WO2008077592A1 - Method for purifying rna on a preparative scale by means of hplc - Google Patents

Method for purifying rna on a preparative scale by means of hplc Download PDF

Info

Publication number
WO2008077592A1
WO2008077592A1 PCT/EP2007/011294 EP2007011294W WO2008077592A1 WO 2008077592 A1 WO2008077592 A1 WO 2008077592A1 EP 2007011294 W EP2007011294 W EP 2007011294W WO 2008077592 A1 WO2008077592 A1 WO 2008077592A1
Authority
WO
WIPO (PCT)
Prior art keywords
rna
separation
porous
vol
hplc
Prior art date
Application number
PCT/EP2007/011294
Other languages
French (fr)
Inventor
Thomas Ketterer
Florian VON DER MüLBE
Ladislaus Reidel
Thorsten Mutzke
Original Assignee
Curevac Gmbh
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39186980&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2008077592(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to JP2009541898A priority Critical patent/JP5307724B2/en
Priority to AT07857017T priority patent/ATE481482T1/en
Priority to DE602007009295T priority patent/DE602007009295D1/en
Priority to KR1020097012957A priority patent/KR101183173B1/en
Priority to DK07857017.3T priority patent/DK2092064T3/en
Application filed by Curevac Gmbh filed Critical Curevac Gmbh
Priority to PL07857017T priority patent/PL2092064T3/en
Priority to CN200780047133.2A priority patent/CN101563457B/en
Priority to CA2670727A priority patent/CA2670727C/en
Priority to US12/520,172 priority patent/US8383340B2/en
Priority to AU2007338360A priority patent/AU2007338360B2/en
Priority to EP07857017A priority patent/EP2092064B1/en
Publication of WO2008077592A1 publication Critical patent/WO2008077592A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • C12N15/101Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers by chromatography, e.g. electrophoresis, ion-exchange, reverse phase
    • 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

