EP2616549A1 - Chemische modifikation von rna an der 2'-position des riboserings mittels aaa-kopplung - Google Patents

Chemische modifikation von rna an der 2'-position des riboserings mittels aaa-kopplung

Info

Publication number
EP2616549A1
EP2616549A1 EP11825711.2A EP11825711A EP2616549A1 EP 2616549 A1 EP2616549 A1 EP 2616549A1 EP 11825711 A EP11825711 A EP 11825711A EP 2616549 A1 EP2616549 A1 EP 2616549A1
Authority
EP
European Patent Office
Prior art keywords
rna
apob
coupling
reaction
chemical modification
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.)
Withdrawn
Application number
EP11825711.2A
Other languages
English (en)
French (fr)
Inventor
Daniel Zewge
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.)
Merck Sharp and Dohme LLC
Original Assignee
Merck Sharp and Dohme LLC
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
Application filed by Merck Sharp and Dohme LLC filed Critical Merck Sharp and Dohme LLC
Publication of EP2616549A1 publication Critical patent/EP2616549A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical

Definitions

  • RNA interference is an evolutionarily conserved cellular mechanism of post-transcriptional gene silencing found in fungi, plants and animals that uses small RNA molecules to inhibit gene expression in a sequence-specific manner.
  • the RNAi machinery can be harnessed to destruct any mRNA of a known sequence. This allows for suppression (knock-down) of any gene from which it was generated and consequently preventing the synthesis of the target protein.
  • Smaller siR A duplexes introduced exogenously were found to be equally effective triggers of RNAi (Zamore, P. D., Tuschl, T., Sharp, P. A., Battel, D. P. Cell 2000, 101, 25-33).
  • Synthetic RNA duplexes can be used to modulate therapeutically relevant biochemical pathways, including ones which are not accessible through traditional small molecule control.
  • RNA modification of RNA leads to improved physical and biological properties such as nuclease stability (Damha et al Drug Discovery Today 2008, 13(19/20), 842-855), reduced immune stimulation (Sioud TRENDS in Molecular Medicine 2006, 12(4), 167-176), enhanced binding (Koller, E. et al Nucl Acids Res. 2006, 34, 4467-4476), enhanced lipophilic character to improve cellular uptake and delivery to the cytoplasm.
  • RNA modifications of RNA have relied heavily on work-intensive, cumbersome, multi-step syntheses of structurally novel nucleoside analogues and their corresponding phosphoramidites prior to RNA assembly.
  • a major emphasis has been placed on chemical modification of the 2'-position of nucleosides.
  • a rigorous approach to structure-activity-relationship (SAR) studies of chemical modifications will obviously require synthesis and evaluation of all four canonical ribonucleosides [adenosine (A), cytidine (C), uridine (U), guanosine (G)].
  • RNA has centered for the most part on simple conjugation chemistry. Conjugation has largely been performed on either the 3' or the 5'-end of the RNA via alkylamine and disulfide linkers. These modifications have allowed conjugation of RNA to various compounds such as cholesterol, fatty acids,
  • poly(ethylene)glycols various delivery vehicles and targeting agents such as poly(amines), peptides, peptidomimetics, and carbohydrates.
  • This invention relates to the post-synthetic chemical modification of RNA at the 2'-postion on the ribose ring via a silver or copper catalyzed Alkyne, Aldehyde, Amine coupling chemistry [AAA or A 3 or AA 3 coupling ("A A 3 coupling” means alkyne, aldehyde, amine coupling)].
  • the invention 1) avoids complex, tedious multi-step syntheses of each desired modified ribonucleoside; 2) allows diverse chemical modifications using high-fidelity chemistry that is completely orthogonal to commonly used alkylamino, carboxylate and disulfide linker reactivities; 3) allows introduction of functional groups that are incompatible with modern automated solid-phase synthesis of RNA and subsequent cieavage-deprotection steps; 4) allows introduction of functional groups useful as targeting ligands; and 5) enables high-throughput structure-activity relationship studies on chemically modified RNA in 96-well format.
