
catctccagg agctccccgc agccccccga gggactcacg gaaatggagg aagaggagga





2581
agaggagcag gacccaggga agccggccct gccactctct cccgaggacc tggagagcct





2641
gaggagcctc cagcggcagc tgcttttccg ccagctgcag aagaactcgg gctgggacac





2701
gatggggtca gagtcagagg ggcccagtgc atgagggcgg ctccccaggg accgcccaga





2761
tcccagcttt gagagaggag tgtgtgtgca cgtattcatc tgtgtgtaca tgtctgcatg





2821
tgtatatgtt cgtgtgtgaa atgtaggctt taaaatgtaa atgtctggat tttaatccca





2881
ggcatccctc ctaacttttc tttgtgcagc ggtctggtta tcgtctatcc ccaggggaat





2941
ccacacagcc cgctcccagg agctaatggt agagcgtcct tgaggctcca ttattcgttc





3001
attcagcatt tattgtgcac ctactatgtg gcgggcattt gggataccaa gataaattgc





3061
atgcggcatg gccccagcca tgaaggaact taaccgctag tgccgaggac acgttaaacg





3121
aacaggatgg gccgggcacg gtggctcacg cctgtaatcc cagcacactg ggaggccgag





3181
gcaggtggat cactctgagg tcaggagttt gagccagcct ggccaacatg gtgaaacccc





3241
atctccacta aaaatagaaa aattagccgg gcatggtgac acatgcctgt agtcctagct





3301
acttgggagg ctgaggcagg agaattgctt gaatctggga ggcagaggtt gcagtgagcc





3361
gagattgtgc cattgcactg cagcctggat gacagagcga gactctatct caaaaaaaaa





3421
aaaaaaaaaa gatggtcacg cgggatgtaa acgctgaatg ggccaggtgc agtggctcat





3481
gcttgtaatc ccagcacttt gggaaggcga ggcaggtgga ttgcttgagc tcaggagttc





3541
aagaccagcc tgggcgacat agtgagacct catctctacc taaatttttt tttagtcagt





3601
catggtggca catgcctgta gtcccagcta ctcgggaggc tgatgccaga tgatcacttg





3661
agcccaggag gtagaggctg cagtgagcta taatggtacc attgcaatcc agcctgggca





3721
gcagagtgag accctgtctc aaaaaaaata aaaaagtaga aagatggagt ggaagcctgc





3781
ccagggttgt gagcatgcac gggaaaggca cccaggtcag gggggatccc cgaggagatg





3841
cctgagctga aggattgtgg ttggggaaag cgtagtccca gcaaggaagc agtttgtggg





3901
taagtgctgg gaggtgagtg gagtgagctt gtcagggagc tgctggtgga gcctggaggg





3961
gaaggaggga ggcagtgaga gagatcgggg tggggggtgg ggggatgtcg ccagagctca





4021
ggggtgggga cagccttgtg cgcatcagtc ctgaggcctg gggcaccttt cgtctgatga





4081
gcctctgcat ggagagaggc tgagggctaa acacagctgg atgtcacctg agttcattta





4141
taggaagaga gaaatgtcga ggtgaaacgt aaaagcatct ggcaggaagg tgagtctgaa





4201
gccctgcacc cgcgttccga ctatcagtgg ggagctgtta gcacgtagga ttcttcagag





4261
cagctgggct ggagctcccc tgagctcagg aagccccagg gtgcaagggc aaggaaatga





4321
ggggtggtgg gtcagtgaag atctgggcag accttgtgtg gggaaggggt gctgctgtga





4381
cttcagggtc tgaggtccaa agacagcatt tgaaaagagg ctctgaagcc agtgtttgaa





4441
gaatttgttc ctgaagtacc tcctgggggt aggctagagg cttctggctt cagggtcctg





4501
aagaacacat tgaggtgccg tctgacactg gaatagggtg cccttcattc ctatgcctga





4561
gtccttaact atatttccaa cctccagtga ggaggagaag attcggaaat gtgacaggag





4621
agcaaacagg acagtttgca tgtgtgtgtg cgcacacata catgtgcgtg aaagattatc





4681
aataaaagtg cataaatttg ttgatctggt aagagtttct agcaggaagg tcgagccact





4741
tactgtaggt caagaagttg ctagttgcgg agttttttct tgcagttaga ctttacctag





4801
tggtagcagg gccaccaaag ctctgtgtcc cagatggtgt atggcccata atccacccaa





4861
cagcagcaaa ggaccaggca aaggagaaca ggagcagaag cctcccagcc actagccttt





4921
tgggctcagt ctctccaata atcctggaga ggggcttcgt tgggtctgga cacctaccat





4981
gcattctgtg acctttccct agcttccaat aaataactgt ttgacgccca gagtacagga





5041
taccacaatg cactcttcct gcgtagagca catgttccca tctgctccca ttcctcagga





5101
accttgaatt ctagctctgc tggcctttga gcccatgcca gtaaatgtcc tgatgggcat





5161
tgcctactat ctccagggca gctgcctttg tcctcctaac agctttattg gagtacagtt





5221
cacttaccat acaatccaca attgaccctg cacaatttga tgccggttta gtatagtcac





5281
agttcagcag ccatcagcac agtcagtctt agagtttact acccccaaaa gaaatccagc





5341
cccccttagt caccacccca acctccccat ccctaggcac ccctaggcta ctttgatctc





5401
tgtagacttg cctcttctgg acatgacata gagaaaggag tcataaattc tccaaggtgt





5461
ctgtttcttc tttaatgtca ttccctgttt ctcctcacat tccctcccca tttcctgggc





5521
ccagtctcac actggtcctt gcttacccta aatgctatta attccatcac tctgagtatg





5581
gtgtttgctg tccgctgaat gccaagagct tcaagagtgt gtgtaaataa agccacacct





5641
ttatttttgt attattctga accatggcta ataaattgtt tcaccaagaa atgtctctct





5701
aagaacaggt gccctccacg ctgtgcccct cccacctctt cagctcgtct cctgagtgtg





5761
cagaggtggt tccggttggg aaagaagcag cggagcatct aaccatgcct gtgtccaggc





5821
cgattatgca cgcagccacc aacaagctcc caactcccgc gtagagtttc atgacttttt





5881
cctgcctact atcttgatcc tagttttttt tttgtttttt ttttttttaa ggaataatta





5941
ctttgattca aaaccagttt ctcttttctg cataggaagg tccttgaagg tgtttagggt





6001
ctaaaaaggg tggtgttcgg tctctgaaac atccattcag cagtttgagc tgggatctct





6061
gaatgcaagg gtatgatgga tatacttctt tcttgctttt gttgtgtttt ggttttttgt





6121
ttgtttttaa gtcagggtct ctctgtcacc aggctgtatt acagtggtgc aatcatggct





6181
cactgcagcc tcgacctccc aggctcaagc catctttcca cctcagcctg ccagtggcta





6241
gaactacagg cgtgcaccac tgtgcccggc taatttgtgt gtatatattt tgtagaaatg





6301
gggtttcacc atgttgtcca ggctggtcac gaactcctgg gctcaagcca tctgcccgcc





6361
tcatcctccc aaagtgctgg gattataggc ctgagcccac cgtgcctggc ctttcctgtt





6421
tatctttgaa aattaaatag ggcataagag agaagaagat gtacttacaa tgcagtgggt





6481
ggttttaact ctatagcctt tgggctctgt ggttggtgct ccccttccta aataaatgag





6541
gtgtatgcag ggccctcttc tgccttagcg ccctgccagc tgggactcca gcaaggcccg





6601
gggcacctga ggacagagtg agatggaggg ccgctgctcc agcagccggg cctgcatccc





6661
acaagtcaac tgtgtcggac agaggatcct tacaaagaag aggcagcagg gttgggggct





6721
ggccagctgc tcgtccgccc taggtagctt gctcatctgt aaagtgggtg gggcaggagt





6781
tcccacctca tggggtcctg gcaagcctgc agtatccccg agtggcacca gcctgcttct





6841
ggggcagagc agtttgtgcc ccctgaggta ccactgatcc tctttccctg ctattaggta





6901
ttgctctctt cctccggtgt ttgccttttc agattataga agtaatatgt gttcccatat





6961
ttggcgtctc tcaggagctc aggaagtact tggctgagtg aacatgtcca ttgtggaaaa





7021
atggcaacaa tatggattcc atgggtatat tttatagaag aatatgaaga aaagcagcta





7081
cccctaaacc cattgcacaa gctgttcatg ttaattctgt acccgacgct ttccccacgg





7141
ggcctcccct cactctgaaa tggcatccag gtccatcttg ccctccacct ctgcatggct





7201
ctccatgccc catcgcctct cccagatcct agcactgggt ccacactctc gccctgtcca





7261
tttaggttga tgaaagcagg cagtcacccg ggtgggccag tcttgcctgt gggaggaaca





7321
tgcagtctcc tgtctcatgg tttgaagtgt gccaggaagc ctggcccagc ccacctcccc





7381
ctggagtcct tcccaggagg aataacccct taggtcattg actataagat gagttcgctc





7441
actggatcct tcctctctga tgagacagga agaaggtaca cagtgaccag gtaggaggag





7501
gagagggagt agaaaggagg gatgcgggtg gctggtccct gcatttgcct gcttccctgc





7561
acgggtgtcc cactggccgc ctctgctcac cagtgtcatg ggattctctc agaagatgaa





7621
aacagcccct gcttttttgc tagaatggct gagctttcat ggaaaggaag ctggacccaa





7681
gcaacagccg actaccgaag gttgcctgga gcagtgcaga tgtgggagga agaagggcct





7741
tggtgcacac tggcttttct tcctgactgc aatgtggcat tgtgccagct acctcctctt





7801
tctcggcctc aggaaaatgg agagaaagca gccctgaagg tggctgtgac gagggaaggg





7861
gcagagggcc tgacagtcaa ccacgcgcta tattttcctg ttcttcctta gggcaagaac





7921
tgcatggcca gactcaggca aggcctaggt gtgggctggg cattgcctac acgtgaagag





7981
atcactccgc gtccctactg cacctgtcac aaagtgcctt ctgatatgcc tggcaaacca





8041
aaatcggtga gcgccagctt gcttccctag aagacatttc taaatattca taacatgctt





8101
gctcaaatca atcaccttat tttacatccg ctccagggag aaatgaagac atggtcctac





8161
gttgttctgt aattattttc tatgtaaatt ttgttccttg ttacaattat atatgtctta





8221
ggggaaagga ccatttcaca tgtgtcacct catgtgattc tcaccacagc cctgtgattg





8281
ctcctgtttt ataaataatg acatagttcc agttgatggc caaagccaca gctaacgaga





8341
ggcagagaga gctcaggctc ccaggagctt ccactctcag accttgcctc ccgggctgcc





8401
ctgagtgaaa cgcctgctta gcatttggca cagccagaag cagcaagcta gggtcacaac





8461
acagagaggg gctgtgtaat actggctgcc tctgtgctaa gaaaaaaaaa aaatcactgt





8521
gtgtttgttt attttggtgc aggcccagtg ttcttgctta gacttaatac tacccttcat





8581
gttaaaataa aaccaaacaa aaacccat






In some embodiments the IL-17RA ASO is complementary to a nucleic acid encoding the following protein sequence:









(SEQ ID NO: 305)


MGAARSPPSAVPGPLLGLLLLLLGVLAPGGASLRLLDHRALVCSQPGLNC





TVKNSTCLDDSWIHPRNLTPSSPKDLQIQLHFAHTQQGDLFPVAHIEWTL





QTDASILYLEGAELSVLQLNTNERLCVRFEFLSKLRHHHRRWRFTFSHFV





VDPDQEYEVTVHHLPKPIPDGDPNHQSKNFLVPDCEHARMKVTTPCMSSG





SLWDPNITVETLEAHQLRVSFTLWNESTHYQILLTSFPHMENHSCFEHMH





HIPAPRPEEFHQRSNVTLTLRNLKGCCRHQVQIQPFFSSCLNDCLRHSAT





VSCPEMPDTPEPIPDYMPLWVYWFITGISILLVGSVILLIVCMTWRLAGP





GSEKYSDDTKYTDGLPAADLIPPPLKPRKVWIIYSADHPLYVDVVLKFAQ





FLLTACGTEVALDLLEEQAISEAGVMTWVGRQKQEMVESNSKIIVLCSRG





TRAKWQALLGRGAPVRLRCDHGKPVGDLFTAAMNMILPDFKRPACFGTYV





VCYFSEVSCDGDVPDLFGAAPRYPLMDRFEEVYFRIQDLEMFQPGRMHRV





GELSGDNYLRSPGGRQLRAALDRFRDWQVRCPDWFECENLYSADDQDAPS





LDEEVFEEPLLPPGTGIVKRAPLVREPGSQACLAIDPLVGEEGGAAVAKL





EPHLQPRGQPAPQPLHTLVLAAEEGALVAAVEPGPLADGAAVRLALAGEG





EACPLLGSPGAGRNSVLFLPVDPEDSPLGSSTPMASPDLLPEDVREHLEG





LMLSLFEQSLSCQAQGGCSRPAMVLTDPHTPYEEEQRQSVQSDQGYISRS





SPQPPEGLTEMEEEEEEEQDPGKPALPLSPEDLESLRSLQRQLLFRQLQK





NSGWDTMGSESEGPSA.






In some embodiments the ASOs used herein include any one or more of the following IL-17RA ASOs:

    • mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA/isp18//isp18//3CholTEG/(SEQ ID NO: 225);
    • mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU/isp18//isp18//3CholTEG/(SEQ ID NO: 288);
    • mGmUmAmGmGmGmCmGmUG*T*G*T*G*G*mGmUmC/isp18//isp18//3CholTEG/(SEQ ID NO: 291); mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA (SEQ ID NO: 225);
    • mCmCmCmAmCmAmGG*G*G*C*A*T*mGmUmAmGmU (SEQ ID NO: 288);
    • mGmUmAmGmGmGmCmGmUG*T*G*T*G*G*mGmUmC (SEQ ID NO: 291); GCUUGGGCAGGTGGUGAA (SEQ ID NO: 225); CCCACAGGGGCATGUAGU (SEQ ID NO: 288); or GUAGGGCGUGTGTGGGUC (SEQ ID NO: 291) or salts thereof.


In some embodiments antisense oligonucleotides are designed to target the nucleotide sequence corresponding to IL-4R, set forth below. Human IL-4R cDNA sequence has been disclosed in Genbank accession number NM_000418.










(SEQ ID NO: 306)










1
gggtctccgc gcccaggaaa gccccgcgcg gcgcgggcca gggaagggcc acccaggggt






61
cccccacttc ccgcttgggc gcccggacgg cgaatggagc aggggcgcgc agataattaa





121
agatttacac acagctggaa gaaatcatag agaagccggg cgtggtggct catgcctata





181
atcccagcac ttttggaggc tgaggcgggc agatcacttg agatcaggag ttcgagacca





241
gcctggtgcc ttggcatctc ccaatggggt ggctttgctc tgggctcctg ttccctgtga





301
gctgcctggt cctgctgcag gtggcaagct ctgggaacat gaaggtcttg caggagccca





361
cctgcgtctc cgactacatg agcatctcta cttgcgagtg gaagatgaat ggtcccacca





421
attgcagcac cgagctccgc ctgttgtacc agctggtttt tctgctctcc gaagcccaca





481
cgtgtatccc tgagaacaac ggaggcgcgg ggtgcgtgtg ccacctgctc atggatgacg





541
tggtcagtgc ggataactat acactggacc tgtgggctgg gcagcagctg ctgtggaagg





601
gctccttcaa gcccagcgag catgtgaaac ccagggcccc aggaaacctg acagttcaca





661
ccaatgtctc cgacactctg ctgctgacct ggagcaaccc gtatccccct gacaattacc





721
tgtataatca tctcacctat gcagtcaaca tttggagtga aaacgacccg gcagatttca





781
gaatctataa cgtgacctac ctagaaccct ccctccgcat cgcagccagc accctgaagt





841
ctgggatttc ctacagggca cgggtgaggg cctgggctca gtgctataac accacctgga





901
gtgagtggag ccccagcacc aagtggcaca actcctacag ggagcccttc gagcagcacc





961
tcctgctggg cgtcagcgtt tcctgcattg tcatcctggc cgtctgcctg ttgtgctatg





1021
tcagcatcac caagattaag aaagaatggt gggatcagat tcccaaccca gcccgcagcc





1081
gcctcgtggc tataataatc caggatgctc aggggtcaca gtgggagaag cggtcccgag





1141
gccaggaacc agccaagtgc ccacactgga agaattgtct taccaagctc ttgccctgtt





1201
ttctggagca caacatgaaa agggatgaag atcctcacaa ggctgccaaa gagatgcctt





1261
tccagggctc tggaaaatca gcatggtgcc cagtggagat cagcaagaca gtcctctggc





1321
cagagagcat cagcgtggtg cgatgtgtgg agttgtttga ggccccggtg gagtgtgagg





1381
aggaggagga ggtagaggaa gaaaaaggga gcttctgtgc atcgcctgag agcagcaggg





1441
atgacttcca ggagggaagg gagggcattg tggcccggct aacagagagc ctgttcctgg





1501
acctgctcgg agaggagaat gggggctttt gccagcagga catgggggag tcatgccttc





1561
ttccaccttc gggaagtacg agtgctcaca tgccctggga tgagttccca agtgcagggc





1621
ccaaggaggc acctccctgg ggcaaggagc agcctctcca cctggagcca agtcctcctg





1681
ccagcccgac ccagagtcca gacaacctga cttgcacaga gacgcccctc gtcatcgcag





1741
gcaaccctgc ttaccgcagc ttcagcaact ccctgagcca gtcaccgtgt cccagagagc





1801
tgggtccaga cccactgctg gccagacacc tggaggaagt agaacccgag atgccctgtg





1861
tcccccagct ctctgagcca accactgtgc cccaacctga gccagaaacc tgggagcaga





1921
tcctccgccg aaatgtcctc cagcatgggg cagctgcagc ccccgtctcg gcccccacca





1981
gtggctatca ggagtttgta catgcggtgg agcagggtgg cacccaggcc agtgcggtgg





2041
tgggcttggg tcccccagga gaggctggtt acaaggcctt ctcaagcctg cttgccagca





2101
gtgctgtgtc cccagagaaa tgtgggtttg gggctagcag tggggaagag gggtataagc





2161
ctttccaaga cctcattcct ggctgccctg gggaccctgc cccagtccct gtccccttgt





2221
tcacctttgg actggacagg gagccacctc gcagtccgca gagctcacat ctcccaagca





2281
gctccccaga gcacctgggt ctggagccgg gggaaaaggt agaggacatg ccaaagcccc





2341
cacttcccca ggagcaggcc acagaccccc ttgtggacag cctgggcagt ggcattgtct





2401
actcagccct tacctgccac ctgtgcggcc acctgaaaca gtgtcatggc caggaggatg





2461
gtggccagac ccctgtcatg gccagtcctt gctgtggctg ctgctgtgga gacaggtcct





2521
cgccccctac aacccccctg agggccccag acccctctcc aggtggggtt ccactggagg





2581
ccagtctgtg tccggcctcc ctggcaccct cgggcatctc agagaagagt aaatcctcat





2641
catccttcca tcctgcccct ggcaatgctc agagctcaag ccagaccccc aaaatcgtga





2701
actttgtctc cgtgggaccc acatacatga gggtctctta ggtgcatgtc ctcttgttgc





2761
tgagtctgca gatgaggact agggcttatc catgcctggg aaatgccacc tcctggaagg





2821
cagccaggct ggcagatttc caaaagactt gaagaaccat ggtatgaagg tgattggccc





2881
cactgacgtt ggcctaacac tgggctgcag agactggacc ccgcccagca ttgggctggg





2941
ctcgccacat cccatgagag tagagggcac tgggtcgccg tgccccacgg caggcccctg





3001
caggaaaact gaggcccttg ggcacctcga cttgtgaacg agttgttggc tgctccctcc





3061
acagcttctg cagcagactg tccctgttgt aactgcccaa ggcatgtttt gcccaccaga





3121
tcatggccca cgtggaggcc cacctgcctc tgtctcactg aactagaagc cgagcctaga





3181
aactaacaca gccatcaagg gaatgacttg ggcggccttg ggaaatcgat gagaaattga





3241
acttcaggga gggtggtcat tgcctagagg tgctcattca tttaacagag cttccttagg





3301
ttgatgctgg aggcagaatc ccggctgtca aggggtgttc agttaagggg agcaacagag





3361
gacatgaaaa attgctatga ctaaagcagg gacaatttgc tgccaaacac ccatgcccag





3421
ctgtatggct gggggctcct cgtatgcatg gaacccccag aataaatatg ctcagccacc





3481
ctgtgggccg ggcaatccag acagcaggca taaggcacca gttaccctgc atgttggccc





3541
agacctcagg tgctagggaa ggcgggaacc ttgggttgag taatgctcgt ctgtgtgttt





3601
tagtttcatc acctgttatc tgtgtttgct gaggagagtg gaacagaagg ggtggagttt





3661
tgtataaata aagtttcttt gtctctttaa aaaaaaaaaa aaaaaaaaaa






In some embodiments the IL-4R ASO is complementary to a nucleic acid encoding the following protein sequence:











(SEQ ID NO: 309)



MGWLCSGLLF PVSCLVLLQV ASSGNMKVLQ EPTCVSDYMS







ISTCEWKMNG PTNCSTELRL LYQLVFLLSE AHTCIPENNG







GAGCVCHLLM DDVVSADNYT LDLWAGQQLL WKGSFKPSEH







VKPRAPGNLT VHTNVSDTLL LTWSNPYPPD NYLYNHLTYA







VNIWSENDPA DFRIYNVTYL EPSLRIAAST LKSGISYRAR







VRAWAQCYNT TWSEWSPSTK WHNSYREPFE QHLLLGVSVS







CIVILAVCLL CYVSITKIKK EWWDQIPNPA RSRLVAIIIQ







DAQGSQWEKR SRGQEPAKCP HWKNCLTKLL PCFLEHNMKR







DEDPHKAAKE MPFQGSGKSA WCPVEISKTV LWPESISVVR







CVELFEAPVE CEEEEEVEEE KGSFCASPES SRDDFQEGRE







GIVARLTESL FLDLLGEENG GFCQQDMGES CLLPPSGSTS







AHMPWDEFPS AGPKEAPPWG KEQPLHLEPS PPASPTQSPD







NLTCTETPLV IAGNPAYRSF SNSLSQSPCP RELGPDPLLA







RHLEEVEPEM PCVPQLSEPT TVPQPEPETW EQILRRNVLQ







HGAAAAPVSA PTSGYQEFVH AVEQGGTQAS AVVGLGPPGE







AGYKAFSSLL ASSAVSPEKC GFGASSGEEG YKPFQDLIPG







CPGDPAPVPV PLFTFGLDRE PPRSPQSSHL PSSSPEHLGL







EPGEKVEDMP KPPLPQEQAT DPLVDSLGSG IVYSALTCHL







CGHLKQCHGQ EDGGQTPVMA SPCCGCCCGD RSSPPTTPLR







APDPSPGGVP LEASLCPASL APSGISEKSK SSSSFHPAPG







NAQSSSQTPK IVNEVSVGPT YMRVS






In some embodiments antisense oligonucleotides are designed to target the nucleotide sequence corresponding to IL-1β, set forth below. Human IL-1β cDNA sequence has been disclosed in Genbank accession number NM_000576.