Definitions

  • the present invention relates to a method for purifying RNA on a preparative scale by means of HPLC and to the use of a porous reversed phase as stationary phase in this method.
  • HPLC abbreviation for "High Performance (High Pressure) Liquid Chromatography” is an established method of separating mixtures of substances, which is widely used in biochemistry, analytical chemistry and clinical chemistry.
  • An HPLC apparatus consists in the simplest case of a pump with eluent reservoir containing the mobile phase, a sample application system, a separation column containing the station- ary phase, and the detector.
  • a fraction collector may also be provided, with which the individual fractions may be separately collected after separation and are thus available for further applications.
  • RNA analysis using ion pair reversed phase HPLC is known from A. Azarani and K. H. Hecker (Nucleic Acids Research, vol. 29, no. 2 e7).
  • the stationary phase here consists of a non-porous alkylated polystyrenedivinylbenzene matrix. Elution proceeds with a buffer system of two eluents.
  • Buffer A consists of an aqueous solution of 0.1 M triethylammonium acetate (TEAA), pH 7.0
  • buffer B consists of an aqueous solution of 0.1 M TEAA, pH 7.0, with 25% acetonitrile.
  • Elution was performed using the following three gradient sys- terns: 38-40% B over 1 minute, to 60% B over 15 minutes, to 66% B over 6 minutes, to 70% B over 0.5 minutes, to 100% B over 0.5 minute, holding at 100% B for 1 minute, to 38% over 1 minute, holding at 38% B for 2 minutes.
  • the following gradient program was used: 38-60% over 30 minutes, to 100% B over 2 minutes, to 38% B over 3 minutes.
  • the following was used as a third gradient program: 38-40% B over 1 minute, to 60% B over 3 minutes, to 100% B over 1 minute, holding at 100% B over 6 minutes, to 38% B over 1 minute, holding at 38% B over 1 minute.
  • the object of the present invention is here to improve a method of this type to the effect that it no longer exhibits the disadvantages of the prior art.
  • RNA is purified by means of HPLC using a porous reversed phase as stationary phase.
  • a porous reversed phase is used.
  • HPLC by means of HPLC
  • HPLC various HPLC methods as well as low or normal pressure liquid chromatography methods, which may be used to carry out the inventive method.
  • HPLC reversed phase HPLC
  • chromatography size exclusion chromatography
  • gel filtration gel filtration
  • affinity chroma- tography gel filtration
  • hydrophobic interaction chromatography hydrophobic interaction chromatography or ion pair chromatography
  • reversed phase HPLC consists of a non-polar stationary phase and a moderately polar mobile phase.
  • One common stationary phase is e.g. a silica which has been treated with e.g.
  • RMe 2 SiCI where R is a straight chain alkyl group such as C 18 H 37 or C 8 H 17 .
  • the retention time is therefore longer for mole- cules which are more non-polar in nature, allowing polar molecules to elute more readily. Retention time is increased by the addition of polar solvent to the mobile phase and decreased by the addition of more hydrophobic solvent.
  • the various techniques mentioned above operate on the principle of hydrophobic interactions which result from repulsive forces between a relatively polar solvent, the relatively non-polar analyte, and the non-polar stationary phase (reversed phase principle).
  • the driving force in the binding of the analyte (the RNA molecule) to the stationary phase is the decrease in the area of the non-polar segment of the analyte molecule exposed to the sol- vent.
  • This hydrophobic effect is dominated by the decrease in free energy from entropy associated with the minimization of the ordered molecule-polar solvent interface.
  • the hydrophobic effect is decreased by adding more non-polar solvent into the mobile phase. This shifts the partition coefficient such that the analyte spends some portion of time moving down the column in the mobile phase, eventually eluting from the column.
  • RNA molecule as an analyte may play an important role in its retention characteristics.
  • an analyte having more apolar functional groups results in a longer retention time because it increases the molecule's hydrophobicity.
  • Very large molecules can result in incomplete interaction between the large analyte surface and the alkyl chain. Retention time increases with hydrophobic surface area which is roughly inversely proportional to solute size. Branched chain compounds elute more rapidly than their corresponding isomers because the overall surface area is decreased.
  • RNA molecule retention Aside from mobile phase hydrophobicity, other mobile phase modifiers can - according to the invention - affect analyte (RNA molecule) retention.
  • the addition of inorganic salts may be considered to cause a linear increase in the surface tension of aqueous solutions, and because the entropy of the analyte-solvent interface is controlled by surface tension, the addition of salts tend to increase the retention time.
  • Another important compo- nent, which may be used according to the invention is pH, since this can change the hydrophobicity of the analyte. For this reason, a buffering agent, such as sodium phosphate or other common buffering agents, may be used to control the pH.
  • An organic acid such as formic acid or most commonly trifluoroacetic acid, may be added to mobile phase.
  • the above modifications may serve multiple purposes: They control pH, neutralize the charge on any residual exposed material on the stationary phase and act as ion pairing agents to neutralize charge on the analyte. The effect varies depending on use but generally improve the chromatography.
  • the term "purification" is understood to mean that the desired RNA in a sample is separated and/or isolated from the impurities present therein.
  • the RNA is present in a purer form than in the originally introduced RNA-containing sample prior to HPLC purification.
  • Undesired constituents of RNA- containing samples which therefore need to be separated may in particular be degraded fragments or fragments which have arisen as a result of premature termination of transcription, or also excessively long transcripts if plasmids are not completely linearised.
  • impurities such as enzymes, for example RNases and polymerases, and nucleotides may be separated.
  • RNA is purified which has a higher purity after purification than the starting material. It is desirable in this respect for the degree of purity to be as close as possible to 100%. A degree of purity of more than 70%, in particular 80%, very particularly 90% and most favourably 99% or more may be achieved in this way.
  • the method according to the invention results in preparative RNA purification.
  • an analytical HPLC method a distinctly smaller quantity is introduced and separated than in a preparative HPLC method.
  • quanti- ties of 8 ng to 1000 ng were introduced.
  • a preparative HPLC method should be understood to mean an HPLC method in which relatively large quantities of RNA are purified.
  • Such relatively large quantities are for example quantities of 0.5 mg or more, in particular 1 .0 mg to 1000 mg or more, very particularly approximately 1.5 mg or more, upscal- ing even to the kg range being possible.
  • the above statements are therefore to be under- stood to mean that these quantities relate to a single HPLC run. If a plurality of HPLC runs are performed, the quantity increases in direct proportion to the number of HPLC runs.
  • the method according to the invention exhibits a series of advantages: using the method according to the in- vention substantially larger quantities of RNA may be separated than is possible with the method known from the prior art. Using the method according to the invention, these relatively high quantities are obtained with a high degree of purity. Separation of degraded fragments or excessively long fragments or of fragments which have arisen as a result of premature termination of transcription is possible. In addition, good separation of further impurities present in RNA samples, such as enzymes, for example RNases and polymerases, and nucleotides is possible. These relatively large quantities of RNA may be recovered within minutes, even from samples having a very high level of contamination with impurities.
  • RNA recovery proceeds reliably, i.e. high purity RNA is recovered with great reliability.
  • a high level of resolution may be achieved.
  • the method according to the invention may additionally easily be operated automatically. It is thus most suitable for routine purification of RNA on a preparative scale.
  • RNA is stable under the conditions of the method according to the invention, i.e. degradation during HPLC purification into RNA fragments and thus the occurrence of new impurities and a reduction in the yield of desired RNA during HPLC purification is avoided.
  • the method according to the invention may thus be performed in a simple, rapid, inexpensive manner, accurately and with reliable results. Isolation of RNA after HPLC separation may proceed simply with a fraction collector, i.e. the recovery of RNA may be achieved very simply, it being possible for direct further processing of the RNA likewise to take place. Detection may finally be performed very sensitively.
  • RNA which is to be purified with the method according to the invention is ribonucleic acid of any type.
  • the RNA is particularly preferably selected from tRNA, rRNA, mRNA or whole-cell RNA.
  • the RNA may also comprise aptamers and ribozymes.
  • the RNA to be isolated may be single-stranded or double-stranded. Single-stranded RNA may optionally form secondary structures by refolding, the RNA to be separated typically being single-stranded.
  • the RNA may be unlabelled or also labelled (with a fluorescent label or a radiolabel or an antibody epitope).
  • a labelled RNA is digoxigenin-labelled ⁇ -actin RNA.
  • the RNA to be purified may be native or non-native RNA.
  • Native RNA is preferably used, which is prepared using a for example cellular or in vitro expression system.
  • the RNA is then isolated and the isolate purified using the method according to the invention.
  • chemically synthesised RNA may also be purified using the method according to the invention.
  • the RNA may also contain non-native nucleotides, wherein the chemical modifications may be present in the backbone of the RNA, for example phosphor- thioate, in the structure of the nucleotide base, for example hypoxanthine, dihydrouracil, pseudouracil, 2-thiouracil, 4-thiouracil, 5-aminouracil, 5-(C1 -C6)-alkyluracil, 5-(C2-C6)- alkenyluracil, 5-(C2-C6)-alkynyluracil, 5-(hydroxymethyl)uracil, 5-chlorouracil, 5- fluorouracil, 5-bromouracil, 5-hydroxycytosine, 5-(C1 -C6)-alkylcytosine, 5-(C2-C6)- alkenylcytosine, 5-(C2-C6)-alkynylcytosine, 5-chlorocytosine, 5-fluorocytosine, 5- bromocytosine, N ⁇ 2
  • modified nucleotides are 2-amino-6-chloropurine-riboside-5'-triphosphate, 2-aminoadenosine-5 '-triphosphate, 2-thiocytidine-5 '-triphosphate, 2-thiouridine-5'- triphosphate, 4-thiouridine-5'-triphosphate, 5-aminoallylcytidine-5'-triphosphate, 5- aminoallyluridine-5'-triphosphate, 5-bromocytidine-5'-triphosphate, 5-bromouridine-5'- triphosphate, 5-iodocytidine-5 '-triphosphate, 5-iodouridine-5'-triphosphate, 5- methylcytidine-5 '-triphosphate, 5-methyluridine-5 '-triphosphate, 6-azacytidine-5'- triphosphate, 6-azauridine-5 '-triphosphate, 6-chloropurine-riboside-5'-triphosphate, 7- deazaadenosine-5 '-triphosphat
  • the RNA to be separated has a size of up to about 15000 nucleotides (as single stranded RNA molecule) or base pairs (as double stranded RNA molecule), in particular 100 to 10000, more preferably 500 to 10000 nucleotides or base pairs, even more preferably 800 to 5000 nucleotides or base pairs and even more preferably 800 to 2000 nucleotides or base pairs.
  • this size of RNA it has proved possible to achieve very good results with regard to purification of the RNA, since the method according to the invention is particularly well suited to RNA of this size.
  • smaller RNA fragments for example with a length of 30-1000, 50- 1000 or 100-1000 or 20-200, 20-100, 20-50 or 20-30 nucleotides may also be separated in this way.
  • RNA to be separated is mRNA
  • this will preferably code for proteins, in particular those which have an antigen character, and for example all recombinantly produced or naturally occurring proteins, which are known to a person skilled in the art from the prior art and are used for therapeutic, diagnostic or research purposes.
  • the antigens are then tumour antigens or antigens of pathogens, for example viral, bacterial or protozoal organisms.
  • proteins include inter alia growth hormones or growth factors, for example for promoting growth in a (transgenic) living organism such as for example TGF and the IGFs ("insulin-like growth factors"), proteins which influence metabolism and/or haematopoiesis, such as for example (alpha-l )-anti-trypsin, LDL receptor, erythropoietin (EPO), insulin, GAT A-1 , etc., or proteins such as for example factors VIII and Xl of the blood clotting system, etc..
  • Such proteins further include enzymes, such as for example
  • proteases such as for example papain, bromelain, keratinases, trypsin, chymotrypsin, pepsin, rennin (chymosin), suizyme, nortase, etc., and protease inhibitors (for example for the treatment of HIV infections), such as for example protease inhibitors se- lected from a group consisting of AG1 776, amprenavir (141W94 or VX-478), atazanavir (BMS-232632), cathepsin S protease inhibitor, D1927, D9120, fosamprenavir (GW-433908 or VX-175), GS 9005, GW640385 (VX-385), HCV protease inhibitor, indinavir (MK-639), L- 756 423
  • the proteins coded from the RNA separated according to the invention may likewise comprise proteins which stimulate or inhibit cell signal transmission, for example cytokines, etc.. Such proteins therefore also comprise for example cytokines of class I of the cytokine family, which comprise 4 posi- tionally conserved cysteine residues (CCCC) and one conserved sequence motif Trp-Ser-X- Trp-Ser (WSXWS), wherein X represents a non-conserved amino acid.
  • cytokines of class I of the cytokine family which comprise 4 posi- tionally conserved cysteine residues (CCCC) and one conserved sequence motif Trp-Ser-X- Trp-Ser (WSXWS), wherein X represents a non-conserved amino acid.
  • Cytokines of class I of the cytokine family comprise the GM-CSF subfamily, for example IL-3, IL-5, GM-CSF, the IL-6 subfamily, for example IL-6, IL-1 1 , IL-12, or the IL-2 subfamily, for example IL-2, IL-4, IL-7, IL-9, IL-15, etc., or the cytokines IL-I , IL- 1 , IL-10 etc..
  • such proteins may also comprise cytokines of the class Il of the cytokine family (interferon receptor family), which likewise comprise 4 positionally conserved cysteine residues (CCCC), but no conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS).
  • Cytokines of class Il of the cytokine family include for example IFN-alpha, IFN-beta, IFN-gamma, etc..
  • the proteins coded by the separated RNA may furthermore also include cytokines of the tumour necrosis family including for example TNF-alpha, TNF-beta, CD40 ligand, Fas ligand, etc., or cytokines of the chemokine family, in particular interferons, interleukins, colony-stimulating factors and tumour necrosis factors, and interact with G protein, for example IL-8, MIP-1 , RANTES, CCR5, CXR4, etc.
  • Such proteins may also be selected from apoptosis factors or apoptosis- related or -linked proteins, including AIF, Apaf, for example Apaf-1 , Apaf-2, Apaf-3, or APO-2 (L), APO-3 (L), apopain, Bad, Bak, Bax, Bcl-2, Bcl-x L , Bcl-x s , bik, CAD, calpain, cas- pases for example caspase-1 , caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, cas- pase-7, caspase-8, caspase-9, caspase-10, caspase-1 1 , ced-3, ced-9, c-Jun, c-Myc, crm A, cytochrome C, CdR1, DcRI , DD, DED, DISC, DNA-PKc 5 , DR3, DR4, DR5, FADD/MORT-1 , FA
  • the proteins coded by RNA separated according to the invention may also be selected from antigens, for example from tumour-specific surface antigens (TSSA), for example 5T4alpha5 betai integrin, 707-AP, AFP, ART-4, B7H4, BAGE, Bcr-abl, MN/C IX-antigen, CAI 25, CAMEL, CAP-1 , CASP-8, beta-catenin/m, CD4, CD19, CD20, CD22, CD25, CDC27/m, CD30, CD33, CD52, CD56, CD80, CDK4/m, CEA, CT, Cyp-B, DAM, EGFR, ErbB3, ELF2M, EMMPRIN, EpCam, ETV6-AML1 , G250, GAGE, GnT-V, Gp100, HAGE, HER-2/new, HLA-A*0201 - R170I, HPV-E7, HSP70-2M, HAST-2, hTERT (or
  • Proteins which may be coded by the RNA separated according to the invention additionally also include proteins or protein sequences which exhibit sequence identity of at least 80% or 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99% to one of the above-described proteins.
  • the mRNA to be separated may exhibit the following modifications relative to a corresponding wild-type mRNA, which may either be present as alternatives or in combination.
  • the G/C content of the region of the modified mRNA coding for a peptide or polypeptide may be greater than the G/C content of the coding region of the wild-type mRNA coding for the peptide or polypeptide, the coded amino acid sequence being unchanged relative to the wild-type.
  • This modification is based on the fact that the sequence order of the mRNA domain to be translated is essential for efficient mRNA translation. In this respect, the composition and sequence of the various nucleotides has an important part to play.
  • sequences with an elevated G(guanosine)/C(cytosine) content are more stable than sequences with an elevated A(adenosine)/U(uracil) content.
  • the codons are varied relative to the wild-type mRNA in such a manner that they have a greater content of G/C nucleotides. Since several codons code for one and the same amino acid (degeneration of the genetic code), it is possible to determine the codons which are most favourable for stability (alternative codon usage). On the other hand it is also possible to provide a translation-optimised RNA with the method according to the invention.
  • a reversed phase is used as the stationary phase for HPLC purification.
  • a nonpolar compound serves as the stationary phases and a polar solvent, such as mixtures of water, which is generally used in the form of buffers, with e.g. acetonitrile and/or methanol, serves as the mo- bile phase for elution.
  • the material packed on the column which is used as stationary phase, may be provided in bead form or as a polymerized "block", i.e. a block which fills a substantial part of the chromatography column.
  • the polymeric stationary phase is porous in its nature, which means that the beads or the block are characterized by pores.
  • the porous reversed phase material is provided with a particle size of 8.0 ⁇ m to 50 ⁇ m, in particular 8.0 to 30, still more preferably 8.0 to 25 ⁇ m.
  • the reversed phase material may be present in the form of small spheres.
  • the method according to the invention may be performed particularly favourably with a porous reversed phase with this particle size, optionally in bead form, wherein particularly good separation results are obtained.
  • the reversed phase used in the method according to the invention is porous and has specific particle sizes. With stationary reversed phases which are not porous and thus differ completely with regard to particle size from the subject matter of the present invention as described for example by A. Azarani and K.H. Hecker, on the other hand, excessively high pressures are built up, such that preparative purification of the RNA is possible only with great difficulty, if at all.
  • the reversed phase has a pore size of 1000 ⁇ to 5000 A, in particular a pore size of 1000 A to 4000 A, more pref- erably 1500 A to 4000 A, 2000 A to 4000 A or 2500 A to 4000 A.
  • Particularly preferred pore sizes for the reversed phases are 1000 A to 2000 A, more preferably 1000 A to 1500 A and most preferably 1000 A to 1200 A or 3500-4500 A.
  • a pore size of 1000 A to 5000 A, in particular a pore size of 1000 A to 4000 A, more pref- erably 1500 A to 4000 A, 2000 A to 4000 A or 2500 A to 4000 A may be suitable to separate a RNA from other components of a mixture, the RNA having a size as mentioned above of up to about 15000 nucleotides (as single stranded RNA molecule) or base pairs (as double stranded RNA molecule), in particular 100 to 10000, more preferably 500 to 10000 nucleotides or base pairs, even more preferably 800 to 5000 nucleotides or base pairs and even more preferably 800 to 2000 nucleotides or base pairs.
  • the pore size of the reversed phase may also be selected in dependence of the size of the RNA to be separated, i.e. a larger pore size may be selected, if larger RNA molecules are to be separated and smaller pore sizes may be selected, if smaller RNA molecules may be selected.
  • a larger pore size may be selected, if larger RNA molecules are to be separated and smaller pore sizes may be selected, if smaller RNA molecules may be selected.
  • a pore size for the reversed phase of about 2000 A to about 5000 A, more preferably of about 2500 to about 4000, most preferably of about 3500 to about 4500 A may thus be used to separate larger RNA molecules, e.g. RNA molecules of 100 to 10000, more preferably 500 to 10000 nucleotides or base pairs, even more preferably 800 to 5000 nucleotides or base pairs and even more preferably 800 to 2000 nucleotides or base pairs.
  • a pore size of for the reversed phases of about 1000 ⁇ to about 2500 A, more preferably of about 1000 A to about 2000 A, and most preferably of about 1000 A to 1200 A may be used to separate smaller RNA molecules, e.g. RNA molecules of about 30-1000, 50-1000 or 100-1000 or 20-200, 20-100, 20-50 or 20- 30 nucleotides may also be separated in this way.
  • any material known to be used as reverse phase stationary phase in particular any polymeric material may be used for the inventive method, if that material can be provided in porous form.
  • the stationary phase may be composed of organic and/or inorganic material.
  • polymers to be used for the present invention are (non-alky lated) polystyrenes, (non-alkylated) polystyrenedivinylbenzenes, silica gel, silica gel modified with non-polar residues, particularly silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, silica gel modified with phenyl ic residues, polymethacrylates, etc..
  • dextran including e.g. Sephadex ® and Sephacryl ® materials, agarose, dextran/agarose mixtures, poly- acrylamide, etc..
  • dextran including e.g. Sephadex ® and Sephacryl ® materials, agarose, dextran/agarose mixtures, poly- acrylamide, etc.
  • the material for the reversed phase is a porous polystyrene polymer, a (non-alkylated) (porous) polystyrenedivinylbenzene polymer, porous silica gel, porous silica gel modified with non-polar residues, particularly porous silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, porous silica gel modified with phenylic residues, porous polymethacrylates, wherein in particular a porous polystyrene polymer or a non-alkylated (porous) polystyrenedivinylbenzene may be used.
  • a non-alkylated porous polystyrenedivinylbenzene which is very particularly preferred for the method according to the invention is one which, without being limited thereto, may have in particular a particle size of 8.0 ⁇ 1.5 ⁇ m, in particular 8.0 ⁇ 0.5 ⁇ m, and a pore size of 1000- 1500 A, in particular 1000-1200 A or 3500-4500 A.
  • This stationary phase described in greater detail above is conventionally located in a column.
  • V2A steel is conventionally used as the material for the column, but other materials may also be used for the column provided they are suitable for the conditions prevailing during HPLC.
  • the column is straight. It is favourable for the HPLC column to have a length of 5 cm to 100 cm and a diameter of 4 mm to 25 mm.
  • Columns used for the method according to the invention may in particular have the following dimensions: 50 mm long and 7.5 mm in diameter or 50 mm long and 4.6 mm in diameter, or 50 mm long and 10 mm in diameter or any other dimension with regard to length and diameter, which is suitable for preparative recovery of RNA, even lengths of several metres and also larger diameters being feasible in the case of upscaling.
