COMPOSITIONS AND METHODS FOR IMMUNOSTIMULATORY RNA OLIGONUCLEOTIDES

Information

  • Patent Application
  • 20090111765
  • Publication Number
    20090111765
  • Date Filed
    September 14, 2006
    17 years ago
  • Date Published
    April 30, 2009
    15 years ago
Abstract
The present invention provides 4-nucleotide (4mer) RNA motifs that confer immunostimulatory activity, in particular, IL-12-inducing activity to a single-stranded RNA oligonucleotide. The present invention also provides single-stranded RNA oligonucleotides, including antisense RNA, with high or low immunostimulatory activity. The present invention further provides the use of the RNA oligonucleotides of the invention for therapeutic purposes.
Description
FIELD OF THE INVENTION

The present invention relates to the field of immunotherapy and drug discovery. The present invention provides a method for determining the immunostimulatory activity of a RNA oligonucleotide. The present invention also provides a method for predicting the immunostimulatory activity of a single-stranded RNA oligonucleotide (ssRNA). The present invention further provides a method for preparing ssRNA oligonucleotides with high or low immunostimulatory activity. Moreover, the present invention provides ssRNA oligonucleotides with immunostimulatory activity and the therapeutic uses thereof. In addition, the present invention provides antisense RNA with gene silencing activity and with either high or low immunostimulatory activity, the methods of their preparation, and their therapeutic uses.


BACKGROUND OF THE INVENTION

The development of synthetic compounds that mimic the presence of viruses may result in potent novel candidate drugs for the treatment of viral infection and cancer. The immune system employs at least four members of the family of Toll-like receptors (TLR3, TLR7, TLR8 and TLR9) to detect viruses based on the presence of certain characteristics of viral nucleic acid [1]. By far the most information is available for TLR9 [2, 3]. TLR9 detects so-called CpG motifs within microbial DNA [4]. Synthetic oligonucleotides containing such CpG motifs (CpG ODN) have been extensively studied over the last 10 years, and the lead compound ODN 2006 (identical with ODN 7909, ProMune) is currently entering clinical phase III for the treatment of cancer. Although CpG ODN represent strong Th1 vaccine adjuvants and show excellent anti-tumor activity in murine models of cancer [5, 6], their application for the treatment of human disease is limited. In mice TLR9 is expressed on B cells and both myeloid and plasmacytoid dendritic cells, whereas in humans expression is restricted to B cells and PDC [7, 8]. As a consequence, in the absence of T cell help, in mice CpG ODN stimulate both IL-12 and IFN-α in mice, while in humans, CpG ODN stimulate only IFN-α.


Recent data suggest that this deficit of CpG ODN in humans can be overcome by RNA oligonucleotides. Unlike TLR9 detecting DNA, TLR3, TLR7 and TLR8 all recognize RNA. TLR3 binds to long double-stranded (ds) RNA [9] and therefore by definition can not be activated by short synthetic oligoribonucleotides (ORN). TLR7 detects short dsRNA (as used for siRNA) [10] and both long and short single-stranded (ss) RNA [10-12]. RNA recognition by TLR8 is limited to ssRNA. Both TLR7 and TLR8 are expressed in human myeloid cells and thus, ORNs that serve as ligands for TLR7 and TLR8 are excellent candidates for eliciting the desired IL-12 response which is missing when CpG ODN is used for stimulation in the human system.


While for TLR9 the optimal sequence motif (CpG motif) has been exactly defined [4, 13, 14], the situation is much less clear for TLR7 and TLR8. It has been suggested that the potency of RNA oligonucleotides depends on a high content of G and U. One group proposed that the presence of the UGUGU motif confers IFN-α-inducing activity to RNA oligonucleotides. Our group defined a 9mer sequence motif [10]. WO 03/086280 discloses that guanosine, particularly guanosine in combination with uracil, are natural ligands of TLR8. Additional TLR8 ligands disclosed in WO 03/086280 include nucleic acid molecules containing one or more copies of GUU, GUG, GGU, GGG, UGG, UGU, UUG, UUU, UUGUGG, UGGUUG, GUGUGU, and GGGUUU.


In none of the studies published to date efforts were undertaken to distinguish potentially distinct sequence requirements for TLR7 and TLR8. However, small molecules (nucleoside analogues) have been proposed that specifically activate TLR7 or TLR8 [15], supporting the idea that TLR7 and TLR8 may indeed have preferences for distinct RNA motifs.


It is an object of the present invention to identify RNA oligonucleotide motifs for stimulating an immune response, in particular, IL-12 induction. It is also an object of the present invention to identify ligands for activating TLR8. It is another object of the present invention to develop a method for determining the immunostimulatory activity, in particular, the IL-12-inducing activity, of a RNA oligonucleotide. It is yet another object of the present invention to develop a method for predicting the immunostimulatory activity, in particular, IL-12-inducing activity, of a RNA oligonucleotide. It is a further object of the invention to develop a method for designing and preparing RNA oligonucleotide having or lacking immunostimulatory activity, in particular, IL-12-inducing activity. It is also an object of the invention to provide RNA oligonucleotides having high immunostimulatory activity which can be used to induce an immune response, in particular, IL-12 production, in patients in need thereof. It is yet another object of the present invention to provide antisense RNA molecules that either have or lack immunostimulatory activity which can be used to treat disorders caused by the expression or overexpression of disease/disorder-related genes.


SUMMARY OF THE INVENTION

The present invention provides a method for determining the immunostimulatory activity of a RNA oligonucleotide, a method for predicting the immunostimulatory activity of a RNA oligonucleotide, a method for preparing a RNA oligonucleotide with high or low immunostimulatory activity, and a method for preparing an antisense RNA oligonucleotide with gene silencing activity and with high or low immunostimulatory activity.


The present application also provides an in vitro method for inducing IL-12 production from a mammalian cell.


The present invention further provides a single-stranded RNA oligonucleotide with immunostimulatory activity, an antisense RNA oligonucleotide with gene silencing activity and with high or low immunostimulatory activity, and the therapeutic uses thereof.


In addition, the present invention provides a pharmaceutical composition comprising one or more of the RNA oligonucleotides of the invention.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1: PBMC of three individual donors were isolated and stimulated with ssRNA-oligonucleotides 9.2sense (5′-AGCUUAACCUGUCCUUCAA-3′) and 9.2antisense (5′-UUGAAGGACAGGUUAAGCU-3′) that were complexed with either Lipofectamine, poly-L-arginine, poly-L-histidine or poly-L-lysine in duplicates. 24 hours after stimulation supernatants were harvested and IL-12p70 was assessed by ELISA. Data are presented as mean values ±SEM.



FIG. 2: PBMC of two different healthy donors were isolated and stimulated with poly-L-arginine complexed ssRNA-oligonucleotide 9.2sense (5′-AGCUUAACCUGUCCUUCAA) in duplicates. Poly-L-Arginine polymers of different sizes were used as indicated. 24 hours after stimulation supernatants were assessed for IL-12p70 production. Data were normalized to the poly-L-arginine polypetide with an approximate number of 88 amino acids (absolute values were 1390 and 396 pg/ml for donor 1 and donor 2). Data from two different donors were summarized and are presented as mean values ±SEM.



FIG. 3: PBMC from two different healthy donors were isolated and stimulated with poly-L-arginine (˜88 AS) complexed ssRNA-oligonucleotide 9.2sense (5′-AGCUUAACCUGUCCUUCAA). Time of complexation was either 0 min, 5 min, 10 min or 20 min. 24 hours after stimulation supernatants were harvested and analyzed for IL-12p70 production. Data from two individual donors were normalized (0 min value set to 1) and depicted as mean values ±SEM.



FIG. 4: PCS-measurement



FIG. 5: PBMC from three different healthy donors were isolated and stimulated with the following stimuli (form left to right): medium, Poly I:C (10 pg/ml), LPS (1 μg/ml), 3M-001 (1 μM), 3M-007 (1 μM), R848 (5 pg/ml), 3M-002 (1 μM), CpG ODN2216 (3 μg/ml), (the following stimuli were all complexed using poly-L-arginine 88) medium, polyA ssRNA, poly U ssRNA, polyA/U dsRNA, polyc ssRNA, polyG ssRNA, poly C/G dsRNA, 9.1 sense ssRNA, 9.1antisene ssRNA, 9.1duplex dsRNA, 9.2sense ssRNA, 9.2 antisene ssRNA, 9.2duplex dsRNA, 9.3sense ssRNA, 9.3antisene ssRNA and 9.3duplex dsRNA. 24 hours after stimulation supernatants were harvested and analyzed for IL-12p70 (A) and TNF-a (B) production. Data from three individual donors were summarized and depicted as mean values ±SEM.



FIG. 6: PBMC from two different healthy donors were isolated. PDC and MDC were depleted from PBMC using MACS. PBMC were reconstituted with either PDC, MDC, MDC and PDC or nothing and subsequently stimulated with poly-L-arginine (˜88 AS) complexed ssRNA-oligonucleotide 9.2sense (5′-AGCUUAACCUGUCCUUCAA). 24 hours after stimulation supernatants were harvested and analyzed for IL-12p70 production (A) and IFN-a production (B). Data from two individual donors were summarized and are depicted as mean values ±SEM.



FIG. 7: A: PBMC were stimulated with either poly-L-arginine (˜88 AS) complexed ssRNA-oligonucleotide 9.2sense (5′-AGCUUAACCUGUCCUUCAA) or poly-L-arginine alone. Six hours after stimulation brefeldin A was added and after three more hours cells were harvested and intracellular IL-12 production was analyzed using FACS. Data from two individual donors are shown. In addition, PBMC from two different donors were isolated. CD14 positive cells were depleted via MACS. One fraction (CD14+) was reconstituted with CD14 positive cells, whereas one fraction was left untreated. Subsequently cells were stimulated with poly-L-arginine (˜88 AS) complexed ssRNA-oligonucleotide 9.2sense (5′-AGCUUAACCUGUCCUUCAA). 24 hours after stimulation supernatants were harvested and analyzed for IL-12p70 production. Data from two individual donors were summarized and are depicted as mean values ±SEM (B). Monocytes were isolated from PBMC via depletion using MACS. Monocytes were stimulated with poly-L-arginine (˜88 AS) complexed with either RNA-oligonucleotides polyA, polyU, polyA/U, polyc, polyG, polyC/G, 9.1sense, 9.1antisense, 9.1duplex, 9.2sense, 9.2antisense, 9.2duplex, 9.3sense, 9.3antisense, 9.3duplex (for a detailed listing see table 1). In addition LPS (1 μg/ml), polyI:C (10 μg/ml), R848 (5 μg/ml) were used to stimulate cells. 24 hours after stimulation supernatants were analyzed for IL-12p70 (C) and TNF-a (D) production. Data from two individual donors were summarized and are depicted as mean values ±SEM.



FIG. 8: PBMC or monocytes from three or two different healthy donors were isolated and stimulated with poly-L-arginine complexed ssRNA oligonucleotides (see table 1). Shortened versions of ssRNA oligonucleotides 9.2sense were tested: 9.2L ssRNA (16mer), 9.2R ssRNA (16mer), 9.2A ssRNA (12mer), 9.2B ssRNA (12mer). RNA oligonucleotides were either tested in their ssRNA conformation or after annealing with the corresponding antisense strand 9.2antisense. 24 hours after stimulation supernatants were analyzed for IL-12p70 production. Data were normalized to ssRNA 9.2sense and are depicted as mean values ±SEM for PBMC (A) and monocytes (B). In addition a panel of self complementary ssRNA oligonucleotides (see table 2) were tested on both PBMC (C) and monocytes (D): ssRNA oligonucleotides were complexed using poly-L-arginine and used to stimulate PBMC from three individual donors and monocytes from two individual donors. 24 hours after stimulation supernatants were analyzed for IL-12p70 production. Data were normalized to ssRNA M20 and are depicted as mean values ±SEM for PBMC (C) and Monocytes (D).



FIG. 9: PBMC of six different healthy donors were isolated and stimulated with poly-L-arginine complexed ssRNA oligonucleotides in duplicates. 44 hours after stimulation IL-12p40 production was assessed in supernatant via ELISA. For all tested ssRNA oligonucleotides (table 3), the mean values of the measured duplicates were normalized to the positive control ssRNA oligonucleotide 9.2sense (5′-AGCUUAACCUGUCCUUCAA) by dividing the mean value of tested oligonucleotide by the mean value of 9.2sense (=IL-12 index of a given oligonucleotide). Next, all individual IL-12 indices were adjusted to the mean value of all IL-12 indices by subtracting the mean value of all IL-12 indices from the individual IL-12 index of a given oligonucleotide (=adjusted IL-12 index). Data from six individual donors were summarized and were assorted in ascending order displaying the corresponding SEM (for a complete listing including numerical data see table 4).



FIG. 10: A statistical analysis was performed to assess a putative significant difference for the adjusted IL-12 indices of all top thirty ssRNA oligonucleotides. A two-tailed Student's t-test was employed to calculate the p-value off all possible ssRNA oligonucleotide combinations. A p-value >0.01 and <0.05 is depicted by a black box, whereas a p-value <0.01 is depicted as a grey box.



FIG. 11: For all possible 1 mer motifs (5′-X-3′), 2mer motifs (5′-XX-3′,5′-X*X-3′,5′-X**X-3′) or 3mer motifs (5′-XXX-3′,5′-XX*X-3′,5′-X*XX-3′) a mean IL-12 index was assigned by calculating a mean IL-12 index of all ssRNA oligonucleotides containing the corresponding motifs (=IL-12 score of a given motif). The IL-12 score of all possible motifs is depicted in FIG. 11 ±SEM: 1mer motifs 5′-X-3′ (FIG. 11A); 2mer motifs 5′-XX-3′ (FIG. 11B1), 5′-X*X-3′ FIG. (11B2), 5′-X**X-3′ (FIG. 11B3) and 3mer motifs 5′-XXX-3′ (FIG. 11C1-11C4), 5′-XX*X-3′ (FIG. 11C5-11C8), 5′-X*XX-3′ (FIG. 11C9-11C12). A two-tailed Student's t-test for unpaired samples was used to analyze a putative significant difference between the respective motifs. A p-value >0.01 and <0.05 is marked by a “*”, whereas a p-value <0.01 is marked by a “**”.



FIG. 12: A calculated IL-12 index was assigned to each oligonucleotide by using the obtained motif-IL-12 scores. For each set of motifs [1mer motifs (5′-X-3′), 2mer motifs (5′-XX-3′,5′-X*X-3′,5′-X**X-3′) or 3mer motifs (5′-XXX-3′,5′-XX*X-3′,5′-X*XX-3′] a predicted IL-12 index was calculated for each ssRNA oligonucleotide. Next, the obtained predicted IL-12 indices were compared to the actual adjusted IL-12 indices. Data are depicted the following way: For all ssRNA oligonucleotides the predicted IL-12 indices are shown as a black bars, whereas data are sorted in ascending order according to the actual IL-12 score that is depicted as a red index line. The y-axis on the left side depicts the scale for the predicted IL-12 score, while the y-axis on the right side depicts the scale for the actual IL-12 score.





DETAILED DESCRIPTION OF THE INVENTION
Definitions

As used herein, “a” and “an” refers to a group or species of entities, rather than one single individual.


Oligonucleotide

As used herein, the term “oligonucleotide” refers to a polynucleotide formed from a plurality of linked nucleoside units. Such oligonucleotides can be obtained from existing nucleic acid sources, including genomic or cDNA, but are preferably produced by synthetic methods including chemical synthesis, in vitro and in vivo transcription. In preferred embodiments each nucleoside unit includes a heterocyclic base and a pentofuranosyl, trehalose, arabinose, 2′-deoxy-2′-substituted arabinose, 2′-O-substituted arabinose or hexose sugar group. The nucleoside residues can be coupled to each other by any of the numerous known internucleoside linkages. Such internucleoside linkages include, without limitation, phosphodiester, phosphorothioate, phosphorodithioate, pyrophosphate, alkylphosphonate, alkylphosphonothioate, phosphotriester, phosphoramidate, siloxane, carbonate, carboalkoxy, acetamidate, carbamate, morpholino, borano, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphorothioate, and sulfone internucleoside linkages. The term “oligonucleotide” also encompasses polynucleosides having one or more stereospecific internucleoside linkage (e.g., (Rp)— or (Sp)-phosphorothioate, alkylphosphonate, or phosphotriester linkages).


The oligonucleotides of the invention can include naturally occurring nucleosides, modified nucleosides, or mixtures thereof. As used herein, the term “modified nucleoside” is a nucleoside that includes a modified heterocyclic base, a modified sugar moiety, or a combination thereof. In some embodiments, the modified nucleoside is a non-natural pyrimidine or purine nucleoside, as herein described. In some embodiments, the modified nucleoside is a 2′-substituted ribonucleoside an arabinonucleoside or a 2′-deoxy-2′-substituted-arabinoside.


As used herein, the term “2′-substituted ribonucleoside” or “2′-substituted arabinoside” includes ribonucleosides or arabinonucleoside in which the hydroxyl group at the 2′ position of the pentose moiety is substituted to produce a 2′-substituted or 2′-O-substituted ribonucleoside. Preferably, such substitution is with a lower alkyl group containing 1-6 saturated or unsaturated carbon atoms, or with an aryl group having 6-10 carbon atoms, wherein such alkyl, or aryl group may be unsubstituted or may be substituted, e.g., with halo, hydroxy, trifluoromethyl, cyano, nitro, acyl, acyloxy, alkoxy, carboxyl, carboalkoxy, or amino groups. Examples of 2′-O-substituted ribonucleosides or 2′-O-substituted-arabinosides include, without limitation 2′-O-methylribonucleosides or 2′-O-methylarabinosides and 2′-O-methoxyethylribonucleosides or 2′-O-methoxyethylarabinosides.


The term “2′-substituted ribonucleoside” or “2′-substituted arabinoside” also includes ribonucleosides or arabinonucleosides in which the 2′-hydroxyl group is replaced with a lower alkyl group containing 1-6 saturated or unsaturated carbon atoms, or with an amino or halo group. Examples of such 2′-substituted ribonucleosides or 2′-substituted arabinosides include, without limitation, 2′-amino, 2′-fluoro, 2′-allyl, and 2′-propargyl ribonucleosides or arabinosides.


The term “oligonucleotide” includes hybrid and chimeric oligonucleotides. A “chimeric oligonucleotide” is an oligonucleotide having more than one type of internucleoside linkage. One preferred example of such a chimeric oligonucleotide is a chimeric oligonucleotide comprising a phosphorothioate, phosphodiester or phosphorodithioate region and non-ionic linkages such as alkylphosphonate or alkylphosphonothioate linkages (see e.g., Pederson et al. U.S. Pat. Nos. 5,635,377 and 5,366,878).


A “hybrid oligonucleotide” is an oligonucleotide having more than one type of nucleoside. One preferred example of such a hybrid oligonucleotide comprises a ribonucleotide or 2′-substituted ribonucleotide region, and a deoxyribonucleotide region (see, e.g., Metelev and Agrawal, U.S. Pat. Nos. 5,652,355, 6,346,614 and 6,143,881).


RNA oligonucleotides discussed herein include otherwise unmodified RNA as well as RNA which have been modified (e.g., to improve efficacy), and polymers of nucleoside surrogates. Unmodified RNA refers to a molecule in which the components of the nucleic acid, namely sugars, bases, and phosphate moieties, are the same or essentially the same as that which occur in nature, preferably as occur naturally in the human body. The art has referred to rare or unusual, but naturally occurring, RNAs as modified RNAs, see, e.g., Limbach et al. 1994, Nucleic Acids Res 22: 2183-2196. Such rare or unusual RNAs, often termed modified RNAs (apparently because these are typically the result of a post-transcriptional modification) are within the term unmodified RNA, as used herein. Modified RNA as used herein refers to a molecule in which one or more of the components of the nucleic acid, namely sugars, bases, and phosphate moieties, are different from that which occurs in nature, preferably different from that which occurs in the human body. While they are referred to as modified “RNAs,” they will of course, because of the modification, include molecules which are not RNAs. Nucleoside surrogates are molecules in which the ribophosphate backbone is replaced with a non-ribophosphate construct that allows the bases to the presented in the correct spatial relationship such that hybridization is substantially similar to what is seen with a ribophosphate backbone, e.g., non-charged mimics of the ribophosphate backbone.


All nucleic acid sequences listed herein are in the 5′ to 3′ direction unless otherwise indicated.


The RNA oligonucleotide of the invention is single-stranded. Furthermore, the ssRNA oligonucleotide of the invention does not contain any palindromic or otherwise self-complementary sequences which allow for the formation of a hairpin (or stem-loop) secondary structure with double-stranded characteristics.


The length of a ssRNA oligonucleotide is the number of nucleotides it contains. A ssRNA oligonucleotide containing n nucleotides can also be called a “n-mer” oligonucleotide.


Enhanced Nuclease Resistance

For increased nuclease resistance and/or binding affinity to the target, an oligonucleotide can include, for example, 2′-modified ribose units and/or phosphorothioate linkage(s) and/or pyrophosphate linkage(s). For example, the 2/hydroxyl group (OH) can be modified or replaced with a number of different “oxy” or “deoxy” substituents.


Examples of “oxy”-2′ hydroxyl group modifications include alkoxy or aryloxy (OR, e.g., R═H, alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar); polyethyleneglycols (PEG), O(CH2CH2O)nCH2CH2OR; “locked” nucleic acids (LNA) in which the 2′ hydroxyl is connected, e.g., by a methylene bridge, to the 4′ carbon of the same ribose sugar; O-AMINE and aminoalkoxy, O(CH2)nAMINE, (e.g., AMINE=NH2; alkylamino, dialkylamino, heterocyclyl amino, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino). It is noteworthy that oligonucleotides containing only the methoxyethyl group (MOE), (OCH2CH2OCH3, a PEG derivative), exhibit nuclease stabilities comparable to those modified with the robust phosphorothioate modification. “Deoxy” modifications include hydrogen (i.e. deoxyribose sugars, which are of particular relevance to the overhang portions of partially ds RNA); halo (e.g., fluoro); amino (e.g. NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); NH(CH2CH2NH)nCH2CH2-AMINE (AMINE=NH2; alkylamino, dialkylamino, heterocyclyl amino, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino), —NHC(O)R(R=alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), cyano; mercapto; alkyl-thio-alkyl; thioalkoxy; and alkyl, cycloalkyl, aryl, alkenyl and alkynyl, which may be optionally substituted with e.g., an amino functionality.


Preferred substitutents are 2∝-methoxyethyl, 2′-OCH3,2′-O-allyl, 2′-C-allyl, and 2′-fluoro. To maximize nuclease resistance, the 2′ modifications can be used in combination with one or more phosphate linker modifications (e.g., phosphorothioate). The so-called “chimeric” oligonucleotides are those that contain two or more different modifications. The inclusion of furanose sugars in the oligonucleotide backbone can also decrease endonucleolytic cleavage. An oligonucleotide agent can be further modified by including a 3′ cationic group, or by inverting the nucleoside at the 3′-terminus with a 3′-3′ linkage. In another alternative, the 3′-terminus can be blocked with an aminoalkyl group, e.g., a 3′ C5-aminoalkyl dT. Other 3′ conjugates can inhibit 3′-5′ exonucleolytic cleavage. While not being bound by theory, a 3′ conjugate, such as naproxen or ibuprofen, may inhibit exonucleolytic cleavage by sterically blocking the exonuclease from binding to the 3′-end of oligonucleotide. Even small alkyl chains, aryl groups, or heterocyclic conjugates or modified sugars (D-ribose, deoxyribose, glucose etc.) can block 3′-5′-exonucleases.


