AU2013201786B2 - Amino-LNA, thio-LNA and alpha-L-oxy-LN - Google Patents

Amino-LNA, thio-LNA and alpha-L-oxy-LN Download PDF

Info

Publication number
AU2013201786B2
AU2013201786B2 AU2013201786A AU2013201786A AU2013201786B2 AU 2013201786 B2 AU2013201786 B2 AU 2013201786B2 AU 2013201786 A AU2013201786 A AU 2013201786A AU 2013201786 A AU2013201786 A AU 2013201786A AU 2013201786 B2 AU2013201786 B2 AU 2013201786B2
Authority
AU
Australia
Prior art keywords
lna
oxy
beta
alpha
oligonucleotides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU2013201786A
Other versions
AU2013201786A1 (en
Inventor
Miriam Frieden
Henrik Frydenlund Hansen
Troels Koch
Nikolaj Dam Mikkelsen
Daniel-Sejer Pedersen
Christoph Rosenbohm
Christensen Signe M.
Charlotte Albaek Thrue
Majken Westergaard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Roche Innovation Center Copenhagen AS
Original Assignee
Roche Innovation Center Copenhagen AS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2011201821A external-priority patent/AU2011201821B2/en
Application filed by Roche Innovation Center Copenhagen AS filed Critical Roche Innovation Center Copenhagen AS
Priority to AU2013201786A priority Critical patent/AU2013201786B2/en
Publication of AU2013201786A1 publication Critical patent/AU2013201786A1/en
Assigned to ROCHE INNOVATION CENTER COPENHAGEN A/S reassignment ROCHE INNOVATION CENTER COPENHAGEN A/S Alteration of Name(s) of Applicant(s) under S113 Assignors: SANTARIS PHARMA A/S
Application granted granted Critical
Publication of AU2013201786B2 publication Critical patent/AU2013201786B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Abstract

Abstract A novel class of pharmaceuticals which comprises a Locked Nucleic Acid (LNA) which can be used in antisense therapy. These novel oligonucleotides have improved antisense properties. The novel oligonucleotides are composed of at least one LNA selected from beta-D-thio/amino-LNA or alpha-L-oxy/thio/amino LNA. The oligonucleotides comprising LNA may also include DNA and/or RNA nucleotides.