  • the dimensions are here geared towards what is technically possible with liquid chromatography.
  • the method according to the invention is performed as an ion pair method, wherein an ion with a lipophilic residue and positive charge is added to the mobile phase as counterion for the negatively charged RNA.
  • an ion pair with lipophilic character is obtained, which is slowed down by the nonpolar stationary phase of the reversed phase system.
  • the precise conditions for the ion pair method have to be empirically worked out for each specific separation problem.
  • the size of the counterion, its concentration and the pH value of the solution contribute greatly to the separation result. This means that the empirically established conditions of an ion pair method cannot be straightforwardly applied to a different separation problem.
  • Alkylammo- nium salts such as triethylammonium acetate, and/or tetraalkylammonium compounds, such as tetrabutylammonium, are favourably added in the method according to the inven- tion.
  • 0.1 M triethylammonium acetate is added and the pH value is adjusted to about 7.
  • RNA on a preparative scale, in a particularly favourable manner.
  • the following may be considered as further buffer solutions: trifluoroace- tic acid, acetic acid, formic acid, acetate buffer, phosphate buffer, tetrabutylammonium bisulfate, tetrabutylammonium bromide and tetrabutylammonium chloride.
  • Selection of the mobile phase depends on the type of separation desired. This means that the mobile phase established for a specific separation, as may be known for example from the prior art, cannot be straightforwardly applied to a different separation problem with a sufficient prospect of success. For each separation problem, the ideal elution conditions, in particular the mobile phase used, have to be determined by empirical testing.
  • a mixture of an aqueous solvent and an organic solvent is used as the mobile phase for eluting the RNA.
  • a buffer to be used as the aqueous solvent which has in particular a pH of 6.0-8.0, for example of about 7, for example. 7.0; preferably the buffer is triethylammonium acetate, particularly preferably a 0.02 M to 0.5 M, in particular 0.08 M to 0.12 M, very particularly an about 0.1 M triethylammonium acetate buffer, which, as described above, also acts as a counterion to the RNA in the ion pair method.
  • the organic solvent which is used in the mobile phase is ace- tonitrile, methanol, ethanol, 1 -propanol, 2-propanol and acetone or a mixture thereof, very particularly preferably acetonitrile.
  • the mobile phase is a mixture of 0.1 M triethylammonium acetate, pH 7, and acetonitrile.
  • the mobile phase it has proven particularly favourable for the method according to the invention for the mobile phase to contain 5.0 vol.% to 25.0 vol.% organic solvent, relative to the mobile phase, and for this to be made up to 100 vol.% with the aqueous solvent.
  • the proportion of organic solvent is increased, in particular by at least 10%, more preferably by at least 50% and most preferably by at least 100%, optionally by at least 200%, relative to the initial vol.% in the mobile phase.
  • the proportion of organic solvent in the mobile phase amounts in the course of HPLC separation to 3 to 9, preferably 4 to 7.5, in particular 5.0 vol.%, in each case relative to the mobile phase.
  • the proportion of organic solvent in the mobile phase is increased in the course of HPLC separation from 3 to 9, in particular 5.0 vol.% to up to 20.0 vol.%, in each case relative to the mobile phase. Still more preferably, the method is performed in such a way that the proportion of organic sol- vent in the mobile phase is increased in the course of HPLC separation from 6.5 to 8.5, in particular 7.5 vol.%, to up to 1 7.5 vol.%, in each case relative to the mobile phase.
  • the mobile phase contains 7.5 vol.% to 17.5 vol.% organic solvent, relative to the mo- bile phase, and for this to be made up to 100 vol.% with the aqueous buffered solvent.
  • elution may proceed isocratically or by means of gradient separation.
  • elution of the RNA proceeds with a single eluent or a constant mixture of a plurality of eluents, wherein the solvents described above in detail may be used as eluent.
  • gradient separation is performed.
  • the composition of the eluent is varied by means of a gradient program.
  • the equipment necessary for gradient separation is known to a person skilled in the art.
  • Gradient elution may here proceed either on the low pressure side by mixing chambers or on the high pressure side by further pumps.
  • the proportion of organic solvent, as described above, is increased relative to the aqueous solvent during gradient separation.
  • the above-described agents may here be used as the aqueous solvent and the likewise above-described agents may be used as the organic solvent.
  • the proportion of organic solvent in the mobile phase may be increased in the course of HPLC separation from 5.0 vol.% to 20.0 vol.%, in each case relative to the mobile phase.
  • the proportion of organic solvent in the mobile phase may be increased in the course of HPLC separation from 7.5 vol.% to 1 7.5 vol.%, in particular 9.5 to 14.5 vol.%, in each case relative to the mobile phase.
  • Eluent A 0.1 M triethylammonium acetate, pH 7
  • Eluent B 0.1 M triethylammonium acetate, pH 7, with 25 vol.% acetonitrile
  • Eluent composition start: 62% A and 38% B (1 st to 3rd minute) increase to 58% B (1 .67% increase in B per minute), (3rd-15th minute) 100% B (15th to 20th minute)
  • purified solvent for HPLC Such purified solvents are commercially obtainable. They may additionally also be filtered with a 1 to 5 ⁇ m microfilter, which is generally mounted in the system upstream of the pump. It is additionally favourable for ail the solvents to be de- gassed prior to use, since otherwise gas bubbles occur in most pumps. If air bubbles occur in the solvent, this may interfere not only with separation but also with the continuous monitoring of outflow in the detector.
  • the solvents may be degassed by heating, by vigor- ous stirring with a magnetic stirrer, by brief evacuation, by ultrasonication or by passing a small stream of helium through the solvent storage vessel.
  • the flow rate of the eluent is so selected that good separation of the RNA from the other constituents contained in the sample to be investigated takes place.
  • the eluent flow rate selected for the method according to the invention may amount to from 1 ml/min to several litres per minute (in the case of upscaling), in particular about 1 to 1000 ml/min, more preferably 5 ml to 500 ml/min, even more preferably more than 100 ml/min, depending on the type and scope of the upscaling. This flow rate may be established and regulated by the pump.
  • Detection proceeds favourably with a UV detector at 254 nm, wherein a reference measurement may be made at 600 nm.
  • a reference measurement may be made at 600 nm.
  • any other detection method may alternatively be used, with which the RNA described above in greater detail may be detected in satisfac- tory and reliable manner.
  • RNA-containing eluted solvent quantities For preparative purification of the RNA with the method according to the invention, it is advisable to collect the RNA-containing eluted solvent quantities. In this respect, it is favourable to carry out this collection in such a way that the eluted solvent is collected in individual separated fractions. This may take place for example with a fraction collector. In this way, the high-purity RNA-containing fractions may be separated from other RNA- containing fractions which still contain undesired impurities, albeit in very small quantities. The individual fractions may be collected for example over 1 minute.
  • the method according to the invention is favourably performed under completely denaturing conditions. This may proceed for example in that sample application takes place at a temperature of 4-12°C, the HPLC method otherwise proceeding at a higher temperature, preferably at 70 0 C or more, particularly preferably at 75°C or more, in particular up to 82 0 C, and very particularly preferably at about 78°C. Heating is typically important for de- naturing the RNA, whereby better separation is ultimately achieved. Resolution falls at lower temperatures, i.e. separation of the RNA from the impurities decreases. Sample application may be performed with two methods, stop-flow injection or loop injection. For stop-flow injection a microsyringe is used which is able to withstand the high pressure applied in HPLC.
  • the sample is injected through a septum in an inlet valve either directly onto the column packing or onto a small drop of inert material immediately over the packing.
  • the system may in this case be under elevated pressure, or the pump may be turned off prior to injection, which is then performed when the pressure has fallen to close to the normal value.
  • a loop injector is used to introduce the sample. This consists of a tubular loop, into which the sample is inserted.
  • the stationary phase is then conveyed out of the pump through the loop, whose outlet leads directly into the column.
  • the sample is entrained in this way by the stationary phase into the column, without solvent flow to the pump being interrupted.
  • the present invention further provides the use of a porous reversed phase in the above- described HPLC method according to the invention.
  • the porous reversed phase has a particle size of 8.0 ⁇ m to 50 ⁇ m, in particular 8.0 to 30, still more preferably 8.0 to 25 ⁇ m.
  • the reversed phase may be present in bead form.
  • the method according to the invention may be performed particularly favourably with a porous reversed phase with this particle size and/or in the form of beads, wherein particularly good separation results are obtained.
  • the reversed phase used for the use according to the invention is porous, since with stationary reversed phases, which are not porous, and thus differ completely with regard to particle size from the subject matter of the present invention, as described for example by A. Azarani and K.H. Hecker, excessively high pressures are built up, such that preparative purification of the RNA by means of HPLC is possible only with great difficulty, if at all.
  • the reversed phase has a pore size of 1000 ⁇ to 5000 A, in particular a pore size of 1000 A to 4000 A, or the above-stated preferred values.
  • Particularly preferred pore sizes for the reversed phases are 1000-1200 A and 3500-4500 A.
  • the material for the reversed phase is a polystyrenedivinylbenzene, wherein in particular non-alkylated polystyrenedivinylbenzene may be used.
  • Stationary phases with polystyrenedivinylbenzene are known per se.
  • the per se known polystyrenedivinylbenzenes already used for HPLC methods, which are commercially obtainable, may be used for the method according to the invention.
  • a non-alkylated porous polystyrenedivinylbenzene which is very particularly preferred for the use according to the invention is one which has in particular a particle size of 8.0 + 1.5 ⁇ m, in particular 8.0 ⁇ 0.5 ⁇ m, and a pore size of 1000- or 4000 A.
  • a particle size of 8.0 + 1.5 ⁇ m in particular 8.0 ⁇ 0.5 ⁇ m
  • a pore size of 1000- or 4000 A With this material for the reversed phase, the advantages described below may be achieved in a particularly favourable manner.
  • this stationary phase described in greater detail above is located in a column.
  • V2A steel is conventionally used as the material for the column, but other materials may also be used for the column provided they are suitable for the conditions prevailing during HPLC recovery.
  • the column is straight. It is favourable for the HPLC column to have a length of 5 cm to 100 cm and a diameter of 4 mm to 25 mm.
  • Columns used for the method according to the invention may in particular have the following dimensions: 50 mm long and 7.5 mm in diameter or 50 mm long and 4.6 mm in diameter, or 50 mm long and 10 mm in diameter or any other dimension with regard to length and diameter, which is suitable for preparative purification of RNA by means of HPLC, even lengths of several metres and also larger diameters being feasible in the case of upscaling.
  • the dimensions are here geared towards what is technically possible with liquid chromatography. Further conditions for the use according to the invention have already been described above in connection with the method according to the invention, such that reference is made thereto to avoid unnecessary repetition.
  • RNA samples in particular the preferred embodiments described in detail hereinafter, exhibits a series of advantages: in the context of the use according to the invention, substantially larger quantities of RNA may be separated than is possible with the methods known from the prior art.
  • the use according to the invention enables these higher quantities to be obtained with a high degree of purity. Separation of degraded fragments or of relatively long fragments or of fragments which have arisen as a result of premature termination of transcription is possible.
  • good separation of further impurities present in RNA samples such as enzymes, for example RNases or polymerases, and nucleotides is possible. These relatively large quantities of RNA may be recovered within minutes, even from samples having a very high level of contamination with impurities.
  • RNA recovery proceeds reliably, i.e. high purity RNA is recovered with great reliability.
  • the use according to the invention enables high resolution to be achieved.
  • the use according to the invention may easily be operated automatically. It is thus most suitable for routine purification of RNA on a preparative scale.
  • RNA is stable under the conditions of the HPLC separation method, i.e. degradation into RNA fragments and thus the occurrence of new impurities or a reduc- tion in the yield of pure RNA during HPLC purification is avoided.
  • Figure 1 shows a chromatogram of a separation of 10 ⁇ g of luciferase-mRNA (analytical);
  • Figure 2 shows a chromatogram of a separation of 1 .5 ⁇ g of luciferase-mRNA (preparative);
  • Figure 3 shows the chromatogram of Figure 2, the collected fractions being highlighted in grey
  • Figure 4 shows verification of the purity of the collected mRNA fractions on an agarose gel
  • Figure 5 shows a chromatogram of a separation of 200 ⁇ g of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 1000 A;
  • Figure 6 shows a chromatogram of a separation of 100 ⁇ g of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 4000 A;
  • Figure 7 shows a chromatogram of a separation of 250 ⁇ g of a 2 kb and of a 4 kb RNA fragment with a stationary matrix, which has a pore size of 4000 A,
  • Figure 8 is a bar chart which demonstrates the improvement in the expression of luciferase from RNA purified with the method according to the invention.
  • Figure 9 shows the sequence of the wild-type of luciferase, specifically the mRNA se- quence, which was used for the exemplary embodiments.
  • Figure 10 shows a chromatogram of a separation of 3 mg of a Flic(GC) RNA preparation (FC-9050) with a stationary matrix, which has a pore size of 4000 A
  • Figure 11 shows a chromatogram of a separation of 1 ,5 mg of a FOLHI (GC) RNA preparation (FO-9334) with a stationary matrix, which has a pore size of 4000 A
  • GC FOLHI
  • Figure 12 shows a chromatogram of a separation of 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849) with a stationary matrix, which has a pore size of 4000 A,
  • Figure 13 shows a chromatogram of a separation of 1 ,5 mg of a PSCA(GC) RNA preparation (PS-8845) with a stationary matrix, which has a pore size of 4000 A,
  • Figure 14 shows a chromatogram of a separation of 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848) with a stationary matrix, which has a pore size of 4000 A,
  • Figure 15 shows a chromatogram of a separation of a mixture of a 2 kb RNA (200 ⁇ g) and a
  • Figure 16 shows an agarose gel analysis corresponding to the chromatogram according to
  • Figure 15 which shows a separation of a mixture of a 2 kb RNA (200 ⁇ g) and a
  • Example 1 Purification of 1 .5 mg of luciferase mRNA by means of the HPLC method according to the invention
  • Luciferase mRNA having a size of 1825 base pairs was used for separation.
  • a porous, non- alkylated polystyrene/divinylbenzene (polystyrenedivinylbenzene) matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 ⁇ m and a pore size of 4000 A.
  • the column used was 5.0 cm long and had a diameter of 7.5 mm.
  • the sampler temperature was 12°C
  • the temperature for the HPLC separation, in particular of the separation column was 78°C, i.e. operations were carried out under completely denaturing conditions.
  • Eluent B 0.1 M triethylammonium acetate, pH 7, with 25 vol.% acetonitrile Eluent composition: start: 62% A and 38% B (1 st to 3rd minute) increase to 58% B (1 .67% increase in B per minute), (3rd-15th minute) 100% B (15th to 20th minute)
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
  • the purpose of purifying the luciferase mRNA on a preparative scale is to remove these undesired impurities.
  • Figures 2 and 3 show the corresponding chromatograms for the separation of luciferase mRNA on a preparative scale (1 .5 mg), wherein the gradient program is shown in broken lines in Figure 2.
  • the collected fractions 1 to 10 are highlighted in grey in Figures 2 and 3; the fractions were collected over a period of 1 minute.
  • This exemplary embodiment shows that, using the method according to the invention, it is possible to purify mRNA on a preparative scale by means of HPLC.
  • Example 2 Separation of 200 ⁇ g of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 1000 A;
  • RNA fragments 200 ⁇ g of a 2 kb and 4 kb RNA fragment were used for separation.
  • a porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 ⁇ m and a pore size of 1000 ⁇ .
  • the column used was 2.5 cm long and had a diameter of 4.6 mm.
  • the sampler temperature was 12°C
  • the temperature for the HPLC separation, in particular of the separa- tion column was 78°C, i.e. operations were carried out under completely denaturing conditions.
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
  • Figure 5 shows the corresponding chromatograms for this separation on a preparative scale (200 ⁇ g), wherein the gradient program is shown in broken lines in Figure 5.
  • the collected fractions are highlighted in grey in Figure 5; the fractions were collected over a period of 1 minute.
  • a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
  • Example 3 Separation of 100 ⁇ g of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 4000 A;
  • RNA fragment 100 ⁇ g of a 2 kb and 4 kb RNA fragment were used for separation.
  • a porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 ⁇ m and a pore size of 4000 A.
  • the column used was 2.5 cm long and had a diameter of 4.6 mm.
  • the sampler temperature was 12°C
  • the temperature for the HPLC separation, in particular of the separa- tion column was 78°C, i.e. operations were carried out under completely denaturing conditions.
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
  • Figure 6 shows the corresponding chromatograms for this separation on a preparative scale (100 ⁇ g), wherein the gradient program is shown in broken lines in Figure 6. The fractions collected are highlighted in grey in Figure 6. As a result, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
  • Example 4 Separation of 250 ⁇ g of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 4000 A;
  • RNA fragments 250 ⁇ g of a 2 kb and 4 kb RNA fragment were used for separation.
  • a porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 ⁇ m and a pore size of 4000 A.
  • the column used was 2.5 cm long and had a diameter of 4.6 mm.
  • the sampler temperature was 12 0 C
  • the temperature for the HPLC separation, in particular of the separation column was 78 0 C, i.e. operations were carried out under completely denaturing con- ditions.
  • Eluent B 0.1 M triethylammonium acetate/25% acetonitri Ie Eluent composition: starting level: 62% A and 38% B (1 st-3rd min) separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd-
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/mi n.
  • Figure 7 shows the corresponding chromatograms for this separation on a preparative scale (250 ⁇ g), wherein the gradient program is shown in broken lines in Figure 7. The fractions collected are highlighted in grey in Figure 7. As a result, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
  • Example 5 Separation of 3 mg of a 1,7 kb RNA (FliC(GC)-preparation, FC9050) with a stationary phase, which has a pore size of 4000 A;
  • RNA 1 ,7 kb RNA
  • FC9050 1 ,7 kb RNA
  • a porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 ⁇ m and a pore size of 4000 A.
  • the column used was 2.5 cm long and had a diameter of 4.6 mm.
  • the sampler temperature was 12°C
  • the temperature for the HPLC separation, in particular of the separation column was 78°C, i.e. operations were carried out under completely denaturing conditions.
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
  • Figure 10 shows the corresponding chromatograms for this separation on a preparative scale (3 mg), wherein the gradient program is shown in broken lines in Figure 10. The fractions collected are highlighted in grey in Figure 10. As can be seen, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
  • Example 6 Separation of 1,5 mg of a FOLHI (GC) RNA preparation (FO-9334), 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849), 1,5 mg of a PSCA(GC) RNA preparation (PS-8845) or 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848) with a stationary phase, which has a pore size of 4000 A;
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
  • Figures 1 1 , 12, 13 and 14 shows the corresponding chromatograms for this separation of 1 ,5 mg of a FOLHI (GC) RNA preparation (FO-9334), 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849), 1 ,5 mg of a PSCA(GC) RNA preparation (PS-8845) or 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848), respectively on a preparative scale (1 ,5 mg), wherein the gradient program is shown in broken lines in these Figures.
  • the fractions collected are high- lighted in grey in Figure 1 1 , 12, 13 and 14. As can be seen, a separation of the RNAs is possible in each case, leading to a highly pure fraction of RNA in each case in preparative scale.
  • Example 7 Separation of a mixture of 200 ⁇ g of a 2 kb RNA and 200 ⁇ g of a 4kb RNA on a non-porous stationary polystyrenedivinylbenzene matrix (Comparison Example)
  • a mixture of 200 ⁇ g of a 2 kb RNA and 200 ⁇ g of a 4kb RNA were used for separation.
  • the columns used were packed with either a 1000 ⁇ non-alkylated porous polystyrenedivinyl- benzene or a non-porous stationary polystyrenedivinylbenzene matrix.
  • the column used for the 1000 A non-alkylated porous polystyrenedivinylbenzene was 2.5 cm long and had a diameter of 4.6 mm.
  • the column used for the non-alkylated non-porous polystyrenedivinylbenzene was 2.5 cm long and had a diameter of 4.0 mm.
  • the sampler temperature was 12°C
  • the temperature for the HPLC separation, in particular of the separation column was 78°C, i.e. operations were carried out under completely denaturing conditions.
  • Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
  • Figures 15 shows the corresponding chromatogram for this separation on a preparative scale (each 200 ⁇ g), wherein the gradient program is shown in broken lines in Figure 15. The fractions collected are highlighted in grey in Figure 15. As can be seen, a separation of both RNAs is not possible using a common non-alkylated porous polystyrenedivinylbenzene as a matrix.
  • Figure 16 shows the agarose gel corresponding to the chromatogram according to Figure 15.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Saccharide Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Steroid Compounds (AREA)