(SEQ ID NO: 307)








1
accaaacctc ttcgaggcac aaggcacaac aggctgctct



gggattctct tcagccaatc





61
ttcattgctc aagtgtctga agcagccatg gcagaagtac



ctgagctcgc cagtgaaatg





121
atggcttatt acagtggcaa tgaggatgac ttgttctttg



aagctgatgg ccctaaacag





181
atgaagtgct ccttccagga cctggacctc tgccctctgg



atggcggcat ccagctacga





241
atctccgacc accactacag caagggcttc aggcaggccg



cgtcagttgt tgtggccatg





301
gacaagctga ggaagatgct ggttccctgc ccacagacct



tccaggagaa tgacctgagc





361
accttctttc ccttcatctt tgaagaagaa cctatcttct



tcgacacatg ggataacgag





421
gcttatgtgc acgatgcacc tgtacgatca ctgaactgca



cgctccggga ctcacagcaa





481
aaaagcttgg tgatgtctgg tccatatgaa ctgaaagctc



tccacctcca gggacaggat





541
atggagcaac aagtggtgtt ctccatgtcc tttgtacaag



gagaagaaag taatgacaaa





601
atacctgtgg ccttgggcct caaggaaaag aatctgtacc



tgtcctgcgt gttgaaagat





661
gataagccca ctctacagct ggagagtgta gatcccaaaa



attacccaaa gaagaagatg





721
gaaaagcgat ttgtcttcaa caagatagaa atcaataaca



agctggaatt tgagtctgcc





781
cagttcccca actggtacat cagcacctct caagcagaaa



acatgcccgt cttcctggga





841
gggaccaaag gcggccagga tataactgac ttcaccatgc



aatttgtgtc ttcctaaaga





901
gagctgtacc cagagagtcc tgtgctgaat gtggactcaa



tccctagggc tggcagaaag





961
ggaacagaaa ggtttttgag tacggctata gcctggactt



tcctgttgtc tacaccaatg





1021
cccaactgcc tgccttaggg tagtgctaag aggatctcct



gtccatcagc caggacagtc





1081
agctctctcc tttcagggcc aatccccagc ccttttgttg



agccaggcct ctctcacctc





1141
tcctactcac ttaaagcccg cctgacagaa accacggcca



catttggttc taagaaaccc





1201
tctgtcattc gctcccacat tctgatgagc aaccgcttcc



ctatttattt atttatttgt





1261
ttgtttgttt tattcattgg tctaatttat tcaaaggggg



caagaagtag cagtgtctgt





1321
aaaagagcct agtttttaat agctatggaa tcaattcaat



ttggactggt gtgctctctt





1381
taaatcaagt cctttaatta agactgaaaa tatataagct



cagattattt aaatgggaat





1441
atttataaat gagcaaatat catactgttc aatggttctg



aaataaactt cactgaag






In some embodiments the IL-1β ASO is complementary to a nucleic acid encoding the following protein sequence:











(SEQ ID NO: 310)



MAEVPELASE MMAYYSGNED DLFFEADGPK QMKCSFQDLD







LCPLDGGIQL RISDHHYSKG FRQAASVVVA MDKLRKMLVP







CPQTFQENDL STFFPFIFEE EPIFFDTWDN EAYVHDAPVR







SLNCTLRDSQ QKSLVMSGPY ELKALHLQGQ DMEQQVVFSM







SFVQGEESND KIPVALGLKE KNLYLSCVLK DDKPTLQLES







VDPKNYPKKK MEKRFVFNKI EINNKLEFES AQFPNWYIST







SQAENMPVFL GGTKGGQDIT DFTMQFVSS






In some embodiments antisense oligonucleotides are designed to target the nucleotide sequence corresponding to CTGF, set forth below. Human CTGF cDNA sequence has been disclosed in Genbank accession number NM_001901.









(SEQ ID NO: 308)








1
aaactcacac aacaactctt ccccgctgag aggagacagc



cagtgcgact ccaccctcca





61
gctcgacggc agccgccccg gccgacagcc ccgagacgac



agcccggcgc gtcccggtcc





121
ccacctccga ccaccgccag cgctccaggc cccgccgctc



cccgctcgcc gccaccgcgc





181
cctccgctcc gcccgcagtg ccaaccatga ccgccgccag



tatgggcccc gtccgcgtcg





241
ccttcgtggt cctcctcgcc ctctgcagcc ggccggccgt



cggccagaac tgcagcgggc





301
cgtgccggtg cccggacgag ccggcgccgc gctgcccggc



gggcgtgagc ctcgtgctgg





361
acggctgcgg ctgctgccgc gtctgcgcca agcagctggg



cgagctgtgc accgagcgcg





421
acccctgcga cccgcacaag ggcctcttct gtgacttcgg



ctccccggcc aaccgcaaga





481
tcggcgtgtg caccgccaaa gatggtgctc cctgcatctt



cggtggtacg gtgtaccgca





541
gcggagagtc cttccagagc agctgcaagt accagtgcac



gtgcctggac ggggcggtgg





601
gctgcatgcc cctgtgcagc atggacgttc gtctgcccag



ccctgactgc cccttcccga





661
ggagggtcaa gctgcccggg aaatgctgcg aggagtgggt



gtgtgacgag cccaaggacc





721
aaaccgtggt tgggcctgcc ctcgcggctt accgactgga



agacacgttt ggcccagacc





781
caactatgat tagagccaac tgcctggtcc agaccacaga



gtggagcgcc tgttccaaga





841
cctgtgggat gggcatctcc acccgggtta ccaatgacaa



cgcctcctgc aggctagaga





901
agcagagccg cctgtgcatg gtcaggcctt gcgaagctga



cctggaagag aacattaaga





961
agggcaaaaa gtgcatccgt actcccaaaa tctccaagcc



tatcaagttt gagctttctg





1021
gctgcaccag catgaagaca taccgagcta aattctgtgg 



agtatgtacc gacggccgat





1081
gctgcacccc ccacagaacc accaccctgc cggtggagtt



caagtgccct gacggcgagg





1141
tcatgaagaa gaacatgatg ttcatcaaga cctgtgcctg



ccattacaac tgtcccggag





1201
acaatgacat ctttgaatcg ctgtactaca ggaagatgta



cggagacatg gcatgaagcc





1261
agagagtgag agacattaac tcattagact ggaacttgaa



ctgattcaca tctcattttt





1321
ccgtaaaaat gatttcagta gcacaagtta tttaaatctg



tttttctaac tgggggaaaa





1381
gattcccacc caattcaaaa cattgtgcca tgtcaaacaa



atagtctatc aaccccagac





1441
actggtttga agaatgttaa gacttgacag tggaactaca



ttagtacaca gcaccagaat





1501
gtatattaag gtgtggcttt aggagcagtg ggagggtacc



agcagaaagg ttagtatcat





1561
cagatagcat cttatacgag taatatgcct gctatttgaa



gtgtaattga gaaggaaaat





1621
tttagcgtgc tcactgacct gcctgtagcc ccagtgacag



ctaggatgtg cattctccag





1681
ccatcaagag actgagtcaa gttgttcctt aagtcagaac



agcagactca gctctgacat





1741
tctgattcga atgacactgt tcaggaatcg gaatcctgtc



gattagactg gacagcttgt





1801
ggcaagtgaa tttgcctgta acaagccaga ttttttaaaa



tttatattgt aaatattgtg





1861
tgtgtgtgtg tgtgtgtata tatatatata tgtacagtta



tctaagttaa tttaaagttg





1921
tttgtgcctt tttatttttg tttttaatgc tttgatattt



caatgttagc ctcaatttct





1981
gaacaccata ggtagaatgt aaagcttgtc tgatcgttca



aagcatgaaa tggatactta





2041
tatggaaatt ctgctcagat agaatgacag tccgtcaaaa



cagattgttt gcaaagggga





2101
ggcatcagtg tccttggcag gctgatttct aggtaggaaa



tgtggtagcc tcacttttaa





2161
tgaacaaatg gcctttatta aaaactgagt gactctatat



agctgatcag ttttttcacc





2221
tggaagcatt tgtttctact ttgatatgac tgtttttcgg



acagtttatt tgttgagagt





2281
gtgaccaaaa gttacatgtt tgcacctttc tagttgaaaa



taaagtgtat attttttcta





2341
taaaaaaaaa aaaaaaaa






In some embodiments the CTGF ASO is complementary to a nucleic acid encoding the following protein sequence:











(SEQ ID NO: 311)



MTAASMGPVR VAFVVLLALC SRPAVGQNCS GPCRCPDEPA







PRCPAGVSLV LDGCGCCRVC AKQLGELCTE RDPCDPHKGL







FCHFGSPANR KIGVCTAKDG APCIFGGTVY RSGESFQSSC







KYQCTCLDGA VGCMPLCSMD VRLPSPDCPF PRRVKLPGKC







CEEWVCDEPK DQTVVGPALA AYRLEDTFGP DPTMIRANCL







VQTTEWSACS KTCGMGISTR VTNDNASCRL EKQSRLCMVR







PCEADLEENI KKGKKCIRTP KISKPIKFEL SGCTSMKTYR







AKFCGVCTDG RCCTPHRTTT LPVEFKCPDG EVMKKNMMFI







KTCACHYNCP GDNDIFESLY YRKMYGDMA 






The invention in some aspects relates to multiplex ASO-SNAs for reducing the expression of multiple gene targets. In some embodiments the mASO-SNAs include ASOs that target IL-17RA and TNFα. These particular mASOs-SNAs are particularly useful for treating inflammatory disorders. A “TNFα antisense oligonucleotide” or “TNF-α ASO” as used herein refers to a nucleic acid based agent which interferes with TNFα activity. In particular, the TNFα antisense inhibitors or TNFα antisense oligonucleotides of the invention reduce the expression of the TNFα gene.


TNF-α (tumor necrosis factor-alpha) is a pleiotropic cytokine produced by activated macrophages/monocytes and lymphocytes which often promotes inflammatory responses leading to a variety of diseases. TNF-α is released from macrophages, monocytes and natural killer cells and play an important role in inflammatory and immune responses, including the recruitment of leukocytes to injured tissues during bacterial and other microbial infections, and following stimulation with inflammatory substances. When present in excessive quantities, TNF-α is known to cause tissue injury, and has been implicated in the pathology associated with inflammatory and autoimmune diseases.


TNF-α mediates biological effects through two distinct membrane-protein receptors, TNF-RI and TNF-RII, which differ in sequence and molecular mass. TNF-RI is reported to be present at low levels in most, if not all, human cell types, and expression of the TNF-RI gene in humans can be upregulated by infection, interferons, and modulators of second messengers, such as phorbol esters. The extracellular portions of both TNF receptors also exist in soluble forms, which are derived from membrane-bound forms of the receptors by proteolytic cleavage at the cell surface. The soluble TNF receptors retain the ability to bind TNF-α in solution. Soluble TNF receptors have been identified in urine and sera from healthy individuals, and have been shown to be elevated in some chronic diseases and following inoculation with agents that induce TNF-α.


In some embodiments, antisense oligonucleotides are designed to target human TNFα, for instance, the nucleotide sequence of SEQ ID NO: 304, set forth below. Human TNF-α cDNA sequence has been published by Nedwin, G. E. et al. (Nucleic Acids Res. 1985, 13, 6361-6373); and is disclosed in Genbank accession number X02910 and NM_000594.










(SEQ ID NO: 304)










   1
gaattccggg tgatttcact cccggctgtc caggcttgtc ctgctacccc acccagcctt






  61
tcctgaggcc tcaagcctgc caccaagccc ccagctcctt ctccccgcag gacccaaaca





 121
caggcctcag gactcaacac agcttttccc tccaacccgt tttctctccc tcaacggact





 181
cagctttctg aagcccctcc cagttctagt tctatctttt tcctgcatcc tgtctggaag





 241
ttagaaggaa acagaccaca gacctggtcc ccaaaagaaa tggaggcaat aggttttgag





 301
gggcatgggg acggggttca gcctccaggg tcctacacac aaatcagtca gtggcccaga





 361
agacccccct cggaatcgga gcagggagga tggggagtgt gaggggtatc cttgatgctt





 421
gtgtgtcccc aactttccaa atccccgccc ccgcgatgga gaagaaaccg agacagaagg





 481
tgcagggccc actaccgctt cctccagatg agctcatggg tttctccacc aaggaagttt





 541
tccgctggtt gaatgattct ttccccgccc tcctctcgcc ccagggacat ataaaggcag





 601
ttgttggcac acccagccag cagacgctcc ctcagcaagg acagcagagg accagctaag





 661
agggagagaa gcaactacag accccccctg aaaacaaccc tcagacgcca catcccctga





 721
caagctgcca ggcaggttct cttcctctca catactgacc cacggcttca ccctctctcc





 781
cctggaaagg acaccatgag cactgaaagc atgatccggg acgtggagct ggccgaggag





 841
gcgctcccca agaagacagg ggggccccag ggctccaggc ggtgcttgtt cctcagcctc





 901
ttctccttcc tgatcgtggc aggcgccacc acgctcttct gcctgctgca ctttggagtg





 961
atcggccccc agagggaaga ggtgagtgcc tggccagcct tcatccactc tcccacccaa





1021
ggggaaatga gagacgcaag agagggagag agatgggatg ggtgaaagat gtgcgctgat





1081
agggagggat gagagagaaa aaaacatgga gaaagacggg gatgcagaaa gagatgtggc





1141
aagagatggg gaagagagag agagaaagat ggagagacag gatgtctggc acatggaagg





1201
tgctcactaa gtgtgtatgg agtgaatgaa tgaatgaatg aatgaacaag cagatatata





1261
aataagatat ggagacagat gtggggtgtg agaagagaga tgggggaaga aacaagtgat





1321
atgaataaag atggtgagac agaaagagcg ggaaatatga cagctaagga gagagatggg





1381
ggagataagg agagaagaag atagggtgtc tggcacacag aagacactca gggaaagagc





1441
tgttgaatgc tggaaggtga atacacagat gaatggagag agaaaaccag acacctcagg





1501
gctaagagcg caggccagac aggcagccag ctgttcctcc tttaagggtg actccctcga





1561
tgttaaccat tctccttctc cccaacagtt ccccagggac ctctctctaa tcagccctct





1621
ggcccaggca gtcagtaagt gtctccaaac ctctttccta attctgggtt tgggtttggg





1681
ggtagggtta gtaccggtat ggaagcagtg ggggaaattt aaagttttgg tcttggggga





1741
ggatggatgg aggtgaaagt aggggggtat tttctaggaa gtttaagggt ctcagctttt





1801
tcttttctct ctcctcttca ggatcatctt ctcgaacccc gagtgacaag cctgtagccc





1861
atgttgtagg taagagctct gaggatgtgt cttggaactt ggagggctag gatttgggga





1921
ttgaagcccg gctgatggta ggcagaactt ggagacaatg tgagaaggac tcgctgagct





1981
caagggaagg gtggaggaac agcacaggcc ttagtgggat actcagaacg tcatggccag





2041
gtgggatgtg ggatgacaga cagagaggac aggaaccgga tgtggggtgg gcagagctcg





2101
agggccagga tgtggagagt gaaccgacat ggccacactg actctcctct ccctctctcc





2161
ctccctccag caaaccctca agctgagggg cagctccagt ggctgaaccg ccgggccaat





2221
gccctcctgg ccaatggcgt ggagctgaga gataaccagc tggtggtgcc atcagagggc





2281
ctgtacctca tctactccca ggtcctcttc aagggccaag gctgcccctc cacccatgtg





2341
ctcctcaccc acaccatcag ccgcatcgcc gtctcctacc agaccaaggt caacctcctc





2401
tctgccatca agagcccctg ccagagggag accccagagg gggctgaggc caagccctgg





2461
tatgagccca tctatctggg aggggtcttc cagctggaga agggtgaccg actcagcgct





2521
gagatcaatc ggcccgacta tctcgacttt gccgagtctg ggcaggtcta ctttgggatc





2581
attgccctgt gaggaggacg aacatccaac cttcccaaac gcctcccctg ccccaatccc





2641
tttattaccc cctccttcag acaccctcaa cctcttctgg ctcaaaaaga gaattggggg





2701
cttagggtcg gaacccaagc ttagaacttt aagcaacaag accaccactt cgaaacctgg





2761
gattcaggaa tgtgtggcct gcacagtgaa gtgctggcaa ccactaagaa ttcaaactgg





2821
ggcctccaga actcactggg gcctacagct ttgatccctg acatctggaa tctggagacc





2881
agggagcctt tggttctggc cagaatgctg caggacttga gaagacctca cctagaaatt





2941
gacacaagtg gaccttaggc cttcctctct ccagatgttt ccagacttcc ttgagacacg





3001
gagcccagcc ctccccatgg agccagctcc ctctatttat gtttgcactt gtgattattt





3061
attatttatt tattatttat ttatttacag atgaatgtat ttatttggga gaccggggta





3121
tcctggggga cccaatgtag gagctgcctt ggctcagaca tgttttccgt gaaaacggag





3181
ctgaacaata ggctgttccc atgtagcccc ctggcctctg tgccttcttt tgattatgtt





3241
ttttaaaata tttatctgat taagttgtct aaacaatgct gatttggtga ccaactgtca





3301
ctcattgctg agcctctgct ccccagggga gttgtgtctg taatcgccct actattcagt





3361
ggcgagaaat aaagtttgct tagaaaagaa acatggtctc cttcttggaa ttaattctgc





3421
atctgcctct tcttgtgggt gggaagaagc tccctaagtc ctctctccac aggctttaag





3481
atccctcgga cccagtccca tccttagact cctagggccc tggagaccct acataaacaa





3541
agcccaacag aatattcccc atcccccagg aaacaagagc ctgaacctaa ttacctctcc





3601
ctcagggcat gggaatttcc aactctggga attc






The nanostructures descried herein may be stable self-assembling nanostructures. For instance the nanostructure may be an antisense oligonucleotide of 18-21 nucleotides in length having a sequence described herein, wherein a hydrophobic group at the 3′ or 5′ terminus self-associates to form the core of the nanostructure in water or other suitable solvents. A hydrophobic group as used herein may include cholesterol, a cholesteryl or modified cholesteryl residue, adamantine, dihydrotesterone, long chain alkyl, long chain alkenyl, long chain alkynyl, olely-lithocholic, cholenic, oleoyl-cholenic, palmityl, heptadecyl, myrisityl, bile acids, cholic acid or taurocholic acid, deoxycholate, oleyl litocholic acid, oleoyl cholenic acid, glycolipids, phospholipids, sphingolipids, isoprenoids, such as steroids, vitamins, such as vitamin E, fatty acids either saturated or unsaturated, fatty acid esters, such as triglycerides, pyrenes, porphyrines, Texaphyrine, adamantane, acridines, biotin, coumarin, fluorescein, rhodamine, Texas-Red, digoxygenin, dimethoxytrityl, t-butyldimethylsilyl, t-butyldiphenylsilyl, cyanine dyes (e.g. Cy3 or Cy5), Hoechst 33258 dye, psoralen, or ibuprofen.


The antisense oligonucleotides typically have a length of 10-30 or 15-20 bases, which is generally long enough to have one complementary sequence in the mammalian genome. Additionally, antisense compounds having a length of at least 12, typically at least 15 nucleotides in length hybridize well with their target mRNA. Thus, the antisense oligonucleotides of the invention are typically in a size range of 8-100 nucleotides, more preferably 12-50 nucleotides in length. In some embodiments of the invention the antisense oligonucleotides are of 18-19 nucleotides in length. The antisense oligonucleotides may include further nucleotides on the 5′ and/or 3′ end of the oligonucleotide. However an antisense oligonucleotide that is limited to 18 nucleotides in length, for example, does not have any additional nucleotides on the 5′ or 3′ end of the molecule. Other non-nucleotide molecules may be linked covalently or non-covalently to the 5′ and/or 3′ end of those oligonucleotides.


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 hexose, 2′-F hexose, 2′-amino ribose, CEt-LNA, 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.


In some embodiments, the modified oligonucleotide is a single-stranded modified oligonucleotide. In some embodiments, the single-stranded modified oligonucleotide consists of 10-30, 10-35, 10-40, 10-45, 10-50, 10-60, 10-70, 10-80, 10-90, 10-100 or more than 100 linked nucleosides and has a gap segment. In some embodiments, a gap segment refers to one or more linked nucleic acids consisting of deoxynucleosides located at the center or near the center of a modified oligonucleotide, such as a single-stranded modified oligonucleotide. In some embodiments, the gap segment consists of 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 2-9, 2-10, 2-20, 2-30, 2-40 2-50, 10-20, 10-30, 10-40 or 10-50 linked deoxynucleosides.


A 5′ wing segment corresponds to the linked nucleic acids (e.g., nucleosides) from the 5′-end of a modified oligonucleotide to the nucleic acid before the first nucleic acid at the 5′-end of the gap segment. A 3′ wing segment corresponds to the linked nucleic acids (e.g., nucleosides) after the last nucleic acid at the 3′ end of the gap segment to the last nucleic acid at the 3′ end of the modified oligonucleotide.


The gap segment is positioned between the 5′ wing segment and the 3′ wing segment. In some embodiments, at least one nucleoside of the 5′ wing segment and/or at least one nucleoside of the 3′ wing segment comprises a modified nucleoside. In some embodiments, the internucleoside linkages within the gap segment and the linkages connecting the gap segment to the 3′ wing segment and/or the 5′ wing segment are all phosphorothioate linkages (*). In some embodiments, the internucleoside linkages connecting the rest of the nucleosides of both the 5′ and 3′ wing segments are phosphodiester linkages. In some embodiments, the nucleosides in the modified oligonucleotide are modified with a 2′ O-methyl group. The nucleosides in the modified oligonucleoside can also be modified with any other modification described herein.


In some embodiments, the nucleobase sequence of the modified oligonucleotide consists of 10-30, 10-35, 10-40, 10-45, 10-50, 10-60, 10-70, 10-80, 10-90, 10-100 or more than 100 linked nucleosides and has a gap segment complementary to an equal length portion of the coding sequence (e.g., cDNA) of the IL-17R, such as IL-17RA (e.g., cDNA sequence of IL-17RA is represented by SEQ ID NO: 302), IL-17RB, IL-17RC, IL-17RD, or IL-17RE) (see e.g., Johansen et al., Br J Dermatol (2009) 160(2):319-24, or a pharmaceutically acceptable salt thereof.


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; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, 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-(guanidiniumalkylhydroxy)-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 antisense oligonucleotides of the invention may be chimeric oligonucleotides. Chimeric antisense 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 mixed backbone or chimeric 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.


The oligonucleotides may include a molecular species at one or both ends, i.e., at the 3′ and/or 5′ end. A molecular species as used herein refers to any compound that is not a naturally occurring or non-naturally occurring nucleotide. Molecular species include but are not limited to a spacer, a lipid, a sterol, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-S-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety, stearyl, C16 alkyl chain, bile acids, cholic acid, taurocholic acid, deoxycholate, oleyl litocholic acid, oleoyl cholenic acid, glycolipids, phospholipids, sphingolipids, isoprenoids, such as steroids, vitamins, such as vitamin E, saturated fatty acids, unsaturated fatty acids, fatty acid esters, such as triglycerides, pyrenes, porphyrines, Texaphyrine, adamantane, acridines, biotin, coumarin, fluorescein, rhodamine, Texas-Red, digoxygenin, dimethoxytrityl, t-butyldimethylsilyl, t-butyldiphenylsilyl, cyanine dyes (e.g. Cy3 or Cy576), Hoechst 33258 dye, psoralen, or ibuprofen.


The molecular species may be attached at various positions of the oligonucleotide. As described above, the molecular species may be linked to the 2′-end, 3′-end or 5′-end of the oligonucleotide, where it also serves the purpose to enhance the stability of the oligomer against 3′- or 5′-exonucleases. Alternatively, it may be linked to an internal nucleotide or a nucleotide on a branch. The molecular species may be attached to a 2′-position of the nucleotide. The molecular species may also be linked to the heterocyclic base of the nucleotide.


The molecular species may be connected to the oligonucleotide by a linker moiety. Optionally the linker moiety is a non-nucleotidic linker moiety. Non-nucleotidic linkers are e.g. abasic residues (dSpacer), oligoethyleneglycol, such as triethyleneglycol or hexaethylenegylcol, or alkane-diol, such as butanediol. The spacer units are preferably linked by phosphodiester, phosphorodithioate or phosphorothioate bonds. The linker units may appear just once in the molecule or may be incorporated several times, e.g. via phosphodiester, phosphorothioate, methylphosphonate, or amide linkages.


The oligonucleotide of the invention (separate from the linkers connecting nucleotides to the molecular species) may also contain non-nucleotidic linkers, in particular abasic linkers (dSpacers), trietyhlene glycol units or hexaethylene glycol units. Further preferred linkers are alkylamino linkers, such as C3, C6, C12 aminolinkers, and also alkylthiol linkers, such as C3 or C6 thiol linkers.


IL-17RA and TNFα play a role in a wide variety of inflammatory disorders. An inflammatory disorder as used herein refers to a disorder in which IL-17 or TNFα activity is detrimental to a particular physiological function in a subject. As used herein, the term “a disorder in which IL-17 or TNFα activity is detrimental” is intended to include diseases and other disorders in which the levels of these cytokines expressed in a subject suffering from the disorder plays a role in the pathophysiology of the disorder or as a factor that contributes to a worsening of or maintenance of the disorder. Accordingly, a disorder in which IL-17 or TNFα activity is detrimental is a disorder in which inhibition of IL-17/IL-17RA or TNFα activity is expected to alleviate at least one symptom and/or progression or worsening of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of IL-17 or TNFα in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of IL-17 or TNFα in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using a probe or an antibody for detecting IL-17 or TNFα message or protein respectively.


Inflammatory disorders include but are not limited to sepsis, infections, autoimmune diseases, cancer, transplant rejection and graft-versus-host disease, transplant rejection, malignancy, a pulmonary disorder, an intestinal disorder, a cardiac disorder, sepsis, a spondyloarthropathy, a metabolic disorder, anemia, pain, a hepatic disorder, a skin disorder, a nail disorder, rheumatoid arthritis, psoriasis, psoriasis in combination with psoriatic arthritis, ulcerative colitis, Crohn's disease, vasculitis, Behcet's disease, ankylosing spondylitis, asthma, chronic obstructive pulmonary disorder (COPD), idiopathic pulmonary fibrosis (IPF), restenosis, diabetes, anemia, pain, a Crohn's disease-related disorder, juvenile rheumatoid arthritis (JRA), a hepatitis C virus infection, psoriatic arthritis, and chronic plaque psoriasis.


The biological role played by IL-17 or TNFα in these diseases is known. Inhibiting IL-17RA and/or TNFα expression in these diseases provides a therapeutic treatment for the disorder.