Description

1 AUSTRALIA Patents Act 1990 Santaris Pharma A/S COMPLETE SPECIFICATION STANDARD PATENT Invention Title: Amino-LNA, thio-LNA and alpha-L-oxy-LN The following statement is a full description of this invention including the best method of performing it known to us:- 2 ANTISENSE DESIGN Cross-Reference to Related Applications This application is a divisional application under S 79B of the Patents Act 1990 of Australian Patent Application No. 2011201821 which is a divisional application of Australian Patent No. 5 2003281969 which corresponds to International Application No. PCT/DK2003/000788 filed November 18, 2003, which claims priority to Danish Provisional Application No. PA200301540 filed October 20, 2003 and Danish Provisional Application No. PA200201774 filed November 18, 2002, the entire contents of which are incorporated herein by reference. Field of Invention 10 The present invention relates to pharmaceuticals comprising antisense oligonucleotides, and novel oligonucleotides having improved antisense properties. Background of the Invention The Professors Imanishi and Wengel independently invented Locked Nucleic Acid (LNA) in 1997 (International Patent Applications WO 99/14226,WO 98/39352; P. Nielsen et al, J. 15 Chem. Soc., Perkin Trans. 1, 1997, 3423; P. Nielsen et al., Chem. Commun., 1997, 9, 825; N. K. Christensen et al., J. Am. Chem. Soc., 1998, 120, 5458; A. A. Koshkin et al., J. Org. Chem., 1998, 63, 2778; A. A Koshkin et al. J. Am. Chem. Soc. 1998, 120, 13252-53; Kumar et al. Bioorg, & Med. Chem. Lett.,1998, 8, 2219-2222; and S. Obika et al., Bioorg. Med. Chem. Lett., 1999, 515). The first LNA monomer was based on the 2'-O-CH 2 -4' 20 bicyclic structure. Due to the configuration of this structure it is called: beta-D-oxy-LNA. This oxy-LNA has since then showed promising biological applications (Braasch & Corey, Biochemistry, 2002, 41(14), 4503-19; Childs et al. PNAS, 2002, 99(17), 11091-96; Crinelli et al., Nucl. Acid. Res., 2002, 30(11), 2435-43; Elayadi et al., Biochemistry, 2002, 41, 9973-9981; Jacobsen et al., Nucl. Acid. Res., 2002, 30(19), in press; Kurreck et al., Nucl. 25 Acid. Res., 2002, 30(9),1911-1918; Simeonov & Nikiforov, Nucl. Acid. Res., 2002, 30(17); Alayadi & Corey, Curr. opinion in Inves. Drugs., 2001, 2(4), 558-61; Obika et al., Bioorg. & Med. Chem., 2001, 9, 1001-11; Braasch & Corey, Chem. & Biol., 2000, 55, 1-7; Wahlestedt et al., PNAS, 2000, 97(10), 5633-38), Freier & Altmann, Nucl. Acid Res., 1997, 25, 4429-43; Cook, 1999, Nucleosides & Nucleotides, 18(6&7), 1141-62. 30 Right after the discovery of oxy-LNA the bicyclic furanosidic structure was chemically derivatised. Thus, the 2'-S-CH 2 -4' (thio-LNA) and the 2'-NH-CH 2 -4' (amino-LNA) bicyclic analogues were disclosed (Singh, S. K., J. Org. Chem., 1998, 63, 6078-79; Kumar et al. Bioorg, & Med. Chem. Lett.,1998, 8, 2219-2222; Singh et al. J. Org. Chem., 1998, 63, 10035-39). The synthesis of the thio-LNA containing uridine as nucleobase has been 3 shown (Singh, S. K., J. Org. Chem., 1998, 63, 6078-79). For amino-LNA the synthesis of the thymidine nucleobase has been disclosed (Kumar et al. Bioorg, & Med. Chem. Lett.,1998, 8, 2219-2222; Singh et al. J. Org. Chem., 1998, 63, 10035-39). A series of LNA-diastereoisomers have been prepared (Rajwanshi et al., J. Chem Commun. 5 1999;2073-2074; Hakansson & Wengel, Bioorg Med Chem Lett 2001; 11(7):935-938; Rajwanshi et al. , Chem Commun., 1999;1395-1396; Wengel at al., Nucleosides Nucleotides Nucleic Acids, 2001; 20(4-7):389-396; Rajwanshi et al., Angew. Chem. Int. Ed., 2000; 39:1656-1659; Petersen et al., J. Amer. Chem. Soc., 2001, 123(30), 7431-32; Sorensen et al., J. Amer. Chem. Soc., 2002, 124(10), 2164-76; Vester et al., J. Amer. 10 Chem. Soc., 2002, 124(46), 13682-13683). In the prior art the synthesis of alpha-L-xylo, xylo-LNA, and alpha-L-oxy-LNA containing thymidine bases have been shown. For the alpha-L-oxy-LNA also the 5-methyl and adenine nucleosides have been synthesised. The melting temperature (Tm) of duplexes containing the LNA distereoisomers have been presented. It turned out that the alpha-L-oxy-LNA has interesting properties. It was shown 15 that the alpha-L-oxy-LNA can be incorporated in complex chimerae comprising DNA/RNA residues and be adapted in the oligo structure and increase the binding. This property of being incorporated in oligonucleotides containing several other monomeric classes and act co-operatively is a property that the alpha-L-oxy-LNA shares with the parent oxy-LNA. Furthermore, it has been demonstrated that a segment of 4 consecutive alpha-L-T 20 monomers can be incorporated in conjunction with a segment of 4 consecutive oxy-LNA-T monomers (Rajwanshi et al., Chem. Commun., 1999, 2073-74). Increased stability of oligonucleotides containing alpha-L-oxy-LNA monomers (MeC, A, T-monomers) have been demonstrated. The alpha-L-oxy-LNA monomers were incorporated into oligonucleotides with alternating alpha-L-oxy-LNA and DNA monomers (mix-mers) and in fully modified 25 alpha-L-oxy-LNA oligomers. The stability was compared to oxy-LNA and to DNA and it was found that alpha-L-oxy-LNA monomers displaced the same protection pattern as oxy-LNA (Sorensen, et al., J.Amer.Chem.Soc., 2002, 124(10), 2164-76). The same alpha-L-oxy LNA containing oligonucleotides were tested in RNase H assays and it was found that the designs disclosed were not efficiently recruiting RNase H. When these examples are taken 30 together also in combination with the data published by Arzumanov et al (Biochemistry 2001, 40, 14645-54) it has not been shown that alpha-L-oxy-LNA containing oligonucleotides efficiently recruits RNase H. Oligonucleotides containing any combination of the diastereoisomers and any other LNA family member has not been demonstrated. 35 Natural dsDNA exists at physiological pH as a B-form helix, whereas dsRNA exists as an A form helix. A helix formed by DNA and RNA exists in an intermediate A/B-form. This morphological difference is originated in the difference in the preferred sugar conformations of the deoxyriboses and the riboses. The furanose ring of deoxyribose exists 4 at room temperature in an equilibrium between C2'-endo (S-type) and C3'-endo (N-type) conformation with an energy barrier of ~2 kcal/mol (Figure 3). For deoxyribose the S-type conformation is slightly lowered in energy (~0.6 kcal/mol) compared to the N-type and explains why DNA is found in the S-type conformation. The conformation leads to the B 5 form helix. For ribose, and RNA, the preference is for the N-type that leads to the A-form helix. The A-form helix is associated with higher hybridisation stability. The oxy-LNA and the LNA analogues are locked in the N-conformation and consequently the oligonucleotides they are forming will be RNA-like. The alpha-L-oxy-LNA is locked in a S-type and therefore the oligonucleotides that it will form will be more DNA like (Sorensen et al., 3. Amer. 10 Chem. Soc., 2002, 124(10), 2164-76; Rajwanshi et al., Angew. Chem. Int. Ed., 2000; 39:1656-1659). Molecular strategies are being developed to modulate unwanted gene expression that either directly causes, participates in, or aggravates a disease state. One such strategy involves inhibiting gene expression with oligonucleotides complementary in sequence to the messenger RNA of a target gene. The messenger RNA strand is a copy of 15 the coding DNA strand and is therefore, as the DNA strand, called the sense strand. Oligonucleotides that hybridise to the sense strand are called antisense oligonucleotides. Binding of these strands to mRNA interferes with the translation process and consequently with gene expression. Zamecnik and co-workers originally described the Antisense strategy and the principle has since then attracted a lot of interest (Zamecnik & Stephenson, PNAS, 20 1978, 75(1), 280-4; Bennet & Cowset, Biochim. Biophys. Acta, 1999, 1489, 19-30; Crooke, 1998, Biotechnol.Genet.Eng Rev., 15, 121-57; Wengel, J. In Antisense Drug Technology; Principles, Strategies, and Applications; Edited by Crooke, S. T., Ed.; Marcel Dekker, Inc.: New York, Basel, 2001; pp 339-357). It has been a long sought goal to develop drugs with the capacity to destroy malignant 25 genes base specifically. The applications of such drugs in e.g. cancer and infections diseases are self-evident. Native oligonucleotides cannot be employed as such mainly due to their instability in cellular media and to too low affinity for the target genes. The wish to develop nucleic acid probes with improved properties in this regard has been the main driver behind the massive synthesis effort in the area of nucleic acid analogue preparation. 30 The most important guideline in this work has been to design the DNA analogues in such a way that the DNA analogue would attain the N-type/"RNA"-like conformation that is associated with the higher affinity of the oligonucleotides to nucleic acids. One of the important mechanisms involved in Antisense is the RNase H mechanism. RNase H is an intra cellular enzyme that cleaves the RNA strand in RNA/DNA duplexes. Therefore, 35 in the search for efficient Antisense oligonucleotides, it has been an important hallmark to prepare oligonucleotides that can activate RNase H. However, the prerequisite for an oligonucleotide in this regard is therefore that the oligo is DNA-like and as stated above most high affinity DNA analogues induces RNA-like oligonucleotides. Therefore, to 5 compensate for the lack of RNase H substrate ability of most DNA analogues (like e.g. 2' OMe DNA analogue and oxy-LNA) the oligonucleotides must have segments/consecutive stretches of DNA and/or phosphorothioates. Depending on the design of the segments of such oligonucleotides they are usually called Gap-mers, if the DNA segment is flanked by 5 the segments of the DNA analogue, Head-mers, if the segment of the DNA analogue is located in the 5' region of the oligonucleotide, and Tail-mers, if the segment of the DNA analogue is located in the 3' region of the oligonucleotide. It should be mentioned that other important mechanisms are involved in Antisense that are not dependent on RNase H activation. For such oligonucleotides the DNA analogues, 10 like LNA, can be placed in any combination design (Childs et al. PNAS, 2002, 99(17), 11091-96; Crinelli et al., Nucl. Acid. Res., 2002, 30(11), 2435-43; Elayadi et al., Biochemistry, 2002, 1, 9973-9981; Kurreck et al., Nucl. Acid. Res., 2002, 30(9), 1911 1918; Alayadi & Corey, Curr. opinion in Inves. Drugs., 2001, 2(4), 558-61; Braasch & Corey, Chem. & Biol., 2000, 55, 1-7). 15 In contrast to the beta-D-oxy-LNA the alpha-L-oxy-LNA has a DNA-like locked conformation and it has been demonstrated that alpha-L-oxy-LNA can activate RNase H (Sorensen et al., J. Amer. Chem. Soc., 2002, 124(10), 2164-76). However, the cleavage rate of RNase H is much lower compared to DNA in the disclosed designs and thus, the oligonucleotides in the disclosed designs have not been shown to be efficient Antisense 20 reagents. Summary of the Invention The present inventors have found a novel class of pharmaceuticals which can be used in antisense therapy. Also, the inventors disclose novel oligonucleotides with improved antisense properties. The novel oligonucleotides are composed of at least one Locked 25 Nucleic Acid (LNA) selected from beta-D-thio/amino-LNA or alpha-L-oxy/thio/amino LNA.The oligonucleotides comprising LNA may also include DNA and/or RNA nucleotides. The present inventors have demonstrated that a,-L-oxy-LNA surprisingly provides the possibility for the design of improved Antisense oligonucleotides that are efficient 30 substrates for RNase H. These novel designs are not previously described and the guidelines developed broaden the design possibilities of potent Antisense oligonucleotides. Also comprised in this invention is the disclosure of Antisense oligonucleotides having other improved properties than the capability of being RNase H substrates. The oligonucleotides comprise any combination of LNA-relatives with DNA/RNA, and their analogues, as well as 35 oxy-LNA. The design of more potent Antisense reagents is a combination of several features. Among the features of these novel oligonucleotide designs are increased 6 enzymatic stability, increased cellular uptake, and efficient ability to recrute RNase H. Also important is the relation between the length and the potency of the oligonucleotides (e.g. a 15-mer having the same potency as a 21-mer is regarded to be much more optimal). The potency of the novel oligonucleotides comprised in this invention is tested in cellular in vitro assays and in vivo assays. It is furthermore showed that the novel designs also improves the in vivo properties such as better pharmacokinetic/pharmacological properties and toxicity profiles. In one aspect, the invention provides a locked nucleic acid (LNA) nucleoside having the formula RN O Base 0 (CH2)n X4 0 R" or n(H2C)- X Base 0OkO R. R" or a pharmaceutically acceptable salt thereof, wherein: X is 0; R' and R" are independently either H or a terminal group; and (CH2)n is a methylene substituted with a C 1 6 alkyl. In another aspect, the invention provides a pharmaceutical composition comprising the LNA antisense oligomer of the invention and a pharmaceutically acceptable carrier.
6A Beta-D-oxy-LNA and the analogues thio-and amino LNA: 0Base - Base 0 Base o= -o- oIo- oIo Oxy-LNA Thio-LNA Amino-LNA LNA diastereoisomers: HO O Base HO 0 Base H HO OBase HOO 0 a-L-oxy-LNA p-D-xylo-LNA a-D-oxy-LNA s Base NH Base HO HO a-L-thio-LNA a-L-amino-LNA Sugar conformations in DNA: B B o - t C2'-endo (S-type) C3'-endo (NI-type) 7 Disclosure of the Invention Thus, the present invention in it broadest scope relates to a pharmaceutical composition comprising a therapeutically active antisense oligonucleotide construct which (i) comprises at least one locked nucleic acid unit selected from the group consisting of amino-LNA and 5 thio-LNA and derivatives thereof; or (ii) comprises at least two consecutively located locked nucleotide units of which at least one is selected from the group consisting of alpha L-oxy-LNA and derivatives thereof. The antisense construct can be in the form of a salt or in the form of prodrug or salts of such prodrug. The invention thus relates to pharmaceutical compositions in which an active ingredient is a pharmaceutically acceptable 10 salt, prodrug (such as an ester) or salts of such prodrug of the above oligonucleotide construct. Both amino- and thio-LNA can be either alpha or beta configuration, and in (i), the oligonucleotide construct encompasses constructs with at least one (such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) units selected from the group consisting of: alpha-L-thio-LNA, beta-D thio-LNA, beta-D-amino-LNA, alpha-L-amino-LNA and derivatives thereof; optionally in 15 combination with at least one (such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) further independently selected locked or non-locked nucleotide units. Examples on these further units are oxy-LNA (such as alpha-L or beta-D), thio/amino LNA (such as alpha-L or beta-D), a nucleotide unit which has a 2'-deoxy erythro-pentofuranosyl sugar moiety (such as a DNA nucleotide), a nucleotide unit which 20 has a ribo-pentofuranosyl sugar moiety (such as a RNA nucleotide); and derivatives thereof. In (ii), the oligonucleotide construct encompasses constructs with at least two (such as 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) consecutively located nucleotide units, of which at least one (such as 1, 2, 3, 4, 5, 6, 7 or more) is alpha-L-oxy LNA units or derivatives thereof. In addition to the alpha-L-oxy LNA units or derivatives thereof, the sequence of 25 consecutively located locked nucleotide units optionally comprises other locked nucleotide units (such as the units defined herein). Besides the essential two consecutively located locked nucleotide units, the construct in (ii) optionally comprises one or more (such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) independently selected locked or non-locked nucleotide units (such as the units defined herein). 30 In an interesting embodiment, the invention relates to a pharmaceutical composition in which the antisense oligonucleotide construct comprises two adjacently located nucleotide sequences A and B, where A represents a sequence of nucleotide units comprising (i) at least one locked nucleotide 35 unit selected from the group consisting of thio-LNA, amino-LNA (both in either alpha-L or beta-D configuration) and derivatives thereof, or (ii) at least two consecutively located locked nucleotide units of which at least one is selected from the group consisting of alpha L-oxy-LNA and derivatives thereof; and 8 B represents one nucleotide unit or a sequence of nucleotide units, with the proviso that at least one nucleotide unit in B has a 2'-deoxy-erythro-pentofuranosyl sugar moiety or a ribo-pentofuranosyl sugar moiety. Sequence A can additionally comprise at least one further locked nucleotide unit (such as 2, 3, 4 or 5 units), preferably selected 5 independently from the group consisting of amino-LNA, thio-LNA (both in either alpha-L or beta-D configuration), alpha-L-oxy-LNA and derivatives thereof. In an other interesting embodiment, the invention relates to a pharmaceutical composition comprising an oligonucleotide construct which contains three adjacently located nucleotide 10 sequences, A, B and C, in the following order (5' to 3'): A-B-C or C-B-A, in which A represents a sequence comprising at least two consecutively located locked nucleotide units, at least one of which is an alpha-L-oxy-LNA unit, and which sequence optionally contains one or more (such as 2, 3, 4 or 5) non-locked nucleotide units (such as 15 deoxyribonucleotide units, ribonucleotide units or derivatives thereof) and/or optionally contains one or more (such as 2, 3, 4 or 5) locked nucleotide units, such as a unit selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA (all in either alpha-L or beta-D configuration) and derivatives thereof; B represents one nucleotide unit or a sequence of nucleotide units, with the proviso that at 20 least one nucleotide unit in B has a 2'-deoxy-erythro-pentofuranosyl sugar moiety or a ribo-pentofuranosyl moiety; and C represents a sequence comprising at least two consecutively located locked nucleotide units, at least one of which is an alpha-L-oxy-LNA unit, and which sequence optionally contains one or more (such as 2, 3, 4 or 5) non-locked nucleotide units (such as 25 deoxyribonucleotide units, ribonucleotide units or derivatives thereof) and/or optionally contains one or more (such as 2, 3, 4 or 5) locked nucleotide units, such as a unit selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA (all in either alpha-L or beta-D configuration) and derivatives thereof. 30 The invention also relates to an oligonucleotide construct which comprises at least one nucleotide sequence comprising one or more nucleotide units selected from the group consisting of amino-LNA, thio-LNA (in all configurations) and derivatives thereof; with the proviso that the following oligonucleotide constructs are excluded: (i) 5'-d(GTGAVATGC), 5'-d(GVGAVAVGC), 5'-d(GTGAXATGC), 5'-d(GXGAXAXGC), 5' 35 d(GXGVXVXGC), in which sequences V represents a beta-D-amino-LNA thymine unit, and X represents a beta-D-methylamino-LNA thymine unit; and (ii) 5'-d(GTGAYATGC), 5'-d(GYGAYAYGC) and 5'-d(GYGYYYYGC) in which sequences Y represents a beta-D-thio-LNA uracil unit.