Similarly, 5′ conjugates can inhibit 5′-3′ exonucleolytic cleavage. While not being bound by theory, a 5′ conjugate, such as naproxen or ibuprofen, may inhibit exonucleolytic cleavage by sterically blocking the exonuclease from binding to the 5′-end of oligonucleotide. Even small alkyl chains, aryl groups, or heterocyclic conjugates or modified sugars (D-ribose, deoxyribose, glucose etc.) can block 3′-5′-exonucleases.


Single-stranded RNA oligonucleotides which contain self-complementary sequences and form a hairpin structure have enhanced nuclease resistance compared to single-stranded oligonucleotides which do not.


5-Phosphate Modifications

The oligonucleotides of the present invention can be 5′ phosphorylated or can include a phosphoryl analog at the 5′ prime terminus. 5′-phosphate modifications of the antisense strand include those which are compatible with RISC mediated gene silencing. Suitable modifications include: 5′-monophosphate ((HO)2(O)P—O-5′); 5′-diphosphate ((HO)2(O)P—O—P(HO)(O)—O-5′); 5′-triphosphate ((HO)2(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure. Other suitable 5′-phosphate modifications will be known to the skilled person.


Tethered Ligands

The RNA oligonucleotides of the present invention also include those with tethered ligands. The properties of a RNA oligonucleotide, including its pharmacological properties, can be influenced and tailored by the introduction of ligands, e.g. tethered ligands.


The ligands may be coupled, preferably covalently, either directly or indirectly via an intervening tether, to the RNA oligonucleotide. In preferred embodiments, the ligand is attached to the oligonucleotide via an intervening tether.


In preferred embodiments, a ligand alters the distribution, targeting or lifetime of a RNA oligonucleotide into which it is incorporated. In preferred embodiments a ligand provides an enhanced affinity for a selected target, e.g., molecule, cell or cell type, a cellular or organ compartment, tissue, organ or region of the body.


Preferred ligands can improve transport, hybridization, and specificity properties and may also improve nuclease resistance of the resultant natural or modified oligoribonucleotide, or a polymeric molecule comprising any combination of monomers described herein and/or natural or modified ribonucleotides.


A wide variety of ligands may be used. Ligands may include agents that allow for the specific targeting of the oligonucleotide; diagnostic compounds or reporter groups which allow for the monitoring of oligonucletotide distribution; cross-linking agents; nuclease-resistance conferring moieties; and natural or unusual nucleobases. General examples include lipophilic moleculeses, lipids, lectins, steroids (e.g., uvaol, hecigenin, diosgenin), terpenes (e.g., triterpenes, e.g., sarsasapogenin, Friedelin, epifriedelanol derivatized lithocholic acid), vitamins, carbohydrates (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid), proteins, protein binding agents, integrin targeting molecules, polycationics, peptides, polyamines, and peptide mimics.


The ligand may be a naturally occurring or recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid. Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacrylic acid), N-isopropylacrylamide polymers, or polyphosphazine. Example of polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic moieties, e.g., cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.


Ligands can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a thyrotropin, melanotropin, surfactant protein A, Mucin carbohydrate, a glycosylated polyaminoacid, transferrin, bisphosphonate, polyglutamate, polyaspartate, or an RGD peptide or RGD peptide mimetic.


Ligands can be proteins, e.g., glycoproteins, lipoproteins, e.g. low density lipoprotein (LDL), or albumins, e.g. human serum albumin (HSA), or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell. Ligands may also include hormones and hormone receptors. They can also include non-peptidic species, such as cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine, multivalent mannose, or multivalent fucose. The ligand can be, for example, a lipopolysaccharide, an activator of p38 MAP kinase, or an activator of NF-κB. The ligand can be a substance, e.g., a drug, which can increase the uptake of the oligonucleotide agent into the cell, for example, by disrupting the cell's cytoskeleton, e.g., by disrupting the cell's microtubules, microfilaments, and/or intermediate filaments. The drug can be, for example, taxon, vincristine, vinblastine, cytochalasin, nocodazole, japlakinolide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.


In one embodiment, the ligand is a lipid or lipid-based molecule. Such a lipid or lipid-based molecule preferably binds a serum protein, e.g., human serum albumin (HSA). An HSA binding ligand allows for distribution of the conjugate to a target tissue, e.g., liver tissue, including parenchymal cells of the liver. Other molecules that can bind HSA can also be used as ligands. For example, neproxin or aspirin can be used. A lipid or lipid-based ligand can (a) increase resistance to degradation of the conjugate, (b) increase targeting or transport into a target cell or cell membrane, and/or (c) can be used to adjust binding to a serum protein, e.g., HSA.


A lipid based ligand can be used to modulate, e.g., control the binding of the conjugate to a target tissue. For example, a lipid or lipid-based ligand that binds to HSA more strongly will be less likely to be targeted to the kidney and therefore less likely to be cleared from the body. A lipid or lipid-based ligand that binds to HSA less strongly can be used to target the conjugate to the kidney.


In another embodiment, the ligand is a moiety, e.g., a vitamin or nutrient, which is taken up by a target cell, e.g., a proliferating cell. These are particularly useful for treating disorders characterized by unwanted cell proliferation, e.g., of the malignant or non-malignant type, e.g., cancer cells. Exemplary vitamins include vitamin A, E, and K. Other exemplary vitamins include the B vitamins, e.g., folic acid, B12, riboflavin, biotin, pyridoxal or other vitamins or nutrients taken up by cancer cells.


In another embodiment, the ligand is a cell-permeation agent, preferably a helical cell-permeation agent. Preferably, the agent is amphipathic. An exemplary agent is a peptide such as tat or antennapedia. If the agent is a peptide, it can be modified, including a peptidylmimetic, invertomers, non-peptide or pseudo-peptide linkages, and use of D-amino acids. The helical agent is preferably an alpha-helical agent, which preferably has a lipophilic and a lipophobic phase.


In a preferred embodiment, the ligand is an antibody or a fragment thereof which is specific for a moiety present in a cell to be targeted. The moiety may be a protein, a carbohydrate structure, a polynucleotide, or a combination thereof. The moiety may be secreted, associated with the plasma membrane (e.g., on the extracellular or intracellular surface), cytosolic, associated with intracellular organelles (e.g., ER, Golgi complex, mitochondria, endosome, lysosome, secretory vesicle) or nuclear. The antibody may be monoclonal or polyclonal. The antibody may be chemeric or humanized. The antibody may be a single chain antibody. The antibody fragment may be a Fab fragment, a F(ab′)2 fragment, or any fragments that retain the antigen-binding specificity of the intact antibody.


Immunostimulatory Activity

As used herein, “immunostimulatory activity” refers to the capability of a molecule or a composition to induce an immune response. In one aspect, the immunostimulatory activity refers to the type I-IFN-inducing activity, in particular, the IFN-α-inducing activity.


As used herein, “inducing an immune response” means initiating or causing an increase in one or more of B-cell activation, T-cell activation, natural killer cell activation, activation of antigen presenting cells (e.g., B cells, dendritic cells, monocytes and macrophages), cytokine production, chemokine production, specific cell surface marker expression, in particular, expression of co-stimulatory molecules. In one aspect, such an immune response involves the production of IL-12, in cells such as monocytes and myeloid dendritic cells (MDC).


As used herein, “IL-12-inducing activity” refers to the capability of a molecule or composition to induce IL-12 production from a cell capable of producing IL-12. Cells capable of producing IL-12 include, but are not limited to, activated B cells, activated T cells, myeloid dendritic cells, monocytes, macrophages, and various cell lines (e.g., B cell lines; monocyte cell lines; cells transfected with expression vectors for TLR-8 such as CHO cells, COS cells, HEK293 cells). Cells capable of producing IL-12 include those that express TLR8.


Gene Silencing Activity

As used herein, “gene silencing” refers to the downregulation or the abolition of the expression of a target gene. Gene silencing as used herein, occurs at the post-transcriptional level. Gene silencing may be directly or indirectly mediated by siRNA, shRNA and antisense RNA.


Both the antisense-strand of the siRNA and the antisense RNA have complementary to the target mRNA and are the effector strand of the gene silencing activity. The term complementary is well understood by those skilled in the art. For example, A is complementary to T, G is complementary to C, 5′-AG-3′ is complementary to 5′-CT-3′.


The degree of complementarity between two oligonucleotides is the percentage of complementary bases in the overlapping region of the two oligonucleotides. The degree of complementarily can be determined manually or automatically by various engines such as BLAST. For example, ATCG has 100% complementarity to CGAT and CGATGG, and 75% complementarity to CGTT and CGTTGG. Furthermore, the degree of complementarity between a RNA oligonucleotide and any sequences present in the public databases (e.g., EMBL, GeneBank) can be determined by the BLAST program.


The degree of complementarity between the antisense strand of the siRNA or the antisense RNA and the target mRNA is at least 80% 81%, 82%, 83%, preferably at least 84%, 85%, 86%, 87%, 88%, more preferably at least 89%, 90%, 91%, 92%, 93%, even more preferably at least 94%, 95%, 96%, 97%, 98%, 99%, and most preferably 100%.


The gene silencing activity of a RNA oligonucleotide can be determined experimentally by methods well known in the art. For Example, the RNA oligonucleotide may be introduced into a cell by a method known in the art such as transfection and transduction; the mRNA level of the target gene can be determined by routine methods such as Northern blot analysis, quantitative PCR, RNase protection assay, and branching DNA; and the protein expression level can be determined by routine methods such as Western blotting, ELISA, and biological activity assays specific to the target protein. Furthermore, the mRNA level of all known and hypothetical genes can be determined at the global level using the microarray technology. Technologies in the field of proteonomics allow for the protein levels of a large number of genes to be determined at the global level as well.


Naked RNA oligonceotide may be transfected into a cell via electroporation. RNA oligonucleotide may be complexed with a complexation agent which facilitates the uptake of the oligonucletide into a cell. Such complexation agents include, but are not limited to cationic lipids (e.g., Lipofectamine, Oligofectamine, DOTAP), cationic peptides, and calcium phosphate.


Antisense RNA

As used herein, “antisense RNA” has the same definition as that established in the art. Antisense RNA is complementary to target mRNA and it thought to interfere with the translation of the target mRNA. Antisense RNA molecules are usually 18-50 nucleotides in length. Antisense RNA may be modified to have enhanced stability, nuclease resistance, target specificity and improved pharmacological properties.


Antisense RNA can be chemically synthesized, produced by in vitro transcription from linear (e.g. PCR products) or circular templates (e.g., viral or non-viral vectors), or produced by in vivo transcription from viral or non-viral vectors.


Disorder/Disease-Related Gene and Antigen

As used herein, “disorder/disease-related gene” refers to a gene that is expressed or overexpressed in a disease/disorder and that is not expressed or expressed in reduced amount under normal condition. For example, a mutant CF gene is expressed in cystic fibrosis patient but not in an individual without cystic fibrosis; ErbB2 (or Her2) is overexpressed in breast cancer cells compared to normal breast cells; a viral gene is expressed in infected cells but not in uninfected cells. The gene product of the disorder/disease-related gene is referred to herein as the “disorder/disease-related antigen”.


Mammal

As used herein, the term “mammal” includes, without limitation, rats, mice, cats, dogs, horses, sheep, cattle, cows, pigs, rabbits, non-human primates, and humans.


Technology Platform

IL-12 and IFN-α are the two key cytokines to drive strong Th1 responses desired for effective immunotherapy of viral infection and cancer. While plasmacytoid dendritic cells (PDC) are responsible for the production of IFN-α, the major source for IL-12 are myeloid cells including monocytes, macrophages and myeloid dendritic cells. In several recent studies, RNA molecules were reported to induce IL-12 production in human immune cells, but the RNA sequence motif and the cellular subset responsible for IL-12 production has not been defined in these studies. Here we used a systematic approach to identify potent RNA motifs for IL-12 induction in TLR8-expressing cells.


We made use of the distinct expression pattern of TLR7 and TLR8 in human PDC (TLR7+TLR8−) and human monocytes (TLR7−TLR8+) [8]. A technology platform was developed that allowed screening of RNA oligonucleotides on a large scale. We designed a RNA oligonucleotide library that contained all possible 4-nucleotide (4-mer) motifs. By applying this library to our experimental system we were able to identify potent sequence motifs for inducing IL-12 production in human monocytes.


Our systematic approach (i.e., technological platform) is based on the following key features: i) the use of poly-L-Arg for complexation and transfection of RNA oligonucleotides; ii) identification of monocytes as the major source for IL-12 within PBMC and analysis of IL-12 production in supernatant of PBMC culture; iii) the generation of a 4mer library in the center of poly adenosine (poly A) RNA oligonucleotides; iv) the development of algorithms that use the 4mer library data matrix for prediction of IL-12-inducing activity of a RNA oligonucleotide. Our technical platform addresses the following key issues that are essential for successful analysis of IL-12-inducing activity of RNA oligonucleotides in PBMC:


The first key issue is the type of RNA transfection in PBMC. In the literature, cationic lipids such as lipofectamine or DOTAP are routinely used for the transfection of RNA oligonucleotides. However, RNA-cationic lipid complexes lead to rapid cell death especially of myeloid cells. Since myeloid cells represent the cellullar source of IL-12 within PBMC (the source of IFN-α are PDC), cationic lipids are not useful in this setting. Searching for alternative techniques we compared the use of the cationic lipid lipofectamine with different types of cationic peptides (poly-His, poly-L-Lys, poly-L-Arg) for complexation of RNA. Only poly-L-Arg, but none of the other cationic polymers, potently supported the IL-12-inducing activity of the RNA oligonucleotides. By changing the length of the poly cationic peptide and the incubation time (i.e., the length of complex formation), a protocol could be established that allowed well-controlled and highly reproducible complex formation. Complex formation was validated by monitoring the size of complexes and the functional activity over a range of concentrations. It was found that poly-L-Arg which contain at least 24 amino acids are effective in introducing a RNA oligonucleotides into myeloid cells. Furthermore, complex formation between a RNA oligonucleotide and poly-L-Arg is achieved after 20 min of incubation. This type of RNA transfection did not affect the viability of myeloid cells and thus could be applied to PBMC without restrictions.


The second key issue of our technological platform was the in vitro system for testing the biological activity of RNA oligonucleotides. Primary monocytes were identified as the cellular source of IL-12 production within PBMC and where found to be stimulated by RNA oligonucleotides directly. Since primary monocytes exclusively express TLR8 but not TLR3, TLR7 or TLR9, a monocyte-based motif search is focused on motifs for TLR8. The use of primary immune cells was preferred over the use of TLR-transfected cell lines for several reasons: a) cell lines such as 293 cells may lack yet undefined adaptor proteins for oligonucleotide recognition by TLR or may lack downstream signaling molecules [11, 16]; b) reporter assays may not reflect the correct type of activity; c) internalization and intracellular distribution of oligonucleotides in cell lines may differ from primary immune cells. Therefore, the value of such cell line-based systems for screening the activity of RNA oligonucleotides is limited.


The third key issue of our technology platform was the generation of an oligonucleotide library. We demonstrate that a minimal length of 19 bases was required for optimal IL-12-inducing activity of RNA oligonucleotides; furthermore, we show that poly adenosine RNA oligonucleotides were completely inactive in terms of IL-12 induction in PBMC. Therefore, the search for the optimal motif for IL-12 induction was performed with a 19mer oligonucleotide on a poly adenosine sequence background. By adding increasing numbers of uridine (U) in the center of such an oligonucleotide we found that a 4mer motif in the center is sufficient to confer marked immunostimulatory activity (data not shown). The library of 193 RNA oligonucleotides used covered all 256 possible 4mer motifs. The reduction from 256 to 193 was possible because of redundant motifs caused by the poly adenosine flanking regions.


The fourth key feature of the technology platform was the generation of a data matrix and its mathematical analysis. Algorithms were developed that allowed the prediction of the IL-12 inducing activity of RNA oligonucleotides. The frequency of a given 4mer motif at a certain site within an oligonucleotide is only 1:256. Although the identification of the most active 4mer motifs can be used to generate potent immunostimulatory oligonucleotides, based on their low frequency of 1:256, the IL-12 indices of the 4mer motifs are less useful to predict the activity of a given oligonucleotide. Therefore, algorithms were established that are based on parts of the 4mer motif, namely 1, 2 or 3 bases either in a row (XXX) or with spacing (X*XX; XX*X). The highest predictive value was obtained by combining the information based on 3 bases (XXX, X*XX, XX*X). This algorithm described in the results section allowed a valuable prediction (r=0.78) of the IL-12 inducing activity of the 4mer motif library.


There is a number of applications for the information generated by using the technology platform: a) the 4mer motif data matrix can be used to design oligonucleotides with high IL-12-inducing activity; b) 4mer motifs with minimal/low IL-12-inducing activity can be used as preselection of potential inhibitory sequence motifs; c) the 3mer-based algorithm can be used to predict the IL-12-inducing activity of a given single-stranded RNA oligonucleotide; d) the 3mer-based algorithm can be used to maximize/optimize the immunostimulatory activity of single-stranded RNA oligonucleotides with other sequence requirements (such as antisense activity); e) use of 3mer-based algorithm to minimize (avoid) the IL-12-inducing activity of single-stranded RNA oligonucleotides with other sequence requirements (such as antisense activity).


In general, two major strategies for the therapeutic development of RNA oligonucleotides can be distinguished: i) gene silencing employing siRNA, shRNA or antisense RNA, and ii) inducing an immune response using immunostimulatory RNA (is RNA).


As shown in an earlier study of ours [10], double-stranded RNA oligonucleotides (including siRNA) are completely inactive in inducing IL-12, although they can be potent inducers of IFN-α. Therefore, only the avoidance of IFN-α-inducing activity is relevant for siRNA design as detailed in our co-pending application. On the other hand, in the preparation of IL-12-inducing single-stranded RNA oligonucleotides, the formation of double-stranded secondary structure via self-complementary sequence (including palindromic sequence) is to be avoided. Indeed, we show in our study that a single-strand 19mer RNA oligonucleotide lost its activity to induce IL-12 when a 12mer or a 16mer complementary strand is attached; furthermore, a palindromic RNA oligonucleotide was inactive in inducing IL-12. This aspect can be mathematically integrated in the algorithm used for the prediction and the design of IL-12-inducing oligonucleotides. On the other hand, the incorporation of a palindromic sequence into an immunostimulatory oligonucleotide can be used to direct the activity of the RNA oligonucleotide to the IFN-α-inducing activity.


It is important to note that a poly adenosine RNA oligonucleotide containing the best 4mer motif (UCGU) is 5-fold more active in inducing IL-12 than one of the most active immunostimulatory oligonucleotides in the literature, RNA9.2sense ([10]). This motif was found to be weak at inducing IFN-α in PDC. On the other hand, the most potent 4mer motif for IFN-α induction identified in our co-pending application, GUUC, shows only weak IL-12-inducing activity. The existence of two distinct optimal motifs for IFN-α and IL-12 induction together with the selective presence of TLR7 (but not TLR8) in PDC and TLR8 (but not TLR7) in monocytes strongly support the concept that two distinct receptors, TLR7 and TLR8, are responsible for IFN-α and IL-12 production, respectively, and that the two receptors have different ligand preference. However, we were able to identified 4mer motifs that are relatively potent at inducing both IFN-α and IL-12 production, suggesting that there are some common structural features that are recognized by both TLR7 and TLR8.


Of note, in a 19mer oligonucleotide, these two types of sequence motifs can be combined for eliciting optimal IFN-α and IL-12 responses at the same time.


The 4mer motifs that are more potent at inducing IFN-α than IL-12 include GCUC, GUCA, GUUC, GGUC, GUCC, GUCU, GUUU, CGUC, GCUU, GUGU.


The 4mer motifs that are more potent at inducing IL-12 than IFN-α include UCGU, GAUA, UGGC, UGCU, UGGU, UGCC, UUGC, UGAC, UAAU, UUAU.


The 4mer motifs that are potent at inducing both IL-12 than IFN-α include GUUG, GGUU, UUGU, GGUA, CUGU, UGUC, UGUA, UGUU, UGUG, UAGU.


Furthermore, one can hypothesize that distinct inhibitory sequence motifs exist for TLR7 and TLR8, and that by combining inhibitory motifs for TLR7 and active motifs for TLR8 or vice versa, the activity of a RNA oligonucleotide can be directed to exclusive TLR7 or TLR8 activity. Combining inhibitory motifs for TLR7 and TLR8 would lead to minimized immunostimulatory activity of a RNA oligonucleotide, which may be desired in the preparation of certain gene silencing oligonucleotides.


Distinct sequence motifs for TLR7 and TLR8 activation have not been previously reported. To date, there are only two publications describing the identification of specific sequence motifs responsible for the immunological activity of RNA [10, 17]. However, in both publications, the sequence motif was identified based on the IFN-α- but not IL-12-inducing activity. In one publication, Judge and colleagues propose that a sequence motif, UGUGU, is responsible for the IFN-α-inducing activity of a RNA oligonucleotide [17]. In our own publication we identified a 9mer sequence motif to be responsible for the immunological activity of the ssRNA oligonucleotide RNA9.2 sense (5′-AGC UUA ACC UGU CCU UCA A-3′, 9mer motif underlined). Heil and colleagues did not claim a specific sequence motif but attributed the immunological activity of RNA oligonucleotides to a high content of G and U within the sequence [11]. Sioud and colleagues tested a large panel of RNA oligonucleotides for TNF-α and IL-6 production in PBMC, but only one sequence was tested for IFN-α induction [18]. In mice, two publications report IFN-α induction by RNA oligonucleotides [12, 19].


In the six studies described above, IL-12 was examined by Heil [11] in human PBMC and by Heil, by Diebold and by Barchet in murine dendritic cells [11, 12, 19] without disclosing any specific sequence motifs. In one other study, sequence-specific induction of IL-12 in primary monocytes by RNA oligonucleotide was reported [16]. In this study, the authors report that the induction of IL-12 by RNA oligonucleotides is CpG specific. Inversion of CG to GC within the sequence motif used (5′-GGUGCAUCGAUGCAGGGGGG-3′; CpG motif underlined, palindrome in bold) abolished the activity of the oligonucleotide. Furthermore, methylation of the C abrogated the activity. The sequence context was defined as an AU dinucleotide at the 5′ of the CG and a poly G sequence at the 3′ end of the RNA oligonucleotide. RNA oligonucleotides with a complete phosphorothioate modification for stabilization against nucleases did not require complexation or transfection; in contrast, unmodified RNA oligonucleotides were only active when transfected with cationic lipids. In addition to the phosphorothioate backbone, RNA oligonucleotides were protected by 2-bis(acetoxyethoxy)-methyl ether. Our own results do not confirm the strict CG dependence of this effect. The best single prediction formula of active motifs in our study is UXGU, in which the X is preferentially C, G or U. Even the motif UGCU (which in their study was an inactive sequence) is among the most active sequence motifs in our study. In contrast to their study, the presence of palindromic sequences in our hands abolish the IL-12 inducing activity. Furthermore, poly G is not necessary in our approach. Together, in their study [16] a number of additional factors (phosphorothioate backbone, palindromic sequence, protection by 2-bis(acetoxyethoxy)-methyl ether) seem to affect the immunological activity of their oligonucleotides in a way that the pure sequence specificity seems to be obscured and can obviously not be separated correctly from other influencing factors.


Several studies report that long single-stranded RNA such as mRNA is immunologically active. Koski and colleagues showed that RNA derived from bacterial but not eukaryotic sources, when transfected into human monocyte-derived dendritic cell precursors, induced IL-12 secretion, and that lack of activity of vertebrate mRNA depends on the presence of a poly(A) tail. Furthermore they report that in vitro-transcribed mRNA mimics the structure of bacterial mRNA in the lack of a long 3′-poly(A) tail [20]. Scheel and colleagues demonstrated that monocytes are activated by protamine-condensed mRNA (long single-stranded RNA) [21, 22].