  • FIGURE 1 Systematic evaluation of the impact on knockdown of the 2'-0-propargylamine chemical modification along positions 1 through 19 of the guide strand of a ApoB (9514) siRNA seq.
  • FIGURE 2 Systematic evaluation of the impact on knockdown of the 2'-0-propargylamine chemical modification along positions 1 through 19 of the guide strand of a ApoB (10162) siRNA seq.
  • FIGURE 3 Synthesis of multi AAA 2'-0-propargylamine chemical modification positions 1 through 19 of the guide strand of Luc 80 siRNA sequence.
  • This invention relates to the post-synthetic chemical modification of RNA at the 2'-postion on the ribose ring via silver or copper catalyzed alkyne, aldehyede, amine coupling chemistry (AAA or A or AA coupling).
  • the invention 1) avoids complex, tedious multi-step syntheses of each desired modified ribonucleoside; 2) allows diverse chemical modifications using high-fidelity chemistry that is completely orthogonal to commonly used alkylamino, carboxylate and disulfide linker reactivities; 3) allows introduction of functional groups that are incompatible with modern automated solid-phase synthesis of RNA and subsequent cieavage-deprotection steps; 4) allows introduction of functional groups useful as targeting ligands; and 5) enables high-throughput structure-activity relationship studies on chemically modified RNA in 96-well format.
  • RNA is unstable towards hydrolysis and can undergo auto-catalytic cleavage via
  • RNA with alkyne functional group at the 2 '-position RNA with alkyne functional group at the 2 '-position.
  • the current invention relates to chemical modification of RNA at the 2' ⁇ position of the ribose ring based on "alkyne, aldehyde, amine coupling" chemistry.
  • alkyne, aldehyde, amine coupling Three-component coupling between aldehydes, alkynes and amines is known. Wei et al. Synlett. 20 ⁇ 4, 1472 - 1483.
  • the invention provides a process for introducing 2'- modifications into RNA, said process comprises a) obtaining RNA with an alkyne functional group at the 2'-position on at least one ribose ring; b) creating a solution of RNA in a solvent; and c) adding an aldehyde, an amine and a metal catalyst to the solution to form a reaction and creating a 2 '-modified RNA.
  • the process is conducted in high-throughput format.
  • the step (a) RNA may be purchased or synthesized.
  • the step (b) solvent is selected from aqueous buffer solutions (including phosphate buffers), aqueous DMSO, CH 3 CN, DMF, DM Ac, NMP and a suitable ionic liquid.
  • the step (b) solvent is aqueous DMSO.
  • the step (c) metal catalyst is selected from silver, copper, ruthenium, iridium, iron, zinc or gold.
  • the step (c) metal catalyst is silver.
  • the step (c) metal catalyst is copper with a suitable ligand to stabilize the Cu(I) oxidation state and/or induce asymmetry during amine addition.
  • the step (c) reaction is performed at temperatures between - 0-300°C for 0 to 18 h.
  • the step (c) reaction is performed at temperatures between 5- 120°C for 0.5 to 18 h.
  • step (c) reaction is performed at temperatures between
  • step (c) reaction is performed at temperatures between 60-90°C for 0.5 to l8 h.
  • the step (c) reaction is performed at temperatures between 65-80 °C for 0.5 to 18 h.
  • the invention provides a process for introducing 2 - modifications into RNA, said process comprises a) obtaining RNA with an alkyne functional group at the 2' ⁇ position on at least one ribose ring of an internal nucleotide; b) creating a solution of RNA in a solvent; and c) adding an aldehyde, an amine and a metal catalyst to the solution to form a reaction and creating a 2 '-modified RNA.
  • the process is conducted in high-throughput format.
  • the step (a) RNA may be purchased or synthesized.
  • the step (b) solvent is selected from aqueous buffer solutions (including phosphate buffers), aqueous DMSO, CH 3 CN, DMF, DMAc, NMP and a suitable ionic liquid.
  • the step (b) solvent is aqueous DMSO.