9 The excluded oligonucleotides are previously disclosed by Singh et al and Kumar et al. (Kumar et al. Bioorg, & Med. Chem. Lett.,1998, 8, 2219-2222; Singh et al. J. Org. Chem., 1998, 63, 10035-39). It has collectively for the excluded LNA- relatives been shown that they can be incorporated into oligonucleotides. However, no biological properties have not 5 been demonstrated or suggested. A presently preferred group of oligonucleotide constructs of the invention comprises two adjacently located nucleotide sequences, A and B, where A represents a sequence of nucleotide units comprising at least one locked nucleotide unit selected from the group 10 consisting of amino-LNA, thio-LNA (both in either alpha-L or beta-D) configuration, and derivatives thereof; and B represents one nucleotide unit or a sequence of nucleotide units, with the proviso that at least one nucleotide unit in B has a 2'-deoxy-erythro pentofuranosyl sugar moiety or a ribo-pentofuranosyl moiety; especially constructs in which B represents a sequence of nucleotide units, said sequence contains a subsequence 15 of at least three nucleotide units having 2'-deoxy-erythro-pentofuranosyl sugar moieties, such as 4, 5, 6, 7, 8, 9 or 10 nucleotide units, said subsequence optionally being spiked with an other nucleotide, preferably an alpha-L-oxy-LNA unit selected from the group consisting of alpha-L-amino-LNA, alpha-L-thio-LNA, alpha-L-oxy-LNA and derivatives thereof. 20 Also interesting is a construct according which comprises three adjacently located nucleotide sequences in the following order (5' to 3'):A-B-C, in which the nucleotide sequences A and B are as defined as above, and C represents a sequence of nucleotide units, which comprises at least one locked nucleotide unit selected 25 from the group consisting of amino-LNA, thio-LNA (both in either alpha-L or beta-D configuration) and derivatives thereof. In the above constructs, it is preferred that A has a length of 2-10 (preferably 2-8, such as 3, 4, 5, 6, 7) nucleotide units; B has a length of 1-10 (preferably 5-8, such as 6 or 7) 30 nucleotide units; and C (if present) has a length of 2-10 (preferably 2-8, such as 3, 4, 5, 6, or 7) nucleotide units; so that the overall length of the construct is 6-30 (preferably 10 20, more preferably 12-18, such as 13, 14, 15, 16 or 17) nucleotide units. A preferred embodiment of the above construct according to the invention is a construct in 35 which A represents a sequence of nucleotide units comprising at least two consecutively located locked nucleotide units (such as 3, 4, 5, 6, 7, 8, 9 or 10 units), at least one of said locked nucleotide units being selected from the group consisting of amino-LNA, thio-LNA and derivatives thereof; C represents a sequence of nucleotide units comprising at least two consecutively located locked nucleotide units (such as 3, 4, 5, 6, 7, 8, 9 or 10 units), 10 at least one of said locked nucleotide units being selected from the group consisting of amino-LNA, thio-LNA (in all configurations) and derivatives thereof, and/or B represents a sequence of least 2 nucleotide units (such as 3, 4, 5, 6, 7, 8, 9 or 10 units), which sequence in addition to the nucleotide unit(s) having 2'-deoxy-erythro-pentofuranosyl 5 sugar moiety(ies) and/or ribo-pentofuranosyl moiety(ies), comprises nucleotides units which are selected independently from the group consisting of: locked nucleotide units (such as alpha-L-oxy-, -thio-, or -amino- nucleotide units) and derivatives thereof. An other embodiment of the invention relates to an oligonucleotide construct which 10 contains three adjacently located nucleotide sequences, A, B and C, in the following order (5' to 3'): A-B-C or C-B-A, in which A represents a sequence comprising at least two consecutively located locked nucleotide units, at least one of which is an alpha-L-oxy-LNA unit, and which sequence optionally contains one or more (such as 2, 3, 4 or 5) non-locked nucleotide units (such as 15 deoxyribonucleotide units, ribonucleotide units or derivatives thereof) and/or optionally contains one or more (such as 2, 3, 4 or 5) locked nucleotide units, such as a unit selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA (all in either alpha or beta configuration) and derivatives thereof; B represents one nucleotide unit or a sequence of nucleotide units, with the proviso that at 20 least one nucleotide unit in B has a 2'-deoxy-erythro-pentofuranosyl sugar moiety or a ribo-pentofuranosyl moiety; and C represents a sequence comprising at least two consecutively located locked nucleotide units, at least one of which is an alpha-L-oxy-LNA unit, and which sequence optionally contains one or more (such as 2, 3, 4 or 5) non-locked nucleotide units (such as 25 deoxyribonucleotide units, ribonucleotide units or derivatives thereof) and/or optionally contains one or more (such as 2, 3, 4 or 5) locked nucleotide units, such as a unit selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA (all in either alpha or beta configuration) and derivatives thereof. It is preferred that A has a length of 2-10 (preferably 2, 3, 4, 5, 6, 7, or 8) nucleotide units; B has a length of 1-10 (preferably 5, 6, 30 7, or 8) nucleotide units; C has a length of 2-10 (preferably 2, 3, 4, 5, 6, 7, or 8) nucleotide units; so that the overall length of the construct is 8-30 (preferably 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) nucleotide units. 35 An other interesting embodiment is a construct in which A represents a sequence of nucleotide units comprising at least three consecutively located locked nucleotide units, at least one of said locked nucleotide units being selected from the group consisting of alpha L-oxy-LNA and derivatives thereof; C represents a sequence of nucleotide units comprising at least three consecutively located locked nucleotide units, at least one of said locked 11 nucleotide units being selected from the group consisting of alpha-L-oxy-LNA and derivatives thereof; and/or B represents a sequence of least 2 nucleotide units (such as 3, 4, 5, 6, 7, 8, 9 or 10 units), which sequence in addition to the nucleotide unit(s) having 2' deoxy-erythro-pentofuranosyl sugar moiety(ies) and/or ribo-pentofuranosyl moiety(ies), 5 comprises nucleotide units which are selected independently from the group consisting of: locked nucleotide units (such as alpha-L-oxy-, -thio-, or -amino- nucleotide units) and derivatives thereof. Especially preferred is a construct in which A and C comprises at least one alpha-L-oxy-LNA or alpha-L-thio-LNA unit located adjacent to B. 10 In a further embodiment, the invention relates to an oligonucleotide which has the formula (in 5' to 3' order): A-B-C-D, in which A represents a sequence of locked nucleotide units; B represents a sequence of non-locked nucleotide units, preferably at least one unit has a 2' deoxy pentofuranose sugar moiety, in which sequence 1 or 2 nucleotide units optionally are substituted with locked nucleotide units, preferably alpha-L-oxy-LNA; C represents a 15 sequence of locked nucleotide units; and D represents a non-locked nucleotide unit or a sequence of non-locked nucleotide units. It is preferred that A has a length of 2-6 (preferably 3, 4 or 5) nucleotide units; B has a length of 4-12 (preferably 6, 7, 8, 9, 10 or 11) nucleotide units; C has a length of 1-5 (preferably 2, 3, or 4) nucleotide units; D has a lenght of 1-3 (preferably 1-2) nucleotide units; and that the overall length of the construct 20 is 8-26 (preferably 12-21) nucleotide units. In presently preferred construct, A has a length of 4 nucleotide units; B has a length of 7-9, preferably 8, nucleotide units; C has a length of 3 nucleotide units; D has a length of 1 nucleotide unit; and the overall length of the construct is 15-17 (preferably 16) nucleotide units. It is further preferred that the locked nucleotide units in A and C are beta-D-oxy-LNA units or derivatives thereof. 25 The oligonucleotide constructs according to the invention can contain naturally occurring phosphordiester internucleoside linkages, as well as other internucleoside linkages as defined in this specification. Examples on internucleoside linkages are linkages selected from the group consisting of -O-P(0) 2 -0- , -O-P(0,S)-0-, -O-P(S) 2 -0-, -NRH-P(0) 2 -0-, 30 P(O,NRH)-O, -O-PO(R")-0-, -O-PO(CH 3 )-O-, and -O-PO(NHRN)-0-, where RH is selected form hydrogen and C 1 4 -alkyl, and R" is selected from C 1 6 -alkyl and phenyl. In a further embodiment, the invention relates to an oligonucleotide construct which comprises at least one locked nucleotide unit selected from the group consisting of amino 35 LNA, thio-LNA (both in either alpha-L or beta-D configuration), alpha-L-oxy-LNA, and derivatives thereof; wherein at least one of the linkages between the nucleotide units is different from the natural occurring phosphordiester (-0-P(0) 2 -0-) linker. Constructs in which the internucleoside linkage (between 3' carbon and 5' carbon on adjacent (3', 5' dideoxy) nucleosides) selected from the group consisting of: -O-P(0,S)-0-, -O-P(S) 2 -0-, - 12 NRH-P(0) 2 O-, -O-P(O,NRH)-0-, -O-PO(R")-O-, -O-PO(CH 3 )-O-, and -O-PO(NHRN)-O, where RH is selected form hydrogen and C 1 4 -alkyl, and R" is selected from C 1 6 -alkyl and phenyl, is presently preferred, and the phoshorothioate internucleoside linkage is presently most preferred. 5 An embodiment of the oligonucleotide constructs according to the invention relates to such constructs that are able to mediate enzymatic inactivation (at least partly) of the target nucleic acid (eg. a RNA molecule) for the construct. Constructs that mediate RNase H cutting of the target are within the scope of the present invention. Thus, the present 10 invention relates to constructs that are able to recruit RNase, especially constructs in which sequence B represents a sequence of nucleotide units that makes the construct able to recruit RNase H when hybridised to a target nucleic acid (such as RNA, mRNA). It should be understood that the invention also relates to a pharmaceutical composition 15 which comprises a least one antisense oligonucleotide construct of the invention as an active ingredient. It should be understood that the pharmaceutical composition according to the invention optionally comprises a pharmaceutical carrier, and that the pharmaceutical composition optionally comprises further antisense compounds, chemotherapeutic compounds, antiinflammatory compounds and/or antiviral compounds. 20 The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be (a) oral (b) pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, (c) 25 topical including epidermal, transdermal, ophthalmic and to mucous membranes including vaginal and rectal delivery; or (d) parenteral including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. 30 Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, sprays, suppositories, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful. Preferred topical formulations include those in which the 35 oligonucleotides of the invention are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Compositions and formulations for oral administration include but is not restricted to powders or granules, microparticulates, nanoparticulates, suspensions or solutions in water or non-aqueous media, capsules, gel capsules, sachets, tablets or minitablets.
13 Compositions and formulations for parenteral, intrathecal or intraventricular administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients. 5 Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self- emulsifying solids and self-emulsifying semisolids. 10 The pharmaceutical formulations of the present invention, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In 15 general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. The compositions of the present invention may be formulated into any of many possible 20 dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels and suppositories. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. Aqueous suspensions may further contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may 25 also contain stabilizers. The antisense nucleotide constructs of the invention encompass, in their brodest scope, any pharmaceutically acceptable salts, esters, or salts of such esters. Furthermore encompasses the invention any other compound, which, upon administration to an animal 30 or a human, is capable of directly or indirectly providing the biologically active metabolite or residue thereof. The invention therefore also encompasses prodrugs of the compounds of the invention and pharmaceutically acceptable salts of such prodrugs, and other bioequivalents. The term prodrug indicates a therapeutic agent that is prepared in an inactive form and that is converted to an active form, a drug, within the body or cells 35 thereof. The pharmaceutically acceptable salts include but are not limited to salts formed with cations; acid addition salts formed with inorganic acids salts formed with organic acids such as, and salts formed from elemental anions.
14 In one embodiment, the present invention employs various penetration enhancers to effect the efficient delivery of nucleic acids, particularly oligonucleotides, to the skin of animals or humans. Most drugs are present in solution in both ionized and nonionized forms. However, usually only lipid soluble or lipophilic drugs readily cross cell membranes. It has 5 been discovered that even non-lipophilic drugs may cross cell membranes if the membrane to be crossed is treated with a penetration enhancer. In addition to aiding the diffusion of non-lipophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs. 10 Pharmaceutical compositions of the invention include a pharmaceutical carrier that may contain a variety of components that provide a variety of functions, including regulation of drug concentration, regulation of solubility, chemical stabilization, regulation of viscosity, absorption enhancement, regulation of pH, and the like. The pharmaceutical carrier may comprise a suitable liquid vehicle or excipient and an optional auxiliary additive or 15 additives. The liquid vehicles and excipients are conventional and commercially available. Illustrative thereof are distilled water, physiological saline, aqueous solutions of dextrose, and the like. For water soluble formulations, the pharmaceutical composition preferably includes a buffer such as a phosphate buffer, or other organic acid salt. For formulations containing weakly soluble antisense compounds, micro-emulsions may be employed. 20 Other components may include antioxidants, such as ascorbic acid, hydrophilic polymers, such as, monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, dextrins, chelating agents, and like components well known to those in the pharmaceutical sciences. The oligonucleotides may be encapsulated in liposomes for therapeutic delivery. 25 In a certain embodiment, the present invention provides pharmaceutical compositions containing (a) one or more antisense compounds and (b) one or more other chemotherapeutic agents which function by a non-antisense mechanism. When used with the compounds of the invention, such chemotherapeutic agents may be used individually 30 (e.g., mithramycin and oligonucleotide), sequentially (e.g., mithramycin and oligonucleotide for a period of time followed by another agent and oligonucleotide), or in combination with one or more other such chemotherapeutic agents or in combination with radiotherapy. 35 Anti-inflammatory drugs, including but not limited to nonsteroidal anti- inflammatory drugs and corticosteroids, and antiviral drugs, may also be combined in compositions of the invention. Two or more combined compounds may be used together or sequentially.
15 In another embodiment, compositions of the invention may contain one or more antisense compounds, particularly oligonucleotides, targeted to a first nucleic acid and one or more additional antisense compounds targeted to a second nucleic acid target. Two or more combined compounds may be used together or sequentially. 5 Dosing is dependent on severity and responsiveness of the disease state to be treated, and the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. 10 Optimum dosages may vary depending on the relative potency of individual oligonucleotides. Generally it can be estimated based on EC50s found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 ug to 25 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once 15 every 2 to 10 years. The repetition rates for dosing can be estimated based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state. 20 The LNA containing antisense compounds of the present invention can be utilized for diagnostics, therapeutics, prophylaxis and as research reagents and kits. For therapeutics, an animal or a human, suspected of having a disease or disorder, which can be treated by modulating the expression of a gene by administering antisense compounds in accordance with this invention. Further provided are methods of treating an animal and humans, 25 suspected of having or being prone to a disease or condition, associated with expression of a target gene by administering a therapeutically or prophylactically effective amount of one or more of the antisense compounds or compositions of the invention. Examples of such a diseases are for example different types of cancer, infectious and inflammatory diseases. 30 In a certain embodiment, the present invention relates to a method of synthesis of a pharmaceutical compositions, a oligonucleotides or a construct according to the present invention. Definitions The term "nucleotide sequence" or "sequence" comprises a plurality (ie. more than one) 35 nucleosides (or derivatives thereof), in which sequence each two adjacent nucleosides (or derivatives thereof) are linked by an internucleoside linker. When the length of a sequence are defined by a range (such as from 2-10 nucleotide units), the range are understood to 16 comprise all integers in that range, i.e. "a sequence of 2-10 nucleotide units" comprises sequences having 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide units. In the present context, the term "oligonucleotide" (or oligo, oligomer) means a successive 5 chain of nucleoside units (i.e. glycosides of heterocyclic bases) connected via internucleoside linkages. By the term "unit" is understood a monomer. 10 The term "at least one" comprises the integers larger than or equal to 1, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 and so forth. The term "locked nucleotide" comprises nucleotides in which the 2' deoxy ribose sugar moiety is modified by introduction of a structure containing a heteroatom bridging from the 15 2' to the 4' carbon atoms. The term includes nucleotides having the following substructures (the oxygen at the 3' and 5' ends illustrates examples of the starting point of the internucleoside linkages): beta-D-LNA derivatives: O Base 0 (C H2)n 6 -X 0 20 alpha-L-LNA derivatives n(H2C)
-
Base
O
17 In both structures, X represents 0, S or N-R (R= H; C1-C6 alkyl such as methyl, ethyl, propyl, i-propyl, butyl, i-butyl, t-butyl and pentyl); and n is an integer 1, 2 or 3, so that the group -(CH2)n- comprises methylen, ethylen or 5 propylen groups. In these alkylene groups (and the -N(C1-C6 alkyl)- group), one or more H atoms can be replaced with substituents, such as one or more substituents selected from the group consisting of halogen atoms (Cl, F, Br, I), Nitro, C1-6 alkyl or C1-6 alkoxy, both optionally halogenated. 10 In the present context, the term "C 1 6 -alkyl" means a linear, cyclic or branched hydrocarbon group having 1 to 6 carbon atoms, such as methyl, ethyl, propyl, iso-propyl, butyl, tert-butyl, iso-butyl, pentyl, cyclopentyl, hexyl, cyclohexyl, in particular methyl, ethyl, propyl, iso-propyl, tert-butyl, iso-butyl and cyclohexyl. "C 1 6 -alkoxy" means -0-(C1 6-alkyl). 15 The term "non-locked nucleotide" comprises nucleotides that do not contain a bridging structure in the ribose sugar moiety. Thus, the term comprises DNA and RNA nucleotide monomers (phosphorylated adenosine, guanosine, uridine, cytidine, deoxyadenosine, deoxyguanosine, deoxythymidine, deoxycytidine) and derivatives thereof as well as other 20 nucleotides having a 2'-deoxy-erythro-pentofuranosyl sugar moiety or a ribo pentofuranosyl moiety. The term "thio-LNA" comprises a locked nucleotide in which X in the above formulas represents S, and n is 1. Thio-LNA can be in both beta-D and alpha-L-configuration. 25 The term "amino-LNA" comprises a locked nucleotide in which X in the above formulas represents -NR-, and n is 1. Amino-LNA can be in both beta-D and alpha-L-configuration. The term "oxy-LNA" comprises a locked nucleotide in which X in the above formulas 30 represents 0 and n is 1. Oxy-LNA can be in both beta-D and alpha-L-configuration. By the term "alpha-L-LNA" as used herein is normally understood alpha-L-oxy-LNA (n=1 in the bridging group), and by the term "LNA" as used herein is understood beta-D-oxy-LNA monomer wherein n in the bridging group is 1. 35 However, derivatives of the above locked LNA's comprise nucleotides in which n is an other integer than 1.
18 By the term "derivatives thereof" in connection with nucleotides (e.g. LNA and derivatives thereof) is understood that the nucleotide, in addition to the bridging of the furan ring, can be further derivatized. For example, the base of the nucleotide, in addition to adenine, guanine, cytosine, uracil and thymine, can be a derivative thereof, or the base can be 5 substituted with other bases. Such bases includes heterocyclic analogues and tautomers thereof. Illustrative examples of nucleobases are xanthine, diaminopurine, 8-oxo-N methyladenine, 7-deazaxanthine, 7-deazaguanine, N4,N 4 -ethanocytosin, N 6
N