Protamine used in the study of Scheel and colleagues for complexation of long single-stranded RNA [21, 22] is a long cationic protein. In the literature, also short cationic peptides were described to complex long nucleic acids resulting for example in enhanced delivery of plasmid DNA [23]. The cationic peptide poly-L-Arg is known to be effectively internalized into the endosomal compartment [24], which facilitates cellular delivery of oligonucleotides complexed to poly-L-Arg [25]. However, poly-L-Arg has never been described for the delivery of RNA oligonucleotides.


In conclusion, the use of poly-L-Arg for the delivery of RNA oligonucleotides to myeloid cells without inducing cell death for the first time allowed a valid search for RNA motif(s) recognized by TLR8-positive (and TLR7-negative) monocytes. The availability of distinct motifs for IL-12 and IFN-α (co-pending application) induction allows for the first time selective and/or combined induction of these two key Th1 cytokines in the human immune system, thereby filling the gap left by CpG DNA oligonucleotides which fail to induce IL-12 in human immune cells. Our results may have great impact on the design, the understanding and the clinical development of immunostimulatory RNA (is RNA).


Method for Determining the Immunostimulatory Activity of an RNA Oligonucleotide

The present invention provides a method for determining the immunostimulatory activity, in particular, the IL-12-inducing activity, of a RNA oligonucleotide, comprising the steps of:

    • (a) complexing the RNA oligonucleotide with a complexation agent;
    • (b) contacting a cell with the complexed RNA oligonucleotide, wherein the cell expresses TLR8; and
    • (c) determining the amount of IL-12 produced by the cell of step (b), an increase of IL-12 production indicating immunostimulatory activity of the RNA oligonucleotide.


In one embodiment of the invention, the complexation agent is a polycationic peptide, preferably poly-L-arginine (poly-L-Arg). In one embodiment, the polycationic peptide, in particular, poly-L-Arg, is at least 24 amino acids in length. The polycationic peptide, in particular, poly-L Arg, may be a heterogeneous mixture of peptides of different lengths.


The cells expressing TLR8 include, but are not limited to, peripheral blood mononuclear cells (PBMC), myeloid dendritic cells (MDC), macrophages, monocytes, B cells, and cells containing exogenous DNA which directs the expression of TLR8 such as transfected CHO, HEK293, and COS cells.


In one embodiment of the invention, the cell is a mammalian cell, preferably a human cell or a cell of human origin.


Method for Predicting the Immunostimulatory Activity of a RNA Oligonucleotide

The present invention provides a method for predicting the immunostimulatory activity, in particular, the IL-12-inducing activity, of a single-stranded RNA oligonucleotide, comprising the steps of:

    • (a) identifying all possible 3-nucleotide (3mer) motifs contained in the oligonucleotide
      • (i) for a 3mer motif which appears in Table 5, assing an IL-12 point score according to Table 5;
      • (ii) for a 3mer motif which does not appear in Table 5, assign an IL-12 point score of 0;
    • (b) assigning an IL-12 point score for each individual 3mer motif;
    • (c) assigning the sum of the IL-12 point scores of individual 3mer motifs as the IL-12 score of the oligonucleotide; and
    • (d) assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 93.53, an intermediate immunostimulatory activity if the IL-12 score is between −28 and 93.53, and a low immunostimulatory activity if the IL-12 score is at most −28, when n=6;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 107.77, an intermediate immunostimulatory activity if the IL-12 score is between −31 and 107.77, and a low immunostimulatory activity if the IL-12 score is at most −31, when n=7;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 121, an intermediate immunostimulatory activity if the IL-12 score is between −34 and 121, and a low immunostimulatory activity if the IL-12 score is at most −34, when n=8;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 121.98, an intermediate immunostimulatory activity if the IL-12 score is between −36 and 121.98, and a low immunostimulatory activity if the IL-12 score is at most −36, when n=9;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 8.4064×n+66.958, an intermediate immunostimulatory activity if the IL-12 score is between 0.0468×n2-2.3103×n −23.244 and 8.4064×n+66.958, and a low immunostimulatory activity if the IL-12 score is at most 0.0468×n2-2.3103×n −23.244, when n is greater than 9,
      • wherein n is the length of the oligonucleotide.


The present invention also provides a method of assigning an IL-12 score to a RNA oligonucleotide comprising steps (a)-(c) described above.


A single-stranded RNA oligonucleotide of the length n is broken up into all possible 3mer motifs of the following configuration. XXX, X*XX, XX*X. This will result in a total number of (3*n)−4 possible 3mer motifs.


For example, the 20mer ssRNA oligonucleotide 5′-AACGCCCGGCUCAUUACGUC-3′ can be broken up into the following 18 3mer motifs 5′-XXX-3′: AAC, ACG, CGC, GCC, CCC, CCG, CGG, GGC, GCU, CUC, UCA, CAU, AUU, UUA, UAC, ACG, CGU, GUC; the following 17 3mer motifs 5′-X*XX-3′: A*AG, A*CC, C*GC, G*CC, C*CG, C*CG, C*GC, G*GU, G*CC, C*UA, U*CU, C*AU, A*UA, U*UC, U*AG, A*CU, C*GC; and the following 17 3mer motifs 5′-XX-*X-3′: AC*G, AG*C, CC*C, GC*C, CC*G, CG*G, CG*C, GC*U, GU*C, CC*A, UA*U, CU*U, AU*A, UA*C, UC*G, AG*U, CU*C


Subsequently all of the obtained 3mer motifs are compared to the IL-12 point score matrix (Table 5).










TABLE 5







IL-12 point score matrix for 3mer motifs. * can



be any one of the nucleotides A, U, G and C.











Sequence
IL-12




5′→ 3′
Point score















AAC
−4








AAU
4







ACA
−4







ACC
−5







ACG
−4







AGA
−3







AGU
7







AUG
8







CAA
−2







CCA
−4







CGA
−4







CGU
6







GAA
−2







GGU
11







GUA
10







GUG
6







GUU
13







UAA
4







UGA
4







UGC
9







UGG
8







UGU
11







UUG
12







AA*C
−3







AA*U
4







AC*A
−4







AC*C
−5







AG*U
6







AU*G
9







AU*U
5







CA*A
−2







CC*A
−4







GU*A
8







GU*G
14







GU*U
9







UA*A
4







UC*U
13







UG*A
5







UG*C
13







UG*U
20







UU*A
4







A*AC
−4







A*AU
3







A*CA
−4







A*CC
−5







A*GU
7







A*UG
8







A*UU
4







C*AA
−2







C*CA
−5







C*GA
−5







G*UA
8







G*UG
11







G*UU
8







U*AA
3







U*AU
8







U*GC
11







U*GU
27







U*UA
5










Whenever a 3mer motif is present in the IL-12 point score matrix, the listed point score is added to the so-called predicted IL-12 score of the oligonucleotide analyzed. Whenever a 3mer motif is absent from the IL-12 point score matrix, the motif get s point score of 0. Therefore, the predicted IL-12 score of a given ssRNA oligonucleotide is the sum of IL-12 points scores of all 3mer motifs that are present in the IL-12 point score matrix.


For example, for the 20mer ssRNA oligonucleotide 5′-AACGCCCGGCUCAUUACGUC-3′ a predicted IL-12 score can be calculated as follows:














3mer motifs in the 20mer




ssRNA oligonucleotide
presence (✓) or absence (Ø)


5′-AACGCCCGGCUCA-
in the IL-12 point score
predicted IL-12


UUACGUC-3′
matrix
score

















AAC

−4


AAU
Ø
0


ACA
Ø
0


ACC
Ø
0


ACG

−8


AGA
Ø
0


AGU
Ø
0


AUG
Ø
0


CAA
Ø
0


CCA
Ø
0


CGA
Ø
0


CGU

6


GAA
Ø
0


GGU
Ø
0


GUA
Ø
0


GUG
Ø
0


GUU
Ø
0


UAA
Ø
0


UGA
Ø
0


UGC
Ø
0


UGG
Ø
0


UGU
Ø
0


UUG
Ø
0


AA*C
Ø
0


AA*U
Ø
0


AC*A
Ø
0


AC*C

−5


AG*U
Ø
0


AU*G
Ø
0


AU*U
Ø
0


CA*A
Ø
0


CC*A
Ø
0


GU*A
Ø
0


GU*G
Ø
0


GU*U
Ø
0


UA*A
Ø
0


UC*U

13


UG*A
Ø
0


UG*C
Ø
0


UG*U
Ø
0


UU*A
Ø
0


A*AC
Ø
0


A*AU
Ø
0


A*CA
Ø
0


A*CC
Ø
0


A*GU

7


A*UG
Ø
0


A*UU
Ø
0


C*AA
Ø
0


C*CA

−5


C*GA
Ø
0


G*UA
Ø
0


G*UG
Ø
0


G*UU
Ø
0


U*AA
Ø
0


U*AU

8


U*GC
Ø
0


U*GU
Ø
0


U*UA
Ø
0


overall

12









Method for Designing and Preparing RNA Oligonucleotides

The present application provides a method for preparing an ssRNA oligonucleotide having immunostimulatory activity, in particular, high IL-12-inducing activity, comprising the steps of:

    • (a) providing candidate oligonucleotide sequence(s);
    • (b) identifying oligonucleotide sequence(s) with high immunostimulatory activity predicted according to the method of prediction described in the previous section;
    • (c) preparing the RNA oligonucleotide(s) identified for high immunostimulatory activity in step (b); and
    • (d) optionally testing the immunostimulatory activity of the RNA oligonucleotide(s) prepared in step (c) according to the method of determination described previously; and
    • (e) further optionally modifying the oligonucleotide(s) to optimize the immunostimulatory activity.


The present application also provides a method for preparing an ssRNA oligonucleotide having low immunostimulatory activity, in particular, low IL-12-inducing activity, comprising the steps of:

    • (a) providing candidate oligonucleotide sequence(s);
    • (b) identifying oligonucleotide sequence(s) with low immunostimulatory activity predicted according to the method of prediction described in the previous section;
    • (c) preparing the RNA oligonucleotide(s) identified for low immunostimulatory activity in step (b); and
    • (d) optionally testing the RNA oligonucleotide(s) prepared in step (c) for the lack of immunostimulatory activity according to the method of determination described previously; and
    • (e) further optionally modifying the oligonucleotide(s) to minimize the immunostimulatory activity.


The present application further provides a method for preparing a single-stranded RNA oligonucleotide having high immunostimulatory activity, in particular, high IL-12-inducing activity, comprising the steps of:

    • (a) providing an oligonucleotide sequence which comprises at least one, preferably at least two, more preferably at least three, even more preferably at least four, of the 4-nucleotide (4-mer) motifs selected from the group consisting of:












UCGU (No. 1), GUUG (No. 2), UGGU (No. 3),








UGGC (No. 4), GGUA (No. 5), UGAU (No. 6),







UGCU (No. 7), UUGC (No. 8), UUGU (No. 9),







UAGU (No. 10), GGUU (No. 11), GUUU (No. 12),







UGUG (No. 13), GUGU (No. 14), UGCC (No. 15),







GUAU (No. 16), GUGC (No. 17), UGUA (No. 18),







UGUC (No. 19), CUGU (No. 20), UGAC (No. 21),







UGUU (No. 22), UAAU (No. 23), GUAG (No. 24),







UCUU (No. 25), UUGG (No. 26), UUUG (No. 27),







GGAU (No. 28), UUUU (No. 29), CGUU (No. 30),







UUAU (No. 31), GUUC (No. 32), GUGG (No. 33),







GGUG (No. 34), UAUU (No. 35), UCUG (No. 36),







GUAC (No. 37), UAGG (No. 38), UCUC (No. 39),







UAGC (No. 40), UAUC (No. 41), CUAU (No. 42),







UACU (No. 43), CGGU (No. 44), UGCG (No. 45),







UUUC (No. 46), UAUG (No. 47), UAAG (No. 48),







UACC (No. 49), UUAG (No. 50), GCUU (No. 51),







CAGU (No. 52), UGAG (No. 53), GAUU (No. 54),







GAGU (No. 55), GUUA (No. 56), UGCA (No. 57),







UUCU (No. 58), GCCU (No. 59), GGUC (No. 60),







GGCU (No. 61), UUAC (No. 62), UCAU (No. 63),







GCGU (No. 64), GCAU (No. 65), GAUG (No. 66),







GUCU (No. 67), CGUA (No. 68), CGAU (No. 69),








    • wherein the nucleotide sequences of the motifs are 5′→3′,

    • wherein the oligonucleotide is between 6 and 64, preferably between 12 and 50, more preferably between 14 and 40, even more preferably between 16 and 36, and most preferably between 18 and 30 nucleotides in length,

    • wherein the oligonucleotide has an IL-12 score of at least 93.53 when n=6; at least 107.77 when n=7; at least 121 when n=8; at least 121.98 when n=9; at least 8.4064×n+66.985 when n is greater than 9, wherein the IL-12 score is assigned according to the method described above,

    • wherein n is the length of the oligonucleotide;

    • (b) preparing the RNA oligonucleotide of step (a); and

    • (c) optionally testing the immunostimulatory activity of the RNA oligonucleotide prepared in step (b) according to the method of determination described previously; and

    • (d) further optionally modifying the oligonucleotide to optimize the immunostimulatory activity.





The ssRNA oligonucleotide can have other functionalities such as the gene silencing activity. The ssRNA oligonucleotide may be an antisense RNA.


The methods provided by the present application can be used to prepare immunostimulatory RNA oligonucleotides and antisense RNA with high or low immunostimulatory activity.


Some of the RNA oligonucleotides which have low immunostimulatory activity, i.e., the non-immunostimulatory oligonucleotides, may in fact have inhibitory activity against immune activation. Such an immunoinhibitory oligonucleotide may be able to prevent immune activation induced by an immunostimulatory oligonucleotide when used in combination.


RNA oligonucleotides can be prepared by methods including, but are not limited to, chemical synthesis, in vitro and in vivo transcription from linear templates (e.g., PCR product) and circular templates (e.g., viral or non-viral vectors).


Method for Preparing Antisense RNA Having High or Low Immunostimulatory Activity

The present invention provides a method for preparing an antisense RNA having gene silencing activity for a target gene and having immunostimulatory activity, in particular, IL-12-inducing activity, comprising the steps of:

    • (a) identifying all potential antisense sequences for a target mRNA;
    • (b) identifying antisense sequences that have gene silencing activity;
    • (c) predicting the immunostimulatory activity for the antisense sequences identified in step (b);
    • (d) identifying antisense RNA sequences which have an IL-12 score of at least 8.4064×n+66.958, wherein the IL-12 score is assigned according to the method described above, and wherein n is the length of the sequence and n is between 18 and 50;
    • (e) decreasing the IL-12 score threshold by 1 if no antisense sequence is identified in step (d), until at least one antisense sequence is identified;
    • (f) preparing the antisense RNA identified in step (d) or (e);
    • (g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense prepared in (f);
    • (h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing and/or immunostimulatory activity.


The present invention also provides an alternative method for preparing an antisense RNA with gene silencing activity and immunostimulatory activity, comprising the steps of:

    • (a) identifying all potential antisense sequences for a target mRNA;
    • (b) predicting the immunostimulatory activity for all of the potential antisense sequences identified in (a);
    • (c) identifying 10 potential antisense sequences with the highest IL-12 scores;
    • (d) identifying antisense sequences with gene silencing activity among the 10 potential antisense sequences identified in step (c);
    • (e) identifying 10 potential antisense sequences with the next highest IL-12 scores if no antisense sequence can be identified in step (d); repeat steps (d) and (e) until at least one antisense sequence is identified;
    • (f) preparing the antisense RNA identified in step (d) or (e);
    • (g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense RNA prepared in (f);
    • (h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing and/or immunostimulatory activity.


The present invention further provides a method for preparing an antisense RNA having gene silencing activity for a target gene and having low (or minimal) immunostimulatory activity, in particular, IL-12-inducing activity, comprising the steps of:

    • (a) identifying all potential antisense sequences for a target mRNA;
    • (b) identifying antisense sequences that have gene silencing activity;
    • (c) predicting the immunostimulatory activity for the antisense sequences identified in step (b);
    • (d) identifying antisense sequences which have an IL-12 score of at most 1.9763×n−30.643, wherein the IL-12 score is assigned according to the method described above, and wherein n is the length of the sequence and n is between 18 and 50;
    • (e) increasing the IL-12 score threshold by 1 if no antisense sequence is identified in step (b), until at least one antisense sequence is identified;
    • (f) preparing the antisense RNA identified in step (d) or (e);
    • (g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense RNA prepared in (f);
    • (h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing activity and/or to minimize the immunostimulatory activity.


The present invention also provide an alternative method for preparing an antisense RNA with gene silencing activity and low (or minimal) immunostimulatory activity, comprising the steps of:

    • (a) identifying all potential antisense sequences for a target mRNA;
    • (b) predicting the immunostimulatory activity for all of the potential antisense sequences identified in (a);
    • (c) identifying 10 potential antisense sequences with the lowest IL-12 scores;
    • (d) identifying antisense sequences with gene silencing activity among the 10 potential antisense sequences identified in step (c);
    • (e) identifying 10 potential antisense sequences with the next lowest IL-12 scores if no antisense sequence can be identified in step (d); repeat steps (d) and (e) until at least one antisense sequence is identified;
    • (f) preparing the antisense RNA identified in step (d) or (e);
    • (g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense RNA prepared in (f);
    • (h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing activity and/or to minimize the immunostimulatory activity.


Candidate (potential) antisense sequences with gene silencing activity for a given gene can be identified using methods known to those skilled in the art. Furthermore, the gene silencing activity of an antisense RNA may be determined experimentally.


The gene silencing activity of an antisense can be determined experimentally by methods well known in the art. For Example, the antisense RNA oligonucleotide may be introduced into a cell by a method known in the art such as transfection and transduction; the mRNA level of the target gene can be determined by routine methods such as Northern blot analysis, quantitative PCR, RNase protection assay, and branching DNA; and the protein expression level can be determined by routine methods such as Western blotting, ELISA, and biological activity assays specific to the target protein. Furthermore, the mRNA level of all known and hypothetical genes can be determined at the global level using the microarray technology. Technologies in the field of proteonomics allow for the protein levels of a large number of genes to be determined at the global level as well.


The antisense RNA can be prepared by methods including, but are not limited to, chemical synthesis, in vitro and in vivo transcription from PCR products and viral or non-viral vectors.


Immunostimulatory RNA Oligonucleotides

The present invention provides an immunostimulatory RNA oligonucleotide having immunostimulatory activity, in particular, IL-12-inducing activity, comprising at least one, preferably at least two, more preferably at least three, even more preferably at least four, even more preferably at least five, and most preferably at least six, of the 4-nucleotide (4-mer) motifs selected from the group consisting of:












UCGU (No. 1), GUUG (No. 2), UGGU (No. 3),








UGGC (No. 4), GGUA (No. 5), UGAU (No. 6),







UGCU (No. 7), UUGC (No. 8), UUGU (No. 9),







UAGU (No. 10), GGUU (No. 11), GUUU (No. 12),







UGUG (No. 13), GUGU (No. 14), UGCC (No. 15),







GUAU (No. 16), GUGC (No. 17), UGUA (No. 18),







UGUC (No. 19), CUGU (No. 20), UGAC (No. 21),







UGUU (No. 22), UAAU (No. 23), GUAG (No. 24),







UCUU (No. 25), UUGG (No. 26), UUUG (No. 27),







GGAU (No. 28), UUUU (No. 29), CGUU (No. 30),







UUAU (No. 31), GUUC (No. 32), GUGG (No. 33),







GGUG (No. 34), UAUU (No. 35), UCUG (No. 36),







GUAC (No. 37), UAGG (No. 38), UCUC (No. 39),







UAGC (No. 40), UAUC (No. 41), CUAU (No. 42),







UACU (No. 43), CGGU (No. 44), UGCG (No. 45),







UUUC (No. 46), UAUG (No. 47), UAAG (No. 48),







UACC (No. 49), UUAG (No. 50), GCUU (No. 51),







CAGU (No. 52), UGAG (No. 53), GAUU (No. 54),







GAGU (No. 55), GUUA (No. 56), UGCA (No. 57),







UUCU (No. 58), GCCU (No. 59), GGUC (No. 60),







GGCU (No. 61), UUAC (No. 62), UCAU (No. 63),







GCGU (No. 64), GCAU (No. 65), GAUG (No. 66),







GUCU (No. 67), CGUA (No. 68), CGAU (No. 69),








    • wherein the nucleotide sequences of the motifs are 5′→3′,

    • wherein the oligonucleotide is between 6 and 64, preferably between 12 and 50, more preferably between 14 and 40, even more preferably between 16 and 36, and most preferably between 18 and 30 nucleotides in length,

    • wherein the oligonucleotide has an IL-12 score of at least 93.53 when n=6; at least 107.77 when n=7; at least 121 when n=8; at least 121.98 when n=9; at least 8.4064×n+66.985 when n is greater than 9, wherein the IL-12 score is assigned according to the method described above,

    • wherein n is the length of the oligonucleotide,

    • and wherein the oligonucleotide is not 5′-UUGAUGUGUUUAGUCGCUA-3′ ([17]), 5′-GCACCACUAGUUGGUUGUC-3′ ([18]), 5′-UGCUAUUGGUGAUUGCCUC-3′ ([18]), 5′-GUUGUAGUUGUACUCCAGC-3′ ([18]), 5′-UGGUUG-3′ (WO 03/086280), 5′-GUUGUGGUUGUGGUUGUG-3′ (WO 03/086280).





In one embodiment, the 4mer motifs are selected from the group consisting of No. 1-11, preferably No. 1-10, No. 1-9, No. 1-8, more preferably No. 1-7, No. 1-6, No. 1-5, No. 1-4, even more preferably No. 1-3, No. 1-2 of the above-listed 4mer motifs, most preferably, the 4mer motif is UCGU.


The immunostimulatory RNA oligonucleotide of the invention may comprise one or more copies of the same 4mer motif, or one or more copies of different 4mer motifs.


The present invention also provide an immunostimulatory RNA oligonucleotide having immunostimulatory, in particular, IL-12-inducing activity, comprising at least one, preferably at least two, more preferably at least three, even more preferably at least four, even more preferably at least five, most preferably at least six, of the 4mer motifs selected from the group consisting of No. 1-11 of the 4mer motifs, wherein the spacer nucleotides which are not part of any of the 4mer motif(s) are identical, and wherein the spacer nucleotide is selected from the group consisting of A, T, C, G and variants and derivatives thereof.


In one embodiment, the spacer nucleotide is A or a derivative thereof.


The immunostimulatory RNA oligonucleotide of the invention can comprise one or more copies of one type of 4mer motif (e.g., UCGU) on a poly A backbone. Examples of such an oligonucleotide includes, but are not limited to:












AAAAAAAUCGUAAAAAAAA








AAAUCGUAAAAAAAAAAAA







AAAAAAAAAAAAUCGUAAA







AAAUCGUAAAAAUCGUAAA







UCGUAAAUCGUAAAUCGUA






In another embodiment, the immunostimulatory RNA oligonucleotide of the invention can comprise one or more copies of more than one type of 4mer motif (e.g., UGAU, GGUA, UGGC, UGGU, GUUG, UCGU) on a poly A backbone. Examples of such an oligonucleotide includes, but are not limited to:












AUGGUAAAGUUGAAAUCGU








AUGGUAAUGGUAAAGUUGA







AAAUGAUAAAGGUAAAAUGGCAAAUGGUAAAGUUGAAAUCGUAAA






The more than one 4mer motifs in an immunostimulatory RNA oligonucleotide may overlap. For example, AAAUGAUGGCAAAAAA.