Abstract

The application describes a method for the preparative purification of RNA, which method is distinguished in that the RNA is purified by means of HPLC using a porous reversed phase as the stationary phase. The use of the porous reversed phase in this HPLC method is also described.

Description

Method for purifying RNA on a preparative scale by means of HPLC
The present invention relates to a method for purifying RNA on a preparative scale by means of HPLC and to the use of a porous reversed phase as stationary phase in this method.
HPLC (abbreviation for "High Performance (High Pressure) Liquid Chromatography") is an established method of separating mixtures of substances, which is widely used in biochemistry, analytical chemistry and clinical chemistry.
An HPLC apparatus consists in the simplest case of a pump with eluent reservoir containing the mobile phase, a sample application system, a separation column containing the station- ary phase, and the detector. In addition, a fraction collector may also be provided, with which the individual fractions may be separately collected after separation and are thus available for further applications.
RNA analysis using ion pair reversed phase HPLC is known from A. Azarani and K. H. Hecker (Nucleic Acids Research, vol. 29, no. 2 e7). The stationary phase here consists of a non-porous alkylated polystyrenedivinylbenzene matrix. Elution proceeds with a buffer system of two eluents. Buffer A consists of an aqueous solution of 0.1 M triethylammonium acetate (TEAA), pH 7.0, and buffer B consists of an aqueous solution of 0.1 M TEAA, pH 7.0, with 25% acetonitrile. Elution was performed using the following three gradient sys- terns: 38-40% B over 1 minute, to 60% B over 15 minutes, to 66% B over 6 minutes, to 70% B over 0.5 minutes, to 100% B over 0.5 minute, holding at 100% B for 1 minute, to 38% over 1 minute, holding at 38% B for 2 minutes. Alternatively, the following gradient program was used: 38-60% over 30 minutes, to 100% B over 2 minutes, to 38% B over 3 minutes. The following was used as a third gradient program: 38-40% B over 1 minute, to 60% B over 3 minutes, to 100% B over 1 minute, holding at 100% B over 6 minutes, to 38% B over 1 minute, holding at 38% B over 1 minute.
This HPLC method thus involved using relatively complicated and costly gradient programs. In addition, only analytical quantities of the RNA up to at most 1000 ng (1 μg or 0.001 mg) can be separated and analysed with this method.
The object of the present invention is here to improve a method of this type to the effect that it no longer exhibits the disadvantages of the prior art.
This is achieved according to the invention by a method for purifying RNA on a preparative scale, which is distinguished in that the RNA is purified by means of HPLC using a porous reversed phase as stationary phase. In the method according to the invention a significant factor is therefore that a porous reversed phase is used.
In the context of the present invention, the term "by means of HPLC" shall include various HPLC methods as well as low or normal pressure liquid chromatography methods, which may be used to carry out the inventive method. These may include reversed phase HPLC (RP-HPLC), chromatography, size exclusion chromatography, gel filtration, affinity chroma- tography, hydrophobic interaction chromatography or ion pair chromatography, whereby reversed phase HPLC is preferred. Without going into details, reversed phase HPLC consists of a non-polar stationary phase and a moderately polar mobile phase. One common stationary phase is e.g. a silica which has been treated with e.g. RMe2SiCI, where R is a straight chain alkyl group such as C18H37 or C8H17. The retention time is therefore longer for mole- cules which are more non-polar in nature, allowing polar molecules to elute more readily. Retention time is increased by the addition of polar solvent to the mobile phase and decreased by the addition of more hydrophobic solvent.
However, the various techniques mentioned above operate on the principle of hydrophobic interactions which result from repulsive forces between a relatively polar solvent, the relatively non-polar analyte, and the non-polar stationary phase (reversed phase principle). The driving force in the binding of the analyte (the RNA molecule) to the stationary phase is the decrease in the area of the non-polar segment of the analyte molecule exposed to the sol- vent. This hydrophobic effect is dominated by the decrease in free energy from entropy associated with the minimization of the ordered molecule-polar solvent interface. The hydrophobic effect is decreased by adding more non-polar solvent into the mobile phase. This shifts the partition coefficient such that the analyte spends some portion of time moving down the column in the mobile phase, eventually eluting from the column.
The characteristics of the specific RNA molecule as an analyte may play an important role in its retention characteristics. In general, an analyte having more apolar functional groups results in a longer retention time because it increases the molecule's hydrophobicity. Very large molecules, however, can result in incomplete interaction between the large analyte surface and the alkyl chain. Retention time increases with hydrophobic surface area which is roughly inversely proportional to solute size. Branched chain compounds elute more rapidly than their corresponding isomers because the overall surface area is decreased.
Aside from mobile phase hydrophobicity, other mobile phase modifiers can - according to the invention - affect analyte (RNA molecule) retention. For example, the addition of inorganic salts may be considered to cause a linear increase in the surface tension of aqueous solutions, and because the entropy of the analyte-solvent interface is controlled by surface tension, the addition of salts tend to increase the retention time. Another important compo- nent, which may be used according to the invention is pH, since this can change the hydrophobicity of the analyte. For this reason, a buffering agent, such as sodium phosphate or other common buffering agents, may be used to control the pH. An organic acid, such as formic acid or most commonly trifluoroacetic acid, may be added to mobile phase. The above modifications may serve multiple purposes: They control pH, neutralize the charge on any residual exposed material on the stationary phase and act as ion pairing agents to neutralize charge on the analyte. The effect varies depending on use but generally improve the chromatography.
For the purposes of the present invention, the term "purification" is understood to mean that the desired RNA in a sample is separated and/or isolated from the impurities present therein. Thus, after HPLC purification the RNA is present in a purer form than in the originally introduced RNA-containing sample prior to HPLC purification. Undesired constituents of RNA- containing samples which therefore need to be separated may in particular be degraded fragments or fragments which have arisen as a result of premature termination of transcription, or also excessively long transcripts if plasmids are not completely linearised. In addition, impurities, such as enzymes, for example RNases and polymerases, and nucleotides may be separated.
Using the method according to the invention, RNA is purified which has a higher purity after purification than the starting material. It is desirable in this respect for the degree of purity to be as close as possible to 100%. A degree of purity of more than 70%, in particular 80%, very particularly 90% and most favourably 99% or more may be achieved in this way.
The method according to the invention results in preparative RNA purification. This differs from an analytical HPLC method, which is described in the above prior art (Azarani & Hecker). In an analytical HPLC method, a distinctly smaller quantity is introduced and separated than in a preparative HPLC method. In the method discussed in the prior art, quanti- ties of 8 ng to 1000 ng were introduced. In contrast, a preparative HPLC method should be understood to mean an HPLC method in which relatively large quantities of RNA are purified. Such relatively large quantities are for example quantities of 0.5 mg or more, in particular 1 .0 mg to 1000 mg or more, very particularly approximately 1.5 mg or more, upscal- ing even to the kg range being possible. The above statements are therefore to be under- stood to mean that these quantities relate to a single HPLC run. If a plurality of HPLC runs are performed, the quantity increases in direct proportion to the number of HPLC runs.
The method according to the invention, in particular the preferred embodiments described in detail hereinafter, exhibits a series of advantages: using the method according to the in- vention substantially larger quantities of RNA may be separated than is possible with the method known from the prior art. Using the method according to the invention, these relatively high quantities are obtained with a high degree of purity. Separation of degraded fragments or excessively long fragments or of fragments which have arisen as a result of premature termination of transcription is possible. In addition, good separation of further impurities present in RNA samples, such as enzymes, for example RNases and polymerases, and nucleotides is possible. These relatively large quantities of RNA may be recovered within minutes, even from samples having a very high level of contamination with impurities. RNA recovery proceeds reliably, i.e. high purity RNA is recovered with great reliability. In the method according to the invention, a high level of resolution may be achieved. The method according to the invention may additionally easily be operated automatically. It is thus most suitable for routine purification of RNA on a preparative scale. RNA is stable under the conditions of the method according to the invention, i.e. degradation during HPLC purification into RNA fragments and thus the occurrence of new impurities and a reduction in the yield of desired RNA during HPLC purification is avoided. The method according to the invention may thus be performed in a simple, rapid, inexpensive manner, accurately and with reliable results. Isolation of RNA after HPLC separation may proceed simply with a fraction collector, i.e. the recovery of RNA may be achieved very simply, it being possible for direct further processing of the RNA likewise to take place. Detection may finally be performed very sensitively.
The "RNA" which is to be purified with the method according to the invention is ribonucleic acid of any type. The RNA is particularly preferably selected from tRNA, rRNA, mRNA or whole-cell RNA. In addition, the RNA may also comprise aptamers and ribozymes. The RNA to be isolated may be single-stranded or double-stranded. Single-stranded RNA may optionally form secondary structures by refolding, the RNA to be separated typically being single-stranded. The RNA may be unlabelled or also labelled (with a fluorescent label or a radiolabel or an antibody epitope). One example of a labelled RNA is digoxigenin-labelled β-actin RNA.
The RNA to be purified may be native or non-native RNA. Native RNA is preferably used, which is prepared using a for example cellular or in vitro expression system. The RNA is then isolated and the isolate purified using the method according to the invention. Option- ally, chemically synthesised RNA may also be purified using the method according to the invention. In any event, the RNA may also contain non-native nucleotides, wherein the chemical modifications may be present in the backbone of the RNA, for example phosphor- thioate, in the structure of the nucleotide base, for example hypoxanthine, dihydrouracil, pseudouracil, 2-thiouracil, 4-thiouracil, 5-aminouracil, 5-(C1 -C6)-alkyluracil, 5-(C2-C6)- alkenyluracil, 5-(C2-C6)-alkynyluracil, 5-(hydroxymethyl)uracil, 5-chlorouracil, 5- fluorouracil, 5-bromouracil, 5-hydroxycytosine, 5-(C1 -C6)-alkylcytosine, 5-(C2-C6)- alkenylcytosine, 5-(C2-C6)-alkynylcytosine, 5-chlorocytosine, 5-fluorocytosine, 5- bromocytosine, N<2>-dimethylguanine, 2,4-diaminopurine, 8-azapurine, a substituted 7- deazapurine, preferably 7-deaza-7-substituted and/or 7-deaza-8-substituted purine, 5- hydroxymethylcytosine, N4-alkylcytosine, for example N4-ethylcytosine, 5- hydroxydeoxycytidine, 5-hydroxymethyldeoxycytidine, N4-alkyldeoxycytidine, for example N4-ethyldeoxycytidine, 6-thiodeoxyguanosine, and deoxyribonucleotides of nitropyrrole, C5-propynylpyrimidines, and diaminopurine, for example 2,6-diaminopurine, inosine, 5- methylcytosine, 2-aminopurine, 2-amino-6-chloropurine, or also in the sugar residue. Further examples of modified nucleotides are 2-amino-6-chloropurine-riboside-5'-triphosphate, 2-aminoadenosine-5 '-triphosphate, 2-thiocytidine-5 '-triphosphate, 2-thiouridine-5'- triphosphate, 4-thiouridine-5'-triphosphate, 5-aminoallylcytidine-5'-triphosphate, 5- aminoallyluridine-5'-triphosphate, 5-bromocytidine-5'-triphosphate, 5-bromouridine-5'- triphosphate, 5-iodocytidine-5 '-triphosphate, 5-iodouridine-5'-triphosphate, 5- methylcytidine-5 '-triphosphate, 5-methyluridine-5 '-triphosphate, 6-azacytidine-5'- triphosphate, 6-azauridine-5 '-triphosphate, 6-chloropurine-riboside-5'-triphosphate, 7- deazaadenosine-5 '-triphosphate, 7-deazaguanosine-5 '-triphosphate, 8-azaadenosine-5'- triphosphate, 8-azidoadenosine-5'-triphosphate, benzimidazole-riboside-5'-triphosphate, N1 -methyladenosine-5'-triphosphate, N1 -methylguanosine-5'-triphosphate, N6- methyladenosine-5 '-triphosphate, O6-methylguanosine-5 '-triphosphate, pseudouridine-5'- triphosphate, puromycine-5 '-triphosphate, xanthosine-5 '-triphosphate.
In a preferred embodiment of the method according to the invention, the RNA to be separated has a size of up to about 15000 nucleotides (as single stranded RNA molecule) or base pairs (as double stranded RNA molecule), in particular 100 to 10000, more preferably 500 to 10000 nucleotides or base pairs, even more preferably 800 to 5000 nucleotides or base pairs and even more preferably 800 to 2000 nucleotides or base pairs. For this size of RNA, it has proved possible to achieve very good results with regard to purification of the RNA, since the method according to the invention is particularly well suited to RNA of this size. Optionally, however, smaller RNA fragments, for example with a length of 30-1000, 50- 1000 or 100-1000 or 20-200, 20-100, 20-50 or 20-30 nucleotides may also be separated in this way.
If the RNA to be separated is mRNA, this will preferably code for proteins, in particular those which have an antigen character, and for example all recombinantly produced or naturally occurring proteins, which are known to a person skilled in the art from the prior art and are used for therapeutic, diagnostic or research purposes. In particular, the antigens are then tumour antigens or antigens of pathogens, for example viral, bacterial or protozoal organisms.
Without being limited thereto, such proteins include inter alia growth hormones or growth factors, for example for promoting growth in a (transgenic) living organism such as for example TGF and the IGFs ("insulin-like growth factors"), proteins which influence metabolism and/or haematopoiesis, such as for example (alpha-l )-anti-trypsin, LDL receptor, erythropoietin (EPO), insulin, GAT A-1 , etc., or proteins such as for example factors VIII and Xl of the blood clotting system, etc.. Such proteins further include enzymes, such as for example
-galactosidase (lacZ), DNA restriction enzymes (for example EcoRI, Hindlll, etc.), Iy- sozymes, etc., or proteases, such as for example papain, bromelain, keratinases, trypsin, chymotrypsin, pepsin, rennin (chymosin), suizyme, nortase, etc., and protease inhibitors (for example for the treatment of HIV infections), such as for example protease inhibitors se- lected from a group consisting of AG1 776, amprenavir (141W94 or VX-478), atazanavir (BMS-232632), cathepsin S protease inhibitor, D1927, D9120, fosamprenavir (GW-433908 or VX-175), GS 9005, GW640385 (VX-385), HCV protease inhibitor, indinavir (MK-639), L- 756 423, Levoprin-ZG, lopinavir (ABT-378), lopinavir/ritonavir (LPV ABT-378/r), MK-944A, mozenavir (DMP450), nelfinavir (AG-1343), NNRTI, P-1946, PL-100, prinomastat, ritonavir (ABT-538), RO033-4649, TMC1 14, saquinavir (Ro-31 -8959), NRTI, tipranavir (PNU- 140690), TLK 19781 , TMC-1 14, vertex 385, VX-950. The proteins coded from the RNA separated according to the invention may likewise comprise proteins which stimulate or inhibit cell signal transmission, for example cytokines, etc.. Such proteins therefore also comprise for example cytokines of class I of the cytokine family, which comprise 4 posi- tionally conserved cysteine residues (CCCC) and one conserved sequence motif Trp-Ser-X- Trp-Ser (WSXWS), wherein X represents a non-conserved amino acid. Cytokines of class I of the cytokine family comprise the GM-CSF subfamily, for example IL-3, IL-5, GM-CSF, the IL-6 subfamily, for example IL-6, IL-1 1 , IL-12, or the IL-2 subfamily, for example IL-2, IL-4, IL-7, IL-9, IL-15, etc., or the cytokines IL-I , IL- 1 , IL-10 etc.. Likewise, such proteins may also comprise cytokines of the class Il of the cytokine family (interferon receptor family), which likewise comprise 4 positionally conserved cysteine residues (CCCC), but no conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS). Cytokines of class Il of the cytokine family include for example IFN-alpha, IFN-beta, IFN-gamma, etc.. The proteins coded by the separated RNA may furthermore also include cytokines of the tumour necrosis family including for example TNF-alpha, TNF-beta, CD40 ligand, Fas ligand, etc., or cytokines of the chemokine family, in particular interferons, interleukins, colony-stimulating factors and tumour necrosis factors, and interact with G protein, for example IL-8, MIP-1 , RANTES, CCR5, CXR4, etc.. Such proteins may also be selected from apoptosis factors or apoptosis- related or -linked proteins, including AIF, Apaf, for example Apaf-1 , Apaf-2, Apaf-3, or APO-2 (L), APO-3 (L), apopain, Bad, Bak, Bax, Bcl-2, Bcl-xL, Bcl-xs, bik, CAD, calpain, cas- pases for example caspase-1 , caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, cas- pase-7, caspase-8, caspase-9, caspase-10, caspase-1 1 , ced-3, ced-9, c-Jun, c-Myc, crm A, cytochrome C, CdR1, DcRI , DD, DED, DISC, DNA-PKc5, DR3, DR4, DR5, FADD/MORT-1 , FAK, Fas (Fas ligand CD95/fas (receptor)), FLICE/MACH, FLIP, fodrin, fos, G-actin, Gas-2, gelsolin, granzymes A/B, ICAD, ICE, JNK, lamin A/B, MAP, MCL-1 , Mdm-2, MEKK-1 , MORT-1 , NEDD, NF-B, NuMa, p53, PAK-2, PARP, perforin, PITSLRE, PKC, pRb, presenilin, prlCE, RAIDD, Ras, RIP, sphingomyelinase, thymidine kinase from Herpes simplex, TRADD, TRAF2, TRAIL, TRAIL-R1 , TRAIL-R2, TRAIL-R3, transglutaminase, etc.. The proteins coded by RNA separated according to the invention may also be selected from antigens, for example from tumour-specific surface antigens (TSSA), for example 5T4alpha5 betai integrin, 707-AP, AFP, ART-4, B7H4, BAGE, Bcr-abl, MN/C IX-antigen, CAI 25, CAMEL, CAP-1 , CASP-8, beta-catenin/m, CD4, CD19, CD20, CD22, CD25, CDC27/m, CD30, CD33, CD52, CD56, CD80, CDK4/m, CEA, CT, Cyp-B, DAM, EGFR, ErbB3, ELF2M, EMMPRIN, EpCam, ETV6-AML1 , G250, GAGE, GnT-V, Gp100, HAGE, HER-2/new, HLA-A*0201 - R170I, HPV-E7, HSP70-2M, HAST-2, hTERT (or hTRT), iCE, IGF-1 R, IL-2R, IL-5, KIAA0205, LAGE, LDLR/FUT, MAGE, MART-1/melan-A, MART-2/Ski, MC1 R, myosin/m, MUC1 , MUM- 1 , -2, -3, NA88-A, PAP, proteinase-3, p190 minor bcr-abl, Pml/RAR, PRAME, PSA, PSM, PSMA, RAGE, RU1 or RU2, SAGE, SART-1 or SART-3, survivin, TEL/AML1 , TGF, TPI/m, TRP-1 , TRP-2, TRP-2/INT2, VEGF and WTl , or from sequences, such as for example NY- Eso-1 or NY-Eso-B. Proteins which may be coded by the RNA separated according to the invention additionally also include proteins or protein sequences which exhibit sequence identity of at least 80% or 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99% to one of the above-described proteins.
The mRNA to be separated may exhibit the following modifications relative to a corresponding wild-type mRNA, which may either be present as alternatives or in combination. On the one hand, the G/C content of the region of the modified mRNA coding for a peptide or polypeptide may be greater than the G/C content of the coding region of the wild-type mRNA coding for the peptide or polypeptide, the coded amino acid sequence being unchanged relative to the wild-type. This modification is based on the fact that the sequence order of the mRNA domain to be translated is essential for efficient mRNA translation. In this respect, the composition and sequence of the various nucleotides has an important part to play. In particular, sequences with an elevated G(guanosine)/C(cytosine) content are more stable than sequences with an elevated A(adenosine)/U(uracil) content. Thus, according to the invention, while retaining the translated amino acid sequence, the codons are varied relative to the wild-type mRNA in such a manner that they have a greater content of G/C nucleotides. Since several codons code for one and the same amino acid (degeneration of the genetic code), it is possible to determine the codons which are most favourable for stability (alternative codon usage). On the other hand it is also possible to provide a translation-optimised RNA with the method according to the invention.
In the method according to the invention, a reversed phase is used as the stationary phase for HPLC purification. For chromatography with reversed phases, a nonpolar compound serves as the stationary phases and a polar solvent, such as mixtures of water, which is generally used in the form of buffers, with e.g. acetonitrile and/or methanol, serves as the mo- bile phase for elution.
The material packed on the column, which is used as stationary phase, may be provided in bead form or as a polymerized "block", i.e. a block which fills a substantial part of the chromatography column. Irrespective of its precise nature, the polymeric stationary phase is porous in its nature, which means that the beads or the block are characterized by pores.
In a preferred embodiment of the method according to the invention, the porous reversed phase material is provided with a particle size of 8.0 μm to 50 μm, in particular 8.0 to 30, still more preferably 8.0 to 25 μm. The reversed phase material may be present in the form of small spheres. The method according to the invention may be performed particularly favourably with a porous reversed phase with this particle size, optionally in bead form, wherein particularly good separation results are obtained. The reversed phase used in the method according to the invention is porous and has specific particle sizes. With stationary reversed phases which are not porous and thus differ completely with regard to particle size from the subject matter of the present invention as described for example by A. Azarani and K.H. Hecker, on the other hand, excessively high pressures are built up, such that preparative purification of the RNA is possible only with great difficulty, if at all.
In a preferred embodiment of the method according to the invention, the reversed phase has a pore size of 1000 Λ to 5000 A, in particular a pore size of 1000 A to 4000 A, more pref- erably 1500 A to 4000 A, 2000 A to 4000 A or 2500 A to 4000 A. Particularly preferred pore sizes for the reversed phases are 1000 A to 2000 A, more preferably 1000 A to 1500 A and most preferably 1000 A to 1200 A or 3500-4500 A. With a reversed phase having these pore sizes, particularly good results are achieved with regard to purification of the RNA using the method according to the invention, in particular the elevated pressures built up in the method according to A. Azarani and K.H. Hecker are thus avoided, whereby preparative separation is made possible in a particularly favourable manner. At pore sizes of below 1000 A separation of RNA molecules becomes poorer.
A pore size of 1000 A to 5000 A, in particular a pore size of 1000 A to 4000 A, more pref- erably 1500 A to 4000 A, 2000 A to 4000 A or 2500 A to 4000 A may be suitable to separate a RNA from other components of a mixture, the RNA having a size as mentioned above of up to about 15000 nucleotides (as single stranded RNA molecule) or base pairs (as double stranded RNA molecule), in particular 100 to 10000, more preferably 500 to 10000 nucleotides or base pairs, even more preferably 800 to 5000 nucleotides or base pairs and even more preferably 800 to 2000 nucleotides or base pairs. However, the pore size of the reversed phase may also be selected in dependence of the size of the RNA to be separated, i.e. a larger pore size may be selected, if larger RNA molecules are to be separated and smaller pore sizes may be selected, if smaller RNA molecules may be selected. This is due to the effect, that the retention of the RNA molecules and the separation not only depends on the interaction of the (reversed) phase but also on the possibility of molecules to get inside the pores of the matrix and thus provide a further retention effect. Without being limited thereto, e.g. a pore size for the reversed phase of about 2000 A to about 5000 A, more preferably of about 2500 to about 4000, most preferably of about 3500 to about 4500 A, may thus be used to separate larger RNA molecules, e.g. RNA molecules of 100 to 10000, more preferably 500 to 10000 nucleotides or base pairs, even more preferably 800 to 5000 nucleotides or base pairs and even more preferably 800 to 2000 nucleotides or base pairs. Alternatively, without being limited thereto, a pore size of for the reversed phases of about 1000 Λ to about 2500 A, more preferably of about 1000 A to about 2000 A, and most preferably of about 1000 A to 1200 A may be used to separate smaller RNA molecules, e.g. RNA molecules of about 30-1000, 50-1000 or 100-1000 or 20-200, 20-100, 20-50 or 20- 30 nucleotides may also be separated in this way.
In general, any material known to be used as reverse phase stationary phase, in particular any polymeric material may be used for the inventive method, if that material can be provided in porous form. The stationary phase may be composed of organic and/or inorganic material. Examples for polymers to be used for the present invention are (non-alky lated) polystyrenes, (non-alkylated) polystyrenedivinylbenzenes, silica gel, silica gel modified with non-polar residues, particularly silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, silica gel modified with phenyl ic residues, polymethacrylates, etc.. or other materials suitable e.g. for gelchroma- tographie or other chromatographic methods as mentioned above, such as dextran, including e.g. Sephadex® and Sephacryl® materials, agarose, dextran/agarose mixtures, poly- acrylamide, etc..
In a particularly preferred embodiment, the material for the reversed phase is a porous polystyrene polymer, a (non-alkylated) (porous) polystyrenedivinylbenzene polymer, porous silica gel, porous silica gel modified with non-polar residues, particularly porous silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, porous silica gel modified with phenylic residues, porous polymethacrylates, wherein in particular a porous polystyrene polymer or a non-alkylated (porous) polystyrenedivinylbenzene may be used. Stationary phases with polystyrenedivinylbenzene are known per se. The per se known polystyrenedi vinyl-benzenes already used for HPLC methods, which are commercially obtainable, may be used for the method according to the invention. A non-alkylated porous polystyrenedivinylbenzene which is very particularly preferred for the method according to the invention is one which, without being limited thereto, may have in particular a particle size of 8.0 ± 1.5 μm, in particular 8.0 ± 0.5 μm, and a pore size of 1000- 1500 A, in particular 1000-1200 A or 3500-4500 A. With this material for the re- versed phases, the above-described advantages of the method according to the invention may be achieved in a particularly favourable manner.
This stationary phase described in greater detail above is conventionally located in a column. V2A steel is conventionally used as the material for the column, but other materials may also be used for the column provided they are suitable for the conditions prevailing during HPLC. Conventionally the column is straight. It is favourable for the HPLC column to have a length of 5 cm to 100 cm and a diameter of 4 mm to 25 mm. Columns used for the method according to the invention may in particular have the following dimensions: 50 mm long and 7.5 mm in diameter or 50 mm long and 4.6 mm in diameter, or 50 mm long and 10 mm in diameter or any other dimension with regard to length and diameter, which is suitable for preparative recovery of RNA, even lengths of several metres and also larger diameters being feasible in the case of upscaling. The dimensions are here geared towards what is technically possible with liquid chromatography.
In a preferred embodiment, the method according to the invention is performed as an ion pair method, wherein an ion with a lipophilic residue and positive charge is added to the mobile phase as counterion for the negatively charged RNA. In this way, an ion pair with lipophilic character is obtained, which is slowed down by the nonpolar stationary phase of the reversed phase system. In practice, the precise conditions for the ion pair method have to be empirically worked out for each specific separation problem. The size of the counterion, its concentration and the pH value of the solution contribute greatly to the separation result. This means that the empirically established conditions of an ion pair method cannot be straightforwardly applied to a different separation problem. If, for example, an ion pair method is known from the prior art, this does not mean that the same conditions may inevi- tably also be applied to the separation problem underlying the present invention, even if ultimately the same or similar conditions may in fact prove to be favourable. Alkylammo- nium salts, such as triethylammonium acetate, and/or tetraalkylammonium compounds, such as tetrabutylammonium, are favourably added in the method according to the inven- tion. Preferably, 0.1 M triethylammonium acetate is added and the pH value is adjusted to about 7. This buffer for the ion pair method solves the separation problem underlying the present invention, i.e. the purification of RNA on a preparative scale, in a particularly favourable manner. The following may be considered as further buffer solutions: trifluoroace- tic acid, acetic acid, formic acid, acetate buffer, phosphate buffer, tetrabutylammonium bisulfate, tetrabutylammonium bromide and tetrabutylammonium chloride.