6 -ethano-2,6 diaminopurine, 5-methylcytosine, 5-(C-C 6 )-alkynylcytosine, 5-fluorouracil, 5-bromouracil, pseudoisocytosine, 2-hydroxy-5-methyl-4-triazolopyridin, isocytosine, isoguanin, inosine, 10 N 6 -alylpurines, N 6 -acylpurines, N 6 - benzylpurine, N 6 -halopurine, N 6 -vinylpurine, N 6 acetylenic purine, N 6 -acyl purine, N 6 -hydroxyalkyl purine, N 6 -thioalkyl purine, N 2 alkylpurines, N 4 -alkylpyrimidines, N 4 - acylpyrimidines, N 4 -benzylpurine, N 4 halopyrimidines, N 4 -vinylpyrimidines, N 4 - acetylenic pyrimidines, N 4 -acyl pyrimidines, N 4 hydroxyalkyl pyrimidines, N 6 -thioalkyl pyrimidines, thymine, cytosine, 6-azapyrimidine, 15 including 6-azacytosine, 2- and/or 4- mercaptopyrimidine, uracil, C 5 -alkylpyrimidines, C 5 benzylpyrimidines, C 5 -halopyrimidines, C 5 -vinylpyrimidine, C 5 -acetylenic pyrimidine, C 5 acyl pyrimidine, C 5 -hydroxyalkyl purine, C 5 - amidopyrimidine, C5-cyanopyrimidine, C nitropyrimidine, C5-aminopyrimdine, N 2 - alkylpurines, N 2 -alkyl-6-thiopurines, 5 azacytidinyl, 5- azauracilyl, trazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and 20 pyrazolopyrimidinyl. Functional oxygen and nitrogen groups on the base can be protected as necessary or desired. Suitable protecting groups are well known to those skilled in the art, and included trimethylsilyl, dimethylhexylsilyl, t-butyldimenthylsilyl, and t butyldiphenylsilyl, trityl, alkyl groups, acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl. Preferred bases include cytosine, methyl cytosine, 25 uracil, thymine, adenine and guanine. In addition to the derivatisation of the base, both locked and non-locked nucleotides can be derivatised on the ribose moiety. For example, a 2' substituent can be introduced, such as a substituent selected from the group consisting of halogen (such as fluor), C1-C9 alkoxy (such as methoxy, ethoxy, n-propoxy or i propoxy), C1-C9 aminoalkoxy (such as aminomethoxy and aminoethoxy), allyloxy, 30 imidazolealkoxy, and polyethyleneglycol, or a 5' substituent (such as a substituent as defined above for the 2' position) can be introduced. By the terms "internucleoside linkage" and "linkage between the nucleotide units" (which is used interchangeably) are to be understood the divalent linker group that forms the 35 covalent linking of two adjacent nucleosides, between the 3' carbon atom on the first nucleoside and the 5' carbon atom on the second nucleoside (said nucleosides being 3',5' dideoxy). The oligonucleotides of the present invention comprises sequences in which both locked and non-locked nucleotides independently can be derivatised on the internucleoside linkage which is a linkage consisting of preferably 2 to 4 groups/atoms selected from -CH 2
-
19 -O-, -S-, -N RH-, >C=0, >C=N RH, >CS ni)2-, -SO-, -S(0)2-, -P(0)2-, -PO(BH3), -P(O,S)-, -P(S) 2 -, -PO(R")-, -PO(OCH 3 )-, and -PO(NHRH)_, where RH is selected form hydrogen and C 1
.
6 -alkyl, and R" is selected from C 1
.
6 -alkyl and phenyl. Illustrative examples of such internucleoside linkages are -CH 2
-CH
2
-CH
2 -, -CH 2
-CO-CH
2
-,,-CH
2
-CHOH
5 CH 2 -, -O-CH 2 -0-, -O-CH 2
-CH
2 -, -O-CH 2 -CH(R5)-, -CH 2
-CH
2 -0-, -NRH-CH 2
-CH
2 -, -CH 2
-CH
2 NRH-, -CH 2
-NRH-CH
2 -, -- CH2-CH2-N -NRHCO , -NRH-CO-NRH, -NRH-CSNRH -NRH-C(=NRH)-NRH, -N -CO-CH2-N -O-CO-O-, -O-CO-CH 2 -0-, -O-CH 2 -CO-O-, -CH 2 CO-NRH-, -O-CO-NRH-, -NRH-CO-CH 2 -, -O-CH2-CO-NRp -O-CH2-CH2-NR,_ -CH=N-O-,
-CH
2 -NRH-0-, -CH 2 -O-N(R5)-, -CH 2 -0-NRH-, -CO-NRH-CH 2 -, -CH 2 -NRH-0-, -CH 2 -NRH-CO-, 10 -O-NRH-CH 2 -, -O-NRH-, -O-CH 2 -S-, -S-CH 2 -O-, -CH 2
-CH
2 -S-, -O-CH 2
-CH
2 -S-, -S-CH 2 CH(R5)-, -S-CH 2
-CH
2 -, -S-CH 2
-CH
2 -0-, -S-CH 2
-CH
2 -S-, -CH 2
-S-CH
2 -, -CH 2
-SO-CH
2 -, -CH 2 S0 2
-CH
2 -, -0-SO-0-, -O-S(O)2-0-, -O-S(0) 2
-CH
2 -, -O-S(O) 2 -NRH-, -N RH-S(0) 2
-CH
2 -, -O-S(0) 2
-CH
2 -, -O-P(O) 2 -0-, -O-P(O,S)-O-, -O-P(S) 2 -0-, -S-P(O) 2 -0-, -S-P(O,S)-O-, -S
P(S)
2 -0-, -O-P(O) 2 -S-, -O-P(O,S)-S-, -O-P(S) 2 -S-, -S-P(O) 2 -S-, -S-P(O,S)-S-, -S-P(S) 2 -S-, 15 -O-PO(R")-O-, -O-PO(OCH 3 )-0-, -O-PO(OCH 2
CH
3 )-0-, -O-PO(OCH 2
CH
2 S-R)-O-,
-O-PO(BH
3 )-O-, -O-PO(NH RN)-0-, -O-P(0) 2 -NRH-, -NRH-P(0) 2 -0-, -O-P(O,NRH)-0-, -CH 2 P(O) 2 -0-, -O-P(0) 2
-CH
2 -, and -O-Si(R") 2 -0-; where R5 is selected from hydrogen and C 1
.
6 alkyl, RH is selected form hydrogen and C 1
.
6 -alkyl, and R" is selected from C 1
.
6 -alkyl and phenyl. 20 CH2-CO-NR- -CH2-NRH-0, -S-CH2-0-, -O-P(0)2-0-, -O-P(0,S)-0-, -O-P(S)2-0-, -NRH_
P(O)
2 -0-, -O-P(O,NRH)-0-, -O-PO(R")-O-, -O-PO(CH 3 )-O-, and -O-PO(NHRN)-0-, where RH is selected from hydrogen and C 1
.
6 -alkyl, and R" is selected from C 1
.
6 -alkyl and phenyl, are especially preferred. 25 The nucleotides units may also contain a 3'-Terminal group or a 5'-terminal group, preferably -OH. By the term "able to recruit RNase H" is understood that the an oligonucleotide construct, 30 in order to elicit RNase H enzyme cleavage of a target nucleic acid (such as target mRNA), must include a segment or subsequence that is of DNA type. This means that at least some nucleotide units of the oligonucleotide construct (or a subsequence thereof) must have 2'-deoxy-erythro-pentofuranosyl sugar moieties. A subsequence having more than three consecutive, linked 2'-deoxy-erythro-pentofuranosyl containing nucleotide units 35 likely is necessary in order to elicit RNase H activity upon hybridisation of an oligonucleotide construct of the invention with a target nucleic acid, such as a RNA. Preferably, a sequence which is able to recruit RNase H contains more than three consecutively located nucleotides having 2'-deoxy-erythro-pentofuranosyl sugar moieties, such as 4,5, 6, 7, 8 or more units. However, such a subsequence of consecutively located 20 nucleotides having 2'-deoxy-erythro-pentofuranosyl sugar moieties can by spiked (ie. one or more (such as 1, 2, 3, 4, or more) nucleotides being replaced) with other nucleotides, preferably alpha-L-oxy, thio- or amino-LNA units or derivatives thereof. 5 The term "pharmaceutically acceptable salt" is well known to the person skilled in the art. Examples of such pharmaceutically acceptable salts are the iodide, acetate, phenylacetate, trifluoroacetate, acrylate, ascorbate, benzoate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, o-acetoxybenzoate, naphthalene-2 10 benzoate, bromide, isobutyrate, phenylbutyrate, g-hydroxybutyrate, b-hydroxybutyrate, butyne-1,4-dioate, hexyne-1,4-dioate, hexyne-1,6-dioate, caproate, caprylate, chloride, cinnamate, citrate, decanoate, formate, fumarate, glycollate, heptanoate, hippurate, lactate, malate, maleate, hydroxymaleate, malonate, mandelate, mesylate, nicotinate, isonicotinate, nitrate, oxalate, phthalate, terephthalate, phosphate, 15 monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, propiolate, propionate, phenylpropionate, salicylate, sebacate, succinate, suberate, sulfate, bisulfate, pyrosulfate, sulfite, bisulfite, sulfonate, benzenesulfonate, p bromophenylsulfonate, chlorobenzenesulfonate, propanesulfonate, ethanesulfonate, 2 hydroxyethanesulfonate, methanesulfonate, naphthalene-I -sulfonate, naphthalene-2 20 sulfonate, p-toluenesulfonate, xylenesulfonate, tartarate, and the like. Legends to figures Figure 1: Stability of oligonucleotides containing beta-D-amino-LNA against SVPD. (Capital letters are LNA, TN stands for beta-D-amino-LNA and small letters are DNA. The oligonucleotide is synthesized on deoxynucleoside-support, t.) 25 Figure 2: Subcellular distribution in MiaPacaII cells of FAM-labeled oligonucleotides (2740, 2774, 2752, 2746) transfected with Lipofectamine2000. Figure 3: Comparison of the uptake of titriated oligonucleotides (thio=2748; 30 amino=2754; oxy=2742) in MiaPacaII and 15PC3 cells at different oligonucleotide concentration with Lipofectamine2000 as transfection agent. Figure 4: Down-regulation of Luciferase expression of oligonucleotides gapmers containing beta-D-amino-LNA or beta-D-thio-LNA and the corresponding beta-D-oxy-LNA gapmer 35 control at 50 nM oligonucleotide concentration.
21 Figure 5: Northern blot analysis of oligonucleotides containing beta-D-amino-LNA (2754 and 2755), beta-D-thio-LNA (2748 and 2749) or beta-D-oxy-LNA (2742) at 400 and 800 nM in 15PC3 cells transfected with Lipofectamine2000. Figure 6: Northern blot analysis of oligonucleotides containing beta-D-amino-LNA 5 (2754), beta-D-thio-LNA (2748), alpha-L-oxy-LNA (2776) or beta-D-oxy-LNA (2742) at 50-400 nM in 15PC3 cells transfected with Lipofectamine2000; comparison with the corresponding mismatch control at 400 nM. Mismatch controls (thio=2750; amino=2756; alpha=2778) were also analyzed at 30-90 nM and compared with the corresponding match at 30 nM. Table containing Northern blot analysis of 10 oligonucleotides containing beta-D-amino-LNA (2754), alpha-L-oxy-LNA (2776) and beta-D-oxy-LNA (2742) at 5-40 nM in 15PC3 cells transfected with Lipofectamine2000; comparison with the corresponding mismatch controls at 20 nM. Figure 7: Serum clearance and biodistribution of titriated 2754=amino, 2748=thio and 15 2742=oxy after 30 min of intravenous bolus injection. 2131 is an oligonucleotide gapmer containing beta-D-oxy-LNA used as a reference. Figure 8: Biodistribution of titriated 2754=amino, 2748=thio and 2742=oxy after 14 days of continuous administration at a 2.5 mg/Kg/day dosage using Alzet osmotic 20 minipumps. Figure 9: Electrophoresis analysis of 32 P-labelled target RNA degradation products mediated by RNaseH and an oligonucleotide containing beta-D-amino-LNA. Aliquots taken at 0, 10, 20 and 30 min for each design. In the drawings, the line is DNA, the rectangle 25 beta-D-amino- or -thio-LNA. Figure 10: Stability of oligonucleotides containing beta-D-thio-LNA against SVPD. (Capital letters are LNA, TS stands for beta-D-thio-LNA and small letters are DNA. The oligonucleotide is synthesized on deoxynucleoside-support, t.) 30 Figure 11: FACS analysis of oligonucleotides containing beta-D-thio-LNA and the corresponding controls. Figure 12: Stability of oligonucleotides containing alpha-L-oxy-LNA against SVPD. (Capital 35 letters are LNA, Ta stands for alpha-L-oxy-LNA and small letters are DNA. The oligonucleotide is synthesized on deoxynucleoside-support, t.) 22 Figure 13: Stability of different oligonucleotides (t 16 , t, 12 , T 16 , Tai 5 T) against S1 endonuclease. (Capital letters are LNA, T" stands for alpha-L-oxy-LNA and small letters are DNA. The oligonucleotide is synthesized on oxy-LNA-support, T.) 5 Figure 14: FACS analysis of oligonucleotides containing alpha-L-oxy-LNA, and the corresponding controls. Figure 15: Gapmers including alpha-L-oxy-LNA (shadowed in gray). 10 Figure 16: Down-regulation of Luciferase expression of oligonucleotides containing alpha L-oxy-LNA at 50nM oligonucleotide concentration. Figure 17: Different mixmers containing alpha-L-oxy-LNA. The numbers stand for the alternate contiguous stretch of DNA or LNA. In the drawing, the line is DNA, the rectangle 15 beta-D-oxy-LNA, the gray shadow corresponds to alpha-L-oxy-LNA residues. Figure 18: Other mixmers containing alpha-L-oxy-LNA. The numbers stand for the alternate contiguous stretch of DNA or alpha-L-oxy-LNA. In the drawing, the line is DNA, the gray shadow corresponds to alpha-L-oxy-LNA residues. 20 Figure 19: Electrophoresis analysis of 32 P-labelled target RNA degradation products mediated by RNaseH and an oligonucleotide containing alpha-L-oxy-LNA. Aliquots taken at 0, 10, 20 and 30 min for each design. In the drawings, the line is DNA, the rectangle beta D-oxy-LNA, the gray shadow corresponds to alpha-L-oxy-LNA residues. 25 Figure 20: Tumor growth in nude mice treated with the indicated doses for 14 days using Alzet osmotic minipumps, both for MiaPacaII and 15PC3. Figure 21: ASAT, ALAT and Alkaline phosphatase levels in mice serum after 14-day 30 treatment using Alzet osmotic minipumps with the indicated oligonucleotides and at the indicated concentrations. 2722 and 2713 are oligonucleotides not relevant to this study. Figure 22: Monitoring the body temperature of the mice during the in vivo 35 experiment. 2722 and 2713 are oligonucleotides not relevant to this study.
23 Figure 23: Special constructs with beta-D- oxy-LNA. The numbers stand for the alternate contiguous stretch of DNA and beta-D-oxy-LNA. In the drawing, the line is DNA, the rectangle is beta-D-oxy-LNA. 5 Figure 24: Down-regulation of Luciferase expression of special constructs containing beta D-oxy-LNA (designs 3-9-3-1) at 2nM oligonucleotide concentration. Experimental Oligonucleotide synthesis Oligonucleotides were synthesized using the phosphoramidite approach on an Expedite 10 8900/MOSS synthesizer (Multiple Olionucleotide Synthesis System) at 1 pM scale. At the end of the synthesis (DMT-on) the oligonucleotides were cleaved from the solid support using aqueous ammonia for 1 h at room temperature, and further deprotected for 4 h at 650C. The crudes were purified by reverse phase HPLC. After the removal of the DMT group, the oligonucleotides were characterized by AE-HPLC or RP-HPLC, and the structure 15 further confirmed by ESI. 3'-Exonuclease stability study Snake venom phosphodiesterase (SVPD, Amersham Pharmacia) assays were performed using 26 pg/mL oligonucleotide, 0.3 pg/mL enzyme at 37 0 C in a buffer of 50 mM Tris-HCI, 10 mM MgC1 2 , pH 8. The enzyme was shown to maintain its activity under these conditions 20 for at least 2 h. Aliquots of the enzymatic digestion were removed at the indicated times, quenched by heat denaturation for 3 min and stored at -20 0 C until analysis by RP-HPLC. S1-Endonuclease stability study S1 endonuclease (Amersham Pharmacia) assays were performed using 1.5 pmol oligonucleotide and 16 U/mL enzyme at 37 0 C in a buffer of 30 mM NaOAc, 100 mM NaCI, 1 25 mM ZnSO 4 , pH 4.6. The enzyme was shown to maintain its activity under these conditions for at least 2 h. Aliquots of the enzymatic digestion were removed at the indicated times, quenched by freezing-drying, and stored at -20 0 C until analysis by either RP-HPLC and ES MS or polyacrylamide electrophoresis. Luciferase assay 30 The X1/5 Hela cell line (ECACC Ref. No: 95051229), which is stably transfected with a "tet off" luciferase system, was used. In the absence of tetracycline the luciferase gene is expressed constitutively. The expression can be measured as light in a luminometer, when the luciferase substrate, luciferin has been added. The X1/5 Hela cell line was grown in Minimun Essential Medium Eagle (Sigma M2279) 35 supplemented with 1x Non Essential Amino Acid (Sigma M7145), 1x Glutamax I 24 (Invitrogen 35050-038), 10 % FBS calf serum, 25 pg/ml Gentamicin (Sigma G1397), 500 pg/ml G418 (Invitrogen 10131-027) and 300 pg/ml Hygromycin B (invitrogen 10687-010). The X1/5 Hela cells were seeded at a density of 8000 cells per well in a white 96 well plate (Nunc 136101) the day before the transfection. Before the transfection, the cells were 5 washed one time with OptiMEM (Invitrogen) followed by addition of 40 pl of OptiMEM with 2pg/ml of Lipofectamine2000 (Invitrogen). The cells were incubated for 7 minutes before addition of the oligonucleotides. 10 pl of oligonucleotide solutions were added and the cells were incubated for 4 hours at 371C and 5 % C0 2 . After the 4 hours of incubation the cells were washed once in OptiMEM and growth medium was added (100 pl). The luciferase 10 expression was measure the next day. Luciferase expression was measured with the Steady-Glo luciferase assay system from Promega. 100 pl of the Steady-Glo reagent was added to each well and the plate was shaken for 30s at 700rpm. The plate was read in Luminoskan Ascent instrument from ThermoLabsystems after 8min of incubation to complete total lysis of the cells. The 15 luciferase expression is measured as Relative Light Units per seconds (RLU/s). The data was processed in the Ascent software (v2.6) and graphs were drawn in SigmaPlot2001. RNaseH assay 25 nM RNA was incubated in the presence of a 10-fold excess of various complementary oligonucleotides in 1 x TMK-glutamate buffer (20 mM Tris acetate, 10 mM magnesium 20 acetate and 200 mM potassium glutamate, pH 7.25) supplied with 1 mM DTT in a reaction volume of 40 pl. The reactions were preincubated for 3 minutes at 651C followed by 15 minutes at 371C before addition of RNase H (Promega, Cat.# 4285). 0.2 U of RNase H was added, and samples were withdrawn (6 pl) to formamide dye (3 pl) on ice at the time points 0, 10, 20 and 30 minutes after RNase H addition. 3 pl of the 0, 10, 20 and 30 25 minutes samples were loaded on a 15 % polyacrylamide gel containing 6M urea and 0.9 x Tris borate/EDTA buffer. The gel was 0.4 mm thick and ran at 35 watt as the limiting parameter for 2 hours. The gel was dried for 60 minutes at 801C, followed by ON exposure on Kodak phosphorscreen. The Kodak phosphorscreen was read in a Bio-Rad FX instrument and the result was analysed in Bio-Rad software Quantity One. 30 Cellular assay: Luciferase target Cell Culture: Cell lines 15PC3 (human prostate cancer) and X1/5 (HeLa cells stably transfected with a Tet-Off luciferase construct) were used, 15PC3 were kindly donated by F. Baas, Neurozintuigen lab, Amsterdam, The Netherlands, X1/5 were purchased from ECACC. 15PC3 were maintained in DMEM + 10% FCS + glutamax + gentamicin and X1/5 35 were maintained in DMEM + 10% FCS + glutamax + gentamicin + hygromycin + G418 and both cell lines were passaged twice weekly.
25 Transfection: Cells were seeded at 150000 cells pr. well in 12-well plates the day before transfection. For transfection with lipid, Lipofectamine 2000 (GIBCO BRL) was mixed with OptiMem and 300 pl of the mixture was added to each well and incubated for 7 min. before addition of 100pl oligo diluted in OptiMem. For each cell line, the optimal Lipofectamine 5 2000 was determined, for X1/5, the optimal Lipofectamine concentration was 2 pg/ml and for 15PC3 the optimal concentration was 10 pg/ml. For transfection without lipid, the cells were washed in OptiMem (GIBCO BRL) and 300 pl OptiMem was added to each well. Working stocks of 200 pM were prepared of each oligonucleotide to be tested and added to each well obtaining the desired concentration. 10 For mock controls, oligonucleotide was substituted with water in both protocols. The cells were incubated with the oligonucleotide for 4 h at 37oC and 5% CO 2 in a humidified atmosphere and subsequently washed in OptiMem before complete growth medium was added. The cells were incubated for an additional 20h. For FACS analysis, cells were harvested by trypsination and washed twice in Cell Wash 15 (BD) and resuspended in 1x Cell Fix (BD). FACS analysis: FACS analysis was performed on a FACSCalibur (BD), settings were adjusted on mock controls. Data analysis was performed using the Cell Quest Pro software (BD). Assisted cellular uptake 20 Transfections were performed in 6 well culture plates on microscope glass coverslips with FAM-labeled oligonucleotides at 400nM. Transfections were done with either DAC30 (Eurogentec) or Lipofectamine 2000 as liposomal transfection agents for 5h in serum free DMEM at 37 0 C. Immediately after the transfection period, the cells were washed with PBS and fixed with 4% paraformaldehyde. 25 Cell lines: Ha-Ras target Prostate cancer cell line 15PC3 and pancreatic carcinoma cell line MiaPacaII were maintained by serial passage in Dulbecco's modified Eagle's medium (DMEM). Cells were grown at 37 0 C and 5 % C0 2 . Media were supplemented with 10% fetal calf serum, 2 mM L glutamine, 100 U/mL penicillin and 100 pg/mL streptomycin. 30 Transfections: Ha-Ras target Cell transfections were performed with 15PC3 cells plated in 6 well culture plates. The cells were plated ( 7 0% confluent) the day before transfection. The transfections were usually performed using lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions, except for using serum free DMEM. Cells were transfected for 5 hours. 35 Afterwards the medium was replaced with fresh DMEM. We also compared Lipofectamine 2000 with DAC30 (Eurogentec). When DAC30 was used the protocol described in Ten Asbroek et al. (NAR 28, 1133-1138) was followed.
26 For Fluorescence studies the cells were plated on glass cover slips in 6 well culture plates. Transfections were performed as described above but using FAM labeled oligonucleotides. At the time of analysis, the cells were fixed on the glass in 4% paraformaldehyde and sealed on microscope glass in Vectashield mounting medium (Vector Laboratories Inc.). 5 Fluorescence microscopy was done with a Vanox Microscope and appropriate filtres. mRNA analysis: Ha-Ras target After 20 hours the cells were harvested in TRIZOL (Invitrogen), 1ml per well. The RNA was isolated according to the manufacturer's instructions for TRIZOL. The RNA was separated on glyoxal gels containing 1% agarose following standard protocols. RNA 10 was subsequently blotted onto Hybond N+ membrane (Amersham) in 20x SSC. After the transfer, the RNA was UV cross-linked, and then the membrane was baked for 2 hours at 80 0 C. Hybridizations and post-hybridization washes were done according to Church and Gilbert (PNAS 81, 1991-1995). The Ha-Ras probe used was generated using Ha-Ras primers according to Sharpe et al. (J.AM.Soc. Nephrol. 11 1600-1606) cloned into pGEM-T 15 Easy vector (Promega). The loadings of the Ha-Ras mRNA levels were corrected by using a 28S probe as described in Ten Asbroek et al. (NAR 28, 1133-1138). Biodistribution studies The animal experiments were approved by the ethical committee and are registered under No. DNL19. 20 Tritium labeling of oligonucleotides was performed using the heat exchange method described by Graham et al. (Graham, M.J., Freier, S.M., Crooke, R.M., Ecker, D.J., Maslova, R.N., and Lesnik, E.A. (1993). Tritium labeling of antisense oligonucleotides was carried out by exchange with tritiated water. Nucleic Acids Res., 21: 3737-3743). The only two introduced differences to the protocol were that only 1mg was labeled per 25 oligonucleotide and that the separation of free tritium from the labeled oligonucleotide was done by 3x G10 30cm Sephadex columns (the columns were made using 10ml plastic pipettes). Radioactivity in all samples was counted after dissolving the samples in Ultima Gold (Packard) scintillation fluid, and using a scintillation counter. For the biodstribution studies, female nude mice (NMRI nu/nu, Charles River Netherlands, 30 Maastricht, The Netherlands) with 15PC3 and MiapacaII xenografts were used. See the in vivo experiment section for further details. Tissue distribution studies of tritiated oligonucleotides were performed according to Bijsterbosch et al. (Bijsterbosch, M.K., Manoharan, M., Rump, E.T., De Vrueh, R.L., van Veghel, R., Tivel, K.L., Biessen, E.A., Bennett, C.F., Cook, P.D., and van Berkel T.J. (1997) 35 In vivo fate of phosphorothioate antisense oligodeoxynucleotides: predominant uptake by scavenger receptors on endothelial liver cells. Nucleic Acids Res., 25: 3290-3296).