Examples of the immunostimulatory RNA olignucleotide of the invention include, but are not limited to:













aaagguuaaaaaagguuaaa
(SEQ ID NO: 246)








aaauuguaaaaaagguuaaa
(SEQ ID NO: 247)







aaauugcaaaaaagguuaaa
(SEQ ID NO: 248)







aaaugcuaaaaaagguuaaa
(SEQ ID NO: 249)







aaaugauaaaaaagguuaaa
(SEQ ID NO: 250)







aaagguaaaaaaagguuaaa
(SEQ ID NO: 251)







aaauggcaaaaaagguuaaa
(SEQ ID NO: 252)







aaaugguaaaaaagguuaaa
(SEQ ID NO: 253)







aaaguugaaaaaagguuaaa
(SEQ ID NO: 254)







aaaucguaaaaaagguuaaa
(SEQ ID NO: 255)







aaagguuaaaaaauuguaaa
(SEQ ID NO: 256)







aaauuguaaaaaauuguaaa
(SEQ ID NO: 257)







aaauugcaaaaaauuguaaa
(SEQ ID NO: 258)







aaaugcuaaaaaauuguaaa
(SEQ ID NO: 259)







aaaugauaaaaaauuguaaa
(SEQ ID NO: 260)







aaagguaaaaaaauuguaaa
(SEQ ID NO: 261)







aaauggcaaaaaauuguaaa
(SEQ ID NO: 262)







aaaugguaaaaaauuguaaa
(SEQ ID NO: 263)







aaaguugaaaaaauuguaaa
(SEQ ID NO: 264)







aaaucguaaaaaauuguaaa
(SEQ ID NO: 265)







aaagguuaaaaaauugcaaa
(SEQ ID NO: 266)







aaauuguaaaaaauugcaaa
(SEQ ID NO: 267)







aaauugcaaaaaauugcaaa
(SEQ ID NO: 268)







aaaugcuaaaaaauugcaaa
(SEQ ID NO: 269)







aaaugauaaaaaauugcaaa
(SEQ ID NO: 270)







aaagguaaaaaaauugcaaa
(SEQ ID NO: 271)







aaauggcaaaaaauugcaaa
(SEQ ID NO: 272)







aaaugguaaaaaauugcaaa
(SEQ ID NO: 273)







aaaguugaaaaaauugcaaa
(SEQ ID NO: 274)







aaaucguaaaaaauugcaaa
(SEQ ID NO: 275)







aaagguuaaaaaaugcuaaa
(SEQ ID NO: 276)







aaauuguaaaaaaugcuaaa
(SEQ ID NO: 277)







aaauugcaaaaaaugcuaaa
(SEQ ID NO: 278)







aaaugcuaaaaaaugcuaaa
(SEQ ID NO: 279)







aaaugauaaaaaaugcuaaa
(SEQ ID NO: 280)







aaagguaaaaaaaugcuaaa
(SEQ ID NO: 281)







aaauggcaaaaaaugcuaaa
(SEQ ID NO: 282)







aaaugguaaaaaaugcuaaa
(SEQ ID NO: 283)







aaaguugaaaaaaugcuaaa
(SEQ ID NO: 284)







aaaucguaaaaaaugcuaaa
(SEQ ID NO: 285)







aaagguuaaaaaaugauaaa
(SEQ ID NO: 286)







aaauuguaaaaaaugauaaa
(SEQ ID NO: 287)







aaauugcaaaaaaugauaaa
(SEQ ID NO: 288)







aaaugcuaaaaaaugauaaa
(SEQ ID NO: 289)







aaaugauaaaaaaugauaaa
(SEQ ID NO: 290)







aaagguaaaaaaaugauaaa
(SEQ ID NO: 291)







aaauggcaaaaaaugauaaa
(SEQ ID NO: 292)







aaaugguaaaaaaugauaaa
(SEQ ID NO: 293)







aaaguugaaaaaaugauaaa
(SEQ ID NO: 294)







aaaucguaaaaaaugauaaa
(SEQ ID NO: 295)







aaagguuaaaaaagguaaaa
(SEQ ID NO: 296)







aaauuguaaaaaagguaaaa
(SEQ ID NO: 297)







aaauugcaaaaaagguaaaa
(SEQ ID NO: 298)







aaaugcuaaaaaagguaaaa
(SEQ ID NO: 299)







aaaugauaaaaaagguaaaa
(SEQ ID NO: 300)







aaagguaaaaaaagguaaaa
(SEQ ID NO: 301)







aaauggcaaaaaagguaaaa
(SEQ ID NO: 302)







aaaugguaaaaaagguaaaa
(SEQ ID NO: 303)







aaaguugaaaaaagguaaaa
(SEQ ID NO: 304)







aaaucguaaaaaagguaaaa
(SEQ ID NO: 305)







aaagguuaaaaaauggcaaa
(SEQ ID NO: 306)







aaauuguaaaaaauggcaaa
(SEQ ID NO: 307)







aaauugcaaaaaauggcaaa
(SEQ ID NO: 308)







aaaugcuaaaaaauggcaaa
(SEQ ID NO: 309)







aaaugauaaaaaauggcaaa
(SEQ ID NO: 310)







aaagguaaaaaaauggcaaa
(SEQ ID NO: 311)







aaauggcaaaaaauggcaaa
(SEQ ID NO: 312)







aaaugguaaaaaauggcaaa
(SEQ ID NO: 313)







aaaguugaaaaaauggcaaa
(SEQ ID NO: 314)







aaaucguaaaaaauggcaaa
(SEQ ID NO: 315)







aaagguuaaaaaaugguaaa
(SEQ ID NO: 316)







aaauuguaaaaaaugguaaa
(SEQ ID NO: 317)







aaauugcaaaaaaugguaaa
(SEQ ID NO: 318)







aaaugcuaaaaaaugguaaa
(SEQ ID NO: 319)







aaaugauaaaaaaugguaaa
(SEQ ID NO: 320)







aaagguaaaaaaaugguaaa
(SEQ ID NO: 321)







aaauggcaaaaaaugguaaa
(SEQ ID NO: 322)







aaaugguaaaaaaugguaaa
(SEQ ID NO: 323)







aaaguugaaaaaaugguaaa
(SEQ ID NO: 324)







aaaucguaaaaaaugguaaa
(SEQ ID NO: 325)







aaagguuaaaaaaguugaaa
(SEQ ID NO: 326)







aaauuguaaaaaaguugaaa
(SEQ ID NO: 327)







aaauugcaaaaaaguugaaa
(SEQ ID NO: 328)







aaaugcuaaaaaaguugaaa
(SEQ ID NO: 329)







aaaugauaaaaaaguugaaa
(SEQ ID NO: 330)







aaagguaaaaaaaguugaaa
(SEQ ID NO: 331)







aaauggcaaaaaaguugaaa
(SEQ ID NO: 332)







aaaugguaaaaaaguugaaa
(SEQ ID NO: 333)







aaaguugaaaaaaguugaaa
(SEQ ID NO: 334)







aaaucguaaaaaaguugaaa
(SEQ ID NO: 335)







aaagguuaaaaaaucguaaa
(SEQ ID NO: 336)







aaauuguaaaaaaucguaaa
(SEQ ID NO: 337)







aaauugcaaaaaaucguaaa
(SEQ ID NO: 338)







aaaugcuaaaaaaucguaaa
(SEQ ID NO: 339)







aaaugauaaaaaaucguaaa
(SEQ ID NO: 340)







aaagguaaaaaaaucguaaa
(SEQ ID NO: 341)







aaauggcaaaaaaucguaaa
(SEQ ID NO: 342)







aaaugguaaaaaaucguaaa
(SEQ ID NO: 343)







aaaguugaaaaaaucguaaa
(SEQ ID NO: 344)







aaaucguaaaaaaucguaaa
(SEQ ID NO: 345)







aaaguucaaaaaagguuaaa
(SEQ ID NO: 346)







aaagucaaaaaaagguuaaa
(SEQ ID NO: 347)







aaagcucaaaaaagguuaaa
(SEQ ID NO: 348)







aaaguugaaaaaagguuaaa
(SEQ ID NO: 349)







aaaguuuaaaaaagguuaaa
(SEQ ID NO: 350)







aaagguuaaaaaagguuaaa
(SEQ ID NO: 351)







aaaguguaaaaaagguuaaa
(SEQ ID NO: 352)







aaaggucaaaaaagguuaaa
(SEQ ID NO: 353)







aaagucuaaaaaagguuaaa
(SEQ ID NO: 354)







aaaguccaaaaaagguuaaa
(SEQ ID NO: 355)







aaaguucaaaaaauuguaaa
(SEQ ID NO: 356)







aaagucaaaaaaauuguaaa
(SEQ ID NO: 357)







aaagcucaaaaaauuguaaa
(SEQ ID NO: 358)







aaaguugaaaaaauuguaaa
(SEQ ID NO: 359)







aaaguuuaaaaaauuguaaa
(SEQ ID NO: 360)







aaagguuaaaaaauuguaaa
(SEQ ID NO: 361)







aaaguguaaaaaauuguaaa
(SEQ ID NO: 362)







aaaggucaaaaaauuguaaa
(SEQ ID NO: 363)







aaagucuaaaaaauuguaaa
(SEQ ID NO: 364)







aaaguccaaaaaauuguaaa
(SEQ ID NO: 365)







aaaguucaaaaaauugcaaa
(SEQ ID NO: 366)







aaagucaaaaaaauugcaaa
(SEQ ID NO: 367)







aaagcucaaaaaauugcaaa
(SEQ ID NO: 368)







aaaguugaaaaaauugcaaa
(SEQ ID NO: 369)







aaaguuuaaaaaauugcaaa
(SEQ ID NO: 370)







aaagguuaaaaaauugcaaa
(SEQ ID NO: 371)







aaaguguaaaaaauugcaaa
(SEQ ID NO: 372)







aaaggucaaaaaauugcaaa
(SEQ ID NO: 373)







aaagucuaaaaaauugcaaa
(SEQ ID NO: 374)







aaaguccaaaaaauugcaaa
(SEQ ID NO: 375)







aaaguucaaaaaaugcuaaa
(SEQ ID NO: 376)







aaagucaaaaaaaugcuaaa
(SEQ ID NO: 377)







aaagcucaaaaaaugcuaaa
(SEQ ID NO: 378)







aaaguugaaaaaaugcuaaa
(SEQ ID NO: 379)







aaaguuuaaaaaaugcuaaa
(SEQ ID NO: 380)







aaagguuaaaaaaugcuaaa
(SEQ ID NO: 381)







aaaguguaaaaaaugcuaaa
(SEQ ID NO: 382)







aaaggucaaaaaaugcuaaa
(SEQ ID NO: 383)







aaagucuaaaaaaugcuaaa
(SEQ ID NO: 384)







aaaguccaaaaaaugcuaaa
(SEQ ID NO: 385)







aaaguucaaaaaaugauaaa
(SEQ ID NO: 386)







aaagucaaaaaaaugauaaa
(SEQ ID NO: 387)







aaagcucaaaaaaugauaaa
(SEQ ID NO: 388)







aaaguugaaaaaaugauaaa
(SEQ ID NO: 389)







aaaguuuaaaaaaugauaaa
(SEQ ID NO: 390)







aaagguuaaaaaaugauaaa
(SEQ ID NO: 391)







aaaguguaaaaaaugauaaa
(SEQ ID NO: 392)







aaaggucaaaaaaugauaaa
(SEQ ID NO: 393)







aaagucuaaaaaaugauaaa
(SEQ ID NO: 394)







aaaguccaaaaaaugauaaa
(SEQ ID NO: 395)







aaaguucaaaaaagguaaaa
(SEQ ID NO: 396)







aaagucaaaaaaagguaaaa
(SEQ ID NO: 397)







aaagcucaaaaaagguaaaa
(SEQ ID NO: 398)







aaaguugaaaaaagguaaaa
(SEQ ID NO: 399)







aaaguuuaaaaaagguaaaa
(SEQ ID NO: 400)







aaagguuaaaaaagguaaaa
(SEQ ID NO: 401)







aaaguguaaaaaagguaaaa
(SEQ ID NO: 402)







aaaggucaaaaaagguaaaa
(SEQ ID NO: 403)







aaagucuaaaaaagguaaaa
(SEQ ID NO: 404)







aaaguccaaaaaagguaaaa
(SEQ ID NO: 405)







aaaguucaaaaaauggcaaa
(SEQ ID NO: 406)







aaagucaaaaaaauggcaaa
(SEQ ID NO: 407)







aaagcucaaaaaauggcaaa
(SEQ ID NO: 408)







aaaguugaaaaaauggcaaa
(SEQ ID NO: 409)







aaaguuuaaaaaauggcaaa
(SEQ ID NO: 410)







aaagguuaaaaaauggcaaa
(SEQ ID NO: 411)







aaaguguaaaaaauggcaaa
(SEQ ID NO: 412)







aaaggucaaaaaauggcaaa
(SEQ ID NO: 413)







aaagucuaaaaaauggcaaa
(SEQ ID NO: 414)







aaaguccaaaaaauggcaaa
(SEQ ID NO: 415)







aaaguucaaaaaaugguaaa
(SEQ ID NO: 416)







aaagucaaaaaaaugguaaa
(SEQ ID NO: 417)







aaagcucaaaaaaugguaaa
(SEQ ID NO: 418)







aaaguugaaaaaaugguaaa
(SEQ ID NO: 419)







aaaguuuaaaaaaugguaaa
(SEQ ID NO: 420)







aaagguuaaaaaaugguaaa
(SEQ ID NO: 421)







aaaguguaaaaaaugguaaa
(SEQ ID NO: 422)







aaaggucaaaaaaugguaaa
(SEQ ID NO: 423)







aaagucuaaaaaaugguaaa
(SEQ ID NO: 424)







aaaguccaaaaaaugguaaa
(SEQ ID NO: 425)







aaaguucaaaaaaguugaaa
(SEQ ID NO: 426)







aaagucaaaaaaaguugaaa
(SEQ ID NO: 427)







aaagcucaaaaaaguugaaa
(SEQ ID NO: 428)







aaaguugaaaaaaguugaaa
(SEQ ID NO: 429)







aaaguuuaaaaaaguugaaa
(SEQ ID NO: 430)







aaagguuaaaaaaguugaaa
(SEQ ID NO: 431)







aaaguguaaaaaaguugaaa
(SEQ ID NO: 432)







aaaggucaaaaaaguugaaa
(SEQ ID NO: 433)







aaagucuaaaaaaguugaaa
(SEQ ID NO: 434)







aaaguccaaaaaaguugaaa
(SEQ ID NO: 435)







aaaguucaaaaaaucguaaa
(SEQ ID NO: 436)







aaagucaaaaaaaucguaaa
(SEQ ID NO: 437)







aaagcucaaaaaaucguaaa
(SEQ ID NO: 438)







aaaguugaaaaaaucguaaa
(SEQ ID NO: 439)







aaaguuuaaaaaaucguaaa
(SEQ ID NO: 440)







aaagguuaaaaaaucguaaa
(SEQ ID NO: 441)







aaaguguaaaaaaucguaaa
(SEQ ID NO: 442)







aaaggucaaaaaaucguaaa
(SEQ ID NO: 443)







aaagucuaaaaaaucguaaa
(SEQ ID NO: 444)







aaaguccaaaaaaucguaaa
(SEQ ID NO: 445)






In one embodiment, immunostimulatory RNA oligonucleotide of the invention does not have gene silencing activity for any known mammalian gene.


The present invention provides a single-stranded RNA oligonucleotide having IFN-α-inducing activity and low IL-12-inducing activity, comprising at least one, preferably at least two, more preferably at least three, even more preferably at least four, of the 4-nucleotide (4-mer) motifs selected from the group consisting of:


GCUC, GUCA, GUUC, GGUC, GUCC, GUCU, GUUU, CGUC, GCUU, GUGU,

wherein the nucleotide sequences of the motifs are 5′→3′,


wherein the oligonucleotide has an IFN-α score of at least 1.4909×n+22.014 and an IL-12 score of at most 2.4194×n −31.914, wherein the IFN-α score is assigned according to the “addition method” described in the co-pending application and the IL-12 score is assigned according to the method described above;


or wherein the oligonucleotide has an IFN-α score of at least 0.58 and an IL-12 score of at most 2.4194×n −31.914, wherein the IFN-α score is assigned according to the “simplified method” described in the co-pending application and the IL-12 score is assigned according to the method described above;


wherein the oligonucleotide is between 6 and 64, preferably between 12 and 50, more preferably between 14 and 40, even more preferably between 16 and 36, and most preferably between 18 and 30 nucleotides in length.


The present application also provides a single-stranded RNA oligonucleotide having IL-12-inducing activity and low IFN-α-inducing activity, comprising at least one, preferably at least two, more preferably at least three, even more preferably at least four, of the 4-nucleotide (4-mer) motifs selected from the group consisting of:


UCGU, GAUA, UGGC, UGCU, UGGU, UGCC, UUGC, UGAC, UAAU, UUAU,

wherein the nucleotide sequences of the motifs are 5′→3′,


wherein the oligonucleotide has an IL-12 score of at least 8.4064×n+66.958 and an IFN-α score of at most 0.5439×n −8.0234, wherein the IFN-α score is assigned according to the “addition method” described in the co-pending application and the IL-12 score is assigned according to the method described above;


or wherein the oligonucleotide has an IL-12 score of at least 8.4064×n+66.958 and an IFN-α score of at most 0.11, wherein the IFN-α score is assigned according to the “simplified method” described in the co-pending application and the IL-12 score is assigned according to the method described above;


wherein the oligonucleotide is between 6 and 64, preferably between 12 and 50, more preferably between 14 and 40, even more preferably between 16 and 36, and most preferably between 18 and 30 nucleotides in length.


The present application further provides a single-stranded RNA oligonucleotide having both IL-12-inducing activity and IFN-α-inducing activity, comprising at least one, preferably at least two, more preferably at least three, even more preferably at least four, of the 4-nucleotide (4-mer) motifs selected from the group consisting of:


GUUG, GGUU, UUGU, GGUA, CUGU, UGUC, UGUA, UGUU, UGUG, UAGU,

wherein the nucleotide sequences of the motifs are 5′→3′,


wherein the oligonucleotide has an IL-12 score of at least 8.4064×n+66.958 and an IFN-α score of at least 1.4909×n+22.014, wherein the IFN-α score is assigned according to the “addition method” described in the co-pending application and the IL-12 score is assigned according to the method described above;


or wherein the oligonucleotide has an IL-12 score of at least 8.4064×n+66.958 and an IFN-α score of at least 0.58, wherein the IFN-α score is assigned according to the “simplified method” described in the co-pending application and the IL-12 score is assigned according to the method described above;


wherein the oligonucleotide is between 6 and 64, preferably between 12 and 50, more preferably between 14 and 40, even more preferably between 16 and 36, and most preferably between 18 and 30 nucleotides in length.


Briefly, the “addition method” for assigning the IFN-α score of an oligonucleotide and thereby predicting its immunostimulatory activity comprises the steps of:

    • (a) identifying all possible 3-nucleotide (3mer) motifs contained in the oligonucleotide;
    • (b) assigning an IFN-α point score for each individual 3mer motif:
      • (i) for a 3mer motif which appears in Table A, assign an IFN-α point score according to Table A;
      • (ii) for a 3mer motif which does not appear in Table A, assign an IFN-α point score of 0;
    • (c) assigning the sum of the IFN-α point scores of individual 3mer motifs as the IFN-α score of the oligonucleotide; and
    • (d) assigning to the oligonucleotide a high immunostimulatory activity if the IFN-α score is at least 23, an intermediate immunostimulatory activity if the IFN-α score is between −4 and 23, and a low immunostimulatory activity if the IFN-α score is at most −4, when n=6;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IFN-α score is at least 26, an intermediate immunostimulatory activity if the IFN-α score is between −4 and 26, and a low immunostimulatory activity if the IFN-α score is at most −4, when n=7;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IFN-α score is at least 28, an intermediate immunostimulatory activity if the IFN-α score is between −5 and 28, and a low immunostimulatory activity if the IFN-α score is at most −5, when n=8;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IFN-α score is at least 30, an intermediate immunostimulatory activity if the IFN-α score is between −5 and 30, and a low immunostimulatory activity if the IFN-α score is at most −5, when n=9;
      • assigning to the oligonucleotide a high immunostimulatory activity if the IFN-α score is at least 1.4909×n+22.014, an intermediate immunostimulatory activity if the IFN-α score is between 0.005×n2−0.2671×n−3.5531 and 1.4909×n+22.014, and a low immunostimulatory activity if the IFN-α score is at most 0.005×n2−0.2671×n−3.5531, when n is greater than 9,
      • wherein n is the length of the oligonucleotide.












TABLE A







3mer motif




(5′→3′)
IFN-α point score









ACA
−2



ACC
−2



AGA
−2



AAC
−1



AUA
−1



UGG
+1



GUA
+3



GUG
+3



GGU
+4



UCA
+4



UUC
+4



UUU
+5



AGU
+6



UUG
+6



GUC
+8



UGU
+8



GUU
+9










The “simplified method” for assigning the IFN-α score of an oligonucleotide and thereby predicting its immunostimulatory activity comprises the steps of:

    • (a) identifying all possible 3-nucleotide (3mer) motifs contained in the oligonucleotide;
    • (b) assigning an IFN-α point score for each individual 3mer motif according to Table B;
    • (c) assigning the highest individual IFN-α point score as the IFN-α score of the oligonucleotide; and
    • (d) assigning to the oligonucleotide a high immunostimulatory activity if the IFN-α score is at least 0.58, an intermediate immunostimulatory activity if the IFN-α score is between 0.11 and 0.58, and a low immunostimulatory activity if the IFN-α score is at most 0.11.













TABLE B







Mean






(IFN-α point


Motif
Occurrences
score)
Sem
p-value



















AAA
2192
0.00
0.01
0.389


AAC
67
−0.14
0.02
**0.001


AAG
67
0.11
0.06
**0.009


AAU
67
0.01
0.03
0.841


ACA
31
−0.17
0.02
**0.007


ACC
19
−0.19
0.01
*0.017


ACG
19
−0.12
0.05
0.128


ACU
19
−0.07
0.05
0.407


AGA
31
−0.14
0.02
*0.021


AGC
19
−0.05
0.09
0.508


AGG
19
−0.01
0.09
0.877


AGU
19
0.59
0.10
**<0.001


AUA
31
−0.10
0.02
0.118


AUC
19
−0.11
0.03
0.176


AUG
19
0.08
0.07
0.318


AUU
19
0.09
0.06
0.253


CAA
67
0.00
0.05
0.913


CAC
7
−0.21
0.01
0.11


CAG
7
−0.13
0.06
0.313


CAU
7
−0.19
0.01
0.143


CCA
19
−0.14
0.05
0.077


CCC
7
−0.21
0.01
0.102


CCG
7
−0.21
0.01
0.11


CCU
7
−0.15
0.03
0.252


CGA
19
−0.14
0.04
0.082


CGC
7
−0.19
0.02
0.155


CGG
7
−0.18
0.03
0.172


CGU
7
0.05
0.11
0.678


CUA
19
−0.09
0.05
0.251


CUC
7
0.16
0.19
0.229


CUG
7
0.00
0.12
0.979


CUU
7
0.08
0.12
0.529


GAA
67
−0.08
0.03
*0.044


GAC
7
−0.19
0.02
0.153


GAG
7
−0.13
0.06
0.306


GAU
7
−0.11
0.01
0.385


GCA
19
−0.12
0.04
0.114


GCC
7
−0.19
0.03
0.149


GCG
7
−0.17
0.03
0.184


GCU
7
0.22
0.21
0.094


GGA
19
−0.13
0.04
0.111


GGC
7
−0.17
0.04
0.194


GGG
7
−0.22
0.00
0.095


GGU
7
0.40
0.18
**0.002


GUA
19
0.27
0.08
**<0.001


GUC
7
0.82
0.10
**<0.001


GUG
7
0.32
0.13
*0.014


GUU
7
0.87
0.18
**<0.001


UAA
67
0.06
0.04
0.135


UAC
7
−0.06
0.08
0.647


UAG
7
0.01
0.08
0.94


UAU
7
−0.03
0.07
0.793


UCA
19
0.35
0.12
**<0.001


UCC
7
−0.03
0.14
0.794


UCG
7
0.02
0.09
0.848


UCU
7
0.10
0.13
0.421


UGA
19
0.04
0.08
0.611


UGC
7
0.01
0.07
0.91


UGG
7
0.10
0.09
0.448


UGU
7
0.69
0.06
**<0.001


UUA
19
0.17
0.10
*0.036


UUC
7
0.37
0.18
**0.005


UUG
7
0.38
0.18
**0.004


UUU
7
0.32
0.17
*0.013









The immunostimulatory RNA oligonucleotide of the invention may be covalently linked to one or more lipophilic groups which enhance the stability and the activity and facilitate the delivery of the RNA oligonucleotides.