Selection of the mobile phase depends on the type of separation desired. This means that the mobile phase established for a specific separation, as may be known for example from the prior art, cannot be straightforwardly applied to a different separation problem with a sufficient prospect of success. For each separation problem, the ideal elution conditions, in particular the mobile phase used, have to be determined by empirical testing.
In a preferred embodiment of the HPLC method according to the invention, a mixture of an aqueous solvent and an organic solvent is used as the mobile phase for eluting the RNA. It is favourable for a buffer to be used as the aqueous solvent which has in particular a pH of 6.0-8.0, for example of about 7, for example. 7.0; preferably the buffer is triethylammonium acetate, particularly preferably a 0.02 M to 0.5 M, in particular 0.08 M to 0.12 M, very particularly an about 0.1 M triethylammonium acetate buffer, which, as described above, also acts as a counterion to the RNA in the ion pair method.
In a preferred embodiment, the organic solvent which is used in the mobile phase is ace- tonitrile, methanol, ethanol, 1 -propanol, 2-propanol and acetone or a mixture thereof, very particularly preferably acetonitrile. With these organic solvents, in particular acetonitrile, purification of the RNA proceeds in particularly favourable manner with the method according to the invention.
In a particularly preferred embodiment of the method according to the invention, the mobile phase is a mixture of 0.1 M triethylammonium acetate, pH 7, and acetonitrile.
It has proven particularly favourable for the method according to the invention for the mobile phase to contain 5.0 vol.% to 25.0 vol.% organic solvent, relative to the mobile phase, and for this to be made up to 100 vol.% with the aqueous solvent. Typically, in the event of gradient separation, the proportion of organic solvent is increased, in particular by at least 10%, more preferably by at least 50% and most preferably by at least 100%, optionally by at least 200%, relative to the initial vol.% in the mobile phase. In a preferred embodiment, in the method according to the invention the proportion of organic solvent in the mobile phase amounts in the course of HPLC separation to 3 to 9, preferably 4 to 7.5, in particular 5.0 vol.%, in each case relative to the mobile phase. More preferably, the proportion of organic solvent in the mobile phase is increased in the course of HPLC separation from 3 to 9, in particular 5.0 vol.% to up to 20.0 vol.%, in each case relative to the mobile phase. Still more preferably, the method is performed in such a way that the proportion of organic sol- vent in the mobile phase is increased in the course of HPLC separation from 6.5 to 8.5, in particular 7.5 vol.%, to up to 1 7.5 vol.%, in each case relative to the mobile phase.
It has proven even more particularly favourable for the method according to the invention for the mobile phase to contain 7.5 vol.% to 17.5 vol.% organic solvent, relative to the mo- bile phase, and for this to be made up to 100 vol.% with the aqueous buffered solvent.
In the case of the method according to the invention elution may proceed isocratically or by means of gradient separation. In isocratic separation, elution of the RNA proceeds with a single eluent or a constant mixture of a plurality of eluents, wherein the solvents described above in detail may be used as eluent.
In a preferred embodiment of the method according to the invention, gradient separation is performed. In this respect, the composition of the eluent is varied by means of a gradient program. The equipment necessary for gradient separation is known to a person skilled in the art. Gradient elution may here proceed either on the low pressure side by mixing chambers or on the high pressure side by further pumps.
Preferably, in the method according to the invention, the proportion of organic solvent, as described above, is increased relative to the aqueous solvent during gradient separation. The above-described agents may here be used as the aqueous solvent and the likewise above-described agents may be used as the organic solvent. For example, the proportion of organic solvent in the mobile phase may be increased in the course of HPLC separation from 5.0 vol.% to 20.0 vol.%, in each case relative to the mobile phase. In particular, the proportion of organic solvent in the mobile phase may be increased in the course of HPLC separation from 7.5 vol.% to 1 7.5 vol.%, in particular 9.5 to 14.5 vol.%, in each case relative to the mobile phase.
The following gradient program has proven particularly favourable for the purification of RNA with the method according to the invention:
Eluent A: 0.1 M triethylammonium acetate, pH 7 Eluent B: 0.1 M triethylammonium acetate, pH 7, with 25 vol.% acetonitrile
Eluent composition: start: 62% A and 38% B (1 st to 3rd minute) increase to 58% B (1 .67% increase in B per minute), (3rd-15th minute) 100% B (15th to 20th minute)
Another example of a gradient program is described below, the same eluent A and B being used:
Eluent composition:
• starting level: 62% A and 38% B (1 st-3rd min) • separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd-
5th min)
• separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th- 14th min)
• rinsing range: 100% B (15th-20th min)
So that the column is not soiled or clogged and detection is not disrupted, it is favourable to use purified solvent for HPLC. Such purified solvents are commercially obtainable. They may additionally also be filtered with a 1 to 5 μm microfilter, which is generally mounted in the system upstream of the pump. It is additionally favourable for ail the solvents to be de- gassed prior to use, since otherwise gas bubbles occur in most pumps. If air bubbles occur in the solvent, this may interfere not only with separation but also with the continuous monitoring of outflow in the detector. The solvents may be degassed by heating, by vigor- ous stirring with a magnetic stirrer, by brief evacuation, by ultrasonication or by passing a small stream of helium through the solvent storage vessel.
The flow rate of the eluent is so selected that good separation of the RNA from the other constituents contained in the sample to be investigated takes place. The eluent flow rate selected for the method according to the invention may amount to from 1 ml/min to several litres per minute (in the case of upscaling), in particular about 1 to 1000 ml/min, more preferably 5 ml to 500 ml/min, even more preferably more than 100 ml/min, depending on the type and scope of the upscaling. This flow rate may be established and regulated by the pump.
Detection proceeds favourably with a UV detector at 254 nm, wherein a reference measurement may be made at 600 nm. However, any other detection method may alternatively be used, with which the RNA described above in greater detail may be detected in satisfac- tory and reliable manner.
For preparative purification of the RNA with the method according to the invention, it is advisable to collect the RNA-containing eluted solvent quantities. In this respect, it is favourable to carry out this collection in such a way that the eluted solvent is collected in individual separated fractions. This may take place for example with a fraction collector. In this way, the high-purity RNA-containing fractions may be separated from other RNA- containing fractions which still contain undesired impurities, albeit in very small quantities. The individual fractions may be collected for example over 1 minute.
The method according to the invention is favourably performed under completely denaturing conditions. This may proceed for example in that sample application takes place at a temperature of 4-12°C, the HPLC method otherwise proceeding at a higher temperature, preferably at 700C or more, particularly preferably at 75°C or more, in particular up to 820C, and very particularly preferably at about 78°C. Heating is typically important for de- naturing the RNA, whereby better separation is ultimately achieved. Resolution falls at lower temperatures, i.e. separation of the RNA from the impurities decreases. Sample application may be performed with two methods, stop-flow injection or loop injection. For stop-flow injection a microsyringe is used which is able to withstand the high pressure applied in HPLC. The sample is injected through a septum in an inlet valve either directly onto the column packing or onto a small drop of inert material immediately over the packing. The system may in this case be under elevated pressure, or the pump may be turned off prior to injection, which is then performed when the pressure has fallen to close to the normal value. In the case of loop injection, a loop injector is used to introduce the sample. This consists of a tubular loop, into which the sample is inserted. By means of a suitable rotary valve, the stationary phase is then conveyed out of the pump through the loop, whose outlet leads directly into the column. The sample is entrained in this way by the stationary phase into the column, without solvent flow to the pump being interrupted.
The present invention further provides the use of a porous reversed phase in the above- described HPLC method according to the invention.
In a preferred embodiment of the method according to the invention, the porous reversed phase has a particle size of 8.0 μm to 50 μm, in particular 8.0 to 30, still more preferably 8.0 to 25 μm. The reversed phase may be present in bead form. The method according to the invention may be performed particularly favourably with a porous reversed phase with this particle size and/or in the form of beads, wherein particularly good separation results are obtained.
The reversed phase used for the use according to the invention is porous, since with stationary reversed phases, which are not porous, and thus differ completely with regard to particle size from the subject matter of the present invention, as described for example by A. Azarani and K.H. Hecker, excessively high pressures are built up, such that preparative purification of the RNA by means of HPLC is possible only with great difficulty, if at all.
In one preferred embodiment of the use according to the invention, the reversed phase has a pore size of 1000 Λ to 5000 A, in particular a pore size of 1000 A to 4000 A, or the above-stated preferred values. Particularly preferred pore sizes for the reversed phases are 1000-1200 A and 3500-4500 A. With a reversed phase having these pore sizes, particularly good results are achieved with regard to purification of the RNA using the method accord- ing to the invention, in particular the elevated pressures built up in the method according to A. Azarani and K.H. Hecker are thus avoided, whereby preparative separation is made possible in a particularly favourable manner. At pore sizes of below 1000 A, separation of the RNA becomes poorer, for which reason such particle sizes are less preferable. However, the pore size may be selected in dependence of the RNA size to be separated, as mentioned above.
In a particularly preferred embodiment of the use according to the invention, the material for the reversed phase is a polystyrenedivinylbenzene, wherein in particular non-alkylated polystyrenedivinylbenzene may be used. Stationary phases with polystyrenedivinylbenzene are known per se. The per se known polystyrenedivinylbenzenes already used for HPLC methods, which are commercially obtainable, may be used for the method according to the invention.
A non-alkylated porous polystyrenedivinylbenzene which is very particularly preferred for the use according to the invention is one which has in particular a particle size of 8.0 + 1.5 μm, in particular 8.0 ± 0.5 μm, and a pore size of 1000- or 4000 A. With this material for the reversed phase, the advantages described below may be achieved in a particularly favourable manner.
For the use according to the invention, this stationary phase described in greater detail above is located in a column. V2A steel is conventionally used as the material for the column, but other materials may also be used for the column provided they are suitable for the conditions prevailing during HPLC recovery. Conventionally the column is straight. It is favourable for the HPLC column to have a length of 5 cm to 100 cm and a diameter of 4 mm to 25 mm. Columns used for the method according to the invention may in particular have the following dimensions: 50 mm long and 7.5 mm in diameter or 50 mm long and 4.6 mm in diameter, or 50 mm long and 10 mm in diameter or any other dimension with regard to length and diameter, which is suitable for preparative purification of RNA by means of HPLC, even lengths of several metres and also larger diameters being feasible in the case of upscaling. The dimensions are here geared towards what is technically possible with liquid chromatography. Further conditions for the use according to the invention have already been described above in connection with the method according to the invention, such that reference is made thereto to avoid unnecessary repetition.
The use according to the invention, in particular the preferred embodiments described in detail hereinafter, exhibits a series of advantages: in the context of the use according to the invention, substantially larger quantities of RNA may be separated than is possible with the methods known from the prior art. The use according to the invention enables these higher quantities to be obtained with a high degree of purity. Separation of degraded fragments or of relatively long fragments or of fragments which have arisen as a result of premature termination of transcription is possible. In addition, good separation of further impurities present in RNA samples, such as enzymes, for example RNases or polymerases, and nucleotides is possible. These relatively large quantities of RNA may be recovered within minutes, even from samples having a very high level of contamination with impurities. RNA recovery proceeds reliably, i.e. high purity RNA is recovered with great reliability. The use according to the invention enables high resolution to be achieved. The use according to the invention may easily be operated automatically. It is thus most suitable for routine purification of RNA on a preparative scale. RNA is stable under the conditions of the HPLC separation method, i.e. degradation into RNA fragments and thus the occurrence of new impurities or a reduc- tion in the yield of pure RNA during HPLC purification is avoided. The use according to the invention may thus be performed in a simple, rapid, inexpensive manner, accurately and with reliable results. Isolation of RNA after HPLC separation may proceed simply with a fraction collector, i.e. the recovery of RNA may be achieved very simply, direct further processing of the RNA likewise being possible. Detection may proceed very sensitively.
The invention is explained in greater detail below with reference to the Figures and an exemplary embodiment, without being limited thereto.
Figures
Figure 1 shows a chromatogram of a separation of 10 μg of luciferase-mRNA (analytical);
Figure 2 shows a chromatogram of a separation of 1 .5 μg of luciferase-mRNA (preparative);
Figure 3 shows the chromatogram of Figure 2, the collected fractions being highlighted in grey;
Figure 4 shows verification of the purity of the collected mRNA fractions on an agarose gel;
Figure 5 shows a chromatogram of a separation of 200 μg of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 1000 A;
Figure 6 shows a chromatogram of a separation of 100 μg of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 4000 A;
Figure 7 shows a chromatogram of a separation of 250 μg of a 2 kb and of a 4 kb RNA fragment with a stationary matrix, which has a pore size of 4000 A,
Figure 8 is a bar chart which demonstrates the improvement in the expression of luciferase from RNA purified with the method according to the invention.
Figure 9 shows the sequence of the wild-type of luciferase, specifically the mRNA se- quence, which was used for the exemplary embodiments.
Figure 10 shows a chromatogram of a separation of 3 mg of a Flic(GC) RNA preparation (FC-9050) with a stationary matrix, which has a pore size of 4000 A, Figure 11 shows a chromatogram of a separation of 1 ,5 mg of a FOLHI (GC) RNA preparation (FO-9334) with a stationary matrix, which has a pore size of 4000 A,
Figure 12 shows a chromatogram of a separation of 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849) with a stationary matrix, which has a pore size of 4000 A,
Figure 13 shows a chromatogram of a separation of 1 ,5 mg of a PSCA(GC) RNA preparation (PS-8845) with a stationary matrix, which has a pore size of 4000 A,
Figure 14 shows a chromatogram of a separation of 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848) with a stationary matrix, which has a pore size of 4000 A,
Figure 15 shows a chromatogram of a separation of a mixture of a 2 kb RNA (200μg) and a
4kb RNA (200μg) with a non-porous stationary matrix;
Figure 16 shows an agarose gel analysis corresponding to the chromatogram according to
Figure 15, which shows a separation of a mixture of a 2 kb RNA (200μg) and a
4kb RNA (200μg) with a non-porous stationary matrix;
Example 1 : Purification of 1 .5 mg of luciferase mRNA by means of the HPLC method according to the invention
Luciferase mRNA having a size of 1825 base pairs was used for separation. A porous, non- alkylated polystyrene/divinylbenzene (polystyrenedivinylbenzene) matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 μm and a pore size of 4000 A. The column used was 5.0 cm long and had a diameter of 7.5 mm. The sampler temperature was 12°C, the temperature for the HPLC separation, in particular of the separation column, was 78°C, i.e. operations were carried out under completely denaturing conditions.
Separation proceeded via a gradient program:
Eluent A: 0.1 M triethylammonium acetate, pH 7
Eluent B: 0.1 M triethylammonium acetate, pH 7, with 25 vol.% acetonitrile Eluent composition: start: 62% A and 38% B (1 st to 3rd minute) increase to 58% B (1 .67% increase in B per minute), (3rd-15th minute) 100% B (15th to 20th minute)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
In order to show the impurities in the luciferase mRNA, first of all an analytical separation was performed, in which only 10 μg of the luciferase mRNA was applied to the column. The result of this analytical HPLC separation is shown in Figure 1 (the gradient is shown with broken lines in the chromatogram). As may clearly be seen, in addition to the desired mRNA of luciferase (retention time 12.760 minutes), impurities are still present (retention times of 12.398 minutes and 13.227 minutes), which flank the luciferase mRNA peak.
The purpose of purifying the luciferase mRNA on a preparative scale is to remove these undesired impurities.
Figures 2 and 3 show the corresponding chromatograms for the separation of luciferase mRNA on a preparative scale (1 .5 mg), wherein the gradient program is shown in broken lines in Figure 2. The collected fractions 1 to 10 are highlighted in grey in Figures 2 and 3; the fractions were collected over a period of 1 minute.
In each case 1 μg of the luciferase mRNA from collected fractions 2 to 10 was applied onto an agarose gel and visualised with the assistance of ethidium bromide, in order to determine the purity of the mRNA fraction.
The result of the agarose gel separation is shown in Figure 4 (M denotes a size standard). Fractions 5 to 8 contain the desired luciferase mRNA without undesired impurities. As a result, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
This exemplary embodiment shows that, using the method according to the invention, it is possible to purify mRNA on a preparative scale by means of HPLC.
Example 2: Separation of 200 μg of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 1000 A;
200 μg of a 2 kb and 4 kb RNA fragment were used for separation. A porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 μm and a pore size of 1000 Λ. The column used was 2.5 cm long and had a diameter of 4.6 mm. The sampler temperature was 12°C, the temperature for the HPLC separation, in particular of the separa- tion column, was 78°C, i.e. operations were carried out under completely denaturing conditions.
Separation proceeds via a gradient program
Eluent A: 0.1 M triethylammonium acetate Eluent B: 0.1 M triethylammonium acetate/25% acetonitrile
Eluent composition: • starting level: 62% A and 38% B (1 st-3rd min) • separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd- 5th min)
• separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th- 14th min) • rinsing range: 100% B (15th-20th min)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
Figure 5 shows the corresponding chromatograms for this separation on a preparative scale (200 μg), wherein the gradient program is shown in broken lines in Figure 5. The collected fractions are highlighted in grey in Figure 5; the fractions were collected over a period of 1 minute. As a result, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
Example 3: Separation of 100 μg of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 4000 A;
100 μg of a 2 kb and 4 kb RNA fragment were used for separation. A porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 μm and a pore size of 4000 A. The column used was 2.5 cm long and had a diameter of 4.6 mm. The sampler temperature was 12°C, the temperature for the HPLC separation, in particular of the separa- tion column, was 78°C, i.e. operations were carried out under completely denaturing conditions.
Separation proceeds via a gradient program
Eluent A: 0.1 M triethylammonium acetate Eluent B: 0.1 M triethylammonium acetate/ 25% acetonitrile
Eluent composition:
• starting level: 62% A and 38% B (1 st-3rd min) • separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd- 5th min)
• separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th- 14th min) • rinsing range: 100% B (15th-20th min)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
Figure 6 shows the corresponding chromatograms for this separation on a preparative scale (100 μg), wherein the gradient program is shown in broken lines in Figure 6. The fractions collected are highlighted in grey in Figure 6. As a result, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
Example 4: Separation of 250 μg of a 2 kb and of a 4 kb RNA fragment with a stationary phase, which has a pore size of 4000 A;
250 μg of a 2 kb and 4 kb RNA fragment were used for separation. A porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 μm and a pore size of 4000 A. The column used was 2.5 cm long and had a diameter of 4.6 mm. The sampler temperature was 120C, the temperature for the HPLC separation, in particular of the separation column, was 780C, i.e. operations were carried out under completely denaturing con- ditions.
Separation proceeds via a gradient program
Eluent A: 0.1 M triethylammonium acetate
Eluent B: 0.1 M triethylammonium acetate/25% acetonitri Ie Eluent composition: starting level: 62% A and 38% B (1 st-3rd min) separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd-
5th min) • separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th- 14th min)
• rinsing range: 100% B (15th-20th min)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/mi n.
Figure 7 shows the corresponding chromatograms for this separation on a preparative scale (250 μg), wherein the gradient program is shown in broken lines in Figure 7. The fractions collected are highlighted in grey in Figure 7. As a result, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
The improvement of the expression of luciferase resulting from purification with the method according to the invention is shown in Figure 8.
Example 5: Separation of 3 mg of a 1,7 kb RNA (FliC(GC)-preparation, FC9050) with a stationary phase, which has a pore size of 4000 A;
3 mg of a 1 ,7 kb RNA (FliC(GC)-preparation, FC9050) were used for separation. A porous, non-alkylated polystyrene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 μm and a pore size of 4000 A. The column used was 2.5 cm long and had a diameter of 4.6 mm. The sampler temperature was 12°C, the temperature for the HPLC separation, in particular of the separation column, was 78°C, i.e. operations were carried out under completely denaturing conditions.
Separation proceeds via a gradient program
Eluent A: 0.1 M triethylammonium acetate Eluent B: 0.1 M triethylammonium acetate/25% acetonitrile
Eluent composition: starting level: 62% A and 38% B (1 st-3rd min) separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd-5th min)
• separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th-14th min)
• rinsing range: 100% B (15th-20th min)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
Figure 10 shows the corresponding chromatograms for this separation on a preparative scale (3 mg), wherein the gradient program is shown in broken lines in Figure 10. The fractions collected are highlighted in grey in Figure 10. As can be seen, a separation of the RNA was successful, leading to a highly pure fraction of RNA in preparative scale.
Example 6: Separation of 1,5 mg of a FOLHI (GC) RNA preparation (FO-9334), 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849), 1,5 mg of a PSCA(GC) RNA preparation (PS-8845) or 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848) with a stationary phase, which has a pore size of 4000 A;
1 ,5 mg of a FOLHI (GC) RNA preparation (FO-9334), 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849), 1 ,5 mg of a PSCA(GC) RNA preparation (PS-8845) or 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848) were used for separation. A porous, non-alkylated polysty- rene/divinylbenzene matrix (conventional commercial product from Polymer Laboratories) was used as the stationary phase. It had a particle size of 8 μm and a pore size of 4000 A. The column used was 2.5 cm long and had a diameter of 4.6 mm. The sampler temperature was 120C, the temperature for the HPLC separation, in particular of the separation column, was 78°C, i.e. operations were carried out under completely denaturing conditions.
Separation proceeds via a gradient program Eluent A: 0.1 M triethylammonium acetate
Eluent B: 0.1 M triethylammonium acetate/25% acetonitrile
Eluent composition: starting level: 62% A and 38% B (1 st-3rd min) • separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd-5th min)
• separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th-14th min) • rinsing range: 100% B (15th-20th min)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
Figures 1 1 , 12, 13 and 14 shows the corresponding chromatograms for this separation of 1 ,5 mg of a FOLHI (GC) RNA preparation (FO-9334), 1 ,5 mg of a KLK3(GC) RNA preparation (KL-8849), 1 ,5 mg of a PSCA(GC) RNA preparation (PS-8845) or 1 ,5 mg of a STEAP(GC) RNA preparation (ST-8848), respectively on a preparative scale (1 ,5 mg), wherein the gradient program is shown in broken lines in these Figures. The fractions collected are high- lighted in grey in Figure 1 1 , 12, 13 and 14. As can be seen, a separation of the RNAs is possible in each case, leading to a highly pure fraction of RNA in each case in preparative scale.
Example 7: Separation of a mixture of 200 μg of a 2 kb RNA and 200 μg of a 4kb RNA on a non-porous stationary polystyrenedivinylbenzene matrix (Comparison Example)
A mixture of 200 μg of a 2 kb RNA and 200 μg of a 4kb RNA were used for separation. The columns used were packed with either a 1000 Λ non-alkylated porous polystyrenedivinyl- benzene or a non-porous stationary polystyrenedivinylbenzene matrix. The column used for the 1000 A non-alkylated porous polystyrenedivinylbenzene was 2.5 cm long and had a diameter of 4.6 mm. The column used for the non-alkylated non-porous polystyrenedivinylbenzene was 2.5 cm long and had a diameter of 4.0 mm. The sampler temperature was 12°C, the temperature for the HPLC separation, in particular of the separation column, was 78°C, i.e. operations were carried out under completely denaturing conditions.
Separation proceeds via a gradient program
Eluent A: 0.1 M triethylammonium acetate Eluent B: 0.1 M triethylammonium acetate/25% acetonitrile
Eluent composition:
• starting level: 62% A and 38% B (1 st-3rd min)
• separation range I: gradient 38%-49.5% B (5.75% increase in B/min) (3rd-5th min)
• separation range II: gradient 49.5%-57% B (0.83% increase in B/min) (5th-14th min)
• rinsing range: 100% B (15th-20th min)
Detection proceeded with a UV detector at 254 nm with a reference measurement at 600 nm. Flow rate was 3 ml/min.
Figures 15 shows the corresponding chromatogram for this separation on a preparative scale (each 200 μg), wherein the gradient program is shown in broken lines in Figure 15. The fractions collected are highlighted in grey in Figure 15. As can be seen, a separation of both RNAs is not possible using a common non-alkylated porous polystyrenedivinylbenzene as a matrix. Figure 16 shows the agarose gel corresponding to the chromatogram according to Figure 15.