The ASO-SNAs and mASO-SNAs may be administered alone or in conjunction with another therapeutic agent for the treatment of an inflammatory disorder. Non-limiting examples of therapeutic agents with which the ASO-SNAs and mASO-SNAs of the invention can be combined include the following: non-steroidal anti-inflammatory drug(s) (NSAIDs); cytokine suppressive anti-inflammatory drug(s) (CSAIDs); CDP-571/BAY-10-3356 (humanized anti-TNFα antibody; Celltech/Bayer); cA2/infliximab (chimeric anti-TNFα antibody; Centocor); 75 kdTNFR-IgG/etanercept (75 kD TNF receptor-IgG fusion protein; Immunex; 55 kdTNF-IgG (55 kD TNF receptor-IgG fusion protein; Hoffmann-LaRoche); IDEC-CE9/SB 210396 (non-depleting primatized anti-CD4 antibody; IDEC/SmithKline; DAB 486-IL-2 and/or DAB 389-IL-2 (IL-2 fusion proteins; Seragen; Anti-Tac (humanized anti-IL-2Ra; Protein Design Labs/Roche); IL-4 (anti-inflammatory cytokine; DNAX/Schering); IL-10 (SCH 52000; recombinant IL-10, anti-inflammatory cytokine; DNAX/Schering); IL-4; IL-10 and/or IL-4 agonists (e.g., agonist antibodies); IL-1 RA (IL-1 receptor antagonist; Synergen/Amgen); anakinra (Kineret/Amgen); TNF-bp/s-TNF (soluble TNF binding protein); R973401 (phosphodiesterase Type IV inhibitor; MK-966 (COX-2 Inhibitor; Iloprost; methotrexate; thalidomide and thalidomide-related drugs (e.g., Celgen); leflunomide (anti-inflammatory and cytokine inhibitor; tranexamic acid (inhibitor of plasminogen activation; T-614 (cytokine inhibitor; prostaglandin E1; Tenidap (non-steroidal anti-inflammatory drug; Naproxen (non-steroidal anti-inflammatory drug; Meloxicam (non-steroidal anti-inflammatory drug); Ibuprofen (non-steroidal anti-inflammatory drug); Piroxicam (non-steroidal anti-inflammatory drug); Diclofenac (non-steroidal anti-inflammatory drug); Indomethacin (non-steroidal anti-inflammatory drug); Sulfasalazine; Azathioprine; ICE inhibitor (inhibitor of the enzyme interleukin-1-beta-converting enzyme); zap-70 and/or Ick inhibitor (inhibitor of the tyrosine kinase zap-70 or Ick); VEGF inhibitor and/or VEGF-R inhibitor (inhibitors of vascular endothelial cell growth factor or vascular endothelial cell growth factor receptor; inhibitors of angiogenesis); corticosteroid anti-inflammatory drugs (e.g., SB203580); TNF-convertase inhibitors; anti-IL-12 antibodies; anti-IL-18 antibodies; interleukin-11; interleukin-13; interleukin-17 inhibitors; gold; penicillamine; chloroquine; hydroxychloroquine; chlorambucil; cyclosporine; cyclophosphamide; total lymphoid irradiation; anti-thymocyte globulin; anti-CD4 antibodies; CD5-toxins; orally-administered peptides and collagen; lobenzarit disodium; Cytokine Regulating Agents (CRAs) HP228 and HP466 (Houghten Pharmaceuticals, Inc.); ICAM-1 antisense phosphorothioate oligodeoxynucleotides (ISIS 2302; Isis Pharmaceuticals, Inc.); soluble complement receptor 1 (TP10; T Cell Sciences, Inc.); prednisone; orgotein; glycosaminoglycan polysulphate; minocycline; anti-IL2R antibodies; marine and botanical lipids (fish and plant seed fatty acids; auranofin; phenylbutazone; meclofenamic acid; flufenamic acid; intravenous immune globulin; zileuton; azaribine; mycophenolic acid (RS-61443); tacrolimus (FK-506); sirolimus (rapamycin); amiprilose (therafectin); cladribine (2-chlorodeoxyadenosine); methotrexate; antivirals; and immune modulating agents.


Toxicity and efficacy of the prophylactic and/or therapeutic protocols of the present invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Prophylactic and/or therapeutic agents that exhibit large therapeutic indices are preferred. While prophylactic and/or therapeutic agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.


The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the prophylactic and/or therapeutic agents for use in humans. The dosage of such agents lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any agent used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. A number of studies have examined the optimal dosages for antisense oligonucleotides.


In certain embodiments, pharmaceutical compositions may comprise, for example, at least about 0.000001% (w/w) of an active compound. In other embodiments, the active compound may comprise between about 2% to about 75% of the weight of the unit (w/w), or between about 25% to about 60%, for example, and any range derivable therein. In some embodiments, the active compound (e.g., oligonucleotide or nanostructure) described herein comprises between 0.000001% and 0.00001%, between 0.00001% and 0.0001%, between 0.0001% and 0.001%, between 0.001% and 0.01%, between 0.01% and 0.1%, between 0.1% and 1%, between 1% and 5%, between 5% and 10%, between 10% and 15%, between 15% and 20%, between 20% and 25%, between 25% and 30%, between 30% and 40%, between 40% and 50% (w/w), and any range derivable in between. In some embodiments, the active compound (e.g., oligonucleotide or nanostructure) described herein comprises 0.00007%, 0.007%, 0.01%, 0.1%, 1% (w/w)


Subject doses of the compounds described herein typically range from about 0.1 μg to 10,000 mg, more typically from about 1 μg/day to 8000 mg, and most typically from about 10 μg to 100 μg. Stated in terms of subject body weight, typical dosages range from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg/body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc., can be administered, based on the numbers described above. The absolute amount will depend upon a variety of factors including the concurrent treatment, the number of doses and the individual patient parameters including age, physical condition, size and weight. These are factors well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to sound medical judgment.


Multiple doses of the molecules of the invention are also contemplated. In some instances, when the molecules of the invention are administered with another therapeutic, for instance, an anti-inflammatory agent, a sub-therapeutic dosage of either the molecules or the other agent, or a sub-therapeutic dosage of both, is used in the treatment of a subject having, or at risk of developing an inflammatory disorder. When the two classes of drugs are used together, the other agent may be administered in a sub-therapeutic dose to produce a desirable therapeutic result. A “sub-therapeutic dose” as used herein refers to a dosage which is less than that dosage which would produce a therapeutic result in the subject if administered in the absence of the other agent. Thus, the sub-therapeutic dose of a therapeutic agent is one which would not produce the desired therapeutic result in the subject in the absence of the administration of the molecules of the invention. Therapeutic doses of agents useful for treating inflammatory disorders are well known in the field of medicine. These dosages have been extensively described in references such as Remington's Pharmaceutical Sciences; as well as many other medical references relied upon by the medical profession as guidance for the treatment of infectious disease, cancer, and autoimmune disease. Therapeutic dosages of oligonucleotides have also been described in the art.


Dosing regimens may be several times a day, daily, every other day, weekly, biweekly any of the times there between or less frequently. The term “biweekly dosing” as used herein, refers to the time course of administering a substance (e.g., an anti-IL-17RA nucleic acid) to a subject once every two weeks. The oligonucleotides may be administered every 7-20 days, every 11-17 days, or every 13-15 days, for example.


In some embodiments, a compound (e.g., oligonucleotide, nanostructure, etc.) described herein is administered for 4, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, 48, 72, 96, 120, 240, 480 hours, or any ranges in between, per dose in a dosing schedule.


The oligonucleotides are administered in effective amounts. The effective amount of a compound of the invention in the treatment of a disease described herein may vary depending upon the specific compound used, the mode of delivery of the compound, and whether it is used alone or in combination. The effective amount for any particular application can also vary depending on such factors as the disease being treated, the particular compound being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular molecule of the invention without necessitating undue experimentation. Combined with the teachings provided herein, by choosing among the various active compounds and weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and preferred mode of administration, an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the particular subject.


In some embodiments, the cell is contacted with an oligonucleotide, a nanostructure, or a mASO-SNA described herein at a concentration of at least 0.001 nM, at least 0.01 nM, at least 0.1 nM, at least 1 nM, at least 10 nM, at least 100 nM, at least 1000 nM, at least 10 μM, at least 100 μM, at least 1000 μM, or above 1000 μM. In some embodiments, the cell is contacted with an oligonucleotide, a nanostructure, or a mASO-SNA described herein at a concentration range of 0.001 nM to 0.01 nM, 0.01 nM to 0.1 nM, 0.1 nM to 1 nM, 1 nM to 10 nM, 10 nM to 100 nM, 100 nM to 1000 nM, 1000 nM to 10 μM, 10 μM to 100 μM, or 100 μM to 1000 μM. In some embodiments, the cell is contacted with an oligonucleotide, a nanostructure, or a mASO-SNA described herein at a concentration of 0.001 nM, 0.01 nM, 0.1 nM, 1 nM, 10 nM, 100 nM, 1000 nM, 10 μM, 100 μM, 1000 μM or above 1000 μM.


The oligonucleotides described herein can be used alone or in conjugates with other molecules such as detection or cytotoxic agents in the detection and treatment methods of the invention, as described in more detail herein.


The oligonucleotide may be, for instance, coupled or conjugated to a detectable label. A detectable label is a moiety, the presence of which can be ascertained directly or indirectly. Generally, detection of the label involves an emission of energy by the label. The label can be detected directly by its ability to emit and/or absorb photons or other atomic particles of a particular wavelength (e.g., radioactivity, luminescence, optical or electron density, etc.). A label can be detected indirectly by its ability to bind, recruit and, in some cases, cleave another moiety which itself may emit or absorb light of a particular wavelength (e.g., epitope tag such as the FLAG epitope, enzyme tag such as horseradish peroxidase, etc.). An example of indirect detection is the use of a first enzyme label which cleaves a substrate into visible products. The label may be of a chemical, peptide or nucleic acid molecule nature although it is not so limited. Other detectable labels include radioactive isotopes such as P32 or H3, luminescent markers such as fluorochromes, optical or electron density markers, etc., or epitope tags such as the FLAG epitope or the HA epitope, biotin, avidin, and enzyme tags such as horseradish peroxidase, β-galactosidase, etc. The label may be bound to an oligonucleotide during or following its synthesis. There are many different labels and methods of labeling known to those of ordinary skill in the art. Examples of the types of labels that can be used in the present invention include enzymes, radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, and bioluminescent compounds. Those of ordinary skill in the art will know of other suitable labels for the oligonucleotides described herein, or will be able to ascertain such, using routine experimentation. Furthermore, the coupling or conjugation of these labels to the oligonucleotides of the invention can be performed using standard techniques common to those of ordinary skill in the art.


Conjugation of the oligonucleotides to a detectable label facilitates, among other things, the use of such agents in diagnostic assays. Another category of detectable labels includes diagnostic and imaging labels (generally referred to as in vivo detectable labels) such as for example magnetic resonance imaging (MRI): Gd(DOTA); for nuclear medicine: 201Tl, gamma-emitting radionuclide 99mTc; for positron-emission tomography (PET): positron-emitting isotopes, (18)F-fluorodeoxyglucose ((18)FDG), (18)F-fluoride, copper-64, gadodiamide, and radioisotopes of Pb(II) such as 203Pb; 111In. In such instances, the use of the oligonucleotide could be observed as the oligonucleotide provides an antisense effect.


The conjugations or modifications described herein employ routine chemistry, which chemistry does not form a part of the invention and which chemistry is well known to those skilled in the art of chemistry. The use of protecting groups and known linkers such as mono- and hetero-bifunctional linkers are well documented in the literature and will not be repeated here.


As used herein, “conjugated” means two entities stably bound to one another by any physiochemical means. It is important that the nature of the attachment is such that it does not impair substantially the effectiveness of either entity. Keeping these parameters in mind, any covalent or non-covalent linkage known to those of ordinary skill in the art may be employed. In some embodiments, covalent linkage is preferred. Noncovalent conjugation includes hydrophobic interactions, ionic interactions, high affinity interactions such as biotin-avidin and biotin-streptavidin complexation and other affinity interactions. Such means and methods of attachment are well known to those of ordinary skill in the art. A variety of methods may be used to detect the label, depending on the nature of the label and other assay components.


Pharmaceutical compositions of the present invention comprise an effective amount of one or more agents, dissolved or dispersed in a pharmaceutically acceptable carrier. The phrases “pharmaceutical or pharmacologically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate. Moreover, for animal (e.g., human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards. The compounds are generally suitable for administration to humans. This term requires that a compound or composition be nontoxic and sufficiently pure so that no further manipulation of the compound or composition is needed prior to administration to humans.


As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art.


The agent may comprise different types of carriers depending on whether it is to be administered in solid, liquid, gel, cream, or aerosol form, and whether it need to be sterile for such routes of administration as injection. The present invention can be administered intravenously, intradermally, intraarterially, intralesionally, intratumorally, intracranially, intrathecally, intraarticularly, intraprostaticaly, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, intramuscularly, intraperitoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, via eyedrops, orally, topically, locally, inhalation (e.g., aerosol inhalation), injection, infusion, continuous infusion, localized perfusion bathing target cells directly, via a catheter, via a lavage, in creams, in gel, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art.


In any case, the composition may comprise various antioxidants to retard oxidation of one or more components. Additionally, the prevention of the action of microorganisms can be brought about by preservatives such as various antibacterial and antifungal agents, including but not limited to parabens (e.g., methylparabens, propylparabens), chlorobutanol, phenol, sorbic acid, thimerosal or combinations thereof.


The agent may be formulated into a composition in a free base, neutral or salt form. Pharmaceutically acceptable salts, include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine.


In embodiments where the composition is in a liquid form, a carrier can be a solvent or dispersion medium comprising but not limited to, water, ethanol, polyol (e.g., glycerol, propylene glycol, liquid polyethylene glycol, etc.), lipids (e.g., triglycerides, vegetable oils, liposomes) and combinations thereof. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin; by the maintenance of the required particle size by dispersion in carriers such as, for example liquid polyol or lipids; by the use of surfactants such as, for example hydroxypropylcellulose; or combinations thereof such methods. In many cases, it will be preferable to include isotonic agents, such as, for example, sugars, sodium chloride or combinations thereof.


The compounds of the invention may be administered directly to a tissue. Direct tissue administration may be achieved by direct injection, topical application, or local application. The compounds may be administered once, or alternatively they may be administered in a plurality of administrations. If administered multiple times, the compounds may be administered via different routes. For example, the first (or the first few) administrations may be made directly into the affected tissue while later administrations may be systemic.


The formulations of the invention are 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.


According to the methods of the invention, the compound may be administered in a pharmaceutical composition. In general, a pharmaceutical composition comprises the compound of the invention and a pharmaceutically-acceptable carrier. As used herein, a pharmaceutically-acceptable carrier means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredients.


Pharmaceutically acceptable carriers include diluents, fillers, salts, buffers, stabilizers, solubilizers and other materials which are well-known in the art. Such preparations may routinely contain salt, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents. When used in medicine, the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention. Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like. Also, pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.


The compounds of the invention may be formulated into preparations in solid, semi-solid, liquid or gaseous forms such as tablets, capsules, powders, granules, ointments, solutions, depositories, inhalants and injections, and usual ways for oral, parenteral or surgical administration. The invention also embraces pharmaceutical compositions which are formulated for local administration, such as by implants.


Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent. Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion.


For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, thermoreversible hydrogels such as pluronic F-127, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Optionally the oral formulations may also be formulated in saline or buffers for neutralizing internal acid conditions or may be administered without any carriers.


Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.


Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art. All formulations for oral administration should be in dosages suitable for such administration.


For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.


For administration by inhalation, the compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. Techniques for preparing aerosol delivery systems are well known to those of skill in the art.


The compounds, when it is desirable to deliver them systemically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.


Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. Lower doses will result from other forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of compounds.


The compositions of the invention may be formulated in a topical composition for administration to the skin or a body cavity. Suitable topical vehicles and vehicle components are well known in the cosmetic and pharmaceutical arts, and include such vehicles (or vehicle components) as water; thermoreversible hydrogels such as pluronic F-127, organic solvents such as alcohols (particularly lower alcohols readily capable of evaporating from the skin such as ethanol), glycols (such as propylene glycol, butylene glycol, and glycerin), aliphatic alcohols (such as lanolin); mixtures of water and organic solvents (such as water and alcohol), and mixtures of organic solvents such as alcohol and glycerin (optionally also with water); lipid-based materials such as fatty acids, acylglycerols (including oils, such as mineral oil, and fats of natural or synthetic origin), phosphoglycerides, sphingolipids and waxes; protein-based materials such as collagen and gelatin; silicone-based materials (both non-volatile and volatile) such as cyclomethicone, demethiconol and dimethicone copolyol (Dow Corning); hydrocarbon-based materials such as petrolatum and squalane; anionic, cationic and amphoteric surfactants and soaps; sustained-release vehicles such as microsponges and polymer matrices; stabilizing and suspending agents; emulsifying agents; and other vehicles and vehicle components that are suitable for administration to the skin, as well as mixtures of topical vehicle components as identified above or otherwise known to the art. The vehicle may further include components adapted to improve the stability or effectiveness of the applied formulation, such as preservatives, antioxidants, skin penetration enhancers, sustained release materials, and the like.


The choice of a suitable vehicle will depend on the particular physical form and mode of delivery that the formulation is to achieve. Examples of suitable forms include liquids (e.g., gargles and mouthwashes, including dissolved forms of the strontium cation as well as suspensions, emulsions and the like); solids and semisolids such as gels, foams, pastes, creams, ointments, “sticks” (as in lipsticks or underarm deodorant sticks), powders and the like; formulations containing liposomes or other delivery vesicles; rectal or vaginal suppositories, creams, foams, gels or ointments; and other forms. Typical modes of delivery include application using the fingers; application using a physical applicator such as a cloth, tissue, swab, stick or brush (as achieved for example by soaking the applicator with the formulation just prior to application, or by applying or adhering a prepared applicator already containing the formulation—such as a treated or premoistened bandage, wipe, washcloth or stick—to the skin); spraying (including mist, aerosol or foam spraying); dropper application (as for example with ear drops); sprinkling (as with a suitable powder form of the formulation); and soaking.


Topical formulations also include formulations for rectal and vaginal administration. Formulations for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter. Formulations suitable for vaginal administration may be presented as tablets, pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.


In yet other embodiments, a delivery vehicle is a biocompatible microparticle or implant that is suitable for implantation into the mammalian recipient. Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the compound, increasing convenience to the subject and the physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides.


In some embodiments the antisense nucleic acids of the invention are formulated as a stable self-assembling nanostructure. The nanostructure includes a IL-17RA antisense oligonucleotide, wherein the antisense oligonucleotide is associated with a core. The core may be a solid or a hollow core, such as a liposomal core. A solid core is a spherical shaped material that does not have a hollow center. The term spherical as used herein refers to a general shape and does not imply or is not limited to a perfect sphere or round shape. It may include imperfections.


Solid cores can be constructed from a wide variety of materials known to those skilled in the art including but not limited to: noble metals (gold, silver), transition metals (iron, cobalt) and metal oxides (silica). In addition, these cores may be inert, paramagnetic, or superparamagnetic. These solid cores can be constructed from either pure compositions of described materials, or in combinations of mixtures of any number of materials, or in layered compositions of materials. In addition, solid cores can be composed of a polymeric core such as amphiphilic block copolymers, hydrophobic polymers such as polystyrene, poly(lactic acid), poly(lactic co-glycolic acid), poly(glycolic acid), poly(caprolactone) and other biocompatible polymers known to those skilled in the art.


The core may alternatively be a hollow core, which has at least some space in the center region of a shell material. Hollow cores include liposomal cores and niosomes. A liposomal core as used herein refers to a centrally located core compartment formed by a component of the lipids or phospholipids that form a lipid bilayer. “Liposomes” are artificial, self closed vesicular structure of various sizes and structures, where one or several membranes encapsulate an aqueous core. Most typically liposome membranes are formed from lipid bilayers membranes, where the hydrophilic head groups are oriented towards the aqueous environment and the lipid chains are embedded in the lipophilic core. Liposomes can be formed as well from other amphiphilic monomeric and polymeric molecules, such as polymers, like block copolymers, or polypeptides. Unilamellar vesicles are liposomes defined by a single membrane enclosing an aqueous space. In contrast, oligo- or multilamellar vesicles are built up of several membranes. Typically, the membranes are roughly 4 nm thick and are composed of amphiphilic lipids, such as phospholipids, of natural or synthetic origin. Optionally, the membrane properties can be modified by the incorporation of other lipids such as sterols or cholic acid derivatives.


The lipid bilayer is composed of two layers of lipid molecules. Each lipid molecule in a layer is oriented substantially parallel to adjacent lipid bilayers, and two layers that form a bilayer have the polar ends of their molecules exposed to the aqueous phase and the non-polar ends adjacent to each other. The central aqueous region of the liposomal core may be empty or filled fully or partially with water, an aqueous emulsion, oligonucleotides, or other therapeutic or diagnostic agents.


Niosomes are vesicles formed from non-ionic surfactant oriented in a bilayer. Niosomes commonly have cholesterol added as an excipient, but other lipid-based and non-lipid-based constituents can also be included. Methods for preparation of niosomes are known in the art. In some embodiments polyethylene glycol (PEG) is included during or following niosome preparation. Niosome vesicles are structurally and functionally analogous to liposomes, but are based on non-ionic surfactant rather than lipid as the primary constiuent. Common non-ionic surfactants used include sorbitans (spans) or polysorbates (tween); however, a wide variety of non-ionic surfactants can be used to prepare niosomes.


“Lipid” refers to its conventional sense as a generic term encompassing fats, lipids, alcohol-ether-soluble constituents of protoplasm, which are insoluble in water. Lipids usually consist of a hydrophilic and a hydrophobic moiety. In water lipids can self organize to form bilayers membranes, where the hydrophilic moieties (head groups) are oriented towards the aqueous phase, and the lipophilic moieties (acyl chains) are embedded in the bilayers core. Lipids can comprise as well two hydrophilic moieties (bola amphiphiles). In that case, membranes may be formed from a single lipid layer, and not a bilayer. Typical examples for lipids in the current context are fats, fatty oils, essential oils, waxes, steroid, sterols, phospholipids, glycolipids, sulpholipids, aminolipids, chromolipids, and fatty acids. The term encompasses both naturally occurring and synthetic lipids. Preferred lipids in connection with the present invention are: steroids and sterol, particularly cholesterol, phospholipids, including phosphatidyl, phosphatidylcholines and phosphatidylethanolamines and sphingomyelins. Where there are fatty acids, they could be about 12-24 carbon chains in length, containing up to 6 double bonds. The fatty acids are linked to the backbone, which may be derived from glycerol. The fatty acids within one lipid can be different (asymmetric), or there may be only 1 fatty acid chain present, e.g. lysolecithins. Mixed formulations are also possible, particularly when the non-cationic lipids are derived from natural sources, such as lecithins (phosphatidylcholines) purified from egg yolk, bovine heart, brain, liver or soybean.


The liposomal core can be constructed from one or more lipids known to those in the art including but not limited to: 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dimyristoyl-sn-phosphatidylcholine (DMPC), 1-palmitoyl-2-oleoyl-sn-phosphatidylcholine (POPC), 1,2-distearoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (DSPG), 1,2-dioleoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (DOPG), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-di-(9Z-octadecenoyl)-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine (DPPE), sphingolipids such as sphingosine, sphingosine phosphate, methylated sphingosines and sphinganines, ceramides, ceramide phosphates, 1-0 acyl ceramides, dihydroceramides, 2-hydroxy ceramides, sphingomyelin, glycosylated sphingolipids, sulfatides, gangliosides, phosphosphingolipids, and phytosphingosines of various lengths and saturation states and their derivatives, phospholipids such as phosphatidylcholines, lysophosphatidylcholines, phosphatidic acids, lysophosphatidic acids, cyclic LPA, phosphatidylethanolamines, lysophosphatidylethanolamines, phosphatidylglycerols, lysophosphatidylglycerols, phosphatidylserines, lysophosphatidylserines, phosphatidylinositols, inositol phosphates, LPI, cardiolipins, lysocardiolipins, bis(monoacylglycero) phosphates, (diacylglycero) phosphates, ether lipids, diphytanyl ether lipids, and plasmalogens of various lengths, saturation states, and their derivatives, sterols such as cholesterol, desmosterol, stigmasterol, lanosterol, lathosterol, diosgenin, sitosterol, zymosterol, zymostenol, 14-demethyl-lanosterol, cholesterol sulfate, DHEA, DHEA sulfate, 14-demethyl-14-dehydrlanosterol, sitostanol, campesterol, ether anionic lipids, ether cationic lipids, lanthanide chelating lipids, A-ring substituted oxysterols, B-ring substituted oxysterols, D-ring substituted oxysterols, side-chain substituted oxysterols, double substituted oxysterols, cholestanoic acid derivatives, fluorinated sterols, fluorescent sterols, sulfonated sterols, phosphorylated sterols, and polyunsaturated sterols of different lengths, saturation states, and their derivatives.