As used herein, the term “lipophilic” or “lipophilic group” broadly refers to any compound or chemical moiety having an affinity for lipids. Lipophilic groups encompass compounds of many different types, including those having aromatic, aliphatic or alicyclic characteristics, and combinations thereof.


In specific embodiments, the lipophilic group is an aliphatic, alicyclic, or polyalicyclic substance, such as a steroid (e.g., sterol) or a branched aliphatic hydrocarbon. The lipophilic group generally comprises a hydrocarbon chain, which may be cyclic or acyclic. The hydrocarbon chain may comprise various substituents and/or at least one heteroatom, such as an oxygen atom. Such lipophilic aliphatic moieties include, without limitation, saturated or unsatarated fatty acids, waxes (e.g., monohydric alcohol esters of fatty acids and fatty diamides), terpenes (e.g., the C10 terpenes, C15 sesquiterpenes, C20 diterpenes, C30 triterpenes, and C40 tetraterpenes), and other polyalicyclic hydrocarbons.


The lipophilic group may be attached by any method known in the art, including via a functional grouping present in or introduced into the RNA oligonucleotide, such as a hydroxy group (e.g., —CO—CH2—OH). Conjugation of the RNA oligonucleotide and the lipophilic group may occur, for example, through formation of an ether or a carboxylic or carbamoyl ester linkage between the hydroxy and an alkyl group R—, an alkanoyl group RCO— or a substituted carbamoyl group KNHCO—. The alkyl group R may be cyclic (e.g., cyclohexyl) or acyclic (e.g., straight-chained or branched; and saturated or unsaturated). Alkyl group R may be a butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl or octadecyl group, or the like. Preferably, the lipophilic group is conjugated to the 5′-hydroxyl group of the terminal nucleotide. In a preferred embodiment, the liphophilic group is 12-hydroxydodeconoic acid bisdecylamide.


In another embodiment, the lipophilic group is a steroid, such as sterol. Steroids are polycyclic compounds containing a perhydro-1,2-cyclopentanophenanthrene ring system. Steroids include, without limitation, bile acids (e.g., cholic acid, deoxycholic acid and dehydrocholic acid), cortisone, digoxigenin, testosterone, cholesterol and cationic steroids, such as cortisone.


In a preferred embodiment, the lipophilic group is cholesterol or a derivative thereof. A “cholesterol derivative” refers to a compound derived from cholesterol, for example by substitution, addition or removal of substituents. The steroid may be attached to the RNA oligonucleotide by any method known in the art. In a preferred embodiment, the liphophilic group is cholesteryl (6-hydroxyhexyl) carbamate.


In another embodiment, the lipophilic group is an aromatic moiety. In this context, the term “aromatic” refers broadly to mono- and polyaromatic hydrocarbons. Aromatic groups include, without limitation, C6-C14 aryl moieties comprising one to three aromatic rings, which may be optionally substituted; “aralkyl” or “arylalkyl” groups comprising an aryl group covalently linked to an alkyl group, either of which may independently be optionally substituted or unsubstituted; and “heteroaryl” groups. As used herein, the term “heteroaryl” refers to groups having 5 to 14 ring atoms, preferably 5, 6, 9, or 10 ring atoms; having 6, 10, or 14π electrons shared in a cyclic array; and having, in addition to carbon atoms, between one and about three heteroatoms selected from the group consisting of nitrogen (N), oxygen (O), and sulfur (S).


As used herein, a “substituted” alkyl, cycloalkyl, aryl, heteroaryl, or heterocyclic group is one having between one and about four, preferably between one and about three, more preferably one or two, non-hydrogen substituents. Suitable substituents include, without limitation, halo, hydroxy, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, acylamino, alkylcarbamoyl, arylcarbamoyl, aminoalkyl, alkoxycarbonyl, carboxy, hydroxyalkyl, alkanesulfonyl, arenesulfonyl, alkanesulfonamido, arenesulfonamido, aralkylsulfonamido, alkylcarbonyl, acyloxy, cyano, and ureido groups.


The lipophilic group can be covalently linked directly or indirectly via a linker to the RNA oligonucleotide. The covalent linkage may or may not comprise a phosphodiester group. And the linker may be of various lengths. The preferred lengths of the linker are known to those skilled in the art and may be determined experimentally.


In one embodiment, the lipophilic group is covalently linked to the 5′ end of the RNA oligonucleotide.


In addition, the immunostimulatory oligonucleotide of the invention may be coupled to a solid support. By “coupled” it is meant that the oligonucleotide is covalently or non-covalently, directly or indirectly, linked to the solid support. Suitable solid supports include, but are not limited to, silicon wafers, synthetic polymer support such as polystyrene, polypropylene, polyglycidylmethacrylate, substituted polystyrene (e.g., aminated or carboxylated polystyrene, polyacrlamides, polyamides, polyvinylchlorides, etc.), glass, agarose, nitrocellulose, nylon and gelatin nanoparticles. Solid support may enhance the stability and the activity of the oligonucleotide, especially short oligonucleotides less than 16 nucleotides in length.


Immunostimulatory RNA Oligonucleotide Conjugates

The present invention also provides immunomodulatory RNA oligonucleotide conjugates, comprising an immunomodulatory RNA oligonucleotide and an antigen conjugated to the oligonucleotide. In some embodiments, the antigen is conjugated to the oligonucleotide at a position other than its 3′ end. In some embodiments, the antigen produces a vaccine effect.


The antigen is preferably selected from the group consisting of disease/disorder-related antigens. The disorder may be a cancer, a dermatological disorder, an immune disorder, a metabolic disorder, a neurological disorder, an ocular disease, an infection, or other hereditary and non-hereditary disorders. The antigen may be a protein, a polypeptide, a peptide, a carbohydrate, or a combination thereof.


The immunostimulatory RNA oligonucleotide may be covalently linked to the antigen, or it is otherwise operatively associated with the antigen. As used herein, the term “operatively associated with” refers to any association that maintains the activity of both the oligonucleotide and the antigen. Non-limiting examples of such operative associations include being part of the same liposome or other such delivery vehicle or reagent. In embodiments wherein the oligonucleotide agent is covalently linked to the antigen, such covalent linkage preferably is at any position on the oligonucleotide that does not interfere with the immunostimulatory activity of the oligonucleotide.


Antisense RNA Oligonucleotide with High or Low Immunostimulatory Activity


The present invention provides an antisense RNA oligonucleotide with gene silencing activity.


In one embodiment, the antisense RNA oligonucleotide has both gene silencing activity and immunostimulatory activity, wherein the oligonucleotide has an IL-12 score of at least 8.4064×n 66.958, wherein the IL-12 score is assigned according to the method describe above, wherein n is the length of the oligonucleotide and n is between 14 and 50, and wherein the oligonucleotide is not 5′-GCACCACUAGUUGGUUGUC-3′ ([18]), 5′-UGCUAUUGGUGAUUGCCUC-3′ ([18]), 5′-GUUGUAGUUGUACUCCAGC-3′ ([18]).


In another embodiment, the antisense RNA oligonucleotide has gene silencing activity and low/minimal immunostimulatory activity, wherein the oligonucleotide has an IL-12 score of at most 1.9763×n−30. 643, wherein the IL-12 score is assigned according to the method describe above, wherein n is the length of the oligonucleotide and n is between 14 and 50, and wherein the oligonucleotide is not 5′-UACCUAACCGGACAUAAUC-3′ ([17]), 5′-UAAACCUUCGAUUCCGACC-3′ ([17]), 5′-UAGCGACUAAACGCAUCAA-3′ ([17]), 5′-AUACGCUCAGACAAAGCUG-3′ ([17]), 5′-AUACGCUCACACAAAGCUG-3′ ([17]), 5′-CUAAUACAGGCCAAUACAU-3′ ([17]), 5′-UAGCGACUAAACACAUCAA-3′ ([17]), 5′-UAACCUUUAGCUCCGACC-3′ ([17]), 5′-AUACCAGGCUCCAAAGCUG-3′ ([17]), 5′-UAGCGACUAAGCGCAUCAA-3′ ([17]), 5′-AUACGCUCAGCCAAAGCUG-3′ ([17]), 5′-AAGGCAGCACGACUUCUUC-3′ ([18]).


The antisense RNA oligonucleotide of the invention may be covalently linked to one or more lipophilic groups which enhance the stability and the activity and facilitate the delivery of the RNA oligonucleotides.


Pharmaceutical Compositions

The present invention provides pharmaceutical compositions comprising one or more of the ssRNA oligonucleotides of the invention and a pharmaceutically acceptable carrier. The more than one RNA oligonucleotides may have the same, similar, or different functionalities including, but are not limited to immunostimulatory activity and gene silencing activity.


For example, an ssRNA oligonucleotide having immunostimulatory activity but lacking gene silencing activity may be combined with an ssRNA oligonucleotide having gene silencing activity and low immunostimulatory activity in a pharmaceutical composition to achieve both immune activation and gene silencing. Such a combination composition may be useful for treating disorders such as cancers and viral infections. Such a combination composition may be necessary when the two activities cannot be optimized on a single RNA oligonucleotide.


In one embodiment, the pharmaceutical composition further comprises a RNA complexation agent. In a preferred embodiment, the complexation agent is a polycationic peptide, preferably poly-L-arginine (poly-L-Arg). In a preferred embodiment, the polycationic peptide, in particular, poly-L-Arg, is at least 24 amino acids in length. The polycationic peptide, in particular, poly-L Arg, may be a heterogeneous mixture of peptides of different lengths.


The pharmaceutical composition of the invention may further comprises another agent such as an agent that stabilizes the RNA oligonucleotide(s), e.g., a protein that complexes with the oligonucleotide agent to form an iRNP. Still other agents include chelators, e.g., EDTA (e.g., to remove divalent cations such as Mg2+), salts, RNAse inhibitors (e.g., a broad specificity RNAse inhibitor such as RNAsin) and so forth.


The pharmaceutical composition of the present invention may further comprise one or more additional pharmaceutically active (or therapeutic) agents which are selected from the group consisting of agents that are used for the treatment of cancer, dermatological disorders, immune disorders, metabolic disorders, neurological disorders, ocular diseases, infections, and other hereditary and non-hereditary disorders in a mammal.


In certain embodiments, the additional pharmaceutically active agent is selected from the group consisting of immunostimulatory RNA oligonucleotides, immunostimulatory DNA oligonucleotides, cytokines, chemokines, growth factors, antibiotics, anti-angiogenic factors, chemotherapeutic agents, anti-viral agents, anti-fungal agents, anti-parasitic agents, and antibodies. In one embodiment, the additional pharmaceutically active agent is natural or recombinant IL-12, or a CpG-containing RNA oligonucleotide capable inducing IL-12 (see e.g., Sugiyama et al. 2005, J Immunol 174:2273-2279). In another embodiment, the additional pharmaceutically active agent is natural or recombinant IFN-α polypeptide, an immunostimulatory RNA oligonucleotide capable of inducing IFN-α (see e.g., our co-pending application), or a CpG-containing or non-CpG-containing DNA oligonucleotide capable of inducing IFN-α (see e.g., WO 01/22990, WO 03/101375). In yet another embodiment, the additional pharmaceutically active agent is natural or recombinant IL-2. In another embodiment, the additional pharmaceutically active agent is an anti-angiogenic factor such as vasostatin or an anti-VEGF antibody. In certain embodiments, the additional pharmaceutically active agent is a cancer-specific agent such as Herceptin, Rituxan, Gleevec, Iressa.


A formulated oligonucleotide composition can assume a variety of states. In some examples, the composition is at least partially crystalline, uniformly crystalline, and/or anhydrous (e.g., less than 80, 50, 30, 20, or 10% water). In another example, the oligonucleotide agent is in an aqueous phase, e.g., in a solution that includes water, this form being the preferred form for administration via inhalation.


The aqueous phase or the crystalline compositions can be incorporated into a delivery vehicle, e.g., a liposome (particularly for the aqueous phase), or a particle (e.g., a microparticle as can be appropriate for a crystalline composition). Generally, the oligonucleotide composition is formulated in a manner that is compatible with the intended method of administration.


The pharmaceutical compositions encompassed by the invention may be administered by any means known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal, and topical (including buccal and sublingual) administration. In preferred embodiments, the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection. The pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.


For oral administration, the oligonucleotide agent useful in the invention will generally be provided in the form of tablets or capsules, as a powder or granules, or as an aqueous solution or suspension.


Tablets for oral use may include the active ingredients mixed with pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.


Capsules for oral use include hard gelatin capsules in which the active ingredient is mixed with a solid diluent, and soft gelatin capsules wherein the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.


For intramuscular, intraperitoneal, subcutaneous and intravenous use, the pharmaceutical compositions of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity. Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride. In a preferred embodiment, the carrier consists exclusively of an aqueous buffer. In this context, “exclusively” means no auxiliary agents or encapsulating substances are present which might affect or mediate uptake of oligonucleotide agent in the cells that harbor the target gene or virus. Such substances include, for example, micellar structures, such as liposomes or capsids, as described below. Although microinjection, lipofection, viruses, viroids, capsids, capsoids, or other auxiliary agents are required to introduce oligonucleotide agent into cell cultures, surprisingly these methods and agents are not necessary for uptake of oligonucleotide agent in vivo. The oligonucleotide agent of the present invention are particularly advantageous in that they do not require the use of an auxiliary agent to mediate uptake of the oligonucleotide agent into the cell, many of which agents are toxic or associated with deleterious side effects. Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, polyvinyl-pyrrolidone and gum tragacanth, and a wetting agent such as lecithin. Suitable preservatives for aqueous suspensions include ethyl and n-propyl p-hydroxybenzoate.


The pharmaceutical compositions can also include encapsulated formulations to protect the oligonucleotide agent against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811; PCT publication WO 91/06309; and European patent publication EP-A-43075.


In general, a suitable dose of a RNA oligonucleotide will be in the range of 0.001 to 500 milligrams per kilogram body weight of the recipient per day (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 100 milligrams per kilogram, about 1 milligrams per kilogram to about 75 milligrams per kilogram, about 10 micrograms per kilogram to about 50 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram). The pharmaceutical composition may be administered once per day, or the oligonucleotide agent may be administered as two, three, four, five, six or more sub-doses at appropriate intervals throughout the day. In that case, the oligonucleotide agent contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage. The dosage unit can also be compounded for delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the oligonucleotide agent over a several day period. Sustained release formulations are well known in the art. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.


The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the infection or disease/disorder, previous treatments, the general health and/or age of the subject, and other diseases/disorders present. Moreover, treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments. Estimates of effective dosages and in vivo half-lives for the individual RNA oligonucleotide agent encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate animal model, as described elsewhere herein.


Toxicity and therapeutic efficacy of the RNA oligonucleotide and the pharmaceutical composition of the 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. oligonucleotide agents that exhibit high therapeutic indices are preferred.


The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosages of compositions of the invention are 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 oligonucleotide 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 of the oligonucleotide agent or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a decreased concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test oligonucleotide agent which 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.


The administering physician can adjust the amount and timing of the administration of the pharmaceutical composition of the invention on the basis of results observed using standard measures of efficacy known in the art or described herein.


Use of the ssRNA Oligonucleotide for Inducing an Immune Response


The present application provides the use of the immunostimulatory ssRNA oligonucleotide of the invention for the preparation of a pharmaceutical composition for inducing an immune response in a mammal.


Inducing an immune response means initiating or causing an increase in one or more of B-cell activation, T-cell activation, natural killer cell activation, activation of antigen presenting cells (e.g., B cells, dendritic cells, monocytes and macrophages), cytokine production, chemokine production, specific cell surface marker expression, in particular, expression of co-stimulatory molecules. In one aspect, such an immune response involves the production of type IL-12 in cells such as monocytes, macrophages, and myeloid dendritic cells.


Use of the RNA Oligonucleotide for Treating Diseases/Disorders

The present invention provides the use of the immunostimulatory ssRNA oligonucleotide of the invention for the preparation of a pharmaceutical composition for preventing and/or treating a disorder selected from immune disorders, infections, and cancers in a mammal, wherein the induction of an immune response is beneficial to the mammal.


The present invention also provides the use of the antisense RNA oligonucleotide of the invention which has both immunostimulatory activity and gene silencing activity for the preparation of a pharmaceutical composition for preventing and/or treating a disorder selected from infections and cancers in a mammal, wherein the induction of an immune response together with the downregulation of a disorder-related gene are beneficial to the mammal.


The present invention further provides the use of the antisense RNA oligonucleotide of the invention which has gene silencing activity and low/minimal immunostimulatory activity for the preparation of a pharmaceutical composition for preventing and/or treating a disorder in a mammal caused by the expression or overexpression of a disorder-related gene, wherein the induction of an immune disorder it to be avoided. The disorder may be selected from cancer, dermatological disorders, immune disorders, metabolic disorders, neurological disorders, ocular diseases, infections, and other hereditary and non-hereditary disorders.


The immune disorders include, but are not limited to, allergy, autoimmune disorders, inflammatory disorders, and immunodeficiency.


Allergies include, but are not limited to, food allergies and respiratory allergies.


Autoimmune diseases include, but are not limited to, diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, encephalomyelitis, myasthenia gravis, systemic lupus erythematosis, automimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, drug eruptions, leprosy reversal reactions, erythema nodosum leprosum, autoimmune uveitis, allergic encephalomyelitis, acute necrotizing hemorrhagic encephalopathy, idiopathic bilateral progressive sensorineural hearing, loss, aplastic anemia, pure red cell anemia, idiopathic thrombocytopenia, polychondritis, Wegener's granulomatosis, chronic active hepatitis, Stevens-Johnson syndrome, idiopathic sprue, lichen planus, Graves' disease, sarcoidosis, primary biliary cirrhosis, uveitis posterior, and interstitial lung fibrosis.


Inflammatory disorders include, without limitation, airway inflammation which includes, without limitation, asthma.


Immunodeficiencies include, but are not limited to, spontaneous immunodeficiency, acquired immunodeficiency (including AIDS), drug-induced immunodeficiency (such as that induced by immunosuppressants used in transplantation and chemotherapeutic agents used for treating cancer).


In one embodiment, the immune disorders include those caused by pathological Th2 responses.


The infections include, but are not limited to viral infections, bacterial infections, anthrax, parasitic infections, fungal infections and prion infection.


Viral infections include, but are not limited to, infection by hepatitis C, hepatitis B, herpes simplex virus (HSV), HIV-AIDS, measles virus, poliovirus, and smallpox virus. Examples of (+) strand RNA viruses which can be targeted for inhibition include, without limitation, picornaviruses, caliciviruses, nodaviruses, coronaviruses, arteriviruses, flaviviruses, and togaviruses. Examples of picornaviruses include enterovirus (poliovirus 1), rhinovirus (human rhinovirus 1A), hepatovirus (hepatitis A virus), cardiovirus (encephalomyocarditis virus), aphthovirus (foot-and-mouth disease virus 0), and parechovirus (human echovirus 22). Examples of caliciviruses include vesiculovirus (swine vesicular exanthema virus), lagovirus (rabbit hemorrhagic disease virus), “Norwalk-like viruses” (Norwalk virus), “Sapporo-like viruses” (Sapporo virus), and “hepatitis E-like viruses” (hepatitis E virus). Betanodavirus (striped jack nervous necrosis virus) is the representative nodavirus. Coronaviruses include coronavirus (avian infections bronchitis virus) and torovirus (Berne virus). Arterivirus (equine arteritis virus) is the representative arteriviridus. Togavirises include alphavirus (Sindbis virus) and rubivirus (Rubella virus). Finally, the flaviviruses include flavivirus (Yellow fever virus), pestivirus (bovine diarrhea virus), and hepacivirus (hepatitis C virus).


In certain embodiments, the viral infections are selected from chornic hepatitis B, chornic hepatitis C, HIV infection, RSV infection, HSV infection, VSV infection, CMV infection, measles virus infection, and influenza infection.


In certain embodiments, the parasitic infections include, but are not limited to, Lesihmania infection and Toxoplasma infection.


In one embodiment, the bacterial infection is M. tuberculosis infection.


Cancers include, but are not limited to biliary tract cancer, brain cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, endometrial cancer, esophageal cancer, gastric cancer, intraepithelial neoplasm, leukemia, lymphoma, liver cancer, lung cancer, melanoma, myelomas, neuroblastoma, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, sarcoma, skin cancer, testicular cancer, thyroid cancer and renal cancer.


In certain embodiments, cancers are selected from hairy cell leukemia, chronic myelogenous leukemia, cutaneous T-cell leukemia, chronic myeloid leukemia, non-Hodgkin's lymphoma, Burkitt lymphoma, multiple myeloma, follicular lymphoma, malignant melanoma, squamous cell carcinoma, renal cell carcinoma, prostate carcinoma, bladder cell carcinoma, breast carcinoma, ovarian carcinoma, non-small cell lung cancer, small cell lung cancer, hepatocellular carcinoma, basaliom, colon carcinoma, cervical dysplasia, malignant glioma and Kaposi's sarcoma (AIDS-related and non-AIDS-related).


Dermatological disorders include, but are not limited to, psoriasis, acne, rosacea, eczema, molluscum contagious, seborrheic keratosis, actinic keratosis, verruca vulgaris.


Metabolic disorders include, but are not limited to, diabetes and obesity.


Ocular diseases include, but are not limited to, age-related macular degeneration.


Neurological disorders include, but are not limited to, Alzeimer' disease, Huntington's disease, Parkinson's disease, and spinal cord injury.


Hereditary diseases include, but are not limited to, cystic fibrosis.


In one embodiment, the pharmaceutical composition is for administration selected from the group consisting of airway, oral, ocular, parenteral (including intraveneous, intradermal, intramuscular, intraperitoneal, and subcutaneous), rectal, vaginal and topical (including buccal and sublingual) administration.


In another embodiment, the pharmaceutical composition is for use in combination with one or more treatments of disorders selected from treatments for cancer, dermatological disorders, immune disorders, metabolic disorders, neurological disorders, ocular diseases, infections, and other hereditary and non-hereditary disorders in a mammal. Such treatments include, but are not limited to, surgery, chemotherapy, radiation therapy, and the administration of pharmaceutically active (or therapeutic) agents such as immunostimulatory RNA oligonucleotides, immunostimulatory DNA oligonucleotides, cytokines, chemokines, growth factors, antibiotics, anti-angiogenic factors, chemotherapeutic agents, anti-viral agents, anti-fungal agents, anti-parasitic agents, and antibodies.


In one embodiment, the pharmaceutically active agent is natural or recombinant IL-12, or a CpG-containing RNA oligonucleotide capable inducing IL-12 (see e.g., Sugiyama et al. 2005, J Immunol 174:2273-2279). In another embodiment, the pharmaceutically active agent is natural or recombinant IFN-α polypeptide, an immunostimulatory RNA oligonucleotide capable of inducing IFN-α (see e.g., our co-pending application), or a CpG-containing or non-CpG-containing DNA oligonucleotide capable of inducing IFN-α (see e.g., WO 01/22990, WO 03/101375). In yet another embodiment, the pharmaceutically active agent is natural or recombinant IL-2. In another embodiment, the pharmaceutically active agent is an anti-angiogenic factor such as vasostatin or an anti-VEGF antibody. In certain embodiments, the pharmaceutically active agent is a cancer-specific agent such as Herceptin, Rituxan, Gleevec, Iressa.