Claims

Claims
1 . A method for purifying RNA on a preparative scale, characterised in that the RNA is purified by means of HPLC or low or normal pressure liquid chromatography meth- ods using a porous reversed phase as stationary phase.
2. A method according to claim 1 , wherein the RNA is selected from among tRNA, rRNA, mRNA or whole-cell RNA, in particular also RNA variants, such as for example aptamers or ribozymes.
3. A method according to one of the preceding claims, characterised in that the RNA has a size of up to about 15000 nucleotides or base pairs.
4. A method according to one of the preceding claims, characterised in that the RNA has a size of up to 100 to 10000 nucleotides or base pairs.
5. A method according to one of the preceding claims, characterised in that the porous reversed phase has a particle size of 8 μm to 50 μm, in particular 8 to 30 and more preferably 8 to 25 μm.
6. A method according to one of the preceding claims, characterised in that the reversed phase has a pore size of 1000 Λ to 5000 Λ.
7. A method according to one of the preceding claims, characterised in that the re- versed phase has a pore size of 1000 A to 4000 A.
8. A method according to one of the preceding claims, characterised in that the porous reversed phase is a porous polystyrene, a porous non-alkylated polystyrene, a poly- styrenedivinylbenzene, a porous non-alkylated polystyrenedivinylbenzene, a porous silica gel, a porous silica gel modified with non-polar residues, a porous silica gel modified with alkyl containing residues, selected from butyl-, octyl and/or octadecyl containing residues, a porous silica gel modified with phenylic residues, or a porous polymethacrylate.
9. A method according to one of the preceding claims, characterised in that the porous reversed phase is formed by beads or occurs as a polymerized block.
10. A method according to one of the preceding claims, characterised in that the HPLC column has a length of more than 5 cm, in particular up to 100 cm and a diameter of more than 4 mm, in particular up to 25 mm, preferably up to 1 m, more preferably up to 100 cm.
1 1 . A method according to one of the preceding claims, characterised in that HPLC purification is performed as an ion pair method, an ion with positive charge being added to the mobile phase as counterion to the negatively charged RNA, or by size exclusion chromatography, gel filtration, affinity chromatography, hydrophobic interaction chromatography or ion pair chromatography.
12. A method according to one of the preceding claims, characterised in that a mixture of an aqueous solvent and an organic solvent is used for HPLC purification for the purpose of elution.
13. A method according to claim 12, characterised in that the aqueous solvent is a buffer.
14. A method according to claim 13, characterised in that the buffer is triethylammo- nium acetate, trifluoroacetic acid, acetic acid, formic acid, acetate buffer, phosphate buffer, tetrabutylammonium bisulfate, tetrabutylammonium bromide and tetrabu- tylammonium chloride.
15. A method according to claim 14, characterised in that the triethylammonium acetate buffer is a 0.1 M triethylammonium acetate buffer.
16. A method according to one of claims 12 to 15, characterised in that the organic solvent is acetonitrile, methanol, ethanol, 1 -propanol, 2-propanol and acetone or a mixture thereof.
1 7. A method according to claim 16, characterised in that the organic solvent is acetoni- trile.
18. A method according to one of claims 12 to 1 7, characterised in that the mobile phase is a mixture of 0.1 M triethylammonium acetate and acetonitrile.
19. A method according to one of claims 12 to 18, characterised in that the mobile phase contains 5.0 vol.% to 25.0 vol.% organic solvent, relative to the mobile phase, and this is made up to 100 vol.% with the aqueous solvent.
20. A method according to claim 19, characterised in that the mobile phase contains 7.5 vol.% to 1 7.5 vol.%, in particular 9.5 to 14.4 vol.%, organic solvent, relative to the mobile phase, and this is made up to 100 vol.% with the aqueous solvent.
21 . A method according to one of the preceding claims, characterised in that the separation proceeds isocratically.
22. A method according to one of claims 1 to 20, characterised in that a gradient sepa- ration proceeds.
23. A method according to claim 22, wherein, in the event of gradient separation, the proportion of organic solvent is increased, in particular by at least 10%, more preferably by at least 50% and most preferably by at least 100%, optionally by at least 200%, relative to the initial vol.% in the mobile phase.
24. A method according to claim 23, characterised in that the proportion of organic solvent in the mobile phase in the course of HPLC separation amounts to from 3 to 9, preferably 4 to 7.5, in particular 5.0 vol.%.
25. A method according to claim 23 or claim 24, characterised in that the proportion of organic solvent in the mobile phase is increased in the course of HPLC separation from 3 to 9, in particular 5.0 vol.%, to up to 20.0 vol.%, in each case relative to the mobile phase.
26. A method according to claim 25, characterised in that the proportion of organic solvent in the mobile phase is increased in the course of HPLC separation from 6.- to 8.5, in particular 7.5 vol.%, to up to 1 7.5 vol.%, in each case relative to the mo bile phase.
27. Use of a porous reversed phase in an HPLC method according to one of claims 1 tc 26.
28. Use according to claim 27, wherein the porous reversed phase is as defined in one of claims 5 to 8.
PCT/EP2007/011294 2006-12-22 2007-12-20 Method for purifying rna on a preparative scale by means of hplc WO2008077592A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
EP07857017A EP2092064B1 (en) 2006-12-22 2007-12-20 Method for purifying rna on a preparative scale by means of hplc
AT07857017T ATE481482T1 (en) 2006-12-22 2007-12-20 METHOD FOR PRELIMINARY RNA PURIFICATION USING HPLC
DE602007009295T DE602007009295D1 (en) 2006-12-22 2007-12-20 PROCESS FOR PREPARANT RNA CLEANING BY HPLC
KR1020097012957A KR101183173B1 (en) 2006-12-22 2007-12-20 Method for purifying rna on a preparative scale by means of hplc
DK07857017.3T DK2092064T3 (en) 2006-12-22 2007-12-20 Method of Preparative RNA Purification by HPLC
JP2009541898A JP5307724B2 (en) 2006-12-22 2007-12-20 Method of purifying RNA using HPLC on a preparative scale
PL07857017T PL2092064T3 (en) 2006-12-22 2007-12-20 Method for purifying rna on a preparative scale by means of hplc
CN200780047133.2A CN101563457B (en) 2006-12-22 2007-12-20 Method for purifying RNA on preparative scale by means of HPLC
CA2670727A CA2670727C (en) 2006-12-22 2007-12-20 Method for purifying rna on a preparative scale by means of hplc
US12/520,172 US8383340B2 (en) 2006-12-22 2007-12-20 Method for purifying RNA on a preparative scale by means of HPLC
AU2007338360A AU2007338360B2 (en) 2006-12-22 2007-12-20 Method for purifying RNA on a preparative scale by means of HPLC

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE102006061015.6 2006-12-22
DE102006061015A DE102006061015A1 (en) 2006-12-22 2006-12-22 Process for the purification of RNA on a preparative scale by HPLC

Publications (1)

Publication Number Publication Date
WO2008077592A1 true WO2008077592A1 (en) 2008-07-03

Family

ID=39186980

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/011294 WO2008077592A1 (en) 2006-12-22 2007-12-20 Method for purifying rna on a preparative scale by means of hplc

Country Status (13)

Country Link
US (1) US8383340B2 (en)
EP (1) EP2092064B1 (en)
JP (1) JP5307724B2 (en)
KR (1) KR101183173B1 (en)
CN (1) CN101563457B (en)
AT (1) ATE481482T1 (en)
AU (1) AU2007338360B2 (en)
CA (1) CA2670727C (en)
DE (2) DE102006061015A1 (en)
DK (1) DK2092064T3 (en)
ES (1) ES2352306T3 (en)
PL (1) PL2092064T3 (en)
WO (1) WO2008077592A1 (en)