In certain embodiments, the diameter of the core is from 1 nm to about 250 nm in mean diameter, about 1 nm to about 240 nm in mean diameter, about 1 nm to about 230 nm in mean diameter, about 1 nm to about 220 nm in mean diameter, about 1 nm to about 210 nm in mean diameter, about 1 nm to about 200 nm in mean diameter, about 1 nm to about 190 nm in mean diameter, about 1 nm to about 180 nm in mean diameter, about 1 nm to about 170 in mean diameter, about 1 nm to about 160 nm in mean diameter, about 1 nm to about 150 nm in mean diameter, about 1 nm to about 140 nm in mean diameter, about 1 nm to about 130 nm in mean diameter, about 1 nm to about 120 nm in mean diameter, about 1 nm to about 110 nm in mean diameter, about 1 nm to about 100 nm in mean diameter, about 1 nm to about 90 nm in mean diameter, about 1 nm to about 80 nm in mean diameter, about 1 nm to about 70 nm in mean diameter, about 1 nm to about 60 nm in mean diameter, about 1 nm to about 50 nm in mean diameter, about 1 nm to about 40 nm in mean diameter, about 1 nm to about 30 nm in mean diameter, or about 1 nm to about 20 nm in mean diameter, or about 1 nm to about 10 nm in mean diameter.


The oligonucleotides may be positioned on the exterior of the core, within the walls of the core and/or in the center of the core. An oligonucleotide that is positioned on the core is typically referred to as coupled to the core. Coupled may be direct or indirect. In some embodiments at least 5, 10, 15, 25, 50, 75, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 5000 or 10,000 oligonucleotides or any range combination thereof are on the exterior of the core. In some embodiments, 1-1000, 10-500, 50-250, or 50-300 oligonucleotides are present on the surface.


The oligonucleotides of the oligonucleotide shell may be oriented in a variety of directions. In some embodiments the oligonucleotides are oriented radially outwards. The orientation of these oligonucleotides can be either 5′ distal/3′ terminal in relation to the core, or 3′ distal/5′terminal in relation to the core, or laterally oriented around the core. In one embodiment one or a multiplicity of different oligonucleotides are present on the same surface of a single SNA. In all cases, at least 1 oligonucleotide is present on the surface but up to 10,000 can be present.


The oligonucleotides may be linked to the core or to one another and/or to other molecules such an active agents either directly or indirectly through a linker. The oligonucleotides may be conjugated to a linker via the 5′ end or the 3′ end, e.g. [Sequence, 5′-3′]-Linker or Linker-[Sequence, 5′-3′]. Some or all of the oligonucleotides of the nanostructure may be linked to one another either directly or indirectly through a covalent or non-covalent linkage. The linkage of one oligonucleotide to another oligonucleotide may be in addition to or alternatively to the linkage of that oligonucleotide to liposomal core.


The oligonucleotide shell may be anchored to the surface of the core through one or multiple of linker molecules, including but not limited to: any chemical structure containing one or multiple thiols, such as the various chain length alkane thiols, cyclic dithiol, lipoic acid, or other thiol linkers known to those skilled in the art.


In an embodiment containing a liposomal core, the oligonucleotide shell may be anchored to the surface of the liposomal core through conjugation to one or a multiplicity of linker molecules including but not limited to: tocopherols, sphingolipids such as sphingosine, sphingosine phosphate, methylated sphingosines and sphinganines, ceramides, ceramide phosphates, 1-0 acyl ceramides, dihydroceramides, 2-hydroxy ceramides, sphingomyelin, glycosylated sphingolipids, sulfatides, gangliosides, phosphosphingolipids, and phytosphingosines of various lengths and saturation states and their derivatives, phospholipids such as phosphatidylcholines, lysophosphatidylcholines, phosphatidic acids, lysophosphatidic acids, cyclic LPA, phosphatidylethanolamines, lysophosphatidylethanolamines, phosphatidylglycerols, lysophosphatidylglycerols, phosphatidylserines, lysophosphatidylserines, phosphatidylinositols, inositol phosphates, LPI, cardiolipins, lysocardiolipins, bis(monoacylglycero) phosphates, (diacylglycero) phosphates, ether lipids, diphytanyl ether lipids, and plasmalogens of various lengths, saturation states, and their derivatives, sterols such as cholesterol, desmosterol, stigmasterol, lanosterol, lathosterol, diosgenin, sitosterol, zymosterol, zymostenol, 14-demethyl-lanosterol, cholesterol sulfate, DHEA, DHEA sulfate, 14-demethyl-14-dehydrlanosterol, sitostanol, campesterol, ether anionic lipids, ether cationic lipids, lanthanide chelating lipids, A-ring substituted oxysterols, B-ring substituted oxysterols, D-ring substituted oxysterols, side-chain substituted oxysterols, double substituted oxysterols, cholestanoic acid derivatives, fluorinated sterols, fluorescent sterols, sulfonated sterols, phosphorylated sterols, and polyunsaturated sterols of different lengths, saturation states, and their derivatives.


The oligonucleotide may also be associated with the core by being embedded within the core (liposomal core) or it may be attached or linked, either indirectly (i.e. non-covalently or covalently through other molecules such a linkers) or directly (i.e. covalently).


The invention also includes articles, which refers to any one or collection of components. In some embodiments the articles are kits. The articles include pharmaceutical or diagnostic grade compounds of the invention in one or more containers. The article may include instructions or labels promoting or describing the use of the compounds of the invention.


As used herein, “promoted” includes all methods of doing business including methods of education, hospital and other clinical instruction, pharmaceutical industry activity including pharmaceutical sales, and any advertising or other promotional activity including written, oral and electronic communication of any form, associated with compositions of the invention in connection with treatment of inflammatory disorders.


“Instructions” can define a component of promotion, and typically involve written instructions on or associated with packaging of compositions of the invention. Instructions also can include any oral or electronic instructions provided in any manner.


Thus the agents described herein may, in some embodiments, be assembled into pharmaceutical or diagnostic or research kits to facilitate their use in therapeutic, diagnostic or research applications. A kit may include one or more containers housing the components of the invention and instructions for use. Specifically, such kits may include one or more agents described herein, along with instructions describing the intended therapeutic application and the proper administration of these agents. In certain embodiments agents in a kit may be in a pharmaceutical formulation or pharmaceutical composition and dosage suitable for a particular application and for a method of administration of the agents.


The kit may be designed to facilitate use of the methods described herein by physicians and can take many forms. Each of the compositions of the kit, where applicable, may be provided in liquid form (e.g., in solution), or in solid form, (e.g., a dry powder). In certain cases, some of the compositions may be constitutable or otherwise processable (e.g., to an active form), for example, by the addition of a suitable solvent or other species (for example, water or a cell culture medium), which may or may not be provided with the kit. As used herein, “instructions” can define a component of instruction and/or promotion, and typically involve written instructions on or associated with packaging of the invention. Instructions also can include any oral or electronic instructions provided in any manner such that a user will clearly recognize that the instructions are to be associated with the kit, for example, audiovisual (e.g., videotape, DVD, etc.), Internet, and/or web-based communications, etc. The written instructions may be in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which instructions can also reflects approval by the agency of manufacture, use or sale for human administration.


The kit may contain any one or more of the components described herein in one or more containers. As an example, in one embodiment, the kit may include instructions for mixing one or more components of the kit and/or isolating and mixing a sample and applying to a subject. The kit may include a container housing agents described herein. The agents may be prepared sterilely, packaged in syringe and shipped refrigerated. Alternatively it may be housed in a vial or other container for storage. A second container may have other agents prepared sterilely. Alternatively the kit may include the active agents premixed and shipped in a syringe, vial, tube, or other container.


The kit may have a variety of forms, such as a blister pouch, a shrink wrapped pouch, a vacuum sealable pouch, a sealable thermoformed tray, or a similar pouch or tray form, with the accessories loosely packed within the pouch, one or more tubes, containers, a box or a bag. The kit may be sterilized after the accessories are added, thereby allowing the individual accessories in the container to be otherwise unwrapped. The kits can be sterilized using any appropriate sterilization techniques, such as radiation sterilization, heat sterilization, or other sterilization methods known in the art. The kit may also include other components, depending on the specific application, for example, containers, cell media, salts, buffers, reagents, syringes, needles, a fabric, such as gauze, for applying or removing a disinfecting agent, disposable gloves, a support for the agents prior to administration etc.


The compositions of the kit may be provided as any suitable form, for example, as liquid solutions or as dried powders. When the composition provided is a dry powder, the powder may be reconstituted by the addition of a suitable solvent, which may also be provided. In embodiments where liquid forms of the composition are sued, the liquid form may be concentrated or ready to use. The solvent will depend on the compound and the mode of use or administration. Suitable solvents for drug compositions are well known and are available in the literature. The solvent will depend on the compound and the mode of use or administration.


The kits, in one set of embodiments, may comprise a carrier means being compartmentalized to receive in close confinement one or more container means such as vials, tubes, and the like, each of the container means comprising one of the separate elements to be used in the method. For example, one of the containers may comprise a positive control for an assay. Additionally, the kit may include containers for other components, for example, buffers useful in the assay.


The present invention also encompasses a finished packaged and labeled pharmaceutical product. This article of manufacture includes the appropriate unit dosage form in an appropriate vessel or container such as a glass vial or other container that is hermetically sealed. In the case of dosage forms suitable for parenteral administration the active ingredient is sterile and suitable for administration as a particulate free solution. In other words, the invention encompasses both parenteral solutions and lyophilized powders, each being sterile, and the latter being suitable for reconstitution prior to injection. Alternatively, the unit dosage form may be a solid suitable for oral, transdermal, topical or mucosal delivery.


In a preferred embodiment, the unit dosage form is suitable for intravenous, intramuscular or subcutaneous delivery. Thus, the invention encompasses solutions, preferably sterile, suitable for each delivery route.


As with any pharmaceutical product, the packaging material and container are designed to protect the stability of the product during storage and shipment. Further, the products of the invention include instructions for use or other informational material that advise the physician, technician or patient on how to appropriately prevent or treat the inflammatory disease or disorder. In other words, the article of manufacture includes instruction means indicating or suggesting a dosing regimen including, but not limited to, actual doses, monitoring procedures and other monitoring information.


More specifically, the invention provides an article of manufacture comprising packaging material, such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like; and at least one unit dosage form of a pharmaceutical agent contained within said packaging material. The invention also provides an article of manufacture comprising packaging material, such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like; and at least one unit dosage form of each pharmaceutical agent contained within said packaging material. The invention further provides an article of manufacture comprising packaging material, such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like; and at least one unit dosage form of each pharmaceutical agent contained within said packaging material. The invention further provides an article of manufacture comprising a needle or syringe, preferably packaged in sterile form, for injection of the formulation, and/or a packaged alcohol pad.


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
Example 1. Liposomal Spherical Nucleic Acid (SNA) Constructs Presenting Antisense Oligonucleotides (ASO) for Specific Knockdown of Interleukin 17 Receptor mRNA

Results


Experiment 1. In Vitro IL-17RA mRNA Knockdown Using Targeted SNAs


HFK cells were treated with 294 SNAs presenting one ASO sequence each of the sequences in Table 1 and human IL-17RA mRNA expression was measured by qRT-PCR. This was performed twice and the three SNAs showing the greatest inhibition of IL-17RA mRNA in multiple assays and having low off-target effects were chosen for further optimization by chemical modification. HFK cells were treated at larger dose ranges with SNAs presenting one ASO sequence of either IL17RA_219, IL17RA_282, IL17RA_285 or a non-complementary control. Targeted SNAs demonstrate target specific mRNA inhibition in a dose-dependent manner while the non-complementary control SNA has no considerable inhibition at all concentrations tested (FIG. 1). Targeted SNAs demonstrate similar IL-17RA mRNA inhibition in HEKa cells. Analyzing the dose-response data as a non-linear fit and calculating the half maximal inhibitory concentration (IC50) results in IC50 values of 124.2, 9.57 and 34.64 nM for IL17RA_285, IL17RA_219 and IL17RA_282, respectively. These results demonstrate that SNAs targeting IL-17RA mRNA can effectively inhibit gene expression in a specific manner in HFK cells.


Experiment 2. IL-17RA mRNA Knockdown Using Topically Applied Targeted SNAs in the Human Skin Equivalent, EpiDerm-FT™


A human skin equivalent model treated topically with SNAs presenting either ASO sequence IL17RA_219 or IL17RA_282 targeting human IL-17RA mRNA resulted in target mRNA specific inhibition. IL17RA_219 and IL17RA_282 were chosen as the lead candidate compounds for further testing after taking into consideration the efficacy data collected in experiment 1. ASOs IL17RA_219 and IL17RA_282 exhibited the best combination of IC50 value, homology to other human mRNA sequences (as determined by BLAST) and cross-species conservation. IL-17RA mRNA expression is decreased in human skin equivalent tissues treated topically with SNAs presenting either ASO IL17RA_219 or ASO IL17RA_282 for 48 hours. Tissues treated with SNAs presenting the control ASO do not demonstrate an appreciable decrease in IL-17RA mRNA expression as measured by qRT-PCR (FIG. 2). Further, IL17RA_219 and IL17RA_282 exhibited no toxicity in HFKs and no cytokine induction in HFKs, HEKa cells and PBMCs from four different donors, up to 20 μM DNA concentration. For these reasons, IL17RA_219 and IL17RA_282 were chosen for further testing. SNAs targeting IL-17RA delivered topically were shown to specifically inhibit mRNA expression in a human skin equivalent model.


Experiment 3. Ex Vivo IL-17RA mRNA Knockdown Using Topically Applied Targeted SNAs in Human Skin Explants


Human skin explants treated topically with SNAs presenting either ASO sequence IL17RA_219 or ASO sequence IL17RA_282 targeting human IL-17RA mRNA resulted in target mRNA specific inhibition. IL-17RA mRNA expression is decreased in human skin explant biopsies treated topically with SNAs presenting either ASO IL17RA_219 or ASO IL17RA_282 for 96 hours. Biopsies treated with SNAs presenting the control ASO do not demonstrate an appreciable decrease in IL-17RA mRNA expression as measured by qRT-PCR (FIG. 3). SNAs targeting IL-17RA delivered topically can specifically inhibit mRNA expression in human skin explants.


Experiment 4. TNF mRNA Knockdown Using a Mixture of SNAs


SNAs presenting either an anti-TNF or an anti-IL17RA ASO sequence were mixed together and co-administered to HFK cells. Mixed SNAs demonstrated target mRNA specific knockdown corresponding to the ASO sequence identity (FIG. 4, FIG. 5).



FIG. 4 shows TNF mRNA expression decreased in HFK cells treated with a mixture of anti-TNF-SNAs and anti-IL17RA-SNAs. FIG. 5 shows IL17RA mRNA expression is decreased in HFK cells treated with a mixture of anti-TNF-SNAs and anti-IL17RA-SNAs. Cells treated with control-SNAs do not demonstrate an appreciable decrease in TNF mRNA levels.


These results demonstrate that mixtures of SNAs targeting more than one mRNA can be co-administered and perform gene knockdown with respect to each target.


Experiment 5. Comparison of TNF mRNA Knockdown and IL17RA Knockdown Using Multiplexed SNAs


SNAs presenting one, two, or three ASO sequences demonstrated target mRNA specific knockdown in HFK cells corresponding to the ASO sequence identity. Only data obtained using SNAs multiplexed with two ASO sequences. SNAs were shown to serve as a platform capable of presenting multiple therapeutic modalities. These results demonstrate that ASO sequences targeting more than one mRNA can be co-delivered on the same SNA particle and perform gene knockdown with respect to each target (FIG. 7, FIG. 8).


When the two gene targets are in a related biological pathway, an additive knockdown effect can be observed for the downstream target (FIG. 8). TNF and IL17A cytokine signaling have been shown to have an additive and synergistic effect in their proinflammatory properties(3). This relationship indicates a shared or highly related signaling pathway, though the exact relationship is not well understood(4). The mRNA of the TNF cytokine as well as the receptor for IL17A, IL17RA were targeted. It is likely that the reduction of secreted TNF cytokine has an effect on IL17RA receptor turnover and expression.


Materials and Methods


Antisense Sequence Design. Antisense sequences, 18 nucleotides in length, targeting human IL-17RA mRNA (GeneBank Accession No. NM_014339.6) were determined based on the following parameters: Sfold Algorithm(2), % GC content, proximity to start codon, and the exclusion of CpG motifs (Table 1). Unless otherwise noted, antisense oligonucleotide sequences are divided into 3 regions: an internal, phosphorothioate linked ‘gap’ region flanked by two external, 2′-O-methyl ‘wings’. This ‘gap-mer’ sequence is modified at the 3′ end with two consecutive hexa(ethylene glycol) spacers and a terminal cholesterol.


Antisense oligonucleotide synthesis. All oligonucleotides were synthesized at the 1 μmole scale employing universal UnyLinker Support 1000 Å (ChemGenes) on the Bioautomation MerMade 48 oligonucleotide synthesizer. The DNA, RNA, 2′-O-Me monomers, hexa(ethylene glycol) spacers and cholesterol modifiers were obtained from ChemGenes Corporation. Coupling time was 1 minute for standard DNA bases and 3 minutes for modified bases. Linkages were either standard phosphodiesters or phosphorothioates made with 0.2 M phenylacetyl disulfide (PADS) in 1:1 lutidine:ACN as the sulfurization agent. Synthesis was performed DMT-off, in the 5′ to 3′ direction. After synthesis, the oligonucleotides were cleaved from the support and deprotected using a 4:1 mixture of ammonium hydroxide and ethanol at 55° C. for 16 hours. The oligonucleotides were purified via high performance liquid chromatography (HPLC) techniques. Molecular weights and extinction coefficients were determined using IDT OligoAnalyzer. Verification of oligonucleotide molecular weight was performed using matrix-assisted laser desorption/ionization (MALDI). Oligonucleotide concentration was determined by UV-absorbance at 260 nm on a microplate reader (BioTek) together with the calculated extinction coefficient from the IDT OligoAnalyzer. All oligonucleotides were sterile filtered using 0.2 μm syringe filters (VWR). The oligonucleotide sequences are listed in Table 1.


Liposome synthesis. Liposomes were synthesized by extrusion of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) hydrated in phosphate buffered saline solution (PBS) (137 mM NaCl, 10 mM phosphate, 2.7 mM KCl, pH 7.4, Hyclone) using 47 mm diameter polycarbonate membranes with 50 nm pores (Sterlitech). Liposome diameters were measured using dynamic light scattering using a Malvern Zetasizer Nano (Malvern Instruments). Lipid concentration was determined using a phospholipid assay kit (Sigma).


Cell Culture. Human neonatal foreskin keratinocytes (HFKs) and adult human epidermal keratinocytes (HEKa) were cultured in M154 media (Life Technologies) supplemented with Human Keratinocyte Growth Supplement (Life Technologies), 0.07 mM CaCl2, 10 μg/mL gentamicin, and 0.25m/mL amphotericin B. Cells were maintained at 37° C. in a 5% CO2 humidified incubator. Fresh primary human peripheral blood mononuclear cells (PBMCs) from four 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.


EpiDerm-FT™ Human Skin Equivalent Culture. The human skin equivalent model, EpiDerm-FT™ (Mattek), tissues were cultured according to the manufacturer's instructions. Briefly, upon receipt, tissues were incubated overnight with supplied maintenance media at 37° C. in a 5% CO2 humidified incubator. The following day, and every day after that for the length of the experiment, the media was replaced with fresh media. The tissues were maintained at 37° C. in a 5% CO2 humidified incubator throughout the length of the experiment.


Human Skin Explant Culture. Fresh human skin explant (Zenbio) was cleaned upon receipt. 8 mm punch biopsies were taken from the explant and placed into trans-well stands for culturing. DMEM supplemented with 2% FBS, 10 μg/mL gentamicin, and 0.25 μg/mL amphotericin B was supplied to the underside of the biopsies and changed every day for the length of the experiment. The biopsies were maintained at 37° C. in a 5% CO2 humidified incubator throughout the length of the experiment.


SNA Synthesis. SNAs were formulated by mixing a 100× molar excess (unless otherwise stated) of cholesterol-modified oligonucleotide to a liposome suspension in PBS and storing them overnight, protected from light, at 4° C. 294 different oligonucleotides (Table 1) were used for the synthesis process, specifically targeting human IL-17RA mRNA, and a control oligonucleotide of the same length and ‘gap-mer’ chemical scheme that does not have any complementarity to known human genes (confirmed by BLAST).


Multiplex SNA Synthesis. SNAs were formulated by mixing a 200× molar excess (unless otherwise stated) of cholesterol-modified oligonucleotide to a liposome suspension in PBS and storing them overnight, protected from light, at 4° C. Four different oligonucleotides were used for the synthesis process: an antisense oligonucleotide targeting human IL-17RA, an antisense oligonucleotide targeting human TNF, an antisense oligonucleotide targeting human IL22RA1, and a same length control oligonucleotide that does not have any complementarity to known genes (confirmed by BLAST). For monoplex SNAs, the oligonucleotide population consisted of 33% targeted antisense oligonucleotide and 66% control oligonucleotide. For multiplex SNAs containing two different targeted antisense oligonucleotides, the oligonucleotide population consisted of 33% of each targeted antisense oligonucleotide and the remaining 33% control oligonucleotide (FIG. 6). For multiplex SNAs containing three different targeted antisense oligonucleotides, the oligonucleotide population consisted of 33% of each targeted antisense oligonucleotide. Control SNAs consisted of 100% of the control oligonucleotide (FIG. 6D).


Cell Culture Studies. HFKs were seeded at passage 5 in 96-well, tissue culture plates at a cell density of 17,000 cells per well. HEKa cells were seeded at passage 5 in 96-well, tissue culture plates at a cell density of 17,000 cells per well. Fresh PBMCs were isolated from cell suspension by centrifugation and seeded in 96-well, tissue culture plates at a cell density of 200,000 cells per well. Cells were allowed to rest in the incubator overnight following plating. Cells were treated with either an IL-17RA targeted antisense SNA or a non-complementary control SNA (confirmed by NCBI Blast), comprising the same ‘gap-mer’ design and 3′-chemical modifications, in fresh maintenance media. Unless otherwise stated, all treatments lasted 24 hours.


For the human skin equivalent model, EpiDerm-FT™, and human skin explant tissues, treatments were applied in biological triplicate. Tissues were treated topically with either an SNA presenting the IL17RA_219 ASO, an SNA presenting the IL17RA_282 ASO, an SNA presenting the non-complementary control ASO or left untreated. The EpiDerm-FT™ tissues were treated for 48 hours and the human skin explant tissues were treated for 96 hours.


For multiplex SNA experiments, HFKs were seeded at passage 5 in 96-well, tissue culture plates at a cell density of 17,000 cells per well. Cells were allowed to rest in the incubator overnight following plating. Cells were treated in triplicate with either an IL-17RA targeted antisense SNA, TNF targeted antisense SNA, a multiplex SNA targeting more than one transcript or a non-complementary control SNA (confirmed by NCBI Blast), comprising the same ‘gap-mer’ design and 3′-chemical modifications, at concentrations of 1000, 100, 10 and 1 nM in fresh maintenance media.


Alamar Blue® Viability Assay. For viability studies, HFKs were seeded at passage 5 in 96-well, tissue culture plates at a cell density of 15,000 cells per well. Cells were allowed to rest in the incubator overnight following plating. Cells were treated in triplicate with either an IL-17RA targeted antisense SNA or a non-complementary control (confirmed by NCBI Blast), comprising the same ‘gap-mer’ design and 3′ chemical modifications, at concentrations of 20, 10, 2 and 1 μM for 24 hours. Briefly, treatments were removed and replace with a 10% solution of AlamarBlue® reagent in M154 maintenance media. Cells were incubated at 37° C. for 3 hours in a 5% CO2 humidified incubator before measuring the fluorescence of each well (excitation 570 nm, emission 585 nm) with a microplate reader (BioTek). Percent viability was calculated by comparing fluorescent values of each treatment to those of the untreated cells.


Cytokine Q-Plex Array. After 24 hours of treatment, the cells in the 96-well tissue culture plates were pelleted by centrifugation; the supernatants were transferred to new plates and stored at −80° C. Cytokine quantification of the supernatants was performed using a Q-Plex chemiluminescent array (Quansys) following the manufacturer's instructions. Each plate was custom built to allow for the detection of IL-12p40, TNF, IP-10, IL-6, RANTES, IL-4, IL-5, IL-10, IL-17, IL-22 and MCP-1 within each well. Briefly, a standard curve was prepared using a cytokine stock of known concentration and diluting it as indicated. The cell culture supernatants were diluted 1:2 using the provided sample buffer. The cell culture samples and standard curve samples were added to the 96-well Q-plex plate and incubated at room temperature for 1 hour with shaking. The plate was then washed three times with the provided wash buffer. Then 50 μL of provided Detection mix was added to each well and the plate was incubated again for 1 hour at room temperature with shaking. Again, the plate was washed three times with wash buffer, followed by incubation with 50 μL of provided Streptavidin-HRP solution for 15 minutes at room temperature with shaking. Finally, the plate was washed six times in wash buffer and 50 μL of a prepared substrate was added to each well. Within 15 minutes, the plate was imaged using a Bio-Rad ChemiDoc XRS+ imager and analyzed using the Q-view software (Quansys). All data was fit to the standard curves for each individual cytokine and plotted in comparison to untreated cells.