Mammals include, but are not limited to, rats, mice, cats, dogs, horses, sheep, cattle, cows, pigs, rabbits, non-human primates, and humans. In a preferred embodiment, the mammal is human.


In Vitro Method for Inducing IL-12 Production

The present invention provides an in vitro method of inducing IL-12 production in a mammalian cell, comprising the steps of:

    • (a) complexing an immunostimulatory RNA oligonucleotide of the invention with a complexation agent; and
    • (b) contacting the cell with the complex prepared in step (a).


The mammalian cell is capable of producing IL-12. In one embodiment, the mammalian cell expresses TLR8. The mammalian cells include, but are not limited to, peripheral blood mononuclear cells (PBMC), myeloid dendritic cells (MDC), B cells, macrophages, monocytes, and cells containing exogenous DNA which directs the expression of TLR7 or TLR8 or both TLR7 or TLR8 such as transfected CHO, HEK293 or COS cells.


In one embodiment of the invention, the complexation agent is a polycationic peptide, preferably poly-L-arginine (poly-L-Arg). In one embodiment, the polycationic peptide, in particular, poly-L-Arg, is at least 24 amino acids in length. The polycationic peptide, in particular, poly-L Arg, may be a heterogeneous mixture of peptides of different lengths.


In a preferred embodiment, the mammal is human.


The present invention is illustrated by the following examples.


EXAMPLES
Example 1
Poly-L-arginine Complexed ssRNA Oligonucleotides are Potent Inductors of IL-12 in PBMC

Given the fact that myeloid cells express high levels of TLR8, it is remarkable that so far little has been reported about RNA-mediated activation of cells of the myeloid lineage. We have previously shown that plasmacytoid dendritic cells are highly sensitive to both short double and single stranded RNA molecules, when cationic lipids are employed to complex respective RNA oligonucleotides. Interestingly, when we addressed the activation of cells of the myeloid lineage in the human system via cationic lipid complexed RNA-oligonucleotides, we were unable to document a significant activation. Indeed, when we used cationic lipids to transfect RNA-oligonucleotides, we observed a considerable cytotoxicity, most prominent in the population of myeloid cells (data not shown). This was in marked contrast to the use of the cationic polypetide poly-L-arginine for complexation of RNA-oligonucleotides. Poly-L-arginine complexed single stranded RNA-oligonucleotides were highly active in terms of IL-12 induction within PBMC, whereas no IL-12 was induced when the cationic lipid Lipofectamine 2000 was used to transfect ssRNA-oligonucleotides into PBMC. Other polycationic polypeptides such as poly-L-lysine and poly-L-histidine were not active in terms of IL-12 induction (FIG. 1).


Example 2
At least 24meric Polypeptides of poly-L-arginine are Required to Achieve Maximal IL-12 Induction in PBMC

In our first set of experiments we used poly-L-arginine consisting of an inhomogeneous mixture of polypetides from 5-15 kDa (approx. 44-132 aminoacids). To decipher the minimal length of poly-L-arginine required for IL-12 induction in PBMC, we compared the activity of the ˜88 mer poly-L-arginine to defined synthetic 32mer, 24mer, 16mer and 8mer polypetides. As depicted in FIG. 2, at least 24 residues of the cationic amino acid L-arginine were required to elicit a substantial IL-12 response in PBMC. Other polycationic peptides containing poly-L-arginine such as homopolymers derived from the HIV TAT peptide were also able to complex ssRNA oligonucleotides. IL-12 induction in PBMC was also detected, yet to a lesser extent (data not shown). Nevertheless, at least 24meric TAT peptides were needed to induce IL-12 in PBMC.


Example 3
Maximal IL-12 Induction is Achieved After 20 Minutes of ssRNA-Complexation with poly-L-Arg

In contrast to cationic lipids, for ssRNA-poly-L-Arg complexation little changes in concentrations of both the nucleic acid and the polycationic peptide (range: 50%-200%) had negligible impact on the activity of the complex (data not shown). Yet when we analyzed the time course of a particle formation and the corresponding immunostimulatory activity, a marked change in immunostimulation was observed. A linear increase in IL-12 production was seen with increasing complexation time. When poly-L-arginine was allowed 20 minutes to complex ssRNA oligonucleotides, a seven fold increase in IL-12 production was observed (FIG. 3). This increase in immunostimulatory activity was paralleled with an increase in particle size (FIG. 4). A plateau in immunostimulatory activity was reached after 20 minutes of complexation. Longer time for complexation did not result in higher immunostimulation (data not shown).


Example 4
Comparing poly-L-arginine ssRNA Oligonucleotides to Other TLR-Ligands

Next we compared poly-L-arginine complexed ssRNA oligonucleotides to established TLR-ligands in terms of immunostimulatory activity. For all TLR-ligands optimal concentrations were used that have been previously reported to activate PBMC. Among all established TLR-ligands only TLR8-activating compounds were able to induce IL-12p70 production within PBMC. This was true for the chimeric TLR7/TLR8 ligands R848 and 3M-007 and the TLR8-specific compound 3M-002. In addition all tested ssRNA oligonucleotides induced a marked IL-12p70 response within PBMC. Of note, whenever ssRNA oligonucleotides were tested in double-stranded conformation the IL-12 production was strongly reduced. Homopolymers of either polyA, polyC, polyG showed no activity, whereas polyU was able to elicit a slight yet consistent IL-12 response. These results indicated that ssRNA oligonucleotides harboring complex motifs rather than simple homopolymeric ssRNA oligonucleotides are are required to induce IL-12 production within PBMC.


Example 5
Maximal IL-12 Production Within PBMC Depends on the Presence of CD14+ Monocytes

We have previously shown that Lipofectamine-complexed ssRNA mediates IFN-a production only in the presence of plasmacytoid dendritic cells [10]. In analogy, when poly-L-arginine was used to complex ssRNA oligonucleotides, IFN-a production was completely abrogated in the absence of PDC (FIG. 6B). However, when PDC were depleted from PBMC no decrease in the ssRNA oligonucleotide mediated IL-12 induction was seen. Interestingly this was also true for the depletion of myeloid dendritic cells (FIG. 6A), thus indicating that within PBMC other cell types than the two major subsets of dendritic cells are responsible for the production of large amounts of IL-12p70 in response to ssRNA oligonucleotide stimulation. In order to decipher the cell type responsible for the observed IL-12 production, we employed intracellular FACS-staining for IL-12 combined with appropriate antibodies for the classical lineage marker. Using this technique, we were able to identify CD14+ monocytes as the main source of IL-12 production within PBMC (FIG. 7A). Moreover, when CD14+ cells were depleted from PBMC using MACS the ssRNA oligonucleotide mediated IL-12 response was completely abrogated (FIG. 7B). Purification of monocytes confirmed that no other cell type was required for IL-12 production in response to ssRNA oligonucleotide stimulation (FIG. 7C). Interestingly, when we compared the chimeric TLR7/TLR8 ligand R848 to ssRNA oligonucleotides in terms of IL-12 induction, we observed an almost five-fold difference in favor of the complexed ssRNA oligonucleotides.


Example 6
Single-Strand Conformation of RNA-Ligands is Required for Potent IL-12 Induction

In above described experiments we had shown that single-strand conformation of RNA oligonucleotides is necessary for maximal IL-12 induction within PBMC. Annealing of the complementary ssRNA oligonucleotide led to a complete abrogation of the IL-12 inducing activity. To address this phenomenon in more detail we performed a set of experiments in which we used shorter versions of a complementary ssRNA oligonucleotide to generate partially double-stranded ssRNA oligonucleotides. For these experiments, the ssRNA oligonucleotide 9.2antisense was used as a standard control oligonucleotide whereas ssRNA oligonucleotides that were complementary to 9.2antisense were designed to achieve partially double stranded compounds (for a detailed listing see table 1). Both within PBMC and in monocytes (FIGS. 8A and B), a marked reduction in IL-12 production was seen with increasing amounts of complementary bases. Already a segment of 12 complementary bases (9.2A/9.2antisense combination) reduced the immunostimulatory activity of 9.2anstisense to less than 20% of the original ssRNA oligonucleotide. An almost complete reduction was obtained as soon as a complete dsRNA conformation was reached. Double stranded conformation of ssRNA oligonucleotides can also be obtained when palindromic stretches are contained within a sequence by either hairpin conformation or self hybridization. To address this possibility we designed a panel of ssRNA oligonucleotides starting with a complete palindromic sequence and respective derivates with increasing amounts of mismatches (see table 2). The completely palindromic sequence and its close derivates proved to be double-stranded 20mer RNA oligonucleotides, when analyzed on a gel (data not shown). This indicated that under these conditions palindromic ssRNA oligonucleotides avidly formed dsRNA by self hybridization rather than hairpin conformation. As anticipated from above findings, palindromic RNA oligonucleotides proved to be poor in inducing IL-12 production within PBMC and monocytes (FIGS. 8C and D). In contrast, when mismatches were introduced to avoid self hybridization a strong IL-12 response could be elicited.


Example 7
Rational Design of a 4mer-Motif Library to Screen for Potent Motifs within ssRNA

Previous experiments have shown that a minimal length of 19 bases is required for maximal IL-12 induction by ssRNA oligonucleotides (see FIG. 8). Since poly adenosine oligonucleotides proved to be inactive in terms of IL-12 induction, we decided to generate a ssRNA oligonucleotide library by placing putative motifs into the centre of poly adenosine RNA oligonucleotides. We generated a library of ssRNA oligonucleotides comprising all possible 4mer motifs in the centre of a poly adenosine oligonucleotide. In view of the fact that the flanking adenosine residues can be part of a 4mer RNA sequence motif, only 193 ssRNA oligonucleotides (Table 3) were needed to cover all 256 possible 4mer motifs.


Example 8
Generation and Processing of Raw Data

All 193 ssRNA oligonucleotides were tested on PBMC of six individual healthy donors using poly-L-Arg for complexation. At 44 hours after stimulation with RNA oligonucleotides, supernatants were collected and IL-12 production was measured by ELISA. Prior to statistical analysis the raw data were processed as follows: for each cell culture plate the mean IL-12 value of the experimental duplicates for each tested ssRNA oligonucleotide were normalized to the ssRNA oligonucleotide RNA9.2sense (5′-AGCUUAACCUGUCCUUCAA-3′). This standard RNA oligonucleotide was included as a positive control on all cell culture plates. Normalization was performed by calculating the ratio of IL-12 induced by the tested oligonucleotide and IL-12 induced by the standard oligonucleotide RNA9.2sense. Thus, for each tested oligonucleotide in an individual donor a mean ratio of IL-12 induction was obtained. In the following, this mean of the ratios is referred to as IL-12 index (one value of IL-12 index per donor). For example testing ssRNA-oligonucleotide ANP175 (5′-AAAAAAAUGCUAAAAAAAA-3′) in donor 3 gave the mean of the two raw values of the duplicates (IL-12 in supernatant) of 1725 pg/ml, whereas the control oligonucleotide RNA9.2sense (5′-AGCUUAACCUGUCCUUCAA-3′) resulted in 908 pg/ml. The corresponding IL-12 index of oligonucleotide ANP175 for donor 3 was calculated to be 1.90 (=1725 pg/ml divided by 908 pg/ml).


Next, the means of all IL-12 indices for every individual donor were calculated. Then the adjusted IL-12 indices were calculated as IL-12 index minus the mean of all IL-12 indices of one individual donor. For example ssRNA oligonucleotide ANP175 of donor 3 (5′-AAAAAAAUGCUAAAAAAAA-3′) had an IL-12 index of 1.90, whereas the mean of all IL-12 indices of donor 3 was 0.44. The adjusted IL-12 index of ANP175 was calculated: 1.90 minus 0.44=1.46. The adjusted IL-12 indices from all six donors were summarized by calculating the means and the corresponding standard error of mean. The data are depicted in ascending order (FIG. 9A, B, C, D). The adjusted IL-12 indices of the top thirty ssRNA oligonucleotides of all six donors were compared using a two-tailed Student's t-test (FIG. 10). For most combinations tested, a significant difference was observed when the interval between the analyzed pairs was at least eight or nine places in the assortment.


Example 9
Calculating an Individual IL-12 Score for 1 mer-, 2mer- and 3mer-Motifs

A mean IL-12 index for all possible 1mer motifs (5′-X-3′), 2mer motifs (5′-XX-3′,5′-X*X-3′, 5′-X**X-3′) or 3mer motifs (5′-XXX-3′,5′-XX*X-3′,5′-X*XX-3′) was obtained by calculating a mean IL-12 index of all ssRNA-oligonucleotides containing the corresponding motifs. This mean IL-12 index is referred to as the IL-12 score of a given motif. For example the 3mer motif 5′-U*GU-3′ was contained in ssRNA-oligonucleotides ANP 154, 166, 178 and 190 with respective adjusted IL-12 indices of 1.57, 4.42, 3.26 and 1.6. The IL-12 score of the 3mer motif 5′-U*GU-3′ was thus calculated to be 2.71 with a standard error of mean of 0.69. The calculation of the IL-12 score of a motif did not account for the position of the motif within the sequence of the corresponding ssRNA oligonucleotides. Multiple occurrences of one motif within the same ssRNA-oligonucleotide was accounted for by adding the corresponding IL-12 index times the number of its occurrence within the oligonucleotide to the calculation of the corresponding IL-12 score of the motif. Consequently an IL-12 score could be assigned to all possible 1mer motifs (5′-X-3′), 2mer motifs (5′-XX-3′,5′-X*X-3′,5′-X**X-3′) or 3mer motifs (5′-XXX-3′,5′-XX*X-3′,5′-X*XX-3′) (FIG. 11).


Example 10
Using the IL-12 Score of 1mer-, 2mer- and 3mer-Motifs to Predict ssRNA-Oligonucleotides with a Low or High IL-12 Index

Next we tested the predictive value of the calculated 1mer-, 2mer- and 3mer-motif IL-12 scores to predict ssRNA oligonucleotides with a low or high IL-12 index. Thus for each ssRNA oligonucleotide the occurrence of a set of motifs was tested and the respective IL-12 scores were assigned to the ssRNA. For example for the panel of 3mer motifs with unspaced sequences (5′-XXX-3′) the ssRNA-oligonucleotide ANP 175 (5′-AAAAAAAAUGGUAAAAAAA-3′) was analyzed the following way:

















Occurrences





within the ssRNA


3mer motif

oligonucleotide
Assigned IL-12


(5′-XXX-3′)
IL-12 score
ANP 175
score


















5′-AAA-3′
−0.0117
11
−0.1287


5′-UAA-3′
+0.3798
1
+0.3798


5′-AAU-3′
+0.4008
1
+0.4008


5′-AUG-3′
+0.7997
1
+0.7997


5′-UGG-3′
+0.8403
1
+0.8403


5′-GUA-3′
+0.9657
1
+0.9657


5′-GGU-3′
+1.0603
1
+1.0603





+4.3180









All ssRNA oligonucleotides were assigned an individual IL-12 score for all possible motif-combinations (1mer-, 2mer- and 3mer-motifs). Next, the prediction that was obtained by using the assigned IL-12 scores were compared to the actual adjusted IL-12 indices for all ssRNA-oligonucleotides and for each motif combination. Data were sorted in ascending order according to the adjusted IL-12 indices. For all predictions, the correlation coefficient was calculated: Using the IL-12 scores of 1mer motifs (5′-X-3′) to predict the actual adjusted IL-12 indices off all ssRNA oligonucleotides a correlation coefficient of 0.6 was obtained (FIG. 12A). When 2mer motifs were used to predict the adjusted IL-12 indices a correlation coefficient of 0.70 was obtained for 5′-XX-3′-motifs, a correlation coefficient of 0.65 for 5′-X*X-3′-motifs and a correlation coefficient of 0.65 for 5′-X**X-3′-motifs (FIG. 12B). Using 3mer motifs to predict the adjusted IL-12 indices, a correlation coefficient of 0.76 was calculated for 5′-XXX-3′-motifs, of 0.75 for 5′-XX*X-3′-motifs and of 0.76 for 5′-X*XX-3′-motifs. When only the IL-12 scores of 3mer motifs were considered that were significantly higher or lower than the IL-12 scores of the other motifs (see FIG. 11C) a correlation coefficient of 0.78 could be obtained (correlations for prediction with different types of motifs see FIG. 12).


Example 11
Determined the Threshold IL-12 Scores for High and Low Immunostimulatory Activity

The immunostimulatory activity of any given RNA oligonucleotide can be predicted using the 3mer-based algorithm as described previously. For research and drug discovery and development purposes, two groups of RNA oligonucleotide are of interest: Group A oligonucleotides which have high or maximal IL-12-inducing activity, and Group B oligonucleotides which have low or minimal IL-12-inducing activity. Among all possible ssRNA oligonucleotides of a certain length, 1% of the oligonucleotides with the highest IL-12 scores are assigned to Group A; where as 1% of the oligonucleotides with the lowest IL-12 scores are assigned to Group B. The cut-off IL-12 score for Group A oligonucleotide is the threshold for high or maximal immunostimulatory activity; the cut-off IL-12 score for Group B oligonucleotide is the threshold for low or minimal immunostimulatory activity.


The IL-12 score thresholds for high/maximal and low/minimal immunostimulatory activity for 19mer ssRNA oligonucleotides are determined as follows:


A pool of all possible sequences of 19mer RNA oligonucleotides consists of 419=274,877,906,944 oligonucleotides. The IFN-α score for every single RNA oligonucleotide in the pool is calculated using the 3mer-based algorithm. All 419 oligonucleotides are ranked based on their calculated predicted IL-12 scores. The threshold for group A is determined to be





8.4064×n+66.958

    • (n=length of the ssRNA oligonucleotide, and n>9).


All ssRNA oligonucleotides with a calculated IL-12 score above the threshold value are grouped into Group A. The Group A threshold for 19mer ssRNA oligonucleotides is 226.68. Non-limiting examples of Group A 19mer ssRNA oligonucleotides include the following:
















Sequence (5′-->3′)
Predicted IL-12 score




















UUUAUAAGUUUGCUGGUGC
243








UUGUUUGUAUGGCUAUCCG
231







UUGCUCUCGUGUGUGGUAU
263







UUGCGUUGUUGGAGUGGUC
310







UGUGUUGUUGGCUCUACAA
246







UGUAGUGGUCGGUGGCCCC
261







UGUAGUGAAGUUGUGUCCG
266







UGAGUUGGUGGACUGUUUG
262







UCUCGUUGGUUACGUUACU
237







UAUGGUGUUUCGUAUAUGU
283










The threshold for Group B oligonucleotides is determined to be:





0.0468×n2−2.3103×n−23.244

    • (n=length of the ssRNA oligonucleotide, and X>9)


All ssRNA oligonucleotides with a calculated IL-12 score below the threshold value are grouped into Group B. The Group B threshold for 19mer ssRNA oligonucleotide is −50.2449. Non-limiting examples of Group B 19mer oligonucleotide include the following:
















Sequence (5′-->3′)
Predicted IL-12 score




















GACCCAACACACACGGGCC
−101








AAAACAAAACGGAACCCAG
−91







AAAAACCAACCCAAGAUCU
−81







UUAGGGCAAACCACCAGAC
−73







CAGACCAACGGAACGCGCA
−72







CGGCCCACCAACCCGGACU
−70







CCCAAGAGAGACGAAACGC
−70







UACCACAGGCCCAAACGGC
−68







AUCCGAGAAACUACCACCA
−68







CCCAAUAACACAAAGCCUA
−67










For ssRNA oligonucleotides between 3 and 9 nucleotides in length, the Group A and Group B threshold values are given below in Table 6:









TABLE 6







Threshold IL-12 scores for Group A and Group


B oligonucleotides 3-9 nucleotides in length.









The predicted IL-12 score


ssRNA
(using the IL12 point score matrix)









oligonucleotide
threshold for GROUP B
threshold for GROUP A


length
ssRNA oligonucelotides
ssRNA oligonucelotides












3
−5
13


4
−15
50


5
−19
66


6
−28
93.53


7
−31
107.77


8
−34
121


9
−36
121.98









REFERENCES



  • 1 Schroder, M. and Bowie, A. G., TLR3 in antiviral immunity: key player or bystander? Trends Immunol 2005.

  • 2 Hemmi, H., Takeuchi, O., Kawai, T., Kaisho, T., Sato, S., Sanjo, H., Matsumoto, M., Hoshino, K., Wagner, H., Takeda, K. and Akira, S., A Toll-like receptor recognizes bacterial DNA. Nature 2000. 408: 740-745.

  • 3 Bauer, S., Kirschning, C. J., Hacker, H., Redecke, V., Hausmann, S., Akira, S., Wagner, H. and Lipford, G. B., Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. Proc Natl Acad Sci USA 2001. 98: 9237-9242.

  • 4 Krieg, A. M., Yi, A. K., Matson, S., Waldschmidt, T. J., Bishop, G. A., Teasdale, R., Koretzky, G. A. and Klinman, D. M., CpG motifs in bacterial DNA trigger direct B-cell activation. Nature 1995. 374: 546-549.

  • 5 Heckelsmiller, K., Beck, S., Rall, K., Sipos, B., Schlamp, A., Tuma, E., Rothenfusser, S., Endres, S, and Hartmann, G., Combined dendritic cell- and CpG oligonucleotide-based immune therapy cures large murine tumors that resist chemotherapy. Eur J Immunol 2002. 32: 3235-3245.

  • 6 Heckelsmiller, K., Rall, K., Beck, S., Schlamp, A., Seiderer, J., Jahrsdorfer, B., Krug, A., Rothenfusser, S., Endres, S, and Hartmann, G., Peritumoral CpG DNA elicits a coordinated response of CD8 T cells and innate effectors to cure established tumors in a murine colon carcinoma model. J Immunol 2002. 169: 3892-3899.

  • 7 Krug, A., Towarowski, A., Britsch, S., Rothenfusser, S., Hornung, V., Bals, R., Giese, T., Engelmann, H., Endres, S., Krieg, A. M. and Hartmann, G., Toll-like receptor expression reveals CpG DNA as a unique microbial stimulus for plasmacytoid dendritic cells which synergizes with CD40 ligand to induce high amounts of IL-12. Eur J Immunol 2001. 31: 3026-3037.

  • 8 Hornung, V., Rothenfusser, S., Britsch, S., Krug, A., Jahrsdorfer, B., Giese, T., Endres, S, and Hartmann, G., Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J Immunol 2002. 168: 4531-4537.

  • 9 Choe, J., Kelker, M. S, and Wilson, I. A., Crystal structure of human toll-like receptor 3 (TLR3) ectodomain. Science 2005. 309: 581-585.

  • 10 Hornung, V., Guenthner-Biller, M., Bourquin, C., Ablasser, A., Schlee, M., Uematsu, S., Noronha, A., Manoharan, M., Akira, S., de Fougerolles, A., Endres, S, and Hartmann, G., Sequence-specific potent induction of IFN-alpha by short interfering RNA in plasmacytoid dendritic cells through TLR7. Nat Med 2005. 11: 263-270.

  • 11 Heil, F., Hemmi, H., Hochrein, H., Ampenberger, F., Kirschning, C., Akira, S., Lipford, G., Wagner, H. and Bauer, S., Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 2004. 303: 1526-1529.

  • 12 Diebold, S. S., Kaisho, T., Hemmi, H., Akira, S, and Reis e Sousa, C., Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 2004. 303: 1529-1531.