Cited By (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014140211A1 (en) 2013-03-15 2014-09-18 Novartis Ag Rna purification methods
WO2015062738A1 (en) 2013-11-01 2015-05-07 Curevac Gmbh Modified rna with decreased immunostimulatory properties
WO2015101416A1 (en) 2013-12-30 2015-07-09 Curevac Gmbh Methods for rna analysis
WO2015149944A2 (en) 2014-04-01 2015-10-08 Curevac Gmbh Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
WO2015188933A1 (en) 2014-06-10 2015-12-17 Curevac Ag Methods and means for enhancing rna production
US9352028B2 (en) 2007-10-09 2016-05-31 Curevac Ag Composition for treating lung cancer, particularly of non-small lung cancers (NSCLC)
US9402887B2 (en) 2007-10-09 2016-08-02 Curevac Ag Composition for treating prostate cancer (PCa)
WO2016170176A1 (en) 2015-04-22 2016-10-27 Curevac Ag Rna containing composition for treatment of tumor diseases
WO2016180430A1 (en) 2015-05-08 2016-11-17 Curevac Ag Method for producing rna
WO2016184822A1 (en) 2015-05-15 2016-11-24 Curevac Ag Prime-boost regimens involving administration of at least one mrna construct
WO2016193206A1 (en) 2015-05-29 2016-12-08 Curevac Ag A method for producing and purifying rna, comprising at least one step of tangential flow filtration
WO2017001058A1 (en) * 2015-07-01 2017-01-05 Curevac Ag Method for analysis of an rna molecule
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US9616084B2 (en) 2009-12-09 2017-04-11 Curevac Ag Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
WO2017066791A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Sugar substituted mrna cap analogs
WO2017066782A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Hydrophobic mrna cap analogs
WO2017066789A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified sugar
WO2017066793A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs and methods of mrna capping
WO2017066781A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified phosphate linkage
WO2017112865A1 (en) 2015-12-22 2017-06-29 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
WO2017109161A1 (en) 2015-12-23 2017-06-29 Curevac Ag Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
WO2017140345A1 (en) 2016-02-15 2017-08-24 Curevac Ag Method for analyzing by-products of rna in vitro transcription
WO2017149139A1 (en) 2016-03-03 2017-09-08 Curevac Ag Rna analysis by total hydrolysis
WO2017191264A1 (en) 2016-05-04 2017-11-09 Curevac Ag Nucleic acid molecules and uses thereof
WO2017212006A1 (en) 2016-06-09 2017-12-14 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2017212007A1 (en) 2016-06-09 2017-12-14 Curevac Ag Cationic carriers for nucleic acid delivery
WO2017212009A1 (en) 2016-06-09 2017-12-14 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2017218704A1 (en) 2016-06-14 2017-12-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
WO2018041921A1 (en) 2016-08-31 2018-03-08 Curevac Ag Mixing device for the production of a liquid nucleic acid composition
WO2018078053A1 (en) 2016-10-26 2018-05-03 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018096179A1 (en) 2016-11-28 2018-05-31 Curevac Ag Method for purifying rna
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
WO2018115525A1 (en) 2016-12-23 2018-06-28 Curevac Ag Lassa virus vaccine
WO2018115507A2 (en) 2016-12-23 2018-06-28 Curevac Ag Henipavirus vaccine
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
WO2018141371A1 (en) 2017-01-31 2018-08-09 Curevac Ag Purification and/or formulation of rna
WO2018170336A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Lipid nanoparticle formulation
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2018167320A1 (en) 2017-03-17 2018-09-20 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
WO2018211038A1 (en) 2017-05-17 2018-11-22 Curevac Ag Method for determining at least one quality parameter of an rna sample
WO2018232120A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
WO2019036638A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods of preparing modified rna
WO2019038332A1 (en) 2017-08-22 2019-02-28 Curevac Ag Bunyavirales vaccine
EP3450561A1 (en) 2013-08-21 2019-03-06 CureVac AG Method for increasing expression of rna-encoded proteins
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
EP3461498A1 (en) 2013-08-21 2019-04-03 CureVac AG Rabies vaccine
WO2019092153A1 (en) 2017-11-08 2019-05-16 Curevac Ag Rna sequence adaptation
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2019115635A1 (en) 2017-12-13 2019-06-20 Curevac Ag Flavivirus vaccine
EP3540060A1 (en) 2013-12-30 2019-09-18 CureVac AG Methods for rna analysis
EP3542802A1 (en) 2013-11-01 2019-09-25 CureVac AG Modified rna with decreased immunostimulatory properties
WO2019193183A2 (en) 2018-04-05 2019-10-10 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
EP3586871A2 (en) 2013-08-21 2020-01-01 CureVac AG Respiratory syncytial virus (rsv) vaccine
WO2020002525A1 (en) 2018-06-27 2020-01-02 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
WO2020002598A1 (en) 2018-06-28 2020-01-02 Curevac Ag Bioreactor for rna in vitro transcription
WO2020061367A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2020061457A1 (en) 2018-09-20 2020-03-26 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof
US10653712B2 (en) 2016-09-14 2020-05-19 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020160397A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Methods of preparing lipid nanoparticles
WO2020160430A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Vortex mixers and associated methods, systems, and apparatuses thereof
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
EP3701963A1 (en) 2015-12-22 2020-09-02 CureVac AG Method for producing rna molecule compositions
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
WO2021123332A1 (en) 2019-12-20 2021-06-24 Curevac Ag Lipid nanoparticles for delivery of nucleic acids
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
WO2021204179A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2021204175A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Lipid nanoparticle composition
EP3916091A2 (en) 2015-05-20 2021-12-01 CureVac AG Dry powder composition comprising long-chain rna
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
WO2021254593A1 (en) 2020-06-15 2021-12-23 Curevac Ag Analysis of nucleic acid mixtures
EP3928800A2 (en) 2015-05-20 2021-12-29 CureVac AG Dry powder composition comprising long-chain rna
WO2022002040A1 (en) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
WO2022037652A1 (en) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022049093A1 (en) 2020-09-01 2022-03-10 CureVac RNA Printer GmbH Manufacturing device for a pharmaceutical product
WO2022112498A1 (en) 2020-11-27 2022-06-02 CureVac RNA Printer GmbH A device for preparing a dna product by means of capillary polymerase chain reaction
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
US11384352B2 (en) 2016-12-13 2022-07-12 Modernatx, Inc. RNA affinity purification
WO2022152141A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Polymer conjugated lipid compounds and lipid nanoparticle compositions
WO2022152109A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2022240960A1 (en) * 2021-05-14 2022-11-17 Crispr Therapeutics Ag Mrna large scale synthesis and purification
WO2022247755A1 (en) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
WO2023007019A1 (en) 2021-07-30 2023-02-02 CureVac SE Cap analogs having an acyclic linker to the guanine derivative nucleobase
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
WO2023044343A1 (en) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Acyclic lipids and methods of use thereof
WO2023044333A1 (en) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Cyclic lipids and methods of use thereof
EP4162950A1 (en) 2021-10-08 2023-04-12 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2023056917A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023056914A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023122752A1 (en) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Constrained lipids and methods of use thereof
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
EP4227319A1 (en) 2018-04-17 2023-08-16 CureVac SE Novel rsv rna molecules and compositions for vaccination
WO2023196931A1 (en) 2022-04-07 2023-10-12 Renagade Therapeutics Management Inc. Cyclic lipids and lipid nanoparticles (lnp) for the delivery of nucleic acids or peptides for use in vaccinating against infectious agents
WO2023196634A2 (en) 2022-04-08 2023-10-12 Flagship Pioneering Innovations Vii, Llc Vaccines and related methods
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
EP4289965A2 (en) 2016-02-12 2023-12-13 CureVac SE Method for analyzing rna
US11866696B2 (en) 2017-08-18 2024-01-09 Modernatx, Inc. Analytical HPLC methods
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2024030856A2 (en) 2022-08-01 2024-02-08 Flagship Pioneering Innovations Vii, Llc Immunomodulatory proteins and related methods
WO2024037578A1 (en) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition of lipid nanoparticles
US11912982B2 (en) 2017-08-18 2024-02-27 Modernatx, Inc. Methods for HPLC analysis
DE202023106198U1 (en) 2022-10-28 2024-03-21 CureVac SE Nucleic acid-based vaccine
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
EP4353257A2 (en) 2015-04-13 2024-04-17 CureVac Manufacturing GmbH Method for producing rna compositions
US11964012B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
WO2024089229A1 (en) 2022-10-28 2024-05-02 CureVac SE Improved formulations comprising lipid-based carriers encapsulating rna

Families Citing this family (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102007001370A1 (en) * 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
US20110172405A1 (en) * 2008-09-17 2011-07-14 Ge Healthcare Bio-Sciences Corp. Method for small rna isolation
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
EP2955230A1 (en) 2010-07-30 2015-12-16 CureVac AG Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
WO2012019168A2 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2012019630A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
DE19177059T1 (en) 2010-10-01 2021-10-07 Modernatx, Inc. RIBONUCLEIC ACID CONTAINING N1-METHYL-PSEUDOURACILE AND USES
WO2012089225A1 (en) 2010-12-29 2012-07-05 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
WO2012116715A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in newborns and infants
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
DE12722942T1 (en) 2011-03-31 2021-09-30 Modernatx, Inc. RELEASE AND FORMULATION OF MANIPULATED NUCLEIC ACIDS
JP6113737B2 (en) 2011-10-03 2017-04-12 モデルナティエックス インコーポレイテッドModernaTX,Inc. Modified nucleosides, nucleotides and nucleic acids and methods for their use
MX2014007233A (en) 2011-12-16 2015-02-04 Moderna Therapeutics Inc Modified nucleoside, nucleotide, and nucleic acid compositions.
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
WO2013120500A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
WO2013120499A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly (a) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
WO2013120497A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
WO2013120498A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
SG11201405545XA (en) 2012-03-27 2014-11-27 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr
ES2654205T3 (en) 2012-03-27 2018-02-12 Curevac Ag Artificial nucleic acid molecules for enhanced protein or peptide expression
MX357803B (en) 2012-03-27 2018-07-24 Curevac Ag Artificial nucleic acid molecules.
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
EP2834259A4 (en) 2012-04-02 2016-08-24 Moderna Therapeutics Inc Modified polynucleotides
CA2892529C (en) 2012-11-26 2023-04-25 Moderna Therapeutics, Inc. Terminally modified rna
MX2015010880A (en) 2013-02-22 2015-12-03 Curevac Gmbh Combination of vaccination and inhibition of the pd-1 pathway.
CN105051213A (en) 2013-03-14 2015-11-11 夏尔人类遗传性治疗公司 Quantitative assessment for cap efficiency of messenger RNA
WO2014144711A1 (en) * 2013-03-15 2014-09-18 Moderna Therapeutics, Inc. Analysis of mrna heterogeneity and stability
EP2983804A4 (en) 2013-03-15 2017-03-01 Moderna Therapeutics, Inc. Ion exchange purification of mrna
WO2014152031A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Ribonucleic acid purification
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP3578663A1 (en) 2013-03-15 2019-12-11 ModernaTX, Inc. Manufacturing methods for production of rna transcripts
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
WO2014186334A1 (en) 2013-05-15 2014-11-20 Robert Kruse Intracellular translation of circular rna
JP7019233B2 (en) 2013-07-11 2022-02-15 モデルナティエックス インコーポレイテッド Compositions and Methods of Use Containing Synthetic polynucleotides and Synthetic sgRNAs Encoding CRISPR-Related Proteins
CA2915712A1 (en) 2013-08-21 2015-02-26 Margit SCHNEE Rabies vaccine
AU2014310934B2 (en) 2013-08-21 2019-09-12 CureVac SE Respiratory syncytial virus (RSV) vaccine
SG10201801428RA (en) 2013-08-21 2018-03-28 Curevac Ag Method for increasing expression of rna-encoded proteins
CN105473157A (en) 2013-08-21 2016-04-06 库瑞瓦格股份公司 Combination vaccine
EP3052511A4 (en) 2013-10-02 2017-05-31 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
DK3090053T3 (en) 2013-12-30 2019-02-25 Curevac Ag Artificial nucleic acid molecules
US11254951B2 (en) 2014-12-30 2022-02-22 Curevac Ag Artificial nucleic acid molecules
CA2927254C (en) 2013-12-30 2023-10-24 Curevac Ag Artificial nucleic acid molecules
WO2015135558A1 (en) 2014-03-12 2015-09-17 Curevac Gmbh Combination of vaccination and ox40 agonists
RU2746406C2 (en) 2014-04-23 2021-04-13 МОДЕРНАТиЭкс, ИНК. Vaccines based on nucleic acids
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
US10407683B2 (en) 2014-07-16 2019-09-10 Modernatx, Inc. Circular polynucleotides
WO2016091391A1 (en) 2014-12-12 2016-06-16 Curevac Ag Artificial nucleic acid molecules for improved protein expression
US10780054B2 (en) 2015-04-17 2020-09-22 Curevac Real Estate Gmbh Lyophilization of RNA
SG11201708867UA (en) 2015-04-30 2017-11-29 Curevac Ag Immobilized poly(n)polymerase
EP4098743A1 (en) 2015-05-29 2022-12-07 CureVac AG Method for adding cap structures to rna using immobilized enzymes
US10501768B2 (en) 2015-07-13 2019-12-10 Curevac Ag Method of producing RNA from circular DNA and corresponding template DNA
US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
US11225682B2 (en) 2015-10-12 2022-01-18 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
WO2017081110A1 (en) 2015-11-09 2017-05-18 Curevac Ag Rotavirus vaccines
WO2017140905A1 (en) 2016-02-17 2017-08-24 Curevac Ag Zika virus vaccine
EP3445850B1 (en) 2016-04-22 2021-10-27 BioNTech SE Methods for providing single-stranded rna
WO2017186928A1 (en) 2016-04-29 2017-11-02 Curevac Ag Rna encoding an antibody
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
EP3808380A1 (en) 2016-12-08 2021-04-21 CureVac AG Rna for treatment or prophylaxis of a liver disease
WO2018104540A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rnas for wound healing
SG11201906297QA (en) 2017-03-24 2019-10-30 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
US11357856B2 (en) 2017-04-13 2022-06-14 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
US10034951B1 (en) 2017-06-21 2018-07-31 New England Biolabs, Inc. Use of thermostable RNA polymerases to produce RNAs having reduced immunogenicity
SG11202005760PA (en) 2017-12-21 2020-07-29 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
US11072808B2 (en) 2018-10-04 2021-07-27 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed RNA
EP3861108A1 (en) 2018-10-04 2021-08-11 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed rna
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
CN115335695A (en) * 2020-03-25 2022-11-11 沃特世科技公司 Device and method for sensitive detection and quantification of biomolecules
AU2021424650A1 (en) 2021-01-27 2023-08-17 New England Biolabs, Inc. Faustovirus capping enzyme, mrna capping enzyme compositions, methods and kits
US11028379B1 (en) 2021-01-27 2021-06-08 New England Biolabs, Inc. FCE mRNA capping enzyme compositions, methods and kits
KR20240017865A (en) 2021-06-04 2024-02-08 트랜슬레이트 바이오 인코포레이티드 Assay for quantitative assessment of mRNA capping efficiency
WO2023014649A1 (en) 2021-08-02 2023-02-09 Modernatx, Inc. Extraction-less reverse phase (rp) chromatography of mrna encapsulated in lipid nanoparticles for mrna purity assessment
KR102646914B1 (en) * 2021-10-01 2024-03-12 서울대학교산학협력단 Direct and simultaneous analysis of all oleoylglycerol isomers and oleic acid and determination of lipase stereoselectivity thereof
WO2023196950A1 (en) 2022-04-07 2023-10-12 New England Biolabs, Inc. Methods of higher fidelity rna synthesis
WO2024030369A1 (en) 2022-08-01 2024-02-08 Modernatx, Inc. Extraction-less reverse phase (rp) chromatography for mrna purity assessment
CN115774075B (en) * 2023-02-15 2023-06-06 江苏耀海生物制药有限公司 Method for analyzing in vitro transcription product component circRNA based on RP-HPLC

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2468048A1 (en) * 1994-02-23 1995-08-31 Pamela Pavco Process for purifying chemically synthesized rna
US20020102563A1 (en) * 1998-04-24 2002-08-01 Gjerde Douglas T. Apparatus and method for separating and purifying polynucleotides
US20050011836A1 (en) * 2003-07-17 2005-01-20 Brian Bidlingmeyer Additives for reversed-phase HPLC mobile phases

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5447922A (en) 1994-08-24 1995-09-05 Bristol-Myers Squibb Company α-phosphonosulfinic squalene synthetase inhibitors
CA2393849A1 (en) 1999-12-22 2001-06-28 Transgenomic, Inc. System and method for automated matched ion polynucleotide chromatography
US7655790B2 (en) * 2002-07-12 2010-02-02 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US7781573B2 (en) * 2006-08-03 2010-08-24 Nam Q Ngo Multi layer chromatography of nucleic acids
DE102007001370A1 (en) * 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2009030254A1 (en) * 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2468048A1 (en) * 1994-02-23 1995-08-31 Pamela Pavco Process for purifying chemically synthesized rna
US20020102563A1 (en) * 1998-04-24 2002-08-01 Gjerde Douglas T. Apparatus and method for separating and purifying polynucleotides
US20050011836A1 (en) * 2003-07-17 2005-01-20 Brian Bidlingmeyer Additives for reversed-phase HPLC mobile phases