RNA Extraction and Quantitative Reverse Transcriptase Polymerase Chain Reaction (qRT-PCR). HFK and HEKa cells were lysed in RLT Buffer (Qiagen) at 24 hours post-transfection. EpiDerm-FT™ tissues and human skin explant tissues were bead homogenized in RLT buffer at 48 and 96 hours of treatment, respectively. RNA was isolated from lysates using the RNEasy 96-well kit (Qiagen) according to the manufacturer's instructions. cDNA was then synthesized from RNA isolates using the cDNA High Capacity Reverse Transcription Kit (Life Technologies). Samples were run on a thermocycler at 25° C. for 10 minutes, 37° C. for 90 minutes, 85° C. for 5 minutes and held at 4° C. to generate cDNA. qPCR was performed using 6 μL of the synthesized cDNA, 4.66 μL LightCycler480 Probes Master Mix (Roche), 0.47 μL target specific FAM-labeled probe and primers, and 0.37 μL human Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) specific HEX-labeled probe and primers per reaction well of a 384-well optical reaction plate (Roche). The primer and probe sets for IL-17RA were purchased from ThermoFisher Scientific (catalogue number Hs01064648_m1). The primer and probe sets for TNF and GAPDH were designed using the known human genome sequence (NCBI reference sequences NM_000594.3 and NM_002046.5, respectively) and were found to be specific by “blastn” analysis (NCBI). The oligonucleotide sequences used for TNF were: forward 5′-GCT GCA CTT TGG AGT GAT CG-3′ (SEQ ID NO: 1), reverse 5′-GTT TGC TAC AAC ATG GGC TAC AG-3′ (SEQ ID NO: 2), probe 5′-FAM-CCC AGG CAG TCA GAT CAT CTT CTC GA-BHQ1-3′ (SEQ ID NO: 3). The oligonucleotide sequences used for GAPDH were: forward 5′-CAA GGT CAT CCA TGA CAA CTT TG-3′ (SEQ ID NO: 4), reverse 5′-GGG CCA TCC ACA GTC TTC T-3′ (SEQ ID NO: 5), probe 5′-HEX-ACC ACA GTC CAT GCC ATC ACT GCC A-BHQ1-3′ (SEQ ID NO: 6). FAM is 6-fluorescein amidite, HEX is hexachloro-fluorescein, and BHQ1 is a black hole quencher 1. qPCR reactions, in technical duplicate, were carried out on the Roche Lightcycler 480 under the following conditions: initial denaturation at 95° C. for 10 minutes and then 50 cycles of denaturation at 95° C. for 10 seconds, annealing at 60° C. for 30 seconds and extension at 72° C. for 1 second. Cp values were obtained by analysis with the 2nd derivative method. Relative gene expression was determined by normalization with the housekeeping gene (GAPDH) and the ΔΔ-Ct method. Each treatment was compared to its respective concentration control.









TABLE 1







Oligonucleotide Sequences


Special bases used in the oligonucleotides are


as follows: mN = 2′ O-methyl RNA, /iSp18/ =


hexa(ethylene glycol) spacer, * = phosphoro-


thioate, /3CholTEG/ = 3′ cholesterol













% IL17RA




SEQ
mRNA


Compound
Oligonucleotide Sequence
ID
Expres-


ID
(5' to 3')
NO:
sion





IL17RA_001
mUmCmGmCmGmGA*G*G*G*C*T*mCmGm
  7
 67



GmCmCmC/iSp18//iSp18//3CholTEG/







IL17RA_002
mGmUmCmGmCmGG*A*G*G*G*C*mUmCm
  8
 35



GmGmCmC/iSp18//iSp18//3CholTEG/







IL17RA_003
mCmGmUmCmGmCG*G*A*G*G*G*mCmUm
  9
 36



CmGmGmC/iSp18//iSp18//3CholTEG/







IL17RA_004
mAmCmGmGmCmGG*G*C*T*G*C*mGmUm
 10
 49



GmCmGmG/iSp18//iSp18//3CholTEG/







IL17RA_005
mGmAmCmGmGmCG*G*G*C*T*G*mCmGm
 11
 40



UmGmCmG/iSp18//iSp18//3CholTEG/







IL17RA_006
mAmCmUmCmUmGC*A*C*C*C*T*mCmGm
 12
 35



AmGmGmU/iSp18//iSp18//3CholTEG/







IL17RA_007
mGmGmGmCmUmGC*C*C*A*G*C*mAmGm
 13
 30



CmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_008
mUmGmUmGmUmGG*G*T*C*T*G*mUmGm
 14
 73



AmGmGmA/iSp18//iSp18//3CholTEG/







IL17RA_009
mGmGmCmGmUmGT*G*T*G*G*G*mUmCm
 15
 43



UmGmUmG/iSp18//iSp18//3CholTEG/







IL17RA_010
mGmGmGmCmGmUG*T*G*T*G*G*mGmUm
 16
 35



CmUmGmU/iSp18//iSp18//3CholTEG/







IL17RA_011
mAmGmGmGmCmGT*G*T*G*T*G*mGmGm
 17
 54



UmCmUmG/iSp18//iSp18//3CholTEG/







IL17RAv012
mUmAmGmGmGmCG*T*G*T*G*T*mGmGm
 18
 49



GmUmCmU/iSp18//iSp18//3CholTEG/







IL17RA_013
mAmGmCmUmCmCT*G*G*A*G*A*mUmGm
 19
 38



UmAmGmC/iSp18//iSp18//3CholTEG/







IL17RA_014
mGmAmGmCmUmCC*T*G*G*A*G*mAmUm
 20
 49



GmUmAmG/iSp18//iSp18//3CholTEG/







IL17RA_015
mGmGmAmGmCmUC*C*T*G*G*A*mGmAm
 21
 88



UmGmUmA/iSp18//iSp18//3CholTEG/







IL17RA_016
mCmCmCmUmCmGG*G*G*G*G*C*mUmGm
 22
 91



CmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_017
mGmGmGmAmGmAG*A*G*T*G*G*mCmAm
 23
105



GmGmGmC/iSp18//iSp18//3CholTEG/







IL17RA_018
mCmGmGmGmAmGA*G*A*G*T*G*mGmCm
 24
106



AmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_019
mAmCmGmAmUmAA*C*C*A*G*A*mCmCm
 25
103



GmCmUmG/iSp18//iSp18//3CholTEG/







IL17RA_020
mGmGmGmAmGmCG*G*G*C*T*G*mUmGm
 26
 77



UmGmGmA/iSp18//iSp18//3CholTEG/







IL17RA_021
mAmCmAmUmAmGT*A*G*G*T*G*mCmAm
 27
 81



CmAmAmU/iSp18//iSp18//3CholTEG/







IL17RA_022
mCmAmCmAmUmAG*T*A*G*G*T*mGmCm
 28
 76



AmCmAmA/iSp18//iSp18//3CholTEG/







IL17RA_023
mGmGmGmUmCmUC*A*C*T*C*T*mGmCm
 29
 57



UmGmCmC/iSp18//iSp18//3CholTEG/







IL17RA_024
mGmGmGmCmAmGG*C*T*T*C*C*mAmCm
 30
 43



UmCmCmA/iSp18//iSp18//3CholTEG/







IL17RA_025
mAmAmAmGmCmUG*T*T*A*G*G*mAmGm
 31
 77



GmAmCmA/iSp18//iSp18//3CholTEG/







IL17RA_026
mUmAmAmAmGmCT*G*T*T*A*G*mGmAm
 32
 75



GmGmAmC/iSp18//iSp18//3CholTEG/







IL17RA_027
mAmUmAmAmAmGC*T*G*T*T*A*mGmGm
 33
 91



AmGmGmA/iSp18//iSp18//3CholTEG/







IL17RA_028
mAmCmCmGmGmCA*T*C*A*A*A*mUmUm
 34
 87



GmUmGmC/iSp18//iSp18//3CholTEG/







IL17RA_029
mAmAmCmCmGmGC*A*T*C*A*A*mAmUm
 35
 93



UmGmUmG/iSp18//iSp18//3CholTEG/







IL17RA_030
mCmUmGmGmAmUT*T*C*T*T*T*mUmGm
 36
 64



GmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_031
mGmCmUmGmGmAT*T*T*C*T*T*mUmUm
 37
 53



GmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_032
mGmGmCmUmGmGA*T*T*T*C*T*mUmUm
 38
 38



UmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_033
mGmGmGmCmUmGG*A*T*T*T*C*mUmUm
 39
 41



UmUmGmG/iSp18//iSp18//3CholTEG/







IL17RA_034
mGmGmGmGmCmUG*G*A*T*T*T*mCmUm
 40
 77



UmUmUmG/iSp18//iSp18//3CholTEG/







IL17RA_035
mGmGmGmAmGmGG*A*A*T*G*T*mGmAm
 41
 58



GmGmAmG/iSp18//iSp18//3CholTEG/







IL17RA_036
mGmCmUmGmAmAG*A*G*G*T*G*mGmGm
 42
 49



AmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_037
mGmAmGmCmCmUG*G*G*A*G*G*mUmCm
 43
 40



GmAmGmG/iSp18//iSp18//3CholTEG/







IL17RA_038
mGmGmGmAmGmGA*T*G*A*G*G*mCmGm
 44
 39



GmGmCmA/iSp18//iSp18//3CholTEG/







IL17RA_039
mUmGmGmGmAmGG*A*T*G*A*G*mGmCm
 45
 31



GmGmGmC/iSp18//iSp18//3CholTEG/







IL17RA_040
mUmUmUmAmGmGA*A*G*G*G*G*mAmG
 46
 60



mCmAmCmC/iSp18//iSp18//3CholTEG/







IL17RA_041
mAmUmUmUmAmGG*A*A*G*G*G*mGmA
 47
 57



mGmCmAmC/iSp18//iSp18//3CholTEG/







IL17RA_042
mUmAmUmUmUmAG*G*A*A*G*G*mGmG
 48
 57



mAmGmCmA/iSp18//iSp18//3CholTEG/







IL17RA_043
mUmUmAmUmUmUA*G*G*A*A*G*mGmG
 49
 23



mGmAmGmC/iSp18//iSp18//3CholTEG/







IL17RA_044
mCmAmUmUmUmAT*T*T*A*G*G*mAmAm
 50
 38



GmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_045
mUmCmAmUmUmUA*T*T*T*A*G*mGmAm
 51
 35



AmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_046
mGmGmGmAmUmGC*A*G*G*C*C*mCmGm
 52
 53



GmCmUmG/iSp18//iSp18//3CholTEG/







IL17RA_047
mAmAmAmGmAmGG*A*T*C*A*G*mUmGm
 53
 71



GmUmAmC/iSp18//iSp18//3CholTEG/







IL17RA_048
mGmAmAmAmGmAG*G*A*T*C*A*mGmUm
 54
 42



GmGmUmA/iSp18//iSp18//3CholTEG/







IL17RA_049
mUmGmGmGmUmUT*A*G*G*G*G*mUmA
 55
 48



mGmCmUmG/iSp18//iSp18//3CholTEG/







IL17RA_050
mAmUmGmGmGmUT*T*A*G*G*G*mGmUm
 56
 43



AmGmCmU/iSp18//iSp18//3CholTEG/







IL17RA_051
mAmAmUmGmGmGT*T*T*A*G*G*mGmGm
 57
 64



UmAmGmC/iSp18//iSp18//3CholTEG/







IL17RA_052
mUmGmCmAmAmUG*G*G*T*T*T*mAmGm
 58
 58



GmGmGmU/iSp18//iSp18//3CholTEG/







IL17RA_053
mGmUmGmCmAmAT*G*G*G*T*T*mUmAm
 59
 64



GmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_054
mUmGmUmGmCmAA*T*G*G*G*T*mUmUm
 60
 79



AmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_055
mUmUmGmUmGmCA*A*T*G*G*G*mUmUm
 61
 73



UmAmGmG/iSp18//iSp18//3CholTEG/







IL17RA_056
mCmUmUmGmUmGC*A*A*T*G*G*mGmUm
 62
 64



UmUmAmG/iSp18//iSp18//3CholTEG/







IL17RA_057
mGmCmUmUmGmUG*C*A*A*T*G*mGmGm
 63
 77



UmUmUmA/iSp18//iSp18//3CholTEG/







IL17RA_058
mAmGmCmUmUmGT*G*C*A*A*T*mGmGm
 64
 11



GmUmUmU/iSp18//iSp18//3CholTEG/







IL17RA_059
mCmAmGmCmUmUG*T*G*C*A*A*mUmGm
 65
 75



GmGmUmU/iSp18//iSp18//3CholTEG/







IL17RA_060
mGmGmGmCmAmUG*G*A*G*A*G*mCmCm
 66
 52



AmUmGmC/iSp18//iSp18//3CholTEG/







IL17RA_061
mUmCmUmGmGmGA*G*A*G*G*C*mGmA
 67
 47



mUmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_062
mGmGmGmAmGmGT*G*G*G*C*T*mGmGm
 68
 59



GmCmCmA/iSp18//iSp18//3CholTEG/







IL17RA_063
mAmCmUmCmCmCT*C*T*C*C*T*mCmCm
 69
 44



UmCmCmU/iSp18//iSp18//3CholTEG/







IL17RA_064
mUmAmCmUmCmCC*T*C*T*C*C*mUmCm
 70
 29



CmUmCmC/iSp18//iSp18//3CholTEG/







IL17RA_065
mGmGmGmAmGmGC*A*A*G*G*T*mCmUm
 71
 60



GmAmGmA/iSp18//iSp18//3CholTEG/







IL17RA_066
mUmCmAmGmGmGC*A*G*C*C*C*mGmGm
 72
 64



GmAmGmG/iSp18//iSp18//3CholTEG/







IL17RA_067
mCmAmCmUmCmCA*C*T*C*A*C*mCmUm
 73
 16



CmCmCmA/iSp18//iSp18//3CholTEG/







IL17RA_068
mGmGmGmUmGmCA*G*G*G*C*T*mUmCm
 74
 26



AmGmAmC/iSp18//iSp18//3CholTEG/







IL17RA_069
mGmCmGmGmGmUG*C*A*G*G*G*mCmUm
 75
 40



UmCmAmG/iSp18//iSp18//3CholTEG/







IL17RA_070
mCmUmGmUmUmUG*C*T*C*T*C*mCmUm
 76
 22



GmUmCmA/iSp18//iSp18//3CholTEG/







IL17RA_071
mCmCmUmGmUmUT*G*C*T*C*T*mCmCm
 77
 61



UmGmUmC/iSp18//iSp18//3CholTEG/







IL17RA_072
mUmCmCmUmGmUT*T*G*C*T*C*mUmCm
 78
 51



CmUmGmU/iSp18//iSp18//3CholTEG/







IL17RA_073
mGmUmCmCmUmGT*T*T*G*C*T*mCmUm
 79
 46



CmCmUmG/iSp18//iSp18//3CholTEG/







IL17RA_074
mUmGmUmCmCmUG*T*T*T*G*C*mUmCm
 80
 45



UmCmCmU/iSp18//iSp18//3CholTEG/







IL17RA_075
mCmUmGmUmCmCT*G*T*T*T*G*mCmUm
 81
 52



CmUmCmC/iSp18//iSp18//3CholTEG/







IL17RA_076
mGmGmCmUmAmGT*G*G*C*T*G*mGmGm
 82
 70



AmGmGmC/iSp18//iSp18//3CholTEG/







IL17RA_077
mGmGmAmAmUmGG*G*A*G*C*A*mGmA
 83
 63



mUmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_078
mGmGmGmCmUmUG*G*G*C*A*G*mGmU
 84
 32



mGmGmUmG/iSp18//iSp18//3CholTEG/







IL17RA_079
mGmGmGmAmUmGG*G*C*T*T*G*mGmGm
 85
 42



CmAmGmG/iSp18//iSp18//3CholTEG/







IL17RA_080
mGmGmUmCmUmCC*A*C*G*G*T*mGmAm
 86
 30



UmGmUmU/iSp18//iSp18//3CholTEG/







IL17RA_081
mGmUmAmAmUmGG*G*T*A*G*A*mUmU
 87
 22



mCmGmUmU/iSp18//iSp18//3CholTEG/







IL17RA_082
mGmGmUmAmAmUG*G*G*T*A*G*mAmU
 88
 40



mUmCmGmU/iSp18//iSp18//3CholTEG/







IL17RA_083
mUmGmGmUmAmAT*G*G*G*T*A*mGmAm
 89
 35



UmUmCmG/iSp18//iSp18//3CholTEG/







IL17RA_084
mCmUmGmGmUmAA*T*G*G*G*T*mAmGm
 90
 37



AmUmUmC/iSp18//iSp18//3CholTEG/







IL17RA_085
mGmGmGmCmGmCA*G*G*T*A*T*mGmUm
 91
 55



GmGmUmG/iSp18//iSp18//3CholTEG/







IL17RA_086
mGmGmCmUmGmAG*T*A*G*A*T*mGmAm
 92
 65



UmCmCmA/iSp18//iSp18//3CholTEG/







IL17RA_087
mCmGmCmGmCmCG*A*A*C*A*G*mGmUm
 93
 63



CmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_088
mGmUmAmGmAmGG*T*T*C*T*C*mAmCm
 94
 57