  • the step (c) metal catalyst is selected from silver, copper, ruthenium, iridium, iron, zinc or gold.
  • the step (c) metal catalyst is silver.
  • the step (c) metal catalyst is copper with a suitable ligand to stabilize the Cu(I) oxidation state and/or induce asymmetry during amine addition.
  • the step (c) reaction is performed at temperatures between - 20-300°C for 0 to 18 h.
  • step (c) reaction is performed at temperatures between 5- 120°C for 0.5 to l8 h.
  • the step (c) reaction is performed at temperatures between 20-100°C for 0.5 to 18 h.
  • step (c) reaction is performed at temperatures between 60-90°C for 0.5 to l8 h.
  • step (c) reaction is performed at temperatures between
  • the invention provides a process for introducing 2'- modifications into RNA, said process comprises a) obtaining RNA with an alkyne functional group at the 2 '-position on at least one ribose ring of an internal nucleotide; b) creating a solution of RNA in a solvent; c) adding an aldehyde, an amine and a metal catalyst to the solution to form a reaction and creating a 2'-modified RNA; and d) purifying the 2'-modified RNA.
  • the step (a) RNA may be purchased or synthesized.
  • the step (c) solvent is selected from aqueous buffer solutions (including phosphate buffers), aqueous DMSO, C3 ⁇ 4CN, DMF, D Ac, NMP and a suitable ionic liquid.
  • the step (c) solvent is aqueous DMSO.
  • the step (c) metal catalyst is selected from silver, copper, ruthenium, iridium, iron, zinc or gold.
  • the step (c) metal catalyst is silver.
  • the step (c) metal catalyst is copper with a suitable ligand to stabilize the Cu(I) oxidation state and/or induce asymmetry during amine addition.
  • step (c) reaction is performed at temperatures between -
  • the step (c) reaction is performed at temperatures between 5- 120°C for 0.5 to 18 h.
  • the step (c) reaction is performed at temperatures between 20-100°C for 0.5 to 18 h.
  • step (c) reaction is performed at temperatures between 60-90°C for 0.5 to 18 h.
  • the step (c) reaction is performed at temperatures between 65-80°C or 0.5 to 18 h.
  • the step (d) purification is performed in high-throughput format on 96-well CIS cartridges (solid-phase extraction) or strong-a ion-exchange-HPLC or reverse-phase HPLC or poly(acrylamide) gel electrophoresis (PAGE) or size-exclusion chromatography.
  • the invention provides a process for introducing 2'- modifications into RNA, said process comprises a) obtaining RNA with an alkyne functional group at the 2'-position on at least one ribose ring of an internal nucleotide; b) creating a solution of RNA in a solvent; c) adding an aldehyde, an amine and a metal catalyst to the solution to form a reaction and creating a 2'-modified RNA; d) cooling the solution and adding a fluoride source; e) heating the solution; f) cooling the solution and adding a diluent; and g) purifying the 2'-modified RNA.
  • the step (a) RNA may be purchased or synthesized.
  • the step (c) solvent is selected from aqueous buffer solutions (including phosphate buffers), aqueous DMSO, CH 3 CN, DMF, DMAc, NMP and a suitable ionic liquid.
  • the step (c) solvent is aqueous DMSO.
  • the step (c) metal catalyst is selected from silver, copper, ruthenium, iridium, iron, zinc or gold.
  • the step (c) metal catalyst is silver.
  • the step (c) metal catalyst is copper with a suitable ligand to stabilize the Cu(I) oxidation state and/or induce asymmetry during amine addition.
  • the step (c) reaction is performed at temperatures between - 20-300°C for 0 to 18 h.
  • the step (c) reaction is performed at temperatures between 5- 120°C for 0.5 to 18 h.
  • the step (c) reaction is performed at temperatures between 20-100°C for 0.5 to 18 h.
  • step (c) reaction is performed at temperatures between 60-90°C for 0.5 to 18 h.
  • step (c) reaction is performed at temperatures between 65-80°C for 0.5 to l8 h.
  • the step (e) fluoride source is Et3N-3HF
  • the step (e) fluoride source is ammonium fluoride.
  • the step (f) diluent is NaCl.