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ANDERSON AMY C ET AL: "HPLC purification of RNA for crystallography and NMR", RNA (NEW YORK), vol. 2, no. 2, 1996, pages 110 - 117, XP002474348, ISSN: 1355-8382 *
AZARANI A ET AL: "RNA analysis by ion-pair reversed-phase high performance liquid chromatography.", NUCLEIC ACIDS RESEARCH 15 JAN 2001, vol. 29, no. 2, 15 January 2001 (2001-01-15), pages E7, XP002474349, ISSN: 1362-4962 *
DICKMAN MARK J ET AL: "Enrichment and analysis of RNA centered on ion pair reverse phase methodology", RNA (COLD SPRING HARBOR), vol. 12, no. 4, April 2006 (2006-04-01), pages 691 - 696, XP002474347, ISSN: 1355-8382 *

Cited By (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9402887B2 (en) 2007-10-09 2016-08-02 Curevac Ag Composition for treating prostate cancer (PCa)
US10434154B2 (en) 2007-10-09 2019-10-08 Curevac Ag Composition for treating prostate cancer (PCa)
US9352028B2 (en) 2007-10-09 2016-05-31 Curevac Ag Composition for treating lung cancer, particularly of non-small lung cancers (NSCLC)
US9616084B2 (en) 2009-12-09 2017-04-11 Curevac Ag Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
EP3521429A1 (en) 2013-03-15 2019-08-07 GlaxoSmithKline Biologicals SA Rna purification methods
WO2014140211A1 (en) 2013-03-15 2014-09-18 Novartis Ag Rna purification methods
EP4043032A1 (en) 2013-08-21 2022-08-17 CureVac AG Rabies vaccine
EP3586871A2 (en) 2013-08-21 2020-01-01 CureVac AG Respiratory syncytial virus (rsv) vaccine
EP3450561A1 (en) 2013-08-21 2019-03-06 CureVac AG Method for increasing expression of rna-encoded proteins
EP3461498A1 (en) 2013-08-21 2019-04-03 CureVac AG Rabies vaccine
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015062738A1 (en) 2013-11-01 2015-05-07 Curevac Gmbh Modified rna with decreased immunostimulatory properties
EP3542802A1 (en) 2013-11-01 2019-09-25 CureVac AG Modified rna with decreased immunostimulatory properties
WO2015101416A1 (en) 2013-12-30 2015-07-09 Curevac Gmbh Methods for rna analysis
US10648017B2 (en) 2013-12-30 2020-05-12 Curevac Real Estate Gmbh Methods for RNA analysis
EP3540060A1 (en) 2013-12-30 2019-09-18 CureVac AG Methods for rna analysis
WO2015149944A2 (en) 2014-04-01 2015-10-08 Curevac Gmbh Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
EP3521456A1 (en) 2014-06-10 2019-08-07 CureVac AG Methods and means for enhancing rna production
WO2015188933A1 (en) 2014-06-10 2015-12-17 Curevac Ag Methods and means for enhancing rna production
EP4353257A2 (en) 2015-04-13 2024-04-17 CureVac Manufacturing GmbH Method for producing rna compositions
EP3173092A2 (en) 2015-04-22 2017-05-31 CureVac AG Rna containing composition for treatment of tumor diseases
EP3326641A1 (en) 2015-04-22 2018-05-30 CureVac AG Rna containing composition for treatment of tumor diseases
WO2016170176A1 (en) 2015-04-22 2016-10-27 Curevac Ag Rna containing composition for treatment of tumor diseases
EP3603661A2 (en) 2015-04-22 2020-02-05 CureVac AG Rna containing composition for treatment of tumor diseases
WO2016180430A1 (en) 2015-05-08 2016-11-17 Curevac Ag Method for producing rna
WO2016184822A1 (en) 2015-05-15 2016-11-24 Curevac Ag Prime-boost regimens involving administration of at least one mrna construct
EP3916091A2 (en) 2015-05-20 2021-12-01 CureVac AG Dry powder composition comprising long-chain rna
EP3928800A2 (en) 2015-05-20 2021-12-29 CureVac AG Dry powder composition comprising long-chain rna
EP3744843A1 (en) 2015-05-29 2020-12-02 CureVac Real Estate GmbH A method for producing and purifying rna, comprising at least one step of tangential flow filtration
DE202016009003U1 (en) 2015-05-29 2021-05-28 Curevac Real Estate Gmbh Composition comprising in vitro transcribed RNA obtainable by a method for the production and purification of RNA with at least one step with a tangential flow filtration
US11834651B2 (en) 2015-05-29 2023-12-05 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
EP4108769A1 (en) 2015-05-29 2022-12-28 CureVac Real Estate GmbH A method for producing and purifying rna, comprising at least one step of tangential flow filtration
WO2016193206A1 (en) 2015-05-29 2016-12-08 Curevac Ag A method for producing and purifying rna, comprising at least one step of tangential flow filtration
EP4239080A2 (en) 2015-07-01 2023-09-06 CureVac Manufacturing GmbH Method for analysis of an rna molecule
WO2017001058A1 (en) * 2015-07-01 2017-01-05 Curevac Ag Method for analysis of an rna molecule
EP3736261A1 (en) 2015-09-17 2020-11-11 ModernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
EP4286012A2 (en) 2015-09-17 2023-12-06 ModernaTX, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
EP4086269A1 (en) 2015-10-16 2022-11-09 ModernaTX, Inc. Mrna cap analogs with modified phosphate linkage
WO2017066791A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Sugar substituted mrna cap analogs
WO2017066782A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Hydrophobic mrna cap analogs
WO2017066789A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified sugar
WO2017066793A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs and methods of mrna capping
WO2017066781A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified phosphate linkage
WO2017112865A1 (en) 2015-12-22 2017-06-29 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
EP3701963A1 (en) 2015-12-22 2020-09-02 CureVac AG Method for producing rna molecule compositions
EP4036079A2 (en) 2015-12-22 2022-08-03 ModernaTX, Inc. Compounds and compositions for intracellular delivery of agents
WO2017109161A1 (en) 2015-12-23 2017-06-29 Curevac Ag Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
EP4289965A2 (en) 2016-02-12 2023-12-13 CureVac SE Method for analyzing rna
WO2017140345A1 (en) 2016-02-15 2017-08-24 Curevac Ag Method for analyzing by-products of rna in vitro transcription
WO2017149139A1 (en) 2016-03-03 2017-09-08 Curevac Ag Rna analysis by total hydrolysis
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
WO2017191264A1 (en) 2016-05-04 2017-11-09 Curevac Ag Nucleic acid molecules and uses thereof
WO2017212007A1 (en) 2016-06-09 2017-12-14 Curevac Ag Cationic carriers for nucleic acid delivery
WO2017212009A1 (en) 2016-06-09 2017-12-14 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2017212006A1 (en) 2016-06-09 2017-12-14 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2017218704A1 (en) 2016-06-14 2017-12-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018033254A2 (en) 2016-08-19 2018-02-22 Curevac Ag Rna for cancer therapy
WO2018041921A1 (en) 2016-08-31 2018-03-08 Curevac Ag Mixing device for the production of a liquid nucleic acid composition
US11202793B2 (en) 2016-09-14 2021-12-21 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US10653712B2 (en) 2016-09-14 2020-05-19 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
WO2018078053A1 (en) 2016-10-26 2018-05-03 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018096179A1 (en) 2016-11-28 2018-05-31 Curevac Ag Method for purifying rna
US11279923B2 (en) 2016-11-28 2022-03-22 Curevac Ag Method for purifying RNA
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11384352B2 (en) 2016-12-13 2022-07-12 Modernatx, Inc. RNA affinity purification
WO2018115525A1 (en) 2016-12-23 2018-06-28 Curevac Ag Lassa virus vaccine
WO2018115507A2 (en) 2016-12-23 2018-06-28 Curevac Ag Henipavirus vaccine
WO2018115527A2 (en) 2016-12-23 2018-06-28 Curevac Ag Mers coronavirus vaccine
WO2018141371A1 (en) 2017-01-31 2018-08-09 Curevac Ag Purification and/or formulation of rna
WO2018170336A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Lipid nanoparticle formulation
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
EP4186888A1 (en) 2017-03-15 2023-05-31 ModernaTX, Inc. Compound and compositions for intracellular delivery of therapeutic agents
WO2018167320A1 (en) 2017-03-17 2018-09-20 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
WO2018211038A1 (en) 2017-05-17 2018-11-22 Curevac Ag Method for determining at least one quality parameter of an rna sample
WO2018232120A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
EP3424524A2 (en) 2017-07-04 2019-01-09 CureVac AG Cancer rna-vaccine
WO2019008001A1 (en) 2017-07-04 2019-01-10 Curevac Ag Novel nucleic acid molecules
US11912982B2 (en) 2017-08-18 2024-02-27 Modernatx, Inc. Methods for HPLC analysis
US11866696B2 (en) 2017-08-18 2024-01-09 Modernatx, Inc. Analytical HPLC methods
WO2019036638A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods of preparing modified rna
WO2019038332A1 (en) 2017-08-22 2019-02-28 Curevac Ag Bunyavirales vaccine
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
WO2019092153A1 (en) 2017-11-08 2019-05-16 Curevac Ag Rna sequence adaptation
WO2019115635A1 (en) 2017-12-13 2019-06-20 Curevac Ag Flavivirus vaccine
WO2019193183A2 (en) 2018-04-05 2019-10-10 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
EP4227319A1 (en) 2018-04-17 2023-08-16 CureVac SE Novel rsv rna molecules and compositions for vaccination
WO2020002525A1 (en) 2018-06-27 2020-01-02 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
EP4273266A2 (en) 2018-06-28 2023-11-08 CureVac RNA Printer GmbH Bioreactor for rna in vitro transcription
WO2020002598A1 (en) 2018-06-28 2020-01-02 Curevac Ag Bioreactor for rna in vitro transcription
WO2020061367A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2020061457A1 (en) 2018-09-20 2020-03-26 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020160430A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Vortex mixers and associated methods, systems, and apparatuses thereof
WO2020160397A1 (en) 2019-01-31 2020-08-06 Modernatx, Inc. Methods of preparing lipid nanoparticles
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
WO2021123332A1 (en) 2019-12-20 2021-06-24 Curevac Ag Lipid nanoparticles for delivery of nucleic acids
US11964011B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11964012B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
EP4147717A1 (en) 2020-02-04 2023-03-15 CureVac SE Coronavirus vaccine
DE112021000012T5 (en) 2020-02-04 2021-11-18 Curevac Ag Coronavirus vaccine
DE202021004130U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
DE202021004123U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
DE202021003575U1 (en) 2020-02-04 2022-01-17 Curevac Ag Coronavirus Vaccine
WO2021204175A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Lipid nanoparticle composition
WO2021204179A1 (en) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
WO2021254593A1 (en) 2020-06-15 2021-12-23 Curevac Ag Analysis of nucleic acid mixtures
WO2022002040A1 (en) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
WO2022037652A1 (en) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022049093A1 (en) 2020-09-01 2022-03-10 CureVac RNA Printer GmbH Manufacturing device for a pharmaceutical product
WO2022112498A1 (en) 2020-11-27 2022-06-02 CureVac RNA Printer GmbH A device for preparing a dna product by means of capillary polymerase chain reaction
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2022137133A1 (en) 2020-12-22 2022-06-30 Curevac Ag Rna vaccine against sars-cov-2 variants
US11918643B2 (en) 2020-12-22 2024-03-05 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2022135993A2 (en) 2020-12-22 2022-06-30 Curevac Ag Pharmaceutical composition comprising lipid-based carriers encapsulating rna for multidose administration
WO2022152141A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Polymer conjugated lipid compounds and lipid nanoparticle compositions
WO2022152109A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
WO2022200575A1 (en) 2021-03-26 2022-09-29 Glaxosmithkline Biologicals Sa Immunogenic compositions
WO2022207862A2 (en) 2021-03-31 2022-10-06 Curevac Ag Syringes containing pharmaceutical compositions comprising rna
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2022240960A1 (en) * 2021-05-14 2022-11-17 Crispr Therapeutics Ag Mrna large scale synthesis and purification
WO2022247755A1 (en) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023007019A1 (en) 2021-07-30 2023-02-02 CureVac SE Cap analogs having an acyclic linker to the guanine derivative nucleobase
WO2023006999A2 (en) 2021-07-30 2023-02-02 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023031392A2 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids comprising phosphatidylserine
WO2023044343A1 (en) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Acyclic lipids and methods of use thereof
WO2023044333A1 (en) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Cyclic lipids and methods of use thereof
EP4162950A1 (en) 2021-10-08 2023-04-12 Suzhou Abogen Biosciences Co., Ltd. Nucleic acid vaccines for coronavirus
WO2023056917A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023056914A1 (en) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023122752A1 (en) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Constrained lipids and methods of use thereof
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023196931A1 (en) 2022-04-07 2023-10-12 Renagade Therapeutics Management Inc. Cyclic lipids and lipid nanoparticles (lnp) for the delivery of nucleic acids or peptides for use in vaccinating against infectious agents
WO2023196634A2 (en) 2022-04-08 2023-10-12 Flagship Pioneering Innovations Vii, Llc Vaccines and related methods
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
WO2024030856A2 (en) 2022-08-01 2024-02-08 Flagship Pioneering Innovations Vii, Llc Immunomodulatory proteins and related methods
WO2024037578A1 (en) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition of lipid nanoparticles
WO2024068545A1 (en) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Influenza virus vaccines
DE202023106198U1 (en) 2022-10-28 2024-03-21 CureVac SE Nucleic acid-based vaccine
WO2024089229A1 (en) 2022-10-28 2024-05-02 CureVac SE Improved formulations comprising lipid-based carriers encapsulating rna
WO2024089638A1 (en) 2022-10-28 2024-05-02 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine

Also Published As

Publication number Publication date
DK2092064T3 (en) 2010-12-13
AU2007338360A1 (en) 2008-07-03
AU2007338360B2 (en) 2011-06-02
ATE481482T1 (en) 2010-10-15
PL2092064T3 (en) 2011-03-31
JP2010512767A (en) 2010-04-30
CA2670727A1 (en) 2008-07-03
KR20090098968A (en) 2009-09-18
JP5307724B2 (en) 2013-10-02
US8383340B2 (en) 2013-02-26
US20100048883A1 (en) 2010-02-25
CN101563457B (en) 2014-03-12
DE102006061015A1 (en) 2008-06-26
ES2352306T3 (en) 2011-02-17
EP2092064B1 (en) 2010-09-15
CN101563457A (en) 2009-10-21
CA2670727C (en) 2012-10-09
EP2092064A1 (en) 2009-08-26
KR101183173B1 (en) 2012-09-14
DE602007009295D1 (en) 2010-10-28

Similar Documents

Publication Publication Date Title
US8383340B2 (en) Method for purifying RNA on a preparative scale by means of HPLC
Eon-Duval et al. Purification of pharmaceutical-grade plasmid DNA by anion-exchange chromatography in an RNase-free process
Ghanem et al. Current trends in separation of plasmid DNA vaccines: a review
Tian et al. Evaluation of silica resins for direct and efficient extraction of DNA from complex biological matrices in a miniaturized format
Smrekar et al. Preparation of pharmaceutical-grade plasmid DNA using methacrylate monolithic columns
Soares et al. Purification of human papillomavirus 16 E6/E7 plasmid deoxyribonucleic acid-based vaccine using an arginine modified monolithic support
JP3714940B2 (en) RNA extraction method and biomaterial analysis method
US7510826B2 (en) Process for the depletion or removal of endotoxins
Hanora et al. Direct capture of plasmid DNA from non-clarified bacterial lysate using polycation-grafted monoliths
JPH09508283A (en) Enhanced transfection efficiency of nucleic acids using isopropanol-containing aqueous solution
Poeckh et al. Adsorption and elution characteristics of nucleic acids on silica surfaces and their use in designing a miniaturized purification unit
WO2008063717A2 (en) Pharmaceutical manufacturing methods
CA2245576C (en) Process for preparing purified nucleic acid and the use thereof
Santos et al. Influenza DNA vaccine purification using pHEMA cryogel support
Krajnc et al. Purification of large plasmids with methacrylate monolithic columns
Spector et al. Polyadenylic acid on poliovirus RNA: IV. Poly (U) in replicative intermediate and double-stranded RNA
Bonturi et al. Sodium citrate and potassium phosphate as alternative adsorption buffers in hydrophobic and aromatic thiophilic chromatographic purification of plasmid DNA from neutralized lysate
Corradini et al. Combined lectin-affinity and metal-interaction chromatography for the separation of glycophorins by high-performance liquid chromatography
AU2005209389A1 (en) Method for chromatographic separation of a nucleic acid mixture
JP4125605B2 (en) Purification of plasmid DNA with hydroxyapatite without acetic acid
Hayakawa et al. Evaluation of the electroosmotic medium pump system for preparative disk gel electrophoresis
AU2003206358B2 (en) Method of separation using aromatic thioether ligands
US20090047734A1 (en) Method of separation of deoxyribonucleic acids
AU2002308753A1 (en) Acetate-free purification of plasmid DNA on hydroxyapatite
JP4073447B2 (en) RNA extraction method, RNA extraction reagent, and biomaterial analysis method

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780047133.2

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07857017

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2007857017

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2670727

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 3565/DELNP/2009

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007338360

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2009541898

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020097012957

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007338360

Country of ref document: AU

Date of ref document: 20071220

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12520172

Country of ref document: US