27 The radioactivity in the different organs was corrected for serum present at the time of sampling as determined by the distribution of 25 1-BSA (personal communication K. Kruijt, University of Leiden, the Netherlands). The oligonucleotides were either administrated by bolus injection in the lower vena cava 5 (circulation for 30 minutes) or using Alzet osmotic minipumps (see in vivo experiment section), for a prolonged systemic circulation. Tissue samples were dissolved in 5 M NaOH at 65 0 C and subsequently mixed with 10 volumes of Ultima Gold scintillation fluid. Serum and urine can be counted by mixing directly with Ultima gold. In vivo experiment 10 The animal experiments were approved by the ethical committee and are registered under No. DNL19. The detailed protocols of the animal studies are described in two publications: Tumor genotype-specific growth inhibition in vivo by antisense oligonucleotides against a polymorphic site of the large subunit of human RNA polymerase II. Fluiter K, ten Asbroek AL, van Groenigen M, Nooij M, Aalders MC, Baas F. Cancer Res 2002 Apr 1;62(7):2024 15 2028 In vivo tumor growth inhibition and biodistribution studies of locked nucleic acid (LNA) antisense oligonucleotides. Fluiter K, ten Asbroek AL, de Wissel MB, Jakobs ME, Wissenbach M, Olsson H, Olsen 0, Oerum H, Baas F. Nucleic Acids Res 2003 Feb 1;31(3):953-962 20 Mice: Female NMRI nu/nu (Charles River Netherlands, Maastricht, The Netherlands). Xenografts: MiaPaca II injected in the right flank s.c. with Matrigel (collaborative biomedical products Bedford, MA); 15PC3 injected in the left flank s.c. with Matrigel. Osmotic pumps: Alzet 1002 (DURECT Corporation, Cupertino, CA) lot no.10045-02 Dosage for 2776, 2778 (alpha-L-oxy-LNA), 2742 and 2744 (beta-D-oxy-LNA): 1 and 2.5 25 mg/kg/day. Control: physiological saline. Temperature and animal ID was monitored using: ELAM chips (IPTT 200) using a DAS 5002 chip reader (BMDS, Seaford, Delaware). Serum samples were taken for ASAT/ALAT and Alkaline Phosphatase determination. Aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT) levels and 30 alkaline phosphatase in serum were determined using standard diagnostic procedures with the H747 (Hitachi/Roche) with the appropriate kits (Roche Diagnostics). The ALAT/ASAT and Alkaline phosphatase Levels were determined approx 20 hours post extraction of serum from the animal.
28 Results Beta-D-Amino-LNA Nuclease stability One of the major difficulties encountered using the naturally occurring phosphodiester 5 oligonucleotides as antisense probes is their rapid degradation by various nucleolytic activities in cells, serum, tissues or culture medium. Since the phosphorus center is the site of nucleolytic attack, many modifications have been introduced in the internucleoside linkage to prevent enzymatic degradation. To date, the most commonly employed synthetic modification is the backbone phosphorothioate analogue, made by replacing one 10 of the non-bridging oxygen atoms of the internucleoside linkage by sulfur. We wanted to evaluate the effect of introducing the novel LNA within an oligonucleotide in the presence of nucleases, and to compare it with the well-studied phosphorothioate oligonucleotides. The study was carried out with oligothymidylates by blocking the 3'-end with the novel LNA relatives. The oligonucleotide is synthesized on deoxynucleoside 15 support (t). From figure 1, we can appreciate the stability properties, which confer beta-D-amino-LNA. Oligonucleotides containing T-monomer of 2'-beta-D-amino-LNA (TN) present a remarkable stability against a 3'-exonuclease. Blocking the 3'-end with just two TN stops the enzyme from degrading the oligonucleotide at least for 2 h. See figure 1. 20 Assisted cellular uptake and subcellular distribution The uptake efficiency of FAM-labeled oligonucleotide containing beta-D-amino-LNA was measured as the mean fluorescence intensity of the transfected cells by FACS analysis. Two different transfection agents were tested (Lipofectamine 2000 and DAC30) in two 25 different cell lines (MiaPacaII and 15PC3). DAC30 Lipofectamine 2000 Ref oligonucleotides % % % % cells uptake cells uptake 2753 TN CN CNgstscsastscsgscstsC N N Nc-FAM - - 100 100 2752 TNs CNs CNsgstscsastscsgscstsC NsC NsT Nsc-FAM 30 30 100 100 2740 TsCsCsgstscsastscsgscstsCsCsTsc-FAM 80 30 100 100 Table 1. Oligonucleotides containing beta-D-amino-LNA used in cellular uptake and subcellular distribution experiments. Residue c is methyl-c both for DNA and LNA.
29 Oligonucleotides both fully thiolated (PS, 2752) and partially thiolated (PO in the flanks and PS in the gap, 2753) containing beta-D-amino-LNA listed in table 1 were transfected with good efficiency, see table 1. Both transfection agents, DAC30 and Lipofectamine, presented good transfection efficiency; however, Lipofectamine was superior. 5 Lipofectamine showed 100% efficiency in all cases: for both oligonucleotides (2753 and 2752) and in both cell lines. Moreover, no significant differences in assisted transfection efficiency were observed between 2752 and 2753. The FAM-labeled oligonucleotide 2752 was also used to assay the subcellular distribution of oligonucleotides containing beta-D-amino-LNA, see figure 2. Most of the staining was 10 detected as nuclear fluorescence that appeared as bright spherical structures (the nucleoli is also stained) in a diffuse nucleoplasmic background, as well as some cytoplasmic staining in bright punctate structures. The observed distribution patterns were similar for 15PC3 and MiaPacaII. The subcellular distribution of beta-D-amino-LNA was comparable to the one observed with 15 beta-D-oxy-LNA, 2740. The uptake efficiency was also measured with tritium-labeled oligonucleotide 2754 (see table 3 and figure 3) at different concentrations 100, 200, 300 and 400 nM, using Lipofectamine2000 as transfection agent, both in MiaPacaII and 15PC3 cells, and compared with the equivalent beta-D-oxy-LNA, 2742 (see table 3). 2754 shows lower 20 uptake than 2742. Antisense activity assay: Luciferase target It has been shown that beta-D-oxy-LNA does not elicit RNaseH activity, which is the most common mode of action for an antisense oligonucleotide targeting the down-stream region 25 of the mRNA. However, this disadvantage can be overcome by creating chimeric oligonucleotides composed of beta-D-oxy-LNA and a DNA gap positioned in the middle of the sequence. A gapmer is based on a central stretch of 4-12 DNA (gap) typically flanked by 1 to 6 residues of 2'-0 modified nucleotides (beta-D-oxy-LNA in our case, flanks). It was of our interest to evaluate the antisense activity of oligonucleotides, which contain 30 beta-D-amino-LNA in a gapmer design, and compare them with beta-D-oxy-LNA/DNA gapmers. The oligonucleotides from table 2 were prepared. We decided to carry out the study with gapmers of 16nt in length and a gap of 7nt, which contain 4 residues of beta-D-amino-LNA in one flank and 4 residues of beta-D-oxy-LNA in the other flank, and a thiolated gap. The 35 FAM group was shown not to affect the antisense ability of the oligonucleotides. Therefore, we prepared a FAM-labelled oligonucleotide to be both tested in the Luciferase assay, and in the Cellular uptake (unassisted).
30 The oligonucleotide, which targets a motif of the mRNA of the Firefly Luciferase, contains two mismatches in the flanks. Two C residues of the 5'-end LNA flank were substituted for two Ts for synthetic reasons. At that point in time, only the T residues were available. Therefore and in order to be able to establish a correct comparison, the corresponding 5 beta-D-oxy-LNA control was also included in the assay. No FAM labeling was necessary in this case. ref sequence design size U-14 FAM-T'TTT ''' astscsgTCTTT Amino-LNA in one flank/ PS gap of 7 16mer 2023-m; TTTTgstscsastscgTCTTT Control with oxy-LNA 16mer 02579 Table 2 Oligonucleotide containing beta-D-amino-LNA used in the antisense activity assay and the oxy-LNA control (Capital letters for LNA and small letters for DNA, TN is beta-D-amino-LNA). Residue c is methyl-c both for LNA. From figure 4, we can see that the oligonucleotide with beta-D-amino-LNA presents good antisense activity at 50 nM oligonucleotide concentration. The inclusion of beta-D-amino 10 LNA in the flanks of an oligonucleotide results in good down-regulation. We can conclude that the antisense activity of an oligonucleotide containing beta-D-amino-LNA is at least as good as the parent all beta-D-oxy-LNA gapmer. Antisense activity assay: Ha-Ras target 15 It was of our interest to further evaluate the antisense activity of oligonucleotides containing beta-D-amino-LNA in a gapmer design, and compare them with beta-D-oxy-LNA gapmers. The oligonucleotides from table 3 were prepared. We decided to carry out the study with oligonucleotides of 16nt in length and a gap of 8nt, which contain 3 residues of beta-D 20 amino-LNA in each flank and a different extent of thiolation. 2754 is fully thiolated (PS), while 2755 is only thiolated in the gap (PO in the flanks and PS in the gap). The oligonucleotides were designed to target a motif of the mRNA of Ha-Ras. Different mismatch controls were also included, 2756 is fully thiolated and 2757 presents thiolation only in the gap, see table 3. Moreover, the corresponding beta-D-oxy-LNA gapmers (see 25 table 3, 2742 is all PS, 2744 is the corresponding mismatch control; 2743 has PS in the gap, 2745 is the corresponding mismatch control) were also tested.
31 ref oligonucleotides 2755 T N CN CNs gscsastscsgscsts N CN TN c PO/PS 25 TNs Ns Ns ~ sNa ~ N Ns AII PS 2755 TNSCNSC sgstscsastscsgscstsC C T c 2743 TCCgstscsastscsgscstsCCTc PO/PS 2742 TsCsCsgstscsastscsgscstsCsC Tsc All PS 2757 TN CN TNgstsasastsasgscscsC N CNCN c Mismatch control 2756 TNs CNsTNsgstsasastsasgscscsC Ns Ns Ns 2745 TCTgstsasastsasgscscsCCCc Mismatch control 2744 TsCsTsgstsasastsasgscscsCsCsCsc Mismatch control Table 3. Oligonucleotides containing beta-D-amino-LNA and beta-D-oxy-LNA used in the antisense activity experiments. Residue c is methyl-c both for DNA and LNA. 5 The Ras family of mammalian proto-oncogenes includes three well-known isoforms termed Ha-Ras (Ha-Ras), Ki-Ras (K-Ras) and N-Ras. The ras proto-oncogenes encode a group of plasma membrane associated G-proteins that bind guanine nucleotides with high affinity and activates several effectors including raf-1, P13-K etc. that are known to activate 10 several distinct signaling cascades involved in the regulation of cellular survival, proliferation and differentiation. Several in vitro (and in vivo) studies have demonstrated that the Ras family of proto oncogenes are involved in the induction of malignant transformation. Consequently, the Ras family is regarded as important targets in development of anticancer drugs, and it has 15 been found that the Ras proteins are either over-expressed or mutated (often leading to constitutive active Ras proteins) in approximately 25 % of all human cancers. Interestingly, the ras gene mutations in most cancer types are frequently limited to only one of the ras genes and are dependent on tumor type and tissue. Mutations in the Ha-Ras gene are mainly restricted to urinary tract and bladder cancer. 20 The inclusion of beta-D-amino-LNA in the flanks of an oligonucleotide results in good down regulation levels. From figure 5, we can see that oligonucleotides with beta-D-amino-LNA present good antisense activity at two different concentrations, 400 and 800 nM. No significant difference in down-regulation can be seen between oligonucleotides 2755 and 2754, which present a different degree in thiolation. We can conclude that the antisense 25 activity of an oligonucleotide containing beta-D-amino-LNA is at least as good as the parent beta-D-oxy-LNA gapmer. From figure 6, a wider range of concentration was tested. There is a potent down-regulation between 50-400 nM for 2754. The specificity was also tested; at 30 nM there is a significant difference in down-regulation between the mismatch 2756 (less potent) and the match 2754. Lower concentrations (5-40 nM) were also included 32 from the table in figure 6. Potent down-regulation is observed even at 5 nM for 2754, and these levels of down-regulation are comparable to the corresponding beta-D-LNA control, 2742. The specificity is also remarkable, if we compare the antisense activity for 2754 at 20 nM (8,7% down-regulation) in comparison with the mismatch containing control 2756 5 (56,2% down-regulation). Biodistribution The biodistribution of oligonucleotides containing beta-D-amino-LNA (tritiated 2754) was 10 also studied, both after i.v. injection and using Alzet osmotic minipumps. 2754 was administered to xenografted mice with 15PC3 tumors on the left side and MiaPacaII tumors on the right side as an intraveneous injection, and the analysis was carried out after 30 min circulation. From figure 7, the serum clearance for 2754 is very rapid, and the biodistribution looks very similar to the biodistribution pattern presented by 15 the reference containing beta-D-oxy-LNA; the kidney and the liver (to lesser extent) are the main sites of uptake, when corrected for tissue weight. Moreover, a group of 4 nude mice xenografted with 15PC3 tumors on the left side and MiaPacaII tumors on the right side were treated for 72 h with Alzet osmotic minipumps with a 2.5 mg/Kg/day dosage of tritiated 2754. After the treatment, the radioactivity 20 present in the different tissues was measured. Figure 8 shows the distribution of 2754 in the tissues as a total uptake and as a specific uptake. It seems that the tissue takes up significantly better amino-LNA than beta-D-oxy LNA. The main sites of uptake were liver, muscle, kidney, skin, bone and heart. When corrected for tissue weight, kidney, heart and liver (lungs and muscle in a lower extent) were the main uptake sites. This 25 pattern differs to a certain extent from the one observed for beta-D-oxy-LNA. It is also noteworthy that the uptake of amino-LNA is significantly better in tumor tissue than for e.g. beta-D-oxy LNA (see Figure 7 and 8). RNase H assay 30 Rnase H is a ubiquitous cellular enzyme that specifically degrades the RNA strand of DNA/RNA hybrids, and thereby inactivates the mRNA toward further cellular metabolic processes. The inhibitory potency of some antisense agents seems to correlate with their ability to elicit ribonuclease H (RNaseH) degradation of the RNA target, which is considered a potent mode of action of antisense oligonucleotides. As such, understanding the 35 mechanisms of catalytic function and substrate recognition for the RNaseH is critical in the design of potential antisense molecules. It was our aim to evaluate the RNaseH activity of gapmers containing beta-D-amino-LNA.
33 From figure 9, we can appreciate a good cleavage activity for an oligonucleotide containing beta-D-amino-LNA, as in table 2. Beta-D-Thio-LNA 5 Nuclease stability As we did for beta-D-amino-LNA, beta-D-thio-LNA was also evaluated against a 3' exonuclease (SVPD). The oligonucleotide is synthesized on deoxynucleoside-support (t). The study was carried out with oligothymidylates by blocking the 3'-end with beta-D-thio LNA. 10 From figure 10, we can see that the incorporation of just one T-monomer of 2'-beta-D thio-LNA (TS) has a significant effect in the nucleolytic resistance of the oligonucleotide towards SVPD. After 2h digestion more than 80% of the oligonucleotide remains, while the corresponding beta-D-oxy-LNA oligonucleotide is digested by the exonuclease, see figure 10. 15 Unassisted cellular uptake The efficiency of FAM-labelled oligonucleotide uptake was measured as the mean fluorescence intensity of the transfected cells by FACS analysis. The transfection without lipid showed distinct differences between the tested oligonucleotides. The uptake as measured from mean fluorescence intensity of transfected 20 cells was dose dependent. Gapmers (16nt in length and gap of 7nt) containing beta-D-thio-LNA in the flanks were analysed and compared with the corresponding beta-D-oxy-LNA gapmers. Beta-D-thio-LNA (one flank with beta-D-thio-LNA and the other one with oxy-LNA, as in table 5) showed higher uptake than oligonucleotides containing only oxy-LNA. The beta-D-thio-LNA 25 oligonucleotides (both all-PO gapmer and gapmer with PS-gap and PO-flanks) had good uptake efficiency. Specially, the all-PO gapmer containing beta-D-thio-LNA was far superior to other all-PO oligonucleotides tested so far, as it can be appreciated from figure 11. Assisted cellular uptake and subcellular distribution The uptake efficiency of FAM-labeled oligonucleotide containing beta-D-thio-LNA was 30 measured as the mean fluorescence intensity of the transfected cells by FACS analysis. Two different transfection agents were tested (Lipofectamine 2000 and DAC30) in two different cell lines (MiaPacaII and 15PC3).
34 DAC30 Lipofectamine 2000 ref oligonucleotides % % % % cells uptake cells uptake 2747 T C C gstscsastscsgscstsC C T c-FAM - - 100 100 2746 T SSC SSC sgstscs astscsg scstsC SSC SST sSc-FAM 80 50 100 100 2740 TsCsCsgstscsastscsgscstsCsCsTsc- FAM 80 30 100 100 Table 4. Oligonucleotides containing beta-D-thio-LNA used in cellular uptake and subcellular distribution experiments. Residue c is methyl-c both for DNA and LNA. 5 Oligonucleotides both fully thiolated (PS, 2746) and partially thiolated (PO in the flanks and PS in the gap, 2747) containing beta-D-thio-LNA listed in table 4 were transfected with good efficiency, see table 4. Both transfection agents, DAC30 and Lipofectamine, presented good transfection efficiency; however, Lipofectamine was superior. Lipofectamine showed 100% efficiency in all cases: for both oligonucleotides (2746 and 10 2747) and in both cell lines. Moreover, no significant differences in assisted transfection efficiency were observed between 2746 and 2747. The FAM-labeled oligonucleotide 2746 was also used to assay the subcellular distribution of oligonucleotides containing beta-D-thio-LNA, see figure 2. Most of the staining was detected as nuclear fluorescence that appeared as bright spherical structures (the nucleoli 15 is also stained) in a diffuse nucleoplasmic background, as well as some cytoplasmic staining in bright punctate structures. The observed distribution patterns were similar for 15PC3 and MiaPacaII. The subcellular distribution of beta-D-thio-LNA was comparable to the one observed with beta-D-oxy-LNA, 2740. 20 The uptake efficiency was also measured with tritium-labeled oligonucleotide 2748 (see table 6 and figure 3) at different concentrations 100, 200, 300 and 400 nM, using Lipofectamine2000 as transfection agent, both in MiaPacaII and 15PC3 cells, and compared with the equivalent beta-D-oxy-LNA, 2742 (see table 6). 2748 shows superior uptake than 2742. 25 Antisense activity assay: Luciferase target We also introduced beta-D-thio-LNA in a gapmer design, and evaluated it in terms of antisense activity. The oligonucleotides from table 5 were prepared. We decided to carry out the study with 30 gapmers of 16nt in length and a gap of 7nt, which contain 4 residues of beta-D-thio-LNA in one flank and 4 residues of oxy-LNA in the other flank, and a thiolated gap.