  • the step (g) purification is performed in high-throughput format on 96-well CI 8 cartridges (solid-phase extraction) or strong-anion-exchange-HPLC or reverse-phase HPLC or poly(acrylamide) gel electrophoresis (PAGE) or size-exclusion chromatography.
  • the instant invention also discloses a method for attaching targeting ligands to RNA utilizing the process described herein.
  • the instant invention further discloses a method for attaching targeting ligands to internal nucleotides in RNA utilizing the process described herein.
  • 2'-modified RNA means a RNA wherein at least one ribose ring is modified at the 2'-position.
  • Alkyne functional group means any chemical compound containing an alkyne functional group.
  • the preferred alkyne functional group is propargyl.
  • High-throughput format means that several operations are run in parallel fashion such as for example in 96-well plate chemical synthesis, 96-well plate purification, 96- well plate chromatographic analysis and 96-well plate mass spectrometric analysis.
  • Internal nucleotide means a nucleotide in an RNA molecule that is not at the 3'- or 5'-end.
  • the internal nucleotides in a 21mer siRNA occur at positions 2-20.
  • RNA means a chemically modified or unmodified ribonucleic acid molecule (single stranded or double stranded) comprising at least 3 nucleotides, including but not limited to miRNA and siRNA. In another embodiment, “RNA” means miRNA. In another
  • RNA means siRNA.
  • Chemical modifications include, for example,
  • the base can be a canonical base (A, G, T and U) or a modified or universal base (including but not limited to inosine and nitroindole). See US2006/0240554.
  • Aldehyde means any chemical compound containing an aldehyde functional group.
  • Amine means any chemical compound containing an amine functional group.
  • Metal catalyst means any chemical form of silver, copper, iridium, ruthenium, iron, zinc or gold. Including solid-supported variants.
  • metal catalyst include Agl, CuBr, C Br-Me2S, Cul, CuSC>4 or CuOAc and a suitable reducing agent such as sodium ascorbate.
  • Ribose ring means the ribose moiety in a ribonucleotide.
  • Targeting ligand means a conjugate delivery moiety capable of delivering the RNA to a target cell of interest.
  • Targeting ligands include, but are not limited to, lipids (cholesterol), sugars (NAG), proteins (transferrin), peptides, poly(ethylene)glycols and antibodies. See Juliano et al., Nucleic Acids Research, 2008, 1-14, doi:10.1093/nar/gkn342.
  • the present invention provides a process for introducing chemical modifications into RNA at the 2'-position on the ribose ring. It is well known in the art that RNA are useful for therapeutic and research purposes.
  • RNA The synthesis of RNA is well known in the art.
  • a suitable 2'-O-propargyl nucleoside phosphoramidite is incorporated into RNA using modern techniques based on the phosphoramidite approach.
  • the crude, solid-support bound protected oligonucleotide is then treated with aqueous methylamine to remove nucleobase and phosphate protecting groups.
  • the crude product is then lyophilized to remove volatiles.
  • the crude product is dissolved in DMSO:H 2 0, treated with a suitable aldehyde, a suitable amine and silver or copper catalyst (scheme 1). After aging an appropriate amount of time, the reaction mixture is treated with fluoride to remove the 2 r -0-teri-butyldimethylsilyl protecting groups.
  • the crude product is then purified to obtain the chemically modified RNA.
  • RNA (-50 nmol) containing at least one alkyne functional group (shown below) in 96-well format was dissolved in DMSO;water (75:25, 40 ⁇ ,).
  • the "alkyne, amine, aldehyde coupling" reaction can be utilized to introduce multiple chemical modifications in one synthetic operation.
  • the A 3 coupling reaction was performed to introduce four units of propargylamines on RNA
  • RNA oligomers with the first nucleotide, Adenine (A), replaced with 2'-0-propargyl-Adenine. Then, a second sequence, in which the second nucleoside (U) was replaced with 2'-0 ⁇ propargyI -uridine was synthesized, keeping all other nucleotides unchanged.
  • L 2'OmeUridine
  • L 2'-0-Propargyl Adenine
  • M 2'-0-Propargyl Cytidine
  • W 2'-0- Propargyl Guanosine
  • Y 2'-0-Propargyl Uridine.
  • Hepal-6 cells were transfected with 10 nM of either the unmodified, modified, or negative control siRNA using a commercial lipid transfection reagent.
  • the target mRNA was assessed for degradation using standard Taqman procedures. Modified Multiplex luciferase report assay for i vitro duration study
  • Multiplex luciferase assay for in vitro duration study is modified from the manufacturer's instruction using HeLa-luc cell line. Briefly, the cell viability and the luciferease expression at the same well are determined by CellTiter-FluorTM (Promega, Cat# G6082) and Bright-GloTM (Promega Cat# E2620) sequentially.
  • HeLa-luc cell line is a stable firefly luciferase reporter expression cell line.
  • Bright-GloTM luciferase assay system contains the stable substrate - luciferin and assay buffer.
  • the luminescent reaction of luciferease and luciferin has high quantum yield and can be detected as luminescence intensity, which represents the luciferase expression level.
  • Target siRNAs containing luciferase coding region is designed to be transfected into the HeLa-luc cells. Once the target is effected, the luciferase expression is reduced accordingly. Therefore, the siRNA silencing efficacy can be determined by the relative luminecence intensity of treated cells.
  • CellTiter-fluor kit measures the conserved and constitutive protease activity within live cells and therefore serves as a marker of cell viability, using a fluorogenic, cell-permeable peptide substrate (glycyl- phenylalanyl-aminofluorocoumarin; GF-AFC).
  • Luciferase stable expressed HeLa-luc cell cells are plated in 96-well plates at density of 4,500 cells per well in 100 ⁇ DMEM media without antibiotics 24 hours prior to transfection.
  • si NA transfection is performed using the RNAiMAXTM (Invitrogen). Briefly, 0,05 ⁇ siRNA are mixed with Opti-MEMmedia and RNAiMAX and incubated at room temperature for 15 rnin. The mix is then added to the cells. The final siRNA concentration is 1 nM. Cell plates for all time points are transfected at same time with a medium change at 6 hours post-transfection into 100 ⁇ of fresh completed DMEM (DMEM + 10% FBS +
  • In vitro duration is determined by the luciferase expression post-transfection at four time points: day 1, day 2, day 5 and day 7. Addition medium changes are performed at day 2 and day 5 into 100 ⁇ , of fresh completed DMEM (DMEM + 10% FBS + Penn/strep). Luciferase levels are determined using the Bright- Glo Luminescence Assay (Promega) and measuring the wells on an Envison instrument (Perkin Elmer) according to manufacturer's instructions.
  • the cell viability of the same treatment wells is measured using CellTiter-fluor kit (Promega) according to manufacturer's instructions.
  • This assay measures the conserved and constitutive protease activity within live cells and therefore servers as a marker of cell viability, using a fluorogenic, cell-permeable peptide substrate (glycyl-phenylalanyl-aminofluorocoumarin; GF-AFC).
  • the fluorescence was measured on the Envision using exciton filter at 405 nm and emission filter at 510 nm.
  • the luciferase expression was normalized to cell viability. The log of this number was calculated to determine the luciferase protein that was degraded (knockdown). A non-targeting siRNA was subtracted from this value to account for non-specific background.
  • RNAs made by the process of the invention are useful in high-throughput structure-activity relationship studies on chemically modified R A in 96-well format.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Saccharide Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP11825711.2A 2010-09-17 2011-09-09 Chemische modifikation von rna an der 2'-position des riboserings mittels aaa-kopplung Withdrawn EP2616549A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38377910P 2010-09-17 2010-09-17
PCT/US2011/050903 WO2012036972A1 (en) 2010-09-17 2011-09-09 Chemical modification of rna at the 2'-position of the ribose ring via aaa coupling

Publications (1)

Publication Number Publication Date
EP2616549A1 true EP2616549A1 (de) 2013-07-24

Family

ID=45831914

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11825711.2A Withdrawn EP2616549A1 (de) 2010-09-17 2011-09-09 Chemische modifikation von rna an der 2'-position des riboserings mittels aaa-kopplung

Country Status (2)

Country Link
EP (1) EP2616549A1 (de)
WO (1) WO2012036972A1 (de)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8114636B2 (en) * 2006-02-10 2012-02-14 Life Technologies Corporation Labeling and detection of nucleic acids

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2012036972A1 *

Also Published As

Publication number Publication date
WO2012036972A1 (en) 2012-03-22

Similar Documents

Publication Publication Date Title
US9441228B2 (en) Post-synthetic chemical modification of RNA at the 2′-position of the ribose ring via “click” chemistry
US20080213891A1 (en) RNAi Agents Comprising Universal Nucleobases
AU2008256871A1 (en) Hydroxymethyl substituted RNA oligonucleotides and RNA complexes
TWI772632B (zh) 髮夾型單股rna分子之製造方法、單股寡rna分子及包含其之套組
Odadzic et al. Synthesis of 2′-O-modified adenosine building blocks and application for RNA interference
AU2018223027A1 (en) Post-synthetic orthogonal amidation plus metal catalyzed azide-alkyne cycloaddition click chemistry on siRNA
GB2479833A (en) Modified nucleotides
US10844376B2 (en) Structurally-enhanced miRNA inhibitor S-TuD
Sato et al. Synthesis and characterization of novel (S)-5′-C-aminopropyl-2′-fluorouridine modified oligonucleotides as therapeutic siRNAs
Kajino et al. (S)-5′-C-Aminopropyl-2′-O-methyl nucleosides enhance antisense activity in cultured cells and binding affinity to complementary single-stranded RNA
WO2011090968A1 (en) Post-synthetic chemical modification of rna at the 2'-position of the ribose ring via "click" chemistry
WO2012036972A1 (en) Chemical modification of rna at the 2'-position of the ribose ring via aaa coupling
US20210032282A1 (en) Nucleic acid complex, method for forming nucleic acid hybridization, pharmaceutical composition, nucleic acid probe, and complementary-strand nucleic acid complex
AU2012254580B2 (en) Process for preparing phosphate compound bearing isotope
Rydzik et al. Synthesis of 2′-modified N6-methyladenosine phosphoramidites and their incorporation into siRNA
EP3187584B1 (de) Verfahren zur nicht-enzymatischen kombination von nukleinsäureketten
Hirano et al. Synthesis and Application of Interstrand Cross‐Linked Duplexes by Covalently Linking a Pair of Abasic Sites
Wu et al. Synthesis of Site‐Specifically Phosphate‐Caged siRNAs
Zatsepin et al. Efficient conjugation and preferential DNA binding of oligonucleotides containing 2′-O-(2-oxoethyl) arabinouridine
Matsubara et al. Synthesis of siRNAs incorporated with cationic peptides R8G7 and R8A7 and the effect of the modifications on siRNA properties
JP6621831B2 (ja) ヌクレオシド誘導体及びその利用
Koyasu et al. Non-terminal conjugation of small interfering RNAs with spermine improves duplex binding and serum stability with position-specific incorporation
AU2013273719B2 (en) UNA amidites for therapeutic oligomers
を持つ機能性オリゴヌクレオチドの開発 Development of chemically functionalized oligonucleotides for DNA-binding proteins labeling and DNA cross-linking
Domingo Investigations into the effect of nucleoside modifications on the physicochemical properties and biological function of DNA and RNA

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130417

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20131017