  • 13 Hartmann, G., Weeratna, R. D., Ballas, Z. K., Payette, P., Blackwell, S., Suparto, I., Rasmussen, W. L., Waldschmidt, M., Sajuthi, D., Purcell, R. H., Davis, H. L. and Krieg, A. M., Delineation of a CpG phosphorothioate oligodeoxynucleotide for activating primate immune responses in vitro and in vivo. J Immunol 2000. 164: 1617-1624.

  • 14 Hartmann, G. and Krieg, A. M., Mechanism and function of a newly identified CpG DNA motif in human primary B cells. J Immunol 2000. 164: 944-953.

  • 15 Gorden, K. B., Gorski, K. S., Gibson, S. J., Kedl, R. M., Kieper, W. C., Qiu, X., Tomai, M. A., Alkan, S. S, and Vasilakos, J. P., Synthetic TLR agonists reveal functional differences between human TLR7 and TLR8. J Immunol 2005. 174: 1259-1268.

  • 16 Sugiyama, T., Gursel, M., Takeshita, F., Coban, C., Conover, J., Kaisho, T., Akira, S., Klinman, D. M. and Ishii, K. J., CpG RNA: identification of novel single-stranded RNA that stimulates human CD14+CD11c+ monocytes. J Immunol 2005.174: 2273-2279.

  • 17 Judge, A. D., Sood, V., Shaw, J. R., Fang, D., McClintock, K. and MacLachlan, I., Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol 2005. 23: 457-462.

  • 18 Sioud, M., Induction of inflammatory cytokines and interferon responses by double-stranded and single-stranded siRNAs is sequence-dependent and requires endosomal localization. J Mol Biol 2005. 348: 1079-1090.

  • 19 Barchet, W., Krug, A., Cella, M., Newby, C., Fischer, J. A., Dzionek, A., Pekosz, A. and Colonna, M., Dendritic cells respond to influenza virus through TLR7- and PKR-independent pathways. Eur J Immunol 2005. 35: 236-242.

  • 20 Koski, G. K., Kariko, K., Xu, S., Weissman, D., Cohen, P. A. and Czerniecki, B. J., Cutting edge: innate immune system discriminates between RNA containing bacterial versus eukaryotic structural features that prime for high-level IL-12 secretion by dendritic cells. J Immunol 2004.172: 3989-3993.

  • 21 Scheel, B., Teufel, R., Probst, J., Carralot, J. P., Geginat, J., Radsak, M., Jarrossay, D., Wagner, H., Jung, G., Rammensee, H. G., Hoerr, I. and Pascolo, S., Toll-like receptor-dependent activation of several human blood cell types by protamine-condensed mRNA. Eur J Immunol 2005. 35: 1557-1566.

  • 22 Scheel, B., Braedel, S., Probst, J., Carralot, J. P., Wagner, H., Schild, H., Jung, G., Rammensee, H. G. and Pascolo, S., Immunostimulating capacities of stabilized RNA molecules. Eur J Immunol 2004. 34: 537-547.

  • 23 Rudolph, C., Plank, C., Lausier, J., Schillinger, U., Muller, R. H. and Rosenecker, J., Oligomers of the arginine-rich motif of the HIV-1 TAT protein are capable of transferring plasmid DNA into cells. J Biol Chem 2003. 278: 11411-11418.

  • 24 Fuchs, S. M. and Raines, R. T., Pathway for polyarginine entry into mammalian cells. Biochemistry 2004. 43: 2438-2444.

  • 25 Lochmann, D., Jauk, E. and Zimmer, A., Drug delivery of oligonucleotides by peptides. Eur J Pharm Biopharm 2004. 58: 237-251.










TABLE 1





Partially annealed ssRNA oligonucleotides
















9.2sense
5′-AGCUUAACCUGUCCUUCAA-3′


9.2antisense
5′-UUGAAGGACAGGUUAAGCU-3′





9.2duplex










9.2R
5′-AGCUUAACCUGUCCUU-3′





9.2R + 9.2antisense










9.2L
5′-UUAACCUGUCCUUCAA-3′





9.2L + 9.2antisense










9.2A
5′ -AGCUUAACCUGU-   3′





9.2A + 9.2as










9.2B
5′-ACCUGUCCUUCA-   3′





9.2B + 9.2as










Complete base pairing is indicated by normal letters, whereas mismatches are indicated by underlined letters.













TABLE 2





Self complementary ssRNA oligonucleotides
















RNA-P20










RNA-P20-1M










RNA-P20-2M










RNA-P20-3′M










RNA-P20-5′M










RNA-P20-6M










RNA-M20










Complete base pairing is indicated by normal letters, whereas mismatches are indicated by underlined letters.














TABLE 3







193 ssRNA oligonucleotide library covering all



possible 256 4mer motifs










Name
Sequence















ANP-Oligo 001
AAAAAAAAAAAAAAAAAAA








ANP-Oligo 002
AAAAAAAACAAAAAAAAAA







ANP-Oligo 003
AAAAAAAAGAAAAAAAAAA







ANP-Oligo 004
AAAAAAAAUAAAAAAAAAA







ANP-Oligo 005
AAAAAAAACCAAAAAAAAA







ANP-Oligo 006
AAAAAAAACGAAAAAAAAA







ANP-Oligo 007
AAAAAAAACUAAAAAAAAA







ANP-Oligo 008
AAAAAAAAGCAAAAAAAAA







ANP-Oligo 009
AAAAAAAAGGAAAAAAAAA







ANP-Oligo 010
AAAAAAAAGUAAAAAAAAA







ANP-Oligo 011
AAAAAAAAUCAAAAAAAAA







ANP-Oligo 012
AAAAAAAAUGAAAAAAAAA







ANP-Oligo 013
AAAAAAAAUUAAAAAAAAA







ANP-Oligo 014
AAAAAAAACACAAAAAAAA







ANP-Oligo 015
AAAAAAAACAGAAAAAAAA







ANP-Oligo 016
AAAAAAAACAUAAAAAAAA







ANP-Oligo 017
AAAAAAAACCCAAAAAAAA







ANP-Oligo 018
AAAAAAAACCGAAAAAAAA







ANP-Oligo 019
AAAAAAAACCUAAAAAAAA







ANP-Oligo 020
AAAAAAAACGCAAAAAAAA







ANP-Oligo 021
AAAAAAAACGGAAAAAAAA







ANP-Oligo 022
AAAAAAAACGUAAAAAAAA







ANP-Oligo 023
AAAAAAAACUCAAAAAAAA







ANP-Oligo 024
AAAAAAAACUGAAAAAAAA







ANP-Oligo 025
AAAAAAAACUUAAAAAAAA







ANP-Oligo 026
AAAAAAAAGACAAAAAAAA







ANP-Oligo 027
AAAAAAAAGAGAAAAAAAA







ANP-Oligo 028
AAAAAAAAGAUAAAAAAAA







ANP-Oligo 029
AAAAAAAAGCCAAAAAAAA







ANP-Oligo 030
AAAAAAAAGCGAAAAAAAA







ANP-Oligo 031
AAAAAAAAGCUAAAAAAAA







ANP-Oligo 032
AAAAAAAAGGCAAAAAAAA







ANP-Oligo 033
AAAAAAAAGGGAAAAAAAA







ANP-Oligo 034
AAAAAAAAGGUAAAAAAAA







ANP-Oligo 035
AAAAAAAAGUCAAAAAAAA







ANP-Oligo 036
AAAAAAAAGUGAAAAAAAA







ANP-Oligo 037
AAAAAAAAGUUAAAAAAAA







ANP-Oligo 038
AAAAAAAAUACAAAAAAAA







ANP-Oligo 039
AAAAAAAAUAGAAAAAAAA







ANP-Oligo 040
AAAAAAAAUAUAAAAAAAA







ANP-Oligo 041
AAAAAAAAUCCAAAAAAAA







ANP-Oligo 042
AAAAAAAAUCGAAAAAAAA







ANP-Oligo 043
AAAAAAAAUCUAAAAAAAA







ANP-Oligo 044
AAAAAAAAUGCAAAAAAAA







ANP-Oligo 045
AAAAAAAAUGGAAAAAAAA







ANP-Oligo 046
AAAAAAAAUGUAAAAAAAA







ANP-Oligo 047
AAAAAAAAUUCAAAAAAAA







ANP-Oligo 048
AAAAAAAAUUGAAAAAAAA







ANP-Oligo 049
AAAAAAAAUUUAAAAAAAA







ANP-Oligo 050
AAAAAAACAACAAAAAAAA







ANP-Oligo 051
AAAAAAACAAGAAAAAAAA







ANP-Oligo 052
AAAAAAACAAUAAAAAAAA







ANP-Oligo 053
AAAAAAACACCAAAAAAAA







ANP-Oligo 054
AAAAAAACACGAAAAAAAA







ANP-Oligo 055
AAAAAAACACUAAAAAAAA







ANP-Oligo 056
AAAAAAACAGCAAAAAAAA







ANP-Oligo 057
AAAAAAACAGGAAAAAAAA







ANP-Oligo 058
AAAAAAACAGUAAAAAAAA







ANP-Oligo 059
AAAAAAACAUCAAAAAAAA







ANP-Oligo 060
AAAAAAACAUGAAAAAAAA







ANP-Oligo 061
AAAAAAACAUUAAAAAAAA







ANP-Oligo 062
AAAAAAACCACAAAAAAAA







ANP-Oligo 063
AAAAAAACCAGAAAAAAAA







ANP-Oligo 064
AAAAAAACCAUAAAAAAAA







ANP-Oligo 065
AAAAAAACCCCAAAAAAAA







ANP-Oligo 066
AAAAAAACCCGAAAAAAAA







ANP-Oligo 067
AAAAAAACCCUAAAAAAAA







ANP-Oligo 068
AAAAAAACCGCAAAAA4AA







ANP-Oligo 069
AAAAAAACCGGAAAAAAAA







ANP-Oligo 070
AAAAAAACCGUAAAAAAAA







ANP-Oligo 071
AAAAAAACCUCAAAAAAAA







ANP-Oligo 072
AAAAAAACCUGAAAAAAAA







ANP-Oligo 073
AAAAAAACCUUAAAAAAAA







ANP-Oligo 074
AAAAAAACGACAAAAAAAA







ANP-Oligo 075
AAAAAAACGAGAAAAAAAA







ANP-Oligo 076
AAAAAAACGAUAAAAAAAA







ANP-Oligo 077
AAAAAAACGCCAAAAAAAA







ANP-Oligo 078
AAAAAAACGCGAAAAAAAA







ANP-Oligo 079
AAAAAAACGCUAAAAAAAA







ANP-Oligo 080
AAAAAAACGGCAAAAAAAA







ANP-Oligo 081
AAAAAAACGGGAAAAAAAA







ANP-Oligo 082
AAAAAAACGGUAAAAAAAA







ANP-Oligo 083
AAAAAAACGUCAAAAAAAA







ANP-Oligo 084
AAAAAAACGUGAAAAAAAA







ANP-Oligo 085
AAAAAAACGUUAAAAAAAA







ANP-Oligo 086
AAAAAAACUACAAAAAAAA







ANP-Oligo 087
AAAAAAACUAGAAAAAAAA







ANP-Oligo 088
AAAAAAACUAUAAAAAAAA







ANP-Oligo 089
AAAAAAACUCCAAAAAAAA







ANP-Oligo 090
AAAAAAACUCGAAAAAAAA







ANP-Oligo 091
AAAAAAACUCUAAAAAAAA







ANP-Oligo 092
AAAAAAACUGCAAAAAAAA







ANP-Oligo 093
AAAAAAACUGGAAAAAAAA







ANP-Oligo 094
AAAAAAACUGUAAAAAAAA







ANP-Oligo 095
AAAAAAACUUCAAAAAAAA







ANP-Oligo 096
AAAAAAACUUGAAAAAAAA







ANP-Oligo 097
AAAAAAACUUUAAAAAAAA







ANP-Oligo 098
AAAAAAAGAACAAAAAAAA







ANP-Oligo 099
AAAAAAAGAAGAAAAAAAA







ANP-Oligo 100
AAAAAAAGAAUAAAAAAAA







ANP-Oligo 101
AAAAAAAGACCAAAAAAAA







ANP-Oligo 102
AAAAAAAGACGAAAAAAAA







ANP-Oligo 103
AAAAAAAGACUAAAAAAAA







ANP-Oligo 104
AAAAAAAGAGCAAAAAAAA







ANP-Oligo 105
AAAAAAAGAGGAAAAAAAA







ANP-Oligo 106
AAAAAAAGAGUAAAAAAAA







ANP-Oligo 107
AAAAAAAGAUCAAAAAAAA







ANP-Oligo 108
AAAAAAAGAUGAAAAAAAA







ANP-Oligo 109
AAAAAAAGAUUAAAAAAAA







ANP-Oligo 110
AAAAAAAGCACAAAAAAAA







ANP-Oligo 111
AAAAAAAGCAGAAAAAAAA







ANP-Oligo 112
AAAAAAAGCAUAAAAAAAA







ANP-Oligo 113
AAAAAAAGCCCAAAAAAAA







ANP-Oligo 114
AAAAAAAGCCGAAAAAAAA







ANP-Oligo 115
AAAAAAAGCCUAAAAAAAA







ANP-Oligo 116
AAAAAAAGCGCAAAAAAAA







ANP-Oligo 117
AAAAAAAGCGGAAAAAAAA







ANP-Oligo 118
AAAAAAAGCGUAAAAAAAA







ANP-Oligo 119
AAAAAAAGCUCAAAAAAAA







ANP-Oligo 120
AAAAAAAGCUGAAAAAAAA







ANP-Oligo 121
AAAAAAAGCUUAAAAAAAA







ANP-Oligo 122
AAAAAAAGGACAAAAAAAA







ANP-Oligo 123
AAAAAAAGGAGAAAAAAAA







ANP-Oligo 124
AAAAAAAGGAUAAAAAAAA







ANP-Oligo 125
AAAAAAAGGCCAAAAAAAA







ANP-Oligo 126
AAAAAAAGGCGAAAAAAAA







ANP-Oligo 127
AAAAAAAGGCUAAAAAAAA







ANP-Oligo 128
AAAAAAAGGGCAAAAAAAA







ANP-Oligo 129
AAAAAAAGGGGAAAAAAAA







ANP-Oligo 130
AAAAAAAGGGUAAAAAAAA







ANP-Oligo 131
AAAAAAAGGUCAAAAAAAA







ANP-Oligo 132
AAAAAAAGGUGAAAAAAAA







ANP-Oligo 133
AAAAAAAGGUUAAAAAAAA







ANP-Oligo 134
AAAAAAAGUACAAAAAAAA







ANP-Oligo 135
AAAAAAAGUAGAAAAAAAA







ANP-Oligo 136
AAAAAAAGUAUAAAAAAAA







ANP-Oligo 137
AAAAAAAGUCCAAAAAAAA







ANP-Oligo 138
AAAAAAAGUCGAAAAAAAA







ANP-Oligo 139
AAAAAAAGUCUAAAAAAAA







ANP-Oligo 140
AAAAAAAGUGCAAAAAAAA







ANP-Oligo 141
AAAAAAAGUGGAAAAAAAA







ANP-Oligo 142
AAAAAAAGUGUAAAAAAAA







ANP-Oligo 143
AAAAAAAGUUCAAAAAAAA







ANP-Oligo 144
AAAAAAAGUUGAAAAAAAA







ANP-Oligo 145
AAAAAAAGUUUAAAAAAAA







ANP-Oligo 146
AAAAAAAUAACAAAAAAAA







ANP-Oligo 147
AAAAAAAUAAGAAAAAAAA







ANP-Oligo 148
AAAAAAAUAAUAAAAAAAA







ANP-Oligo 149
AAAAAAAUACCAAAAAAAA







ANP-Oligo 150
AAAAAAAUACGAAAAAAAA







ANP-Oligo 151
AAAAAAAUACUAAAAAAAA







ANP-Oligo 152
AAAAAAAUAGCAAAAAAAA







ANP-Oligo 153
AAAAAAAUAGGAAAAAAAA







ANP-Oligo 154
AAAAAAAUAGUAAAAAAAA







ANP-Oligo 155
AAAAAAAUAUCAAAAAAAA







ANP-Oligo 156
AAAAAAAUAUGAAAAAAAA







ANP-Oligo 157
AAAAAAAUAUUAAAAAAAA







ANP-Oligo 158
AAAAAAAUCACAAAAAAAA







ANP-Oligo 159
AAAAAAAUCAGAAAAAAAA







ANP-Oligo 160
AAAAAAAUCAUAAAAAAAA







ANP-Oligo 161
AAAAAAAUCCCAAAAAAAA







ANP-Oligo 162
AAAAAAAUCCGAAAAAAAA







ANP-Oligo 163
AAAAAAAUCCUAAAAAAAA







ANP-Oligo 164
AAAAAAAUCGCAAAAAAAA







ANP-Oligo 165
AAAAAAAUCGGAAAAAAAA







ANP-Oligo 166
AAAAAAAUCGUAAAAAAAA







ANP-Oligo 167
AAAAAAAUCUCAAAAAAAA







ANP-Oligo 168
AAAAAAAUCUGAAAAAAAA







ANP-Oligo 169
AAAAAAAUCUUAAAAAAAA







ANP-Oligo 170
AAAAAAAUGACAAAAAAAA







ANP-Oligo 171
AAAAAAAUGAGAAAAAAAA







ANP-Oligo 172
AAAAAAAUGAUAAAAAAAA







ANP-Oligo 173
AAAAAAAUGCCAAAAAAAA







ANP-Oligo 174
AAAAAAAUGCGAAAAAAAA







ANP-Oligo 175
AAAAAAAUGCUAAAAAAAA







ANP-Oligo 176
AAAAAAAUGGCAAAAAAAA







ANP-Oligo 177
AAAAAAAUGGGAAAAAAAA







ANP-Oligo 178
AAAAAAAUGGUAAAAAAAA







ANP-Oligo 179
AAAAAAAUGUCAAAAAAAA







ANP-Oligo 180
AAAAAAAUGUGAAAAAAAA







ANP-Oligo 181
AAAAAAAUGUUAAAAAAAA







ANP-Oligo 182
AAAAAAAUUACAAAAAAAA







ANP-Oligo 183
AAAAAAAUUAGAAAAAAAA







ANP-Oligo 184
AAAAAAAUUAUAAAAAAAA







ANP-Oligo 185
AAAAAAAUUCCAAAAAAAA







ANP-Oligo 186
AAAAAAAUUCGAAAAAAAA







ANP-Oligo 187
AAAAAAAUUCUAAAAAAAA







ANP-Oligo 188
AAAAAAAUUGCAAAAAAAA







ANP-Oligo 189
AAAAAAAUUGGAAAAAAAA







ANP-Oligo 190
AAAAAAAUUGUAAAAAAAA







ANP-Oligo 191
AAAAAAAUUUCAAAAAAAA







ANP-Oligo 192
AAAAAAAUUUGAAAAAAAA







ANP-Oligo 193
AAAAAAAUUUUAAAAAAAA


















TABLE 4







Adjusted IL-12 indices of the 193 ssRNA



oligonucleotide library














IL-12





Adjusted
index


Name
Sequence
Mean
SEM














ANP-Oligo 018
AAAAAAAACCGAAAAAAAA
−0.68
0.12






ANP-Oligo 053
AAAAAAACACCAAAAAAAA
−0.67
0.11





ANP-Oligo 020
AAAAAAAACGCAAAAAAAA
−0.66
0.12





ANP-Oligo 051
AAAAAAACAAGAAAAAAAA
−0.66
0.11





ANP-Oligo 062
AAAAAAACCACAAAAAAAA
−0.66
0.11





ANP-Oligo 029
AAAAAAAAGCCAAAAAAAA
−0.66
0.11





ANP-Oligo 009
AAAAAAAAGGAAAAAAAAA
−0.66
0.12





ANP-Oligo 017
AAAAAAAACCCAAAAAAAA
−0.66
0.12





ANP-Oligo 075
AAAAAAACGAGAAAAAAAA
−0.65
0.11





ANP-Oligo 015
AAAAAAAACAGAAAAAAAA
−0.65
0.12





ANP-Oligo 054
AAAAAAACACGAAAAAAAA
−0.65
0.11





ANP-Oligo 080
AAAAAAACGGCAAAAAAAA
−0.65
0.11





ANP-Oligo 102
AAAAAAAGACGAAAAAAAA
−0.64
0.09





ANP-Oligo 042
AAAAAAAAUCGAAAAAAAA
−0.64
0.1





ANP-Oligo 050
AAAAAAACAACAAAAAAAA
−0.64
0.11





ANP-Oligo 069
AAAAAAACCGGAAAAAAAA
−0.64
0.1





ANP-Oligo 128
AAAAAAAGGGCAAAAAAAA
−0.64
0.1





ANP-Oligo 014
AAAAAAAACACAAAAAAAA
−0.64
0.11





ANP-Oligo 021
AAAAAAAACGGAAAAAAAA
−0.63
0.11





ANP-Oligo 003
AAAAAAAAGAAAAAAAAAA
−0.63
0.11





ANP-Oligo 117
AAAAAAAGCGGAAAAAAAA
−0.63
0.09





ANP-Oligo 006
AAAAAAAACGAAAAAAAAA
−0.63
0.12





ANP-Oligo 081
AAAAAAACGGGAAAAAAAA
−0.63
0.1





ANP-Oligo 105
AAAAAAAGAGGAAAAAAAA
−0.62
0.09





ANP-Oligo 066
AAAAAAACCCGAAAAAAAA
−0.62
0.11





ANP-Oligo 027
AAAAAAAAGAGAAAAAAAA
−0.62
0.1





ANP-Oligo 032
AAAAAAAAGGCAAAAAAAA
−0.62
0.1





ANP-Oligo 077
AAAAAAACGCCAAAAAAAA
−0.62
0.1





ANP-Oligo 005
AAAAAAAACCAAAAAAAAA
−0.62
0.12





ANP-Oligo 104
AAAAAAAGAGCAAAAAAAA
−0.61
0.08





ANP-Oligo 099
AAAAAAAGAAGAAAAAAAA
−0.61
0.1





ANP-Oligo 008
AAAAAAAAGCAAAAAAAAA
−0.61
0.12





ANP-Oligo 057
AAAAAAACAGGAAAAAAAA
−0.6
0.1





ANP-Oligo 101
AAAAAAAGACCAAAAAAAA
−0.6
0.08





ANP-Oligo 026
AAAAAAAAGACAAAAAAAA
−0.6
0.1





ANP-Oligo 074
AAAAAAACGACAAAAAAAA
−0.6
0.09





ANP-Oligo 098
AAAAAAAGAACAAAAAAAA
−0.6
0.1





ANP-Oligo 030
AAAAAAAAGCGAAAAAAAA
−0.6
0.1





ANP-Oligo 033
AAAAAAAAGGGAAAAAAAA
−0.59
0.09





ANP-Oligo 065
AAAAAAACCCCAAAAAAAA
−0.59
0.1





ANP-Oligo 078
AAAAAAACGCGAAAAAAAA
−0.59
0.1





ANP-Oligo 114
AAAAAAAGCCGAAAAAAAA
−0.58
0.08





ANP-Oligo 116
AAAAAAAGCGCAAAAAAAA
−0.58
0.07





ANP-Oligo 110
AAAAAAAGCACAAAAAAAA
−0.58
0.09





ANP-Oligo 002
AAAAAAAACAAAAAAAAAA
−0.57
0.12





ANP-Oligo 063
AAAAAAACCAGAAAAAAAA
−0.57
0.09





ANP-Oligo 068
AAAAAAACCGCAAAAAAAA
−0.56
0.08





ANP-Oligo 007
AAAAAAAACUAAAAAAAAA
−0.56
0.11





ANP-Oligo 071
AAAAAAACCUCAAAAAAAA
−0.56
0.09





ANP-Oligo 056
AAAAAAACAGCAAAAAAAA
−0.54
0.11





ANP-Oligo 126
AAAAAAAGGCGAAAAAAAA
−0.54
0.07





ANP-Oligo 125
AAAAAAAGGCCAAAAAAAA
−0.54
0.08





ANP-Oligo 113
AAAAAAAGCCCAAAAAAAA
−0.54
0.06





ANP-Oligo 090
AAAAAAACUCGAAAAAAAA
−0.54
0.07





ANP-Oligo 055
AAAAAAACACUAAAAAAAA
−0.53
0.11





ANP-Oligo 123
AAAAAAAGGAGAAAAAAAA
−0.51
0.08





ANP-Oligo 019
AAAAAAAACCUAAAAAAAA
−0.51
0.14





ANP-Oligo 129
AAAAAAAGGGGAAAAAAAA
−0.51
0.08





ANP-Oligo 072
AAAAAAACCUGAAAAAAAA
−0.51
0.07





ANP-Oligo 162
AAAAAAAUCCGAAAAAAAA
−0.5
0.07





ANP-Oligo 119
AAAAAAAGCUCAAAAAAAA
−0.48
0.07





ANP-Oligo 001
AAAAAAAAAAAAAAAAAAA
−0.48
0.12





ANP-Oligo 059
AAAAAAACAUCAAAAAAAA
−0.48
0.07





ANP-Oligo 024
AAAAAAAACUGAAAAAAAA
−0.48
0.1





ANP-Oligo 041
AAAAAAAAUCCAAAAAAAA
−0.48
0.15





ANP-Oligo 086
AAAAAAACUACAAAAAAAA
−0.46
0.07





ANP-Oligo 060
AAAAAAACUAGAAAAAAAA
−0.46
0.08





ANP-Oligo 047
AAAAAAAAUUCAAAAAAAA
−0.45
0.13





ANP-Oligo 130
AAAAAAAGGGUAAAAAAAA
−0.44
0.08





ANP-Oligo 122
AAAAAAAGGACAAAAAAAA
−0.43
0.06





ANP-Oligo 011
AAAAAAAAUCAAAAAAAAA
−0.43
0.1





ANP-Oligo 161
AAAAAAAUCCCAAAAAAAA
−0.42
0.05





ANP-Oligo 089
AAAAAAACUCCAAAAAAAA
−0.42
0.07





ANP-Oligo 023
AAAAAAAACUCAAAAAAAA
−0.41
0.06





ANP-Oligo 004
AAAAAAAAUAAAAAAAAAA
−0.4
0.12





ANP-Oligo 158
AAAAAAAUCACAAAAAAAA
−0.39
0.09





ANP-Oligo 095
AAAAAAACUUCAAAAAAAA
−0.39
0.07





ANP-Oligo 120
AAAAAAAGCUGAAAAAAAA
−0.38
0.04





ANP-Oligo 064
AAAAAAACCAUAAAAAAAA
−0.38
0.09





ANP-Oligo 052
AAAAAAACAAUAAAAAAAA
−0.37
0.12





ANP-Oligo 016
AAAAAAAACAUAAAAAAAA
−0.37
0.14





ANP-Oligo 177
AAAAAAAUGGGAAAAAAAA
−0.35
0.08





ANP-Oligo 048
AAAAAAAAUUGAAAAAAAA
−0.35
0.1





ANP-Oligo 091
AAAAAAACUCUAAAAAAAA
−0.33
0.07





ANP-Oligo 038
AAAAAAAAUACAAAAAAAA
−0.32
0.13





ANP-Oligo 028
AAAAAAAAGAUAAAAAAAA
−0.32
0.08





ANP-Oligo 111
AAAAAAAGCAGAAAAAAAA
−0.32
0.12





ANP-Oligo 031
AAAAAAAAGCUAAAAAAAA
−0.32
0.12





ANP-Oligo 093
AAAAAAACUGGAAAAAAAA
−0.31
0.04





ANP-Oligo 025
AAAAAAAACUUAAAAAAAA
−0.31
0.08





ANP-Oligo 150
AAAAAAAUACGAAAAAAAA
−0.3
0.12





ANP-Oligo 067
AAAAAAACCCUAAAAAAAA
−0.3
0.1





ANP-Oligo 045
AAAAAAAAUGGAAAAAAAA
−0.29
0.13





ANP-Oligo 159
AAAAAAAUCAGAAAAAAAA
−0.28
0.09





ANP-Oligo 073
AAAAAAACCUUAAAAAAAA
−0.28
0.07





ANP-Oligo 137
AAAAAAAGUCCAAAAAAAA
−0.27
0.05





ANP-Oligo 083
AAAAAAACGUCAAAAAAAA
−0.27
0.11





ANP-Oligo 165
AAAAAAAUCGGAAAAAAAA
−0.26
0.17





ANP-Oligo 103
AAAAAAAGACUAAAAAAAA
−0.25
0.09





ANP-Oligo 012
AAAAAAAAUGAAAAAAAAA
−0.23
0.11





ANP-Oligo 061
AAAAAAACAUUAAAAAAAA
−0.23
0.08





ANP-Oligo 043
AAAAAAAAUCUAAAAAAAA
−0.22
0.1





ANP-Oligo 035
AAAAAAAAGUCAAAAAAAA
−0.22
0.14





ANP-Oligo 084
AAAAAAACGUGAAAAAAAA
−0.2
0.07





ANP-Oligo 107
AAAAAAAGUACAAAAAAAA
−0.2
0.11





ANP-Oligo 010
AAAAAAAAGUAAAAAAAAA
−0.16
0.1





ANP-Oligo 070
AAAAAAACCGUAAAAAAAA
−0.16
0.06





ANP-Oligo 146
AAAAAAAUAACAAAAAAAA
−0.16
0.14





ANP-Oligo 163
AAAAAAAUCCUAAAAAAAA
−0.15
0.09





ANP-Oligo 096
AAAAAAACUUGAAAAAAAA
−0.14
0.09





ANP-Oligo 036
AAAAAAAAGUGAAAAAAAA
−0.12
0.17





ANP-Oligo 164
AAAAAAAUCGCAAAAAAAA
−0.1
0.08





ANP-Oligo 097
AAAAAAACUUUAAAAAAAA
−0.1
0.1





ANP-Oligo 079
AAAAAAACGCUAAAAAAAA
−0.09
0.1





ANP-Oligo 013
AAAAAAAAUUAAAAAAAAA
−0.09
0.12





ANP-Oligo 185
AAAAAAAUUCCAAAAAAAA
−0.08
0.14





ANP-Oligo 186
AAAAAAAUUCGAAAAAAAA
−0.08
0.09





ANP-Oligo 049
AAAAAAAAUUUAAAAAAAA
−0.07
0.07





ANP-Oligo 039
AAAAAAAAUAGAAAAAAAA
−0.05
0.15





ANP-Oligo 040
AAAAAAAAUAUAAAAAAAA
−0.04
0.22





ANP-Oligo 087
AAAAAAACUAGAAAAAAAA
−0.03
0.09





ANP-Oligo 092
AAAAAAACUGCAAAAAAAA
0
0.06





ANP-Oligo 100
AAAAAAAGAAUAAAAAAAA
0.01
0.19





ANP-Oligo 022
AAAAAAAACGUAAAAAAAA
0.02
0.12





ANP-Oligo 076
AAAAAAACGAUAAAAAAAA
0.02
0.06





ANP-Oligo 138
AAAAAAAGUCGAAAAAAAA
0.02
0.18





ANP-Oligo 139
AAAAAAAGUCUAAAAAAAA
0.05
0.08





ANP-Oligo 112
AAAAAAAGCAUAAAAAAAA
0.08
0.12





ANP-Oligo 108
AAAAAAAGUAGAAAAAAAA
0.08
0.16





ANP-Oligo 160
AAAAAAAUCAUAAAAAAAA
0.1
0.14





ANP-Oligo 118
AAAAAAAGCGUAAAAAAAA
0.1
0.14





ANP-Oligo 127
AAAAAAAGGCUAAAAAAAA
0.11
0.09





ANP-Oligo 182
AAAAAAAUUACAAAAAAAA
0.11
0.13





ANP-Oligo 131
AAAAAAAGGUCAAAAAAAA
0.12
0.16





ANP-Oligo 115
AAAAAAAGCCUAAAAAAAA
0.15
0.08





ANP-Oligo 187
AAAAAAAUUCUAAAAAAAA
0.18
0.12





ANP-Oligo 044
AAAAAAAAUCGAAAAAAAA
0.2
0.09





ANP-Oligo 106
AAAAAAAGAGUAAAAAAAA
0.21
0.22





ANP-Oligo 037
AAAAAAAAGUUAAAAAAAA
0.21
0.17





ANP-Oligo 058
AAAAAAACAGUAAAAAAAA
0.22
0.08





ANP-Oligo 171
AAAAAAAUGAGAAAAAAAA
0.22
0.23





ANP-Oligo 109
AAAAAAAGUAAAAAAAAAA
0.22
0.21





ANP-Oligo 121
AAAAAAAGCUUAAAAAAAA
0.24
0.17





ANP-Oligo 191
AAAAAAAUUUCAAAAAAAA
0.27
0.15





ANP-Oligo 156
AAAAAAAUAUGAAAAAAAA
0.27
0.09





ANP-Oligo 147
AAAAAAAUAAGAAAAAAAA
0.27
0.36





ANP-Oligo 149
AAAAAAAUACCAAAAAAAA
0.27
0.2





ANP-Oligo 183
AAAAAAAUUAGAAAAAAAA
0.27
0.33





ANP-Oligo 174
AAAAAAAUGCGAAAAAAAA
0.28
0.23





ANP-Oligo 082
AAAAAAACGGUAAAAAAAA
0.29
0.2





ANP-Oligo 088
AAAAAAACUAUAAAAAAAA
0.3
0.15





ANP-Oligo 151
AAAAAAAUACUAAAAAAAA
0.3
0.2





ANP-Oligo 155
AAAAAAAUAUCAAAAAAAA
0.33
0.14





ANP-Oligo 152
AAAAAAAUAGCAAAAAAAA
0.35
0.23





ANP-Oligo 167
AAAAAAAUCUCAAAAAAAA
0.37
0.16





ANP-Oligo 134
AAAAAAAGUACAAAAAAAA
0.42
0.24





ANP-Oligo 153
AAAAAAAUAGGAAAAAAAA
0.42
0.31





ANP-Oligo 168
AAAAAAAUCUGAAAAAAAA
0.45
0.16





ANP-Oligo 157
AAAAAAAUAUUAAAAAAAA
0.46
0.22





ANP-Oligo 132
AAAAAAAGGUGAAAAAAAA
0.47
0.1





ANP-Oligo 141
AAAAAAAGUGGAAAAAAAA
0.5
0.45





ANP-Oligo 184
AAAAAAAUUAUAAAAAAAA
0.56
0.33





ANP-Oligo 143
AAAAAAAGUUCAAAAAAAA
0.56
0.29





ANP-Oligo 085
AAAAAAACGUUAAAAAAAA
0.57
0.18





ANP-Oligo 193
AAAAAAAUUUUAAAAAAAA
0.62
0.18





ANP-Oligo 124
AAAAAAAGGAUAAAAAAAA
0.63
0.23





ANP-Oligo 192
AAAAAAAUUUGAAAAAAAA
0.65
0.12





ANP-Oligo 189
AAAAAAAUUGGAAAAAAAA
0.66
0.15





ANP-Oligo 135
AAAAAAAGUAGAAAAAAAA
0.67
0.34





ANP-Oligo 169
AAAAAAAUCUUAAAAAAAA
0.67
0.36





ANP-Oligo 148
AAAAAAAUAAUAAAAAAAA
0.7
0.34





ANP-Oligo 181
AAAAAAAUGUUAAAAAAAA
0.79
0.23





ANP-Oligo 170
AAAAAAAUGACAAAAAAAA
0.86
0.39





ANP-Oligo 179
AAAAAAAUGUCAAAAAAAA
0.9
0.23





ANP-Oligo 094
AAAAAAACUGUAAAAAAAA
0.9
0.22





ANP-Oligo 046
AAAAAAAAUGUAAAAAAAA
0.97
0.26





ANP-Oligo 140
AAAAAAAGUGCAAAAAAAA
0.99
0.55





ANP-Oligo 136
AAAAAAAGUAUAAAAAAAA
1.04
0.33





ANP-Oligo 173
AAAAAAAUGCCAAAAAAAA
1.22
0.3





ANP-Oligo 142
AAAAAAAGUGUAAAAAAAA
1.23
0.54





ANP-Oligo 180
AAAAAAAUGUGAAAAAAAA
1.31
0.37





ANP-Oligo 145
AAAAAAAGUUUAAAAAAAA
1.32
0.5





ANP-Oligo 133
AAAAAAAGGUUAAAAAAAA
1.51
0.61





ANP-Oligo 154
AAAAAAAUAGUAAAAAAAA
1.57
0.43





ANP-Oligo 190
AAAAAAAUUGUAAAAAAAA
1.6
0.36





ANP-Oligo 188
AAAAAAAUUGCAAAAAAAA
1.62
0.19





ANP-Oligo 175
AAAAAAAUGCUAAAAAAAA
1.85
0.4





ANP-Oligo 172
AAAAAAAUGAUAAAAAAAA
1.92
0.83





ANP-Oligo 034
AAAAAAAAGGUAAAAAAAA
2.21
0.36





ANP-Oligo 176
AAAAAAAUGGCAAAAAAAA
2.41
0.46





ANP-Oligo 178
AAAAAAAUGGUAAAAAAAA
3.26
0.98





ANP-Oligo 144
AAAAAAAGUUGAAAAAAAA
4.39
2.19





ANP-Oligo 166
AAAAAAAUCGUAAAAAAAA
4.42
0.6








Claims
  • 1-37. (canceled)
  • 38. A method for predicting the immunostimulatory activity of a single-stranded RNA oligonucleotide, comprising the steps of: (a) identifying all possible 3-nucleotide (3mer) motifs contained in the oligonucleotide;(b) assigning an IL-12 point score for each individual 3mer motif (i) for a 3mer motif which appears in Table 5, assing an IL-12 point score according to Table 5;(ii) for a 3mer motif which does not appear in Table 5, assign an IL-12 point score of 0;(c) assigning the sum of the IL-12 point scores of individual 3mer motifs as the IL-12 score of the oligonucleotide; and(d) assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 93.53, an intermediate immunostimulatory activity if the IL-12 score is between −28 and 93.53, and a low immunostimulatory activity if the IL-12 score is at most −28, when n=6; assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 107.77, an intermediate immunostimulatory activity if the IL-12 score is between −31 and 107.77, and a low immunostimulatory activity if the IL-12 score is at most −31, when n=7;assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 121, an intermediate immunostimulatory activity if the IL-12 score is between −34 and 121, and a low immunostimulatory activity if the IL-12 score is at most −34, when n=8;assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 121.98, an intermediate immunostimulatory activity if the IL-12 score is between −36 and 121.98, and a low immunostimulatory activity if the IL-12 score is at most −36, when n=9;assigning to the oligonucleotide a high immunostimulatory activity if the IL-12 score is at least 8.4064×n+66.958, an intermediate immunostimulatory activity if the IL-12 score is between 0.0468×n2−2.3103×n−23.244 and 8.4064×n+66.958, and a low immunostimulatory activity if the IL-12 score is at most 0.0468×n−2.3103×n−23.244, when n is greater than 9,wherein n is the length of the oligonucleotide.
  • 39. A method for determining the immunostimulatory activity of a RNA oligonucleotide, comprising the steps of: (a) complexing the RNA oligonucleotide with a complexation agent;(b) contacting a cell with the complexed RNA oligonucleotide, wherein the cell expresses TLR8; and(c) determining the amount of IL-12 produced by the cell of step (b), an increase of IL-12 production indicating immunostimulatory activity of the RNA oligonucleotide.
  • 40. The method of any one of claims 39, wherein the complexation agent is poly-L-arginine (poly-L-Arg).
  • 41. The method of claim 39, wherein the cell is a CD14+ monocyte.
  • 42. A method for preparing a single-stranded RNA oligonucleotide having high immunostimulatory activity, comprising the steps of: (a) providing candidate oligonucleotide sequence(s);(b) identifying oligonucleotide sequence(s) with high immunostimulatory activity predicted according to the method of claim 38;(c) preparing the RNA oligonucleotide(s) identified for high immunostimulatory activity in step (b); and(d) optionally testing the immunostimulatory activity of the RNA oligonucleotide(s) prepared in step (c) according to the method of claim 39; and(e) further optionally modifying the oligonucleotide(s) to optimize the immunostimulatory activity.
  • 43. A method for preparing a single-stranded RNA oligonucleotide having low immunostimulatory activity, comprising the steps of: (a) providing candidate oligonucleotide sequence(s);(b) identifying oligonucleotide sequence(s) with low immunostimulatory activity predicted according to the method of claim 38;(c) preparing the RNA oligonucleotide(s) identified for low immunostimulatory activity in step (b); and(d) optionally testing the RNA oligonucleotide(s) prepared in step (c) for the lack of immunostimulatory activity according to the method of claim 39; and(e) further optionally modifying the oligonucleotide(s) to minimize the immunostimulatory activity.
  • 44. A method for preparing a single-stranded RNA oligonucleotide having high immunostimulatory activity, comprising the steps of: (a) providing an oligonucleotide sequence which comprises at least one of the 4-nucleotide (4-mer) motifs selected from the group consisting of:
  • 45. A method for preparing an antisense RNA oligonucleotide having gene silencing activity and high immunostimulatory activity, comprising the steps of: (a) identifying all potential antisense sequences for a target mRNA;(b) identifying antisense sequences that have gene silencing activity;(c) predicting the immunostimulatory activity for the antisense sequences identified in step (b) according to the method of claim 38;(d) identifying antisense RNA sequences which have an IL-12 score of at least 8.4064×n+66.958, wherein the IL-12 score is assigned according to the method a defined in claim 1 (a)-(c), and wherein n is the length of the sequence and n is between 18 and 50;(e) decreasing the IL-12 score threshold by 1 if no antisense sequence is identified in step (d), until at least one antisense sequence is identified;(f) preparing the antisense RNA identified in step (d) or (e);(g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense prepared in (f);(h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing and/or immunostimulatory activity.
  • 46. A method for preparing an antisense RNA oligonucleotide having gene silencing activity and high immunostimulatory activity, comprising the steps of: (a) identifying all potential antisense sequences for a target mRNA;(b) predicting the immunostimulatory activity for all of the potential antisense sequences identified in (a) according to the method of claim 38;(c) identifying 10 potential antisense sequences with the highest IL-12 scores;(d) identifying antisense sequences with gene silencing activity among the 10 potential antisense sequences identified in step (c);(e) identifying 10 potential antisense sequences with the next highest IL-12 scores if no antisense sequence can be identified in step (d); repeat steps (d) and (e) until at least one antisense sequence is identified;(f) preparing the antisense RNA identified in step (d) or (e);(g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense RNA prepared in (f);(h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing and/or immunostimulatory activity.
  • 47. A method for preparing an antisense RNA oligonucleotide having gene silencing activity and low immunostimulatory activity, comprising the steps of: (a) identifying all potential antisense sequences for a target mRNA;(b) identifying antisense sequences that have gene silencing activity;(c) predicting the immunostimulatory activity for the antisense sequences identified in step (b) according to the method of claim 38;(d) identifying antisense sequences which have an IL-12 score of at most 1.9763×n −30.643, wherein the IL-12 score is assigned according to the method as defined in claim 38(a)-(c), and wherein n is the length of the sequence and n is between 18 and 50;(e) increasing the IL-12 score threshold by 1 if no antisense sequence is identified in step (b), until at least one antisense sequence is identified;(f) preparing the antisense RNA identified in step (d) or (e);(g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense RNA prepared in (f);(h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing activity and/or to minimize the immunostimulatory activity.
  • 48. A method for preparing an antisense RNA oligonucleotide having gene silencing activity and low immunostimulatory activity, comprising the steps of: (a) identifying all potential antisense sequences for a target mRNA;(b) predicting the immunostimulatory activity for all of the potential antisense sequences identified in (a) according to the method of claim 38;(c) identifying 10 potential antisense sequences with the lowest IL-12 scores;(d) identifying antisense sequences with gene silencing activity among the 10 potential antisense sequences identified in step (c);(e) identifying 10 potential antisense sequences with the next lowest IL-12 scores if no antisense sequence can be identified in step (d); repeat steps (d) and (e) until at least one antisense sequence is identified;(f) preparing the antisense RNA identified in step (d) or (e);(g) optionally testing the gene silencing and/or the immunostimulatory activity of the antisense RNA prepared in (f);(h) further optionally modify the antisense RNA prepared in (f) to optimize the gene silencing activity and/or to minimize the immunostimulatory activity.
  • 49. A single-stranded RNA oligonucleotide having immunostimulatory activity comprising at least one of the 4-nucleotide (4-mer) motifs selected from the group consisting of:
  • 50. The oligonucleotide of claim 49, wherein the 4mer motifs are selected from the group consisting of number 1-11 of the 4mer motifs.
  • 51. A single-stranded RNA oligonucleotide having immunostimulatory activity comprising at least one of the 4mer motifs selected from the group consisting of number 1-11 of the 4mer motifs, wherein the spacer nucleotides which are not part of any of the 4mer motif(s) are identical, and wherein the spacer nucleotide is selected from the group consisting of A, T, C, G and variants and derivatives thereof.
  • 52. The oligonucleotide of claim 51, wherein the spacer nucleotide is A or a derivative thereof.
  • 53. The oligonucleotide of any one of claims 49-52, wherein the oligonucleotide does not have gene silencing activity for any known mammalian gene.
  • 54. An antisense RNA oligonucleotide having both gene silencing activity and immunostimulatory activity, wherein the oligonucleotide has an IL-12 score of at least 8.4064×n+66.958, wherein the IL-12 score is assigned according to the method as defined in claim 38(a)-(c), wherein n is the length of the oligonucleotide and n is between 14 and 50,and wherein the oligonucleotide is not
  • 55. An antisense RNA oligonucleotide having gene silencing activity and low immunostimulatory activity, wherein the oligonucleotide has an IL-12 score of at most 1.9763×n −30.643, wherein the IL-12 score is assigned according to the method as defined in claim 38(a)-(c), wherein n is the length of the oligonucleotide and n is between 14 and 50,and wherein the oligonucleotide is not
  • 56. A pharmaceutical composition comprising one or more of the RNA oligonucleotides of any one of claims 49-55 and a pharmaceutically acceptable carrier.
  • 57. Use of the single-stranded RNA oligonucleotide of any one of claims 49-53 for the preparation of a pharmaceutical composition for inducing an immune response in a mammal.
  • 58. Use of the RNA oligonucleotide of any one of claims 49-53 for the preparation of a pharmaceutical composition for preventing and/or treating a disorder selected from the group consisting of immune disorders, infections, and cancers in a mammal.
  • 59. Use of the RNA oligonucleotide of claim 54 for the preparation of a pharmaceutical composition for preventing and/or treating a disorder selected from the group consisting of infections and cancers in a mammal.
  • 60. Use of the RNA oligonucleotide of claim 55 for the preparation of a pharmaceutical composition for preventing and/or treating a disorder selected from the group consisting of cancer, dermatological disorders, immune disorders, metabolic disorders, neurological disorders, ocular diseases, infections, and other hereditary and non-hereditary disorders in a mammal.
  • 61. An in vitro method of inducing IL-12 production by a mammalian cell, comprising the steps of: (a) complexing the RNA oligonucleotide of any one of claims 49-53 with a complexation agent; and(b) contacting the cell with the complex prepared in step (a).
Priority Claims (1)
Number Date Country Kind
05020019.5 Sep 2005 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP2006/008972 9/14/2006 WO 00 11/7/2008
Provisional Applications (1)
Number Date Country
60717359 Sep 2005 US