35 The FAM group was shown not to affect the antisense ability of the oligonucleotides. Therefore, we prepared a FAM-labelled oligonucleotide to be both tested in the Luciferase assay, and in the Cellular uptake (unassisted). The oligonucleotide, which is directed against a motif of the mRNA of the firefly luciferase, 5 contains two mismatches in the flanks. Two C residues of the 5'-end LNA flank were substituted for two Ts for synthetic reasons. At that point in time, only the T residues were available. Therefore and in order to be able to establish a correct comparison, the corresponding oxy-LNA control was also included in the assay. No FAM labeling was necessary in this case. 10 ref sequence design size U-16 TsTsTsTsgstscsastscsgTCTTT-FAM Thio-LNA in one flank/ PS gap of 7 16mer 2023-m; TTTTgstscsastscsgTCTTT Control with oxy-LNA 16mer 02579 Table 5 Oligonucleotide containing beta-D-thio-LNA used in the antisense activity assay and the corresponding oxy-LNA control (Capital letters for LNA and small letters for DNA, Ts is beta-D-thio-LNA). Residue c is methyl-c both for LNA. 15 From figure 4, it can be seen that the oligonucleotide with beta-D-thio-LNA presents good antisense activity at 50 nM oligonucleotide concentration. Therefore, the inclusion of beta D-thio-LNA in the flanks of an oligonucleotide results in good down-regulation, and is at least as good as the parent all beta-D-oxy-LNA gapmer. Antisense activity assay: Ha-Ras target 20 It was of our interest to further evaluate the antisense activity of oligonucleotides containing beta-D-thio-LNA in a gapmer design, and compare them with beta-D-oxy-LNA gapmers. The oligonucleotides from table 6 were prepared. We decided to carry out the study with oligonucleotides of 16nt in length and a gap of 8nt, which contain 3 residues of beta-D 25 thio-LNA in each flank and a different extent of thiolation. 2748 is fully thiolated (PS), while 2749 is only thiolated in the gap (PO in the flanks and PS in the gap). The oligonucleotides were designed to target a motif of the mRNA of Ha-Ras. Different mismatch controls were also included, 2750 is fully thiolated and 2751 presents thiolation only in the gap, see table 6. Moreover, the corresponding beta-D-oxy-LNA gapmers (see 30 table 6, 2742 is all PS, 2744 is the corresponding mismatch control; 2743 has PS in the gap, 2745 is the corresponding mismatch control) were also tested.
36 ref oligonucleotides 2749 T C C gstscsastscsgscstsC C T c PO/PS 274 S S S STc All PS 2748 T S C S C sgstscsastscsgscstsC S C ST s cAIP 2743 TCCgstscsastscsgscstsCCTc PO/PS 2742 TsCsCsgstscsastscsgscstsCsCsTsc All PS 2751 T C T gstsasastsasgscscsC C C c Mismatch control 275 S Sc c Sc Mismatch control 2750 T S C J TsgstsasastsasgscscsC S C S C s c Msacoto 2745 TCTgstsasastsasgscscsCCCc Mismatch control 2744 TsCsTsgstsasastsasgscscsCsCsCsc Mismatch control Table 6. Oligonucleotides containing beta-D-thio-LNA and beta-D-oxy-LNA used in the antisense activity experiments. Residue c is methyl-c both for DNA and LNA. 5 The inclusion of beta-D-thio-LNA in the flanks of an oligonucleotide results in good down regulation levels. From figure 5, we can see that oligonucleotides with beta-D-thio-LNA present good antisense activity at two different concentrations, 400 and 800 nM. No significant difference in down-regulation can be seen between oligonucleotides 2749 and 2748, which present a different degree in thiolation. However, 2749 presents better levels 10 of down-regulation, both at 400 and 800 nM. We can conclude that the antisense activity of an oligonucleotide containing beta-D-thio-LNA lies in the range of the parent beta-D-oxy LNA gapmer. From figure 6, a wider range of concentration was tested. There is a potent down-regulation between 50-400 nM for 2748. The specificity was also tested; at 30 nM there is a significant difference in down-regulation between the mismatch 2750 (less 15 potent) and the match 2748. Biodistribution The biodistribution of oligonucleotides containing beta-D-thio-LNA (tritiated 2748) was also studied, both after i.v. injection and using Alzet osmotic minipumps. 20 2748 was administered to xenografted mice with 15PC3 tumors on the left side and MiaPacaII tumors on the right side as an intraveneous injection, and the analysis was carried out after 30 min circulation. From figure 7, the serum clearance for 2748 is very rapid, and the biodistribution looks very similar to the biodistribution pattern presented by the reference containing beta-D-oxy-LNA; the kidney and the liver (to lesser extent) are 25 the main sites of uptake, when corrected for tissue weight. Moreover, a group of 4 nude mice xenografted with 15PC3 tumors on the left side and MiaPacaII tumors on the right side were treated for 72 h with Alzet osmotic minipumps with a 2.5 mg/Kg/day dosage. After the treatment, the radioactivity present in the 37 different tissues was measured. Figure 8 shows the distribution of 2748 in the tissues as a total uptake and as a specific uptake. The main sites of uptake were liver, muscle, kidney, skin and bone. When corrected for tissue weight, kidney and liver were the main uptake sites. 5 RNaseH assay We also evaluated gapmer designs that contain beta-D-thio-LNA, as in table 5, for their ability to recruit RNaseH activity. From figure 9, we can see that a beta-D-thio-LNA gapmer recruits RnaseH activity. 10 Alpha-L-oxy LNA Nuclease stability The stabilization properties of alpha-L-oxy-LNA were also evaluated. The study was carried out with oligothymidylates by blocking the 3'-end with alpha-L-oxy-LNA. The oligonucleotide is synthesized on deoxynucleoside-support (t). From figure 12, we can see 15 that the introduction of just one alpha-L-T (T ) at the 3'-end of the oligonucleotide represents already a gain of 40% stability (after 2h digestion) with respect to the oxy version, for which there was actually no gain. The addition of two modifications contributes even more to the stability of the oligonucleotide. 20 Furthermore, we investigated the effect on stability against S1-endonuclease of alpha-L oxy-LNA for a 16mer fully modified oligothymidylates. The increased stability of these modified oligonucleotides relative to their deoxynucleotide and phosphorothioate backbone relatives was compared in order to carefully assess the contribution of the alpha-L-oxy LNA modification. 25 After 2 h digestion, most of the alpha-L-oxy-LNA oligonucleotide remained (over 80% of the full-length product remained), while neither the oligodeoxynucleotide nor the DNA phosphorothioate analogue could be detected after 30 min digestion (see figure 13). The same kinetic study against S1-endonuclease was carried out with a fully modified oxy-LNA oligonucleotide, which was also very resistant against the S1-endonuclease. Over an 8 5 % 30 of the full-length product remained after 2 h digestion (see figure 13). In conclusion, beta-D-oxy-LNA, beta-D-amino-LNA, beta-D-thio-LNA and alpha-L-oxy-LNA stabilize oligonucleotides against nucleases. An order of efficiency in stabilization can be established: DNA phosphorothioates << oxy-LNA < ax-L-oxy-LNA < beta-D-amino-LNA < 35 beta-D-thio-LNA.
38 Unassisted cellular uptake The efficiency of FAM-labelled oligonucleotide uptake was measured as the mean fluorescence intensity of the transfected cells by FACS analysis. The uptake as measured from mean fluorescence intensity of transfected cells was dose dependent. Gapmers (16nt 5 in length and gap of 7nt) containing ax-L-oxy-LNA in the flanks were analysed and compared with the corresponding beta-D-oxy-LNA gapmer. a,-L-oxy-LNA (in both flanks) showed higher uptake than the oligonucleotide containing only beta-D-oxy-LNA. Both all PO and gapmer with PS-gap had good uptake efficiency; especially the all-PO gapmer was far superior than other all PO oligonucleotides tested so far, see figure 14 for FACS 10 analysis. Assisted cellular uptake and subcellular distribution The uptake efficiency of FAM-labeled oligonucleotides containing alpha-L-oxy-LNA was measured as the mean fluorescence intensity of the transfected cells by FACS analysis. Two different transfection agents were tested (Lipofectamine 2000 and DAC30) in two 15 different cancer cell lines (MiaPacaII and 15PC3). DAC30 Lipofectamine 2000 ref oligonucleotides % % % % cells uptake cells uptake 2773 TaCaCagstscsastscsgscstsCaCaTaC-FAM - - 100 100 2774 TasCasCasgstscsastscsgscstsCasCasTasc-FAM 80 30 100 100 2740 TsCsCsgstscsastscsgscstsCsCsTsc-FAM 80 30 100 100 Table 7. Oligonucleotides containing alpha-L-oxy-LNA used in cellular uptake and subcellular distribution experiments. Residue c is methyl-c both for DNA and LNA. 20 Oligonucleotides both fully thiolated (PS, 2774) and partially thiolated (PO in the flanks and PS in the gap, 2773) containing alpha-L-oxy-LNA listed in table 7 were transfected with good efficiency, see table 7. Both transfection agents, DAC30 and Lipofectamine, presented good transfection efficiency; however, Lipofectamine was superior. Lipofectamine showed 100% efficiency in all cases: for both oligonucleotides (2773 and 25 2774) and in both cell lines. Moreover, no significant differences in assisted transfection efficiency were observed between 2773 and 2774. The FAM-labeled oligonucleotide 2774 was also used to assay the subcellular distribution of oligonucleotides containing alpha-L-oxy-LNA, see figure 2. Most of the staining was detected as nuclear fluorescence that appeared as bright spherical structures (the nucleoli 30 is also stained) in a diffuse nucleoplasmic background, as well as some cytoplasmic 39 staining in bright punctate structures. The observed distribution patterns were similar for 15PC3 and MiaPacaII. The subcellular distribution of alpha-L-oxy-LNA was comparable to the one observed with beta-D-oxy-LNA, 2740. 5 Antisense activity: Luciferase target Gapmers containing alpha-L-oxy-LNA We also wanted to see the antisense acticvity in a gapmer oligonucleotides containing alpha-L-oxy-LNA (16nt in length with a thiolated 7nt gap). Two different designs were 10 evaluated. First, we substituted two oxy-LNA residues for two alpha-L-oxy-LNAs in a gapmer against a motif of the mRNA of the firefly luciferase, and placed the alpha-L-oxy-LNA in the junctions, see figure 15. Then, we substituted both flanks with alpha-L-oxy-LNA in the same construct, see figure 15 15. Previously, different oligonucleotides were tested and compared with the corresponding FAM-labelled molecules, and no significant difference was appreciated between the free and FAM-labelled ones. Therefore, we included oligonucleotides from the Unassisted Cellular Uptake assay in the Luciferase assay study, assuming that the antisense activity 20 will not be affected by the presence of the FAM group. From figure 16, the oligonucleotide with alpha-L-oxy-LNA in the junctions shows potent antisense activity. It is actually 5-fold better than the corresponding all oxy-LNA gapmer (gap of 7nt), and slightly better than a gapmer with an optimised 9nt gap with oxy-LNA. The second design (all alpha-L-oxy-LNAs in both flanks) presents at least as good down 25 regulation levels as the observed for beta-D-oxy-LNA gapmers. We can also conclude that the presence of the alpha-L-oxy-LNA in a gapmer construct shows good-antisense activity level. alpha-L-oxy-LNA reveals to be a potent tool enabling the construction of different gapmers, which show good antisense activity. The placement of alpha-L-oxy-LNA in the 30 junctions results in a very potent oligonucleotide. Short-sized gapmers containing alpha-L-oxy-LNA As a general rule, the length of the construct is usually designed to range from 15-25 nucleotide units, in order to ensure that optimal identification and binding takes place with a unique sequence in the mammalian genome and not with similar genetically redundant 35 elements. Statistical analyses specify 11-15 base paired human sequences as the theoretical lower limits for sufficient recognition of a single genomic region. In practice, however, a longer oligonucleotide is commonly used to compensate for low melting transitions, especially for thiolated oligonucleotides that have lower affinity.
40 As a significant increase in affinity is achieved by the introduction of oxy-LNA or novel LNA relatives, the design of potent and short antisense oligonucleotides (<15nt) should be enabled. The alpha-L-oxy-LNA can play an important role in enabling the design of short molecules 5 by maintaining the required high-affinity, but also an optimal gap size. 12 and 14mers against a motif of the mRNA of the firefly luciferase were evaluated. The results are shown in figure 16. The presence of alpha-L-oxy-LNA in the flanks of a 12 (gap of 7nt) and 14 mer (gap of 8nt) correspond to good levels of down-regulation. From figure 16. 10 In conclusion, alpha-L-oxy-LNA is a potent tool in enabling the design of short antisense oligonucleotides with significant down-regulation levels. Mixmers containing alpha-L-oxy-LNA We also considered other designs containing alpha-L-oxy-LNA against a motif of the mRNA of the firefly luciferase, which we called mixmers. They consist of an alternate composition 15 of DNA, alpha-L-oxy-LNA and beta-D-oxy-LNA. The following figure illustrates the chosen designs. We named the mixmers by the alternate number of units of each alpha-L-oxy LNA, beta-D-oxy-LNA or DNA composition. See figure 17 and table 8 for the different designs. ref sequence mixer 2023-q TTCCgTascsastscegTascTTT 4-1-1-5-1-1-3 a 2023-r TaTaCaCagTascsastscegTascTaTaT 4-1-1-5-1-1-3 b 2023-t TTCCgstscAastscegTCTTT 4-3-1-3-5 a 2023-u TTCCagstscAastscegTaCTTT 4-3-1-3-5 b Table 8 Mixmers containing alpha-L-oxy-LNA used in this study (Capital letters for LNA and small letters for DNA, T" is alpha-L-oxy-LNA). Residue c is methyl-c both for LNA. 20 In design 4-1-1-5-1-1-3 (figure 17, table 8), we placed two alpha-L-oxy-LNA residues interrupting the gap, being the flanks beta-D-oxy-LNA. Furthermore, we interrupted the gap with two alpha-L-oxy-LNA residues, and substituted both flanks with alpha-L-oxy-LNA. The presence of alpha-L-oxy-LNA might introduce a flexible transition between the North 25 locked flanks (oxy-LNA) and the alpha-L-oxy-LNA residue by spiking in deoxynucleotide residues. It is also interesting to study design 4-3-1-3-5 (figure 17, table 8), where an alpha-L-oxy LNA residue interrupts the DNA stretch. In addition to the alpha-L-oxy-LNA in the gap, we also substituted two oxy-LNA residues at the edges of the flanks with two alpha-L-oxy-LNA 30 residues.
41 The presence of just one beta-D-oxy-LNA residue (design 4-3-1-3-5) interrupting the stretch of DNAs in the gap results in a dramatic loss of down-regulation. Just by using alpha-L-oxy-LNA instead, the design shows significant down-regulation at 50nM oligonucleotide concentration, see figure 16. The placement of alpha-L-oxy-LNA in the 5 junctions and one alpha-L-oxy-LNA in the middle of the gap also shows down-regulation, see figure 16. The interruption of the gap with two beta-D-oxy-LNAs (design 4-1-1-5-1-1-3) relates also with a loss in antisense activity. Again the fully substitution of beta-D-oxy-LNA for alpha-L oxy-LNA gives significant antisense activity, see figure 916. 10 alpha-L-oxy-LNA reveals to be a potent tool enabling the construction of different mixmers, which are able to present high levels of antisense activity. Other designs Other mixmers containing alpha-L-oxy-LNA were studied, see figure 18. Furthermore, 15 mixmers, such as in table 8 and figure 17, but with no thiolation, were also tested. Antisense activity assay: Ha-Ras target It was of our interest to further evaluate the antisense activity of oligonucleotides containing alpha-L-oxy-LNA in a gapmer design, and compare them with beta-D-oxy-LNA 20 gapmers. The oligonucleotides from table 9 were prepared. We decided to carry out the study with oligonucleotides of 16nt in length and a gap of 8nt, which contain 3 residues of alpha-L oxy-LNA in each flank and a different extent of thiolation. 2776 is fully thiolated (PS), while 2775 is only thiolated in the gap (PO in the flanks and PS in the gap). The oligonucleotides 25 were designed to target a motif of the mRNA of Ha-Ras. Different mismatch controls were also included, 2778 is fully thiolated and 2777 presents thiolation only in the gap, see table 9. Moreover, the corresponding beta-D-oxy-LNA gapmers (see table 9, 2742 is all PS, 2744 is the corresponding mismatch control; 2743 has PS in the gap, 2745 is the corresponding mismatch control) were also tested.
42 ref oligonucleotides 2775 TaCaCagstscsastScSgscstsCaCaTac PO/PS 2776 TasCasCasgstscsastscsgscstsCasCasTasc All PS 2743 TCCgstscsastscsgscstsCCTc PO/PS 2742 TsCsCsgstscsastscsgscstsCsCsTsc All PS 2777 TaCaTagstsasastsasgscscsCaCaCaC Mismatch control 2778 TasCasTasgstsasastsasgscscsCasCasCasc Mismatch control 2745 TCTgstsasastsasgscscsCCCc Mismatch control 2744 TsCsTsgstsasastsasgscscsCsCsCsc Mismatch control Table 9. Oligonucleotides containing alpha-L-oxy-LNA and beta-D-oxy-LNA used in the antisense activity experiments. Residue c is methyl-c both for DNA and LNA. 5 The inclusion of alpha-L-oxy-LNA in the flanks of an oligonucleotide results in good down-regulation levels. From figure 6, we can see that the oligonucleotide 2776 with alpha-L-oxy-LNA present good antisense activity at a different range of concentrations, 50 nM-400 nM. No significant difference in down-regulation can be seen between 2776 and 2742. We can conclude that the antisense activity of an 10 oligonucleotide containing alpha-L-oxy-LNA is at least as good as the parent beta-D oxy-LNA gapmer. The specificity was also tested; at 30 nM there is a significant difference in down-regulation between the mismatch 2778 (less potent) and the match 2776. Lower concentrations (5-40 nM) were also included from the table in figure 6. Potent down-regulation is observed even at 5 nM for 2776 in comparison with the 15 corresponding beta-D-oxy-LNA control, 2742. The specificity is also remarkable, if we compare the antisense activity for 2776 at 20 nM (2,6% down-regulation) in comparison with the mismatch containing control 2778 (77% down-regulation). RNaseH assay We also evaluated gapmer designs that contain alpha-L-oxy-LNA for their ability to recruit 20 RNaseH activity. alpha-L-oxy-LNA gapmer and mixmer designs recruit RnaseH activity, see figure 19. In vivo experiment Nude mice were injected s.c. with MiaPaca II cells (right flank) and 15PC3 cells (left flank) one week prior to the start of oligonucleotide treatment to allow xenograft growth. The 25 anti Ha-Ras oligonucleotides (2742 and 2776, table 10) and control oligonucleotides (2744 and 2778, table 10) were administrated for 14 days using Alzet osmotic minipumps (model 1002) implanted dorsally. Two dosages were used: 1 and 2.5 mg/Kg/day. During treatment the tumor growth was monitored. Tumor growth was almost inhibited 43 completely at 2.5 mg/Kg/day and even at 1 mg/Kg/day dose with 2742 and 2776 in 15PC3 cells, figure 20. The specificity with control oligonucleotides (2744 and 2778, containing mismatches) increased as the dose decreased. At 1 mg/Kg/day dose the experiment presented a good specificity, particularly for alpha-L-oxy-LNA oligonucleotides (2742 and 5 2744). In MiaPacaII xenograft tumors, the effect of the oligonucleotides is in general comparable with those on the 15PC3 xenografts, except for the fact that the specificity seemed to be a bit lower. It can be concluded that the oligonucleotide containing alpha-L oxy-LNA are as potent, or maybe even better, as the one containing beta-D-oxy-LNA in tumor growth inhibition in the concentration range tested. ref oligonucleotides 2776 Ta Cc match 2778 TasCasTasgstsasastsasgscscsCasCasCasc Mismatch control 2742 TsCsCsgstscsastscsgscstsCsCsTsc match 2744 TsCsTsgstsasastsasgscscsCsCsCsc Mismatch control Table 10. Oligonucleotides containing alpha-L-oxy-LNA and beta-D-oxy-LNA used in the in vivo experiment. Residue c is methyl-c both for DNA and LNA. 15 Toxicity levels The levels of aspartate aminotransferase (ASAT), alanine aminotransferase (ALAT) and alkaline phosphatase in the serum were determined, in order to study the possible effects of this 14-day treatment in the nude mice. Serum samples were taken from each mouse after the 14-day experiment. From figure 21, ALAT levels in the serum varied between 20 250-500 U/L. ASAT levels were in the range of 80-150 U/L. The mice did not seem externally to be sick, and no big changes in behavior were observed. During treatment the body temperature of the mice was also monitored (figure 22). The body temperature did not change significantly during the treatment, not even at high dose 2,5 mg/Kg/day, which is an indication that no major toxicity effects are occurring. In some cases, the body 25 temperature of the mice was a bit higher, divided in two groups. These effects cannot be explained by the fact of one oligonucleotide behaving differently or one dosage being too high.
44 Specific beta-D-oxy-LNA constructs Luciferase target: Antisense activity assay Design 3-9-3-1 has a deoxynucleoside residue at the 3'-end, see table 11 and figure 23. 5 It shows significant levels of down-regulation, in the same range than an optimised (9nt) fully thiolated gapmer. Moreover, only partial thiolation is needed for these mixmers to work as good as the fully thiolated gapmer, see figure 24. ref sequence mixer 2023-1; 02574 TT CcgstscsastscsgstsCTTt 3-9-3-1 2023-k; 02575 TT CcgstscsastscsgstsCTTSt 3-9-3-1 2023-j; 02576 TsTsCscsgstscsastscsgstsCsTsTst 3-9-3-1 Table 11 Special beta-D-oxy-LNA constructs (Capital letters for LNA and small letters for DNA). Residue c is methyl-c for LNA. Other oligonucleotides containing novel LNA monomers (beta-D-amino-, beta-D-thio- and 10 alpha-L-LNA) and bearing a deoxynucleoside residue at the 3'-end were tested in different assays, see tables 3, 6, 9 and 10 for more detail. Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, 15 or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps. Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the 20 present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the filed relevant to the present invention as it existed before the priority date of each claim of this application.