AmUmUmC/iSp18//iSp18//3CholTEG/







IL17RA_089
mCmCmGmCmCmCG*G*G*C*T*C*mCmGm
 95
 83



CmAmGmG/iSp18//iSp18//3CholTEG/







IL17RA_090
mCmCmCmUmGmGG*C*A*G*G*C*mUmUm
 96
 67



CmCmAmC/iSp18//iSp18//3CholTEG/







IL17RA_091
mCmAmAmUmGmGG*T*T*T*A*G*mGmGm
 97
 66



GmUmAmG/iSp18//iSp18//3CholTEG/







IL17RA_092
mGmCmAmAmUmGG*G*T*T*T*A*mGmGm
 98
 64



GmGmUmA/iSp18//iSp18//3CholTEG/







IL17RA_093
mGmGmAmGmGmAT*G*A*G*G*C*mGmGm
 99
 68



GmCmAmG/iSp18//iSp18//3CholTEG/







IL17RA_094
mGmUmGmCmGmGC*C*C*C*C*A*mUmGm
100
 79



GmCmCmC/isp18//isp18//3CholTEG/







IL17RA_095
mCmGmUmGmCmGG*C*C*C*C*C*mAmUm
101
 74



GmGmCmC/isp18//isp18//3CholTEG/







IL17RA_096
mGmCmGmUmGmCG*G*C*C*C*C*mCmAm
102
 72



UmGmGmC/isp18//isp18//3CholTEG/







IL17RA_097
mUmGmCmGmUmGC*G*G*C*C*C*mCmCm
103
 77



AmUmGmG/isp18//isp18//3CholTEG/







IL17RA_098
mCmUmGmCmGmUG*C*G*G*C*C*mCmCm
104
 58



CmAmUmG/isp18//isp18//3CholTEG/







IL17RA_099
mCmGmGmGmCmUG*C*G*T*G*C*mGmGm
105
 83



CmCmCmC/isp18//isp18//3CholTEG/







IL17RA_100
mGmCmGmGmGmCT*G*C*G*T*G*mCmGm
106
 84



GmCmCmC/isp18//isp18//3CholTEG/







IL17RA_101
mGmGmCmGmGmGC*T*G*C*G*T*mGmCm
107
 75



GmGmCmC/isp18//isp18//3CholTEG/







IL17RA_102
mUmCmGmCmAmGG*G*A*G*G*C*mGmCm
108
 87



CmAmCmC/isp18//isp18//3CholTEG/







IL17RA_103
mGmUmCmGmCmAG*G*G*A*G*G*mCmGm
109
 72



CmCmAmC/isp18//isp18//3CholTEG/







IL17RA_104
mUmUmCmGmAmUG*T*G*A*G*C*mCmAm
110
 74



CmGmGmG/isp18//isp18//3CholTEG/







IL17RA_105
mCmCmAmUmUmCG*A*T*G*T*G*mAmGm
111
 76



CmCmAmC/isp18//isp18//3CholTEG/







IL17RA_106
mGmAmUmAmAmCT*C*T*G*C*A*mCmCm
112
 84



CmUmCmG/isp18//isp18//3CholTEG/







IL17RA_107
mGmGmCmUmUmGG*G*C*A*G*G*mUmG
113
 65



mGmUmGmA/isp18//isp18//3CholTEG/







IL17RA_108
mUmGmGmGmCmUT*G*G*G*C*A*mGmGm
114
 37



UmGmGmU/isp18//isp18//3CholTEG/







IL17RA_109
mAmAmUmGmGmGT*A*G*A*T*T*mCmGm
115
 44



UmUmCmC/isp18//isp18//3CholTEG/







IL17RA_110
mUmAmAmUmGmGG*T*A*G*A*T*mUmCm
116
 62



GmUmUmC/isp18//isp18//3CholTEG/







IL17RA_111
mUmCmCmCmCmCG*C*C*A*G*T*mGmCm
117
 57



CmAmGmC/isp18//isp18//3CholTEG/







IL17RA_112
mCmCmCmUmCmUG*A*C*T*C*T*mGmAm
118
 57



CmCmCmC/isp18//isp18//3CholTEG/







IL17RA_113
mGmGmGmCmCmCC*T*C*T*G*A*mCmUm
119
 85



CmUmGmA/isp18//isp18//3CholTEG/







IL17RA_114
mUmGmGmGmCmCC*C*T*C*T*G*mAmCm
120
 76



UmCmUmG/isp18//isp18//3CholTEG/







IL17RA_115
mCmUmGmGmGmCC*C*C*T*C*T*mGmAm
121
 64



CmUmCmU/isp18//isp18//3CholTEG/







IL17RA_116
mAmGmGmGmUmCT*C*A*C*T*C*mUmGm
122
  8



CmUmGmC/isp18//isp18//3CholTEG/







IL17RA_117
mCmAmGmGmGmUC*T*C*A*C*T*mCmUm
123
 55



GmCmUmG/isp18//isp18//3CholTEG/







IL17RA_118
mAmCmAmGmGmGT*C*T*C*A*C*mUmCm
124
 59



UmGmCmU/isp18//isp18//3CholTEG/







IL17RA_119
mGmAmCmAmGmGG*T*C*T*C*A*mCmUm
125
 60



CmUmGmC/isp18//isp18//3CholTEG/







IL17RA_120
mAmGmAmCmAmGG*G*T*C*T*C*mAmCm
126
 70



UmCmUmG/isp18//isp18//3CholTEG/







IL17RA_121
mGmAmGmAmCmAG*G*G*T*C*T*mCmAm
127
 83



CmUmCmU/isp18//isp18//3CholTEG/







IL17RA_122
mUmGmAmGmAmCA*G*G*G*T*C*mUmCm
128
 78



AmCmUmC/isp18//isp18//3CholTEG/







IL17RA_123
mGmCmAmGmGmCT*T*C*C*A*C*mUmCm
129
 60



CmAmUmC/isp18//isp18//3CholTEG/







IL17RA_124
mCmUmCmCmAmCT*C*A*C*C*T*mCmCm
130
 62



CmAmGmC/isp18//isp18//3CholTEG/







IL17RA_125
mAmCmUmCmCmAC*T*C*A*C*C*mUmCm
131
 78



CmCmAmG/isp18//isp18//3CholTEG/







IL17RA_126
mGmCmUmCmAmCT*C*C*A*C*T*mCmAm
132
 91



CmCmUmC/isp18//isp18//3CholTEG/







IL17RA_127
mAmGmCmUmCmAC*T*C*C*A*C*mUmCm
133
 89



AmCmCmU/isp18//isp18//3CholTEG/







IL17RA_128
mGmAmCmAmAmGC*T*C*A*C*T*mCmCm
134
 86



AmCmUmC/isp18//isp18//3CholTEG/







IL17RA_129
mUmCmCmCmCmCC*A*C*C*C*C*mCmCm
135
 59



AmCmCmC/isp18//isp18//3CholTEG/







IL17RA_130
mAmGmGmGmCmUT*C*A*G*A*C*mUmCm
136
 58



AmCmCmU/isp18//isp18//3CholTEG/







IL17RA_131
mCmAmGmGmGmCT*T*C*A*G*A*mCmUm
137
 65



CmAmCmC/isp18//isp18//3CholTEG/







IL17RA_132
mGmCmAmGmGmGC*T*T*C*A*G*mAmCm
138
 51



UmCmAmC/isp18//isp18//3CholTEG/







IL17RA_133
mUmGmCmAmGmGG*C*T*T*C*A*mGmAm
139
 57



CmUmCmA/isp18//isp18//3CholTEG/







IL17RA_134
mGmUmGmCmAmGG*G*C*T*T*C*mAmGm
140
 84



AmCmUmC/isp18//isp18//3CholTEG/







IL17RA_135
mCmGmGmGmUmGC*A*G*G*G*C*mUmUm
141
 66



CmAmGmA/isp18//isp18//3CholTEG/







IL17RA_136
mCmGmCmGmGmGT*G*C*A*G*G*mGmCm
142
 43



UmUmCmA/isp18//isp18//3CholTEG/







IL17RA_137
mAmCmGmCmGmGG*T*G*C*A*G*mGmGm
143
 39



CmUmUmC/isp18//isp18//3CholTEG/







IL17RA_138
mAmAmCmGmCmGG*G*T*G*C*A*mGmGm
144
 51



GmCmUmU/isp18//isp18//3CholTEG/







IL17RA_139
mGmAmAmCmGmCG*G*G*T*G*C*mAmGm
145
 59



GmGmCmU/isp18//isp18//3CholTEG/







IL17RA_140
mCmUmCmUmCmCT*G*T*C*A*C*mAmUm
146
 55



UmUmCmC/isp18//isp18//3CholTEG/







IL17RA_141
mGmCmUmCmUmCC*T*G*T*C*A*mCmAm
147
 58



UmUmUmC/isp18//isp18//3CholTEG/







IL17RA_142
mUmGmCmUmCmUC*C*T*G*T*C*mAmCm
148
 59



AmUmUmU/isp18//isp18//3CholTEG/







IL17RA_143
mUmUmGmCmUmCT*C*C*T*G*T*mCmAm
149
 53



CmAmUmU/isp18//isp18//3CholTEG/







IL17RA_144
mUmUmUmGmCmUC*T*C*C*T*G*mUmCm
150
 51



AmCmAmU/isp18//isp18//3CholTEG/







IL17RA_145
mGmUmUmUmGmCT*C*T*C*C*T*mGmUm
151
 56



CmAmCmA/isp18//isp18//3CholTEG/







IL17RA_146
mUmGmUmUmUmGC*T*C*T*C*C*mUmGm
152
 53



UmCmAmC/isp18//isp18//3CholTEG/







IL17RA_147
mCmCmUmGmUmUT*G*C*T*C*T*mCmCm
153
 71



UmGmUmC/isp18//isp18//3CholTEG/







IL17RA_148
mAmGmCmUmGmAA*G*A*G*G*T*mGmGm
154
 64



GmAmGmG/isp18//isp18//3CholTEG/







IL17RA_149
mGmAmGmCmUmGA*A*G*A*G*G*mUmG
155
 73



mGmGmAmG/isp18//isp18//3CholTEG/







IL17RA_150
mCmGmAmGmCmUG*A*A*G*A*G*mGmU
156
 64



mGmGmGmA/isp18//isp18//3CholTEG/







IL17RA_151
mAmCmGmAmGmCT*G*A*A*G*A*mGmGm
157
 86



UmGmGmG/isp18//isp18//3CholTEG/







IL17RA_152
mGmGmCmAmGmGC*T*T*C*C*A*mCmUm
158
 66



CmCmAmU/isp18//isp18//3CholTEG/







IL17RA_153
mUmGmGmGmCmAG*G*C*T*T*C*mCmAm
159
 63



CmUmCmC/isp18//isp18//3CholTEG/







IL17RA_154
mCmUmGmGmGmCA*G*G*C*T*T*mCmCm
160
 42



AmCmUmC/isp18//isp18//3CholTEG/







IL17RA_155
mCmCmUmGmGmGC*A*G*G*C*T*mUmCm
161
 45



CmAmCmU/isp18//isp18//3CholTEG/







IL17RA_156
mCmCmAmCmCmUC*T*G*C*A*C*mAmCm
162
 77



UmCmAmG/isp18//isp18//3CholTEG/







IL17RA_157
mGmCmCmUmGmGG*A*G*G*T*C*mGmAm
163
 64



GmGmCmU/isp18//isp18//3CholTEG/







IL17RA_158
mAmGmCmCmUmGG*G*A*G*G*T*mCmGm
164
 71



AmGmGmC/isp18//isp18//3CholTEG/







IL17RA_159
mUmGmAmGmCmCT*G*G*G*A*G*mGmUm
165
 80



CmGmAmG/isp18//isp18//3CholTEG/







IL17RA_160
mUmUmGmAmGmCC*T*G*G*G*A*mGmGm
166
 95



UmCmGmA/isp18//isp18//3CholTEG/







IL17RA_161
mCmUmUmGmAmGC*C*T*G*G*G*mAmGm
167
 92



GmUmCmG/isp18//isp18//3CholTEG/







IL17RA_162
mUmUmGmGmGmAG*G*A*T*G*A*mGmG
168
 78



mCmGmGmG/isp18//isp18//3CholTEG/







IL17RA_163
mUmUmUmGmGmGA*G*G*A*T*G*mAmG
169
 74



mGmCmGmG/isp18//isp18//3CholTEG/







IL17RA_164
mGmUmGmGmGmAT*G*C*A*G*G*mCmCm
170
 92



CmGmGmC/isp18//isp18//3CholTEG/







IL17RA_165
mUmGmUmGmGmGA*T*G*C*A*G*mGmCm
171
 85



CmCmGmG/isp18//isp18//3CholTEG/







IL17RA_166
mUmUmGmUmGmGG*A*T*G*C*A*mGmGm
172
 89



CmCmCmG/isp18//isp18//3CholTEG/







IL17RA_167
mAmCmUmCmCmUG*C*C*C*C*A*mCmCm
173
 75



CmAmCmU/isp18//isp18//3CholTEG/







IL17RA_168
mAmGmGmCmUmGG*T*G*C*C*A*mCmUm
174
 82



CmGmGmG/isp18//isp18//3CholTEG/







IL17RA_169
mGmAmGmGmAmUC*A*G*T*G*G*mUmAm
175
 84



CmCmUmC/isp18//isp18//3CholTEG/







IL17RA_170
mAmAmGmAmGmGA*T*C*A*G*T*mGmGm
176
 80



UmAmCmC/isp18//isp18//3CholTEG/







IL17RA_171
mGmCmAmUmGmGA*G*A*G*C*C*mAmUm
177
 80



GmCmAmG/isp18//isp18//3CholTEG/







IL17RA_172
mGmGmGmAmAmAG*A*G*G*A*T*mCmAm
178
 82



GmUmGmG/isp18//isp18//3CholTEG/







IL17RA_173
mAmGmGmGmAmAA*G*A*G*G*A*mUmC
179
 88



mAmGmUmG/isp18//isp18//3CholTEG/







IL17RA_174
mCmAmUmGmGmAG*A*G*C*C*A*mUmGm
180
 84



CmAmGmA/isp18//isp18//3CholTEG/







IL17RA_175
mGmGmCmAmUmGG*A*G*A*G*C*mCmAm
181
 88



UmGmCmA/isp18//isp18//3CholTEG/







IL17RA_176
mGmGmGmGmCmAT*G*G*A*G*A*mGmCm
182
 86



CmAmUmG/isp18//isp18//3CholTEG/







IL17RA_177
mUmGmGmGmGmCA*T*G*G*A*G*mAmGm
183
 56



CmCmAmU/isp18//isp18//3CholTEG/







IL17RA_178
mAmUmGmGmGmGC*A*T*G*G*A*mGmAm
184
 84



GmCmCmA/isp18//isp18//3CholTEG/







IL17RA_179
mAmUmCmUmGmGG*A*G*A*G*G*mCmG
185
 80



mAmUmGmG/isp18//isp18//3CholTEG/







IL17RA_180
mGmAmUmCmUmGG*G*A*G*A*G*mGmC
186
 80



mGmAmUmG/isp18//isp18//3CholTEG/







IL17RA_181
mGmGmAmUmCmUG*G*G*A*G*A*mGmG
187
 85



mCmGmAmU/isp18//isp18//3CholTEG/







IL17RA_182
mCmCmCmUmCmUC*C*T*C*C*T*mCmCm
188
 78



UmAmCmC/isp18//isp18//3CholTEG/







IL17RA_183
mUmCmCmCmUmCT*C*C*T*C*C*mUmCm
189
 74



CmUmAmC/isp18//isp18//3CholTEG/







IL17RA_184
mCmUmCmCmCmUC*T*C*C*T*C*mCmUm
190
 77



CmCmUmA/isp18//isp18//3CholTEG/







IL17RA_185
mCmUmAmCmUmCC*C*T*C*T*C*mCmUm
191
 93



CmCmUmC/isp18//isp18//3CholTEG/







IL17RA_186
mUmCmUmAmCmUC*C*C*T*C*T*mCmCm
192
 96



UmCmCmU/isp18//isp18//3CholTEG/







IL17RA_187
mUmUmCmUmAmCT*C*C*C*T*C*mUmCm
193
 91



CmUmCmC/isp18//isp18//3CholTEG/







IL17RA_188
mUmUmUmCmUmAC*T*C*C*C*T*mCmUm
194
 80



CmCmUmC/isp18//isp18//3CholTEG/







IL17RA_189
mCmUmUmUmCmUA*C*T*C*C*C*mUmCm
195
 89



UmCmCmU/isp18//isp18//3CholTEG/







IL17RA_190
mCmCmUmUmUmCT*A*C*T*C*C*mCmUm
196
 86



CmUmCmC/isp18//isp18//3CholTEG/







IL17RA_191
mCmUmCmCmUmUT*C*T*A*C*T*mCmCm
197
 83



CmUmCmU/isp18//isp18//3CholTEG/







IL17RA_192
mUmCmCmCmUmCC*T*T*T*C*T*mAmCm
198
 82



UmCmCmC/isp18//isp18//3CholTEG/







IL17RA_193
mUmCmCmCmUmCG*T*C*A*C*A*mGmCm
199
 74



CmAmCmC/isp18//isp18//3CholTEG/







IL17RA_194
mGmCmUmCmUmCT*C*T*G*C*C*mUmCm
200
 79



UmCmGmU/isp18//isp18//3CholTEG/







IL17RA_195
mGmAmGmCmUmCT*C*T*C*T*G*mCmCm
201
 80



UmCmUmC/isp18//isp18//3CholTEG/







IL17RA_196
mGmGmGmCmUmUC*A*G*A*C*T*mCmAm
202
 40



CmCmUmU/isp18//isp18//3CholTEG/







IL17RA_197
mCmUmCmCmGmCA*G*G*T*A*G*mUmUm
203
 60



GmUmCmC/isp18//isp18//3CholTEG/







IL17RA_198
mGmGmCmUmCmCG*C*A*G*G*T*mAmGm
204
 20



UmUmGmU/isp18//isp18//3CholTEG/







IL17RA_199
mGmGmGmCmUmCC*G*C*A*G*G*mUmAm
205
 34



GmUmUmG/isp18//isp18//3CholTEG/







IL17RA_200
mAmCmAmCmCmCA*C*A*G*G*G*mGmCm
206
 10



AmUmGmU/isp18//isp18//3CholTEG/







IL17RA_201
mUmCmCmCmCmGA*C*C*A*G*C*mGmGm
207
 76



GmUmCmU/isp18//isp18//3CholTEG/







IL17RA_202
mCmUmCmCmCmCG*A*C*C*A*G*mCmGm
208
 47



GmGmUmC/isp18//isp18//3CholTEG/







IL17RA_203
mCmGmUmAmGmGG*C*G*T*G*T*mGmUm
209
 31



GmGmGmU/isp18//isp18//3CholTEG/







IL17RA_204
mUmCmGmUmAmGG*G*C*G*T*G*mUmGm
210
 70



UmGmGmG/isp18//isp18//3CholTEG/







IL17RA_205
mCmUmCmGmUmAG*G*G*C*G*T*mGmUm
211
 47



GmUmGmG/isp18//isp18//3CholTEG/







IL17RA_206
mCmCmUmCmGmUA*G*G*G*C*G*mUmGm
212
 58



UmGmUmG/isp18//isp18//3CholTEG/







IL17RA_207
mUmCmCmUmCmGT*A*G*G*G*C*mGmUm
213
 36



GmUmGmU/isp18//isp18//3CholTEG/







IL17RAv208
mCmUmCmCmUmCG*T*A*G*G*G*mCmGm
214
 32



UmGmUmG/isp18//isp18//3CholTEG/







IL17RA_209
mGmGmGmCmGmUC*A*A*A*C*A*mGmUm
215
 33



UmAmUmU/isp18//isp18//3CholTEG/







IL17RA_210
mUmCmCmAmAmUA*A*A*G*C*T*mGmUm
216
 84



UmAmGmG/isp18//isp18//3CholTEG/







IL17RA_211
mCmUmCmCmAmAT*A*A*A*G*C*mUmGm
217
 71



UmUmAmG/isp18//isp18//3CholTEG/







IL17RA_212
mAmCmUmCmCmAA*T*A*A*A*G*mCmUm
218
 87



GmUmUmA/isp18//isp18//3CholTEG/







IL17RA_213
mUmAmCmUmCmCA*A*T*A*A*A*mGmCm
219
 78



UmGmUmU/isp18//isp18//3CholTEG/







IL17RA_214
mUmGmCmCmCmUA*T*T*T*A*A*mUmUm
220
 84



UmUmCmA/isp18//isp18//3CholTEG/







IL17RA_215
mAmUmGmCmCmCT*A*T*T*T*A*mAmUm
221
 87



UmUmUmC/isp18//isp18//3CholTEG/







IL17RA_216
mUmAmUmGmCmCC*T*A*T*T*T*mAmAm
222
 81



UmUmUmU/isp18//isp18//3CholTEG/







IL17RA_217
mCmUmCmAmUmUT*A*T*T*T*A*mGmGm
223
 87



AmAmGmG/isp18//isp18//3CholTEG/







IL17RA_218
mCmCmUmCmAmUT*T*A*T*T*T*mAmGm
224
 85



GmAmAmG/isp18//isp18//3CholTEG/







IL17RA_219
mGmCmUmUmGmGG*C*A*G*G*T*mGmGm
225
 36



UmGmAmA/isp18//isp18//3CholTEG/







IL17RA_220
mUmCmCmAmCmUC*A*C*C*T*C*mCmCm
226
 46



AmGmCmA/isp18//isp18//3CholTEG/







IL17RA_221
mUmCmAmCmUmCC*A*C*T*C*A*mCmCm
227
 63



UmCmCmC/isp18//isp18//3CholTEG/







IL17RA_222
mUmCmCmCmCmAC*C*C*C*T*G*mAmGm
228
 73



CmUmCmU/isp18//isp18//3CholTEG/







IL17RA_223
mAmGmAmGmGmAT*C*A*G*T*G*mGmUm
229
 76



AmCmCmU/isp18//isp18//3CholTEG/







IL17RA_224
mGmCmUmCmCmGC*A*G*G*T*A*mGmUm
230
 56



UmGmUmC/isp18//isp18//3CholTEG/







IL17RA_225
mCmUmCmAmCmUC*C*A*C*T*C*mAmCm
231
 68



CmUmCmC/isp18//isp18//3CholTEG/







IL17RA_226
mAmUmGmGmGmCT*T*G*G*G*C*mAmGm
232
 54



GmUmGmG/isp18//isp18//3CholTEG/







IL17RA_227
mAmUmGmGmGmUA*G*A*T*T*C*mGmUm
233
 57



UmCmCmA/isp18//isp18//3CholTEG/







IL17RA_228
mCmAmCmUmCmUT*G*A*A*G*C*mUmCm
234
 79



UmUmGmG/isp18//isp18//3CholTEG/







IL17RA_229
mCmGmUmCmAmAA*C*A*G*T*T*mAmUm
235
 81



UmUmAmU/isp18//isp18//3CholTEG/







IL17RA_230
mGmGmCmUmGmGA*T*T*T*C*T*mUmUm
236
 38



UmGmGmG/iSp18//iSp18//3CholTEG/







IL17RA_231
mCmUmCmCmUmCG*T*A*G*G*G*mCmGm
237
 32



UmGmUmG/isp18//isp18//3CholTEG/







IL17RA_232
mGmGmCmAmGmGT*G*G*T*G*A*mAmCm
238
 57



GmGmUmC/isp18//isp18//3CholTEG/







IL17RA_233
mGmGmGmCmAmGG*T*G*G*T*G*mAmAm
239
 69



CmGmGmU/isp18//isp18//3CholTEG/







IL17RA_234
mUmGmGmGmCmAG*G*T*G*G*T*mGmAm
240
 69



AmCmGmG/isp18//isp18//3CholTEG/







IL17RA_235
mUmUmGmGmGmCA*G*G*T*G*G*mUmGm
241
 62



AmAmCmG/isp18//isp18//3CholTEG/







IL17RA_236
mCmUmUmGmGmGC*A*G*G*T*G*mGmUm
242
 49



GmAmAmC/isp18//isp18//3CholTEG/







IL17RA_237
mCmAmCmAmGmGG*G*C*A*T*G*mUmAm
243
  5



GmUmCmC/isp18//isp18//3CholTEG/







IL17RA_238
mCmCmAmCmAmGG*G*G*C*A*T*mGmUm
244
  6



AmGmUmC/isp18//isp18//3CholTEG/







IL17RA_239
mCmCmCmAmCmAG*G*G*G*C*A*mUmGm
245
  5



UmAmGmU/isp18//isp18//3CholTEG/







IL17RA_240
mCmAmCmCmCmAC*A*G*G*G*G*mCmAm
246
 12



UmGmUmA/isp18//isp18//3CholTEG/







IL17RA_241
mUmAmCmAmCmCC*A*C*A*G*G*mGmGm
247
 16



CmAmUmG/isp18//isp18//3CholTEG/







IL17RA_242
mGmUmAmCmAmCC*C*A*C*A*G*mGmGm
248
   7



GmCmAmU/isp18//isp18//3CholTEG/







IL17RA_243
mAmGmUmAmCmAC*C*C*A*C*A*mGmGm
249
  4



GmGmCmA/isp18//isp18//3CholTEG/







IL17RA_244
mCmAmGmUmAmCA*C*C*C*A*C*mAmGm
250
  4



GmGmGmC/isp18//isp18//3CholTEG/







IL17RA_245
mCmCmAmGmUmAC*A*C*C*C*A*mCmAm
251
  4



GmGmGmG/isp18//isp18//3CholTEG/







IL17RA_246
mGmUmAmGmGmGC*G*T*G*T*G*mUmGm
252
 26



GmGmUmC/isp18//isp18//3CholTEG/







IL17RA_247
mCmCmUmCmCmUC*G*T*A*G*G*mGmCm
253
 15



GmUmGmU/isp18//isp18//3CholTEG/







IL17RA_248
mUmCmCmUmCmCT*C*G*T*A*G*mGmGm
254
 16



CmGmUmG/isp18//isp18//3CholTEG/







IL17RA_249
mCmUmCmCmUmCC*T*C*G*T*A*mGmGm
255
 18



GmCmGmU/isp18//isp18//3CholTEG/







IL17RA_250
mGmCmUmCmCmUC*C*T*C*G*T*mAmGm
256
 49



GmGmCmG/isp18//isp18//3CholTEG/







IL17RA_251
mGmCmUT*G*G*G*C*A*G*G*T*G*G*T*m
257
 49



GmAmA/isp18//isp18//3CholTEG/







IL17RA_252
mUmCmCT*C*G*T*A*G*G*G*C*G*T*G*m
258
 48



UmGmU/isp18//isp18//3CholTEG/







IL17RA_253
mGmCmUmUG*G*G*C*A*G*G*T*G*G*mU
259
 49



mGmAmA/isp18//isp18//3CholTEG/







IL17RA_254
mAmCmAmCC*C*A*C*A*G*G*G*G*C*mA
260
 82



mUmGmU/isp18//isp18//3CholTEG/







IL17RA_255
mCmGmUmAG*G*G*C*G*T*G*T*G*T*mG
261
 51



mGmGmU/isp18//isp18//3CholTEG/







IL17RA_256
mUmCmCmUC*G*T*A*G*G*G*C*G*T*mG
262
 51



mUmGmU/isp18//isp18//3CholTEG/







IL17RA_257
mGmCmUmUmGG*G*C*A*G*G*T*G*mGm
263
 17



UmGmAmA/isp18//isp18//3CholTEG/







IL17RA_258
mAmCmAmCmCC*A*C*A*G*G*G*G*mCmA
264
 82



mUmGmU/isp18//isp18//3CholTEG/







IL17RA_259
mCmGmUmAmGG*G*C*G*T*G*T*G*mUmG
265
 48



mGmGmU/isp18//isp18//3CholTEG/







IL17RA_260
mUmCmCmUmCG*T*A*G*G*G*C*G*mUmG
266
 48



mUmGmU/isp18//isp18//3CholTEG/







IL17RA_261
mGmCmUmUmGmGmGmCmAG*G*T*G*G*
267
 14



T*mGmAmA/isp18//isp18//3CholTEG/







IL17RA_262
mAmCmAmCmCmCmAmCmAG*G*G*G*C*
268
 11



A*mUmGmU/isp18//isp18//3CholTEG/







IL17RA_263
mUmCmCmUmCmGmUmAmGG*G*C*G*T*
269
 18



G*mUmGmU/isp18//isp18//3CholTEG/







IL17RA_264
mGmCmUT*G*G*G*C*A*mGmGmUmGmGm
270
 16



UmGmAmA/isp18//isp18//3CholTEG/







IL17RA_265
mAmCmAC*C*C*A*C*A*mGmGmGmGmCm
271
 14



AmUmGmU/isp18//isp18//3CholTEG/







IL17RA_266
mCmGmUA*G*G*G*C*G*mUmGmUmGmU
272
 31



mGmGmGmU/isp18//isp18//3CholTEG/







IL17RA_267
mUmCmCT*C*G*T*A*G*mGmGmCmGmUm
273
 31



GmUmGmU/isp18//isp18//3CholTEG/







IL17RA_268
mGmCmUmUmGmGmGC*A*G*G*T*G*mGm
274
  9



UmGmAmA/isp18//isp18//3CholTEG/







IL17RA_269
mAmCmAmCmCmCmAC*A*G*G*G*G*mCm
275
 12



AmUmGmU/isp18//isp18//3CholTEG/







IL17RA_270
mCmGmUmAmGmGmGC*G*T*G*T*G*mUm
276
 33



GmGmGmU/isp18//isp18//3CholTEG/







IL17RA_271
mUmCmCmUmCmGmUA*G*G*G*C*G*mUm
277
 33



GmUmGmU/isp18//isp18//3CholTEG/







IL17RA_272
mGmCmUmUmGG*G*C*A*G*G*mUmGmG
278
 13



mUmGmAmA/isp18//isp18//3CholTEG/







IL17RA_273
mAmCmAmCmCC*A*C*A*G*G*mGmGmCm
279
 34



AmUmGmU/isp18//isp18//3CholTEG/







IL17RA_274
mCmGmUmAmGG*G*C*G*T*G*mUmGmUm
280
 46



GmGmGmU/isp18//isp18//3CholTEG/







IL17RA_275
mUmCmCmUmCG*T*A*G*G*G*mCmGmUm
281
 47



GmUmGmU/isp18//isp18//3CholTEG/







IL17RA_276
mAmCmAC*C*C*A*C*A*G*G*G*G*C*A*m
282
 82



UmGmU/isp18//isp18//3CholTEG/







IL17RA_277
mCmGmUA*G*G*G*C*G*T*G*T*G*T*G*m
283
 48



GmGmU/isp18//isp18//3CholTEG/







IL17RA_278
mAmCmCmCmAmCA*G*G*G*G*C*mAmUm
284
  7



GmUmAmG/isp18//isp18//3CholTEG/







IL17RA_279
mCmGmUmAmGmGmGmCmGT*G*T*G*T*G
285
 18



*mGmGmU/isp18//isp18//3CholTEG/







IL17RA_280
mCmCmCmAmCmAmGmGmGG*C*A*T*G*T
286
  7



*mAmGmU/isp18//isp18//3CholTEG/







IL17RA_281
mCmCmCmAmCA*G*G*G*G*C*mAmUmGm
287
 10



UmAmGmU/isp18//isp18//3CholTEG/







IL17RA_282
mCmCmCmAmCmAmGG*G*G*C*A*T*mGm
288
  3



UmAmGmU/isp18//isp18//3CholTEG/







IL17RA_283
mGmUmAmGmGmGmCG*T*G*T*G*T*mGm
289
 33



GmGmUmC/isp18//isp18//3CholTEG/







IL17RA_284
mGmUmAG*G*G*C*G*T*mGmUmGmUmGm
290
 31



GmGmUmC/isp18//isp18//3CholTEG/







IL17RA_285
mGmUmAmGmGmGmCmGmUG*T*G*T*G*
291
 18



G*mGmUmC/isp18//isp18//3CholTEG/







IL17RA_286
mGmUmAmGmGG*C*G*T*G*T*mGmUmGm
292
 46



GmGmUmC/isp18//isp18//3CholTEG/







IL17RA_287
mCmCmCA*C*A*G*G*G*mGmCmAmUmGm
293
 12



UmAmGmU/isp18//isp18//3CholTEG/







IL17RA_288
mGmUmAG*G*G*C*G*T*G*T*G*T*G*G*m
294
 48



GmUmC/isp18//isp18//3CholTEG/







IL17RA_289
mGmUmAmGG*G*C*G*T*G*T*G*T*G*mG
295
 51



mGmUmC/isp18//isp18//3CholTEG/







IL17RA_290
mCmCmCA*C*A*G*G*G*G*C*A*T*G*T*m
296
 57



AmGmU/isp18//isp18//3CholTEG/







IL17RA_291
mCmCmCmAC*A*G*G*G*G*C*A*T*G*mU
297
 35



mAmGmU/isp18//isp18//3CholTEG/







IL17RA_292
mCmCmCmAmCA*G*G*G*G*C*A*T*mGmU
298
 10



mAmGmU/isp18//isp18//3CholTEG/







IL17RA_293
mGmUmAmGmGG*C*G*T*G*T*G*T*mGmG
299
 55



mGmUmC/isp18//isp18//3CholTEG/







Control
mGmUmUmUmCmAC*C*A*C*C*C*mAmAm
300




UmUmCmC/iSp18//iSp18//3CholTEG/
















TABLE 2







Oligonucleotide Sequences


Special bases used in the oligonucleotides are


as follows: mN = 2' O-methyl RNA, /iSp18/ =


hexa(ethylene glycol) spacer phosphoramidite,


* = phosphorothioate, /3CholTEG/ = 3′


cholesterol









Com-

SEQ


pound

ID


ID
Oligonucleotide Sequence (5′ to 3′)
NO:





Control
mGmUmUmUmCmAC*C*A*C*C*C*mAmAmUmUmCmC/
300



iSp18//iSp18//3CholTEG/






Anti-
mUmGmGmGmAmGT*A*G*A*C*A*mAmGmGmUmAmC/
301


TNF
iSp18//iSp18//3CholTEG/






IL17RA_
mGmCmUmUmGmGG*C*A*G*G*T*mGmGmUmGmAmA/
225


219
iSp18//iSp18//3CholTEG/









Example 2. IL-17RA mRNA Knockdown in Human Skin Using SNAs Formulated in a Gel Vehicle

Methods and Materials


Tissue Culture. Healthy human ex vivo skin, freshly acquired from a single donor was received the day of experiment initiation and prepared immediately. Prior to use, the skin was inspected, noting any areas of extensive stretch marks or scarring, and any sections with visible holes or damage were removed. A dermatome was used to excise full thickness skin from the donor sample. The resulting tissue contained an intact stratum corneum, epidermis, and dermis. No subcutaneous tissue from the surgical explant remained. The skin was carefully cut to the appropriate size and care was taken not to stretch the skin when it was placed into the Franz cells. The skin was then clamped into the Franz cell. Next, a stir bar and receiving medium that had been pre-warmed to 37° C. were added to each Franz cell. The receiving medium consisted of Dulbecco's modified Eagle's medium, supplemented with 2% fetal bovine serum, 50 U/mL penicillin, 50 mg/mL streptomycin, 0.25 μg/mL amphotericin B and 10 μg/mL gentamycin. The Franz cells, explants and receiving medium were allowed to equilibrate for a minimum of 30 minutes, after which the temperature of the media in each cell was verified to be 37±1° C. Any air bubbles that were introduced during assembly of the cells were removed. The integrity of the skin was confirmed prior to initiating the study by inverting the cell. If leaks were identified, the skin and donor chamber were repositioned until leaks were absent. Over the course of the experiment, the skin was maintained at ambient humidity and at an epidermal surface temperature of 32° C., which was regulated using 37° C. water-jacketing of the receptor chamber containing cell culture media that contacts the dermal portion of the explant.


SNA Synthesis. The oligonucleotide was synthesized at the 1 mmole scale employing standard UniLinker support (ChemGenes). The DNA, 2′-O-Me RNA monomers and hexa(ethylene glycol) spacers were obtained from ChemGenes Corporation. The cholesterol modifier was obtained from Glen Research. Linkages were either standard phosphodiesters or phosphorothioates prepared with a solution of 0.2 M phenylacetyl disulfide (PADS) in a mixture of 1:1 lutidine:ACN. Synthesis was performed DMT-off, in the 3′ to 5′ direction. After synthesis, the oligonucleotide was cleaved from the support and de-protected using a 4:1 mixture of ammonium hydroxide and ethanol at 55° C. for 16 hours. The oligonucleotide was purified via high performance liquid chromatography (HPLC) techniques. Molecular weights and extinction coefficients were calculated using the IDT OligoAnalyzer. The verification of the oligonucleotide product molecular weight was performed using electrospray ionization mass spectrometry (ESI-MS). Finally, the oligonucleotide concentration was determined by UV-absorbance at 260 nm on a microplate reader (BioTek).


Liposomes were formulated by first dissolving 250 mg 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) in chloroform to a final concentration of 50 mg/mL. The solvent was then removed under nitrogen to form a thin lipid film. The film was lyophilized overnight to ensure all solvent was removed. The lipid film was subsequently hydrated with 10 mM phosphate buffered saline (PBS) and sonication/freeze-fracture were used to form large, unilamellar vesicles. These vesicles were then continuously homogenized through a micro-fluidizer at up to 25 kpsi, until the desired mean number diameter of 20 nm was achieved. Liposome concentrations were determined using a choline quantification assay and the particle size and dispersion were measured by dynamic light scattering (DLS).


SNAs targeting human IL-17RA, compound IL17RA_282, were formulated by mixing a 30-fold molar excess of cholesterol-modified oligonucleotide, SEQ ID NO: 288, to the liposome suspension in PBS followed by overnight incubation at 4° C.


Drug Application to Skin. A gel vehicle was used to apply IL17RA_282 to the skin. The pre-formulated SNAs were mixed into the gel vehicle, resulting in three drug product strengths of 0, 0.00007 and 0.007% w/w (oligonucleotide weight). The composition of these drug products can be found in Table 3.


These drug products were then applied topically to the skin explants. The drug products were briefly mixed prior to dosing using a pipette tip. 40 μL of each drug product was dosed topically to each Franz cell using a positive displacement pipette. The pipette tip was used to spread the drug product evenly covering the entire exposed surface area of the skin (1.0 cm2). After dosing, the sampling port was occluded to prevent solvent evaporation during the study. The actual dosing time was recorded and the explants were maintained at ambient humidity and at an epidermal surface temperature of 32° C. for 24 hours.


RNA Extraction and qRT-PCR. At the conclusion of the 24 hour incubation period, the Franz cells were disassembled and the skin was carefully removed from each cell. A cotton swab wetted with PBS was used to remove the drug product and then the application area was blotted dry. Three samples were cut from the 1.0 cm2 dosing area using a 4 mm biopsy punch. The skin was not stretched while the biopsy punches were taken. The biopsy punches were then lysed in RLT buffer (Qiagen) with a bead homogenizer. RNA was isolated from lysates using the RNEasy 96-well kit (Qiagen) according to the manufacturer's instructions. cDNA was then synthesized from RNA isolates using the cDNA high capacity reverse transcription kit (Life Technologies). cDNA was prepared on a thermocycler with the following temperature program: 25° C. for 10 minutes, 37° C. for 90 minutes, 85° C. for 5 minutes followed by a 4° C. hold. The resulting cDNA was diluted 8 fold with nuclease-free water. qPCR was performed using 6 μL of the diluted cDNA, 4.66 μL LightCycler480 Probes Master Mix (Roche), 0.47 μL human IL-17RA specific FAM-labeled probe and primers, and 0.37 μL human Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) specific HEX-labeled probe and primers per reaction well of a 384-well optical reaction plate (Roche). The primer and probe set for IL-17RA was purchased commercially (Hs01064648 ml, Thermo Fisher Scientific). The primer and probe set for GAPDH was designed using the known human genome sequence (NCBI reference sequences NM_002046.5) and was found to be specific by “blastn” analysis (NCBI). The oligonucleotide sequences used for GAPDH were: forward 5′-CAA GGT CAT CCA TGA CAA CTT TG-3′ (SEQ ID NO: 4), reverse 5′-GGG CCA TCC ACA GTC TTC T-3′ (SEQ ID NO: 5), probe 5′-HEX-ACC ACA GTC CAT GCC ATC ACT GCC A-BHQ1-3′ (SEQ ID NO: 6). qPCR reactions, in technical duplicate, were carried out on the Roche Lightcycler 480 under the following conditions: initial denaturation at 95° C. for 10 minutes and then 50 cycles of denaturation at 95° C. for 10 seconds, annealing at 60° C. for 30 seconds and extension at 72° C. for 1 second. Cp values were obtained by analysis with the 2nd derivative method. Relative gene expression was determined by normalization with the housekeeping gene (GAPDH) and the ΔΔ-Ct method. Statistical analysis was done by performing a one-way ANOVA.


Protein Isolation and Western Blot. Total protein was isolated from the biopsy lysates with acetone precipitation. Lysates from each treatment group were pooled before isolating the protein to increase protein yield. Briefly, four volumes of ice cold acetone were added to the lysate and incubated at −20° C. for 30 minutes. The samples were centrifuged and decanted, and the pellets were washed with one volume ice cold ethanol. Again, the samples were centrifuged and the pellets were allowed to air dry. The pellets were re-suspended in one volume Laemmli sample buffer containing 1% β-mercaptoethanol. To ensure the samples were completely dissolved, they were boiled at 95° C. for 10 minutes and subsequently centrifuged to remove any undissolved debris.


The protein isolates were loaded into a 10% SDS-PAGE gel, run and transferred overnight (22 V) to an Immobilon PSQ PVDF membrane (ISEQ00010, Millipore). The membrane was blocked for 1 hour with 5% dry milk in tris buffered saline containing Tween 20 (the resulting buffer is referred to as TBST) followed by a 5 minute wash in TBST. Human-specific antibodies targeting IL-17RA or β-actin protein were used to probe for the presence of those specific proteins. IL-17RA protein was detected using an anti-IL-17RA rabbit monoclonal primary antibody, and was shown to detect a protein with a predicted molecular weight of 150 kDa (12661, Cell Signaling). This antibody was diluted in blocking buffer (5% dry milk in TBST) at a ratio of 1:500 prior to use. β-actin protein was detected using an anti-β-actin rabbit monoclonal primary antibody, and was shown to detect a protein with a predicted molecular weight of 45 kDa (4970, Cell Signaling). This antibody was diluted in blocking buffer at a ratio of 1:1000 prior to use. Primary antibody incubation occurred overnight at 4° C.


After the incubation period, the membrane was rinsed twice with TBST, followed by two consecutive washes with TBST of 15 minutes and 5 minutes. Next, the membrane was incubated with an anti-rabbit HRP secondary antibody (7074, Cell Signaling) diluted in blocking buffer at a ratio of 1:1000 prior to use. The secondary antibody incubation occurred for 90 minutes at room temperature. Then, the membrane was rinsed twice with TBST, followed by two consecutive washes with TBST for 15 minutes and 5 minutes. Then, the membrane was further washed twice with distilled water for 5 minutes. Subsequently, GE Amersham Prime ECL reagent (RPN2232) was added to the membrane and incubated at room temperature for 5 minutes. Then, the membrane was imaged using an exposure interval of 20 seconds for up to 2 minutes on a BioRad GelDoc Imager. Finally, images were analyzed by densitometry and IL-17RA signal was normalized to β-actin signal to compared expression levels across treatments.









TABLE 3







Drug Product Compositions











Gel Strength Formulations (w/w)












Ingredient
0%
0.00007%
0.007%
















SEQ ID NO: 288
0
0.00007
0.007



DOPC
0
0.0003801
0.03801



Diethylene glycol
25
25
25



monoethyl ether






(Transcutol P)






Glycerin
5
5
5



Hydroxyethyl Cellulose
1
1
1



Methylparaben
0.15
0.15
0.15



Propylparaben
0.05
0.05
0.05



Disodium EDTA
0.1
0.1
0.1



Sodium Metabisulfite
0.2
0.2
0.2



Water
68.5
68.5
68.45











Results


The IL-17RA mRNA targeted SNA (compound IL17RA_282) formulated as a drug product in a gel vehicle showed dose-dependent knockdown of IL-17RA mRNA in healthy human skin explant after only 24 hours of treatment (FIG. 9). Knockdown was observed when compared to both untreated skin and the 0% strength drug product. Treatment with the 0.007% strength drug product resulted in statistically significant IL-17RA mRNA inhibition compared to untreated (p=0.0011) and 0% strength drug product (p<0.0001) by one-way ANOVA. The IL-17RA mRNA targeted SNA (compound IL17RA_282) formulated as a drug product in a gel vehicle showed inhibition of IL-17RA protein expression in healthy human skin explant after only 24 hours of treatment (FIG. 10). Knockdown was observed when compared to both untreated skin and the 0% strength drug product. The graph represents the analyzed densitometry data collected from the western blot (inset). There was insufficient material to perform a protein-level analysis for the skin samples treated with 0.0007% gel.


Discussion and Conclusions


Clinical data suggests that the IL-17 signaling pathway plays a significant role in sustaining the psoriasis disease state. While current monoclonal antibody therapies, like brodalumab, provide very effective treatment options, they also present with troubling side-effect profiles. This presents a therapeutic need for a safer alternative. Antisense technology can reduce the expression of mRNA in cells; however, antisense oligonucleotides generally have poor uptake into cells and skin. In contrast, it has been found that when oligonucleotides are arranged in the SNA geometry, they exhibit skin penetration properties and increased cellular uptake. Given these advantages and the therapeutic needs for psoriasis treatment, an antisense SNA based therapy targeting human IL-17RA in the skin may prove an effective treatment for psoriasis.


It has been shown here that when the IL-17RA mRNA targeted SNA (compound IL17RA_282) formulated as a drug product in a gel vehicle is applied topically to healthy human skin explants, there is a pharmacodynamic effect seen at both the level of IL-17RA mRNA and protein. Increasing strength of the drug product results in decreased IL-17RA expression.


The response observed in this study represents only a 24 hour treatment. It is expected that longer treatment with multiple doses would further enhance the inhibition observed. Given the importance of IL-17 signaling in maintaining the diseased state observed in psoriasis, a strong topical drug product candidate for the treatment of psoriasis is presented.


Example 3. IL-17RA mRNA Knockdown in Human Psoriatic Skin Using SNAs Formulated in a Gel Vehicle

Methods and Materials


Tissue Culture. Four skin biopsies (3 mm diameter) were obtained from each of 8 different patients with mild to moderate plaque psoriasis. Biopsies were taken directly from psoriatic plaques. Due to the limited number of biopsies that can be taken from a single patient, only 4 biopsies were taken per individual. Biopsies were taken from volunteers at about 45 minute intervals and transported to the laboratory (rolled in moist sterile gauze). The time between taking biopsies and starting the incubation was between 5 and 7.5 hours. The biopsies were positioned in the membrane of an insert-well, and incubated in 12-well plates at 32° C. with ambient CO2 and humidity. The membranes were immersed in 1 mL cell culture medium (Dulbecco's modified Eagle's medium, 1% fetal bovine serum, 1.25 μg/mL amphotericin B, 50 μg/mL gentamicin, and 0.1 U/mL penicillin/streptomycin) such that the dermis contacted the medium in the well and the epidermis was exposed to air. There was 1 biopsy per well. The lid of the 12-well plate was elevated using two sterile 1 mL pipette tips to limit moisture build-up. The total incubation time of the drug product treated skin was 21 hours and 15 minutes.


SNA Synthesis. The oligonucleotide, SEQ ID NO: 288, was synthesized via standard solid-phase phosphoramidite chemistry. The molecule was synthesized with successive cycles of de-protection, monomer addition, capping, and oxidation. The DNA, and 2′-O-Me RNA monomers were obtained from Sigma Aldrich Biochemie (Germany). The hexa(ethylene glycol) spacers and cholesterol modifier were obtained from ChemGenes Corporation. Linkages were either standard phosphodiesters or phosphorothioates prepared with a solution of 0.2 M phenylacetyl disulfide (PADS) in a mixture of 1:1 lutidine:ACN. Synthesis was performed DMT-off, in the 3′ to 5′ direction. After synthesis, the oligonucleotide was cleaved from the support and de-protected using a 3:1 mixture of ammonium hydroxide and ethanol at 65° C. for 17 hours. The oligonucleotide was purified via anion exchange high performance liquid chromatography (AX-HPLC) techniques. Then the oligonucleotide solution was lyophilized completely to a dry powder. Verification of the oligonucleotide product molecular weight was performed using electrospray ionization mass spectrometry (ESI-MS) and dual-MS sequencing was performed to confirm the oligonucleotide sequence. Oligonucleotide concentration was determined by UV-absorbance at 260 nm.


Liposomes were formulated by Avanti Polar Lipids (Alabama, USA). First, 500 g 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) was dissolved in chloroform to a final concentration of 50 mg/mL. The solvent was then removed under nitrogen to form a thin lipid film. The film was lyophilized overnight to ensure all solvent was removed. The lipid film was subsequently hydrated with Hyclone sterile, nuclease-free water (SH30538.03, GE) at a concentration of 100 mg/mL and sonication/freeze-fracture were used to form large, unilamellar vesicles. These vesicles were then continuously homogenized through a micro-fluidizer at up to 25 kpsi, until the mean number diameter of 20 nm was achieved. The liposome solution was further concentrated by tangential flow filtration (TFF) to a final lipid concentration of 208 mg/mL. Lipid concentrations were determined using a choline quantification assay and nuclear magnetic resonance spectroscopy. The particle size and dispersion were measured by dynamic light scattering (DLS).


SNAs targeting human IL-17RA, were formulated by mixing a 30-fold molar excess of cholesterol-modified full length oligonucleotide to the liposome suspension in Hyclone sterile, nuclease-free water (SH30538.03, GE) followed by overnight incubation at 4° C.


Drug Application to the Skin. The pre-formulated SNAs targeting human IL-17RA were mixed into a gel vehicle, resulting in four IL17RA_282 gel strengths of 0, 0.01, 0.1 and 1.0% w/w (full length oligonucleotide weight). The composition of these drug products can be found in Table 4.


The IL17RA_282 gels were then applied topically to the skin explants in replicates of eight. During the sample processing, one sample in the 1.0% group was lost, resulting in only 7 replicates for that group. Each replicate was taken from a different patient for accurate representation and comparison of a patient specific response. The IL17RA_282 gels were briefly mixed prior to dosing using a pipette tip. 10 μL of each drug product was dosed topically to each 3 mm biopsy using a positive displacement pipette. The pipette tip was used to spread the drug product evenly covering the entire exposed surface area of the skin. The actual dosing time was recorded and the explants were maintained at 32° C. and ambient humidity for 21 hours.


At the conclusion of the 21 hour incubation period, the biopsies were removed from each membrane. A cotton swab wetted with PBS was used to remove the drug product and then the application area was blotted dry. The biopsy punches were lysed in RLT buffer (Qiagen) with a bead homogenizer and stored frozen at −80° C. until shipped on dry ice to the research facility.


RNA Extraction and qRT-PCT. RNA was isolated from lysates using the RNEasy 96-well kit (Qiagen) according to the manufacturer's instructions. cDNA was then synthesized from RNA isolates using the cDNA high capacity reverse transcription kit (Life Technologies). cDNA was prepared on a thermocycler with the following temperature program: 25° C. for 10 minutes, 37° C. for 90 minutes, 85° C. for 5 minutes followed by a 4° C. hold. The resulting cDNA was diluted 8 fold with nuclease-free water. qPCR was performed using 6 μL of the diluted cDNA, 4.66 μL LightCycler480 Probes Master Mix (Roche), 0.47 μL human IL-17RA specific FAM-labeled probe and primers, and 0.37 μL human Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) specific HEX-labeled probe and primers per reaction well of a 384-well optical reaction plate (Roche). The primer and probe set for IL-17RA was purchased commercially (Hs01064648_m1, Thermo Fisher Scientific). The primer and probe set for GAPDH was designed using the known human genome sequence (NCBI reference sequences NM_002046.5) and was found to be specific by “blastn” analysis (NCBI). The oligonucleotide sequences used for GAPDH were: forward 5′-CAA GGT CAT CCA TGA CAA CTT TG-3′ (SEQ ID NO: 4), reverse 5′-GGG CCA TCC ACA GTC TTC T-3′ (SEQ ID NO: 5), probe 5′-HEX-ACC ACA GTC CAT GCC ATC ACT GCC A-BHQ1-3′ (SEQ ID NO: 6). qPCR reactions, in technical duplicate, were carried out on the Roche Lightcycler 480 under the following conditions: initial denaturation at 95° C. for 10 minutes and then 50 cycles of denaturation at 95° C. for 10 seconds, annealing at 60° C. for 30 seconds and extension at 72° C. for 1 second. Cp values were obtained by analysis with the 2nd derivative method. Relative gene expression was determined by normalization with the housekeeping gene (GAPDH) and the ΔΔ-Ct method. Statistical analysis was done by performing a one sample t-test.









TABLE 4







Drug Product Compositions









Gel Strength Formulations (w/w)











Ingredient
0%
0.01%
0.1%
1.0%














SEQ ID NO: 288
0
0.01419
0.1419
1.419


DOPC
0
0.2655
2.655
26.55


Diethylene glycol
25
25
25
25


monoethyl ether






(Transcutol P)






Glycerin
5
5
5
5


Hydroxyethyl Cellulose
1
1
1
1


Methylparaben
0.15
0.15
0.15
0.15


Propylparaben
0.05
0.05
0.05
0.05


Disodium EDTA
0.1
0.1
0.1
0.1


Sodium Metabisulfite
0.2
0.2
0.2
0.2


Water
68.5
68.2
65.7
40.5









Results


The IL17RA_282 gels showed dose-dependent knockdown of IL-17RA mRNA in human psoriatic skin biopsies after 21 hours of treatment (FIG. 11). Knockdown was observed when comparing the IL17RA_282-containing gels to the 0% gel strength formulation. Treatment with the IL17RA_282 gels resulted in statistically significant reduction of IL-17RA mRNA.


Discussion and Conclusions


It was shown here that when the IL17RA_282 gel is applied topically to ex vivo human psoriatic skin biopsies, there is a pharmacodynamic effect seen at the level of IL-17RA mRNA. The response observed in this study represents only a 21 hour treatment. It is expected that longer treatment with multiple doses would further enhance the inhibition observed. Given the importance of IL-17 signaling in maintaining the diseased state observed in psoriasis, a strong topical drug product candidate for the treatment of psoriasis is presented.


Example 4. Multiplex SNAs Targeting Three Different mRNAs

Materials and Methods


Multiplex SNA Synthesis. SNAs were formulated by mixing a 100× molar excess (unless otherwise stated) of cholesterol-modified oligonucleotide to a liposome suspension in PBS and storing them overnight, protected from light, at 4° C. Four different oligonucleotides were used for the synthesis process: an antisense oligonucleotide targeting human IL-4R, an antisense oligonucleotide targeting human IL-1β, an antisense oligonucleotide targeting human CTGF, and a same length control oligonucleotide that does not have any complementarity to known genes (confirmed by BLAST). For monoplex SNAs, the oligonucleotide population consisted of 33% targeted antisense oligonucleotide and 66% control oligonucleotide. For multiplex SNAs containing two different targeted antisense oligonucleotides, the oligonucleotide population consisted of 33% of each targeted antisense oligonucleotide and the remaining 33% control oligonucleotide (FIG. 12). For multiplex SNAs containing three different targeted antisense oligonucleotides, the oligonucleotide population consisted of 33% of each targeted antisense oligonucleotide. Control SNAs consisted of 100% of the control oligonucleotide (FIG. 9).


Cell Culture Studies. Primary human foreskin keratinocytes (HFKs) were seeded at passage 5 in 96-well, tissue culture plates at a cell density of 17,000 cells per well. Cells were allowed to rest in the incubator overnight following plating. Cells were treated in triplicate with either an IL-4R targeted antisense SNA, IL-1β targeted antisense SNA, CTGF targeted antisense SNA, a multiplex SNA targeting more than one transcript or a non-complementary control SNA (confirmed by NCBI Blast), comprising the same ‘gap-mer’ design and 3′-chemical modifications, at concentrations of 1000, 100, 10 and 1 nM in fresh maintenance media. Unless otherwise stated, all treatments lasted 24 hours.


RNA Extraction and Quantitative Reverse Transcriptase Polymerase Chain Reaction (qRT-PCR). HFK cells were lysed in RLT Buffer (Qiagen) at 24 hours post-transfection. RNA was isolated from lysates using the RNEasy 96-well kit (Qiagen) according to the manufacturer's instructions. cDNA was then synthesized from RNA isolates using the cDNA High Capacity Reverse Transcription Kit (Life Technologies). Samples were run on a thermocycler at 25° C. for 10 minutes, 37° C. for 90 minutes, 85° C. for 5 minutes and held at 4° C. to generate cDNA. qPCR was performed using 6 μL of the synthesized cDNA, 4.66 μL LightCycler480 Probes Master Mix (Roche), 0.47 μL target specific FAM-labeled probe and primers, and 0.37 μL human Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) specific HEX-labeled probe and primers per reaction well of a 384-well optical reaction plate (Roche). The primer and probe sets for IL-4R, IL-1β and CTGF were purchased from ThermoFisher Scientific (catalogue numbers Hs00166237_m1, Hs01555410_m1 and Hs01026927_g1, respectively). The primer and probe set for GAPDH was designed using the known human genome sequence (NCBI reference sequence NM_002046.5) and was found to be specific by “blastn” analysis (NCBI). The oligonucleotide sequences used for GAPDH were: forward 5′-CAA GGT CAT CCA TGA CAA CTT TG-3′ (SEQ ID NO: 4), reverse 5′-GGG CCA TCC ACA GTC TTC T-3′ (SEQ ID NO: 5), probe 5′-HEX-ACC ACA GTC CAT GCC ATC ACT GCC A-BHQ1-3′ (SEQ ID NO: 6). qPCR reactions, in technical duplicate, were carried out on the Roche Lightcycler 480 under the following conditions: initial denaturation at 95° C. for 10 minutes and then 50 cycles of denaturation at 95° C. for 10 seconds, annealing at 60° C. for 30 seconds and extension at 72° C. for 1 second. Cp values were obtained by analysis with the 2nd derivative method. Relative gene expression was determined by normalization with the housekeeping gene (GAPDH) and the ΔΔ-Ct method. Each treatment was compared to its respective concentration control.


Results


Seven different SNA configurations were made to test whether three different mRNAs can be targeted using single SNA construct. Three mRNAs were chosen that were not functionally dependent on each other for expression and were not in a single known pathway. In certain cases, it may be advantageous to target mRNAs that are part of the same pathway, e.g. TNF, IL17, IL22, to control a specific pathway. SNAs were synthesized targeting each individual mRNA alone, three pairs of mRNAs and one SNA configuration that can target all three mRNAs. Control oligonucleotide was used to ensure that each SNA configuration has the same number of targeted and non-targeted oligonucleotides.


The results from cell culture experiments demonstrate that single SNA can be formulated that targets three different mRNAs. The mRNAs chosen were interleukin 4 receptor (IL4R), interleukin 1 beta (IL1b) and connective tissue growth factor (CTGF). These targets were chosen because in other experiments it was noted that knocking down any one of these mRNAs didn't significantly reduce the levels of the other two. The results show that when antisense oligonucleotides targeting any single mRNA is present on the SNA, the expression of that mRNA is reduced in a dose dependent manner (FIG. 13 for IL4R, FIG. 14 for IL1b, FIG. 15 for CTGF). This configuration can be extended to target more than three mRNAs. The total antisense loading per SNA may be the theoretical limit to the number of mRNAs that can be targeted with a single SNA configuration.


REFERENCES





    • 1. Banga R J, Chernyak N, Narayan S P, Nguyen S T, Mirkin C A. Liposomal Spherical Nucleic Acids. J. Am. Chem. Soc. 2014 136: 9866-9869.

    • 2. Ding Y and Lawrence C E. A Statistical Sampling Algorithm for RNA Secondary Structure





Prediction. Nucleic Acids Res. 2003: 31: 7280-7301.

    • 3. Chabaud M1, Fossiez F, Taupin J L, Miossec P. 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. 1998 Jul. 1; 161(1):409-14.
    • 4. Miossec P. Interleukin-17 in rheumatoid arthritis: if T cells were to contribute to inflammation and destruction through synergy. Arthritis Rheum. 2003 March; 48(3):594-601.


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 multiplex antisense oligonucleotide spherical nucleic acid (mASO-SNA), comprising a core having an oligonucleotide shell comprised of an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a first gene and an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a second gene, wherein the core is a liposome core and the oligonucleotide shell is positioned on the exterior of the core,wherein each antisense oligonucleotide comprises at least two modified nucleosides,wherein each antisense oligonucleotide further comprises a molecular species at the 5′ end, 3′ end, or 5′ and 3′ end of the antisense oligonucleotide,wherein the molecular species is a lipid moiety connected to the antisense oligonucleotide by a linker, wherein the linker comprises hexaethylene glycol.
  • 2. The mASO-SNA of claim 1, wherein the first gene and the second gene are associated with a disease.
  • 3. The mASO-SNA of claim 1, wherein the disease is an inflammatory disorder.
  • 4. The mASO-SNA of claim 1, wherein the disease is psoriasis.
  • 5. The mASO-SNA of claim 1, wherein the first gene is associated with a target pathway and the second gene is associated with the target pathway.
  • 6. The mASO-SNA of claim 1, further comprising an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a third gene.
  • 7. The mASO-SNA of claim 1, wherein the first gene is an Interleukin 17 receptor (IL-17RA).
  • 8. The mASO-SNA of claim 1, wherein the second gene is TNF.
  • 9. The mASO-SNA of claim 1, wherein the antisense oligonucleotide targeted to the first gene and the antisense oligonucleotide targeted to the second gene are present in an approximate equimolar amount in the oligonucleotide shell.
  • 10. A method for treating a disorder, comprising: administering to a subject having a disorder a multiplex antisense oligonucleotide spherical nucleic acid (mASO-SNA), comprising a core having an oligonucleotide shell comprised of an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a first gene and an antisense oligonucleotide 10 to 30 linked nucleosides in length targeted to a second gene, wherein the core is a liposome core and the oligonucleotide shell is positioned on the exterior of the core in an effective amount to treat the disorder, wherein each antisense oligonucleotide comprises at least two modified nucleosides,wherein each antisense oligonucleotide further comprises a molecular species at the 5′ end, 3′ end, or 5′ and 3′ end of the antisense oligonucleotide,wherein the molecular species is a lipid moiety connected to the antisense oligonucleotide by a linker, wherein the linker comprises hexaethylene glycol.
  • 11. The method of claim 10, wherein the mASO-SNA produces simultaneous mRNA knock-down of the first and second gene.
  • 12. The mASO-SNA of claim 6, wherein the first gene is associated with a target pathway, the second gene is associated with the target pathway, and the third gene is associated with the target pathway.
  • 13. The mASO-SNA of claim 1, wherein the first gene is associated with a target pathway and the second gene is associated with a second target pathway which is not the target pathway associated with the first gene.
  • 14. The mASO-SNA of claim 6, wherein the first gene is associated with a target pathway, the second gene is associated with a second target pathway, and the third gene is associated with a third target pathway, wherein each of the: target pathway; second target pathway; and third target pathway are different target pathways.
  • 15. The mASO-SNA of claim 1, wherein the first gene is an interleukin-4 receptor (IL-4R).
  • 16. The mASO-SNA of claim 1, wherein the second gene is interleukin-1 beta (IL-1β).
  • 17. The mASO-SNA of claim 6, wherein the third gene is connective tissue growth factor (CTGF).
  • 18. The mASO-SNA of claim 6, wherein the antisense oligonucleotide targeted to the first gene, the antisense oligonucleotide targeted to the second gene, and the antisense oligonucleotide targeted to the third gene are present in an approximate equimolar amount in the oligonucleotide shell.
  • 19. A pharmaceutical composition, comprising the mASO-SNA of claim 1.
  • 20. The mASO-SNA of claim 1, wherein the lipid moiety is a cholesterol moiety.
  • 21. The mASO-SNA of claim 1, wherein each antisense oligonucleotide comprises a gap segment consisting of 2 to 8 linked deoxynucleosides, a 5′ wing segment consisting of linked nucleosides, and a 3′ wing segment consisting of linked nucleosides, wherein the gap segment is positioned between the 5′ wing segment and the 3′ wing segment, wherein at least one nucleoside of each wing segment comprises a modified nucleotide.
RELATED APPLICATIONS

This application is a national stage filing under 35 U.S.C. § 371 of International Patent Application Serial No. PCT/US2017/031428, entitled “LIPOSOMAL SPHERICAL NUCLEIC ACID (SNA) CONSTRUCTS PRESENTING ANTISENSE OLIGONUCLEOTIDES (ASO) FOR SPECIFIC KNOCKDOWN OF INTERLEUKIN 17 RECEPTOR MRNA” filed May 5, 2017, which claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application Ser. No. 62/333,082, entitled “LIPOSOMAL SPHERICAL NUCLEIC ACID (SNA) CONSTRUCTS PRESENTING ANTISENSE OLIGONUCLEOTIDES (ASO) FOR SPECIFIC KNOCKDOWN OF INTERLEUKIN 17 RECEPTOR MRNA”, filed May 6, 2016, the entire contents of each of which are incorporated by reference herein.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2017/031428 5/5/2017 WO
Publishing Document Publishing Date Country Kind
WO2017/193087 11/9/2017 WO A
US Referenced Citations (188)
Number Name Date Kind
5264618 Felgner et al. Nov 1993 A
5472881 Beebe et al. Dec 1995 A
5512439 Hornes et al. Apr 1996 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
6197525 Yao et al. Mar 2001 B1
6214806 Krieg et al. Apr 2001 B1
6218371 Krieg et al. Apr 2001 B1
6339068 Krieg et al. Jan 2002 B1
6361944 Mirkin et al. Mar 2002 B1
6429199 Krieg et al. Aug 2002 B1
6482923 Shi et al. Nov 2002 B1
6506564 Mirkin et al. Jan 2003 B1
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
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
7217412 Chen et al. May 2007 B2
7256264 Goddard et al. Aug 2007 B2
7332586 Franzen et al. Feb 2008 B2
7404969 Chen et al. Jul 2008 B2
7473763 Goddard et al. Jan 2009 B2
7514099 Chen et al. Apr 2009 B2
7544482 Goddard et al. Jun 2009 B2
7611857 Medlock et al. Nov 2009 B2
7638603 Shi et al. Dec 2009 B2
7655761 Gorman et al. Feb 2010 B2
7696150 Shaughnessy Apr 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
8133734 Shi et al. Mar 2012 B2
8188254 Uhlmann et al. May 2012 B2
8202979 McSwiggen et al. Jun 2012 B2
8273866 McSwiggen et al. Sep 2012 B2
8323686 Mirkin 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
8933046 Machuy et al. Jan 2015 B2
9139827 Mirkin et al. Sep 2015 B2
9389236 Fandl et al. Jul 2016 B2
9506056 Mirkin et al. Nov 2016 B2
9532948 Mirkin et al. Jan 2017 B2
9617541 Mirkin et al. Apr 2017 B2
9844562 Mirkin et al. Dec 2017 B2
9889209 Mirkin et al. Feb 2018 B2
10098958 Mirkin et al. Oct 2018 B2
10182988 Mirkin et al. Jan 2019 B2
10208310 Mader et al. Feb 2019 B2
10370656 Mirkin et al. Aug 2019 B2
10391116 Mirkin et al. Aug 2019 B2
10398784 Mirkin et al. Sep 2019 B2
10434064 Radovic-Moreno et al. Oct 2019 B2
10653780 Hope et al. May 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
11123294 Radovic-Moreno et al. Sep 2021 B2
11213593 Mirkin et al. Jan 2022 B2
11364304 Mirkin et al. Jun 2022 B2
20020039568 Moore et al. Apr 2002 A1
20020172711 Martin et al. Nov 2002 A1
20030036568 Raoof Feb 2003 A1
20030147966 Franzen et al. Aug 2003 A1
20030170162 Nayfeh et al. Sep 2003 A1
20040023335 Jing et al. Feb 2004 A1
20040023382 Dean Feb 2004 A1
20040053384 Sligar et al. Mar 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
20050130911 Uhlmann et al. Jun 2005 A1
20050208572 Shaughnessy Sep 2005 A1
20060014191 Lao 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
20060292174 De Los Rios et al. Dec 2006 A1
20070065868 Jing Mar 2007 A1
20070280936 Moore et al. Dec 2007 A1
20080194463 Weller 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
20090209629 Mirkin et al. Aug 2009 A1
20090275735 Golstein 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
20100136682 Mirkin et al. Jun 2010 A1
20100143246 Shi et al. Jun 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
20110305734 Edelson et al. Dec 2011 A1
20120082616 Trawick Apr 2012 A1
20120149843 Chien et al. Jun 2012 A1
20120244230 Mirkin et al. Sep 2012 A1
20130004520 Andersson et al. Jan 2013 A1
20130034599 Thaxton et al. Feb 2013 A1
20130089614 Zhang 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
20130172404 Mirkin et al. Jul 2013 A1
20130178610 Mirkin et al. Jul 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
20140179767 Rozet et al. Jun 2014 A1
20140294927 Thaxton et al. Oct 2014 A1
20150031745 Mirkin et al. Jan 2015 A1
20150080320 Desai Mar 2015 A1
20150086985 Giljohann et al. Mar 2015 A1
20150352138 Mirkin et al. Dec 2015 A1
20160053260 Mirkin et al. Feb 2016 A1
20160123964 Tumeh et al. May 2016 A1
20160186178 Radovic-Moreno et al. Jun 2016 A1
20160194642 Gryaznov et al. Jul 2016 A1
20160310425 Mirkin et al. Oct 2016 A1
20170044544 Mirkin et al. Feb 2017 A1
20170137809 Mirkin et al. May 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
20170306331 Mader et al. Oct 2017 A1
20180042848 Gryaznov et al. Feb 2018 A1
20180085390 Mirkin et al. Mar 2018 A1
20180193484 Mirkin et al. Jul 2018 A1
20180214376 Giljohann Aug 2018 A1
20180311176 Ozsolak 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
20190142739 Patel 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
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
20220064649 Anderson et al. Mar 2022 A1
20220088059 Patel et al. Mar 2022 A1
20220348917 Sarett Nov 2022 A1
Foreign Referenced Citations (125)
Number Date Country
1335884 Feb 2002 CN
1341660 Mar 2002 CN
1357042 Jul 2002 CN
101218254 Jul 2008 CN
101824088 Sep 2010 CN
102036652 Apr 2011 CN
102165061 Aug 2011 CN
102197051 Sep 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
1 899 911 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
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 1996029408 Sep 1996 WO
WO 199804740 Feb 1998 WO
WO 1998023284 Jun 1998 WO
WO-9927086 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 2005030259 Apr 2005 WO
WO 2005063201 Jul 2005 WO
WO 2005063288 Jul 2005 WO
WO 2005108616 Nov 2005 WO
WO 2005116226 Dec 2005 WO
WO 2006015560 Feb 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 200842156 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 2010081049 Jul 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 2011053940 May 2011 WO
WO 2011072133 Jun 2011 WO
WO 2011078672 Jun 2011 WO
WO 2011079290 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 2013086207 Jun 2013 WO
WO 2014025795 Feb 2014 WO
WO 2014123935 Aug 2014 WO
WO 2014169264 Oct 2014 WO
WO 2015013675 Jan 2015 WO
WO 2015153975 Oct 2015 WO
WO 2015187966 Dec 2015 WO
WO 2015195628 Dec 2015 WO
WO 2016057549 Apr 2016 WO
WO 2016115320 Jul 2016 WO
WO 2016149323 Sep 2016 WO
WO 2017011662 Jan 2017 WO
WO 2017035278 Mar 2017 WO
WO 2017161032 Sep 2017 WO
WO 2017193081 Nov 2017 WO
WO 2018152327 Aug 2018 WO
WO 2018209270 Nov 2018 WO
WO 2019118883 Jun 2019 WO
WO 2019168558 Sep 2019 WO
WO 2020168005 Aug 2020 WO
WO 2021202557 Oct 2021 WO
WO 2022032182 Feb 2022 WO
WO 2022036013 Feb 2022 WO
WO 2022147541 Jul 2022 WO
WO 2022150369 Jul 2022 WO
Non-Patent Literature Citations (193)
Entry
Rubenstein et al, Antisense oligonucleotide intralesional therapy for human PC-3 prostate tumors carried in athymic nude mice, Journal of Surgical Oncology, 1996, 62: 194-200 (Year: 1996).
Hurst et al, Maximizing DNA Loading on a Range of Gold Nanoparticle Sizes, Anal.Chem., 2006, 78: 8313-8318 (Year: 2006).
IDT catalog, 2023, pp. 1-2 (Year: 2023).
International Preliminary Report on Patentability dated Nov. 15, 2018 in connection with PCT/US2017/031428.
Partial Supplementary European Search Report dated Nov. 22, 2019 in connection with EP 17793513.7.
Extended European Search Report dated Mar. 27, 2020 in connection with EP 17793513.7.
Agbasi-Porter et al., Transcription inhibition using oligonucleotide-modified gold nanoparticles, Bioconjugate Chem., 17(5):1178-83 (2006).
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.
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.
Cload et al., Polyether tethered oligonucleotide probes. J. Am. Chem. Soc. 1991;113(16): 6324-6.
Cormode, D.P. et al., “Nanocrystal Core High-Density Lipoproteins: A Multimodality Contrast Agent Platform,” Nano Lett., 2008, 8 (11), 3715-3723.
DeMesmaeker et al., Antisense oligonucleotides. Acc. Chem. Res. 1995;28(9): 366-74.
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.
Fattal et al., Biodegradable polyalkylcyanoacrylate nanoparticles for the delivery of oligonucleotides. J. Controlled Release. 1998;53:137-143.
Ghosh et al., Gold nanoparticles in delivery applications. Adv. Drug Deliv. Rev. 2008;60(11):1307-15.
Hames et al., Gene Probes 1 A Practical Approach, IRL Press, New York (1995).
Hayashi, Ultrafine particles. J. Vac. Sci. Technol. 1987;5(4):1375-1384.
Hellstrom et al., Epitaxial growth of DNA-assembled nanoparticle superlattices on patterned substrates. Nano Lett. 2013;13(12):6084-90. doi: 10.1021/nl4033654. Epub Nov. 20, 2013.
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.
Kolarova et al., Preparation of magnetic oligo (dT) particles. Biotechniques. 1996;20: 196-8.
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., Chip-based scanometric detection of mercuric ion using DNA-functionalized gold nanoparticles. Anal. Chem. 2008;80(17):6805-8.
Li et al., “Molecular spherical nucleic acids,” PNAS pp. 1-5 (2018).
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., 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.
Li et al., Reversible and Chemically Programmable Micelle Assembly with DNA Block-Copolymer Amphiphiles, Nano Lett. 2004;4:1055.
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.
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.
Marinakos et al., Template Synthesis of One-Dimensional Au, Au-Poly(pyrrole), and Poly(pyrrole) Nanoparticle Arrays. Chem. Mater. 1998;10:1214-19.
Matijevic et al., Fine Particles Part II: Formation Mechanisms and Applications. MRS Bulletin pp. 16-47 (1990).
Maye et al., A simple method for kinetic control of DNA-induced nanoparticle assembly. J. Am. Chem. Soc. 2006; 128: 14020-1.
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.
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.
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.
Park et al., DNA-programmable nanoparticle cystrallization. Nature. 2008;451: 553-6.
Patel et al., Scavenger receptors mediate cellular uptake of polyvalent oligonucleotide-functionalized gold nanoparticles, Bioconj. Chem., 21:2250 (2010).
Patil et al., DNA-based therapeutics and DNA delivery systems: a comprehensive review. AAPS J., 2005;7(1): E61-77.
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.
Storhoff et al., Sequence-Dependent Stability of DNA-Modified Gold Nanoparticles. Langmuir. 2002;18: 6666-6670.
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).
Tondelli et al., “Highly efficient cellular uptake of c-myb antisense oligonucleotides through specifically desianed polymeric nanospheres,” Nucl. Acids Res. 26:5425-5431 (1998).
Wagner et al., Gene inhibition using antisense oligodeoxynucleotides. Nature, 1994;372: 333-5.
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.
Yamane et al., On the complexing of desoxyribonucleic acid (DNA) by mercuric ion. J. Am. Chem. Soc., 1961;83:2599-2607.
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.
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).
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.
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.
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., 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.
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.
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.
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.
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.
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.
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):3818-21. 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.
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.
He et al., Phospholipid-stabilized Au-nanoparticles. Biomacromolecules. May-Jun. 2005;6(3):1224-5.
Hurst, S. et al., Maximizing DNA Loading on a Range of Gold Nanoparticle Sizes, Anal. Chem., 2006, 78 (24), 8313-8318.
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.
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.
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., 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., 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.
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.
Li et al., Nanofabrication by DNA self-assembly. Materials Today. Elsevier Science. May 1, 2009;12(5)24-32.
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.
Luo et al., Locally instilled tumor necrosis factor α 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.
Massich et al., Regulating immune response using polyvalent nucleic acid-gold nanoparticle conjugates. Mol Pharm. Nov.-Dec. 2009;6(6):1934-40.
Matsunaga, T. et al., Biomagnetic Nanoparticle Formation and Application, Supramolecular Science, 1998, 5 (3-4), 391-394.
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/nl1041947. 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.
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.
Nam et al., Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins. Science. Sep. 26, 2003;301(5641):1884-6.
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.
Pan et al., Dendrimer-Modified Magnetic Nanoparticles Enhance Efficiency of Gene Delivery System. Cancer Res. 2007;67:8156-8163.
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.
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.
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.
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.
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.
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.
Thompson et al., Smart lipids for programmable nanomaterials. Nano Lett. Jul. 14, 2010;10(7):2690-3. doi: 10.1021/nl101640k.
Tiwari et al., Functionalized gold nanoparticles and their biomedical applications. Nanomaterials. 2011;1:31-63. doi: 10.3390/nano1010031.
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.
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.
Xiao et al., Mannosylated bioreducible nanoparticle-mediated macrophage-specific TNF-α RNA interference for IBD therapy. Biomaterials. Oct. 2013;34(30):7471-82. doi: 10.1016/j.biomaterials.2013.06.008. Epub Jun. 29, 2013.
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.
Yin et al., Supramolecular self-assembled nanoparticles mediate oral delivery of therapeutic TNF-α 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 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 cross-linking 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.
PCT/US2017/031428, Nov. 15, 2018, International Preliminary Report on Patentability.
EP 17793513.7, Nov. 22, 2019, Partial Supplementary European Search Report mailed.
EP 17793513.7, Mar. 27, 2020, Extended European Search Report.
Cottrez et al., Genes specifically modulated in sensitized skins allow the detection of sensitizers in a reconstructed human skin model. Development of the SENS-IS assay. Toxicol In Vitro. Jun. 2015;29(4):787-802. doi: 10.1016/j.tiv.2015.02.012. Epub Feb. 24, 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.
Kapadia et al., Spherical Nucleic Acid Nanoparticles: Therapeutic Potential. BioDrugs. Aug. 2018;32(4):297-309. doi: 10.1007/s40259-018-0290-5. Author Manuscript. 24 pages.
Manoharan et al., Lipidic nucleic acids. Tetrahedron Letters. May 22, 1995;36(21):3651-4.
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.
Periyasamy et al., Nanomaterials for the local and targeted delivery of osteoarthritis drugs. J Nanomater. 2012:2012:Article 673968. 13 pages. doi:0.1155/2012/673968.
Ward-Kavanagh et al., The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity. May 17, 2016;44(5):1005-19. doi: 10.1016/j.immuni.2016.04.019.
Yao et al. Human IL-17: a novel cytokine derived from T cells. J Immunol. Dec. 15, 1995; 155(12):5483-6.
U.S. Appl. No. 15/301,467, filed Oct. 3, 2016, Gryaznov.
U.S. Appl. No. 16/099,404, filed Nov. 6, 2018, Anderson et al.
U.S. Appl. No. 16/099,385, filed Nov. 6, 2018, Kang et al.
U.S. Appl. No. 17/639,938, filed Mar. 3, 2022, Sarett.
Inoue, Oligonucleotide therapeutics: past, present and future. Drug Delivery System. Jan. 2016;31(1):10-23.
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.
Somiya et al., Potential of a non-cationic liposomes-based delivery system for nucleic acid medicines. Drug Delivery System. Jan. 2016;31(1):35-43.
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.
Skakuj et al., Conjugation Chemistry-Dependent T-Cell Activation with Spherical Nucleic Acids. J Am Chem Soc. Jan. 31, 2018;140(4):1227-1230 and Supplemental Information. doi: 10.1021/jacs.7b12579. Epub Jan. 22, 2018. 13 pages.
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.
Chan et al., Lipid-anchored DNA mediates vesicle fusion as observed by lipid and content mixing. Biointerphases. Jun. 2008;3(2):FA17. doi: 10.1116/1.2889062.
Ming et al., Bioconjugates for targeted delivery of therapeutic oligonucleotides. Adv Drug Deliv Rev. Jun. 29, 2015;87:81-9. doi: 10.1016/j.addr.2015.02.002. Epub Feb. 15, 2015.
Raouane et al., Lipid conjugated oligonucleotides: a useful strategy for delivery. Bioconjug Chem. Jun. 20, 2012;23(6):1091-104. doi: 10.1021/bc200422w. Epub Mar. 15, 2012.
U.S. Appl. No. 17/066,134, filed Oct. 8, 2020, Radovic-Moreno et al.
U.S. Appl. No. 16/095,134, filed Oct. 19, 2018, Patel et al.
U.S. Appl. No. 17/231,896, filed Nov. 6, 2018, Anderson et al.
U.S. Appl. No. 16/099,409, filed Nov. 6, 2018, Nallagatla et al.
U.S. Appl. No. 16/608,685, filed Oct. 25, 2019, Kang et al.
U.S. Appl. No. 17/253,102, filed Dec. 16, 2020, Anderson.
U.S. Appl. No. 17/430,277, filed Aug. 11, 2021, Bexon et al.
U.S. Appl. No. 16/976,168, filed Aug. 27, 2020, Kang et al.
U.S. Appl. No. 17/605,784, filed Oct. 22, 2021, Anderson.
U.S. Appl. No. 17/639,938, filed Mar. 3, 2022, Sarett et al.
U.S. Appl. No. 17/011,658, filed Sep. 3, 2020, Mirkin et al.
U.S. Appl. No. 18/019,857, filed Feb. 6, 2023, Anderson et al.
U.S. Appl. No. 18/020,501, filed Feb. 9, 2023, Anderson et al.
Related Publications (1)
Number Date Country
20200308579 A1 Oct 2020 US
Provisional Applications (1)
Number Date Country
62333082 May 2016 US