Claims (21)

1. A locked nucleic acid (LNA) nucleoside having the formula R'0 Base 0 (CH2)n X4 0 R" or n(H2C)- X Base /0 R. R" or a pharmaceutically acceptable salt thereof, wherein: X is 0; R' and R" are independently either H or a terminal group; and (CH2)n is a methylene substituted with a C 1 6 alkyl.
2. The LNA nucleoside or pharmaceutically acceptable salt thereof according to claim 1, wherein (CH 2 )n is a methylene substituted with methyl.
3. The LNA nucleoside or pharmaceutically acceptable salt thereof according to claim 1 or claim 2, which has the formula R'0 Base 0 (2)n- -- X 0 R" wherein: X is 0; R' and R" are independently either H or a terminal group; and (CH2)n is a methylene substituted with methyl. 46
4. The LNA nucleoside or pharmaceutically acceptable salt thereof according to claim 1 or claim 2, which has the formula n(H2C)- X Base 0 R'V R" wherein: X is 0; R' and R" are independently either H or a terminal group; and (CH 2 )n is a methylene substituted with methyl.
5. A locked nucleic acid (LNA) antisense oligomer comprising at least one locked nucleotide of formula o, )Base (CHA) or Base wherein: X is 0; and (CH2)n is a methylene substituted with a C 1 6 alkyl.
6. The LNA antisense oligomer according to claim 5, wherein (CH2)n is a methylene substituted with a methyl group.
7. The LNA antisense oligomer according to claim 5 or claim 6, wherein at least one internucleoside linkage thereof is phosphorothioate.
8. The LNA antisense oligomer according to claim 5 or claim 6, wherein all internucleoside linkages thereof are phosphorothioate. 47
9. The LNA antisense oligomer according to any one of claims 5 to 8, wherein the oligomer is 10 to 18 nucleotides in length.
10. The LNA antisense oligomer according to any one of claims 5 to 8, wherein the oligomer is 10 to 15 nucleotides in length.
11. The LNA antisense oligomer according to any one of claims 5 to 10 comprising six, seven or eight consecutive DNA nucleotides flanked 5' by one or more locked nucleotides and 3' by one or more locked nucleotides.
12. The LNA antisense oligomer according to claim 11 comprising six, seven or eight consecutive DNA nucleotides flanked 5' by one or more locked nucleotides and 3' by one or more locked nucleotides, wherein the locked nucleotides at 5' and 3' ends have the formula: Base (CH 2 )j' or Base wherein: X is 0; and (CH 2 )n is a methylene substituted with a C 1 6 alkyl.
13. The LNA antisense oligomer according to claim 12, wherein (CH2)n is a methylene substituted with a methyl group in at least one of the locked nucleotides.
14. The LNA antisense oligomer according to any one of claims 5 to 8 having 8 to 12 DNA nucleotides flanked by one to four locked nucleotides.
15. The LNA antisense oligomer according to claim 10, wherein the oligomer consist of 12 to 14 nucleotide units. 48
16. The LNA antisense oligomer according to any one of claims 5 to 13 having a length of 12 to 14 nucleotide units and at least eight consecutive DNA nucleotides flanked 5' by one to three locked nucleotides and 3' by one to three locked nucleotides, wherein the locked nucleotides have the formula: Base (CH 2 2J wherein: X is 0; and (CH 2 )n is a methylene substituted with methyl group; and wherein all the internucleoside linkages thereof are phosphorothioate linkages.
17. The LNA antisense oligomer according to claim 16, wherein the number of consecutive DNA nucleotides is eight.
18. The LNA antisense oligomer according to claim 16, wherein the number of consecutive DNA nucleotides is nine.
19. The LNA antisense oligomer according to claim 16, wherein the number of consecutive DNA nucleotides is ten.
20. A pharmaceutical composition comprising the LNA antisense oligomer according to any one of claims 5 to 19 and a pharmaceutically acceptable carrier.
21. The LNA nucleoside according to any one of claims 1 to 4, or the LNA antisense oligomer according to any one of claims 5 to 19, or the pharmaceutical composition according to claim 20, substantially as hereinbefore described with reference to the accompanying drawings and/or examples.
AU2013201786A 2002-11-18 2013-03-21 Amino-LNA, thio-LNA and alpha-L-oxy-LN Expired AU2013201786B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013201786A AU2013201786B2 (en) 2002-11-18 2013-03-21 Amino-LNA, thio-LNA and alpha-L-oxy-LN

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
DKPA200201774 2002-11-18
DKPA200301540 2003-10-20
AU2011201821A AU2011201821B2 (en) 2002-11-18 2011-04-20 Amino-LNA, thio-LNA and alpha-L-oxy-LN
AU2013201786A AU2013201786B2 (en) 2002-11-18 2013-03-21 Amino-LNA, thio-LNA and alpha-L-oxy-LN

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2011201821A Division AU2011201821B2 (en) 2002-11-18 2011-04-20 Amino-LNA, thio-LNA and alpha-L-oxy-LN

Publications (2)

Publication Number Publication Date
AU2013201786A1 AU2013201786A1 (en) 2013-04-11
AU2013201786B2 true AU2013201786B2 (en) 2015-04-02

Family

ID=48083751

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2013201763A Expired AU2013201763B2 (en) 2002-11-18 2013-03-21 Amino-LNA, thio-LNA and alpha-L-oxy-LN
AU2013201786A Expired AU2013201786B2 (en) 2002-11-18 2013-03-21 Amino-LNA, thio-LNA and alpha-L-oxy-LN

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2013201763A Expired AU2013201763B2 (en) 2002-11-18 2013-03-21 Amino-LNA, thio-LNA and alpha-L-oxy-LN

Country Status (1)

Country Link
AU (2) AU2013201763B2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000056748A1 (en) * 1999-03-18 2000-09-28 Exiqon A/S Xylo-lna analogues
WO2000066604A2 (en) * 1999-05-04 2000-11-09 Exiqon A/S L-ribo-lna analogues
EP1152009B1 (en) * 1999-02-12 2005-01-26 Sankyo Company, Limited Novel nucleosides and oligonucleotide analogues

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU7406700A (en) * 1999-10-04 2001-05-10 Exiqon A/S Design of high affinity rnase h recruiting oligonucleotide
AU3041701A (en) * 1999-12-23 2001-07-09 Exiqon A/S Therapeutic uses of lna-modified oligonucleotides
EP1251183A3 (en) * 2001-04-18 2003-12-10 Exiqon A/S Improved helper probes for detection of a target sequence by a capture oligonucleotide

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1152009B1 (en) * 1999-02-12 2005-01-26 Sankyo Company, Limited Novel nucleosides and oligonucleotide analogues
WO2000056748A1 (en) * 1999-03-18 2000-09-28 Exiqon A/S Xylo-lna analogues
WO2000066604A2 (en) * 1999-05-04 2000-11-09 Exiqon A/S L-ribo-lna analogues

Also Published As

Publication number Publication date
AU2013201763B2 (en) 2015-05-07
AU2013201763A1 (en) 2013-04-11
AU2013201786A1 (en) 2013-04-11

Similar Documents

Publication Publication Date Title
US9951333B2 (en) Antisense design
AU2017232083B2 (en) Amino-LNA, thio-LNA and alpha-L-oxy-LN
AU2013201786B2 (en) Amino-LNA, thio-LNA and alpha-L-oxy-LN

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired