WO2009127230A1 - MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION - Google Patents

MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION Download PDF

Info

Publication number
WO2009127230A1
WO2009127230A1 PCT/EP2008/003033 EP2008003033W WO2009127230A1 WO 2009127230 A1 WO2009127230 A1 WO 2009127230A1 EP 2008003033 W EP2008003033 W EP 2008003033W WO 2009127230 A1 WO2009127230 A1 WO 2009127230A1
Authority
WO
WIPO (PCT)
Prior art keywords
syndrome
deficiency
type
rna
disease
Prior art date
Application number
PCT/EP2008/003033
Other languages
French (fr)
Inventor
Andreas Thess
Jochen Probst
Ingmar Hoerr
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
Application filed by Curevac Gmbh filed Critical Curevac Gmbh
Priority to PCT/EP2008/003033 priority Critical patent/WO2009127230A1/en
Publication of WO2009127230A1 publication Critical patent/WO2009127230A1/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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/17Immunomodulatory nucleic acids
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • C12N2320/53Methods for regulating/modulating their activity reducing unwanted side-effects

Definitions

  • the present invention relates to a modified (m)RNA suitable for suppressing and/or avoiding an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA and an immunosuppressive composition comprising this RNA.
  • the invention furthermore relates to a pharmaceutical composition containing said modified (m)RNA.
  • the invention also relates to the use of said modified (m)RNA or immunosuppressive composition (for the preparation of a medicament, e.g. a pharmaceutical composition) and/or the use of the pharmaceutical composition for suppressing and/or avoiding an immune response in a mammal when administering said pharmaceutical composition for the treatment of various diseases.
  • the invention relates to kits containing the immunosuppressive composition and/or the pharmaceutical composition.
  • the innate immune system as part of the immune system is the dominant system of host defense in most organisms and comprises barriers such as humoral and chemical barriers including, e.g., inflammation, the complement system and cellular barriers.
  • the innate immune system is typically based on a small number of receptors, called pattern recognition receptors.
  • Such pathogen-associated molecular patterns include viral nucleic acids, components of bacterial and fungal walls, flagellar proteins, and more.
  • TLR Toll-like receptor
  • TLRs 1 - 10 Toll-like receptors
  • TLRs Toll-like receptors in human include TLR1 -TLR2 (known ligand: Triacyl lipopeptide), TLRl - TLR6 (known ligand: Diacyl lipopeptide), TLR2 (known ligand: Peptidoglycan), TLR3 (known ligand: dsRNA), TLR4 (known ligand: LPS (lipopolysaccharide) of Gram-negative bacteria)), TLR5 (known ligand: bacterial flagellin(s)), TLR7/8 (known ligands: imidazoquinolines, guanosine analogs and ssRNA), TLR9 (known ligands: CpG DNA of bacteria, viruses and protozoans and malaria pigment hemozoin (product of digestion of haemoglobin)) and TLRIO.
  • TLR1 -TLR2 known ligand: Triacyl lipopeptide
  • TLRl - TLR6 known ligand: Diacyl lipopeptide
  • TLRs After recognition of microbial pathogens, these TLRs typically trigger intracellular signalling pathways that result in induction of inflammatory cytokines (e.g. TNF-alpha, IL-6, IL-1 -beta and IL-12), type I interferon (IFN-beta and multiple IFN- alpha) and chemokines (Kawai, T. and S. Akira (2006). "TLR signaling.” Cell Death Differ 13(5): 816-25).
  • inflammatory cytokines e.g. TNF-alpha, IL-6, IL-1 -beta and IL-12
  • IFN-beta and multiple IFN- alpha type I interferon
  • chemokines Yamanes, T. and S. Akira (2006). "TLR signaling.” Cell Death Differ 13(5): 816-25).
  • the innate immune system Apart from the adaptive immune system, which adapted over time to recognize particular pathogens or antigens more efficiently, the innate immune system and accordingly the ability of therapeutic agents to trigger an adequate but non-excessive innate immune response represents is a crucial key element with respect to the compatibility of many treatments. In some cases even a therapeutic agent may be desired, which allows to suppress an innate immune response or diminish it up to a certain extent.
  • a suitable therapeutic agent is dependent on the disease to be treated. Hitherto, conventional methods of treatment may, inter alia, comprise the use of nucleic acid sequences, such as DNA or RNA.
  • Nucleic acid sequences represent ligands of the Toll-like receptor (TLR) familiy as defined above, particularly of Toll-like receptors TLR3 (known ligand: dsRNA), TLR7/8 (known ligands: imidazoquinolines, guanosine analogs and ssRNA), and TLR9 (known ligands: CpG DNA of bacteria, viruses and protozoans and malaria pigment hemozoin (product of digestion of haemoglobin)).
  • TLR3 known ligand: dsRNA
  • TLR7/8 known ligands: imidazoquinolines, guanosine analogs and ssRNA
  • TLR9 known ligands: CpG DNA of bacteria, viruses and protozoans and malaria pigment hemozoin (product of digestion of haemoglobin)
  • nucleic acid sequences such as DNA or RNA are used to incorporate required genetic information into the cell, particularly in gene therapeutic method of treatments, whereby those nucleic acids
  • DNA viruses may likewise be used as a DNA vehicle. Such viruses achieve a very high transfection rate, because of their infectious properties.
  • the viruses used are typically genetically modified in such a manner that no functional infectious particles are formed in the transfected cell. Despite these precautions, however, it is not possible to rule out the risk of uncontrolled propagation of the introduced gene. The same risk generally also arises, when using DNA, even if not derived from DNA viruses, e.g. due to uncontrolled recombination events of the introduced gene.
  • the gene may be also affected by inactivation of essential elements thereof, such as promoters, enhancers or silencers, which lead to a different expression pattern, e.g. a silencing of the entire gene, wherein one particular risk occurs if the DNA is integrated into a gene which is involved in the regulation of cell growth.
  • RNA expression systems may have considerable advantages over DNA expression systems, e.g., in immune response, immunization or vaccination. These advantages include, inter alia, that RNA introduced into a cell is not integrated into the genome. Another advantage includes that no viral sequences, such as promoters etc., are required for active transcription, when RNA is used as a coding nucleic acid. Another risk of using DNA as an agent to induce an immune response (e.g.
  • RNA sequences as well as DNA sequences may sometimes lead to excessive immune responses or even to toxicities associated with excessive cytokine release and associated inflammatory syndromes.
  • therapeutic agents may be required, which are neutral, i.e. do not trigger an innate (and also no adaptive) immune response, or even immunosuppressive, i.e. lower an existing immune response of the innate immune system.
  • RNA sequences may be the avoidance of any adjuvant, when administering the RNA sequence, which triggers an innate immune response.
  • transfecting naked RNA sequences typically does not abolish the intrinsic properties of RNA per se to trigger an immune response due to its recognition by Toll-like receptors 3, 7 and 8.
  • the immune stimulation is usually already triggered upon transfection of the RNA into the organism, e.g. in the endosom upon endocytosis. Accordingly, the intrinsic properties of RNA may be changed on molecular level.
  • RNA may be dependent or at least strongly influenced by specific molecular properties of the RNA (or DNA), such as structure, binding capacity, content of CpGs, (post- transcriptional) modification of nucleotides, etc..
  • specific molecular properties of the RNA (or DNA) such as structure, binding capacity, content of CpGs, (post- transcriptional) modification of nucleotides, etc.
  • Another alternative is to alter the modification rate of the nucleosides of the RNA, in order to change immunomodulatory properties of the RNA to be administered.
  • RNA sequences by using an RNA comprising N6-methyl-adenosine, 2-thiouridine, 5-methyl- cytidine, 5-methyl-uridine or pseudouridines.
  • Kariko eta/. (2005, supra) stressed that the role of nucleoside modifications on the immuno-stimulatory potential and on the translation efficiency of RNA, however, is not known. Accordingly, there appears to be no common approach, to provide nucleic acid modifications, which both confer a lowered immunostimulatory property and allow translation of the nucleic acid molecule in vivo. However, this is particularly necessary for the use of longer RNA sequences, such as coding RNA, e.g. mRNA, sequences.
  • WO 2008/019486 discloses nucleic acids with 2'-O-methyl modifications at uridine, guanosine and/or adenosine residues, which confer a lowered immunostimulatory property with respect to the unmodified nucleic acid.
  • WO 2008/019486 discloses short nucleic acid sequences, which are administered in combination with (short) antisense oligonucleotides or siRNAs to confer the lowered immunostimulatory properties.
  • WO 2008/019486 does not disclose the use of longer RNA sequences, such as coding RNA, e.g. mRNA, sequences or the translation of any such oligonucleotide.
  • siRNAs small interfering RNAs
  • Sioud et al. (Eur. J. Immunol., 2006, 36: 1222-1230) also report on chemical modifications, wherein those modifications are carried out on small interfering RNAs (siRNAs).
  • siRNAs small interfering RNAs
  • Sioud et al. (2006, supra) report, that replacement of 2'-hydroxyl uridines with either 2'-fluoro, 2'-deoxy or 2'-O-methyl-uridines is capable to abrogate the immune reaction of siRNAs.
  • siRNAs are typically short double stranded nucleic acid molecules having a length of about 21 to 25 nucleotides.
  • siRNAs are intended to silence gene expression by binding to specific target sequences and thus, by their very nature, those sequences do not have to encode a protein sequence or have to show any capability for translation.
  • WO 2006/009784 also discloses chemical modifications of siRNA molecules, wherein the si RNA strands essentially consist of ribonucleotides with 2'-hydroxyl substitutions on the ribonucleotide ribose.
  • WO 2006/009784 does not disclose other modifications, which may modify the immune response or discusses such modifications with longer RNA molecules, such as e.g. coding RNA, e.g. mRNA, molecules.
  • WO 2007/031877 discloses modifications of siRNAs.
  • WO 2007/031877 describes replacement of nucleotides by at least one nucleotide having a sugar with a 2'- modification, wherein the modified nucleotide shall not be a locked nucleotide or a 2'-O- methyl nucleotide.
  • WO 2007/031877 discloses in detail the use of 2'-modifications selected from 2'-deoxy, 2'-fluoro, 2'-amino, 2'-methoxyethyl, 2'-O-allyl, 2'-propinyl, 2'- aminopropargyl, 2'-O-(3-aminopropyl), 2'-O-propyl, 2'-O-butyl, 2'-O-alkenyl, 2'-O-alkinyl, 2'-methoxyethyl, 2'-aminopropargyl, 2'-O-(3-aminopropyl) and 2'-amino.
  • WO 2007/031877 focusses on siRNA, but does not give any indication as to whether such modifications are suitable for translation of longer RNA molecules or provides such specific modifications.
  • modified nucleotides per se do not necessarily trigger or inhibit an immune response, as inhibiting or lowering the immune response by modification of nucleotides appears to be strongly dependent on the nature of the single modified nucleotide.
  • modifications of nucleotides per se do also not appear to provide an overall solution to the above problem. Accordingly, there is still the need in the art for providing an RNA, preferably a longer RNA such as a coding RNA, e.g. a mRNA, sequence, which is capable to confer both a lowered immunostimulatory property with respect to the unmodified RNA and allows translation of the RNA molecule in vivo.
  • a modified (m)RNA or an immunossuppressive composition comprising at least one such modified (m)RNA, wherein at least one nucleoside of the modified (m)RNA has been modified with: a) a chemical modification at the 4-, 5-or 6-position of the pyrimidine base of the nucleosides of cytidine and/or uridine; b) a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides of adenosine, inosine and/or guanosine; and/or c) a chemical modification at the 2'-position of the sugar of the nucleosides of adenosine, inosine, guanosine, cytidine and/or uridine, wherein the at least one modified (m)RNA is suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administer
  • An immunosuppressive composition according to the present invention shall be understood as a composition, which is capable to suppress and/or avoid an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, preferably an innate immune response as defined herein, due to the at least one modified (m)RNA as contained in the inventive immunosuppressive composition.
  • a mammal as mentioned throughout the description of the present invention may be selected from any mammal, preferably from a mammal, selected from the group comprising, without being limited thereto, e.g. goat, cattle, swine, dog, cat, donkey, monkey, ape, a rodent such as a mouse, hamster, rabbit, and, in particular, human.
  • an (m)RNA is a nucleic acid chain formed by a number of nucleotides typically selected from adenosine-5'-monophosphate, guanosine-5'-monophosphate, inosine-5'-monophosphate, cytidine-5'-monophosphate and/or uridine-5'-monophosphate. Those nucleotides are linked to each other via their monophosphate. Nucleotides comprise nucleosides and a 5'-monophosphate as a structural component, wherein the nucleosides are typically formed by a nucleobase, i.e.
  • a modification of a nucleoside of the modified (m)RNA is always intended to mean a modification in the nucleoside structure of the respective nucleotide of said (m)RNA.
  • At least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response typically exhibited when administering the corresponding unmodified (m)RNA in a mammal may be modified with a chemical modification at the 5- or 6-position of the pyrimidine base of the nucleosides cytidine and/or uridine.
  • a chemical modification at the 4-, 5- or 6-position of the base pyrimidine of the nucleosides cytidine and/or uridine may be selected from the group consisting of:
  • At least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA may be alternatively modified with a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine.
  • such chemical modifications at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine may be selected from the group consisting of:
  • innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA may be modified with at least one chemical modification at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine, when incorporated in the RNA sequence.
  • chemical modifications at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine may be selected from the group consisting of:
  • At least one nucleoside of the modified (m)RNA (of the inventive immunosuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA has been modified at the 4,- 5- or 6- position of the base pyrimidine of the nucleosides cytidine and/or uridine and at the T- position of the ribose sugar according to modifications a) and c) as defined above, more preferably as shown in the following:
  • At least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA has been modified at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine and at the 2'-position of the ribose sugar according to modifications b) and c) as defined above, more preferably as shown in the following:
  • At least one nucleoside of the modified (m)RNA suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, has been modified leading to chemically modified nucleotides (of the (m)RNA) selected from the following group: 4-thio-uridine-5'- (mono)phosphate, 2-Aminopurine-riboside-5'-(mono)phosphate, 5-Aminoallylcytidine-5'- (mono)phosphate , 5-Aminoallyluridine-5'-(mono)phosphate , 5-Bromocytidine-5'- (mono)phosphate, 5-Bromo-2'-deoxycytidine-5'-(mono)phosphate, 5-Bromouridine-5'-
  • At least one nucleoside of the modified (m)RNA has been modified leading to chemically modified nucleotides selected from the following group: 4-thio-uridine-5'-(mono)phosphate, 5-Aminoallyluridine- 5'-(mono)phosphate, 5-Bromo-2'-deoxycytidine-5'-(mono)phosphate, 5-Bromouridine-5'- (mono)phosphate, 5-Bromo-2'-deoxyuridine-5'-(mono)phosphate, 5-lodouridine-5'- (mono)phosphate, 5-lodo-2'-deoxyuridine-5'-(mono)phosphate, 5-Propynyl-2'- deoxyuridine-5'-(mono)phosphate, 6-Azacytidine-5'-(mono)phosphate, 8-Azidoadenosine- 5'-
  • chemically modified nucleotides 5- methylcytidine-5'-(mono)phosphate, 5-methyluridine-5'-(mono)phosphate do not form part of the present disclosure and are explicitely disclaimed herewith from the at least one modified (m)RNA according to the present invention and the inventive immunosuppressive composition.
  • the at least one modified (m)RNA comprises more than one chemically modified nucleoside selected from the chemically modified nucleosides as defined above, more preferably at least two, three or four of these chemically modified nucleosides, or even more.
  • any natively occurring nucleoside which is contained in the natively occurring template (m)RNA of the at least one modified (m)RNA (of the inventive immunosuppressive composition) and which is to be substituted in the context of the present invention with a chemically modified nucleoside as defined above, preferably substitutes a corresponding nucleoside, i.e.
  • adenosine of a natively occurring AMP (adenosine-5'-(mono)phosphate) will be substituted with a chemically modified adenosine as defined above (e.g. leading to 8-Azidoadenosine-5'-(mono)phosphate, etc.)
  • cytidine of a natively occurring CMP (cytidine-5'-(mono)phosphate) will be substituted with a chemically modified cytidine as defined above (e.g.
  • guanosine of a natively occurring GMP guanosine-5'- (mono)phosphate
  • a chemically modified guanosine as defined above e.g. leading to 7-Deazaguanosine-5'-(mono)phosphate, etc.
  • uridine of a natively occurring UMP uridine-5'-(mono)phosphate
  • a chemically modified uridine as defined above e.g. leading to 5-Aminoallyluridine-5'- (mono)phosphate, etc.
  • TMP e.g.
  • the at least one modified (m)RNA comprises between 0.1 % and 100% chemically modified nucleotides as defined above, wherein more preferably between 0.1 % and 100% of each natively occurring non-modified adenosine, guanosine, uridine and/or cytidine nucleoside, respectively, of the corresponding non-modified (m)RNA (template) may be modified using any of the corresponding chemically modified adenosine, guanosine, uridine and/or cytidine nucleosides, respectively, as defined above.
  • adenosine, guanosine, uridine and/or cytidine nucleosides natively occurring in the corresponding non-modified (m)RNA (template) are modified in the at least one modified (m)RNA (of the inventive immunosuppressive composition) using the above defined (corresponding) chemically modified adenosine, guanosine, uridine and/or cytidine nucleoside, respectively, in a range of between 0,1 % and 20%, between 10% and 30%, between 20% and 40%, between 30% and 50%, between 40% and 60%, between 50% and 70%, between 60% and 80%, between 70% and 90%, or between 80% and 100% or at least 10%, more preferably at least 30%, more preferably at least 40%, more preferably at least 60%, more preferably at least 70%, more preferably at least 80% and more preferably at least 90% and most preferably 100% of all natively occurring adenosine, guanosine, uridine and
  • the above selection may be applied to the entire sequence as well as to the content of the single adenosine, guanosine, uridine and/or cytidine nucleosides, respectively, of the modified (m)RNA sequence, i.e. preferably between 0.1 % and 100% in the above ranges of the adenosine, guanosine, uridine and/or cytidine nucleosides, respectively, natively occurring in the corresponding non-modified (m)RNA template sequence are chemically modified in the at least one modified (m)RNA (of the inventive immunosuppressive composition) using the above defined chemically modified nucleosides.
  • the modified (m)RNA may exlusively contain chemically modified adenosine, guanosine, uridine and/or cytidine nucleosides as defined above in the above ranges or a mixture thereof in the above ranges.
  • RNA sequences In order to determine the percentage to which two (modified and/or natively occurring) RNA sequences (nucleic or amino acid) are identical, the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. gaps can be inserted into the sequence of the first (modified) RNA sequence and the component at the corresponding position of the second (modified) RNA sequence can be compared. If a position in the first (modified) RNA sequence is occupied by the same component as is the case at a position in the second (modified) RNA sequence, the two sequences are identical at this position.
  • the percentage to which two (modified) RNA sequences are identical is a function of the number of identical positions divided by the total number of positions. The same, of course also applies accordingly to amino acid sequences encoded by these RNA sequences.
  • the percentage to which two sequences (either amino or nucleic acid sequences) are identical can be determined using a mathematical algorithm.
  • a preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et a/. (1993), PNAS USA, 90:5873-5877 or Altschul et a/. (1997), Nucleic Acids Res, 25:3389- 3402. Such an algorithm is integrated in the BLAST or NBLAST program. Sequences which are identical to the sequences of the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) (or to the coding region thereof) to a certain extent can be identified by these programmes.
  • RNA sequences (corresponding to the at least one modified (m)RNA of the inventive immunosuppressive composition) encoding amino acid sequences which have (a) conservative substitution(s) compared to the physiological sequence in particular fall under the term variants.
  • Substitutions in which encoded amino acids which originate from the same class are exchanged for one another are called conservative substitutions and are encompassed herein.
  • these are encoded amino acids, encoded aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or encoded amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function. This means that e.g.
  • an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g. serine (threonine) by threonine (serine) or leucine (isoleucine) by isoleucine (leucine)).
  • Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g.
  • CD spectra circular dichroism spectra
  • the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be any RNA, preferably, without being limited thereto, a short RNA oligonucleotide, a coding RNA, a siRNA, an antisense RNA, or riboswitches, ribozymes or aptamers.
  • the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a single- or a double-stranded RNA (which may also be regarded as an RNA (molecule) due to non-covalent association of two single- stranded RNA (molecules)) or a partially double-stranded RNA (which is typically formed by a longer and a shorter single-stranded RNA molecule or by two single stranded RNA- molecules, which are about equal in length, wherein one single-stranded RNA molecule is in part complementary to the other single-stranded RNA molecule and both thus form a double-stranded RNA molecule in this region).
  • a single- or a double-stranded RNA which may also be regarded as an RNA (molecule) due to non-covalent association of two single- stranded RNA (molecules)
  • a partially double-stranded RNA which is typically formed by a longer and a shorter single-stranded
  • the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a single- stranded RNA.
  • the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a circular or linear RNA, preferably a linear RNA. More preferably, the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a (linear) single-stranded RNA.
  • the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a ribosomal RNA (rRNA), a transfer RNA (tRNA), a messenger RNA (mRNA), or a viral RNA (vRNA), more preferably an mRNA.
  • rRNA ribosomal RNA
  • tRNA transfer RNA
  • mRNA messenger RNA
  • vRNA viral RNA
  • an mRNA is typically an RNA, which is composed of several structural elements, e.g. an optional 5'-UTR region, an upstream positioned ribosomal binding site followed by a coding region, an optional 3'-UTR region, which may be followed by a poly-A tail (and/or a poly-C-tail).
  • An mRNA may occur as a mono-, di-, or even multicistronic RNA, i.e. an RNA which carries the coding sequences of one, two or more proteins.
  • Such coding sequences in di-, or even multicistronic mRNA may be separated by at least one IRES sequence, e.g. as defined herein.
  • an inventive modified (m)RNA as defined herein, comprises a length of about 5 to about 20000, or 100 to about 20000 nucleotides, preferably of about 250 to about 20000 nucleotides, more preferably of about 500 to about 10000, even more preferably of about 500 to about 5000.
  • the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a coding RNA.
  • a coding RNA may be any RNA as defined above.
  • such a coding RNA may be a single- or a double-stranded RNA, more preferably a single-stranded RNA, and/or a circular or linear RNA, more preferably a linear RNA.
  • the coding RNA may be a (linear) single-stranded RNA.
  • the coding RNA may be a ((linear) single-stranded) messenger RNA (mRNA).
  • the coding RNA used as the inventive at least one modified (m)RNA (of the immunosuppressive composition) according to the present invention, may encode a protein or a peptide, which may be selected, without being restricted thereto, e.g. from therapeutically active proteins or peptides, antibodies, antigens, allergens, etc., from epitopes therof, i.e.
  • the at least one modified coding RNA (molecule) of the inventive immunosuppressive composition is to be transported into a cell, a tissue or an organism and the protein is expressed subsequently in this cell, tissue or organism.
  • therapeutically active proteins encoded by the at least one modified (m)RNA (of the immunosuppressive composition) according to the present invention may be selected from any recombinant or isolated proteins known to a skilled person from the prior art. Without being restricted thereto therapeutically active proteins may comprise proteins, capable of stimulating or inhibiting the signal transduction in the cell,
  • Therapeutically active proteins as encoded by the at least one modified (m)RNA may be selected from apoptotic factors or apoptosis related proteins including AIF, Apaf e.g. Apaf-1 , Apaf-2, Apaf-3, oder APO-2
  • Therapeutically active proteins as encoded by the at least one modified (m)RNA may also be selected from recombinant proteins, including proteins selected from the group consisting of 0ATL3, 0FC3, 0PA3, 0PD2, 4-1 BBL, 5T4, 6Ckine, 707-AP, 9D7, A2M, AA, AAAS, AACT, AASS, ABAT, ABCA1, ABCA4, ABCBI, ABCBIl, ABCB2, ABCB4, ABCB7, ABCC2, ABCC6, ABCC8, ABCD1, ABCD3, ABCG5, ABCG8, ABL1, ABO, ABR ACAA1, ACACA, ACADL, ACADM, ACADS, ACADVL, ACAT1, ACCPN, ACE, ACHE, ACHM3, ACHM1, ACLS, ACPI, ACTA1, ACTC, ACTN4, ACVRL1, AD2, ADA, ADAMTS13, ADAMTS2, ADFN, ADH 1 B,
  • CRYGD CSA, CSE, CSF1R, CSF2RA, CSF2RB, CSF3R, CSF1R, CST3, CSTB, CT, CT7, CT- 9/BRD6, CTAA1, CTACK, CTEN, CTH, CTHM, CTLA4, CTM, CTNNB1, CTNS, CTPA, CTSB, CTSC, CTSK, CTSL, CTS1, CUBN, CVD1, CX3CL1, CXCL1, CXCL10, CXCLl 1, CXCL12, CXCL13, CXCL16, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, CYB5, CYBA, CYBB, CYBB5, , CYFRA 21-1, CYLD, CYLD1, CYMD, CYP11B1,
  • CYP11B2 CYP17, CYP17A1, CYP19, CYP19A1, CYP1A2, CYP1B1, CYP21A2, CYP27A1, CYP27B1, CYP2A6, CYP2C, CYP2C19, CYP2C9, CYP2D, CYP2D6, CYP2D7P1, CYP3A4, CYP7B1, CYPB1, CYP11B1, CYP1A1, CYP1B1, CYRAA, D40,DADl, DAM, DAM-10/MAGE-B1, DAM-6/MAGE-B2, DAX1, DAZ, DBA, DBH, DBI, DBT, DCC, DC- CK1, DCK, DCR, DCX, DDB 1, DDB2, DDIT3, DDU, DECR1, DEK-CAN, DEM, DES,
  • GPI GPI, GP1BA, GPNlLW, GPNMB/m, GPSC, GPX1, GRHPR, GRK1, GRO, GRO, GRO, GRPR, GSE, GSM1, GSN, GSR, GSS, GTD, GTS, GUCA1A, GUCY2D, GULOP, GUSB, GUSM, GUST, GYPA, GYPC, GYSl, GYS2, H0KPP2, H0MG2, HADHA, HADHB, HAGE, HAGH, HAL, HAST-2, HB 1, HBA2, HBAl, HBB, HBBP1, HBD, HBE1, HBG2, HBGl, HBQl, HBZ, HBZP, HCA, HCC-I, HCC-4, HCF2, HCG, HCL2,
  • MELANA MELAS, MENl MSLN, MET, MF4, MG50, MG50/PXDN, MGAT2, MGAT5, MGC1 MGCR, MGCT, MGI, MGP, MHC2TA, MHS2, MHS4, MIC2, MIC5, MIDI, MIF, MIP, MIP-5/HCC-2, MITF, MJD, MKI67, MKKS, MKS1, MLHI, MLL, MLLT2, MLLT3, MLLT7, MLLT1, MLS, MLYCD, MMAIa, MMP 11, MMVP1, MN/CA IX-Antigen, MNG1, MN1, MOC31, MOCS2, MOCS1, MOG, MORC, MOS, MOV18, MPD1, MPE, MPFD, MPI, MPIF-1, MPL, MPO, MPS3C, MPZ, MRE11A, MROS, MRP1, MRP2, MRP3, MRSD, MRX14, MRX2,
  • PCBD PCBD, PCCA, PCCB, PCK2, PCK1, PCLD, PCOSI, PCSK1, PDB1, PDCN, PDE6A, PDE6B, PDEF, PDGFB, PDGFR, PDGFRL, PDHA1, PDR, PDX1, PECAMI, PEE1, PEO1, PEPD, PEX10, PEX12, PEX13, PEX3, PEX5, PEX6, PEX7, PEX1, PF4, PFBI, PFC, PFKFB1, PFKM, PGAM2, PGD, PGK1, PGK1P1, PGL2, PGR, PGS, PHA2A, PHB, PHEX, PHGDH, PHKA2, PHKA1, PHKB, PHKG2, PHP, PHYH, Pl, PI3, PIGA, PIM1 -KINASE, PINI,
  • PIP5K1B PITX2, PITX3, PKD2, PKD3, PKD1, PKDTS, PKHD1, PKLR, PKP1, PKU1, PLA2G2A, PLA2G7, PLAT, PLEC1, PLG, PLI, PLOD, PLP1, PMEL17, PML, PML/RAR ⁇ , PMM2, PMP22, PMS2, PMS1, PNKD, PNLIP, POF1, POLA, POLH, POMC, PON2, PON1, PORC, POTE, POU1F1, POU3F4, POU4F3, POU1F1, PPAC, PPARG, PPCD, PPGB, PPH1, PPKB, PPMX, PPOX, PPP1R3A, PPP2R2B, PPTI, PRAME, PRB, PRB3, PRCA1, PRCC, PRD, PRDX5/m, PRF1, PRG4, PRKAR1A, PRKCA, PRKDC, PRKWNK4, PR
  • REG1A REHOBE, REN, RENBP, RENS1, RET, RFX5, RFXANK, RFXAP, RGR, RHAG, RHAMM/CD168, RHD, RHO, Rip-1, RLBP1, RLN2, RLN1, RLS, RMD1, RMRP, ROM1, ROR2, RP, RP1, RP14, RP17, RP2, RP6, RP9, RPDl, RPE65, RPGR, RPGRIP1, RP1, RP10, RPS19, RPS2, RPS4X, RPS4Y, RPS6KA3, RRAS2, RS1, RSN, RSS, RU1, RU2, RUNX2,RUNXI, RWS, RYR1, S-100, SAA1, SACS, SAG, SAGE, SALL1, SARDH, SART1,
  • SART2 SART3, SAS, SAX1, SCA2, SCA4, SCA5, SCA7, SCA8, SCA1, SCC, SCCD, SCF, SCLC1, SCN1A, SCN1B, SCN4A, SCN5A, SCNNlA, SCNN1B, SCNN1G, SCO2, SCP1, SCZD2, SCZD3, SCZD4, SCZD6, SCZD1, SDF-1 ⁇ / ⁇ SDHA, SDHD, SDYS, SEDL, SERPENA7, SERPINA3, SERPINA6, SERPINA1, SERPINC1, SERPIND1, SERPINE1, SERPINF2, SERPING1, SERPINM, SFTPAl, SFTPB, SFTPC, SFTPD, SGCA, SGCB, SGCD,
  • VDR VDR, VEGF, VEGFR-2, VEGFR-1, VEGFR-2/FLK-1 , VHL, VIM, VMD2, VMD1 , VMGLOM, VNEZ, VNF, VP, VRNI, VWF, VWS, WAS, WBS2, WFS2, WFSl, WHCR, WHN, WISP3, WMS, WRN, WS2A, WS2B, WSN, WSS, WT2, WT3, WT1 , WTS, WWS, XAGE, XDH, XIC, XIST, XK, XM, XPA, XPC, XRCC9, XS, ZAP70, ZFHX1 B, ZFX, ZFY, ZIC2, ZIC3, ZNF145, ZNF261 , ZNF35, ZNF41 , ZNF6, ZNF198, and ZWS1.
  • therapeutically active proteins as encoded by the at least one modified (m)RNA may also be selected from growth hormones or growth factors, for example for promoting growth in a (transgenic) living being, such as, for example, TGF ⁇ and the IGFs (insulin- like growth factors), proteins that influence the metabolism and/or haematopoiesis, such as, for example, ⁇ - anti-trypsin, LDL receptor, erythropoietin (EPO), insulin, GATA-I, etc., or proteins such as, for example, factors VIII and Xl of the blood coagulation system, etc.
  • growth hormones or growth factors for example for promoting growth in a (transgenic) living being, such as, for example, TGF ⁇ and the IGFs (insulin- like growth factors), proteins that influence the metabolism and/or haematopoiesis, such as, for example, ⁇ - anti-trypsin, LDL receptor, erythropoietin (EPO), insulin, GATA-I,
  • Such proteins further include enzymes, such as, for example, ⁇ -galactosidase (lacZ), DNA restriction enzymes (e.g. EcoRI, Hindlll, etc.), lysozymes, etc., or proteases, such as, for example, papain, bromelain, keratinases, trypsin, chymotrypsin, pepsin, renin (chymosin), suizyme, nortase, etc..
  • These proteins may be provided by the at least one modified (m)RNA (of the inventive immunosuppressive composition), which is characterized by an increased level of expression. Accordingly, the invention provides a technology which allows to substitute proteins which are defective in the organism to be treated (e.g. either due to mutations, due to defective or missing expression) and thereby effective and increased expression of proteins, which are not functional in the organism to be treated, as e.g. occurring in monogenetic disorders, without leading to an innate immune response.
  • therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from proteases etc. which allow to cure a specific disease due to e.g. (over)expression of a dysfunctional or exogenous proteins causing disorders or diseases.
  • the invention may be used to therapeutically introduce the at least one modified (m)RNA (of the inventive immunosuppressive composition) into the organism, which attacks a pathogenic organism (virus, bacteria etc).
  • RNA encoding therapeutic proteases may be used to cleave viral proteins which are essential to the viral assembly or other essential steps of virus production.
  • Therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from proteins which modulate various intracellular pathways by e.g. signal transmission modulation (inhibition or stimulation) which may influence pivotal intracellular processes like apoptosis, cell growth etc, in particular with respect to the organism's immune system. Accordingly, immune modulators, e.g. cytokines, lymphokines, monokines, interferones etc. may be expressed efficiently by the at least one modified (m)RNA (of the inventive immunosuppressive composition).
  • signal transmission modulation inhibition or stimulation
  • immune modulators e.g. cytokines, lymphokines, monokines, interferones etc.
  • these proteins therefore also include, for example, cytokines of class I of the cytokine family that contain 4 position-specific conserved cysteine residues (CCCC) and a conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS), wherein X represents an unconserved amino acid.
  • cytokines of class I of the cytokine family that contain 4 position-specific conserved cysteine residues (CCCC) and a conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS), wherein X represents an unconserved amino acid.
  • Cytokines of class I of the cytokine family include the GM-CSF sub-family, for example IL-3, IL-5, GM-CSF, the IL-6 sub-family, for example IL-6, IL-1 1 , IL-12, or the IL-2 sub-family, for example IL-2, IL-4, IL-7, IL-9, IL-15, etc., or the cytokines IL-1 ⁇ , IL-I ⁇ , IL-10 etc.
  • such proteins can also include cytokines of class Il of the cytokine family (interferon receptor family), which likewise contain 4 position-specific 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- ⁇ , IFN- ⁇ , IFN- ⁇ , etc.
  • Proteins coded for by the at least one modified (m)RNA (of the inventive immunosuppressive composition) used according to the invention can further include also cytokines of the tumour necrosis family, for example TNF- ⁇ , TNF- ⁇ , TNF-RI, TNF-RII, CD40, Fas, etc., or cytokines of the chemokine family, which contain 7 transmembrane helices and interact with G-protein, for example IL-8, MIP-1 , RANTES, CCR5, CXR4, etc.
  • cytokines of the tumour necrosis family for example TNF- ⁇ , TNF- ⁇ , TNF-RI, TNF-RII, CD40, Fas, etc.
  • cytokines of the chemokine family which contain 7 transmembrane helices and interact with G-protein, for example IL-8, MIP-1 , RANTES, CCR5, CXR4, etc.
  • Such proteins can 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, BcI-X 1 , BcI-X 5 , bik, CAD, calpain, caspases, for example caspase-1 , caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, caspase-
  • the at least one modified (m)RNA may also code for antigen specific T cell receptors.
  • TCR is a molecule found on the surface of T lymphocytes (or T cells) that is generally responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules. It is a heterodimer consisting of an alpha and beta chain in 95% of T cells, while 5% of T cells have TCRs consisting of gamma and delta chains. Engagement of the TCR with antigen and MHC results in activation of its T lymphocyte through a series of biochemical events mediated by associated enzymes, co-receptors and specialized accessory molecules. Hence, these proteins allow to specifically target specific antigen and may support the functionality of the immune system due to their targeting properties.
  • MHC major histocompatibility complex
  • transfection of cells in vivo by administering the at least one modified (m)RNA (of the inventive immunosuppressive composition) coding for these receptors or, preferably, an ex vivo cell transfection approach (e.g. by transfecting specifically certain immune cells), may be pursued.
  • the T cell receptor molecules introduced recognize specific antigens on MHC molecule and may thereby support the immune system's awareness of antigens to be attacked.
  • therapeutically active proteins as encoded by the at least one modified (m)RNA may also be selected from (efficiently expressed) antigens which elicit only an adaptive immune response, whereas the underlying non-modified RNA provokes an innate and an adaptive immune reaction as such, i.e. the innate immune respnse may be suppressed.
  • This may be particularly advantageous with respect to the treatment of patients both suffering from allergic diseases or autoimmune diseases and another disease, such as cancer diseases or
  • the invention may allow to provide vaccines based on the at least one modified (m)RNA (of the inventive immunosuppressive composition), which expresses increased levels of the antigenic protein or peptide without leading to an (increased) immune response.
  • These vaccines may be used for the provision of tumour vaccines providing tumour antigens or antigens derived from pathogenic microorganisms causing e.g. infectious diseases.
  • the term "antigen" refers to a substance which is recognized by the immune system and is capable of triggering an antigen-specific immune response, e.g. by formation of antibodies as part of an adaptive immune response.
  • antigens may also comprise epitopes.
  • epitopes of a protein or an antigen typically, are fragments of such protein or peptide structures having 5 to 15, preferably 6 to 9, amino acids.
  • the antigen may be furthermore a "self” or non-self” antigen or an allergy antigen, i.e. an antigen, which causes an allergy in a human and may be derived from either a human or other sources. Antigens can be classified in the order of their origins.
  • Exogenous antigens are typically antigens that enter the cell or the body from outside (the cell or the body), for example by inhalation, ingestion or injection, etc.. These antigens are internalized by antigen- presenting cells ("APCs", such as dendritic cells or macrophages) and processed into fragments. APCs then present the fragments to T helper cells (e.g. CD4 + ) by the use of MHC Il molecules on their surface. Recognition of these antigen fragments by T cells leads to activation of the T cells and secretion of cytokines.
  • APCs antigen- presenting cells
  • T helper cells e.g. CD4 +
  • Cytokines are substances that can activate proliferation of immune cells such as cytotoxic T cells, B cells or macrophages.
  • endogenous antigens are antigens which typically have been generated within the cell, e.g. as a result of normal cell metabolism. Fragments of these antigens are presented on MHC I molecules on the surface of APCs. These antigens are recognized by activated antigen-specific cytotoxic CD8 + T cells. After recognition, those T cells react in secretion of different toxins that cause lysis or apoptosis of the antigen- presenting cell.
  • Endogenous antigens comprise antigens, e.g. proteins or peptides encoded by a foreign nucleic acid inside the cell as well as proteins or peptides encoded by the genetic information of the cell itself, or antigens from intracellularly occurring viruses.
  • Specifically preferred antigens coded for by the at least one modified (m)RNA (of the inventive immunosuppressive composition) can be selected from the following antigens: tumour-specific surface antigens (TSSAs), for example 5T4, ⁇ 5 ⁇ 1 -integrin, 707-AP, AFP,
  • TSSAs tumour-specific surface antigens
  • 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 -
  • Pml/RAR ⁇ PRAME, PSA, PSM, PSMA, RAGE, RUI or RU2, SAGE, SART-1 or SART-3, survivin, TEL/AML1, TGF ⁇ , TPI/m, TRP-1, TRP-2, TRP-2/INT2, VEGF and WT1, or from sequences such as, for example, NY-Eso-1 or NY-Eso-B.
  • TSA tumor-specific antigens
  • TAA tumor-associated-antigens
  • tumor antigens can also occur on the surface of the tumor in the form of, e.g., a mutated receptor. In this case, they can be recognized by antibodies, particularly by antibodies as defined above. Examples of tumor antigens are shown in Tables 1 and 2 below. These tables illustrate specific (protein) antigens (i.e. "tumor antigens") with respect to the cancer disease, they are associated with. According to the invention, the terms "cancer diseases” and “tumor diseases” are used synonymously herein.
  • the (protein) antigens, as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) are selected from the group consisting of 5T4, 707-AP, 9D7, AFP, AIbZIP HPGI, alpha-5-beta-1 -integrin, alpha-5-beta-6-integrin, alpha-actinin-4/m, alpha- methylacyl-coenzyme A racemase, ART-4, ARTC1/m, B7H4, BAGE-1 , BCL-2, bcr/abl, beta-eaten in/m, BING-4, BRCA1/m, BRCA2/m, CA 15-3/CA 27-29, CA 19-9, CA72-4, CA125, calreticulin, CAMEL, CASP-8/m, cathepsin B, cathepsin L, CD19, CD20, CD22, CD25, CDE30, CD33, CD4, CD52, CD55, CD56
  • the (protein) antigens as encoded by the at least one modified (m)RNA are selected from the group consisting of MAGE-A1 , MAGE-A6, melan-A, GP100, tyrosinase, survivin, CEA, Her-2/neu, WTl, PRAME, EGFRI (epidermal growth factor receptor 1), mucin-1 and SEC61 G, hTERT, 5T4, NY-Eso1 , and
  • TRP-2 more preferably from sequences of group consisting of MAGE-A1 [accession number M77481 ], MAGE-A6 [accession number NM_005363], melan-A [accession number NM_00551 1 ], GP100 [accession number M77348], tyrosinase [accession number NM_000372], survivin [accession number AF077350], CEA [accession number NMJD04363], Her-2/neu [accession number M1 1730], WT1 [accession number
  • NM_000378 PRAME [accession number NM_006115], EGFRI (epidermal growth factor receptor 1 ) [accession number AF288738], mucin-1 [accession number NM_002456] and SEC61 G [accession number NM_014302], hTERT [accession number NM_198253], 5T4 [accession number NM_006670], NY-Eso1 [accession number NM_001327], TRP-2 [accession number NM_001922], MAGE-A2: [accession number NM_153488], MAGE-
  • A3 [accession number NM_005362]
  • MAGE-C1 [accession number NM_005462]
  • MAGE-C2 [accession number NM_016249].
  • Therapeutically active proteins that can be coded for by the at least one modified (m)RNA (of the inventive immunosuppressive composition) further include also those proteins or protein sequences that have a sequence identity of at least 80% or 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99%, with one of the therapeutically active proteins described above, e.g. their native sequence.
  • the modified nucleosides and their native (non-modified) analog are considered to be "identical" herein.
  • the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode an antibody.
  • an antibody may be selected from any antibody, e.g. any recombinantly produced or naturally occurring antibodies, known in the art, in particular antibodies suitable for therapeutic, diagnostic or scientific purposes, or antibodies which have been identified in relation to specific diseases, such as e.g. cancer diseases or viral diseases.
  • the term "antibody” is used in its broadest sense and specifically covers monoclonal and polyclonal antibodies (including agonist, antagonist, and blocking or neutralizing antibodies) and antibody species with polyepitopic specificity.
  • the term "antibody” typically comprises any antibody known in the art (e.g. IgM, IgD, IgG, IgA and IgE antibodies), such as naturally occurring antibodies, antibodies generated by immunization in a host organism, antibodies which were isolated and identified from naturally occurring antibodies or antibodies generated by immunization in a host organism and recombinantly produced by biomolecular methods known in the art, as well as chimeric antibodies, human antibodies, humanized antibodies, bispecific antibodies, intrabodies, i.e. antibodies expressed in cells and optionally localized in specific cell compartments, and fragments and variants of the aforementioned antibodies.
  • an antibody consists of a light chain and a heavy chain both having variable and constant domains.
  • the light chain consists of an N-terminal variable domain, V L , and a C-terminal constant domain, Q.
  • the heavy chain of the IgG antibody for example, is comprised of an N-terminal variable domain, V H , and three constant domains, C H 1 , C H 2 und C H 3.
  • Single chain antibodies may be encoded by the RNA of the modified (m)RNA of the invention as well, preferably by a single-stranded RNA, more preferably by an mRNA.
  • the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode a polyclonal antibody.
  • polyclonal antibody typically means mixtures of antibodies directed to specific antigens or immunogens or epitopes of a protein which were generated by immunization of a host organism, such as a mammal, e.g. including goat, cattle, swine, dog, cat, donkey, monkey, ape, a rodent such as a mouse, hamster and rabbit.
  • Polyclonal antibodies are generally not identical, and thus usually recognize different epitopes or regions from the same antigen.
  • RNAs of the modified (m)RNA (of the inventive immunosuppressive composition) will be applied, each encoding a specific (monoclonal) antibody being directed to specific antigens or immunogens or epitopes of a protein.
  • the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode a monoclonal antibody.
  • the term "monoclonal antibody” herein typically refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed to a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed to different determinants (epitopes), each monoclonal antibody is directed to a single epitope or a determinant on the antigen. For example, monoclonal antibodies as defined above may be made by the hybridoma method first described by Kohler and Milstein,
  • Monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et a/., Nature, 348:552-554 (1990), for example, and the produced in vitro as recombinant proteins. According to Kohler and Milstein, an immunogen (antigen) of interest is injected into a host such as a mouse and B-cell lymphocytes produced in response to the immunogen are harvested after a period of time.
  • the B-cells are combined with myeloma cells obtained from mouse and introduced into a medium which permits the B-cells to fuse with the myeloma cells, producing hybridomas. These fused cells (hybridomas) are then placed in separate wells in microtiter plates and grown to produce monoclonal antibodies. The monoclonal antibodies are tested to determine which of them are suitable for detecting the antigen or epitope of interest. After being selected, the monoclonal antibodies can be grown in cell cultures or by injecting the hybridomas into mice. However, for the purposes of the present invention, the peptide sequences of these monoclonal antibodies have to be sequenced and modified (m)RNA sequences encoding these antibodies may be prepared according to procedures well known in the art.
  • non-human monoclonal or polyclonal antibodies such as murine antibodies may also be encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention.
  • m modified RNA
  • such antibodies are typically only of limited use, since they generally induce an immune response by production of human antibodies directed to the said non-human antibodies, in the human body. Therefore, a particular non-human antibody can only be administered once to the human.
  • chimeric, humanized non- human and human antibodies are also envisaged encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention.
  • “Chimeric” antibodies which may be encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention, are preferably antibodies in which the constant domains of an antibody described above are replaced by sequences of antibodies from other organisms, preferably human sequences.
  • “Humanized” (non-human) antibodies which may be also encoded by the at least one modified (m)RNA (molecule) of the modified (m)RNA of the present invention, are antibodies in which the constant and variable domains (except for the hypervariable domains) described above of an antibody are replaced by human sequences.
  • the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode human antibodies, i.e.
  • human antibodies having only human sequences can be isolated from human tissues or from immunized non-human host organisms which are transgene for the human IgG gene locus, sequenced RNA sequences may be prepared according to procedures well known in the art. Additionally, human antibodies can be provided by the use of a phage display.
  • the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode bispecific antibodies.
  • "Bispecific" antibodies in context of the invention are preferably antibodies which act as an adaptor between an effector and a respective target, e.g. for the purposes of recruiting effector molecules such as toxins, drugs, cytokines etc., targeting effector cells such as CTL, NK cells, makrophages, granulocytes, etc. (see for review: Kontermann R. E., Acta Pharmacol. Sin, 2005, 26(1 ): 1 -9).
  • Bispecific antibodies as described herein are, in general, configured to recognize, e.g.
  • bispecificity means herewith that the antigen-binding regions of the antibodies are specific for two different epitopes.
  • different antigens, immunogens or epitopes, etc. can be brought close together, what, optionally, allows a direct interaction of the two components.
  • different cells such as effector cells and target cells can be connected via a bispecific antibody.
  • antibodies or fragments thereof which bind, on the one hand, a soluble antigen as described herein, and, on the other hand, an antigen or receptor on the surface of a tumor cell.
  • the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may also encode intrabodies, wherein these intrabodies may be antibodies as defined above. Since these antibodies are intracellular expressed antibodies, i.e. antibodies which are encoded by nucleic acids localized in specific areas of the cell and also expressed there, such antibodies may be termed intrabodies.
  • Antibodies as encoded by the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may preferably comprise full-length antibodies, i.e. antibodies composed of the full heavy and full light chains, as described above. However, derivatives of antibodies such as antibody fragments, variants or adducts may be encoded by the above defined at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention.
  • the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may also encode antibody fragments selected from Fab, Fab', F(ab') 2 , Fc, Facb, pFc', Fd and Fv fragments of the aforementioned antibodies.
  • antibody fragments are known in the art.
  • a Fab fragment, antigen binding
  • a scFv single chain variable fragment
  • the domains are linked by an artificial linkage, in general a polypeptide linkage such as a peptide composed of 15-25 glycine, proline and/or serine residues.
  • Antibodies as defined above, may also be directed against antigens or epitopes thereof as defined above.
  • the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode fragments and/or variants of the aforementioned therapeutically active proteins or antibodies, etc., wherein the fragments and/or variants may have a sequence identity to one of the aforementioned therapeutically active proteins, or antibodies, etc. of at least 70%, 80% or 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99% over the whole length of the (coding) nucleic acid (or amino acid) sequences encoding these therapeutically active proteins, or antibodies, etc..
  • a "fragment of a therapeutically active protein, or antibody, etc.” in the context of the present invention is to be understood as a truncated therapeutically active protein, or antibody, etc. of the therapeutically active proteins, or antibodies, etc. defined above, i.e. an amino acid sequence which is N-terminally, C-terminally and/or intrasequentially truncated compared to the amino acid sequence of the original (native) protein. Especially, fragments including an epitope of those therapeutically active proteins, antibodies, etc., are preferred.
  • a “variant” in the context of the present invention refers to an therapeutically active protein, or antibody, etc. as defined above, wherein nucleic acids of the encoding modified mRNA sequence are exchanged, i.e.
  • a therapeutically active protein, or antibody, etc. having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s).
  • the fragments and/or variants have the same biological function or specific activity compared to the full-length native therapeutically active proteins, or antibodies, etc., e.g. specific binding capacity (e.g. of particular antigens), catalytic activity (e.g. of therapeutically active proteins), etc..
  • biological function of antibodies as described herein also comprises neutralization of antigens, complement activation or opsonization. Thereby, antibodies typically recognize either native epitopes on the cell surface or free antigens.
  • Antibodies as defined above can interact with the eel I -presenting antigens and initiate different defense mechanisms.
  • the antibody can initiate signaling mechanisms in the targeted cell that leads to the cell's self-destruction (apoptosis).
  • apoptosis On the other hand, it can mark the cell in such a way that other components or effector cells of the body's immune system can recognize and attack.
  • the attack mechanisms are referred to as antibody-dependent complement-mediated cytotoxicity (CMC) and antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC involves a recognition of the antibody by immune cells that engage the antibody-marked cells and either through their direct action, or through the recruitment of other cell types, lead to the tagged-cell's death.
  • CMC is a process where a cascade of different complement proteins becomes activated, usually when several antibodies are in close proximity to each other, either resulting in cell lysis or attracting other immune cells to this location for effector cell function.
  • the antibody can bind an antigen and neutralize the same.
  • Such neutralization reaction leads in general to blocking of the antibody.
  • the antibody can bind only one antigen, or, in case of a bispecific antibody, two antigens.
  • scFv antibody fragments are useful for neutralization reactions because they don't contain the functionalities of the constant domain of an antibody.
  • the complex system of complement proteins can be activated via binding of an antibody which is independent of the Fc part of an antibody.
  • End products of the complement cascade result in lysis of the cell and generation of an inflammatory milieu.
  • pathogens or other non-cellular particles are made accessible to phagocytes via binding the constant domain of an antibody.
  • cells recognized as foreign can be lysed via antibody- dependent cell-mediated cytotoxicity (ADCC).
  • ADCC antibody- dependent cell-mediated cytotoxicity
  • NK-cells can display lysis functions by activating Fc receptors.
  • the modified (m)RNA may occur as a mono-, di-, or even multicistronic RNA, i.e. an RNA which carries the coding sequences of one, two or more therapeutically active proteins, or antibodies, etc. as defined above.
  • the at least one coding modified (m)RNA encodes at least one, e.g. two, three or more of an therapeutically active protein, or antibody, etc. as defined above
  • each of the at least one modified (m)RNAs preferably encodes a (preferably different) therapeutically active protein, or antibody, etc. as defined above.
  • each therapeutically active protein, or antibody etc. as defined above encoded by the at least one modified (m)RNA may be selected independently.
  • the modified (m)RNA (of the immunosuppressive composition) according to the invention may be at least a single, bi- or even multicistronic modified (m)RNA, i.e. it ay be at least one modified (m)RNA which carries two, three or even more of the coding sequences of a therapeutically active protein, or antibody, etc. as defined above as defined herein.
  • Such coding sequences of the at least one (preferably different) therapeutically active protein, or antibody, etc. as defined above may be separated by at least one IRES (internal ribosomal entry site) sequence, as defined below.
  • IRES internal ribosomal entry site
  • IRES internal ribosomal entry site
  • a so-called IRES (internal ribosomal entry site) sequence as defined above can function as a sole ribosome binding site, but it can also serve to provide a bi- or even multicistronic modified (m)RNA as defined above which encodes several proteins which are to be translated by the ribosomes independently of one another.
  • IRES sequences which can be used according to the invention are those from picornaviruses (e.g.
  • FMDV pestiviruses
  • CFFV pestiviruses
  • PV polioviruses
  • ECMV encephalomyocarditis viruses
  • FMDV foot and mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MLV mouse leukoma virus
  • SIV simian immunodeficiency viruses
  • CrPV cricket paralysis viruses
  • the immunsuppressive composition of the present invention may comprise a mixture of at least one monocistronic modified (m)RNA as defined above, and at least one bi- or even multicistronic modified (m)RNA as defined above.
  • the at least one monocistronic modified (m)RNA and/or the at least one bi- or even multicistronic modified (m)RNA preferably encode different therapeutically active proteins and/or antibodies as defined herein.
  • the at least one monocistronic modified (m)RNA and the at least one bi- or even multicistronic modified (m)RNA may preferably also encode (in part) identical therapeutically active proteins and/or antibodies as defined herein.
  • a mixture of at least one monocistronic modified (m)RNA, as defined above, and at least one bi- or even multicistronic modified modified (m)RNA or of a mixture of more than one monocistronic modified (m)RNA, wherein the modified (m)RNA encodes at least one (preferably different) therapeutically active protein or antibody as defined above may be advantageous e.g. for a staggered, e.g. time dependent, administration of the immunosuppressive composition of the present invention to a patient in need thereof.
  • the components of such an immunosuppressive composition of the present invention, particularly the at least one (different) modified (m)RNAs encoding the at least one (preferably different) therapeutically active protein and/or antibody may be e.g. contained in (different parts of) a kit of parts composition or may be e.g. administered separately as components of different immunosuppressive compositions according to the present invention.
  • the at least one modified (m)RNA (of the immunosuppressive composition) according to the invention may be a short RNA oligonucleotide.
  • Short RNA oligonucleotides in the context of the present invention may comprise any RNA as defined above.
  • the short RNA oligonucleotide may be a single- or a double-stranded RNA oligonuclotide, more preferably a single-stranded RNA oligonucleotide.
  • the short RNA oligonucleotide may be a linear single-stranded RNA oligonucleotide.
  • the short RNA oligonucleotides as used herein may comprise a length as defined above in general for RNA molecules, more preferably a length of 5 to 100, of 5 to 50, or of 5 of 30, and even more preferably a length of 20 to 100, of 20 to 80, or of 20 of 60 nucleotides.
  • the at least one modified (m)RNA (of the immunosuppressive composition) according to the invention may be in the form of siRNA.
  • a siRNA is of interest particularly in connection with the phenomenon of RNA interference. Attention was drawn to the phenomenon of RNA interference in the course of immunological research. In recent years, an RNA-based defence mechanism has been discovered, which occurs both in the kingdom of the fungi and in the plant and animal kingdom and acts as an "immune system of the genome". The system was originally described in various species independently of one another, first in C.
  • RNA-mediated virus resistance in plants RNA-mediated virus resistance in plants
  • PTGS posttranscriptional gene silencing
  • RNA interference in eukaryotes are accordingly based on a common procedure.
  • the in vitro technique of RNA interference (RNAi) is based on double-stranded RNA molecules (dsRNA), which trigger the sequence- specific suppression of gene expression (Zamore (2001 ) Nat. Struct. Biol. 9: 746-750; Sharp (2001 ) Genes Dev. 5:485-490: Hannon (2002) Nature 41 : 244-251 ).
  • a siRNA as used for the modified (m)RNA according to the present invention typically comprises a (single- or) double stranded, preferably a double-stranded, RNA sequence with about 8 to 30 nucleotides, preferably 17 to 25 nucleotides, even more preferably from 20 to 25 and most preferably from 21 to 23 nucleotides.
  • siRNAs can also be directed against nucleotide sequences (of the RNA or genomic sequence) of a (therapeutically relevant) protein, adjuvant protein or an antigen described hereinbefore either in their coding region or in their non-coding region, in particular in the 5' non-coding region of the RNA or genomic sequence, for example, therefore, against non-coding regions of the RNA having a regulatory function.
  • the target sequence of the siRNA can therefore lie in the translated (coding) and/or untranslated region (3'-and/or 5') and/or in the region of the control elements.
  • the target sequence of a siRNA can also lie in the overlapping region of untranslated and translated sequence; in particular, the target sequence can comprise at least one nucleotide upstream of the start triplet of the coding region of the (m)RNA.
  • the at least one modified (m)RNA (of the immunosuppressive composition) may be an antisense RNA.
  • an antisense RNA is preferably a (single-stranded) RNA molecule transcribed off the coding, rather than the template, strand of DNA, so that it is complementary to the sense (messenger) RNA.
  • An antisense RNA as defined herein typically forms a duplex between the sense and antisense RNA molecules and is thus capable to block transcription of the coding strand.
  • An antisense RNA as used as the at least one modified (m)RNA (of the immunosuppressive composition) can be directed e.g.
  • the target sequence of the antisense RNA can therefore lie in the translated and/or untranslated region of the RNA and/or in the region of the control elements.
  • the target sequence of a antisense RNA can also lie in the overlapping region of untranslated and translated sequence; in particular, the target sequence can comprise at least one nucleotide upstream of the start triplet of the coding region of the RNA.
  • the antisense RNA as used herein as the at least one inventive modified (m)RNA comprises a length as defined above in general for RNA molecules, more preferably a length of 1000 to 5000, of 500 to 5000, of 5 to 5000, or of 5 to 1000, 5 to 500, 5 to 250, of 5 to 100, of 5 to 50 or of 5 to 30 nucleotides, and even more preferably a length of 20 to 100, of 20 to 80, or of 20 to 60 nucleotides.
  • the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) as defined above, particularly when occurring as a coding RNA, a short RNA oligonucleotide, a siRNA, an antisense RNA, riboswitches, ribozymes or aptamers, may be further GC-modified or even further modified. Some modifications may be, dependent on the type of RNA, be more suitable for an RNA in general, or, e.g. in the case of GC-modified (m)RNA sequences, be more suitable for coding RNA, preferably an mRNA. Such further modifications as defined herein preferably lead to a stabilized modified (m)RNA. According to one embodiment, such a stabilized modified (m)RNA may be prepared by modifying the G/C content of the coding region of the modified (m)RNA.
  • the G/C content of the coding region of the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) is altered, preferably increased, compared to the G/C content of the coding region of the corresponding native inventive modified (m)RNA (of the inventive immunosuppressive composition).
  • the encoded amino acid sequence of this G/C-increased modified (m)RNA is preferably not altered compared to the corresponding native modified (m)RNA.
  • Such alteration of the GC-sequence may be termed in the following GC- stabilization.
  • This G/C-stabilization of the modified (m)RNA of the immunosuppressive composition of the present invention is based on the fact that the sequence of any (m)RNA region to be translated is important for efficient translation of that (m)RNA.
  • the sequence of various nucleotides is important.
  • sequences having an increased G (guanosine)/C (cytosine) content are more stable than sequences having an increased A (adenosine)/U (uracil) content.
  • the codons of the inventive modified (m)RNA (of the inventive immunosuppressive composition) are therefore varied compared to its native modified (m)RNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides.
  • the most favorable codons for the stability can be determined (so-called alternative codon usage).
  • the modified (m)RNA sequence there are various possibilities for G/C-modification of the modified (m)RNA sequence, compared to its native sequence.
  • amino acids which are encoded by codons which contain exclusively G or C nucleotides no G/C-modification of the codon is necessary.
  • the codons for Pro (CCC or CCG), Arg (CGC or CGG), Ala (GCC or GCG) and GIy (GGC or GGG) require no G/C-modification, since no A or U is present.
  • codons which contain A and/or U nucleotides can be G/C-modified by substitution of other codons which code for the same amino acids but contain no A and/or U. Examples of these are:
  • the codons for Pro can be G/C-modified from CCU or CCA to CCC or CCG; the codons for Arg can be G/C-modified from CGU or CGA or AGA or AGG to CGC or CGG; the codons for Ala can be G/C-modified from GCU or GCA to GCC or GCG; the codons for GIy can be G/C-modified from GGU or GGA to GGC or GGG.
  • the codons for Phe can be G/C-modified from UUU to UUC; the codons for Leu can be G/C-modified from UUA, UUG, CUU or CUA to CUC or CUG; the codons for Ser can be G/C-modified from UCU or UCA or AGU to UCC, UCG or AGC; the codon for Tyr can be G/C-modified from UAU to UAC; the codon for Cys can be G/C-modified from UGU to UGC; the codon for His can be G/C-modified from CAU to CAC; the codon for GIn can be G/C-modified from CAA to CAG; the codons for He can be G/C-modified from AUU or AUA to AUC; the codons for Thr can be G/C-modified from ACU or ACA to ACC or ACG; the codon for Asn can be G/C-modified from AAU to AAC; the codon for Lys can
  • the G/C content of the coding region of the GC-stabilized inventive modified (m)RNA is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the coding region of the native modified (m)RNA which codes for a protein.
  • at least 60%, more preferably at least 70 %, even more preferably at least 80% and most preferably at least 90%, 95% or even 100% of the substitutable codons in the region coding for a protein or the whole sequence of the native modified (m)RNA sequence are substituted, thereby increasing the GC/content of said sequence.
  • the G/C content of the native modified (m)RNA is particularly preferable to increase the G/C content of the native modified (m)RNA to the maximum (i.e. 100% of the substitutable codons), in particular in the region coding for a protein, compared to the native sequence.
  • a further preferred modification of the native inventive modified (m)RNA is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the corresponding G/C-stabilized or native modified (m)RNA sequence may be translated to a significantly poorer degree than in the case, where codons coding for relatively "frequent" tRNAs are present.
  • the region which codes for a therapeutically active protein, or antibody, etc., as defined above is GC-stabilized compared to the corresponding region of the native modified (m)RNA such that at least one codon of the native sequence which codes for a tRNA which is relatively rare in the cell is exchanged for a codon which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA.
  • the native modified (m)RNA sequences are GC-stabilized such that codons for which frequently occurring tRNAs are available are inserted.
  • This preferred embodiment allows provision of a particularly efficiently translated and GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition).
  • the nucleotide sequence of any desired modified (m)RNA as defined above can be GC-stabilized with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence coded by the GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition) preferably not being further modified compared to the native modified (m)RNA sequence.
  • the source code in Visual Basic 6.0 development environment used: Microsoft
  • Visual Studio Enterprise 6.0 with Servicepack 3 is also described in WO 02/098443.
  • the A/U content in the environment of the ribosome binding site of the (optionally already GC-stabilized) inventive modified (m)RNA is increased compared to the A/U content in the environment of the ribosome binding site of its particular native (m)RNA.
  • This modification (an increased A/U content around the ribosome binding site) increases the efficiency of ribosome binding to the modified (m)RNA.
  • An effective binding of the ribosomes to the ribosome binding site (Kozak sequence: GCCGCCACCAUGG (SEQ ID NO: 1 ), the AUG forms the start codon) in turn has the effect of an efficient translation of the modified (m)RNA.
  • inventive modified (m)RNA may be further modified with respect to potentially destabilizing sequence elements.
  • the coding region and/or the 5 1 and/or 3' untranslated region of this modified (m)RNA may be further modified compared to the particular native modified (m)RNA such that is contains no destabilizing sequence elements, the coded amino acid sequence of the modified (m)RNA preferably not being modified compared to its particular native modified (m)RNA.
  • DSE destabilizing sequence elements
  • one or more such further modifications compared to the corresponding region of the native modified (m)RNA can therefore be carried out, so that no or substantially no destabilizing sequence elements are contained there.
  • DSE present in the untranslated regions (3'- and/or 5'-UTR) can also be eliminated from the inventive modified (m)RNA (of the inventive immunosuppressive composition) by such further modifications.
  • Such destabilizing sequences are e.g. AU-rich sequences (AURES), which occur in 3'-UTR sections of numerous unstable RNAs (Caput et a/., Proc. Natl. Acad. Sci. USA 1986, 83: 1670 to 1674).
  • the inventive modified (m)RNA (of the inventive immunosuppressive composition) is therefore preferably further modified compared to the native modified (m)RNA such that the modified (m)RNA contains no such destabilizing sequences.
  • sequence motifs which are recognized by possible endonucleases, e.g. the sequence GAACAAG, which is contained in the 3'-UTR segment of the gene which codes for the transferrin receptor (Binder eta/., EMBO J.
  • inventive modified (m)RNA of the inventive immunosuppressive composition
  • inventive modified (m)RNA has, in a further modified form of the modified (m)RNA, at least one IRES as defined above and/or at least one 5' and/or 3' stabilizing sequence, e.g. to enhance ribosome binding or to allow expression of different encoded proteins as defined above located on at least one (bi- or even multicistronic) inventive modified (m)RNA (of the inventive immunosuppressive composition).
  • the inventive modified (m)RNA (of the inventive immunosuppressive composition), preferably when occurring as an mRNA, furthermore preferably has at least one 5' and/or 3' stabilizing sequence.
  • These stabilizing sequences in the 5' and/or 3' untranslated regions have the effect of increasing the half-life of the modified (m)RNA in the cytosol.
  • These stabilizing sequences can have 100% sequence homology to naturally occurring sequences which occur in viruses, bacteria and eukaryotes, but can also be partly or completely synthetic.
  • the untranslated sequences (UTR) of the globin gene e.g.
  • stabilizing sequences which can be used in the present invention for a further stabilized modified (m)RNA as contained in the inventive immunosuppressive composition.
  • Another example of a stabilizing sequence has the general formula (C/U)CCAN x CCC(U/A)Py x UC(C/U)CC (SEQ ID NO: 2), which is contained in the 3'UTR of the very stable RNA which codes for globin, (l)-collagen, 15-lipoxygenase or for tyrosine hydroxylase (cf. Holcik et al., Proc. Natl. Acad. Sci. USA 1997, 94: 2410 to 2414).
  • inventive modified (m)RNA (of the inventive immunosuppressive composition) is therefore preferably present as globin UTR (untranslated regions)-stabilized RNA, in particular as globin UTR-stabilized modified (m)RNA.
  • globin UTR untranslated regions
  • m modified RNA
  • any of the above modifications may be applied to the inventive modified (m)RNA (of the inventive immunosuppressive composition), and further to any modified (m)RNA as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the respective modified (m)RNA.
  • a person skilled in the art will be able to take his choice accordingly.
  • the inventive modified (m)RNA (of the inventive immunosuppressive composition) may be prepared using any naturally or synthetic DNA or RNA sequence available in the art as a template, i.e. any suitable (desoxy)ribonucleic acid.
  • Such naturally or synthetic DNA or RNA sequences may be obtained from any synthetic or naturally occurring source, which is available to a skilled person, e.g. may be derived from a protein or peptide library or may be transcribed from a nucleic acid library, such as a cDNA library, or may be obtained from any living or dead tissue, from a sample obtained from e.g. a human, animal or bacterial source.
  • inventive modified (m)RNA (of the inventive immunosuppressive composition) may be prepared synthetically by methods known to a person skilled in the art, e.g., by solid phase synthesis or any other suitable method for preparing nucleic acid sequences, particularly RNA sequences. Furthermore, substitutions, additions or eliminations of bases in these sequences are preferably carried out using a DNA matrix for preparation of the modified (m)RNA of the immunosuppressive composition of the present invention or by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation strategy (see e.g.
  • the modification(s) of the inventive modified (m)RNA can furthermore be introduced into the RNA by means of methods known to a person skilled in the art. Suitable methods are, for example, synthesis methods using (automatic or semi-automatic) oligonucleotide synthesis devices, biochemical methods, such as, for example, in vitro transcription methods, etc..
  • the inventive modified (m)RNA of the immunosuppressive composition of the present invention can likewise be stabilized by the use of nanoplexes (nanoparticular systems), lipoplexes (liposomal systems), or the use of polyplexes or cationic polymers, e.g. by associating or complexing the modified (m)RNA with, or binding it, thereto.
  • nanoplexes nanoparticular systems
  • Such nanoplexes involve use of polyacrylates, polyamides, polystyrene, cyanoacrylates, polylactat (PLA), poly(lactic-co-glycolic acid) (PLGA), polyethyl, etc., as carrier systems for the transport of nucleic acids into cells or tissues.
  • Lipoplexes or liposomal systems typically involve use of cationic lipids, which are capable to mimick a cell membrane. Thereby, the positively charged moiety of the lipids interacts with the negatively charged moiety of the nucleic acids and thus enables fusion with the cell membrane.
  • Lipoplexes or liposomal systems include e.g. DOTMA, DOPE, DOSPA, DOTAP, DC-Choi, EDMPC, etc..
  • Polyplexes (cationic polymers) typically form a complex with negatively charged nucleic acids leading to a condensation of nucleic acids and protecting these nucleic acids against degradation. Transport into cells using polyplexes (cationic polymers) typically occurs via receptor mediated endocytosis.
  • Polyplexes may also include polycations selected from from oligoarginines having formula (I): (Arg)
  • Polyplexes include furthermore e.g. poly-L-lysine (PLL), chitosan, polyethylenimine (PEI), polydimethylaminoethylmethacrylate (PD-MAEMA), polyamidoamine (PAMAM).
  • PLL poly-L-lysine
  • PEI polyethylenimine
  • PD-MAEMA polydimethylaminoethylmethacrylate
  • PAMAM polyamidoamine
  • Cationic compounds may furthermore include any further (poly)cationic peptide or protein.
  • protamine, nucleoline, spermin or spermidine as the polycationic, nucleic-acid-binding protein to inventive modified (m)RNA of the immunosuppressive composition of the present invention is particularly effective.
  • use of other cationic peptides or proteins is likewise possible, such as histones, cationic polysaccharides, for example chitosan, polybrene, polyethyleneimine (PEI), etc., or cationic lipids, e.g. oligofectamine as a lipid based complexation reagent.
  • the procedure for stabilizing the modified (m)RNA is using polycationic compounds in general described in EP-A-1083232, the disclosure of which is incorporated by reference into the present invention in its entirety.
  • the present invention also provides a pharmaceutical composition, comprising an inventive immunosuppressive composition as defined above and optionally a pharmaceutically acceptable carrier, adjuvant, and/or vehicle.
  • an inventive pharmaceutical composition comprises a modified (m)RNA as defined above, e.g. as component of an inventive immunosuppressive composition as defined above, wherein at least one nucleoside of the modified (m)RNA of the inventive immunosuppressive composition has been modified as defined above, i.e.
  • At least one nucleoside comprises: a) a chemical modification at the 4-, 5-or 6-position of the pyrimidine base of the nucleosides cytidine and/or uridine as defined above; b) a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine as defined above; and/or c) a chemical modification at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine as defined above, and wherein the modified (m)RNA is suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA.
  • the inventive pharmaceutical composition due to the content of the modified (m)RNA as defined herein, is suitable for suppressing and/or avoiding an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, i.e. not an overall immune response but the innate immune response caused by administering the unmodified RNA as such is reduced or even avoided with the modified (m)RNA as defined herein.
  • the inventive pharmaceutical composition may comprise a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier typically includes the liquid or non-liquid basis of the inventive pharmaceutical composition.
  • the term "(compatible) pharmaceutically acceptable carrier" used here preferably includes the liquid or non-liquid basis of the inventive pharmaceutical composition.
  • compatible means that the constituents of the inventive pharmaceutical composition are capable of being mixed with the pharmaceutically active component, i.e. with the modified (m)RNA as defined herein, in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the inventive pharmaceutical composition under usual use conditions.
  • Pharmaceutically acceptable carriers must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated.
  • the pharmaceutically acceptable carrier will typically comprise one or more (compatible) pharmaceutically acceptable liquid carriers.
  • the composition may comprise as (compatible) pharmaceutically acceptable liquid carriers e.g. pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g phosphate, citrate etc. buffered solutions, vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid, etc..
  • a buffer preferably an aqueous buffer, may be used, containing a sodium salt, preferably at least 50 mM of a sodium salt, a calcium salt, preferably at least 0,01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt.
  • the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc..
  • examples of sodium salts include e.g.
  • examples of the optional potassium salts include e.g. KCl, Kl, KBr, K 2 CO 3 , KHCO 3 , K 2 SO 4
  • examples of calcium salts include e.g. CaCI 2 , CaI 2 , CaBr 2 , CaCO 3 , CaSO 4 , Ca(OH) 2 .
  • organic anions of the aforementioned cations may be contained in the buffer.
  • the buffer suitable for injection purposes as defined above may contain salts selected from sodium chloride (NaCI), calcium chloride (CaCI 2 ) and optionally potassium chloride (KCI), wherein further anions may be present additional to the chlorides.
  • the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCI), at least 3 itiM potassium chloride (KCI) and at least 0,01 mM calcium chloride (CaCI 2 ).
  • the injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e.
  • the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects.
  • Reference media are e.g. in ,,/n vivd' methods occurring liquids such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in "in vitrd' methods, such as common buffers or liquids.
  • Such common buffers or liquids are known to a skilled person. Ringer solution or Ringer-Lactate solution is particularly preferred as a liquid basis.
  • the pharmaceutically acceptable carrier will typically comprise one or more (compatible) pharmaceutically acceptable solid carriers.
  • the composition may comprise as (compatible) pharmaceutically acceptable solid carriers e.g. one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person.
  • suitable pharmaceutically acceptable solid carriers are e.g.
  • sugars such as, for example, lactose, glucose and sucrose
  • starches such as, for example, corn starch or potato starch
  • cellulose and its derivatives such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate
  • powdered tragacanth malt
  • gelatin gelatin
  • tallow solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulphate, etc.
  • a (compatible) pharmaceutically acceptable carrier as defined above is determined in principle by the manner in which the pharmaceutical composition according to the invention is to be administered.
  • the inventive pharmaceutical composition can be administered, for example, systemically or topically.
  • Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, , intradermal, intranasal, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • Routes for local administration in general include topically, rectally, nasally, buccally, vaginally routes or administration or via an implanted reservoir, but also transdermal, intramuscular or subcutaneous injection.
  • the inventive pharmaceutical composition may be administered by parenteral injection, more preferably by subcutaneous, intravenous, intramuscular, intradermal, intraarticular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or via infusion techniques.
  • Sterile injectable forms of the inventive pharmaceutical compositions may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenteral ly-acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di- glycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutical ly-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation of the inventive pharmaceutical composition.
  • the inventive pharmaceutical composition as defined above may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • the active ingredient i.e. the inventive modified (m)RNA (of the inventive immunosuppressive composition) as contained in the inventive pharmaceutical composition, is combined with emulsifying and suspending agents.
  • certain sweetening, flavoring or coloring agents may also be added.
  • Pharmaceutically acceptable carriers for the preparation of unit dose forms, which can be used for oral administration are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art.
  • inventive pharmaceutical composition may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including diseases of the skin or of any other accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs.
  • inventive pharmaceutical compositions may be formulated in a suitable ointment containing the inventive modified (m)RNA (of the inventive immunosuppressive composition) as contained in the inventive pharmaceutical composition, suspended or dissolved in one or more carriers.
  • Carriers for topical administration include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the inventive pharmaceutical composition can be formulated in a suitable lotion or cream.
  • suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the inventive pharmaceutical composition typically comprises a "safe and effective amount" of the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) as contained in the inventive pharmaceutical composition.
  • a "safe and effective amount” means an amount of the at least one modified (m)RNA as defined herein in the inventive pharmaceutical composition as defined above that is sufficient to significantly induce a positive modification of a disease or disorder as defined herein.
  • a "safe and effective amount” is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • a "safe and effective amount" of the at least one modified (m)RNA as defined herein will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the activity of the specific modified (m)RNA as defined herein employed, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying doctor.
  • the inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine.
  • emulsifiers such as, for example, Tween ® ; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet- forming agents; stabilizers; antioxidants; preservatives.
  • the inventive pharmaceutical composition may be provided as a vaccine.
  • Such an inventive vaccine is typically composed like the inventive pharmaceutical composition and preferably allows to provide an active or a passive adaptive immune response of a patient to be treated, e.g. by using a modified (m)RNA, encoding an antibody as defined above.
  • the inventive vaccine may contain any of the above mentioned antibodies as a further component of the vaccine.
  • the inventive vaccine may also comprise a pharmaceutically acceptable carrier, adjuvant, and/or vehicle as defined above for the inventive pharmaceutical composition.
  • a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered.
  • the inventive vaccine can be administered, for example, systemically or locally as defined above. More preferably, vaccines may be administered by an intradermal, subcutaneous, or intramuscular route. Inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid) form.
  • the suitable amount of the inventive vaccine to be administered can be determined by routine experiments with animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non-human primate models.
  • Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4. Suitable carriers for injection include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices.
  • the modified (m)RNA as defined herein and/or the inventive immunosuppressive composition may be used for the preparation of an inventive pharmaceutical composition or an inventive vaccine, preferably all as defined herein, for the prophylaxis, treatment, and/or amelioration of any of the diseases and disorders as defined herein.
  • the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g.
  • cervical carcinoma cervical cancer
  • an inventive pharmaceutical composition or vaccine may be provided in this context, which contains a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of cancer or tumour diseases.
  • modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases.
  • infectious diseases are typically selected from influenza, malaria, SARS, yellow fever, AIDS, Lyme borreliosis, Leishmaniasis, anthrax, meningitis, viral infectious diseases such as AIDS, Condyloma acuminata, hollow warts, Dengue fever, three-day fever, Ebola virus, cold, early summer meningoencephalitis (FSME), flu, shingles, hepatitis, herpes simplex type I, herpes simplex type II, Herpes zoster, influenza, Japanese encephalitis, Lassa fever, Marburg virus, measles, foot-and-mouth disease, mononucleosis, mumps, Norwalk virus infection, Ffeiffer's glandular fever, smallpox, polio (childhood lameness), pseudo-croup, fifth disease, rabies, warts, West Nile fever, chickenpox, cytomegalic virus
  • an inventive pharmaceutical composition or vaccine may be provided in this context, which contains a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of infectious diseases.
  • the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. autoimmune diseases.
  • Autoimmune diseases as defined herein may involve antibody-mediated or T-cell mediated immunity and/or can be broadly divided into systemic and organ-specific or localised autoimmune disorders, depending on the principal clinico-pathologic features of each disease.
  • Autoimmune diseases may be divided into the categories of systemic syndromes, including systemic lupus erythematosus (SLE), Sjogren's syndrome, Scleroderma, Rheumatoid Arthritis including juvenile (rheumatoid) arthritis, and polymyositis or local syndromes which may be endocrinologic (type I diabetes (Diabetes mellitus Type 1), Hashimoto's thyroiditis, Addison's disease etc.), dermatologic (pemphigus vulgaris), haematologic (autoimmune haemolytic anaemia), neural (multiple sclerosis) or can involve virtually any circumscribed mass of body tissue.
  • SLE systemic lupus erythematosus
  • Sjogren's syndrome Scleroderma
  • Rheumatoid Arthritis including juvenile (rheumatoid) arthritis
  • polymyositis or local syndromes which may be endocrinologic (type I diabetes (Diabetes mellit
  • the autoimmune diseases to be treated may be selected from the group consisting of type I autoimmune diseases or type Il autoimmune diseases or type III autoimmune diseases or type IV autoimmune diseases, such as, for example, multiple sclerosis (MS), rheumatoid arthritis, diabetes, type I diabetes (Diabetes mellitus Type 1 ), chronic polyarthritis, Basedow's disease, autoimmune forms of chronic hepatitis, colitis ulcerosa, type I allergy diseases, type Il allergy diseases, type III allergy diseases, type IV allergy diseases, fibromyalgia, hair loss, alopecia, alopecia areata, Bechterew's disease, Crohn's disease, Myasthenia gravis, neurodermitis, Polymyalgia rheumatica, progressive systemic sclerosis (PSS), rheumatic arthritis, psoriasis, vasculitis, etc, type Il diabetes, graft versus host disease, transplanted organ rejection, asthma, acquired hemophilia, anky
  • the autoreaction may be due to a T-CeII bypass.
  • a normal immune system requires the activation of B-cells by T-cells before the former can produce antibodies in large quantities.
  • This requirement of a T-cell can be by-passed in rare instances, such as infection by organisms producing super-antigens, which are capable of initiating polyclonal activation of B-cells, or even of T-cells, by directly binding to the ⁇ -subunit of T-cell receptors in a non-specific fashion.
  • autoimmune diseases from a Molecular Mimicry.
  • An exogenous antigen may share structural similarities with certain host antigens; thus, any antibody produced against this antigen (which mimics the self-antigens) can also, in theory, bind to the host antigens and amplify the immune response.
  • the most striking form of molecular mimicry is observed in Group A beta- haemolytic streptococci, which shares antigens with human myocardium, and is responsible for the cardiac manifestations of Rheumatic Fever.
  • the present invention allows therefore to provide an inventive immunosuppressive composition or pharmaceutical composition or vaccine containing a modified (m)RNA coding for an autoantigen, which typically allows the immune system to be desensitized by triggering an adaptive immune response towards this specific autoantigen.
  • an immunization against such an autoantigen preferably leads to a decrease in the overall immune response and therefore forms a basis for treatments of allergic diseases or disorders or autoimmune diseases or disorders.
  • the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. allergic disorders or diseases.
  • Allergy is a condition that typically involves an abnormal, acquired immunological hypersensitivity to certain foreign antigens or allergens. Allergies normally result in a local or systemic inflammatory response to these antigens or allergens and leading to an immunity in the body against these allergens. Allergens in this context include e.g. grasses, pollens, molds, drugs, or numerous environmental triggers, etc.
  • Type I hypersensitivity is characterised by excessive activation of mast cells and basophils by IgE, resulting in a systemic inflammatory response that can result in symptoms as benign as a runny nose, to life-threatening anaphylactic shock and death.
  • allergies include, without being limited thereto, allergic asthma (leading to swelling of the nasal mucosa), allergic conjunctivitis (leading to redness and itching of the conjunctiva), allergic rhinitis ("hay fever"), anaphylaxis, angiodema, atopic dermatitis (eczema), urticaria (hives), eosinophilia, respiratory, allergies to insect stings, skin allergies (leading to and including various rashes, such as eczema, hives (urticaria) and (contact) dermatitis), food allergies, allergies to medicine, etc.
  • allergenital asthma leading to swelling of the nasal mucosa
  • allergic conjunctivitis leading to redness and itching of the conjunctiva
  • allergic rhinitis (“hay fever")
  • anaphylaxis angiodema
  • atopic dermatitis eczema
  • urticaria urticaria
  • eosinophilia
  • an inventive pharmaceutical composition or vaccine may be provided, which contains a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of allergic disorders or diseases, which allows a desensitizing of the immune reaction or which triggers a specific immune response, which, in turn, also allows a desensitizing of the immune reaction.
  • a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of allergic disorders or diseases, which allows a desensitizing of the immune reaction or which triggers a specific immune response, which, in turn, also allows a desensitizing of the immune reaction.
  • modified (m)RNA as defined herein, the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. genetic diseases, which are caused by genetic defects, e.g. due to gene mutations resulting in loss of protein activity or regulatory mutations which do not allow transcription or translation of the protein. Frequently, these diseases lead to metabolic disorders or other symptoms, e.g. muscle dystrophy.
  • the present invention allows to treat these diseases by providing the modified (m)RNA of the inventive immunosuppressive composition, which allows sufficient level of the protein to be translated without triggering an innate immune response due to administration of RNA.
  • the following diseases may be treated: 3-beta-hydroxysteroid dehydrogenase deficiency (type II); 3-ketothiolase deficiency; 6-mercaptopurine sensitivity; Aarskog-Scott syndrome; Abetalipoproteinemia; Acatalasemia; Achondrogenesis; Achondrogenesis- hypochondrogenesis; Achondroplasia; Achromatopsia; Acromesomelic dysplasia (Hunter- Thompson type); ACTH deficiency; Acyl-CoA dehydrogenase deficiency (short-chain, medium chain, long chain); Adenomatous polyposis coli; Adenosin-deaminase deficiency; Adenylosuccinase deficiency; Adhalinopathy
  • Pseudohypoparathyroidism Pseudovaginal perineoscrotal hypospadias; Pseudovitamin D deficiency rickets; Pseudoxanthoma elasticum (autosomal dominant; autosomal recessive); Pulmonary alveolar proteinosis; Pulmonary hypertension; Purpura fulminans; Pycnodysostosis; Pyropoikilocytosis; Pyruvate carboxylase deficiency; Pyruvate dehydrogenase deficiency; Rabson-Mendenhall syndrome; Refsum disease; Renal cell carcinoma; Renal tubular acidosis; Renal tubular acidosis with deafness; Renal tubular acidosis-osteopetrosis syndrome; Reticulosis (familial histiocytic); Retinal degeneration; Retinal dystrophy; Retinitis pigmentosa; Retinit
  • Preferred diseases to be treated which have a genetic inherited background and which are typically caused by a single gene defect and are inherited according to Mendel's laws are preferably selected from the group consisting of autosomal-recessive inherited diseases, such as, for example, adenosine deaminase deficiency, familial hypercholesterolemia, Canavan's syndrome, Gaucher's disease, Fanconi anaemia, neuronal ceroid lipofuscinoses, mucoviscidosis (cystic fibrosis), sickle cell anaemia, phenylketonuria, alcaptonuria, albinism, hypothyreosis, galactosaemia, alpha-1 -anti-trypsin deficiency, Xeroderma pigmentosum, Ribbing's syndrome, mucopolysaccharidoses, cleft lip, jaw, palate, Laurence Moon Biedl Bardet sydrome, short rib polydactylia syndrome,
  • X syndrome muscular dystrophy (Duchenne and Becker-Kiener type), haemophilia A and B, G6PD deficiency, Fabry's disease, mucopolysaccharidosis, Nome's syndrome, Retinitis pigmentosa, septic granulomatosis, X-SCID, ornithine transcarbamylase deficiency, Lesch- Nyhan syndrome, or from autosomal-dominant inherited diseases, such as, for example, hereditary angiooedema, Marfan syndrome, neurofibromatosis, type I progeria, Osteogenesis imperfecta, Klippel-Trenaumay syndrome, Sturge-Weber syndrome, Hippel- Lindau syndrome and tuberosis sclerosis.
  • hereditary angiooedema Marfan syndrome
  • neurofibromatosis type I progeria
  • Osteogenesis imperfecta Klippel-Trenaumay syndrome
  • Sturge-Weber syndrome Hippel- Lindau syndrome
  • the present invention also allows treatment of diseases, which have not been inherited, or which may not be summarized under the above categories.
  • Such dieseases may include e.g. the treatment of patients, which are in need of a specific protein factor, e.g. a specific therapeutically active protein as mentioned above.
  • This may e.g. include dialysis patients, e.g. patients which undergo a (regular) a kidney or renal dialysis, and which may be in need of specific therapeutically active proteins as defined above, e.g. EPO, etc..
  • the invention furthermore relates also to the use of a modified (m)RNA as described herein, or of an inventive immunosuppressive composition or of a pharmaceutical composition or of a vaccine as described herein, for the prophylaxis, treatment, and/or amelioration of diseases or disorders as mentioned above. It also includes in particular the use of the modified (m)RNA as described herein, or of an inventive immunosuppressive composition or of a pharmaceutical composition or of a vaccine described herein for inoculation or the use of these components as an inoculant.
  • such a method for prophylaxis, treatment, and/or amelioration of the above-mentioned diseases or disorders, or an inoculation method for preventing the above-mentioned diseases typically comprises administering the described pharmaceutical composition to a patient in need thereof (e.g. suffering from any of the above diseases or showing symptoms thereof), in particular to a human being, preferably in a "safe and effective amount" and in one of the above formulations as described above for inventive pharmaceutical compositions.
  • the administration mode also may be as described above for inventive pharmaceutical compositions.
  • the present invention relates also to an in vitro transcription method for the preparation of inventive modified (m)RNA (of the inventive immunosuppressive composition), comprising the following steps: a) preparation/provision of a (desoxy)ribonucleic acid as a template for the inventive modified (m)RNA (of the inventive immunosuppressive composition), in particular a template as described above; b) addition of the (desoxy)ribonucleic acid to an in vitro transcription medium comprising a RNA polymerase, a suitable buffer, a nucleic acid mix, comprising one or more chemically modified nucleosides selected from the chemically modified nucleosides as defined above as replacement (partially or completely) for one or more of the naturally occurring nucleosides A, G, U and/or C, and optionally one or more naturally occurring nucleosides A, G, U and/or C if not all of the naturally occurring nucleosides A, G, U and/or C are to be replaced, and optionally an RNase inhibitor; c
  • a (desoxy)ribonucleic acid as described in step a) of the in vitro transcription method according to the invention can be any nucleic acid as described above that may be used as a template for the preparation of the modified (m)RNA of the present invention.
  • DNA sequences are used, for example genomic DNA or fragments thereof, or plasmids, or RNA sequences (corresponding thereto), for example mRNA sequences, preferably in linearized form.
  • the in vitro transcription reaction can usually be carried out using a vector having a RNA polymerase binding site. To this end there can be used any vectors known in the art, for example commercially available vectors (see above).
  • the (desoxy)ribonucleic acid sequences used can be transcribed later, as desired, depending on the chosen RNA polymerase.
  • a (desoxy)ribonucleic acid sequence used for in vitro transcription and coding for a (therapeutically active) protein, antigen or antibody as defined above is typically cloned into a vector, for example via a multiple cloning site of the vector used.
  • the clone Before the transcription, the clone is typically cleaved with restriction enzymes at the site at which the future 3' end of the modified (m)RNA is to be located, using a suitable restriction enzyme, and the fragment is purified. This prevents the future modified (m)RNA from containing vector sequences, and a modified (m)RNA of defined length may be obtained.
  • the (desoxy)ribonucleic acid as transcription template by polymerase chain reaction (PCR).
  • one of the primers used typically contains the sequence of a RNA polymerase binding site. It is further preferred for the 5 1 end of the primer used to have a length of approximately from 10 to 50 further nucleotides, more preferably from 15 to 30 further nucleotides and most preferably of approximately 20 nucleotides.
  • the (desoxy)ribonucleic acid e.g. the specific DNA or RNA template, is typically purified and free of RNase in order to ensure a high yield. Purification can be carried out by any process known in the art, for example with a caesium chloride gradient or ion-exchange process.
  • the (desoxy)ribonucleic acid is added to an in vitro transcription medium.
  • a suitable in vitro transcription medium first contains a (desoxy)ribonucleic acid as prepared under step a), for example approximately from 0.1 to 10 ⁇ g, preferably approximately from 1 to 5 ⁇ g, more preferably 2.5 ⁇ g and most preferably approximately 1 ⁇ g, of such a nucleic acid.
  • a suitable in vitro transcription medium further optionally contains a reducing agent, e.g. DTT, more preferably approximately from 1 to 20 ⁇ l of 50 mM DTT, yet more preferably approximately 5 ⁇ l of 50 mM DTT.
  • the in vitro transcription medium further contains nucleotides (AMP, GMP, UMP and/or CMP), for example a nucleotide mix.
  • nucleotides AMP, GMP, UMP and/or CMP
  • the nucleotides preferably comprise chemically modified nucleosides as defined above.
  • Such (chemically modified) nucleotides may serve as replacement for one or more of the naturally occurring nucleotides AMP, GMP, UMP and/or CMP, and optionally one or more naturally occurring nucleotides AMP, GMP, UMP and/or CMP, if not all of the naturally occurring nucleotides AMP, GMP, UMP and/or CMP are to be replaced.
  • the nucleotides AMP, GMP, UMP and/or CMP are typically present in the nucleotide mix in a concentration of typically approximately from 0.1 to 1 O mM per nucleotide, preferably from 0.1 to 1 mM per nucleotide, preferably approximately 4 mM in total.
  • Modified nucleotides as described above are typically added in such an amount that the native nucleotide is replaced completely by the (modified) nucleotide(s) comprising a chemically modified nucleoside as defined above.
  • a suitable in vitro transcription medium likewise contains a RNA polymerase, e.g. T7-RNA polymerase (e.g. T7-Opti mRNA Kit, Cure Vac, Tubingen, Germany), T3-RNA polymerase or SP6, typically approximately from 10 to 500 U, preferably approximately from 25 to 250 U, more preferably approximately from 50 to 150 U, and most preferably approximately 100 U of RNA polymerase.
  • T7-RNA polymerase e.g. T7-Opti mRNA Kit, Cure Vac, Tubingen, Germany
  • T3-RNA polymerase or SP6 typically approximately from 10 to 500 U, preferably approximately from 25 to 250 U, more preferably approximately from 50 to 150 U, and most preferably approximately 100 U of RNA polymerase.
  • the in vitro transcription medium is further preferably kept free of RNase in order to avoid degradation of the transcribed RNA.
  • a suitable in vitro transcription medium therefore optionally contains in addition a RNase inhibitor.
  • the (desoxy)ribonucleic acid is incubated and transcribed in the in vitro transcription medium, typically for approximately from 30 to 120 minutes, preferably for approximately from 40 to 90 minutes and most preferably for approximately 60 minutes, at approximately from 30 to 45°C, preferably at from 37 to 42°C.
  • the incubation temperature is governed by the RNA polymerase that is used, for example in the case of T7 RNA polymerase it is approximately 37°C.
  • the nucleic acid obtained by the transcription is preferably a modified (m)RNA as defined herein for the inventive immunosuppressive composition, more preferably an mRNA.
  • step d) of the in vitro transcription method according to the invention can optionally take place in step d) of the in vitro transcription method according to the invention.
  • any suitable process known in the art can be used, for example chromatographic purification processes, e.g. affinity chromatography, gel filtration, etc.
  • chromatographic purification processes e.g. affinity chromatography, gel filtration, etc.
  • non-incorporated, i.e. excess, nucleotides can be removed from the in vitro transcription medium.
  • Any suitable method known in the prior art e.g. chromatographic purification methods, e.g. affinity chromatography, gel filtration etc., can be used for this.
  • non- incorporated i.e.
  • RNA can be removed from the in vitro transcription medium and a clean modified (m)RNA can be obtained.
  • the reaction mixture containing the transcribed RNA can typically be digested with DNase in order to remove the DNA template still contained in the reaction mixture.
  • the transcribed RNA can be subsequently or alternatively precipitated with LiCI. Purification of the transcribed modified (m)RNA can then take place via IP RP-HPLC. This renders it possible in particular to separate longer and shorter fragments from one another effectively.
  • the purification takes place via a method for purification of RNA on a preparative scale, which is distinguished in that the RNA is purified by means of HPLC using a porous reverse phase as the stationary phase (PURE Messenger).
  • a reverse phase can be employed as the stationary phase for the HPLC purification.
  • a non-polar compound typically serves as stationary phase
  • a polar solvent such as mixtures of water, which is usually employed in the form of buffers, with acetonitrile and/or methanol, serves as the mobile phase for the elution.
  • the porous reverse phase has a particle size of 8.0 ⁇ 2 ⁇ m, preferably ⁇ 1 ⁇ m, more preferably +/- 0.5 ⁇ m.
  • the reverse phase material can be in the form of beads.
  • the purification can be carried out in a particularly favourable manner with a porous reverse phase having this particle size, optionally in the form of beads, particularly good separation results being obtained.
  • the reverse phase employed is preferably porous since with stationary reverse phases which are not porous, such as are described e.g. by Azarani A. and Hecker K.H., pressures which are too high are built up, so that preparative purification of the modified (m)RNA is possible, if at all, only with great difficulty.
  • the reverse phase preferably has a pore size of from 200 to 5,000 , in particular a pore size of from 300 to 4,000 .
  • Particularly preferred pore sizes for the reverse phases are 200 - 400 , 800 - 1,200 and 3,500 - 4,500 .
  • the material for the reverse phase is preferably a polystyrene-divinylbenzene, and non-alkylated polystyrene-divinylbenzenes can be employed in particular. Stationary phases with polystyrene-divinylbenzene are known per se.
  • the polystyrene-divinylbenzenes which are known per se and already employed for HPLC methods and are commercially obtainable can be used.
  • a non-alkylated porous polystyrene-divinylbenzene which in particular has a particle size of 8.0 ⁇ 0.5 ⁇ m and a pore size of 250 - 300 , 900 - 1,100 or 3,500 - 4,500 is very particularly preferably used for the purification in method step d).
  • the advantages described above can be achieved in a particularly favourable manner with this material for the reverse phases.
  • the HPLC purification can be carried out by the ion pair method, an ion having a positive charge being added to the mobile phase as a counter-ion to the negatively charged RNA.
  • An ion pair having a lipophilic character which is slowed down by the non-polar stationary phase of the reverse phase system, is formed in this manner.
  • the precise conditions for the ion pair method must be worked out empirically for each concrete separation problem.
  • the size of the counter-ion, its concentration and the pH of the solution contribute greatly towards the result of the separation.
  • alkylammonium salts such as triethylammonium acetate and/or tetraalkylammonium compounds, such as tetrabutylammonium
  • 0.1 M triethylammonium acetate is added and the pH is adjusted to about 7.
  • the choice of mobile phase depends on the nature of the desired separation. This means that the mobile phase found for a specific separation, such as can be known, for example, from the prior art, cannot be transferred readily to another separation problem with adequate prospect of success.
  • the ideal elution conditions, in particular the mobile phase used must be determined for each separation problem by empirical experiments.
  • a mixture of an aqueous solvent and an organic solvent can be employed as the mobile phase for elution of the modified (m)RNA by the HPLC method.
  • a buffer which has, in particular, a pH of about 7, for example 6.5 - 7.5, e.g. 7.0, is used as the aqueous solvent; preferably, the buffer triethylammonium acetate is used, particularly preferably a 0.1 M triethylammonium acetate buffer which, as described above, also acts as a counter-ion to the RNA in the ion pair method.
  • the organic solvent employed in the mobile phase can be acetonitrile, methanol or a mixture of these two, very particularly preferably acetonitrile.
  • the purification of the modified (m)RNA in method step d) using an HPLC method as described is carried out in a particularly favourable manner with these organic solvents.
  • the mobile phase is particularly preferably a mixture of 0.1 M triethylammonium acetate, pH 7, and acetonitrile. It has emerged to be likewise particularly favourable if the mobile phase contains 5.0 vol.% to 20.0 vol.% of organic solvent, based on the mobile phase, and the remainder to make up 100 vol.% is the aqueous solvent.
  • the mobile phase contains 9.5 vol.% to 14.5 vol.% of organic solvent, based on the mobile phase, and the remainder to make up 100 vol.% is the aqueous solvent.
  • Elution of the modified (m)RNA can subsequently be carried out isocratically or by means of a gradient separation.
  • elution of the modified (m)RNA is carried out with a single eluting agent or a mixture of several eluting agents which remains constant, it being possible for the solvents described above in detail to be employed as the eluting agent.
  • kits particularly kits of parts, comprising as components the modified (m)RNA, and/or the inventive immunosuppressive composition, and/or an inventive pharmaceutical composition, e.g. an inventive vaccine, and optionally technical instructions with information on the administration and dosage of these components.
  • kits preferably kits of parts, may applied e.g. for any of the above mentioned applications or uses.
  • Kits of parts as a special form of kits, may be furthermore used, when a time staggered treatment is envisaged, as described above, wherein the single parts of such a kit may contain either the same or different active immunosuppressive compositions and/or inventive vaccines according to the present invention.
  • Figure 1 depicts the luciferase construct (T7TS-Ppluc(wt)-A70) (SEQ ID NO: 3), which was used as a template for introducing the different chemically modified nucleic acids as defined herein in order to evaluate their immunosuppressive properties in comparison to unmodified RNA and a control (no RNA).
  • the coding sequence is underlined.
  • the construct furthermore contained a poly- A tail of about 70 adenosines at the 3 1 end.
  • FIG 2 depicts the immunosuppressive effect of the chemically modified (m)RNA according to the present invention. As can be seen in Figure 2, the modified
  • (m)RNAs modified with indicated chemically modified nucleosides lead to an abrogation or at least significant reduction in secretion of TNF-alpha in hPBMCs, i.e. an abrogation or at least significant reduction of the immunostimulatory effect, when compared to non-modified (m)RNA.
  • Figure 3 depicts the immunosuppressive effect of the chemically modified (m)RNA according to the present invention.
  • the modified (m)RNAs modified with indicated chemically modified nucleosides lead to an abrogation or at least significant reduction in secretion of IL-6 in hPBMCs, i.e. an abrogation or at least significant reduction of the immunostimulatory effect, when compared to non-modified (m)RNA.
  • the template sequences for the preparation of the modified RNA sequences are sequences comprising a coding sequence of luciferase.
  • the coding sequence was transferred into an RNActive construct (CureVac GmbH, Tubingen, Germany), which has been modified with a poly-A-tag, (A70).
  • the luciferase construct T7TS-Ppluc(wt)-A70 (SEQ ID NO: 3) was obtained as a template for the inventive modifications (see Fig. 1 ).
  • the luciferase construct (T7TS- Ppluc(wt)-A70) (SEQ ID NO: 3) (see Fig. 1 ) was linearized and subsequently in vitro transcribed by means of T7 polymerase (T7-Opti mRNA Kit, CureVac, Tubingen,
  • nucleotides comprising chemically modified nucleosides (2- Aminopurine-riboside-5'-(mono)phosphate; 2-Thiouridine-5'-(mono)phosphate ; 4- Thiouridine-5'-(mono)phosphate; 5-Aminoallylcytidine-5'-(mono)phosphate ; 5- Aminoallyluridine-5'-(mono)phosphate ; 5-Bromocytidine-5'-(mono)phosphate ; 5- Bromo-2'-deoxycytidine-5'-(mono)phosphate ; 5-Bromouridine-5'-(mono)phosphate;
  • nucleotide (mono)phosphates of AMP, GMP, CMP, and UMP.
  • the one remaining nucleotide is the nucleotide to be modified and comprises one of the above mentioned chemically modified nucleosides.
  • the DNA template was then degraded by
  • hPBMC peripheral blood mononuclear cells
  • hPBMc were thawed by resuspending in PBS, followed by two washes in PBS.
  • hPBMC were then suspended in X-Vivo 15, 1 % glutamine, 1 % Pen/Strep at a density of 5 x 10E5 / ml.
  • 50 ⁇ l of protamine-complexed mRNA (final concentration 3 ⁇ g RNA / ml) were added to stimulate/suppress cytokine release over night at 37°C. 5.
  • TNFalpha- and IL6-quantification ELISA
  • Gemini plate reader (Ex 540 nm, Em 590 nm, cutoff 590 nm).
  • nucleoside of the modified (m)RNAs has been modified.
  • the corresponding nucleotides are selected from 2- Aminopurine-riboside-5'-(mono)phosphate; 2-Thiouridine-5'-(mono)phosphate ; 4-
  • lymphocyte medium L-Glutamine (200 mM), Penicilline (Pen, 10000 U / ml) and Streptomycine (Strep, 10000 U / ml) were from Lonza.
  • biotinylated II6 und TNF detection antibodies as well as Streptavidin-coupled horseradish peroxidase (Strept-HRP) were from BD Pharmingen.

Abstract

The present invention relates to a modified (m)RNA suitable for suppressing and/or avoiding an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA and an immunosuppressive composition comprising this RNA. The invention furthermore relates to a pharmaceutical composition containing said modified (m)RNA. The invention also relates to the use of said modified (m)RNA or immunosuppressive composition (for the preparation of a medicament, e.g. a pharmaceutical composition) and/or the use of the pharmaceutical composition for suppressing and/or avoiding an immune response in a mammal when administering said pharmaceutical composition for the treatment of various diseases. Finally, the invention relates to kits containing the immunosuppressive composition and/or the pharmaceutical composition.

Description

Modified (m)RNA for suppressing or avoiding an immunostimulatory response and immunosuppressive composition
The present invention relates to a modified (m)RNA suitable for suppressing and/or avoiding an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA and an immunosuppressive composition comprising this RNA. The invention furthermore relates to a pharmaceutical composition containing said modified (m)RNA. The invention also relates to the use of said modified (m)RNA or immunosuppressive composition (for the preparation of a medicament, e.g. a pharmaceutical composition) and/or the use of the pharmaceutical composition for suppressing and/or avoiding an immune response in a mammal when administering said pharmaceutical composition for the treatment of various diseases. Finally, the invention relates to kits containing the immunosuppressive composition and/or the pharmaceutical composition.
Responses of the innate immune system play an important role in the treatment and prevention of numerous diseases. Such innate immune responses may lead to high levels of inflammatory cytokines and type I interferones. However, in many cases an innate immune activation represents a significant and undesirable side effect due to the toxicities associated with excessive cytokine release and associated inflammatory syndromes. In this context, the innate immune system as part of the immune system is the dominant system of host defense in most organisms and comprises barriers such as humoral and chemical barriers including, e.g., inflammation, the complement system and cellular barriers. The innate immune system is typically based on a small number of receptors, called pattern recognition receptors. They recognize conserved molecular patterns that distinguish foreign organism, like viruses, bacteria, fungi and parasites from cells of their hosts. Such pathogen-associated molecular patterns include viral nucleic acids, components of bacterial and fungal walls, flagellar proteins, and more.
The first family of pattern recognition receptors studied in detail and, up to date, the most prominent one, was the Toll-like receptor (TLR) familiy. TLRs are transmembrane proteins which recognize ligands of the extracellular milieu or of the lumen of endosomes. Following ligand-binding they transduce the signal via cytoplasmic adaptor proteins which leads to triggering of a host-defence response and entailing production of antimicrobial peptides, proinflammatory chemokines and cytokines, antiviral cytokines etc. (see e.g. Meylan, E., J. Tschopp, et al. (2006). "Intracellular pattern recognition receptors in the host response." Nature 442(7098): 39-44). To date, at least 10 members of Toll-like receptors (TLRs 1 -10) have been identified in human and 13 (TLRs 1 -13) in mice. Those Toll-like receptors (TLRs) in human include TLR1 -TLR2 (known ligand: Triacyl lipopeptide), TLRl - TLR6 (known ligand: Diacyl lipopeptide), TLR2 (known ligand: Peptidoglycan), TLR3 (known ligand: dsRNA), TLR4 (known ligand: LPS (lipopolysaccharide) of Gram-negative bacteria)), TLR5 (known ligand: bacterial flagellin(s)), TLR7/8 (known ligands: imidazoquinolines, guanosine analogs and ssRNA), TLR9 (known ligands: CpG DNA of bacteria, viruses and protozoans and malaria pigment hemozoin (product of digestion of haemoglobin)) and TLRIO. After recognition of microbial pathogens, these TLRs typically trigger intracellular signalling pathways that result in induction of inflammatory cytokines (e.g. TNF-alpha, IL-6, IL-1 -beta and IL-12), type I interferon (IFN-beta and multiple IFN- alpha) and chemokines (Kawai, T. and S. Akira (2006). "TLR signaling." Cell Death Differ 13(5): 816-25).
Apart from the adaptive immune system, which adapted over time to recognize particular pathogens or antigens more efficiently, the innate immune system and accordingly the ability of therapeutic agents to trigger an adequate but non-excessive innate immune response represents is a crucial key element with respect to the compatibility of many treatments. In some cases even a therapeutic agent may be desired, which allows to suppress an innate immune response or diminish it up to a certain extent. The choice of a suitable therapeutic agent, of course, is dependent on the disease to be treated. Hitherto, conventional methods of treatment may, inter alia, comprise the use of nucleic acid sequences, such as DNA or RNA. Nucleic acid sequences represent ligands of the Toll-like receptor (TLR) familiy as defined above, particularly of Toll-like receptors TLR3 (known ligand: dsRNA), TLR7/8 (known ligands: imidazoquinolines, guanosine analogs and ssRNA), and TLR9 (known ligands: CpG DNA of bacteria, viruses and protozoans and malaria pigment hemozoin (product of digestion of haemoglobin)). Typically, nucleic acid sequences such as DNA or RNA are used to incorporate required genetic information into the cell, particularly in gene therapeutic method of treatments, whereby those nucleic acids may unspecifically stimulate the innate immune repsonse. For these purposes, various methods have been developed for introducing DNA into cells, such as calcium phosphate transfection, polyprene transfection, protoplast fusion, electroporation, microinjection and lipofection. DNA viruses may likewise be used as a DNA vehicle. Such viruses achieve a very high transfection rate, because of their infectious properties. The viruses used are typically genetically modified in such a manner that no functional infectious particles are formed in the transfected cell. Despite these precautions, however, it is not possible to rule out the risk of uncontrolled propagation of the introduced gene. The same risk generally also arises, when using DNA, even if not derived from DNA viruses, e.g. due to uncontrolled recombination events of the introduced gene. This entails a risk of the DNA being inserted into an intact gene of the host cell's genome e.g. by recombination, with the consequence that this gene may be mutated and thus completely or partially inactivated or give rise to misinformation. The gene may be also affected by inactivation of essential elements thereof, such as promoters, enhancers or silencers, which lead to a different expression pattern, e.g. a silencing of the entire gene, wherein one particular risk occurs if the DNA is integrated into a gene which is involved in the regulation of cell growth.
A conceivable solution to the problem of uncontrolled recombination or propagation of the introduced DNA sequences may be the use of RNA as an approach for curative methods as outlined above. RNA expression systems may have considerable advantages over DNA expression systems, e.g., in immune response, immunization or vaccination. These advantages include, inter alia, that RNA introduced into a cell is not integrated into the genome. Another advantage includes that no viral sequences, such as promoters etc., are required for active transcription, when RNA is used as a coding nucleic acid. Another risk of using DNA as an agent to induce an immune response (e.g. as a vaccine) is the induction of pathogenic anti-DNA antibodies in the patient into whom the foreign DNA has been introduced, so bringing about a (possibly fatal) immune response. Furthermore, the use of (m)RNA does appear to induce production of antibodies as up to date, no anti-(m)RNA antibodies have yet been detected or are known in the art with the exception of antibodies against tRNAs and rRNAs (see e.g. Lupus erythematosus). It is assumed that the underlying mechanism is due to the fact that RNA is substantially more straightforwardly degraded in vivo, i.e. in the patient's body.
However, RNA sequences as well as DNA sequences may sometimes lead to excessive immune responses or even to toxicities associated with excessive cytokine release and associated inflammatory syndromes. In some cases, such as gene therapy or the treatment of allergies, therapeutic agents may be required, which are neutral, i.e. do not trigger an innate (and also no adaptive) immune response, or even immunosuppressive, i.e. lower an existing immune response of the innate immune system.
One possibility to reduce or lower the immunogenicity of RNA sequences may be the avoidance of any adjuvant, when administering the RNA sequence, which triggers an innate immune response. However, transfecting naked RNA sequences typically does not abolish the intrinsic properties of RNA per se to trigger an immune response due to its recognition by Toll-like receptors 3, 7 and 8. It is also to be noted that the immune stimulation is usually already triggered upon transfection of the RNA into the organism, e.g. in the endosom upon endocytosis. Accordingly, the intrinsic properties of RNA may be changed on molecular level. By way of example, it is known in the art, that immunogenicity of RNA (and of DNA) may be dependent or at least strongly influenced by specific molecular properties of the RNA (or DNA), such as structure, binding capacity, content of CpGs, (post- transcriptional) modification of nucleotides, etc.. As a further alternative, one may thus avoid specific nucleotides, which are known or assumed to trigger or enhance an innate immune response. Another alternative is to alter the modification rate of the nucleosides of the RNA, in order to change immunomodulatory properties of the RNA to be administered.
As an example, Kariko eta/. (Immunity, Vol. 23, 165-175, August 2005) were able to show that RNA signals through human TLR3, TLR7 and TLR8, but surprisingly incorporation of modified nucleosides N6-methyl-adenosine, 2-thiouridine, 5-methyl-cytidine, 5-methyl- uridine or pseudouridine ablated immunostimulatory activity to at least a certain extent instead of triggering an innate immune response. Furthermore, dendritic cells (DCs) exposed to such a modified (m)RNA expressed less cytokines and activation markers than those treated with an unmodified (m)RNA. Similarly, the PCT Application WO 2007/024708 A2, corresponding to Kariko eta/. (2005, supra), shows modifications of RNA sequences by using an RNA comprising N6-methyl-adenosine, 2-thiouridine, 5-methyl- cytidine, 5-methyl-uridine or pseudouridines. However, Kariko eta/. (2005, supra) stressed that the role of nucleoside modifications on the immuno-stimulatory potential and on the translation efficiency of RNA, however, is not known. Accordingly, there appears to be no common approach, to provide nucleic acid modifications, which both confer a lowered immunostimulatory property and allow translation of the nucleic acid molecule in vivo. However, this is particularly necessary for the use of longer RNA sequences, such as coding RNA, e.g. mRNA, sequences.
Likewise, WO 2008/019486 discloses nucleic acids with 2'-O-methyl modifications at uridine, guanosine and/or adenosine residues, which confer a lowered immunostimulatory property with respect to the unmodified nucleic acid. However, WO 2008/019486 discloses short nucleic acid sequences, which are administered in combination with (short) antisense oligonucleotides or siRNAs to confer the lowered immunostimulatory properties. WO 2008/019486 does not disclose the use of longer RNA sequences, such as coding RNA, e.g. mRNA, sequences or the translation of any such oligonucleotide.
Sioud et al. (Eur. J. Immunol., 2006, 36: 1222-1230) also report on chemical modifications, wherein those modifications are carried out on small interfering RNAs (siRNAs). Particularly, Sioud et al. (2006, supra) report, that replacement of 2'-hydroxyl uridines with either 2'-fluoro, 2'-deoxy or 2'-O-methyl-uridines is capable to abrogate the immune reaction of siRNAs. However, siRNAs are typically short double stranded nucleic acid molecules having a length of about 21 to 25 nucleotides. Such (short) siRNAs are intended to silence gene expression by binding to specific target sequences and thus, by their very nature, those sequences do not have to encode a protein sequence or have to show any capability for translation. WO 2006/009784 also discloses chemical modifications of siRNA molecules, wherein the si RNA strands essentially consist of ribonucleotides with 2'-hydroxyl substitutions on the ribonucleotide ribose. WO 2006/009784 does not disclose other modifications, which may modify the immune response or discusses such modifications with longer RNA molecules, such as e.g. coding RNA, e.g. mRNA, molecules.
Furthermore, WO 2007/031877 discloses modifications of siRNAs. WO 2007/031877 describes replacement of nucleotides by at least one nucleotide having a sugar with a 2'- modification, wherein the modified nucleotide shall not be a locked nucleotide or a 2'-O- methyl nucleotide. WO 2007/031877 discloses in detail the use of 2'-modifications selected from 2'-deoxy, 2'-fluoro, 2'-amino, 2'-methoxyethyl, 2'-O-allyl, 2'-propinyl, 2'- aminopropargyl, 2'-O-(3-aminopropyl), 2'-O-propyl, 2'-O-butyl, 2'-O-alkenyl, 2'-O-alkinyl, 2'-methoxyethyl, 2'-aminopropargyl, 2'-O-(3-aminopropyl) and 2'-amino. Likewise, WO 2007/031877 focusses on siRNA, but does not give any indication as to whether such modifications are suitable for translation of longer RNA molecules or provides such specific modifications.
Similarly, Judge et al. (HUMAN GENE TERAPY 19:000-000, February 2008) report that chemical modification of siRNAs with nucleotides comprising 2'-OH-substitutions such as 2'-O-methyl (2'-OMe) and 2'-O-fluoro (2'-OF) and 2'deoxy, can lead to an abrogation of immune stimulation in these siRNA molecules. For such an effect, more than 90% of the nucleotides of these siRNAs were chemically modified according to the pattern of purines and pyrimidines in a given siRNA sequence. However, Judge et al. (2008, supra) did not show that such chemical modifications are capable to suppress the immunostimulatory properties of mRNA.
As a consequence, modified nucleotides per se do not necessarily trigger or inhibit an immune response, as inhibiting or lowering the immune response by modification of nucleotides appears to be strongly dependent on the nature of the single modified nucleotide. However, since there appear to be innumerable possibilities to modify such nucleotides, modifications of nucleotides per se do also not appear to provide an overall solution to the above problem. Accordingly, there is still the need in the art for providing an RNA, preferably a longer RNA such as a coding RNA, e.g. a mRNA, sequence, which is capable to confer both a lowered immunostimulatory property with respect to the unmodified RNA and allows translation of the RNA molecule in vivo.
This object is solved by the subject matter of the present invention, particularly by a modified (m)RNA or an immunossuppressive composition comprising at least one such modified (m)RNA, wherein at least one nucleoside of the modified (m)RNA has been modified with: a) a chemical modification at the 4-, 5-or 6-position of the pyrimidine base of the nucleosides of cytidine and/or uridine; b) a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides of adenosine, inosine and/or guanosine; and/or c) a chemical modification at the 2'-position of the sugar of the nucleosides of adenosine, inosine, guanosine, cytidine and/or uridine, wherein the at least one modified (m)RNA is suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA.
An immunosuppressive composition according to the present invention shall be understood as a composition, which is capable to suppress and/or avoid an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, preferably an innate immune response as defined herein, due to the at least one modified (m)RNA as contained in the inventive immunosuppressive composition. In this context, a mammal as mentioned throughout the description of the present invention may be selected from any mammal, preferably from a mammal, selected from the group comprising, without being limited thereto, e.g. goat, cattle, swine, dog, cat, donkey, monkey, ape, a rodent such as a mouse, hamster, rabbit, and, in particular, human.
In this context, an (m)RNA is a nucleic acid chain formed by a number of nucleotides typically selected from adenosine-5'-monophosphate, guanosine-5'-monophosphate, inosine-5'-monophosphate, cytidine-5'-monophosphate and/or uridine-5'-monophosphate. Those nucleotides are linked to each other via their monophosphate. Nucleotides comprise nucleosides and a 5'-monophosphate as a structural component, wherein the nucleosides are typically formed by a nucleobase, i.e. a pyrimidine (uracil or cytosine) or a purine (adenine or guanine) base, and a sugar. Accordingly, a modification of a nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition) is always intended to mean a modification in the nucleoside structure of the respective nucleotide of said (m)RNA.
According to a first modification a), at least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response typically exhibited when administering the corresponding unmodified (m)RNA in a mammal, may be modified with a chemical modification at the 5- or 6-position of the pyrimidine base of the nucleosides cytidine and/or uridine. Without being limited thereto, such chemical modifications at the 4-, 5- or 6-position of the base pyrimidine of the nucleosides cytidine and/or uridine may be selected from the group consisting of:
Figure imgf000009_0001
Figure imgf000010_0001
According to a second modification b), at least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, may be alternatively modified with a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine. Without being limited thereto, such chemical modifications at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine may be selected from the group consisting of:
Figure imgf000010_0002
According to a third modification c), at least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an
(innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, may be modified with at least one chemical modification at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine, when incorporated in the RNA sequence. Without being limited thereto, such chemical modifications at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine may be selected from the group consisting of:
No. Modification at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine
41 2'-deoxy-
42 2/-amino-2'-deoxy- / 2'-amino-
43 2'-fluoro-2'-deoxy- / 2'-fluoro-
44 2'-O-methyl-2'-deoxy- / 2'-O-methyl-
According to a particularly preferred embodiment, at least one nucleoside of the modified (m)RNA (of the inventive immunosuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, has been modified at the 4,- 5- or 6- position of the base pyrimidine of the nucleosides cytidine and/or uridine and at the T- position of the ribose sugar according to modifications a) and c) as defined above, more preferably as shown in the following:
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
163 5-taurinomethyl-2'-amino-
164 5-taurinomethyl-2'-fluoro-
165 5-taurinomethyl-2'-O-methyl-
166 5-taurinomethy)-2-thiouridine-2'-deoxy-
167 5-taurinomethyl-2-thiouridine-2'-amino-
168 5-taurinomethyl-2-thiouridine-2'-fluoro-
169 5-taurinomethyl-2-thiouridine-2'-O-methyl-
170 5-isopentenylaminomethyl-2'-deoxy-
171 5-isopentenylaminomethyl-2'-amino-
172 5-isopentenylaminomethyl-2'-fluoro-
173 5-isopentenylaminomethyl-2'-O-methyl-
174 5-isopentenylaminomethyl-2-thio-2'-deoxy-
175 5-isopentenylaminomethyl-2-thio-2'-amino-
1 76 5-isopentenylaminomethyl-2-thio-2'-fluoro-
1 77 5-isopentenylaminomethyl-2-thio-2'-O-methyl-
178 5-aminopropyl-2'-deoxy-
1 79 5-aminopropyl-2'-amino-
180 5-aminopropyl-2'-fluoro-
181 5-aminopropyl-2'-O-methyl-
182 5-methoxy-ethoxy-methyl-2'-deoxy-
183 5-methoxy-ethoxy-methyl-2'-amino-
184 5-methoxy-ethoxy-methyl-2/-fluoro-
185 5-methoxy-ethoxy-methyl-2'-O-methyl-
186 6-aza-2'-deoxy-
187 6-aza-2'-amino-
188 6-aza-2'-fluoro-
189 6-aza-2'-O-methyl-
According to another particularly preferred embodiment, at least one nucleoside of the modified (m)RNA (of the inventive immunossuppressive composition), suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, has been modified at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine and at the 2'-position of the ribose sugar according to modifications b) and c) as defined above, more preferably as shown in the following:
Figure imgf000014_0001
194 7-Deaza-2'-deoxy-
195 7-Deaza-2'-amino-
196 7-Deaza-2'-fluoro-
197 7-Deaza-2'-O-methyl-
198 8-Aza-2'-deoxy-
199 8-Aza-2'-amino-
200 8-Aza-2'-fluoro-
201 8-Aza-2'-O-methyl-
202 8-Azido-2'-deoxy-
203 8-Azido-2 '-ami no-
204 8-Azido-2'-fluoro-
205 8-Azido-2'-O-methyl-
According to an even more particularly preferred embodiment, at least one nucleoside of the modified (m)RNA, suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, has been modified leading to chemically modified nucleotides (of the (m)RNA) selected from the following group: 4-thio-uridine-5'- (mono)phosphate, 2-Aminopurine-riboside-5'-(mono)phosphate, 5-Aminoallylcytidine-5'- (mono)phosphate , 5-Aminoallyluridine-5'-(mono)phosphate , 5-Bromocytidine-5'- (mono)phosphate, 5-Bromo-2'-deoxycytidine-5'-(mono)phosphate, 5-Bromouridine-5'-
((mmoonnoo))pphhoosspphhaattee,, 5-Bromo-2'-deoxyuridine-5'-(mono)phosphate, 5-lodocytidine-5'- (mono)phosphate, 5-lodo-2'-deoxycytidine-5'-(mono)phosphate, 5-lodouridine-5'- (mono)phosphate, 5-lodo-2'-deoxyuridine-5'-(mono)phosphate, 5-Propynyl-2'- deoxycytidine-5'-(mono)phosphate, 5-Propynyl-2'-deoxyuridine-5'-(mono)phosphate, 5- formylcytidine-5'-(mono)phosphate, 5,2'-0-dimethylcytidine-5'-(mono)phosphate, 5- hydroxymethylcytidine-5'-(mono)phosphate, 5-formyl-2'-0-methylcytidine-5'-
(mono)phosphate, 5,2'-0-dimethyluridine-5'-(mono)phosphate, 5-methyl-2-thiouridine-5'- (mono)phosphate, 5-hydroxyuridine-5'-(mono)phosphate, 5-methoxyuridine-5'-
(mono)phosphate, uridine 5-oxyacetic acid-5'-(mono)phosphate, uridine 5-oxyacetic acid methyl ester-5'-(mono)phosphate, 5-(carboxyhydroxymethyl)uridine-5'-(mono)phosphate, 5- (carboxyhydroxymethyl)uridine methyl ester-5'-(mono)phosphate, 5- methoxycarbonylmethyluridine-5'-(mono)phosphate, 5-methoxycarbonylmethyl-2'-0- methyluridine-5'-(mono)phosphate, 5-methoxycarbonylmethyl-2-thiouridine-5'-
(mono)phosphate, 5-aminomethyl-2-thiouridine-5'-(mono)phosphate, 5- methylaminomethyluridine-5'-(mono)phosphate, 5-methylaminomethyl-2-thiouridine-5'- (mono)phosphate, 5-methylaminomethyl-2-selenouridine-5'-(rnono)phosphate, 5- carbamoylmethyluridine-5'-(mono)phosphate, 5-carbamoylmethyl-2'-0-methyluridine-5'- (mono)phosphate, 5-carboxymethylaminomethyluridine-5'-(mono)phosphate, 5- carboxymethylaminomethyl-2'-0-methyluridine-5'-(mono)phosphate, 5- carboxymethylaminomethyl-2-thiouridine-5'-(mono)phosphate, 5-carboxymethyluridine-5'- (mono)phosphate, 5-methyldihydrouridine-5'-(mono)phosphate, 5-taurinomethyluridine-5'- (mono)phosphate, 5-taurinomethyl-2-thiouridine-5'-(mono)phosphate, 5-
(isopentenylaminomethyl)uridine-5'-(mono)phosphate, 5-(isopentenylaminomethyl)-2- thiouridine-5'-(mono)phosphate, 5-(isopentenylaminomethyl)-2'-0-methyluridine-5'-
(mono)phosphate, 6-Azacytidine-5'-(mono)phosphate, 7-Deazaadenosine-5'- (mono)phosphate, 7-Deazaguanosine-5'-(mono)phosphate, 8-Azaadenosine-5'-
(mono)phosphate, 8-Azidoadenosine-5'-(mono)phosphate, Pseudouridine-51-
(mono)phosphate, 2'-Amino-2'-deoxycytidine-(mono)phosphate, 2'-Fluorothymidine-5'- (mono)phosphate, inosine-5'-(mono)phosphate, 2'-O-Methyl-inosine-5'-(mono)phosphate.
According to a most particularly preferred embodiment, at least one nucleoside of the modified (m)RNA, suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal, has been modified leading to chemically modified nucleotides selected from the following group: 4-thio-uridine-5'-(mono)phosphate, 5-Aminoallyluridine- 5'-(mono)phosphate, 5-Bromo-2'-deoxycytidine-5'-(mono)phosphate, 5-Bromouridine-5'- (mono)phosphate, 5-Bromo-2'-deoxyuridine-5'-(mono)phosphate, 5-lodouridine-5'- (mono)phosphate, 5-lodo-2'-deoxyuridine-5'-(mono)phosphate, 5-Propynyl-2'- deoxyuridine-5'-(mono)phosphate, 6-Azacytidine-5'-(mono)phosphate, 8-Azidoadenosine- 5'-(mono)phosphate, 2'-Fluorothymidine-5'-(mono)phosphate, inosine-5'-(mono)phosphate and 2 '-O-Methyl-i nosi ne-5 ' -(mono)phosphate.
According to a particularly preferred embodiment, chemically modified nucleotides 5- methylcytidine-5'-(mono)phosphate, 5-methyluridine-5'-(mono)phosphate do not form part of the present disclosure and are explicitely disclaimed herewith from the at least one modified (m)RNA according to the present invention and the inventive immunosuppressive composition.
According to a further preferred embodiment, the at least one modified (m)RNA (of the inventive immunosuppressive composition) comprises more than one chemically modified nucleoside selected from the chemically modified nucleosides as defined above, more preferably at least two, three or four of these chemically modified nucleosides, or even more. Typically, any natively occurring nucleoside, which is contained in the natively occurring template (m)RNA of the at least one modified (m)RNA (of the inventive immunosuppressive composition) and which is to be substituted in the context of the present invention with a chemically modified nucleoside as defined above, preferably substitutes a corresponding nucleoside, i.e. adenosine of a natively occurring AMP (adenosine-5'-(mono)phosphate) will be substituted with a chemically modified adenosine as defined above (e.g. leading to 8-Azidoadenosine-5'-(mono)phosphate, etc.), cytidine of a natively occurring CMP (cytidine-5'-(mono)phosphate) will be substituted with a chemically modified cytidine as defined above (e.g. leading to 5-Bromo-2'-deoxycytidine-5'- (mono)phosphate, etc.), guanosine of a natively occurring GMP (guanosine-5'- (mono)phosphate) will be substituted with a chemically modified guanosine as defined above (e.g. leading to 7-Deazaguanosine-5'-(mono)phosphate, etc.) or with an inosine, and uridine of a natively occurring UMP (uridine-5'-(mono)phosphate) will be substituted with a chemically modified uridine as defined above (e.g. leading to 5-Aminoallyluridine-5'- (mono)phosphate, etc.) or by a modified TMP (e.g. 2'-Fluorothymidine-5'-(mono)phosphate, etc.), which carries the above modifications described for UMP, etc.. Further modifications may also be envisaged, e.g. GC-stabilization of the at least one modified (m)RNA (of the inventive immunosuppressive composition), as defined below. In the context of a GC- stabilization of the at least one modified (m)RNA (of the inventive immunosuppressive composition) the GC-stabilized (m)RNA may be regarded as the natively occurring template (m)RNA, to be modified with chemically modified nucleosides as defined above. Preferably, the at least one modified (m)RNA (of the inventive immunosuppressive composition) comprises between 0.1 % and 100% chemically modified nucleotides as defined above, wherein more preferably between 0.1 % and 100% of each natively occurring non-modified adenosine, guanosine, uridine and/or cytidine nucleoside, respectively, of the corresponding non-modified (m)RNA (template) may be modified using any of the corresponding chemically modified adenosine, guanosine, uridine and/or cytidine nucleosides, respectively, as defined above. In other words, preferably between 0.1 % and 100% of the adenosine, guanosine, uridine and/or cytidine nucleosides natively occurring in the corresponding non-modified (m)RNA (template) are modified in the at least one modified (m)RNA (of the inventive immunosuppressive composition) using the above defined (corresponding) chemically modified adenosine, guanosine, uridine and/or cytidine nucleoside, respectively, in a range of between 0,1 % and 20%, between 10% and 30%, between 20% and 40%, between 30% and 50%, between 40% and 60%, between 50% and 70%, between 60% and 80%, between 70% and 90%, or between 80% and 100% or at least 10%, more preferably at least 30%, more preferably at least 40%, more preferably at least 60%, more preferably at least 70%, more preferably at least 80% and more preferably at least 90% and most preferably 100% of all natively occurring adenosine, guanosine, uridine and/or cytidine nucleosides, respectively. The above selection may be applied to the entire sequence as well as to the content of the single adenosine, guanosine, uridine and/or cytidine nucleosides, respectively, of the modified (m)RNA sequence, i.e. preferably between 0.1 % and 100% in the above ranges of the adenosine, guanosine, uridine and/or cytidine nucleosides, respectively, natively occurring in the corresponding non-modified (m)RNA template sequence are chemically modified in the at least one modified (m)RNA (of the inventive immunosuppressive composition) using the above defined chemically modified nucleosides. Accordingly, the modified (m)RNA (of the inventive immunosuppressive composition) may exlusively contain chemically modified adenosine, guanosine, uridine and/or cytidine nucleosides as defined above in the above ranges or a mixture thereof in the above ranges.
In order to determine the percentage to which two (modified and/or natively occurring) RNA sequences (nucleic or amino acid) are identical, the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. gaps can be inserted into the sequence of the first (modified) RNA sequence and the component at the corresponding position of the second (modified) RNA sequence can be compared. If a position in the first (modified) RNA sequence is occupied by the same component as is the case at a position in the second (modified) RNA sequence, the two sequences are identical at this position. The percentage to which two (modified) RNA sequences are identical is a function of the number of identical positions divided by the total number of positions. The same, of course also applies accordingly to amino acid sequences encoded by these RNA sequences.
The percentage to which two sequences (either amino or nucleic acid sequences) are identical can be determined using a mathematical algorithm. A preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et a/. (1993), PNAS USA, 90:5873-5877 or Altschul et a/. (1997), Nucleic Acids Res, 25:3389- 3402. Such an algorithm is integrated in the BLAST or NBLAST program. Sequences which are identical to the sequences of the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) (or to the coding region thereof) to a certain extent can be identified by these programmes.
Those RNA sequences (corresponding to the at least one modified (m)RNA of the inventive immunosuppressive composition) encoding amino acid sequences which have (a) conservative substitution(s) compared to the physiological sequence in particular fall under the term variants. Substitutions in which encoded amino acids which originate from the same class are exchanged for one another are called conservative substitutions and are encompassed herein. In particular, these are encoded amino acids, encoded aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or encoded amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function. This means that e.g. an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g. serine (threonine) by threonine (serine) or leucine (isoleucine) by isoleucine (leucine)). Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g. using CD spectra (circular dichroism spectra) (Urry, 1985, Absorption, Circular Dichroism and ORD of Polypeptides, in: Modern Physical Methods in Biochemistry, Neuberger eta/, (ed.), Elsevier, Amsterdam).
The at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be any RNA, preferably, without being limited thereto, a short RNA oligonucleotide, a coding RNA, a siRNA, an antisense RNA, or riboswitches, ribozymes or aptamers. Furthermore, the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a single- or a double-stranded RNA (which may also be regarded as an RNA (molecule) due to non-covalent association of two single- stranded RNA (molecules)) or a partially double-stranded RNA (which is typically formed by a longer and a shorter single-stranded RNA molecule or by two single stranded RNA- molecules, which are about equal in length, wherein one single-stranded RNA molecule is in part complementary to the other single-stranded RNA molecule and both thus form a double-stranded RNA molecule in this region). Preferably, the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a single- stranded RNA. Furthermore, the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a circular or linear RNA, preferably a linear RNA. More preferably, the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a (linear) single-stranded RNA. The at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a ribosomal RNA (rRNA), a transfer RNA (tRNA), a messenger RNA (mRNA), or a viral RNA (vRNA), more preferably an mRNA. The present invention allows all of these RNAs to be transfected into the cell. In the context of the present invention, an mRNA is typically an RNA, which is composed of several structural elements, e.g. an optional 5'-UTR region, an upstream positioned ribosomal binding site followed by a coding region, an optional 3'-UTR region, which may be followed by a poly-A tail (and/or a poly-C-tail). An mRNA may occur as a mono-, di-, or even multicistronic RNA, i.e. an RNA which carries the coding sequences of one, two or more proteins. Such coding sequences in di-, or even multicistronic mRNA may be separated by at least one IRES sequence, e.g. as defined herein. Preferably, such an inventive modified (m)RNA (of the immunosuppressive composition) as defined herein, comprises a length of about 5 to about 20000, or 100 to about 20000 nucleotides, preferably of about 250 to about 20000 nucleotides, more preferably of about 500 to about 10000, even more preferably of about 500 to about 5000.
Coding RNA
According to a first embodiment, the at least one modified (m)RNA (of the immunosuppressive composition) of the present invention may be a coding RNA. Such a coding RNA may be any RNA as defined above. Preferably, such a coding RNA may be a single- or a double-stranded RNA, more preferably a single-stranded RNA, and/or a circular or linear RNA, more preferably a linear RNA. Even more preferably, the coding RNA may be a (linear) single-stranded RNA. Most preferably, the coding RNA may be a ((linear) single-stranded) messenger RNA (mRNA). The coding RNA, used as the inventive at least one modified (m)RNA (of the immunosuppressive composition) according to the present invention, may encode a protein or a peptide, which may be selected, without being restricted thereto, e.g. from therapeutically active proteins or peptides, antibodies, antigens, allergens, etc., from epitopes therof, i.e. from the above therapeutically active proteins or peptides, antibodies, antigens, allergens, etc., or from any other protein or peptide suitable for a specific (therapeutic) application, wherein the at least one modified coding RNA (molecule) of the inventive immunosuppressive composition is to be transported into a cell, a tissue or an organism and the protein is expressed subsequently in this cell, tissue or organism.
a) Therapeutically active proteins or peptides
In this context, therapeutically active proteins encoded by the at least one modified (m)RNA (of the immunosuppressive composition) according to the present invention may be selected from any recombinant or isolated proteins known to a skilled person from the prior art. Without being restricted thereto therapeutically active proteins may comprise proteins, capable of stimulating or inhibiting the signal transduction in the cell,
Therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may be selected from apoptotic factors or apoptosis related proteins including AIF, Apaf e.g. Apaf-1 , Apaf-2, Apaf-3, oder APO-2
(L), APO-3 (L), Apopain, Bad, Bak, Bax, Bcl-2, Bcl-xL, Bcl-xs, bik, CAD, Calpain, Caspase e.g. Caspase-1 , Caspase-2, Caspase-3, Caspase-4, Caspase-5, Caspase-6, Caspase-7, Caspase-8, Caspase-9, Caspase-10, Caspase-1 1 , ced-3, ced-9, c-Jun, c-Myc, crm A, cytochrom C, CdR1 , DcR1 , 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, granzyme A/B, ICAD, ICE, JNK, lamin A/B, MAP, MCL-1 , Mdm-2, MEKK- 1 , MORT-1, NEDD, NF-kappaB, NuMa, p53, PAK-2, PARP, perforin, PITSLRE, PKCdelta, pRb, presenilin, prlCE, RAIDD, Ras, RIP, sphingomyelinase, thymidinkinase from herpes simplex, TRADD, TRAF2, TRAIL-R1 , TRAIL-R2, TRAIL-R3, transglutaminase, etc..
Therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from recombinant proteins, including proteins selected from the group consisting of 0ATL3, 0FC3, 0PA3, 0PD2, 4-1 BBL, 5T4, 6Ckine, 707-AP, 9D7, A2M, AA, AAAS, AACT, AASS, ABAT, ABCA1, ABCA4, ABCBI, ABCBIl, ABCB2, ABCB4, ABCB7, ABCC2, ABCC6, ABCC8, ABCD1, ABCD3, ABCG5, ABCG8, ABL1, ABO, ABR ACAA1, ACACA, ACADL, ACADM, ACADS, ACADVL, ACAT1, ACCPN, ACE, ACHE, ACHM3, ACHM1, ACLS, ACPI, ACTA1, ACTC, ACTN4, ACVRL1, AD2, ADA, ADAMTS13, ADAMTS2, ADFN, ADH 1 B,
ADH1C, ADLDH3A2, ADRB2, ADRB3, ADSL, AEZ, AFA, AFD1, AFP, AGA, AGL, AGMX2, AGPS, AGSI, AGT, AGTR1, AGXT, AH02, AHCY, AHDS, AHHR, AHSG, AIC, AIED, AIH2, AIH3, AIM-2, AIPL1, AIRE, AK1, ALAD, ALAS2, ALB, HPG1, ALDH2, ALDH3A2, ALDH4A1, ALDH5A1, ALDH1A1, ALDOA, ALDOB, ALMS1, ALPL, ALPP, ALS2, ALX4, AMACR, AMBP, AMCD, AMCD1, AMCN, AMELX, AMELY, AMGL, AMH,
AMHR2, AMPD3, AMPD1, AMT, ANC, ANCR, ANK1, ANOP1, AOM, AP0A4, AP0C2, AP0C3, AP3B1, APC, aPKC, APOA2, APOA1, APOB, APOC3, APOC2, APOE, APOH, APP, APRT, APS1, AQP2, AR, ARAF1, ARG1, ARHGEF12, ARMET, ARSA, ARSB, ARSC2, ARSE, ART-4, ARTC1/m, ARTS, ARVD1, ARX, AS, ASAH, ASAT, ASD1, ASL, ASMD, ASMT, ASNS, ASPA, ASS, ASSP2, ASSP5, ASSP6, AT3, ATD, ATHS, ATM, ATP2A1,
ATP2A2, ATP2C1, ATP6B1, ATP7A, ATP7B, ATP8B1, ATPSK2, ATRX, ATXN1, ATXN2, ATXN3, AUTS1, AVMD, AVP, AVPR2, AVSD1, AXIN1, AXIN2, AZF2, B2M, B4GALT7, B7H4, BAGE, BAGE-1, BAX, BBS2, BBS3, BBS4, BCA225, BCAA, BCH, BCHE, BCKDHA, BCKDHB, BCL10, BCL2, BCL3, BCL5, BCL6, BCPM, BCR, BCR/ABL, BDC, BDE, BDMF, BDMR, BEST1, beta-Catenin/m, BF, BFHD, BFIC, BFLS, BFSP2, BGLAP,BGN, BHD,
BHR1, BING-4, BIRC5, BJS, BLM, BLMH, BLNK, BMPR2, BPGM, BRAF, BRCA1, BRCA1/m, BRCA2, BRCA2/m, BRCD2, BRCD1, BRDT, BSCL, BSCL2, BTAA, BTD, BTK, BUBl, BWS, BZX, C0L2A1, C0L6A1, C1NH, C1QA, C1QB, C1QG, C1S, C2, C3, C4A, C4B, C5, C6, C7, C7orf2, C8A, C8B, C9, CA125, CA15-3/CA 27-29, CA195, CA19-9, CA72-4, CA2, CA242, CA50, CABYR, CACD, CACNA2D1, CACNA1A, CACNA1 F,
CACNA1S, CACNB2, CACNB4, CAGE, CA1, CALB3, CALCA, CALCR, CALM, CALR, CAM43, CAMEL, CAP-1, CAPN3, CARD15, CASP-5/m, CASP-8, CASP-8/m, CASR, CAT, CATM, CAV3, CB1, CBBM, CBS, CCA1, CCAL2, CCAL1, CCAT, CCL-1, CCL-Il, CCL- 12, CCL-13, CCL-14, CCL-15, CCL-16, CCL-17, CCL-18, CCL-19, CCL-2, CCL-20, CCL- 21, CCL-22, CCL-23, CCL-24, CCL-25, CCL-27, CCL-3, CCL-4, CCL-5, CCL-7, CCL-8,
CCM1, CCNB1, CCND1, CCO, CCR2, CCR5, CCT, CCV, CCZS, CD1, CD19, CD20, CD22, CD25, CD27, CD27L, cD3, CD30, CD30, CD30L, CD33, CD36, CD3E, CD3G, CD3Z, CD4, CD40, CD40L, CD44, CD44v, CD44v6, CD52, CD55, CD56, CD59, CD80, CD86, CDANI, CDAN2, CDAN3, CDC27, CDC27/m, CDC2L1, CDHl, CDK4, CDK4/m, CDKN1C, CDKN2A, CDKN2A/m, CDKN1A, CDKN1C, CDL1, CDPDl, CDR1, CEA, CEACAMl, CEACAM5, CECR, CECR9, CEPA, CETP, CFNS, CFTR, CGF1, CHAC, CHED2, CHED1, CHEK2, CHM, CHML, CHR39C, CHRNA4, CHRNAI, CHRNBl, CHRNE, CHS, CHS1, CHST6, CHX10, CIAS1, CIDX, CKN1, CLA2, CLA3, CLAI, CLCA2,
CLCN1, CLCN5, CLCNKB, CLDN16, CLP, CLN2, CLN3, CLN4, CLN5, CLN6, CLN8, C1QA, C1QB, C1QG, C1R, CLS, CMCWTD, CMDJ, CMD1A, CMD1B, CMH2, MH3, CMH6, CMKBR2, CMKBR5, CML28, CML66, CMM, CMT2B, CMT2D, CMT4A, CMT1A, CMTX2, CMTX3, C-MYC, CNA1, CND, CNGA3, CNGA1, CNGB3, CNSN, CNTF, COA- 1/m, COCH, COD2, COD1, COHI, COL10A, COL2A2, COL11A2, COL17A1, COL1A1,
COL1A2, COL2A1, COL3A1, COL4A3, COL4A4, COL4A5, COL4A6, COL5A1, COL5A2, COL6A1, COL6A2, COL6A3, COL7A1, COL8A2, COL9A2, COL9A3, COL11A1, COL1A2, COL23A1, COL1A1, COLQ, COMP, COMT, CORD5, CORD1, COX10, COX-2, CP, CPB2, CPO, CPP, CPS1, CPT2, CPT1A, CPX, CRAT, CRB1, CRBM, CREBBP, CRH, CRHBP, CRS, CRV, CRX, CRYAB, CRYBA1, CRYBB2, CRYGA, CRYGC,
CRYGD, CSA, CSE, CSF1R, CSF2RA, CSF2RB, CSF3R, CSF1R, CST3, CSTB, CT, CT7, CT- 9/BRD6, CTAA1, CTACK, CTEN, CTH, CTHM, CTLA4, CTM, CTNNB1, CTNS, CTPA, CTSB, CTSC, CTSK, CTSL, CTS1, CUBN, CVD1, CX3CL1, CXCL1, CXCL10, CXCLl 1, CXCL12, CXCL13, CXCL16, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, CYB5, CYBA, CYBB, CYBB5, , CYFRA 21-1, CYLD, CYLD1, CYMD, CYP11B1,
CYP11B2, CYP17, CYP17A1, CYP19, CYP19A1, CYP1A2, CYP1B1, CYP21A2, CYP27A1, CYP27B1, CYP2A6, CYP2C, CYP2C19, CYP2C9, CYP2D, CYP2D6, CYP2D7P1, CYP3A4, CYP7B1, CYPB1, CYP11B1, CYP1A1, CYP1B1, CYRAA, D40,DADl, DAM, DAM-10/MAGE-B1, DAM-6/MAGE-B2, DAX1, DAZ, DBA, DBH, DBI, DBT, DCC, DC- CK1, DCK, DCR, DCX, DDB 1, DDB2, DDIT3, DDU, DECR1, DEK-CAN, DEM, DES,
DF,DFN2, DFN4, DFN6, DFNA4, DFNA5, DFNB5, DGCR, DHCR7, DHFR, DHOF, DHS, DIAI, DIAPH2, DIAPH1, DIH1, DIO1, DISCI, DKC1, DLAT, DLD, DLL3, DLX3, DMBT1, DMD, DM1, DMPK, DMWD, DNAM, DNASE1, DNMT3B, DPEP1, DPYD, DPYS, DRD2, DRD4, DRPLA, DSCR1, DSG1, DSP, DSPP, DSS, DTDP2, DTR, DURS1, DWS, DYS, DYSF, DYT2, DYT3, DYT4, DYT2, DYT1, DYX1, EBAF, EBM, EBNA, EBP,
EBR3, EBS1, ECA1, ECB2, ECE1, ECGF1, ECT, ED2, ED4, EDA, EDAR, ECA1, EDN3, EDNRB, EECI, EEF1A1L14, EEGV1, EFEMP1, EFTUD2/m, EGFR, EGFR/Heri, EGI, EGR2, EIF2AK3, elF4G, EKV, El IS, ELA2, ELF2, ELF2M, ELK1, ELN, ELONG, EMD, EML1, EMMPRIN, EMX2, ENA-78, ENAM, END3, ENG, ENO1, ENPPl, ENUR2, ENUR1, EOS, EP300, EPB41, EPB42, EPCAM, EPD, EphA1, EphA2, EphA3, EphrinA2, EphrinA3, EPHX1, EPM2A, EPO,EPOR, EPX, ERBB2, ERCC2 ERCC3,ERCC4, ERCC5, ERCC6, ERVR, ESR1, ETFA, ETFB, ETFDH, ETM1, ETV6-AML1, ETV1, EVC, EVR2, EVR1, EWSR1, EXT2, EXT3, EXT1, EYA1, EYCL2, EYCL3, EYCL1, EZH2, F10, F11, F12, F13A1, F13B, F2, F5,
F5F8D, F7, F8, F8C, F9, FABP2, FACL6, FAH, FANCA, FANCB, FANCC, FANCD2, FANCF, FasL,FBN2, FBN1, FBP1, FCG3RA,FCGR2A, FCGR2B, FCGR3A, FCHL, FCMD, FCP1, FDPSL5, FECH, FEO, FEOM1, FES, FGA, FGB, FGD1, FGF2, FGF23, FGF5, FGFR2, FGFR3, FGFR1, FGG, FGS1, FH, FICI, FIH, F2, FKBP6, FLNA, FLT4, FMO3,FMO4, FMR2, FMR1, FN, FN1/m, FOXC1, FOXE1, FOXL2, FOXO1A, FPDMM,
FPF, Fra-1, FRAXF, FRDA, FSHB, FSHMD1A, FSHR, FTH1, FTHL17, FTL, FTZF1, FUCA1, FUT2, FUT6, FUT1, FY, G250, G250/CAIX, G6PC, G6PD, G6PT1, G6PT2, GAA, GABRA3, GAGE-1, GAGE-2, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7b, GAGE-8, GALC, GALE, GALK1, GALNS, GALT, GAMT, GAN, GAST, GASTRIN17, GATA3, GATA, GBA, GBE, GC, GCDH, GCGR, GCH1, GCK, GCP-2, GCS1, G-CSF, GCSH, GCSL, GCY,
GDEP,GDF5, GDH, GDNF, GDXY, GFAP, GFND, GGCX, GGT1, GH2, GH1, GHR, GHRHR, GHS, GIF, GINGF, GIP, GJA3, GJA8, GJB2, GJB3, GJB6, GJB1, GK, GLA, GLB, GLBl, GLC3B, GLC1B, GLC1C, GLDC, GLI3, GLP1, GLRAl, GLUD1, GMl (fuc-GM1), GM2A, GM-CSF, GMPR, GNAI2, GNAS, GNAT1, GNB3, GNE, GNPTA, GNRH, GNRHl, GNRHR, GNS, GnT-V, gp100, GPlBA, GP1BB, GP9, GPC3, GPD2, GPDSl,
GPI, GP1BA, GPNlLW, GPNMB/m, GPSC, GPX1, GRHPR, GRK1, GRO, GRO, GRO, GRPR, GSE, GSM1, GSN, GSR, GSS, GTD, GTS, GUCA1A, GUCY2D, GULOP, GUSB, GUSM, GUST, GYPA, GYPC, GYSl, GYS2, H0KPP2, H0MG2, HADHA, HADHB, HAGE, HAGH, HAL, HAST-2, HB 1, HBA2, HBAl, HBB, HBBP1, HBD, HBE1, HBG2, HBGl, HBHR, HBPl, HBQl, HBZ, HBZP, HCA, HCC-I, HCC-4, HCF2, HCG, HCL2,
HCL1, HCR, HCVS, HD, HPN, HER2, HER2/NEU, HER3, HERV-K-MEL, HESXl, HEXA, HEXB, HFl, HFE, HFI, HGD, HHC2, HHC3, HHG, HK1 HLA-A, HLA-A*0201-R170l, HLA-A11/m, HLA-A2/m, HLA-DPBl HLA-DRA, HLCS, HLXB9, HMBS, HMGA2, HMGCL, HMI, HMN2, HMOXl, HMSl HMW-MAA, HND, HNE, HNF4A, HOAC, HOMEOBOX NKX 3.1, HOM-TES-14/SCP-1 , HOM-TES-85, HOXA1 HOXD13, HP,
HPC1, HPD, HPE2, HPE1, HPFH, HPFH2, HPRTl, HPSl, HPT, HPV-E6, HPV-E7, HR, HRAS, HRD, HRG, HRPT2, HRPT1, HRX, HSDl 1B2, HSDl 7B3, HSDI 7B4, HSD3B2, HSD3B3, HSN1, HSP70-2M, HSPG2, HST-2, HTC2, HTCl, hTERT, HTN3, HTR2C, HVBS6, HVBSl, HVEC, HV1S, HYAL1, HYR, I-309, IAB, IBGCI, IBM2, ICAM1, ICAM3, iCE, ICHQ, ICR5, ICRl, ICS 1, IDDM2, IDDM1, IDS, IDUA, IF, IFNa/b, IFNGRl, IGADl, IGER, IGF-1R, IGF2R, IGFl, IGH, IGHC, IGHG2, IGHGl, IGHM, IGHR, IGKC, IHGI, IHH, IKBKG, ILl, IL-I RA, ILIO, IL-Il, IL12, IL12RB1, IL13, IL-13Rα2, IL-15, IL- 16, IL-I 7, IL18, IL-Ia, IL-I α, IL-Ib, IL-I β, IL1RAPL1, IL2, IL24, IL-2R, IL2RA, IL2RG, IL3,
IL3RA,IL4, IL4R,IL4R, IL-5, IL6, IL-7, IL7R, IL-8, IL-9, Immature laminin receptor, IMMP2L, INDX, INFGR1, INFGR2, INFα, IFNβlNFγ, INS, INSR, INVS, IP-IO, IP2, IPFI, IPl, IRF6, IRSl, ISCW, ITGA2, ITGA2B, ITGA6, ITGA7, ITGB2, ITGB3, ITGB4, ITIHI, ITM2B, IV, IVD, JAG1, JAK3, JBS, JBTS1, JMS, JPD, KALI, KAL2, KALI, KLK2, KLK4, KCNAl, KCNE2, KCNEI, KCNH2, KCNJ1, KCNJ2, KCNJ1, KCNQ2, KCNQ3, KCNQ4,
KCNQ1, KCS, KERA, KFM, KFS, KFSD, KHK, ki-67, KIAA0020, KIAA0205, KIAA0205/m, KIFIB, KIT, KK-LC-I, KLK3, KLKBl, KM-HN-I, KMS, KNG, KNO, K-RAS/m, KRAS2, KREV1, KRT1, KRT10, KRTl 2, KRT13, KRTl 4, KRT14L1, KRTl 4L2, KRT14L3,KRT16, KRT16L1, KRT16L2, KRTl 7, KRTl 8, KRT2A, KRT3, KRT4, KRT5, KRT6 A, KRT6B, KRT9, KRTHB1, KRTHB6, KRTl, KSA, KSS, KWE, KYNU, L0H19CR1, LlCAM, LAGE, LAGE-1,
LALL, LAMA2, LAMA3, LAMB3, LAMB1, LAMC2, LAMP2, LAP, LCA5, LCAT, LCCS, LCCS 1, LCFS2, LCS1, LCT, LDHA, LDHB, LDHC, LDLR, LDLR/FUT, LEP, LEWISY, LGCR, LGGF-PBP, LGIl, LGMD2H, LGMD1A, LGMD1B, LHB, LHCGR, LHON, LHRH, LHX3, LIF, LIGI, LIMM, LIMP2, LIPA, LIPA, LIPB, LIPC, LIVIN, L1CAM, LMAN1, LMNA, LMX1B, LOLR, LOR, LOX, LPA, LPL, LPP, LQT4, LRP5, LRS 1, LSFC, LT-β , LTBP2,
LTC4S, LYL1, XCLl, LYZ, M344, MA50, MAA, MADH4, MAFD2, MAFD1, MAGE, MAGE-Al, MAGE-A10, MAGE-A12, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A9, MAGEBl, MAGE-BlO, MAGE-Bl 6, MAGE-B17, MAGE-B2, MAGE-B3, MAGE-B4, MAGE-B5, MAGE-B6, MAGE-C1, MAGE-C2, MAGE-C3, MAGE-D1, MAGE- D2, MAGE-D4, MAGE-El, MAGE-E2, MAGE-Fl, MAGE-Hl, MAGEL2, MGB1, MGB2,
MAN2A1, MAN2B1, MANBA, MANBB, MAOA, MAOB, MAPK8IP1, MAPT, MART-1, MART-2, MART2/m, MATlA, MBL2, MBP, MBS1, MCIR, MC2R, MC4R, MCC, MCCC2, MCCC1, MCDRI, MCF2, MCKD, MCLl, MClR, MCOLNl, MCOP, MCOR, MCP-1, MCP-2, MCP-3, MCP-4, MCPH2, MCPHI, MCS, M-CSF, MDB, MDCR, MDM2, MDRV, MDS 1, ME1, ME1/m, ME2, ME20, ME3, MEAX, MEB, MEC CCL-28, MECP2, MEFV,
MELANA, MELAS, MENl MSLN, MET, MF4, MG50, MG50/PXDN, MGAT2, MGAT5, MGC1 MGCR, MGCT, MGI, MGP, MHC2TA, MHS2, MHS4, MIC2, MIC5, MIDI, MIF, MIP, MIP-5/HCC-2, MITF, MJD, MKI67, MKKS, MKS1, MLHI, MLL, MLLT2, MLLT3, MLLT7, MLLT1, MLS, MLYCD, MMAIa, MMP 11, MMVP1, MN/CA IX-Antigen, MNG1, MN1, MOC31, MOCS2, MOCS1, MOG, MORC, MOS, MOV18, MPD1, MPE, MPFD, MPI, MPIF-1, MPL, MPO, MPS3C, MPZ, MRE11A, MROS, MRP1, MRP2, MRP3, MRSD, MRX14, MRX2, MRX20, MRX3, MRX40, MRXA, MRX1, MS, MS4A2, MSD, MSH2, MSH3, MSH6, MSS, MSSE, MSX2, MSX1, MTATP6, MTC03, MTCOl, MTCYB, MTHFR,
MTM1, MTMR2, MTND2, MTND4, MTND5, MTND6, MTND1, MTP, MTR, MTRNR2, MTRNR1, MTRR,MTTE, MTTG, MTTI, MTTK, MTTL2, MTTL1, MTTN, MTTP, MTTS1, MUC1,MUC2, MUC4, MUC5AC, MUM-1, MUM-1/m, MUM-2, MUM-2/m, MUM-3, MUM-3/m, MUT, mutant p21 ras, MUTYH, MVK, MX2, MX11, MY05A, MYB, MYBPC3, MYC, MYCL2, MYH6, MYH7, MYL2, MYL3, MYMY, MYO15A, MYO1G, MYO5A,
MYO7A, MYOC, Myosin/m, MYP2, MYP1, NA88-A, N-acetylglucosaminyltransferase-V, NAGA, NAGLU, NAMSD, NAPB, NAT2, NAT, NBIA1, NBS1, NCAM, NCF2, NCF1, NDN , NDP, NDUFS4, NDUFS7, NDUFS8, NDUFV1, NDUFV2, NEB, NEFH, NEM1, Neo-PAP, neo-PAP/m, NEU1, NEUROD1, NF2, NF1, NFYQm, NGEP, NHS, NKS1, NKX2E, NM, NME1, NMP22, NMTC, NODAL, NOG, NOS3, NOTCH3, NOTCH1, NP,
NPC2, NPC1, NPHL2, NPHP1, NPHS2, NPHS1, NPM/ALK, NPPA, NQO1, NR2E3, NR3C1, NR3C2, NRAS, NRAS/m, NRL, NROB1, NRTN, NSE, NSX, NTRK1, NUMA1, NXF2, NY-CO1, NY-ESO1, NY-ESO-B, NY-LU-12, ALDOA, NYS2, NYS4, NY-SAR-35, NYS1, NYX, OA3, OA1, OAP, OASD, OAT, OCA1, OCA2, OCD1, OCRL, OCRL1, OCT, ODDD, ODT1, OFC1, OFD1, OGDH, OGT, OGT/m, OPA2, OPA1, OPD1, OPEM,
OPG, OPN, OPN 1LW, OPN 1MW, OPN 1SW, OPPG, OPTB 1, TTD, ORM1, ORP1, OS- 9, OS-9/m, OSM LIF, OTC, OTOF, OTSC1, OXCT1, OYTES1, P15, P190 MINOR BCR- ABL, P2RY12, P3, P16, P40, P4HB, P-501, P53, P53/m, P97, PABPN1, PAFAH1B1, PAFAH1P1, PAGE-4, PAGE-5, PAH, PAI-1, PAI-2, PAK3, PAP, PAPPA, PARK2, PART-1, PATE, PAX2, PAX3, PAX6, PAX7, PAX8, PAX9, PBCA, PBCRA1, PBT, PBX1, PBXP1, PC,
PCBD, PCCA, PCCB, PCK2, PCK1, PCLD, PCOSI, PCSK1, PDB1, PDCN, PDE6A, PDE6B, PDEF, PDGFB, PDGFR, PDGFRL, PDHA1, PDR, PDX1, PECAMI, PEE1, PEO1, PEPD, PEX10, PEX12, PEX13, PEX3, PEX5, PEX6, PEX7, PEX1, PF4, PFBI, PFC, PFKFB1, PFKM, PGAM2, PGD, PGK1, PGK1P1, PGL2, PGR, PGS, PHA2A, PHB, PHEX, PHGDH, PHKA2, PHKA1, PHKB, PHKG2, PHP, PHYH, Pl, PI3, PIGA, PIM1 -KINASE, PINI,
PIP5K1B, PITX2, PITX3, PKD2, PKD3, PKD1, PKDTS, PKHD1, PKLR, PKP1, PKU1, PLA2G2A, PLA2G7, PLAT, PLEC1, PLG, PLI, PLOD, PLP1, PMEL17, PML, PML/RARα, PMM2, PMP22, PMS2, PMS1, PNKD, PNLIP, POF1, POLA, POLH, POMC, PON2, PON1, PORC, POTE, POU1F1, POU3F4, POU4F3, POU1F1, PPAC, PPARG, PPCD, PPGB, PPH1, PPKB, PPMX, PPOX, PPP1R3A, PPP2R2B, PPTI, PRAME, PRB, PRB3, PRCA1, PRCC, PRD, PRDX5/m, PRF1, PRG4, PRKAR1A, PRKCA, PRKDC, PRKWNK4, PRNP, PROC, PRODH, PROM1, PROP1, PROS1, PRST, PRP8, PRPF31, PRPF8, PRPH2, PRPS2, PRPSl, PRS, PRSS7, PRSS1, PRTN3, PRX, PSA, PSAP, PSCA, PSEN2, PSENl,
PSG1, PSGR, PSM, PSMA, PSORSI, PTC, PTCH, PTCHl, PTCH2, PTEN, PTGS1, PTH, PTHR1, PTLAH, PTOS1, PTPN12, PTPNI I, PTPRK, PTPRK/m, PTS, PUJO, PVR, PVRL1, PWCR, PXE, PXMP3, PXR1, PYGL, PYGM, QDPR, RAB27A, RAD54B, RAD54L, RAG2, RAGE, RAGE-1, RAG1, RAP1, RARA, RASA1, RBAF600/m, RB1, RBP4, RBP4, RBS, RCAI, RCAS1, RCCP2, RCD1, RCV1, RDH5, RDPA, RDS, RECQL2, RECQL3, RECQL4,
REG1A, REHOBE, REN, RENBP, RENS1, RET, RFX5, RFXANK, RFXAP, RGR, RHAG, RHAMM/CD168, RHD, RHO, Rip-1, RLBP1, RLN2, RLN1, RLS, RMD1, RMRP, ROM1, ROR2, RP, RP1, RP14, RP17, RP2, RP6, RP9, RPDl, RPE65, RPGR, RPGRIP1, RP1, RP10, RPS19, RPS2, RPS4X, RPS4Y, RPS6KA3, RRAS2, RS1, RSN, RSS, RU1, RU2, RUNX2,RUNXI, RWS, RYR1, S-100, SAA1, SACS, SAG, SAGE, SALL1, SARDH, SART1,
SART2 , SART3, SAS, SAX1, SCA2, SCA4, SCA5, SCA7, SCA8, SCA1, SCC, SCCD, SCF, SCLC1, SCN1A, SCN1B, SCN4A, SCN5A, SCNNlA, SCNN1B, SCNN1G, SCO2, SCP1, SCZD2, SCZD3, SCZD4, SCZD6, SCZD1, SDF-1α/β SDHA, SDHD, SDYS, SEDL, SERPENA7, SERPINA3, SERPINA6, SERPINA1, SERPINC1, SERPIND1, SERPINE1, SERPINF2, SERPING1, SERPINM, SFTPAl, SFTPB, SFTPC, SFTPD, SGCA, SGCB, SGCD,
SGCE, SGM1, SGSH, SGY-1, SH2D1A, SHBG, SHFM2, SHFM3, SHFM1, SHH, SHOX, Sl, SIAL, SIALYL LEWISX , SIASD, SII, SIM1, SIRT2/m, SIX3, SJS1, SKP2, SLC10A2, SLC12A1, SLC12A3, SLC17A5, SLC19A2, SLC22A1L, SLC22A5, SLC25A13, SLC25A15, SLC25A20, SLC25A4, SLC25A5, SLC25A6, SLC26A2, SLC26A3, SLC26A4, SLC2A1, SLC2A2, SLC2A4, SLC3A1, SLC4A1, SLC4A4, SLC5A1, SLC5A5, SLC6A2, SLC6A3,
SLC6A4, SLC7A7, SLC7A9, SLC11A1, SLOS, SMA, SMAD1, SMAL, SMARCB1, SMAX2, SMCR, SMCY, SM1, SMN2, SMN1, SMPD1, SNCA, SNRPN, SOD2, SOD3, SOD1, SOS1, SOST, SOX9, SOX10, Sp17, SPANXC, SPG23, SPG3A, SPG4, SPG5A, SPG5B, SPG6, SPG7, SPINK1, SPINK5, SPPK, SPPM, SPSMA, SPTA1, SPTB, SPTLC1, SRC, SRD5A2, SRPX, SRS, SRY, βhCG, SSTR2, SSX1, SSX2 (HOM-MEL-40/SSX2), SSX4, ST8,
STAMP-1, STAR, STARP1, STATH, STEAP, STK2, STKl 1, STn/ KLH, STO, STOM, STS, SUOX, SURF1, SURVIVIN-2B, SYCP1, SYM1, SYN1, SYNS1, SYP, SYT/SSX, SYT-SSX-1, SYT-SSX-2, TA-90, TAAL6, TACSTD1, TACSTD2, TAG72, TAF7L, TAF1, TAGE, TAG-72, TALI, TAM, TAP2, TAP1, TAPVR1 , TARC, TARP, TAT, TAZ, TBP, TBX22, TBX3, TBX5, TBXA2R, TBXASI , TCAP, TCF2, TCF1, TCIRG1 , TCL2, TCL4, TCLIA, TCN2, TCOF1 , TCR, TCRA, TDD, TDFA, TDRD1 , TECK, TECTA, TEK, TEL/AML1 , TELAB1 , TEX15, TF, TFAP2B, TFE3, TFR2, TG, TGFα, TGFβ, TGFβl, TGFβi , TGFβR2, TGFβRE, TGFγ, TGFβRII, TGIF, TGM-4, TGM1, TH, THAS, THBD, THC, THC2, THM, THPO, THRA,
THRB, TIMM8A, TIMP2, TIMP3, TIMP1 , TITF1 , TKCR, TKT, TLP, TLR1, TLR10, TLR2, TLR3, TLR4, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLX1, TM4SF1, TM4SF2, TMC1, TMD, TMIP, TNDM, TNF, TNFRSF1 1 A, TNFRSF1A, TNFRSF6, TNFSF5, TNFSF6, TNFα, TNFβ, TNNI3, TNNT2, TOC, TOP2A, TOP1 , TP53, TP63, TPA, TPBG, TPI, TPI/m, TPH, TPM3, TPM1 , TPMT, TPO, TPS, TPTA, TRA, TRAG3, TRAPPC2, TRC8, TREH, TRG, TRH,
TRIM32, TRIM37, TRPI , TRP2, TRP-2/6b, TRP-2/INT2, Trp-p8, TRPS1 , TS, TSC2, TSC3, TSCl, TSGl Ol, TSHB, TSHR, TSP-180, TST, TTGA2B, TTN, TTPA, TTR, TU M2-PK, TULP1, TWIST, TYH, TYR, TYROBP, TYROBP, TYRP1 , TYS, UBE2A, UBE3A, UBE1 , UCHL1 , UFS, UGT1 A, ULR, UMPK, UMPS, UOX, UPA, UQCRC1 , URO5, UROD, UPK1 B, UROS, USH2A, USH3A, USH1 A, USH1 C, USP9Y, UV24, VBCH, VCF, VDI,
VDR, VEGF, VEGFR-2, VEGFR-1, VEGFR-2/FLK-1 , VHL, VIM, VMD2, VMD1 , VMGLOM, VNEZ, VNF, VP, VRNI, VWF, VWS, WAS, WBS2, WFS2, WFSl, WHCR, WHN, WISP3, WMS, WRN, WS2A, WS2B, WSN, WSS, WT2, WT3, WT1 , WTS, WWS, XAGE, XDH, XIC, XIST, XK, XM, XPA, XPC, XRCC9, XS, ZAP70, ZFHX1 B, ZFX, ZFY, ZIC2, ZIC3, ZNF145, ZNF261 , ZNF35, ZNF41 , ZNF6, ZNF198, and ZWS1.
Additionally, therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from growth hormones or growth factors, for example for promoting growth in a (transgenic) living being, such as, for example, TGFα and the IGFs (insulin- like growth factors), proteins that influence the metabolism and/or haematopoiesis, such as, for example, α- anti-trypsin, LDL receptor, erythropoietin (EPO), insulin, GATA-I, etc., or proteins such as, for example, factors VIII and Xl of the blood coagulation system, etc. Such proteins further include enzymes, such as, for example, β-galactosidase (lacZ), DNA restriction enzymes (e.g. EcoRI, Hindlll, etc.), lysozymes, etc., or proteases, such as, for example, papain, bromelain, keratinases, trypsin, chymotrypsin, pepsin, renin (chymosin), suizyme, nortase, etc.. These proteins may be provided by the at least one modified (m)RNA (of the inventive immunosuppressive composition), which is characterized by an increased level of expression. Accordingly, the invention provides a technology which allows to substitute proteins which are defective in the organism to be treated (e.g. either due to mutations, due to defective or missing expression) and thereby effective and increased expression of proteins, which are not functional in the organism to be treated, as e.g. occurring in monogenetic disorders, without leading to an innate immune response.
Alternatively, therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from proteases etc. which allow to cure a specific disease due to e.g. (over)expression of a dysfunctional or exogenous proteins causing disorders or diseases. Accordingly, the invention may be used to therapeutically introduce the at least one modified (m)RNA (of the inventive immunosuppressive composition) into the organism, which attacks a pathogenic organism (virus, bacteria etc). E.g. RNA encoding therapeutic proteases may be used to cleave viral proteins which are essential to the viral assembly or other essential steps of virus production.
Therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from proteins which modulate various intracellular pathways by e.g. signal transmission modulation (inhibition or stimulation) which may influence pivotal intracellular processes like apoptosis, cell growth etc, in particular with respect to the organism's immune system. Accordingly, immune modulators, e.g. cytokines, lymphokines, monokines, interferones etc. may be expressed efficiently by the at least one modified (m)RNA (of the inventive immunosuppressive composition). Preferably, these proteins therefore also include, for example, cytokines of class I of the cytokine family that contain 4 position-specific conserved cysteine residues (CCCC) and a conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS), wherein X represents an unconserved amino acid. Cytokines of class I of the cytokine family include the GM-CSF sub-family, for example IL-3, IL-5, GM-CSF, the IL-6 sub-family, for example IL-6, IL-1 1 , IL-12, or the IL-2 sub-family, for example IL-2, IL-4, IL-7, IL-9, IL-15, etc., or the cytokines IL-1 α, IL-I β, IL-10 etc. By analogy, such proteins can also include cytokines of class Il of the cytokine family (interferon receptor family), which likewise contain 4 position-specific 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-α, IFN-β, IFN-γ, etc. Proteins coded for by the at least one modified (m)RNA (of the inventive immunosuppressive composition) used according to the invention can further include also cytokines of the tumour necrosis family, for example TNF-α, TNF-β, TNF-RI, TNF-RII, CD40, Fas, etc., or cytokines of the chemokine family, which contain 7 transmembrane helices and interact with G-protein, for example IL-8, MIP-1 , RANTES, CCR5, CXR4, etc. Such proteins can 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, BcI-X1, BcI-X5, bik, CAD, calpain, caspases, for example caspase-1 , caspase-2, caspase-3, caspase-4, caspase-5, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, caspase-
11 , ced-3, ced-9, c-Jun, c-Myc, crm A, cytochrome C, CdR1 , DcR1 , DD, DED, DISC, DNA-PKcs, 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-KB, NuMa, p53, PAK-2, PARP, perforin, PITSLRE, PKCδ, pRb, presenilin, prlCE, RAIDD, Ras, RIP, sphingomyelinase, thymidine kinase from Herpes simplex, TRADD, TRAF2, TRAIL, TRAIL-R1 , TRAIL-R2, TRAIL-R3, transglutaminase, etc.
Additionally, the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also code for antigen specific T cell receptors. The T cell receptor or
TCR is a molecule found on the surface of T lymphocytes (or T cells) that is generally responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules. It is a heterodimer consisting of an alpha and beta chain in 95% of T cells, while 5% of T cells have TCRs consisting of gamma and delta chains. Engagement of the TCR with antigen and MHC results in activation of its T lymphocyte through a series of biochemical events mediated by associated enzymes, co-receptors and specialized accessory molecules. Hence, these proteins allow to specifically target specific antigen and may support the functionality of the immune system due to their targeting properties.
Accordingly, transfection of cells in vivo by administering the at least one modified (m)RNA (of the inventive immunosuppressive composition) coding for these receptors or, preferably, an ex vivo cell transfection approach (e.g. by transfecting specifically certain immune cells), may be pursued. The T cell receptor molecules introduced recognize specific antigens on MHC molecule and may thereby support the immune system's awareness of antigens to be attacked.
According to a further alternative, therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) may also be selected from (efficiently expressed) antigens which elicit only an adaptive immune response, whereas the underlying non-modified RNA provokes an innate and an adaptive immune reaction as such, i.e. the innate immune respnse may be suppressed. This may be particularly advantageous with respect to the treatment of patients both suffering from allergic diseases or autoimmune diseases and another disease, such as cancer diseases or
(viral) infectious diseases, in order to avoid an innate immune response as far as possible. Insofar, the invention may allow to provide vaccines based on the at least one modified (m)RNA (of the inventive immunosuppressive composition), which expresses increased levels of the antigenic protein or peptide without leading to an (increased) immune response. These vaccines may be used for the provision of tumour vaccines providing tumour antigens or antigens derived from pathogenic microorganisms causing e.g. infectious diseases. According to the present invention, the term "antigen" refers to a substance which is recognized by the immune system and is capable of triggering an antigen-specific immune response, e.g. by formation of antibodies as part of an adaptive immune response. Therefore, it is desirable in the context of the present invention, to suppress and/or avoid such an innate immunostimulatory response which arises, when the unmodified RNA is used. Such antigens may also comprise epitopes. In the context of the present invention, epitopes of a protein or an antigen (sometimes also called "antigen determinants"), typically, are fragments of such protein or peptide structures having 5 to 15, preferably 6 to 9, amino acids. The antigen may be furthermore a "self" or non-self" antigen or an allergy antigen, i.e. an antigen, which causes an allergy in a human and may be derived from either a human or other sources. Antigens can be classified in the order of their origins. Accordingly, there are two major classes of antigens: exogenous and endogenous antigens. Exogenous antigens are typically antigens that enter the cell or the body from outside (the cell or the body), for example by inhalation, ingestion or injection, etc.. These antigens are internalized by antigen- presenting cells ("APCs", such as dendritic cells or macrophages) and processed into fragments. APCs then present the fragments to T helper cells (e.g. CD4+) by the use of MHC Il molecules on their surface. Recognition of these antigen fragments by T cells leads to activation of the T cells and secretion of cytokines. Cytokines are substances that can activate proliferation of immune cells such as cytotoxic T cells, B cells or macrophages. In contrast, endogenous antigens are antigens which typically have been generated within the cell, e.g. as a result of normal cell metabolism. Fragments of these antigens are presented on MHC I molecules on the surface of APCs. These antigens are recognized by activated antigen-specific cytotoxic CD8+ T cells. After recognition, those T cells react in secretion of different toxins that cause lysis or apoptosis of the antigen- presenting cell. Endogenous antigens comprise antigens, e.g. proteins or peptides encoded by a foreign nucleic acid inside the cell as well as proteins or peptides encoded by the genetic information of the cell itself, or antigens from intracellularly occurring viruses.
Specifically preferred antigens coded for by the at least one modified (m)RNA (of the inventive immunosuppressive composition) can be selected from the following antigens: tumour-specific surface antigens (TSSAs), for example 5T4, α5β1 -integrin, 707-AP, AFP,
ART-4, B7H4, BAGE, β-catenin/m, Bcr-abl, MN/C IX antigen, CA125, CAMEL, CAP-1,
CASP-8, β-catenin/m, CD4, CD19, CD20, CD22, CD25, CDC27/m, CD 30, 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 -
R1 70I, 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-I , -2, -3, NA88-A, PAP, NY-ESOI , proteinase-3, p190 minor bcr-abl,
Pml/RARα, PRAME, PSA, PSM, PSMA, RAGE, RUI or RU2, SAGE, SART-1 or SART-3, survivin, TEL/AML1, TGFβ, TPI/m, TRP-1, TRP-2, TRP-2/INT2, VEGF and WT1, or from sequences such as, for example, NY-Eso-1 or NY-Eso-B.
One class of endogenous antigens, which may be encoded by therapeutically active proteins as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) is the class of tumor antigens. Those antigens are presented by the MHC I molecules on the surface of tumor cells. This class can be divided further in tumor-specific antigens (TSAs) and tumor-associated-antigens (TAAs). TSAs can only be presented by tumor cells and never by normal "healthy" cells. They typically result from a tumor specific mutation. TAAs, which are more common, are usually presented by both tumor and healthy cells. These antigens are recognized and the antigen-presenting cell can be destroyed by cytotoxic T cells. Additionally, tumor antigens can also occur on the surface of the tumor in the form of, e.g., a mutated receptor. In this case, they can be recognized by antibodies, particularly by antibodies as defined above. Examples of tumor antigens are shown in Tables 1 and 2 below. These tables illustrate specific (protein) antigens (i.e. "tumor antigens") with respect to the cancer disease, they are associated with. According to the invention, the terms "cancer diseases" and "tumor diseases" are used synonymously herein.
Table 1 : Anti ens ex ressed in cancer diseases
Figure imgf000033_0001
Figure imgf000034_0001
cancer, lymphoma
Figure imgf000035_0001
Figure imgf000036_0001
lun cancer sarcoma, leukemia
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Table 2: Mutant antigens expressed in cancer diseases
Figure imgf000040_0001
Figure imgf000041_0001
In a preferred embodiment according to the present invention, the (protein) antigens, as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) are selected from the group consisting of 5T4, 707-AP, 9D7, AFP, AIbZIP HPGI, alpha-5-beta-1 -integrin, alpha-5-beta-6-integrin, alpha-actinin-4/m, alpha- methylacyl-coenzyme A racemase, ART-4, ARTC1/m, B7H4, BAGE-1 , BCL-2, bcr/abl, beta-eaten in/m, BING-4, BRCA1/m, BRCA2/m, CA 15-3/CA 27-29, CA 19-9, CA72-4, CA125, calreticulin, CAMEL, CASP-8/m, cathepsin B, cathepsin L, CD19, CD20, CD22, CD25, CDE30, CD33, CD4, CD52, CD55, CD56, CD80, CDC27/m, CDK4/m, CDKN2A/m, CEA, CLCA2, CML28, CML66, COA-1/m, coactosin-like protein, collage
XXIII, COX-2, CT-9/BRD6, Cten, cyclin B1 , cyclin D1 , cyp-B, CYPB1, DAM-10, DAM-6, DEK-CAN, EFTUD2/m, EGFR, ELF2/m, EMMPRIN, EpCam, EphA2, EphA3, ErbB3, ETV6- AML1 , EZH2, FGF-5, FN, Frau-1 , G250, GAGE-1 , GAGE-2, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE7b, GAGE-8, GDEP, GnT-V, gpl OO, GPC3, GPNMB/m, HAGE, HAST-2, hepsin, Her2/neu, HERV-K-MEL, HLA-A*0201 -R17l, HLA-AH/m, HLA-A2/m, HNE, homeobox NKX3.1 , HOM-TES-14/SCP-1 , HOM-TES-85, HPV-E6, HPV-E7, HSP70-2M, HST-2, hTERT, iCE, IGF-I R, IL-13Ra2, IL-2R, IL-5, immature laminin receptor, kallikrein- 2, kallikrein-4, Ki67, KIAA0205, KIAAO2O5/m, KK-LC-1 , K-Ras/m, LAGE-A1 , LDLR-FUT, MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A6, MAGE-A9, MAGE-A10, MAGE- A12, MAGE-Bl , MAGE-B2, MAGE-B3, MAGE-B4, MAGE-B5, MAGE-B6, MAGE-B10,
MAGE-B16, MAGE-B1 7, MAGE-C1 , MAGE-C2, MAGE-C3, MAGE-D1 , MAGE-D2, MAGE-D4, MAGE-E1 , MAGE-E2, MAGE-Fl, MAGE-H1 , MAGEL2, mammaglobin A, MART-1/melan-A, MART-2, MART-2/m, matrix protein 22, MC1 R, M-CSF, ME1/m, mesothelin, MG50/PXDN, MMP1 1, MN/CA IX-antigen, MRP-3, MUC-1 , MUC-2, MUM- 1/m, MUM-2/m, MUM-3/m, myosin class l/m, NA88-A, N-acetylglucosaminyltransferase- V, Neo-PAP, Neo-PAP/m, NFYQm, NGEP, NMP22, NPM/ALK, N-Ras/m, NSE, NY-ESO- 1, NY-ESO-B, OA1 , OFA-iLRP, OGT, OGT/m, OS-9, OS-9/m, osteocalcin, osteopontin, p15, p190 minor bcr-abl, p53, p53/m, PAGE-4, PAI-I , PAI-2, PART-1, PATE, PDEF, Pim- 1 -Kinase, Pin-1 , Pml/PARalpha, POTE, PRAME, PRDX5/m, prostein, proteinase-3, PSA, PSCA, PSGR, PSM, PSMA, PTPRK/m, RAGE-1 , RBAF600/m, RHAMM/CD168, RU1 , RU2,
S-100, SAGE, SART-1 , SART-2, SART-3, SCC, SIRT2/m, Sp17, SSX-I, SSX-2/HOM-MEL- 40, SSX-4, STAMP-1, STEAP, survivin, survivin-2B, SYT-SSX-1, SYT-SSX-2, TA-90, TAG- 72, TARP, TEL-AML1 , TGFbeta, TGFbetaRII, TGM-4, TPI/m, TRAG-3, TRG, TRP-1 , TRP- 2/6b, TRP/INT2, TRP-p8, tyrosinase, UPA, VEGF, VEGFR-2/FLK-1 , and WT1.
In a particularly preferred embodiment according to the present invention, the (protein) antigens as encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) are selected from the group consisting of MAGE-A1 , MAGE-A6, melan-A, GP100, tyrosinase, survivin, CEA, Her-2/neu, WTl, PRAME, EGFRI (epidermal growth factor receptor 1), mucin-1 and SEC61 G, hTERT, 5T4, NY-Eso1 , and
TRP-2, more preferably from sequences of group consisting of MAGE-A1 [accession number M77481 ], MAGE-A6 [accession number NM_005363], melan-A [accession number NM_00551 1 ], GP100 [accession number M77348], tyrosinase [accession number NM_000372], survivin [accession number AF077350], CEA [accession number NMJD04363], Her-2/neu [accession number M1 1730], WT1 [accession number
NM_000378], PRAME [accession number NM_006115], EGFRI (epidermal growth factor receptor 1 ) [accession number AF288738], mucin-1 [accession number NM_002456] and SEC61 G [accession number NM_014302], hTERT [accession number NM_198253], 5T4 [accession number NM_006670], NY-Eso1 [accession number NM_001327], TRP-2 [accession number NM_001922], MAGE-A2: [accession number NM_153488], MAGE-
A3: [accession number NM_005362], MAGE-C1 : [accession number NM_005462], and MAGE-C2: [accession number NM_016249].
Therapeutically active proteins that can be coded for by the at least one modified (m)RNA (of the inventive immunosuppressive composition) further include also those proteins or protein sequences that have a sequence identity of at least 80% or 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99%, with one of the therapeutically active proteins described above, e.g. their native sequence. In this case, the modified nucleosides and their native (non-modified) analog are considered to be "identical" herein.
Antibodies As a further alternative, the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode an antibody. According to the present invention, such an antibody may be selected from any antibody, e.g. any recombinantly produced or naturally occurring antibodies, known in the art, in particular antibodies suitable for therapeutic, diagnostic or scientific purposes, or antibodies which have been identified in relation to specific diseases, such as e.g. cancer diseases or viral diseases. Herein, the term "antibody" is used in its broadest sense and specifically covers monoclonal and polyclonal antibodies (including agonist, antagonist, and blocking or neutralizing antibodies) and antibody species with polyepitopic specificity. According to the invention, the term "antibody" typically comprises any antibody known in the art (e.g. IgM, IgD, IgG, IgA and IgE antibodies), such as naturally occurring antibodies, antibodies generated by immunization in a host organism, antibodies which were isolated and identified from naturally occurring antibodies or antibodies generated by immunization in a host organism and recombinantly produced by biomolecular methods known in the art, as well as chimeric antibodies, human antibodies, humanized antibodies, bispecific antibodies, intrabodies, i.e. antibodies expressed in cells and optionally localized in specific cell compartments, and fragments and variants of the aforementioned antibodies. In general, an antibody consists of a light chain and a heavy chain both having variable and constant domains. The light chain consists of an N-terminal variable domain, VL, and a C-terminal constant domain, Q. In contrast, the heavy chain of the IgG antibody, for example, is comprised of an N-terminal variable domain, VH, and three constant domains, CH1 , CH2 und CH3. Single chain antibodies may be encoded by the RNA of the modified (m)RNA of the invention as well, preferably by a single-stranded RNA, more preferably by an mRNA.
According to a first alternative, the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode a polyclonal antibody. In this context, the term, "polyclonal antibody" typically means mixtures of antibodies directed to specific antigens or immunogens or epitopes of a protein which were generated by immunization of a host organism, such as a mammal, e.g. including goat, cattle, swine, dog, cat, donkey, monkey, ape, a rodent such as a mouse, hamster and rabbit. Polyclonal antibodies are generally not identical, and thus usually recognize different epitopes or regions from the same antigen. Thus, in such a case, typically a mixture (a composition) of different RNAs of the modified (m)RNA (of the inventive immunosuppressive composition) according to the invention will be applied, each encoding a specific (monoclonal) antibody being directed to specific antigens or immunogens or epitopes of a protein.
According to a further alternative, the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode a monoclonal antibody. The term "monoclonal antibody" herein typically refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed to a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed to different determinants (epitopes), each monoclonal antibody is directed to a single epitope or a determinant on the antigen. For example, monoclonal antibodies as defined above may be made by the hybridoma method first described by Kohler and Milstein,
Nature, 256:495 (1975), or may be made by recombinant DNA methods, e.g. as described in U.S. Pat. No. 4,816,567. "Monoclonal antibodies" may also be isolated from phage libraries generated using the techniques described in McCafferty et a/., Nature, 348:552-554 (1990), for example, and the produced in vitro as recombinant proteins. According to Kohler and Milstein, an immunogen (antigen) of interest is injected into a host such as a mouse and B-cell lymphocytes produced in response to the immunogen are harvested after a period of time. The B-cells are combined with myeloma cells obtained from mouse and introduced into a medium which permits the B-cells to fuse with the myeloma cells, producing hybridomas. These fused cells (hybridomas) are then placed in separate wells in microtiter plates and grown to produce monoclonal antibodies. The monoclonal antibodies are tested to determine which of them are suitable for detecting the antigen or epitope of interest. After being selected, the monoclonal antibodies can be grown in cell cultures or by injecting the hybridomas into mice. However, for the purposes of the present invention, the peptide sequences of these monoclonal antibodies have to be sequenced and modified (m)RNA sequences encoding these antibodies may be prepared according to procedures well known in the art.
For therapeutical purposes in humans, non-human monoclonal or polyclonal antibodies, such as murine antibodies may also be encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention. However, such antibodies are typically only of limited use, since they generally induce an immune response by production of human antibodies directed to the said non-human antibodies, in the human body. Therefore, a particular non-human antibody can only be administered once to the human. To solve this problem, chimeric, humanized non- human and human antibodies are also envisaged encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention. "Chimeric" antibodies, which may be encoded by the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention, are preferably antibodies in which the constant domains of an antibody described above are replaced by sequences of antibodies from other organisms, preferably human sequences. ,,Humanized" (non-human) antibodies, which may be also encoded by the at least one modified (m)RNA (molecule) of the modified (m)RNA of the present invention, are antibodies in which the constant and variable domains (except for the hypervariable domains) described above of an antibody are replaced by human sequences. According to another alternative, the at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode human antibodies, i.e. antibodies having only human sequences. Such human antibodies can be isolated from human tissues or from immunized non-human host organisms which are transgene for the human IgG gene locus, sequenced RNA sequences may be prepared according to procedures well known in the art. Additionally, human antibodies can be provided by the use of a phage display.
In addition, the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode bispecific antibodies. "Bispecific" antibodies in context of the invention are preferably antibodies which act as an adaptor between an effector and a respective target, e.g. for the purposes of recruiting effector molecules such as toxins, drugs, cytokines etc., targeting effector cells such as CTL, NK cells, makrophages, granulocytes, etc. (see for review: Kontermann R. E., Acta Pharmacol. Sin, 2005, 26(1 ): 1 -9). Bispecific antibodies as described herein are, in general, configured to recognize, e.g. two different antigens, immunogens, epitopes, drugs, cells (or receptors on cells), or other molecules (or structures) as described above. Bispecificity means herewith that the antigen-binding regions of the antibodies are specific for two different epitopes. Thus, different antigens, immunogens or epitopes, etc. can be brought close together, what, optionally, allows a direct interaction of the two components. For example, different cells such as effector cells and target cells can be connected via a bispecific antibody. Encompassed, but not limited, by the present invention are antibodies or fragments thereof which bind, on the one hand, a soluble antigen as described herein, and, on the other hand, an antigen or receptor on the surface of a tumor cell.
According to the invention, the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may also encode intrabodies, wherein these intrabodies may be antibodies as defined above. Since these antibodies are intracellular expressed antibodies, i.e. antibodies which are encoded by nucleic acids localized in specific areas of the cell and also expressed there, such antibodies may be termed intrabodies.
Antibodies as encoded by the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may preferably comprise full-length antibodies, i.e. antibodies composed of the full heavy and full light chains, as described above. However, derivatives of antibodies such as antibody fragments, variants or adducts may be encoded by the above defined at least one modified (m)RNA (of the inventive immunosuppressive composition) according to the invention.
The at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may also encode antibody fragments selected from Fab, Fab', F(ab')2, Fc, Facb, pFc', Fd and Fv fragments of the aforementioned antibodies. In general, antibody fragments are known in the art. For example, a Fab ("fragment, antigen binding") fragment is composed of one constant and one variable domain of each of the heavy and the light chain. The two variable domains bind the epitope on specific antigens. The two chains are connected via a disulfide linkage. A scFv ("single chain variable fragment") fragment, for example, typically consists of the variable domains of the light and heavy chains. The domains are linked by an artificial linkage, in general a polypeptide linkage such as a peptide composed of 15-25 glycine, proline and/or serine residues.
Antibodies, as defined above, may also be directed against antigens or epitopes thereof as defined above.
According to the present invention, the at least one coding modified (m)RNA (of the inventive immunosuppressive composition) according to the invention may encode fragments and/or variants of the aforementioned therapeutically active proteins or antibodies, etc., wherein the fragments and/or variants may have a sequence identity to one of the aforementioned therapeutically active proteins, or antibodies, etc. of at least 70%, 80% or 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99% over the whole length of the (coding) nucleic acid (or amino acid) sequences encoding these therapeutically active proteins, or antibodies, etc.. A "fragment of a therapeutically active protein, or antibody, etc." in the context of the present invention is to be understood as a truncated therapeutically active protein, or antibody, etc. of the therapeutically active proteins, or antibodies, etc. defined above, i.e. an amino acid sequence which is N-terminally, C-terminally and/or intrasequentially truncated compared to the amino acid sequence of the original (native) protein. Especially, fragments including an epitope of those therapeutically active proteins, antibodies, etc., are preferred. A "variant" in the context of the present invention refers to an therapeutically active protein, or antibody, etc. as defined above, wherein nucleic acids of the encoding modified mRNA sequence are exchanged, i.e. a therapeutically active protein, or antibody, etc. having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s). Preferably, the fragments and/or variants have the same biological function or specific activity compared to the full-length native therapeutically active proteins, or antibodies, etc., e.g. specific binding capacity (e.g. of particular antigens), catalytic activity (e.g. of therapeutically active proteins), etc.. In this context, the term "biological function" of antibodies as described herein also comprises neutralization of antigens, complement activation or opsonization. Thereby, antibodies typically recognize either native epitopes on the cell surface or free antigens. Antibodies as defined above can interact with the eel I -presenting antigens and initiate different defense mechanisms. On the one hand, the antibody can initiate signaling mechanisms in the targeted cell that leads to the cell's self-destruction (apoptosis). On the other hand, it can mark the cell in such a way that other components or effector cells of the body's immune system can recognize and attack. The attack mechanisms are referred to as antibody-dependent complement-mediated cytotoxicity (CMC) and antibody-dependent cellular cytotoxicity (ADCC). ADCC involves a recognition of the antibody by immune cells that engage the antibody-marked cells and either through their direct action, or through the recruitment of other cell types, lead to the tagged-cell's death. CMC is a process where a cascade of different complement proteins becomes activated, usually when several antibodies are in close proximity to each other, either resulting in cell lysis or attracting other immune cells to this location for effector cell function. In the neutralization of an antigen, the antibody can bind an antigen and neutralize the same. Such neutralization reaction, in turn, leads in general to blocking of the antibody. Thus, the antibody can bind only one antigen, or, in case of a bispecific antibody, two antigens. In particular, scFv antibody fragments are useful for neutralization reactions because they don't contain the functionalities of the constant domain of an antibody. In the complement activation, the complex system of complement proteins can be activated via binding of an antibody which is independent of the Fc part of an antibody. End products of the complement cascade result in lysis of the cell and generation of an inflammatory milieu. In the opsonization, pathogens or other non-cellular particles are made accessible to phagocytes via binding the constant domain of an antibody. Alternatively, cells recognized as foreign can be lysed via antibody- dependent cell-mediated cytotoxicity (ADCC). In particular, NK-cells can display lysis functions by activating Fc receptors.
If the at least one modified (m)RNA (of the immunosuppressive composition) according to the invention is a coding RNA as defined above, the modified (m)RNA may occur as a mono-, di-, or even multicistronic RNA, i.e. an RNA which carries the coding sequences of one, two or more therapeutically active proteins, or antibodies, etc. as defined above. If the at least one coding modified (m)RNA encodes at least one, e.g. two, three or more of an therapeutically active protein, or antibody, etc. as defined above, each of the at least one modified (m)RNAs preferably encodes a (preferably different) therapeutically active protein, or antibody, etc. as defined above. In any case, each therapeutically active protein, or antibody etc. as defined above encoded by the at least one modified (m)RNA may be selected independently.
According to another particularly preferred embodiment, the modified (m)RNA (of the immunosuppressive composition) according to the invention, may be at least a single, bi- or even multicistronic modified (m)RNA, i.e. it ay be at least one modified (m)RNA which carries two, three or even more of the coding sequences of a therapeutically active protein, or antibody, etc. as defined above as defined herein. Such coding sequences of the at least one (preferably different) therapeutically active protein, or antibody, etc. as defined above may be separated by at least one IRES (internal ribosomal entry site) sequence, as defined below. In this context, a so-called IRES (internal ribosomal entry site) sequence as defined above can function as a sole ribosome binding site, but it can also serve to provide a bi- or even multicistronic modified (m)RNA as defined above which encodes several proteins which are to be translated by the ribosomes independently of one another. Examples of IRES sequences which can be used according to the invention are those from picornaviruses (e.g. FMDV), pestiviruses (CFFV), polioviruses (PV), encephalomyocarditis viruses (ECMV), foot and mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), mouse leukoma virus (MLV), simian immunodeficiency viruses (SIV) or cricket paralysis viruses (CrPV).
According to a further particularly preferred embodiment, the immunsuppressive composition of the present invention, may comprise a mixture of at least one monocistronic modified (m)RNA as defined above, and at least one bi- or even multicistronic modified (m)RNA as defined above. The at least one monocistronic modified (m)RNA and/or the at least one bi- or even multicistronic modified (m)RNA preferably encode different therapeutically active proteins and/or antibodies as defined herein. However, the at least one monocistronic modified (m)RNA and the at least one bi- or even multicistronic modified (m)RNA may preferably also encode (in part) identical therapeutically active proteins and/or antibodies as defined herein. Use of a mixture of at least one monocistronic modified (m)RNA, as defined above, and at least one bi- or even multicistronic modified modified (m)RNA or of a mixture of more than one monocistronic modified (m)RNA, wherein the modified (m)RNA encodes at least one (preferably different) therapeutically active protein or antibody as defined above may be advantageous e.g. for a staggered, e.g. time dependent, administration of the immunosuppressive composition of the present invention to a patient in need thereof. The components of such an immunosuppressive composition of the present invention, particularly the at least one (different) modified (m)RNAs encoding the at least one (preferably different) therapeutically active protein and/or antibody, may be e.g. contained in (different parts of) a kit of parts composition or may be e.g. administered separately as components of different immunosuppressive compositions according to the present invention.
Short RNA oligonucleotide
In a second embodiment, the at least one modified (m)RNA (of the immunosuppressive composition) according to the invention may be a short RNA oligonucleotide. Short RNA oligonucleotides in the context of the present invention may comprise any RNA as defined above. Preferably, the short RNA oligonucleotide may be a single- or a double-stranded RNA oligonuclotide, more preferably a single-stranded RNA oligonucleotide. Even more preferably, the short RNA oligonucleotide may be a linear single-stranded RNA oligonucleotide. Also preferably, the short RNA oligonucleotides as used herein may comprise a length as defined above in general for RNA molecules, more preferably a length of 5 to 100, of 5 to 50, or of 5 of 30, and even more preferably a length of 20 to 100, of 20 to 80, or of 20 of 60 nucleotides.
siRNA
In a third embodiment, the at least one modified (m)RNA (of the immunosuppressive composition) according to the invention may be in the form of siRNA. A siRNA is of interest particularly in connection with the phenomenon of RNA interference. Attention was drawn to the phenomenon of RNA interference in the course of immunological research. In recent years, an RNA-based defence mechanism has been discovered, which occurs both in the kingdom of the fungi and in the plant and animal kingdom and acts as an "immune system of the genome". The system was originally described in various species independently of one another, first in C. elegans, before it was possible to identify the underlying mechanisms of the processes as being identical: RNA-mediated virus resistance in plants, PTGS (posttranscriptional gene silencing) in plants, and RNA interference in eukaryotes are accordingly based on a common procedure. The in vitro technique of RNA interference (RNAi) is based on double-stranded RNA molecules (dsRNA), which trigger the sequence- specific suppression of gene expression (Zamore (2001 ) Nat. Struct. Biol. 9: 746-750; Sharp (2001 ) Genes Dev. 5:485-490: Hannon (2002) Nature 41 : 244-251 ). In the transfection of mammalian cells with long dsRNA, the activation of protein kinase R and RnaseL brings about unspecific effects, such as, for example, an interferon response (Stark et al. (1998) Annu. Rev. Biochem. 67: 227-264; He and Katze (2002) Viral Immunol. 15: 95-1 19). These unspecific effects are avoided when shorter, for example 21 - to 23-mer, so-called siRNA (small interfering RNA), is used, because unspecific effects are not triggered by siRNA that is shorter than 30 bp (Elbashir et al. (2001) Nature 411 : 494-498). Recently, dsRNA molecules have also been used in vivo (McCaffrey et al. (2002), Nature 418: 38-39; Xia et al. (2002), Nature Biotech. 20: 1006-1010; Brummelkamp et al. (2002), Cancer Cell 2: 243-247). Thus, a siRNA as used for the modified (m)RNA according to the present invention typically comprises a (single- or) double stranded, preferably a double-stranded, RNA sequence with about 8 to 30 nucleotides, preferably 17 to 25 nucleotides, even more preferably from 20 to 25 and most preferably from 21 to 23 nucleotides. In principle, all the sections having a length of from 17 to 29, preferably from 19 to 25, most preferably from 21 to 23 base pairs that occur in the coding or non-coding (3'-and/or 5') region of a RNA sequence or a genomic sequence can serve as target sequence for such a siRNA. Equally, siRNAs can also be directed against nucleotide sequences (of the RNA or genomic sequence) of a (therapeutically relevant) protein, adjuvant protein or an antigen described hereinbefore either in their coding region or in their non-coding region, in particular in the 5' non-coding region of the RNA or genomic sequence, for example, therefore, against non-coding regions of the RNA having a regulatory function. The target sequence of the siRNA can therefore lie in the translated (coding) and/or untranslated region (3'-and/or 5') and/or in the region of the control elements. The target sequence of a siRNA can also lie in the overlapping region of untranslated and translated sequence; in particular, the target sequence can comprise at least one nucleotide upstream of the start triplet of the coding region of the (m)RNA. Antisense RNA
According to a fourth embodiment, the at least one modified (m)RNA (of the immunosuppressive composition) according to the invention may be an antisense RNA. In the context of the present invention, an antisense RNA is preferably a (single-stranded) RNA molecule transcribed off the coding, rather than the template, strand of DNA, so that it is complementary to the sense (messenger) RNA. An antisense RNA as defined herein typically forms a duplex between the sense and antisense RNA molecules and is thus capable to block transcription of the coding strand. An antisense RNA as used as the at least one modified (m)RNA (of the immunosuppressive composition) can be directed e.g. against nucleotide sequences of a (therapeutically relevant) protein, that do not lie in the coding region, in particular in the 5' non-coding region of the RNA, for example, therefore, against non-coding regions of the RNA having a regulatory function. The target sequence of the antisense RNA can therefore lie in the translated and/or untranslated region of the RNA and/or in the region of the control elements. The target sequence of a antisense RNA can also lie in the overlapping region of untranslated and translated sequence; in particular, the target sequence can comprise at least one nucleotide upstream of the start triplet of the coding region of the RNA. Preferably, the antisense RNA as used herein as the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) comprises a length as defined above in general for RNA molecules, more preferably a length of 1000 to 5000, of 500 to 5000, of 5 to 5000, or of 5 to 1000, 5 to 500, 5 to 250, of 5 to 100, of 5 to 50 or of 5 to 30 nucleotides, and even more preferably a length of 20 to 100, of 20 to 80, or of 20 to 60 nucleotides.
GC-Modifications of the modified (m)RNA According to one embodiment, the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) as defined above, particularly when occurring as a coding RNA, a short RNA oligonucleotide, a siRNA, an antisense RNA, riboswitches, ribozymes or aptamers, may be further GC-modified or even further modified. Some modifications may be, dependent on the type of RNA, be more suitable for an RNA in general, or, e.g. in the case of GC-modified (m)RNA sequences, be more suitable for coding RNA, preferably an mRNA. Such further modifications as defined herein preferably lead to a stabilized modified (m)RNA. According to one embodiment, such a stabilized modified (m)RNA may be prepared by modifying the G/C content of the coding region of the modified (m)RNA.
In a particularly preferred embodiment of the present invention, the G/C content of the coding region of the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) (in the following "native modified (m)RNA") is altered, preferably increased, compared to the G/C content of the coding region of the corresponding native inventive modified (m)RNA (of the inventive immunosuppressive composition). In this context, the encoded amino acid sequence of this G/C-increased modified (m)RNA is preferably not altered compared to the corresponding native modified (m)RNA. Such alteration of the GC-sequence may be termed in the following GC- stabilization.
This G/C-stabilization of the modified (m)RNA of the immunosuppressive composition of the present invention is based on the fact that the sequence of any (m)RNA region to be translated is important for efficient translation of that (m)RNA. Thus, the the sequence of various nucleotides is important. In particular, sequences having an increased G (guanosine)/C (cytosine) content are more stable than sequences having an increased A (adenosine)/U (uracil) content. According to the invention, the codons of the inventive modified (m)RNA (of the inventive immunosuppressive composition) are therefore varied compared to its native modified (m)RNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides. In respect to the fact that several codons code for one and the same amino acid (so-called degeneration of the genetic code), the most favorable codons for the stability can be determined (so-called alternative codon usage).
Depending on the amino acid to be encoded by the modified (m)RNA, there are various possibilities for G/C-modification of the modified (m)RNA sequence, compared to its native sequence. In the case of amino acids which are encoded by codons which contain exclusively G or C nucleotides, no G/C-modification of the codon is necessary. Thus, the codons for Pro (CCC or CCG), Arg (CGC or CGG), Ala (GCC or GCG) and GIy (GGC or GGG) require no G/C-modification, since no A or U is present. In contrast, codons which contain A and/or U nucleotides can be G/C-modified by substitution of other codons which code for the same amino acids but contain no A and/or U. Examples of these are:
the codons for Pro can be G/C-modified from CCU or CCA to CCC or CCG; the codons for Arg can be G/C-modified from CGU or CGA or AGA or AGG to CGC or CGG; the codons for Ala can be G/C-modified from GCU or GCA to GCC or GCG; the codons for GIy can be G/C-modified from GGU or GGA to GGC or GGG.
In other cases, although A or U nucleotides cannot be eliminated from the codons, it is however possible to decrease the A and U content by using codons which contain a lower content of A and/or U nucleotides. Examples of these are:
the codons for Phe can be G/C-modified from UUU to UUC; the codons for Leu can be G/C-modified from UUA, UUG, CUU or CUA to CUC or CUG; the codons for Ser can be G/C-modified from UCU or UCA or AGU to UCC, UCG or AGC; the codon for Tyr can be G/C-modified from UAU to UAC; the codon for Cys can be G/C-modified from UGU to UGC; the codon for His can be G/C-modified from CAU to CAC; the codon for GIn can be G/C-modified from CAA to CAG; the codons for He can be G/C-modified from AUU or AUA to AUC; the codons for Thr can be G/C-modified from ACU or ACA to ACC or ACG; the codon for Asn can be G/C-modified from AAU to AAC; the codon for Lys can be G/C-modified from AAA to AAG; the codons for VaI can be G/C-modified from GUU or GUA to GUC or GUG; the codon for Asp can be G/C-modified from GAU to GAC; the codon for GIu can be G/C-modified from GAA to GAG; the stop codon UAA can be G/C-modified to UAG or UGA.
In the case of the codons for Met (AUG) and Trp (UGG), on the other hand, there is no possibility of sequence modification. The substitutions listed above can be used either individually or in all possible combinations to increase the G/C content of the inventive modified (m)RNA (of the inventive immunosuppressive composition) compared to its particular native modified (m)RNA (i.e. the original sequence). Thus, for example, all codons for Thr occurring in the native sequence can be G/C-modified to ACC (or ACG). Preferably, however, for example, combinations of the above substitution possibilities are used:
substitution of all codons coding for Thr in the original sequence (native modified (m)RNA) to ACC (or ACG) and substitution of all codons originally coding for Ser to UCC (or UCG or AGC); substitution of all codons coding for lie in the original sequence to AUC and substitution of all codons originally coding for Lys to AAG and substitution of all codons originally coding for Tyr to UAC; substitution of all codons coding for VaI in the original sequence to GUC (or GUG) and substitution of all codons originally coding for GIu to GAG and substitution of all codons originally coding for Ala to GCC (or GCG) and substitution of all codons originally coding for Arg to CGC (or CGG); substitution of all codons coding for VaI in the original sequence to GUC (or GUG) and substitution of all codons originally coding for GIu to GAG and substitution of all codons originally coding for Ala to GCC (or GCG) and substitution of all codons originally coding for GIy to GGC (or GGG) and substitution of all codons originally coding for Asn to AAC; substitution of all codons coding for VaI in the original sequence to GUC (or GUG) and substitution of all codons originally coding for Phe to UUC and substitution of all codons originally coding for Cys to UGC and substitution of all codons originally coding for Leu to CUG (or CUC) and substitution of all codons originally coding for GIn to CAG and substitution of all codons originally coding for Pro to CCC (or CCG); etc.
Preferably, the G/C content of the coding region of the GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition) is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the coding region of the native modified (m)RNA which codes for a protein. According to a specific embodiment at least 60%, more preferably at least 70 %, even more preferably at least 80% and most preferably at least 90%, 95% or even 100% of the substitutable codons in the region coding for a protein or the whole sequence of the native modified (m)RNA sequence are substituted, thereby increasing the GC/content of said sequence.
In this context, it is particularly preferable to increase the G/C content of the native modified (m)RNA to the maximum (i.e. 100% of the substitutable codons), in particular in the region coding for a protein, compared to the native sequence.
According to the invention, a further preferred modification of the native inventive modified (m)RNA (of the inventive immunosuppressive composition) is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells. Thus, if so-called "rare codons" are present in a native modified (m)RNA sequence to an increased extent, the corresponding G/C-stabilized or native modified (m)RNA sequence may be translated to a significantly poorer degree than in the case, where codons coding for relatively "frequent" tRNAs are present.
According to the invention, in the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition), the region which codes for a therapeutically active protein, or antibody, etc., as defined above, is GC-stabilized compared to the corresponding region of the native modified (m)RNA such that at least one codon of the native sequence which codes for a tRNA which is relatively rare in the cell is exchanged for a codon which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA. By this modification, the native modified (m)RNA sequences are GC-stabilized such that codons for which frequently occurring tRNAs are available are inserted. In other words, according to the invention, by this modification all codons of the native sequence which code for a tRNA which is relatively rare in the cell can in each case be exchanged for a codon which codes for a tRNA which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA.
Which tRNAs occur relatively frequently in the cell and which, in contrast, occur relatively rarely is known to a person skilled in the art; cf. e.g. Akashi, Curr. Opin. Genet. Dev. 2001 , 1 1 (6): 660-666. The codons which use for the particular amino acid the tRNA which occurs the most frequently, e.g. the GIy codon, which uses the tRNA which occurs the most frequently in the (human) cell, are particularly preferred.
According to the invention, it is particularly preferable to link the sequential G/C content which is increased, in particular maximized, in the GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition), with the "frequent" codons without modifying the amino acid sequence of the protein encoded by the coding region of the (m)RNA. This preferred embodiment allows provision of a particularly efficiently translated and GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition).
The determination of the necessary GC modification of a GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition) as described above (increased
G/C content; exchange of codons) can be carried out using the computer program as explained in WO 02/098443 - the specific disclosure content of which is included in its full scope in the present invention. Using this computer program, the nucleotide sequence of any desired modified (m)RNA as defined above can be GC-stabilized with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence coded by the GC-stabilized inventive modified (m)RNA (of the inventive immunosuppressive composition) preferably not being further modified compared to the native modified (m)RNA sequence. Alternatively, it is also possible to modify only the G/C content or only the codon usage compared to the original sequence. The source code in Visual Basic 6.0 (development environment used: Microsoft
Visual Studio Enterprise 6.0 with Servicepack 3) is also described in WO 02/098443.
In a further preferred embodiment of the present invention, the A/U content in the environment of the ribosome binding site of the (optionally already GC-stabilized) inventive modified (m)RNA (of the inventive immunosuppressive composition) is increased compared to the A/U content in the environment of the ribosome binding site of its particular native (m)RNA. This modification (an increased A/U content around the ribosome binding site) increases the efficiency of ribosome binding to the modified (m)RNA. An effective binding of the ribosomes to the ribosome binding site (Kozak sequence: GCCGCCACCAUGG (SEQ ID NO: 1 ), the AUG forms the start codon) in turn has the effect of an efficient translation of the modified (m)RNA.
According to a further embodiment of the present invention the inventive modified (m)RNA (of the inventive immunosuppressive composition) may be further modified with respect to potentially destabilizing sequence elements. Particularly, the coding region and/or the 51 and/or 3' untranslated region of this modified (m)RNA may be further modified compared to the particular native modified (m)RNA such that is contains no destabilizing sequence elements, the coded amino acid sequence of the modified (m)RNA preferably not being modified compared to its particular native modified (m)RNA. It is known that, for example, in sequences of eukaryotic RNAs destabilizing sequence elements (DSE) occur, to which signal proteins bind and regulate enzymatic degradation of RNA in vivo. For further stabilization of the modified (m)RNA, optionally in the region which encodes for a protein, one or more such further modifications compared to the corresponding region of the native modified (m)RNA can therefore be carried out, so that no or substantially no destabilizing sequence elements are contained there. According to the invention, DSE present in the untranslated regions (3'- and/or 5'-UTR) can also be eliminated from the inventive modified (m)RNA (of the inventive immunosuppressive composition) by such further modifications.
Such destabilizing sequences are e.g. AU-rich sequences (AURES), which occur in 3'-UTR sections of numerous unstable RNAs (Caput et a/., Proc. Natl. Acad. Sci. USA 1986, 83: 1670 to 1674). The inventive modified (m)RNA (of the inventive immunosuppressive composition) is therefore preferably further modified compared to the native modified (m)RNA such that the modified (m)RNA contains no such destabilizing sequences. This also applies to those sequence motifs which are recognized by possible endonucleases, e.g. the sequence GAACAAG, which is contained in the 3'-UTR segment of the gene which codes for the transferrin receptor (Binder eta/., EMBO J. 1994, 13: 1969 to 1980). These sequence motifs are also preferably removed according to the invention in the inventive modified (m)RNA (of the inventive immunosuppressive composition). Also preferably according to the invention, the inventive modified (m)RNA (of the inventive immunosuppressive composition) has, in a further modified form of the modified (m)RNA, at least one IRES as defined above and/or at least one 5' and/or 3' stabilizing sequence, e.g. to enhance ribosome binding or to allow expression of different encoded proteins as defined above located on at least one (bi- or even multicistronic) inventive modified (m)RNA (of the inventive immunosuppressive composition).
According to the invention, the inventive modified (m)RNA (of the inventive immunosuppressive composition), preferably when occurring as an mRNA, furthermore preferably has at least one 5' and/or 3' stabilizing sequence. These stabilizing sequences in the 5' and/or 3' untranslated regions have the effect of increasing the half-life of the modified (m)RNA in the cytosol. These stabilizing sequences can have 100% sequence homology to naturally occurring sequences which occur in viruses, bacteria and eukaryotes, but can also be partly or completely synthetic. The untranslated sequences (UTR) of the globin gene, e.g. from Homo sapiens or Xenopus laevis may be mentioned as an example of stabilizing sequences which can be used in the present invention for a further stabilized modified (m)RNA as contained in the inventive immunosuppressive composition. Another example of a stabilizing sequence has the general formula (C/U)CCANxCCC(U/A)PyxUC(C/U)CC (SEQ ID NO: 2), which is contained in the 3'UTR of the very stable RNA which codes for globin, (l)-collagen, 15-lipoxygenase or for tyrosine hydroxylase (cf. Holcik et al., Proc. Natl. Acad. Sci. USA 1997, 94: 2410 to 2414). Such stabilizing sequences can of course be used individually or in combination with one another and also in combination with other stabilizing sequences known to a person skilled in the art. The inventive modified (m)RNA (of the inventive immunosuppressive composition) is therefore preferably present as globin UTR (untranslated regions)-stabilized RNA, in particular as globin UTR-stabilized modified (m)RNA. Particularly preferred in the context of the present invention is the 3'-UTR derived from the alpha-Globin gene.
Any of the above modifications may be applied to the inventive modified (m)RNA (of the inventive immunosuppressive composition), and further to any modified (m)RNA as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the respective modified (m)RNA. A person skilled in the art will be able to take his choice accordingly.
According to the invention, the inventive modified (m)RNA (of the inventive immunosuppressive composition) may be prepared using any naturally or synthetic DNA or RNA sequence available in the art as a template, i.e. any suitable (desoxy)ribonucleic acid. Such naturally or synthetic DNA or RNA sequences may be obtained from any synthetic or naturally occurring source, which is available to a skilled person, e.g. may be derived from a protein or peptide library or may be transcribed from a nucleic acid library, such as a cDNA library, or may be obtained from any living or dead tissue, from a sample obtained from e.g. a human, animal or bacterial source. Alternatively, the inventive modified (m)RNA (of the inventive immunosuppressive composition) may be prepared synthetically by methods known to a person skilled in the art, e.g., by solid phase synthesis or any other suitable method for preparing nucleic acid sequences, particularly RNA sequences. Furthermore, substitutions, additions or eliminations of bases in these sequences are preferably carried out using a DNA matrix for preparation of the modified (m)RNA of the immunosuppressive composition of the present invention or by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation strategy (see e.g. Maniatis et a/., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 3rd ed., Cold Spring Harbor, NY, 2001 ). The modification(s) of the inventive modified (m)RNA (of the inventive immunosuppressive composition) can furthermore be introduced into the RNA by means of methods known to a person skilled in the art. Suitable methods are, for example, synthesis methods using (automatic or semi-automatic) oligonucleotide synthesis devices, biochemical methods, such as, for example, in vitro transcription methods, etc.. In this connection there can preferably be used in the case of (relatively short) sequences, whose length generally does not exceed from 50 to 100 nucleotides, synthesis methods using (automatic or semi-automatic) oligonucleotide synthesis devices as well as in vitro transcription methods. In the case of (relatively long) sequences, for example sequences having a length of more than 50 to 100 nucleotides, biochemical methods are preferably suitable, such as, for example, in vitro transcription methods. However, even longer inventive modified (m)RNA molecules as defined herein may be synthesized synthetically by coupling various synthesized fragments covalently. The inventive modified (m)RNA of the immunosuppressive composition of the present invention can likewise be stabilized by the use of nanoplexes (nanoparticular systems), lipoplexes (liposomal systems), or the use of polyplexes or cationic polymers, e.g. by associating or complexing the modified (m)RNA with, or binding it, thereto. Such nanoplexes (nanoparticular systems) involve use of polyacrylates, polyamides, polystyrene, cyanoacrylates, polylactat (PLA), poly(lactic-co-glycolic acid) (PLGA), polyethyl, etc., as carrier systems for the transport of nucleic acids into cells or tissues. Lipoplexes or liposomal systems typically involve use of cationic lipids, which are capable to mimick a cell membrane. Thereby, the positively charged moiety of the lipids interacts with the negatively charged moiety of the nucleic acids and thus enables fusion with the cell membrane. Lipoplexes or liposomal systems include e.g. DOTMA, DOPE, DOSPA, DOTAP, DC-Choi, EDMPC, etc.. Polyplexes (cationic polymers) typically form a complex with negatively charged nucleic acids leading to a condensation of nucleic acids and protecting these nucleic acids against degradation. Transport into cells using polyplexes (cationic polymers) typically occurs via receptor mediated endocytosis. Thereby, the DNA is coupled to a distinct molecule, such as transferrin, via e.g. the polyplex poly-L-lysine (PLL), which binds to a surface receptor and triggers endocytosis. Polyplexes (cationic polymers) may also include polycations selected from from oligoarginines having formula (I): (Arg)|,(Lys)m;(His)n;(Orn)o;(Xaa)x/ wherein I + m + n +o + x = 8-15, and I, m, n or o independently of each other may be any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 or 15, provided that the overall content of Arg, Lys, His and Orn represents at least 70% of all amino acids of the oligopeptide; and Xaa may be any amino acid selected from native (= naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x may be any number selected from 0, 1 , 2, 3 or 4, provided, that the overall content of Xaa does not exceed 30 % of all amino acids of the oligopeptide. Particularly preferred are oligoarginines Arg7, Argg, Arg9, but also Lys7, LyS8, Lys9, etc.. Polyplexes (cationic polymers) include furthermore e.g. poly-L-lysine (PLL), chitosan, polyethylenimine (PEI), polydimethylaminoethylmethacrylate (PD-MAEMA), polyamidoamine (PAMAM). Cationic compounds may furthermore include any further (poly)cationic peptide or protein. In particular the use of protamine, nucleoline, spermin or spermidine as the polycationic, nucleic-acid-binding protein to inventive modified (m)RNA of the immunosuppressive composition of the present invention is particularly effective. Furthermore, the use of other cationic peptides or proteins is likewise possible, such as histones, cationic polysaccharides, for example chitosan, polybrene, polyethyleneimine (PEI), etc., or cationic lipids, e.g. oligofectamine as a lipid based complexation reagent. The procedure for stabilizing the modified (m)RNA is using polycationic compounds in general described in EP-A-1083232, the disclosure of which is incorporated by reference into the present invention in its entirety.
According to a further embodiment, the present invention also provides a pharmaceutical composition, comprising an inventive immunosuppressive composition as defined above and optionally a pharmaceutically acceptable carrier, adjuvant, and/or vehicle.
As a first component, an inventive pharmaceutical composition comprises a modified (m)RNA as defined above, e.g. as component of an inventive immunosuppressive composition as defined above, wherein at least one nucleoside of the modified (m)RNA of the inventive immunosuppressive composition has been modified as defined above, i.e. at least one nucleoside comprises: a) a chemical modification at the 4-, 5-or 6-position of the pyrimidine base of the nucleosides cytidine and/or uridine as defined above; b) a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides adenosine, inosine and/or guanosine as defined above; and/or c) a chemical modification at the 2'-position of the sugar of the nucleosides adenosine, inosine, guanosine, cytidine and/or uridine as defined above, and wherein the modified (m)RNA is suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA.
Accordingly, the inventive pharmaceutical composition, due to the content of the modified (m)RNA as defined herein, is suitable for suppressing and/or avoiding an innate immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA, i.e. not an overall immune response but the innate immune response caused by administering the unmodified RNA as such is reduced or even avoided with the modified (m)RNA as defined herein. Furthermore, the inventive pharmaceutical composition may comprise a pharmaceutically acceptable carrier. In the context of the inventive pharmaceutical composition, a pharmaceutically acceptable carrier typically includes the liquid or non-liquid basis of the inventive pharmaceutical composition. The term "(compatible) pharmaceutically acceptable carrier" used here preferably includes the liquid or non-liquid basis of the inventive pharmaceutical composition. The term "compatible" as used herein means that the constituents of the inventive pharmaceutical composition are capable of being mixed with the pharmaceutically active component, i.e. with the modified (m)RNA as defined herein, in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the inventive pharmaceutical composition under usual use conditions. Pharmaceutically acceptable carriers must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated.
If the inventive pharmaceutical composition is provided in liquid form, the pharmaceutically acceptable carrier will typically comprise one or more (compatible) pharmaceutically acceptable liquid carriers. The composition may comprise as (compatible) pharmaceutically acceptable liquid carriers e.g. pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g phosphate, citrate etc. buffered solutions, vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid, etc.. Particularly for injection of the inventive pharmaceutical composition, a buffer, preferably an aqueous buffer, may be used, containing a sodium salt, preferably at least 50 mM of a sodium salt, a calcium salt, preferably at least 0,01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt. According to a preferrred embodiment, the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc.. Without being limited thereto, examples of sodium salts include e.g. NaCI, NaI, NaBr, Na2CO3, NaHCO3, Na2SO4, examples of the optional potassium salts include e.g. KCl, Kl, KBr, K2CO3, KHCO3, K2SO4, and examples of calcium salts include e.g. CaCI2, CaI2, CaBr2, CaCO3, CaSO4, Ca(OH)2. Furthermore, organic anions of the aforementioned cations may be contained in the buffer. According to a more preferred embodiment, the buffer suitable for injection purposes as defined above, may contain salts selected from sodium chloride (NaCI), calcium chloride (CaCI2) and optionally potassium chloride (KCI), wherein further anions may be present additional to the chlorides. Typically, the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCI), at least 3 itiM potassium chloride (KCI) and at least 0,01 mM calcium chloride (CaCI2). The injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e. the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects. Reference media are e.g. in ,,/n vivd' methods occurring liquids such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in "in vitrd' methods, such as common buffers or liquids. Such common buffers or liquids are known to a skilled person. Ringer solution or Ringer-Lactate solution is particularly preferred as a liquid basis.
If the inventive pharmaceutical composition is provided in solid form, the pharmaceutically acceptable carrier will typically comprise one or more (compatible) pharmaceutically acceptable solid carriers. The composition may comprise as (compatible) pharmaceutically acceptable solid carriers e.g. one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person. Some examples of such (compatible) pharmaceutically acceptable solid carriers are e.g. sugars, such as, for example, lactose, glucose and sucrose; starches, such as, for example, corn starch or potato starch; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulphate, etc..
The choice of a (compatible) pharmaceutically acceptable carrier as defined above is determined in principle by the manner in which the pharmaceutical composition according to the invention is to be administered. The inventive pharmaceutical composition can be administered, for example, systemically or topically. Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, , intradermal, intranasal, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Routes for local administration in general include topically, rectally, nasally, buccally, vaginally routes or administration or via an implanted reservoir, but also transdermal, intramuscular or subcutaneous injection.
Preferably, the inventive pharmaceutical composition may be administered by parenteral injection, more preferably by subcutaneous, intravenous, intramuscular, intradermal, intraarticular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or via infusion techniques. Sterile injectable forms of the inventive pharmaceutical compositions may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenteral ly-acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di- glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutical ly-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation of the inventive pharmaceutical composition. The inventive pharmaceutical composition as defined above may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient, i.e. the inventive modified (m)RNA (of the inventive immunosuppressive composition) as contained in the inventive pharmaceutical composition, is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. Pharmaceutically acceptable carriers for the preparation of unit dose forms, which can be used for oral administration, are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art.
The inventive pharmaceutical composition may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including diseases of the skin or of any other accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs. For topical applications, the inventive pharmaceutical compositions may be formulated in a suitable ointment containing the inventive modified (m)RNA (of the inventive immunosuppressive composition) as contained in the inventive pharmaceutical composition, suspended or dissolved in one or more carriers. Carriers for topical administration include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the inventive pharmaceutical composition can be formulated in a suitable lotion or cream. In the context of the present invention, suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
The inventive pharmaceutical composition typically comprises a "safe and effective amount" of the at least one inventive modified (m)RNA (of the inventive immunosuppressive composition) as contained in the inventive pharmaceutical composition. As used herein, a "safe and effective amount" means an amount of the at least one modified (m)RNA as defined herein in the inventive pharmaceutical composition as defined above that is sufficient to significantly induce a positive modification of a disease or disorder as defined herein. At the same time, however, a "safe and effective amount" is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment. A "safe and effective amount" of the at least one modified (m)RNA as defined herein will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the activity of the specific modified (m)RNA as defined herein employed, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying doctor. The inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine.
Further additives which may be included in the inventive pharmaceutical compositions are emulsifiers, such as, for example, Tween®; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet- forming agents; stabilizers; antioxidants; preservatives.
According to a specific embodiment, the inventive pharmaceutical composition may be provided as a vaccine. Such an inventive vaccine is typically composed like the inventive pharmaceutical composition and preferably allows to provide an active or a passive adaptive immune response of a patient to be treated, e.g. by using a modified (m)RNA, encoding an antibody as defined above. Alternatively or additionally the inventive vaccine may contain any of the above mentioned antibodies as a further component of the vaccine.
The inventive vaccine may also comprise a pharmaceutically acceptable carrier, adjuvant, and/or vehicle as defined above for the inventive pharmaceutical composition. In the specific context of the inventive vaccine, the choice of a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered. The inventive vaccine can be administered, for example, systemically or locally as defined above. More preferably, vaccines may be administered by an intradermal, subcutaneous, or intramuscular route. Inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid) form. The suitable amount of the inventive vaccine to be administered can be determined by routine experiments with animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non-human primate models. Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4. Suitable carriers for injection include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices.
According to a further preferred object of the present invention, the modified (m)RNA as defined herein and/or the inventive immunosuppressive composition may be used for the preparation of an inventive pharmaceutical composition or an inventive vaccine, preferably all as defined herein, for the prophylaxis, treatment, and/or amelioration of any of the diseases and disorders as defined herein.
Accordingly, the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. cancer or tumour diseases, preferably selected from melanomas, malignant melanomas, colon carcinomas, lymphomas, sarcomas, blastomas, renal carcinomas, gastrointestinal tumours, gliomas, prostate tumours, bladder cancer, rectal tumours, stomach cancer, oesophageal cancer, pancreatic cancer, liver cancer, mammary carcinomas (= breast cancer), uterine cancer, cervical cancer, acute myeloid leukaemia (AML), acute lymphoid leukaemia (ALL), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), hepatomas, various virus-induced tumours such as, for example, papilloma virus-induced carcinomas (e.g. cervical carcinoma = cervical cancer), adenocarcinomas, herpes virus-induced tumours (e.g. Burkitt's lymphoma, EBV-induced B-cell lymphoma), heptatitis B-induced tumours (hepatocell carcinomas), HTL V-1 - and HTLV-2-induced lymphomas, acoustic neuroma, lung carcinomas (= lung cancer = bronchial carcinoma), small-cell lung carcinomas, pharyngeal cancer, anal carcinoma, glioblastoma, rectal carcinoma, astrocytoma, brain tumours, retinoblastoma, basalioma, brain metastases, medulloblastomas, vaginal cancer, pancreatic cancer, testicular cancer, Hodgkin's syndrome, meningiomas, Schneeberger disease, hypophysis tumour, Mycosis fungoides, carcinoids, neurinoma, spinalioma, Burkitt's lymphoma, laryngeal cancer, renal cancer, thymoma, corpus carcinoma, bone cancer, non-Hodgkin's lymphomas, urethral cancer, CUP syndrome, head/neck tumours, oligodendroglioma, vulval cancer, intestinal cancer, colon carcinoma, oesophageal carcinoma (= oesophageal cancer), wart involvement, tumours of the small intestine, craniopharyngeomas, ovarian carcinoma, genital tumours, ovarian cancer (= ovarian carcinoma), pancreatic carcinoma (= pancreatic cancer), endometrial carcinoma, liver metastases, penile cancer, tongue cancer, gall bladder cancer, leukaemia, plasmocytoma, lid tumour, prostate cancer (= prostate tumours), etc.. With regard to the present invention, e.g. an inventive pharmaceutical composition or vaccine may be provided in this context, which contains a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of cancer or tumour diseases.
Furthermore, the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases. Such infectious diseases, preferably to (viral, bacterial or protozoological) infectious diseases, are typically selected from influenza, malaria, SARS, yellow fever, AIDS, Lyme borreliosis, Leishmaniasis, anthrax, meningitis, viral infectious diseases such as AIDS, Condyloma acuminata, hollow warts, Dengue fever, three-day fever, Ebola virus, cold, early summer meningoencephalitis (FSME), flu, shingles, hepatitis, herpes simplex type I, herpes simplex type II, Herpes zoster, influenza, Japanese encephalitis, Lassa fever, Marburg virus, measles, foot-and-mouth disease, mononucleosis, mumps, Norwalk virus infection, Ffeiffer's glandular fever, smallpox, polio (childhood lameness), pseudo-croup, fifth disease, rabies, warts, West Nile fever, chickenpox, cytomegalic virus (CMV), bacterial infectious diseases such as miscarriage (prostate inflammation), anthrax, appendicitis, borreliosis, botulism, Camphylobacter, Chlamydia trachomatis (inflammation of the urethra, conjunctivitis), cholera, diphtheria, donavanosis, epiglottitis, typhus fever, gas gangrene, gonorrhoea, rabbit fever, Heliobacter pylori, whooping cough, climatic bubo, osteomyelitis, Legionnaire's disease, leprosy, listeriosis, pneumonia, meningitis, bacterial meningitis, anthrax, otitis media, Mycoplasma hominis, neonatal sepsis (Chorioamnionitis), noma, paratyphus, plague, Reiter's syndrome, Rocky Mountain spotted fever, Salmonella paratyphus, Salmonella typhus, scarlet fever, syphilis, tetanus, tripper, tsutsugamushi disease, tuberculosis, typhus, vaginitis (colpitis), soft chancre, and infectious diseases caused by parasites, protozoa or fungi, such as amoebiasis, bilharziosis, Chagas disease, Echinococcus, fish tapeworm, fish poisoning (Ciguatera), fox tapeworm, athlete's foot, canine tapeworm, candidosis, yeast fungus spots, scabies, cutaneous Leishmaniosis, lambliasis (giardiasis), lice, malaria, microscopy, onchocercosis (river blindness), fungal diseases, bovine tapeworm, schistosomiasis, sleeping sickness, porcine tapeworm, toxoplasmosis, trichomoniasis, trypanosomiasis (sleeping sickness), visceral Leishmaniosis, nappy/diaper dermatitis or miniature tapeworm, or prion diseases, including e.g. Creutzfeld- Jakob disease, BSE, scrapie, Kuru, etc.. With regard to the present invention, e.g. an inventive pharmaceutical composition or vaccine may be provided in this context, which contains a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of infectious diseases.
Likewise, the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. autoimmune diseases. Autoimmune diseases as defined herein may involve antibody-mediated or T-cell mediated immunity and/or can be broadly divided into systemic and organ-specific or localised autoimmune disorders, depending on the principal clinico-pathologic features of each disease. Autoimmune diseases may be divided into the categories of systemic syndromes, including systemic lupus erythematosus (SLE), Sjogren's syndrome, Scleroderma, Rheumatoid Arthritis including juvenile (rheumatoid) arthritis, and polymyositis or local syndromes which may be endocrinologic (type I diabetes (Diabetes mellitus Type 1), Hashimoto's thyroiditis, Addison's disease etc.), dermatologic (pemphigus vulgaris), haematologic (autoimmune haemolytic anaemia), neural (multiple sclerosis) or can involve virtually any circumscribed mass of body tissue. The autoimmune diseases to be treated may be selected from the group consisting of type I autoimmune diseases or type Il autoimmune diseases or type III autoimmune diseases or type IV autoimmune diseases, such as, for example, multiple sclerosis (MS), rheumatoid arthritis, diabetes, type I diabetes (Diabetes mellitus Type 1 ), chronic polyarthritis, Basedow's disease, autoimmune forms of chronic hepatitis, colitis ulcerosa, type I allergy diseases, type Il allergy diseases, type III allergy diseases, type IV allergy diseases, fibromyalgia, hair loss, alopecia, alopecia areata, Bechterew's disease, Crohn's disease, Myasthenia gravis, neurodermitis, Polymyalgia rheumatica, progressive systemic sclerosis (PSS), rheumatic arthritis, psoriasis, vasculitis, etc, type Il diabetes, graft versus host disease, transplanted organ rejection, asthma, acquired hemophilia, ankylosing spondylitis, antiphospholipid syndrome, autoimmune hepatitis, autoimmune hemolytic anemia, Behcet's syndrome, cardiomyopathy, celiac sprue dermatitis, chronic fatigue immune dysfunction syndrome (CFIDS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, cicatricial pemphigoid, CREST syndrome, cold agglutinin disease, dermatomyositis, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia, fibromyositis, Guillain-Barre syndrome, idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura, IgA nephropathy, inflammatory bowel disease (including Crohn's disease and ulcerative colitis), juvenile arthritis, lichen planus, myasthenia gravis, multiple sclerosis, mixed connective tissue disease, polyarteritis nodosa, polychondritis, polyglandular syndromes, polymyalgia rheumatica, primary agammaglobulinemia, primary biliary cirrhosis, psoriasis, Raynaud's phenomena, Reiter's syndrome, rheumatoid arthritis (RA), Sjorgen's syndrome, sarcoidosis, stiff-man syndrome, systemic lupus erythematosus (SLE), Takayasu arthritis, temporal arteritis/giant cell arteritis, uveitis, vasculitis, and vitiligo.
While the exact mode as to why the immune system induces a immune reaction against autoantigens has not been elucidated so far, there are several findings with regard to the etiology. Accordingly, the autoreaction may be due to a T-CeII bypass. A normal immune system requires the activation of B-cells by T-cells before the former can produce antibodies in large quantities. This requirement of a T-cell can be by-passed in rare instances, such as infection by organisms producing super-antigens, which are capable of initiating polyclonal activation of B-cells, or even of T-cells, by directly binding to the β-subunit of T-cell receptors in a non-specific fashion. Another explanation deduces autoimmune diseases from a Molecular Mimicry. An exogenous antigen may share structural similarities with certain host antigens; thus, any antibody produced against this antigen (which mimics the self-antigens) can also, in theory, bind to the host antigens and amplify the immune response. The most striking form of molecular mimicry is observed in Group A beta- haemolytic streptococci, which shares antigens with human myocardium, and is responsible for the cardiac manifestations of Rheumatic Fever. The present invention allows therefore to provide an inventive immunosuppressive composition or pharmaceutical composition or vaccine containing a modified (m)RNA coding for an autoantigen, which typically allows the immune system to be desensitized by triggering an adaptive immune response towards this specific autoantigen. In this specific context, an immunization against such an autoantigen preferably leads to a decrease in the overall immune response and therefore forms a basis for treatments of allergic diseases or disorders or autoimmune diseases or disorders.
Additionally, the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. allergic disorders or diseases. Allergy is a condition that typically involves an abnormal, acquired immunological hypersensitivity to certain foreign antigens or allergens. Allergies normally result in a local or systemic inflammatory response to these antigens or allergens and leading to an immunity in the body against these allergens. Allergens in this context include e.g. grasses, pollens, molds, drugs, or numerous environmental triggers, etc. Without being bound to theory, several different disease mechanisms are supposed to be involved in the development of allergies. According to a classification scheme by P. GeII and R. Coombs the word "allergy" was restricted to type I hypersensitivities, which are caused by the classical IgE mechanism. Type I hypersensitivity is characterised by excessive activation of mast cells and basophils by IgE, resulting in a systemic inflammatory response that can result in symptoms as benign as a runny nose, to life-threatening anaphylactic shock and death. Well known types of allergies include, without being limited thereto, allergic asthma (leading to swelling of the nasal mucosa), allergic conjunctivitis (leading to redness and itching of the conjunctiva), allergic rhinitis ("hay fever"), anaphylaxis, angiodema, atopic dermatitis (eczema), urticaria (hives), eosinophilia, respiratory, allergies to insect stings, skin allergies (leading to and including various rashes, such as eczema, hives (urticaria) and (contact) dermatitis), food allergies, allergies to medicine, etc. With regard to the present invention, e.g. an inventive pharmaceutical composition or vaccine may be provided, which contains a modified (m)RNA as defined herein, encoding for a therapeutically active protein or antibody as defined above suitable for the treatment of allergic disorders or diseases, which allows a desensitizing of the immune reaction or which triggers a specific immune response, which, in turn, also allows a desensitizing of the immune reaction.
Furthermore, the modified (m)RNA as defined herein, the modified (m)RNA as defined herein, the inventive immunosuppressive composition, the inventive pharmaceutical composition or the inventive vaccine, containing the modified (m)RNA, or the modified (m)RNA may be used for (the preparation of a medicament for) the prophylaxis, treatment, and/or amelioration of e.g. genetic diseases, which are caused by genetic defects, e.g. due to gene mutations resulting in loss of protein activity or regulatory mutations which do not allow transcription or translation of the protein. Frequently, these diseases lead to metabolic disorders or other symptoms, e.g. muscle dystrophy. Accordingly, the present invention allows to treat these diseases by providing the modified (m)RNA of the inventive immunosuppressive composition, which allows sufficient level of the protein to be translated without triggering an innate immune response due to administration of RNA. Insofar, the following diseases may be treated: 3-beta-hydroxysteroid dehydrogenase deficiency (type II); 3-ketothiolase deficiency; 6-mercaptopurine sensitivity; Aarskog-Scott syndrome; Abetalipoproteinemia; Acatalasemia; Achondrogenesis; Achondrogenesis- hypochondrogenesis; Achondroplasia; Achromatopsia; Acromesomelic dysplasia (Hunter- Thompson type); ACTH deficiency; Acyl-CoA dehydrogenase deficiency (short-chain, medium chain, long chain); Adenomatous polyposis coli; Adenosin-deaminase deficiency; Adenylosuccinase deficiency; Adhalinopathy; Adrenal hyperplasia, congenital (due to 1 1 - beta-hydroxylase deficiency; due to 17-alpha-hydroxylase deficiency; due to 21 - hydroxylase deficiency); Adrenal hypoplasia, congenital, with hypogonadotropic hypogonadism; Adrenogenital syndrom; Adrenoleukodystrophy; Adrenomyeloneuropathy; Afibrinogenemia; Agammaglobulinemia; Alagille syndrome; Albinism (brown, ocular, oculocutaneous, rufous); Alcohol intolerance, acute; Aldolase A deficiency; Aldosteronism, glucocorticoid-remediable; Alexander disease; Alkaptonuria; Alopecia universalis; Alpha-1 - antichymotrypsin deficiency; Alpha-methylacyl-CoA racemase deficiency; Alpha- thalassemia/mental retardation syndrome; Alport syndrome; Alzheimer disease-1 (APP- related); Alzheimer disease-3; Alzheimer disease-4; Amelogenesis imperfecta; Amyloid neuropathy (familial, several allelic types); Amyloidosis (Dutch type; Finnish type; hereditary renal; renal; senile systemic); Amytrophic lateral sclerosis; Analbuminemia; Androgen insensitivity; Anemia (Diamond-Blackfan); Anemia (hemolytic, due to PK deficiency); Anemia (hemolytic, Rh-null, suppressor type); Anemia (neonatal hemolytic, fatal and nearfatal); Anemia (sideroblastic, with ataxia); Anemia (sideroblastic/hypochromic); Anemia due to G6PD deficiency; Aneurysm (familial arterial); Angelman syndrome; Angioedema; Aniridia; Anterior segment anomalies and cataract; Anterior segment mesenchymal dysgenesis; Anterior segment mesenchymal dysgenesis and cataract; Antithrombin III deficiency; Anxiety-related personality traits; Apert syndrome; Apnea (postanesthetic); ApoA-l and apoC-lll deficiency (combined); Apolipoprotein A-Il deficiency; Apolipoprotein B-100 (ligand-defective); Apparent mineralocorticoid excess (hypertension due to); Argininemia; Argininosuccinicaciduria; Arthropathy (progressive pseudorheumatoid, of childhood); Aspartylglucosaminuria; Ataxia (episodic); Ataxia with isolated vitamin E deficiency; Ataxia-telangiectasia; Atelosteogenesis II; ATP-dependent DNA ligase I deficiency; Atrial septal defect with atrioventricular conduction defects; Atrichia with papular lesions; Autism (succinylpurinemic); Autoimmune polyglandular disease, type I; Autonomic nervous system dysfunction; Axenfeld anomaly; Azoospermia; Bamforth-Lazarus syndrome; Bannayan-Zonana syndrome; Barthsyndrome; Bartter syndrome (type 2 or type 3); Basal cell carcinoma ; Basal cell nevus syndrome; BCG infection; Beare-Stevenson cutis gyrata syndrome; Becker muscular dystrophy; Beckwith- Wiedemann syndrome; Bernard-Soulier syndrome (type B; type C); Bethlem myopathy; Bile acid malabsorption, primary ; Biotinidase deficiency; Bladder cancer; Bleeding disorder due to defective thromboxane A2 receptor; Bloom syndrome; Brachydactyly (type Bl or type C); Branchiootic syndrome; Branchiootorenal syndrome; Breast cancer (invasive intraductal; lobular; male, with Reifenstein syndrome; sporadic); Breast cancer-1 (early onset); Breast cancer-2 (early onset); Brody myopathy; Brugada syndrome; Brunner syndrome; Burkitt lymphoma; Butterfly dystrophy (retinal); C1 q deficiency (type A; type B; type C ); C1 r/C1 s deficiency; C1 s deficiency, isolated; C2 deficiency ; C3 deficiency; C3b inactivator deficiency; C4 deficiency; C8 deficiency, type II; C9 deficiency; Campomelic dysplasia with autosomal sex reversal; Camptodactyly-arthropathy-coxa varapericarditis syndrome; Canavan disease; Carbamoylphosphate synthetase I deficiency; Carbohydrate-deficient glycoprotein syndrome (type I; type Ib; type II); Carcinoid tumor of lung; Cardioencephalomyopathy (fatal infantile, due to cytochrome c oxidase deficiency); Cardiomyopathy (dilated; X-linked dilated; familial hypertrophic; hypertrophic); Carnitine deficiency (systemic primary); Carnitine-acylcarnitine translocase deficiency; Carpal tunnel syndrome (familial); Cataract (cerulean; congenital; crystalline aculeiform; juvenile-onset; polymorphic and lamellar; punctate; zonular pulverulent); Cataract, Coppock-like; CD59 deficiency; Central core disease; Cerebellar ataxia; Cerebral amyloid angiopathy; Cerebral adenopathy with subcortical infarcts and leukoencephalopathy; Cerebral cavernous malformations-1; Cerebrooculofacioskeletal syndrome; Cerebrotendinous xanthomatosis; Cerebrovascular disease; Ceroid lipofuscinosis (neuronal, variant juvenile type, with granular osmiophilic deposits); Ceroid lipofuscinosis (neuronal-1, infantile); Ceroid- lipofuscinosis (neuronal-3, juvenile); Char syndrome; Charcot-Marie-Tooth disease; Charcot-Marie-Tooth neuropathy; Charlevoix-Saguenay type; Chediak-Higashi syndrome; Chloride diarrhea (Finnish type); Cholestasis (benign recurrent intrahepatic); Cholestasis (familial intrahepatic); Cholestasis (progressive familial intrahepatic); Cholesteryl ester storage disease; Chondrodysplasia punctata (brachytelephalangic; rhizomelic; X-linked dominant; X-linked recessive; Grebe type); Chondrosarcoma; Choroideremia; Chronic granulomatous disease (autosomal, due to deficiency of CYBA); Chronic granulomatous disease (X-linked); Chronic granulomatous disease due to deficiency of NCF-1; Chronic granulomatous disease due to deficiency of NCF-2; Chylomicronemia syndrome, familial; Citrullinemia; classical Cockayne syndrome-1 ; Cleft lip, cleft jaw, cleft palate; Cleft lip/palate ectodermal dysplasia syndrome; Cleidocranial dysplasia; CMO Il deficiency; Coats disease; Cockayne syndrome-2, type B; Coffin-Lowry syndrome; Colchicine resistance; Colon adenocarcinoma; Colon cancer; Colorblindness (deutan; protan; tritan); Colorectal cancer; Combined factor V and VIII deficiency; Combined hyperlipemia (familial); Combined immunodeficiency (X-linked, moderate); Complex I deficiency; Complex neurologic disorder; Cone dystrophy-3; Cone-rod dystrophy 3; Cone-rod dystrophy 6; Cone-rod retinal dystrophy-2; Congenital bilateral absence of vas deferens; Conjunctivitis, ligneous; Contractural arachnodactyly; Coproporphyria; Cornea plana congenita; Corneal clouding; Corneal dystrophy (Avellino type; gelatinous drop-like; Groenouw type I; lattice type I; Reis-Bucklers type); Cortisol resistance; Coumarin resistance; Cowden disease; CPT deficiency, hepatic (type I; type II); Cramps (familial, potassium-aggravated); Craniofacial-deafness-hand syndrome; Craniosynostosis (type 2); Cretinism; Creutzfeldt-Jakob disease ; Crigler-Najjar syndrome; Crouzon syndrome; Currarino syndrome; Cutis laxa; Cyclic hematopoiesis; Cyclic ichthyosis; Cylindromatosis; Cystic fibrosis; Cystinosis (nephropathic); Cystinuria (type II; type III); Daltonism; Darier disease; D-bifunctional protein deficiency; Deafness, autosomal dominant 1 ; Deafness, autosomal dominant 1 1 ; Deafness, autosomal dominant 12; Deafness, autosomal dominant 15; Deafness, autosomal dominant 2; Deafness, autosomal dominant 3; Deafness, autosomal dominant 5; Deafness, autosomal dominant 8; Deafness, autosomal dominant 9; Deafness, autosomal recessive 1 ; Deafness, autosomal recessive 2; Deafness, autosomal recessive 21; Deafness, autosomal recessive 3; Deafness, autosomal recessive 4; Deafness, autosomal recessive 9; Deafness, nonsyndromic sensorineural 13; Deafness, X-linked 1; Deafness, X-linked 3; Debrisoquine sensitivity; Dejerine-Sottas disease; Dementia (familial Danish); Dementia (frontotemporal, with parkinsonism); Dent disease; Dental anomalies; Dentatorubro-pallidoluysian atrophy; Denys-Drash syndrome; Dermatofibrosarcoma protuberans; Desmoid disease; Diabetes insipidus (nephrogenic ); Diabetes insipidus (neurohypophyseal); Diabetes mellitus (insulin-resistant); Diabetes mellitus (rare form); Diabetes mellitus (type II); Diastrophic dysplasia; Dihydropyrimidinuria; Dosage-sensitive sex reversal; Doyne honeycomb degeneration of retina; Dubin-Johnson syndrome; Duchenne muscular dystrophy; Dyserythropoietic anemia with thrombocytopenia; Dysfibrinogenemia (alpha type; beta type; gamma type); Dyskeratosis congenita-1; Dysprothrombinemia; Dystonia (DOPAresponsive); Dystonia (myoclonic); Dystonia-1 (torsion); Ectodermal dysplasia; Ectopia lentis; Ectopia pupillae; Ectrodactyly (ectodermal dysplasia, and cleft lip/palate syndrome 3); Ehlers-Danlos syndrome (progeroid form); Ehlers-Danlos syndrome (type I; type II; type III; type IV; type Vl; type VII); Elastin Supravalvar aortic stenosis; Elliptocytosis-1 ; Elliptocytosis-2; Elliptocytosis-3; Ellis-van Creveld syndrome; Emery-Dreifuss muscular dystrophy; Emphysema; Encephalopathy; Endocardial fibroelastosis-2; Endometrial carcinoma; Endplate acetylcholinesterase deficiency; Enhanced S-cone syndrome; Enlarged vestibular aqueduct; Epidermolysis bullosa; Epidermolysis bullosa dystrophica (dominant or recessive); Epidermolysis bullosa simplex; Epidermolytic hyperkeratosis; Epidermolytic palmopla ar keratoderma; Epilepsy (generalize; juvenile; myoclonic; nocturnal frontal lobe; progressive myoclonic); Epilepsy, benign, neonatal (typei or type2); Epiphyseal dysplasia (multiple); Episodic ataxia (type 2); Episodic ataxia/myokymia syndrome; Erythremias (alpha-; dysplasia); Erythrocytes is; Erythrokeratoderma; Estrogen resistance; Exertional myoglobinuria due to deficiency of LDH-A; Exostoses, multiple (type 1 ; type 2); Exudative vitreoretinopathy, X-linked; Fabry disease; Factor H deficiency; Factor VII deficiency; Factor X deficiency; Factor Xl deficiency; Factor XII deficiency; Factor XIIIA deficiency; Factor XIIIB deficiency; Familial Mediterranean fever; Fanconi anemia; Fanconi-Bickel syndrome; Farber lipogranulomatosis; Fatty liver (acute); Favism; Fish-eye disease; Foveal hypoplasia; Fragile X syndrome; Frasier syndrome; Friedreich ataxia; fructose-bisphosphatase Fructose intolerance; Fucosidosis; Fumarase deficiency; Fundus albipunctatus; Fundus flavimaculatus; G6PD deficiency; GABA-transaminase deficiency; Galactokinase deficiency with cataracts; Galactose epimerase deficiency; Galactosemia; Galactosialidosis; GAMT deficiency; Gardner syndrome; Gastric cancer; Gaucher disease; Generalized epilepsy with febrile seizures plus; Germ cell tumors; Gerstmann-Straussler disease; Giant cell hepatitis (neonatal); Giant platelet disorder; Giant-cell fibroblastoma; Gitelman syndrome; Glanzmann thrombasthenia (type A; type B); Glaucoma 1 A; Glaucoma 3A; Glioblastoma multiforme; Glomerulosclerosis (focal segmental); Glucose transport defect (blood-brain barrier); Glucose/galactose malabsorption; Glucosidase I deficiency; Glutaricaciduria (type I; type MB; type HC); Gluthation synthetase deficiency; Glycerol kinase deficiency; Glycine receptor (alpha-1 polypeptide); Glycogen storage disease I; Glycogen storage disease II; Glycogen storage disease III; Glycogen storage disease IV; Glycogen storage disease Vl; Glycogen storage disease VII; Glycogenosis (hepatic, autosomal); Glycogenosis (X-linked hepatic); GM1 -gangliosidosis; GM2 -gangliosidosis; Goiter (adolescent multinodular); Goiter (congenital); Goiter (nonendemic, simple); Gonadal dysgenesis (XY type); Granulomatosis, septic; Graves disease; Greig cephalopolysyndactyly syndrome; Griscelli syndrome; Growth hormone deficient dwarfism; Growth retardation with deafness and mental retardation; Gynecomastia (familial, due to increased aromatase activity); Gyrate atrophy of choroid and retina with ornithinemia (B6 responsive or unresponsive); Hailey-Hailey disease; Haim- Munk syndrome; Hand-foot-uterus syndrome; Harderoporphyrinuria; HDL deficiency (familial); Heart block (nonprogressive or progressive); Heinz body anemia; HELLP syndrome; Hematuria (familial benign); Heme oxygenase-1 deficiency; Hemiplegic migraine; Hemochromotosis; Hemoglobin H disease; Hemolytic anemia due to ADA excess; Hemolytic anemia due to adenylate kinase deficiency; Hemolytic anemia due to band 3 defect; Hemolytic anemia due to glucosephosphate isomerase deficiency; Hemolytic anemia due to glutathione synthetase deficiency; Hemolytic anemia due to hexokinase deficiency; Hemolytic anemia due to PGK deficiency; Hemolytic-uremic syndrome; Hemophagocytic lymphohistiocytosis; Hemophilia A; Hemophilia B; Hemorrhagic diathesis due to factor V deficiency; Hemosiderosis (systemic, due to aceruloplasminemia); Hepatic lipase deficiency; Hepatoblastoma; Hepatocellular carcinoma; Hereditary hemorrhagic telangiectasia-1 ; Hereditary hemorrhagic telangiectasia- 2; Hermansky-Pudlak syndrome; Heterotaxy (X-linked visceral); Heterotopia (periventricular); Hippel-Lindau syndrom; Hirschsprung disease; Histidine-rich glycoprotein Thrombophilia due to HRG deficiency; HMG-CoA lyase deficiency; Holoprosencephaly-2; Holoprosencephaly-3; Holoprosencephaly-4; Holoprosencephaly-5; Holt-Oram syndrome; Homocystinuria; Hoyeraal-Hreidarsson; HPFH (deletion type or nondeletion type); HPRT- related gout; Huntington disease; Hydrocephalus due to aqueductal stenosis; Hydrops fetalis; Hyperbetalipoproteinemia; Hypercholesterolemia, familial; Hyperferritinemia- cataract syndrome; Hyperglycerolemia; Hyperglycinemia; Hyperimmunoglobulinemia D and periodic fever syndrome; Hyperinsulinism; Hyperinsulinism-hyperammonemia syndrome; Hyperkalemic periodic paralysis; Hyperlipoproteinemia; Hyperlysinemia; Hypermethioninemia (persistent, autosomal, dominant, due to methionine, adenosyltransferase I/Ill deficiency); Hyperornithinemia- hyperammonemiahomocitrullinemia syndrome; Hyperoxaluria; Hyperparathyroidism; Hyperphenylalaninemia due to pterin-4acarbinolamine dehydratase deficiency; Hyperproinsulinemia; Hyperprolinemia; Hypertension; Hyperthroidism (congenital); Hypertriglyceridemia; Hypoalphalipoproteinemia; Hypobetalipoproteinemia;
Hypocalcemia; Hypochondroplasia; Hypochromic microcytic anemia; Hypodontia; Hypofibrinogenemia; Hypoglobulinemia and absent B cells; Hypogonadism (hypergonadotropic); Hypogonadotropic (hypogonadism); Hypokalemic periodic paralysis; Hypomagnesemia; Hypomyelination (congenital); Hypoparathyroidism; Hypophosphatasia (adult; childhood; infantile; hereditary); Hypoprothrombinemia; Hypothyroidism (congenital; hereditary congenital; nongoitrous); lchthyosiform erythroderma ; Ichthyosis ; Ichthyosis bullosa of Siemens ; lgG2 deficiency; lmmotile cilia syndrome-1; Immunodeficiency (T-cell receptor/CD3 complex); Immunodeficiency (X-linked, with hyper-lgM); Immunodeficiency due to defect in CD3-gamma; Immunodeficiency- centromeric instabilityfacial anomalies syndrome; Incontinentia pigmenti; Insensitivity to pain (congenital, with anhidrosis); Insomnia (fatal familial); lnterleukin-2 receptor deficiency (alpha chain); Intervertebral disc disease; Iridogoniodysgenesis; Isolated growth hormone deficiency (lllig type with absent GH and Kowarski type with bioinactive GH); Isovalericacidemia ; Jackson -Weiss sydnrome; Jensen syndrome; Jervell and Lange-Nielsen syndrome; Joubert syndrom; Juberg-Marsidi syndrome; Kallmann syndrome; Kanzaki disease; Keratitis; Keratoderma (palmoplanar); Keratosis palmoplantaris striata I; Keratosis palmoplantaris striata II; Ketoacidosis due to SCOT deficiency; Keutel syndrome; Klippel- Trenaurnay syndrom; Kniest dysplasia; Kostmann neutropenia; Krabbe disease; Kurzripp- Polydaktylie syndrom; Lacticacidemia due to PDXl deficiency; Langer mesomelic dysplasia; Laron dwarfism; Laurence-Moon-Biedl-Bardet syndrom; LCHAD deficiency; Leber congenital amaurosis; Left-right axis malformation; Leigh syndrome; Leiomyomatosis (diffuse, with Alport syndrome); Leprechaunism; Leri- Weill dyschondrosteosis; Lesch- Nyhan syndrome; Leukemia (acute myeloid; acute promyelocytic; acute T-cell lymphoblastic; chronic myeloid; juvenile myelomonocytic; Leukemia-1 (T-cell acute lymphocytic); Leukocyte adhesion deficiency; Leydig cell adenoma; Lhermitte-Duclos syndrome; Liddle syndrome; Li-Fraumeni syndrome; Lipoamide dehydrogenase deficiency; Lipodystrophy; Lipoid adrenal hyperplasia; Lipoprotein lipase deficiency; Lissencephaly (X- linked); Lissencephaly-1; liver Glycogen storage disease (type 0); Long QT syndrome-1 ; Long QT syndrome-2; Long QT syndrome-3; Long QT syndrome-5; Long QT syndrome-6; Lowe syndrome; Lung cancer; Lung cancer (nonsmall cell); Lung cancer (small cell); Lymphedema; Lymphoma (B-cell non-Hodgkin); Lymphoma (diffuse large cell); Lymphoma (follicular); Lymphoma (MALT); Lymphoma (mantel cell); Lymphoproliferative syndrome (X- linked); Lysinuric protein intolerance; Machado-Joseph disease; Macrocytic anemia refractory (of 5q syndrome); Macular dystrophy; Malignant mesothelioma; Malonyl-CoA decarboxylase deficiency ; Mannosidosis, (alpha- or beta- ); Maple syrup urine disease (type Ia; type Ib; type II); Marfan syndrome; Maroteaux-Lamy syndrome; Marshall syndrome; MASA syndrome; Mast cell leukemia; Mastocytosis with associated hematologic disorder; McArdle disease; McCune-Albright polyostotic fibrous dysplasia; McKusick-Kaufman syndrome; McLeod phenotype ; Medullary thyroid carcinoma; Medulloblastoma; Meesmann corneal dystrophy; Megaloblastic anemia-1 ; Melanoma; Membroproliferative glomerulonephritis ; Meniere disease; Meningioma (NF2-related; SIS-related); Menkes disease; Mental retardation (X-linked); Mephenytoin poor metabolizer; Mesothelioma; Metachromatic leukodystrophy; Metaphyseal chondrodysplasia (Murk Jansen type; Schmid type); Methemoglobinemia; Methionine adenosyltransferase deficiency (autosomal recessive); Methylcobalamin deficiency (cbl G type); Methylmalonicaciduria (mutase deficiency type); Mevalonicaciduria; MHC class Il deficiency; Microphthalmia (cataracts, and iris abnormalities); Miyoshi myopathy; MODY; Mohr-Tranebjaerg syndrome; Molybdenum cofactor deficiency (type A or type B); Monilethrix; Morbus Fabry; Morbus Gaucher; Mucopolysaccharidosis; Mucoviscidosis; Muencke syndrome; Muir-Torre syndrome; Mulibrey nanism; Multiple carboxylase deficiency (biotinresponsive); Multiple endocrine neoplasia; Muscle glycogenosis; Muscular dystrophy (congenital merosindeficient); Muscular dystrophy (Fukuyama congenital); Muscular dystrophy (limb- girdle); Muscular dystrophy )Duchenne-like); Muscular dystrophy with epidermolysis bullosa simplex; Myasthenic syndrome (slow-channel congenital); Mycobacterial infection (atypical, familial disseminated); Myelodysplastic syndrome; Myelogenous leukemia; Myeloid malignancy; Myeloperoxidase deficiency; Myoadenylate deaminase deficiency; Myoglobinuria/hemolysis due to PGK deficiency; Myoneurogastrointestinal encephalomyopathy syndrome; Myopathy (actin; congenital; desmin-related; cardioskeletal; distal; nemaline); Myopathy due to CPT Il deficiency; Myopathy due to phosphoglycerate mutase deficiency; Myotonia congenita; Myotonia levior; Myotonic dystrophy; Myxoid liposarcoma; NAGA deficiency; Nailpatella syndrome; Nemaline myopathy 1 (autosomal dominant); Nemaline myopathy 2 (autosomal recessive); Neonatal hyperparathyroidism; Nephrolithiasis; Nephronophthisis (juvenile); Nephropathy (chronic hypocomplementemic); Nephrosis-1; Nephrotic syndrome; Netherton syndrome; Neuroblastoma; Neurofibromatosis (type 1 or type 2); Neurolemmomatosis; neuronal-5 Ceroid-lipofuscinosis; Neuropathy; Neutropenia (alloimmune neonatal); Niemann-Pick disease (type A; type B; type CI ; type D); Night blindness (congenital stationary); Nijmegen breakage syndrome; Noncompaction of left ventricular myocardium; Nonepidermolytic palmoplanar keratoderma; Norrie disease; Norum disease; Nucleoside phosphorylase deficiency; Obesity; Occipital hornsyndrome; Ocular albinism (Nettleship-Falls type); Oculopharyngeal muscular dystorphy; Oguchi disease; Oligodontia; Omenn syndrome; Opitz G syndrome; Optic nerve coloboma with renal disease; Ornithine transcarbamylase deficiency; Oroticaciduria; Orthostatic intolerance; OSMED syndrome; Ossification of posterior longitudinal ligament of spine; Osteoarthrosis; Osteogenesis imperfecta; Osteolysis; Osteopetrosis (recessive or idiopathic); Osteosarcoma; Ovarian carcinoma; Ovarian dysgenesis; Pachyonychia congenita Qackson-Lawler type or Jadassohn-Lewandowsky type); Paget disease of bone; Pallister-Hall syndrome; Pancreatic agenesis; Pancreatic cancer; Pancreatitis; Papillon- Lefevre syndrome; Paragangliomas ; Paramyotonia congenita; Parietal foramina; Parkinson disease (familial or juvenile); Paroxysmal nocturnal hemoglobinuria; Pelizaeus-Merzbacher disease; Pendred syndrome; Perineal hypospadias; Periodic fever; Peroxisomal biogenesis disorder; Persistent hyperinsulinemic hypoglycemia of infancy; Persistent Mullerian duct syndrome (type II); Peters anomaly; Peutz-Jeghers syndrome ; Pfeiffer syndrome; Phenylketonuria; Phosphoribosyl pyrophosphate synthetaserelated gout; Phosphorylase kinase deficiency of liver and muscle; Piebaldism; Pilomatricoma; Pinealoma with bilateral retinoblastoma; Pituitary ACTH secreting adenoma; Pituitary hormone deficiency; Pituitary tumor; Placental steroid sulfatase deficiency; Plasmin inhibitor deficiency; Plasminogen deficiency (types I and II); Plasminogen Tochigi disease; Platelet disorder; Platelet glycoprotein IV deficiency; Platelet-activating factor acetylhydrolase deficiency; Polycystic kidney disease; Polycystic lipomembranous osteodysplasia with sclerosing leukenencephalophathy; Polydactyly, postaxial; Polyposis; Popliteal pterygium syndrome; Porphyria (acute hepatic or acute intermittent or congenital erythropoietic); Porphyria cutanea tarda; Porphyria hepatoerythropoietic ; Porphyria variegata; Prader-Willi syndrome; Precocious puberty; Premature ovarian failure; Progeria Typ I; Progeria Typ II; Progressive external ophthalmoplegia; Progressive intrahepatic cholestasis-2; Prolactinoma (hyperparathyroidism, carcinoid syndrome); Prolidase deficiency; Propionicacidemia; Prostate cancer; Protein S deficiency; Proteinuria; Protoporphyria (erythropoietic); Pseudoachondroplasia; Pseudohermaphroditism; Pseudohypoaldosteronism;
Pseudohypoparathyroidism; Pseudovaginal perineoscrotal hypospadias; Pseudovitamin D deficiency rickets; Pseudoxanthoma elasticum (autosomal dominant; autosomal recessive); Pulmonary alveolar proteinosis; Pulmonary hypertension; Purpura fulminans; Pycnodysostosis; Pyropoikilocytosis; Pyruvate carboxylase deficiency; Pyruvate dehydrogenase deficiency; Rabson-Mendenhall syndrome; Refsum disease; Renal cell carcinoma; Renal tubular acidosis; Renal tubular acidosis with deafness; Renal tubular acidosis-osteopetrosis syndrome; Reticulosis (familial histiocytic); Retinal degeneration; Retinal dystrophy; Retinitis pigmentosa; Retinitis punctata albescens; Retinoblastoma; Retinol binding protein deficiency; Retinoschisis; Rett syndrome; Rh(mod) syndrome; Rhabdoid predisposition syndrome; Rhabdoid tumors ; Rhabdomyosarcoma; Rhabdomyosarcoma (alveolar); Rhizomelic chondrodysplasia punctata; Ribbing-Syndrom; Rickets (vitamin D-resistant); Rieger anomaly; Robinow syndrome; Rothmund-Thomson syndrome; Rubenstein-Taybi syndrome; Saccharopinuria; Saethre-Chotzen syndrome; SaIIa disease; Sandhoff disease (infantile, juvenile, and adult forms); Sanfilippo syndrome (type A or type B); Schindler disease; Schizencephaly; Schizophrenia (chronic); Schwannoma (sporadic); SCID (autosomal recessive, T-negative/Bpositive type); Secretory pathway w/TMD; SED congenita; Segawa syndrome; Selective T-cell defect; SEMD (Pakistani type); SEMD (Strudwick type); Septooptic dysplasia; Severe combined immunodeficiency (B cellnegative); Severe combined immunodeficiency (T-cell negative, B-cel I/natural killer cell- positive type); Severe combined immunodeficiency (Xlinked); Severe combined immunodeficiency due to ADA deficiency; Sex reversal (XY, with adrenal failure); Sezary syndrome; Shah-Waardenburg syndrome; Short stature; Shprintzen-Goldberg syndrome; Sialic acid storage disorder; Sialidosis (type I or type II); Sialuria; Sickle cell anemia; Simpson-Golabi-Behmel syndrome; Situs ambiguus; Sjogren-Larsson syndrome; Smith- Fineman-Myers syndrome; Smith-Lemli-Opitz syndrome (type I or type II); Somatotroph! noma; Sorsby fundus dystrophy; Spastic paraplegia; Spherocytosis; Spherocytosis-1; Spherocytosis-2; Spinal and bulbar muscular atrophy of Kennedy; Spinal muscular atrophy; Spinocerebellar ataxia; Spondylocostal dysostosis; Spondyloepiphyseal dysplasia tarda; Spondylometaphyseal dysplasia (Japanese type); Stargardt disease-1 ; Steatocystoma multiplex; Stickler syndrome; Sturge-Weber syndrom; Subcortical laminal heteropia; Subcortical laminar heterotopia; Succinic semialdehyde dehydrogenase deficiency; Sucrose intolerance; Sutherland-Haan syndrome; Sweat chloride elevation without CF; Symphalangism; Synostoses syndrome; Synpolydactyly; Tangier disease; Tay- Sachs disease; T-cell acute lymphoblastic leukemia; T-cell immunodeficiency; T-cell prolymphocyte leukemia; Thalassemia (alpha- or delta-); Thalassemia due to Hb Lepore; Thanatophoric dysplasia (types I or II); Thiamine-responsive megaloblastic anemia syndrome; Thrombocythemia; Thrombophilia (dysplasminogenemic); Thrombophilia due to heparin cofactor Il deficiency; Thrombophilia due to protein C deficiency; Thrombophilia due to thrombomodulin defect; Thyroid adenoma; Thyroid hormone resistance; Thyroid iodine peroxidase deficiency; Tietz syndrome; Tolbutamide poor metabolizer; Townes- Brocks syndrome; Transcobalamin Il deficiency; Treacher Collins mandibulofacial dysostosis; Trichodontoosseous syndrome; Trichorhinophalangeal syndrome; Trichothiodystrophy; Trifunctional protein deficiency (type I or type II); Trypsinogen deficiency; Tuberous sclerosis-1; Tuberous sclerosis-2; Turcot syndrome; Tyrosine phosphatase; Tyrosinemia; Ulnar-mammary syndrome; Urolithiasis (2,8-dihydroxyadenine); Usher syndrome (type I B or type 2A); Venous malformations; Ventricular tachycardia; Virilization; Vitamin K-dependent coagulation defect; VLCAD deficiency; Vohwinkel syndrome; von Hippel-Lindau syndrome; von Willebrand disease; Waardenburg syndrome; Waardenburg syndrome/ocular albinism; Waardenburg-Shah neurologic variant; Waardenburg-Shah syndrome; Wagner syndrome; Warfarin sensitivity; Watson syndrome; Weissenbacher-Zweymuller syndrome; Werner syndrome; Weyers acrodental dysostosis; White sponge nevus; Williams-Beuren syndrome; Wilms tumor (typei ); Wilson disease; Wiskott-Aldrich syndrome; Wolcott-Rallison syndrome; Wolfram syndrome; Wolman disease; Xanthinuria (type I); Xeroderma pigmentosum; X-SCID; Yemenite deaf-blind hypopigmentation syndrome; ypocalciuric hypercalcemia (type I); Zellweger syndrome; Zlotogora-Ogur syndrome.
Preferred diseases to be treated which have a genetic inherited background and which are typically caused by a single gene defect and are inherited according to Mendel's laws are preferably selected from the group consisting of autosomal-recessive inherited diseases, such as, for example, adenosine deaminase deficiency, familial hypercholesterolemia, Canavan's syndrome, Gaucher's disease, Fanconi anaemia, neuronal ceroid lipofuscinoses, mucoviscidosis (cystic fibrosis), sickle cell anaemia, phenylketonuria, alcaptonuria, albinism, hypothyreosis, galactosaemia, alpha-1 -anti-trypsin deficiency, Xeroderma pigmentosum, Ribbing's syndrome, mucopolysaccharidoses, cleft lip, jaw, palate, Laurence Moon Biedl Bardet sydrome, short rib polydactylia syndrome, cretinism, Joubert's syndrome, type Il progeria, brachydactylia, adrenogenital syndrome, and X-chromosome inherited diseases, such as, for example, colour blindness, e.g. red/green blindness, fragile X syndrome, muscular dystrophy (Duchenne and Becker-Kiener type), haemophilia A and B, G6PD deficiency, Fabry's disease, mucopolysaccharidosis, Nome's syndrome, Retinitis pigmentosa, septic granulomatosis, X-SCID, ornithine transcarbamylase deficiency, Lesch- Nyhan syndrome, or from autosomal-dominant inherited diseases, such as, for example, hereditary angiooedema, Marfan syndrome, neurofibromatosis, type I progeria, Osteogenesis imperfecta, Klippel-Trenaumay syndrome, Sturge-Weber syndrome, Hippel- Lindau syndrome and tuberosis sclerosis.
The present invention also allows treatment of diseases, which have not been inherited, or which may not be summarized under the above categories. Such dieseases may include e.g. the treatment of patients, which are in need of a specific protein factor, e.g. a specific therapeutically active protein as mentioned above. This may e.g. include dialysis patients, e.g. patients which undergo a (regular) a kidney or renal dialysis, and which may be in need of specific therapeutically active proteins as defined above, e.g. EPO, etc..
In the context of the above, the invention furthermore relates also to the use of a modified (m)RNA as described herein, or of an inventive immunosuppressive composition or of a pharmaceutical composition or of a vaccine as described herein, for the prophylaxis, treatment, and/or amelioration of diseases or disorders as mentioned above. It also includes in particular the use of the modified (m)RNA as described herein, or of an inventive immunosuppressive composition or of a pharmaceutical composition or of a vaccine described herein for inoculation or the use of these components as an inoculant. According to one particularly preferred embodiment of the present invention, such a method for prophylaxis, treatment, and/or amelioration of the above-mentioned diseases or disorders, or an inoculation method for preventing the above-mentioned diseases, typically comprises administering the described pharmaceutical composition to a patient in need thereof (e.g. suffering from any of the above diseases or showing symptoms thereof), in particular to a human being, preferably in a "safe and effective amount" and in one of the above formulations as described above for inventive pharmaceutical compositions. The administration mode also may be as described above for inventive pharmaceutical compositions.
The present invention relates also to an in vitro transcription method for the preparation of inventive modified (m)RNA (of the inventive immunosuppressive composition), comprising the following steps: a) preparation/provision of a (desoxy)ribonucleic acid as a template for the inventive modified (m)RNA (of the inventive immunosuppressive composition), in particular a template as described above; b) addition of the (desoxy)ribonucleic acid to an in vitro transcription medium comprising a RNA polymerase, a suitable buffer, a nucleic acid mix, comprising one or more chemically modified nucleosides selected from the chemically modified nucleosides as defined above as replacement (partially or completely) for one or more of the naturally occurring nucleosides A, G, U and/or C, and optionally one or more naturally occurring nucleosides A, G, U and/or C if not all of the naturally occurring nucleosides A, G, U and/or C are to be replaced, and optionally an RNase inhibitor; c) incubation of the (desoxy)ribonucleic acid in the in vitro transcription medium and in vitro transcription of the (desoxy)ribonucleic acid; d) optionally purification and removal of the unincorporated nucleotides from the in vitro transcription medium.
A (desoxy)ribonucleic acid as described in step a) of the in vitro transcription method according to the invention can be any nucleic acid as described above that may be used as a template for the preparation of the modified (m)RNA of the present invention. For this purpose typically DNA sequences are used, for example genomic DNA or fragments thereof, or plasmids, or RNA sequences (corresponding thereto), for example mRNA sequences, preferably in linearized form. The in vitro transcription reaction can usually be carried out using a vector having a RNA polymerase binding site. To this end there can be used any vectors known in the art, for example commercially available vectors (see above). Preference is given, for example, to those vectors that have a SP6 or a 17 or T3 binding site upstream and/or downstream of the cloning site. Accordingly, the (desoxy)ribonucleic acid sequences used can be transcribed later, as desired, depending on the chosen RNA polymerase. A (desoxy)ribonucleic acid sequence used for in vitro transcription and coding for a (therapeutically active) protein, antigen or antibody as defined above is typically cloned into a vector, for example via a multiple cloning site of the vector used. Before the transcription, the clone is typically cleaved with restriction enzymes at the site at which the future 3' end of the modified (m)RNA is to be located, using a suitable restriction enzyme, and the fragment is purified. This prevents the future modified (m)RNA from containing vector sequences, and a modified (m)RNA of defined length may be obtained.
Alternatively, it is also possible to prepare the (desoxy)ribonucleic acid as transcription template by polymerase chain reaction (PCR). To this end, one of the primers used typically contains the sequence of a RNA polymerase binding site. It is further preferred for the 51 end of the primer used to have a length of approximately from 10 to 50 further nucleotides, more preferably from 15 to 30 further nucleotides and most preferably of approximately 20 nucleotides. Prior to the in vitro transcription, the (desoxy)ribonucleic acid, e.g. the specific DNA or RNA template, is typically purified and free of RNase in order to ensure a high yield. Purification can be carried out by any process known in the art, for example with a caesium chloride gradient or ion-exchange process.
According to method step b), the (desoxy)ribonucleic acid is added to an in vitro transcription medium. A suitable in vitro transcription medium first contains a (desoxy)ribonucleic acid as prepared under step a), for example approximately from 0.1 to 10 μg, preferably approximately from 1 to 5 μg, more preferably 2.5 μg and most preferably approximately 1 μg, of such a nucleic acid. A suitable in vitro transcription medium further optionally contains a reducing agent, e.g. DTT, more preferably approximately from 1 to 20 μl of 50 mM DTT, yet more preferably approximately 5 μl of 50 mM DTT. The in vitro transcription medium further contains nucleotides (AMP, GMP, UMP and/or CMP), for example a nucleotide mix. In the case of the present invention the nucleotides preferably comprise chemically modified nucleosides as defined above. Such (chemically modified) nucleotides may serve as replacement for one or more of the naturally occurring nucleotides AMP, GMP, UMP and/or CMP, and optionally one or more naturally occurring nucleotides AMP, GMP, UMP and/or CMP, if not all of the naturally occurring nucleotides AMP, GMP, UMP and/or CMP are to be replaced. The nucleotides AMP, GMP, UMP and/or CMP are typically present in the nucleotide mix in a concentration of typically approximately from 0.1 to 1 O mM per nucleotide, preferably from 0.1 to 1 mM per nucleotide, preferably approximately 4 mM in total. Modified nucleotides as described above (approximately 1 mM per nucleotide, preferably approximately 4 mM in total), are typically added in such an amount that the native nucleotide is replaced completely by the (modified) nucleotide(s) comprising a chemically modified nucleoside as defined above. It is, however, also possible to use mixtures of one or more modified nucleotides as defined above and one or more naturally occurring nucleotides instead of a particular nucleotide, that is to say one or more chemically modified nucleotides as described above can occur as a replacement for one or more of the naturally occurring nucleotides AMP, GMP, UMP and/or CMP and optionally additionally one or more naturally occurring nucleotides AMP, GMP, UMP and/or CMP, if not all the naturally occurring nucleotides AMP, GMP, UMP and/or CMP are to be replaced. By selective addition of the desired base to the in vitro transcription medium, the content, that is to say the occurrence and amount, of the desired nucleotide modification in the transcribed modified RNA sequence can therefore be controlled. A suitable in vitro transcription medium likewise contains a RNA polymerase, e.g. T7-RNA polymerase (e.g. T7-Opti mRNA Kit, Cure Vac, Tubingen, Germany), T3-RNA polymerase or SP6, typically approximately from 10 to 500 U, preferably approximately from 25 to 250 U, more preferably approximately from 50 to 150 U, and most preferably approximately 100 U of RNA polymerase. The in vitro transcription medium is further preferably kept free of RNase in order to avoid degradation of the transcribed RNA. A suitable in vitro transcription medium therefore optionally contains in addition a RNase inhibitor.
In a step c), the (desoxy)ribonucleic acid is incubated and transcribed in the in vitro transcription medium, typically for approximately from 30 to 120 minutes, preferably for approximately from 40 to 90 minutes and most preferably for approximately 60 minutes, at approximately from 30 to 45°C, preferably at from 37 to 42°C. The incubation temperature is governed by the RNA polymerase that is used, for example in the case of T7 RNA polymerase it is approximately 37°C. The nucleic acid obtained by the transcription is preferably a modified (m)RNA as defined herein for the inventive immunosuppressive composition, more preferably an mRNA.
After the incubation, purification of the reaction can optionally take place in step d) of the in vitro transcription method according to the invention. To this end, any suitable process known in the art can be used, for example chromatographic purification processes, e.g. affinity chromatography, gel filtration, etc. By means of the purification, non-incorporated, i.e. excess, nucleotides can be removed from the in vitro transcription medium. Any suitable method known in the prior art, e.g. chromatographic purification methods, e.g. affinity chromatography, gel filtration etc., can be used for this. By the purification, non- incorporated, i.e. excess nucleotides and template DNA can be removed from the in vitro transcription medium and a clean modified (m)RNA can be obtained. For example, after the transcription the reaction mixture containing the transcribed RNA can typically be digested with DNase in order to remove the DNA template still contained in the reaction mixture. The transcribed RNA can be subsequently or alternatively precipitated with LiCI. Purification of the transcribed modified (m)RNA can then take place via IP RP-HPLC. This renders it possible in particular to separate longer and shorter fragments from one another effectively. Preferably, in this context the purification takes place via a method for purification of RNA on a preparative scale, which is distinguished in that the RNA is purified by means of HPLC using a porous reverse phase as the stationary phase (PURE Messenger). For example, for the purification in step d) of the inventive in vitro transcription method, a reverse phase can be employed as the stationary phase for the HPLC purification. For the chromatography with reverse phases, a non-polar compound typically serves as stationary phase, and a polar solvent, such as mixtures of water, which is usually employed in the form of buffers, with acetonitrile and/or methanol, serves as the mobile phase for the elution. Preferably, the porous reverse phase has a particle size of 8.0 ± 2 μm, preferably ±1 μm, more preferably +/- 0.5 μm. The reverse phase material can be in the form of beads. The purification can be carried out in a particularly favourable manner with a porous reverse phase having this particle size, optionally in the form of beads, particularly good separation results being obtained. The reverse phase employed is preferably porous since with stationary reverse phases which are not porous, such as are described e.g. by Azarani A. and Hecker K.H., pressures which are too high are built up, so that preparative purification of the modified (m)RNA is possible, if at all, only with great difficulty. The reverse phase preferably has a pore size of from 200 to 5,000 , in particular a pore size of from 300 to 4,000 . Particularly preferred pore sizes for the reverse phases are 200 - 400 , 800 - 1,200 and 3,500 - 4,500 . With a reverse phase having these pore sizes, particularly good results are achieved in respect of the purification of the modified (m)RNA in process step d). The material for the reverse phase is preferably a polystyrene-divinylbenzene, and non-alkylated polystyrene-divinylbenzenes can be employed in particular. Stationary phases with polystyrene-divinylbenzene are known per se. For the purification in method step d), the polystyrene-divinylbenzenes which are known per se and already employed for HPLC methods and are commercially obtainable can be used. A non-alkylated porous polystyrene-divinylbenzene which in particular has a particle size of 8.0 ± 0.5 μm and a pore size of 250 - 300 , 900 - 1,100 or 3,500 - 4,500 is very particularly preferably used for the purification in method step d). The advantages described above can be achieved in a particularly favourable manner with this material for the reverse phases. The HPLC purification can be carried out by the ion pair method, an ion having a positive charge being added to the mobile phase as a counter-ion to the negatively charged RNA. An ion pair having a lipophilic character, which is slowed down by the non-polar stationary phase of the reverse phase system, is formed in this manner. In practices, the precise conditions for the ion pair method must be worked out empirically for each concrete separation problem. The size of the counter-ion, its concentration and the pH of the solution contribute greatly towards the result of the separation. In a favourable manner, alkylammonium salts, such as triethylammonium acetate and/or tetraalkylammonium compounds, such as tetrabutylammonium, are added to the mobile phase. Preferably, 0.1 M triethylammonium acetate is added and the pH is adjusted to about 7. The choice of mobile phase depends on the nature of the desired separation. This means that the mobile phase found for a specific separation, such as can be known, for example, from the prior art, cannot be transferred readily to another separation problem with adequate prospect of success. The ideal elution conditions, in particular the mobile phase used, must be determined for each separation problem by empirical experiments. A mixture of an aqueous solvent and an organic solvent can be employed as the mobile phase for elution of the modified (m)RNA by the HPLC method. In this context, it is favourable if a buffer which has, in particular, a pH of about 7, for example 6.5 - 7.5, e.g. 7.0, is used as the aqueous solvent; preferably, the buffer triethylammonium acetate is used, particularly preferably a 0.1 M triethylammonium acetate buffer which, as described above, also acts as a counter-ion to the RNA in the ion pair method. The organic solvent employed in the mobile phase can be acetonitrile, methanol or a mixture of these two, very particularly preferably acetonitrile. The purification of the modified (m)RNA in method step d) using an HPLC method as described is carried out in a particularly favourable manner with these organic solvents. The mobile phase is particularly preferably a mixture of 0.1 M triethylammonium acetate, pH 7, and acetonitrile. It has emerged to be likewise particularly favourable if the mobile phase contains 5.0 vol.% to 20.0 vol.% of organic solvent, based on the mobile phase, and the remainder to make up 100 vol.% is the aqueous solvent. It is very particularly favourable for the method according to the invention if the mobile phase contains 9.5 vol.% to 14.5 vol.% of organic solvent, based on the mobile phase, and the remainder to make up 100 vol.% is the aqueous solvent. Elution of the modified (m)RNA can subsequently be carried out isocratically or by means of a gradient separation. In the case of an isocratic separation, elution of the modified (m)RNA is carried out with a single eluting agent or a mixture of several eluting agents which remains constant, it being possible for the solvents described above in detail to be employed as the eluting agent. According to a final embodiment, the present invention also provides kits, particularly kits of parts, comprising as components the modified (m)RNA, and/or the inventive immunosuppressive composition, and/or an inventive pharmaceutical composition, e.g. an inventive vaccine, and optionally technical instructions with information on the administration and dosage of these components. Such kits, preferably kits of parts, may applied e.g. for any of the above mentioned applications or uses. Kits of parts, as a special form of kits, may be furthermore used, when a time staggered treatment is envisaged, as described above, wherein the single parts of such a kit may contain either the same or different active immunosuppressive compositions and/or inventive vaccines according to the present invention.
The following Figures are intended to illustrate the invention further. They are not intended to limit the subject matter of the invention thereto.
Figure 1 : depicts the luciferase construct (T7TS-Ppluc(wt)-A70) (SEQ ID NO: 3), which was used as a template for introducing the different chemically modified nucleic acids as defined herein in order to evaluate their immunosuppressive properties in comparison to unmodified RNA and a control (no RNA). The coding sequence is underlined. The construct furthermore contained a poly- A tail of about 70 adenosines at the 31 end.
Figure 2: depicts the immunosuppressive effect of the chemically modified (m)RNA according to the present invention. As can be seen in Figure 2, the modified
(m)RNAs modified with indicated chemically modified nucleosides, lead to an abrogation or at least significant reduction in secretion of TNF-alpha in hPBMCs, i.e. an abrogation or at least significant reduction of the immunostimulatory effect, when compared to non-modified (m)RNA.
Figure 3: depicts the immunosuppressive effect of the chemically modified (m)RNA according to the present invention. As can be seen in Figure 3, the modified (m)RNAs modified with indicated chemically modified nucleosides, lead to an abrogation or at least significant reduction in secretion of IL-6 in hPBMCs, i.e. an abrogation or at least significant reduction of the immunostimulatory effect, when compared to non-modified (m)RNA.
Examples:
The following examples are intended to illustrate the invention further. They are not intended to limit the subject matter of the invention thereto.
1. Preparation of template nucleic acid:
In the following experiment a template sequence for the preparation of the modified mRNA sequences of the inventive immunosuppressive composition was prepared. According to one particular example, the template sequences for the preparation of the modified RNA sequences are sequences comprising a coding sequence of luciferase. Starting from the native coding sequence of luciferase, the coding sequence was transferred into an RNActive construct (CureVac GmbH, Tubingen, Germany), which has been modified with a poly-A-tag, (A70). As a result, the luciferase construct (T7TS-Ppluc(wt)-A70) (SEQ ID NO: 3) was obtained as a template for the inventive modifications (see Fig. 1 ).
2. In vitro transcription
For the introduction of chemically modified nucleosides as defined above into the template as prepared according to Example 1 , the luciferase construct (T7TS- Ppluc(wt)-A70) (SEQ ID NO: 3) (see Fig. 1 ) was linearized and subsequently in vitro transcribed by means of T7 polymerase (T7-Opti mRNA Kit, CureVac, Tubingen,
Germany). To this end, nucleotides comprising chemically modified nucleosides (2- Aminopurine-riboside-5'-(mono)phosphate; 2-Thiouridine-5'-(mono)phosphate ; 4- Thiouridine-5'-(mono)phosphate; 5-Aminoallylcytidine-5'-(mono)phosphate ; 5- Aminoallyluridine-5'-(mono)phosphate ; 5-Bromocytidine-5'-(mono)phosphate ; 5- Bromo-2'-deoxycytidine-5'-(mono)phosphate ; 5-Bromouridine-5'-(mono)phosphate;
5-Bromo-2'-deoxyuridine-5'-(mono)phosphate ; 5-lodocytidine-5'-(mono)phosphate ; 5-lodo-2'-deoxycytidine-5'-(mono)phosphate ; 5-lodouridine-5'-(mono)phosphate; 5-lodo-2'-deoxyuridine-5'-(mono)phosphate ; 5-Methylcytidine-5'-(mono)phosphate ; 5-Methyluridine-5'-(mono)phosphate ; 5-Methyluridine-5'-(mono)phosphate ; 5- Propynyl-2'-deoxycytidine-5'-(mono)phosphate ; 5-Propynyl-2'-deoxyuridine-5'- (mono)phosphate; 6-Azacytidine-5'-(mono)phosphate ; 7-Deazaadenosine-5'- (mono)phosphate ; 7-Deazaguanosine-5'-(mono)phosphate ; 8-Azaadenosine-5'- (mono)phosphate ; 8-Azidoadenosine-5'-(mono)phosphate ; N6-Methyladenosine-
5'-(mono)phosphate ; Pseudouridine-5'-(mono)phosphate; 2'-Amino-2'- deoxycytidine-(mono)phosphate ; 2'-Fluorothymidine-5'-(mono)phosphate; 2'-O- Methyl inosine-5'-(mono)phosphate) were used. All mRNA transcripts contained a poly-A tail about 70 bases long. The modified mRNAs were obtained in the in vitro transcription reaction by adding said template, an NMP-mix and one nucleotide, comprising one of the above mentioned chemically modified nucleosides to the reaction. The NMP-mix contained three non-modified nucleoside
(mono)phosphates of AMP, GMP, CMP, and UMP. The one remaining nucleotide is the nucleotide to be modified and comprises one of the above mentioned chemically modified nucleosides. The DNA template was then degraded by
DNAsel digestion and the DNA was recovered by LiCl precipitation.
3. Isolation of human peripheral blood mononuclear cells hPBMC were isolated by centrifugation on Ficoll (20 min at 2000 rpm) and subsequently washed two times in PBS. hPBMC were then resuspended in FCS, 10%
DMSO at a density of 5 x 10E7 / ml. 1 ml aliquots were frozen and stored at -800C.
4. Stimulation/Supression of cytokine release in hPBMC with modified mRNA
In order to evaluate the effect of different chemically modified nuleotides on stimulation/supression of cytokine release in hPBMC modified mRNA and protamine were mixed in water (20 μl, 1/1 w/w) and then diluted with 180 μl X-vivo 15 to a
RNA concentration of 15 μg/ml. hPBMc were thawed by resuspending in PBS, followed by two washes in PBS. hPBMC were then suspended in X-Vivo 15, 1 % glutamine, 1 % Pen/Strep at a density of 5 x 10E5 / ml. After seeding hPBMC at 1 x 10E5 per well in 96 well plates, 50 μl of protamine-complexed mRNA (final concentration 3 μg RNA / ml) were added to stimulate/suppress cytokine release over night at 37°C. 5. TNFalpha- and IL6-quantification (ELISA)
Maxisorb plates were coated over night (4°C) with capture antibody (1 μg/ml) and subsequently blocked with 1 % BSA for 1 hour at room temperature (RT). After three washes with 0.05% Tween, 50 μl (TNF ) or 10 μl (Il6) hPBMC supernatant, adjusted with complete X-Vivo 15 to 100 μl, were added to the wells. Binding was allowed to proceed for two hours (RT) and terminated by three washes with 0.05% Tween. 100 μl / well of a mixture of Strept-HRP (diluted 1/1000) and biotinylated detection antibody (0,5 μg/ml) were added. Incubation for one hour at RT was followed by three washes with 0.05% Tween. Finally, 100 μl / well of Amplex Red HRP substrate (50 μM), 0.014% H2O2 were added. Fluorescence was measured in a Spectramax
Gemini plate reader (Ex 540 nm, Em 590 nm, cutoff 590 nm).
As clearly can be seen in Figures 2 and 3, at least one nucleoside of the modified (m)RNAs has been modified. The corresponding nucleotides are selected from 2- Aminopurine-riboside-5'-(mono)phosphate; 2-Thiouridine-5'-(mono)phosphate ; 4-
Thiouridine-5'-(mono)phosphate; 5-Aminoallylcytidine-5'-(mono)phosphate ; 5- Aminoallyluridine-5'-(mono)phosphate ; 5-Bromocytidine-5'-(mono)phosphate ; 5- Bromo-2'-deoxycytidine-5'-(mono)phosphate ; 5-Bromouridine-5'-(mono)phosphate; 5-Bromo-2'-deoxyuridine-5'-(mono)phosphate ; 5-lodocytidine-5'-(mono)phosphate ; 5-lodo-2'-deoxycytidine-5'-(mono)phosphate ; 5-lodouridine-5'-(mono)phosphate;
5-lodo-2'-deoxyuridine-5'-(mono)phosphate ; 5-Methylcytidine-5'-(mono)phosphate ; 5-Methyluridine-5'-(mono)phosphate ; 5-Methyluridine-5'-(mono)phosphate ; 5- Propynyl-2'-deoxycytidine-5'-(mono)phosphate ; 5-Propynyl-2'-deoxyuridine-5'- (mono)phosphate; 6-Azacytidine-5'-(mono)phosphate ; 7-Deazaadenosine-5'- (mono)phosphate ; 7-Deazaguanosine-5'-(mono)phosphate ; 8-Azaadenosine-5'-
(mono)phosphate ; 8-Azidoadenosine-5'-(mono)phosphate ; N6-Methyladenosine- 5'-(mono)phosphate ; Pseudouridine-5'-(mono)phosphate; 2'-Amino-2'- deoxycytidine-(mono)phosphate ; 2'-Fluorothymidine-5'-(mono)phosphate; and 2'- O-Methyl inosine-5'-(mono)phosphate lead to a reduced secretion of IL-6 (Fig. 3) and TNF-alpha (Fig. 2), i.e. a reduced immunostimulatory effect, when compared to non-modified (m)RNA [or a control (no RNA)]. This effect is particularly strong, when chemical modifications are introduced leading to following nucleotides: 4- Thiouridine-5'-(mono)phosphate; 5-Aminoallyluridine-5'-(mono)phosphate ; 5- Bromo-2'-deoxycytidine-5'-(mono)phosphate ; 5-Bromouridine-5'-(mono)phosphate ; 5-Bromo-2'-deoxyuridine-5'-(mono)phosphate ; 5-lodouridine-5'-(mono)phosphate ; 5-lodo-2'-deoxyuridine-5'-(mono)phosphate ; 5-Methyluridine-5'-(mono)phosphate ; 5-Propynyl-2'-deoxyuridine-5'-(mono)phosphate ; 6-Azacytidine-5'- (mono)phosphate ; 8-Azidoadenosine-5'-(mono)phosphate; Pseudouridine-5'-
(mono)phosphate; 2'-Fluorothymidine-5'-(mono)phosphate; 2'-O-Methyl inosine-51- (mono)phosphate.
6. Materials Materials used in the above experiment are inter alia:
- Ficoll, Dulbecco's Phosphate Buffered Saline (PBS), BSA, Tween-20 and DMSO were from Sigma.
- ,,buffy coat" blood was from the department of Transfusion Medicine, Tubingen University Hospital. - FCS was from HyClone.
- Protaminchloride for injection was from Valeant.
- X-Vivo 15 lymphocyte medium, L-Glutamine (200 mM), Penicilline (Pen, 10000 U / ml) and Streptomycine (Strep, 10000 U / ml) were from Lonza.
- 96 well tissue culture plates were from Sarstedt. - 96 well Maxisorb plates were from Nunc.
- II6 and TNF capture antibodies, biotinylated II6 und TNF detection antibodies as well as Streptavidin-coupled horseradish peroxidase (Strept-HRP) were from BD Pharmingen.
- Amplex Red (20 mM in DMSO) was from Invitrogen.

Claims

Claims
1 . Immunosuppressive composition comprising at least one modified (m)RNA, wherein at least one nucleoside of the modified (m)RNA has been modified with: a) a chemical modification at the 4-, 5- or 6-position of the pyrimidine base of the nucleosides of cytidine and/or uridine; b) a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides of adenosine, inosine and/or guanosine; and/or c) a chemical modification at the 2'-position of the sugar of the nucleosides of adenosine, inosine, guanosine, cytidine and/or uridine, wherein the at least one modified (m)RNA is suitable for suppressing and/or avoiding an (innate) immunostimulatory response in a mammal typically exhibited when administering the corresponding unmodified (m)RNA.
2. Immunosuppressive composition according to claim 1 , wherein at least one nucleoside of the modified (m)RNA has been modified with: a chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides of adenosine, inosine and/or guanosine; and a chemical modification at the 2'-position of the sugar of the nucleosides of adenosine, inosine, guanosine, cytidine and/or uridine.
3. Immunosuppressive composition according to claim 1, wherein at least one nucleoside of the modified (m)RNA has been modified with: a chemical modification at the 4-, 5-or 6-position of the pyrimidine base of the nucleosides of cytidine and/or uridine; and a chemical modification at the 2'-position of the sugar of the nucleosides of adenosine, inosine, guanosine, cytidine and/or uridine.
4. Immunosuppressive composition according to any of claims 1 to 3, wherein the at least one modified (m)RNA is a coding RNA, a short RNA oligonucleotide, an si RNA, an antisense RNA, or riboswitches, ribozymes or aptamers.
5. Immunosuppressive composition according to any of claims 1 to 4, wherein the at least one modified (m)RNA is an mRNA.
6. Immunosuppressive composition according to claim 4, wherein the coding RNA encodes a therapeutically active protein or peptide, including apoptotic factors or apoptosis related proteins, recombinant proteins, growth hormones or growth factors, proteins that influence the metabolism and/or haematopoiesis, proteases, restriction enzymes, proteins which modulate various intracellular pathways by signal transmission modulation, immune modulators, including cytokines, lymphokines, monokines, interferones, antigen specific T cell receptors and antigens, or an antibody, an antigen, an allergen or an epitope thereof.
7. Immunosuppressive composition according to any of claims 1 to 4, wherein the at least one chemical modification at the 4-, 5- or 6-position of the base pyrimidine of the nucleosides cytidine and/or uridine is selected from the group consisting of 4- thio-, 5-iodo- / (5-I-), 5-bromo- / (5-Br-), 5-aminoallyl-, 5-fluoro- / (5-F-), 5-hydroxy-, 5-hydro- / (5-H-), 5-nitro-, 5-propynyl- / (5-(C≡C-CH3)-), 5-methyl-, 5-methyl-2-thio-,
5-formyl-, 5-hydroxymethyl-, 5-methoxy-, ,5-oxyacetic acid methyl ester-, 5- oxyacetic acid-, 5-carboxyhydroxymethyl-, 5-(carboxyhydroxymethyl)pyrimidine methyl ester-, 5-methoxycarbonylmethyl-, 5-methoxycarbonylmethyl-2-thio, 5- aminomethyl-, 5-aminomethyl-2-thio-, 5-aminomethyl-2-seleno-, 5- methylaminomethyl-, 5-carbamoylmethyl-, 5-carboxymethylaminomethyl-, 5- carboxymethylaminomethyl-2-thio-, 5-carboxymethyl-, 5-methyldihydro-, 5- taurinomethyl-, 5-taurinomethyl-2-thiouridine, 5-isopentenylaminomethyl-, 5- isopentenylaminomethyl-2-thio-, 5-aminopropyl- / (5-(C3H6NH3)-), 5-methoxy- ethoxy-methyl- / (5-(CH2-O-C2H4-O-CH3)-), and 6-aza-modifications.
8. Immunosuppressive composition according to any of claims 1 to 4, wherein the at least one chemical modification at the 2-, 6-, 7- or 8-position of the purine base of the nucleosides of adenosine and/or guanosine is selected from the group consisting of 2-Amino-, 7-Deaza-, 8-Aza-, and 8-Azido-modifications.
9. Immunosuppressive composition according to any of claims 1 to 4, wherein the at least one chemical modification at the 2'-position of the sugar of the nucleosides of adenosine, guanosine, cytidine and/or uridine is selected from the group consisting of 2'-deoxy-, 2'-amino-, 2'-fluoro-, and 2'-O-methyl-modifications.
10. Immunosuppressive composition according to any of claims 1 to 9, wherein at least one nucleoside of the at least one modified (m)RNA has been modified leading to chemically modified nucleotides (of the (m)RNA) selected from the group consisting of: 4-thio-uridine-5'-(mono)phosphate, 2-Aminopurine-riboside-5'-
(mono)phosphate, 5-Aminoallylcytidine-5'-(mono)phosphate, 5-Aminoallyluridine- 5'-(mono)phosphate, 5-Bromocytidine-5'-(mono)phosphate, 5-Bromo-2'- deoxycytidine-5'-(mono)phosphate, 5-Bromouridine-5'-(mono)phosphate, 5-Bromo- 2'-deoxyuridine-5'-(mono)phosphate, 5-lodocytidine-5'-(mono)phosphate, 5-lodo- 2'-deoxycytidine-5'-(mono)phosphate, 5-lodouridine-5'-(mono)phosphate, 5-lodo-
2'-deoxyuridine-5'-(mono)phosphate, 5-Propynyl-2'-deoxycytidine-5'-
(mono)phosphate, 5-Propynyl-2'-deoxyuridine-5'-(mono)phosphate, 5- formylcytidine-5'-(mono)phosphate, 5,2'-0-dimethylcytidine-5'-(mono)phosphate, 5-hydroxymethylcytidine-5'-(mono)phosphate, 5-formyl-2'-0-methylcytidine-5'- (mono)phosphate, 5,2'-0-dimethyluridine-5'-(mono)phosphate, 5-methyl-2- thiouridine-5'-(mono)phosphate, 5-hydroxyuridine-5'-(mono)phosphate, 5- methoxyuridine-5'-(mono)phosphate, uridine 5-oxyacetic acid-5'-(mono)phosphate, uridine 5-oxyacetic acid methyl ester-5'-(mono)phosphate, 5- (carboxyhydroxymethyl)uridine-5'-(mono)phosphate, 5- (carboxyhydroxymethyl)uridine methyl ester-5'-(mono)phosphate, 5- methoxycarbonylmethyluridine-5'-(mono)phosphate, 5-methoxycarbonylmethyl-2'- C>-methyluridine-5'-(mono)phosphate, 5-methoxycarbonylmethyl-2-thiouridine-5'- (mono)phosphate, 5-aminomethyl-2-thiouridine-5'-(mono)phosphate, 5- methylaminomethyluridine-5'-(mono)phosphate, 5-methylaminomethyl-2- thiouridine-5'-(mono)phosphate, 5-methylaminomethyl-2-selenouridine-5'-
(mono)phosphate, 5-carbamoylmethyluridine-5'-(mono)phosphate, 5- carbamoylmethyl-2'-0-methyluridine-5'-(mono)phosphate, 5- carboxymethylaminomethyluridine-5'-(mono)phosphate, 5- carboxymethylaminomethyl-2'-0-methyluridine-5'-(mono)phosphate, 5- carboxymethylaminomethyl-2-thiouridine-5'-(mono)phosphate, 5- carboxymethyluridine-5'-(mono)phosphate, 5-methyldihydrouridine-5'-
(mono)phosphate, 5-taurinomethyluridine-5'-(mono)phosphate, 5-taurinomethyl-2- thiouridine-5'-(mono)phosphate, 5-(isopentenylaminomethyl)uridine-5'- (mono)phosphate, 5-(isopentenylaminomethyl)-2-thiouridine-5'-(mono)phosphate, 5-(isopentenylaminomethyl)-2'-0-methyluridine-5'-(mono)phosphate, 6-
Azacytidi ne-5 '-(mono)phosphate, 7-Deazaadenosi ne-5 '-(mono)phosphate, 7- Deazaguanosine-5'-(mono)phosphate, 8-Azaadenosine-5'-(mono)phosphate, 8- Azidoadenosine-5'-(mono)phosphate, Pseudouridine-5'-(mono)phosphate, 2'-
Amino-2'-deoxycytidine-(mono)phosphate, 2'-Fluorothymidine-5'-(mono)phosphate/ inosine-5'-(mono)phosphate, 2'-O-Methyl-inosine-5'-(mono)phosphate.
11 . Immunosuppressive composition according to any of claims 1 to 10, wherein the at least one modified (m)RNA has been modified leading to chemically modified nucleotides selected from the group consisting of 4-thio-uridine-5'-
(mono)phosphate, 5-Aminoallyluridine-5'-(mono)phosphate, 5-Bromo-2'- deoxycytidine-5'-(mono)phosphate, 5-Bromouridine-5'-(mono)phosphate, 5-Bromo-
2'-deoxyuridine-5'-(mono)phosphate, 5-lodouridine-5'-(mono)phosphate, 5-lodo-2'- deoxyuridine-5'-(mono)phosphate, 5-Propynyl-2'-deoxyuridine-5'-(mono)phosphate,
6-Azacytidine-5'-(mono)phosphate, 8-Azidoadenosine-5'-(mono)phosphate, 2'-
Fluorothyrnidine-5'-(rnono)phosphate, inosine-5'-(mono)phosphate and 2'-O-
Methyl-inosine-5'-(mono)phosphate.
12. Immunosuppressive composition according to any of claims 1 to 11 , wherein the modified (m)RNA comprises a length of 250 to 20000 nucleotides.
13. Immunosuppressive composition according to any of claims 1 to 12, wherein the at least one modified (m)RNA is a GC-stabilized modified (m)RNA.
14. Immunosuppressive composition according to claim 13, wherein the G/C content of the coding region of the GC-stabilized modified (m)RNA is increased compared with the G/C content of the coding region of the native modified (m)RNA, the coded amino acid sequence of the GC-stabilized modified (m)RNA preferably not being altered compared with the coded amino acid sequence of the native modified
(m)RNA.
15. Immunosuppressive composition according to any of claims 13 to 14, wherein the A/U content in the environment of the ribosome binding site of the modified (m)RNA is increased compared with the A/U content in the environment of the ribosome binding site of the native modified (m)RNA.
16. Immunosuppressive composition according to any of claims 13 to 15, wherein the coding region and/or the 5' and/or 3' untranslated region of the modified (m)RNA is further modified compared to the native modified (m)RNA such that it contains no destabilizing sequence elements, the coded amino acid sequence of the further modified (m)RNA preferably not being modified compared to the native modified (m)RNA.
17. Immunosuppressive composition according to any of claims 13 to 16, wherein the at least one modified mRNA has a 51 cap structure and/or a poly(A) tail, preferably of 10 to 200 adenosine nucleotides, and/or a poly(C) tail, preferably of 10 to 200 cytosine nucleotides, and/or at least one IRES and/or at least one 5' and/or 3' stabilizing sequence.
18. Immunosuppressive composition according to any of claims 1 to 1 7, wherein the at least one modified (m)RNA is complexed with one or more polycations, selected from polycations, including poly-L-lysine (PLL), chitosan, polyethylenimine (PEI), polydimethylaminoethylmethacrylate (PD-MAEMA), polyamidoamine (PAMAM), protamine, nucleoline, spermin or spermidine, histones, cationic polysaccharides, chitosan, polybrene, and cationic lipids, including oligofectamine, preferably with protamine or oligofectamine, most preferably with protamine, or from polycations selected from from oligoarginines having formula (I): (Arg)ι;(Lys)m;(His)n;(Orn)o;(Xaa)x, including Arg7, Argg, Arg9, LyS7, LyS8, Lys9, wherein I
+ m + n +o + x = 8-15, and I, m, n or o independently of each other is any number selected from 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 or 15, provided that the overall content of Arg, Lys, His and Orn represents at least 70% of all amino acids of the oligopeptide; and Xaa is any amino acid selected from native (= naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x is any number selected from 0, 1 , 2, 3 or 4, provided, that the overall content of Xaa does not exceed 30 % of all amino acids of the oligopeptide.
19. Pharmaceutical composition, comprising an immunosuppressive composition according to any of claims 1 to 18 or a modified (m)RNA according to any of claims 1 to 18 as defined according to any of claims 1 to 18, and optionally a pharmaceutically acceptable carrier.
20. Pharmaceutical composition according to claim 19, wherein the pharmaceutical composition is a vaccine.
21. Pharmaceutical composition according to claim 19 or 20, wherein the modified (m)RNA encodes an antibody.
22. Use of an immunosuppressive composition according to any of claims 1 to 18 or a modified (m)RNA as defined in any of claims 1 to 18, for preparing a pharmaceutical composition for prophylaxis, treatment, and/or amelioration of diseases and disorders selected from cancers or tumor diseases, infectious diseases, including viral, bacterial or protozoological infectious diseases, autoimmune diseases, allergic disorders or diseases, monogenetic diseases, or the treatment of patients in need of a specific therapeutically active protein.
23. Use according to claim 22, wherein the cancer or tumour diseases are selected from melanomas, malignant melanomas, colon carcinomas, lymphomas, sarcomas, blastomas, renal carcinomas, gastrointestinal tumours, gliomas, prostate tumours, bladder cancer, rectal tumours, stomach cancer, oesophageal cancer, pancreatic cancer, liver cancer, mammary carcinomas (= breast cancer), uterine cancer, cervical cancer, acute myeloid leukaemia (AML), acute lymphoid leukaemia (ALL), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), hepatomas, various virus-induced tumours such as, for example, papilloma virus- induced carcinomas (e.g. cervical carcinoma = cervical cancer), adenocarcinomas, herpes virus-induced tumours (e.g. Burkitt's lymphoma, EBV-induced B-cell lymphoma), heptatitis B-induced tumours (hepatocell carcinomas), HTLV-1 - and
HTLV-2-induced lymphomas, acoustic neuroma, lung carcinomas (= lung cancer = bronchial carcinoma), small-cell lung carcinomas, pharyngeal cancer, anal carcinoma, glioblastoma, rectal carcinoma, astrocytoma, brain tumours, retinoblastoma, basalioma, brain metastases, medulloblastomas, vaginal cancer, pancreatic cancer, testicular cancer, Hodgkin's syndrome, meningiomas, Schneeberger disease, hypophysis tumour, Mycosis fungoides, carcinoids, neurinoma, spinalioma, Burkitt's lymphoma, laryngeal cancer, renal cancer, thymoma, corpus carcinoma, bone cancer, non-Hodgkin's lymphomas, urethral cancer, CUP syndrome, head/neck tumours, oligodendroglioma, vulval cancer, intestinal cancer, colon carcinoma, oesophageal carcinoma (= oesophageal cancer), wart involvement, tumours of the small intestine, craniopharyngeomas, ovarian carcinoma, genital tumours, ovarian cancer (= ovarian carcinoma), pancreatic carcinoma (= pancreatic cancer), endometrial carcinoma, liver metastases, penile cancer, tongue cancer, gall bladder cancer, leukaemia, plasmocytoma, lid tumour, and prostate cancer (= prostate tumours).
24. Use according to claim 22, wherein the infectious diseases, including viral, bacterial or protozoological infectious diseases, selected from influenza, malaria, SARS, yellow fever, AIDS, Lyme borreliosis, Leishmaniasis, anthrax, meningitis, viral infectious diseases such as AIDS, Condyloma acuminata, hollow warts, Dengue fever, three-day fever, Ebola virus, cold, early summer meningoencephalitis (FSME), flu, shingles, hepatitis, herpes simplex type I, herpes simplex type II, Herpes zoster, influenza, Japanese encephalitis, Lassa fever, Marburg virus, measles, foot-and- mouth disease, mononucleosis, mumps, Norwalk virus infection, Pfeiffer's glandular fever, smallpox, polio (childhood lameness), pseudo-croup, fifth disease, rabies, warts, West Nile fever, chickenpox, cytomegalic virus (CMV), bacterial infectious diseases such as miscarriage (prostate inflammation), anthrax, appendicitis, borreliosis, botulism, Camphylobacter, Chlamydia trachomatis (inflammation of the urethra, conjunctivitis), cholera, diphtheria, donavanosis, epiglottitis, typhus fever, gas gangrene, gonorrhoea, rabbit fever, Heliobacter pylori, whooping cough, climatic bubo, osteomyelitis, Legionnaire's disease, leprosy, listeriosis, pneumonia, meningitis, bacterial meningitis, anthrax, otitis media, Mycoplasma hominis, neonatal sepsis (Chorioamnionitis), noma, paratyphus, plague, Reiter's syndrome, Rocky Mountain spotted fever, Salmonella paratyphus, Salmonella typhus, scarlet fever, syphilis, tetanus, tripper, tsutsugamushi disease, tuberculosis, typhus, vaginitis (colpitis), soft chancre, and infectious diseases caused by parasites, protozoa or fungi, such as amoebiasis, bilharziosis, Chagas disease, Echinococcus, fish tapeworm, fish poisoning (Ciguatera), fox tapeworm, athlete's foot, canine tapeworm, candidosis, yeast fungus spots, scabies, cutaneous Leishmaniosis, lambliasis (giardiasis), lice, malaria, microscopy, onchocercosis (river blindness), fungal diseases, bovine tapeworm, schistosomiasis, sleeping sickness, porcine tapeworm, toxoplasmosis, trichomoniasis, trypanosomiasis (sleeping sickness), visceral Leishmaniosis, nappy/diaper dermatitis, miniature tapeworm, or prion diseases, including Creutzfeld-Jakob disease, BSE, scrapie, and Kuru.
25. Use according to claim 22, wherein the autoimmune diseases are selected from antibody-mediated or T-cell mediated immunity, systemic and organ-specific or localised autoimmune disorders, including type I autoimmune diseases or type Il autoimmune diseases or type III autoimmune diseases or type IV autoimmune diseases, including systemic lupus erythematosus (SLE), Sjogren's syndrome, Scleroderma, Rheumatoid Arthritis including juvenile (rheumatoid) arthritis, polymyositis, endocrinologic autoimmune disorders including type I diabetes
(Diabetes mellitus Type 1 ), Hashimoto's thyroiditis, and Addison's disease, dermatologic autoimmune disorders including pemphigus vulgaris, haematologic autoimmune disorders including autoimmune haemolytic anaemia, neural autoimmune disorders including multiple sclerosis, multiple sclerosis (MS), rheumatoid arthritis, diabetes, type I diabetes (Diabetes mellitus Type 1 ), chronic polyarthritis, Basedow's disease, autoimmune forms of chronic hepatitis, colitis ulcerosa, type I allergy diseases, type Il allergy diseases, type III allergy diseases, type IV allergy diseases, fibromyalgia, hair loss, alopecia, alopecia areata, Bechterew's disease, Crohn's disease, Myasthenia gravis, neurodermitis, Polymyalgia rheumatica, progressive systemic sclerosis (PSS), rheumatic arthritis, psoriasis, vasculitis, etc, type Il diabetes, graft versus host disease, transplanted organ rejection, asthma, acquired hemophilia, ankylosing spondylitis (Morbus Beschterew or spondylitis ankylosans), antiphospholipid syndrome, autoimmune hepatitis, autoimmune hemolytic anemia, Behcet's syndrome, cardiomyopathy, celiac sprue dermatitis, chronic fatigue immune dysfunction syndrome (CFIDS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, cicatricial pemphigoid, CREST syndrome, cold agglutinin disease, dermatomyositis, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia, fibromyositis, Guillain-Barre syndrome, idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura, IgA nephropathy, inflammatory bowel disease (including Crohn's disease and ulcerative colitis), juvenile arthritis, lichen planus, myasthenia gravis, multiple sclerosis, mixed connective tissue disease, polyarteritis nodosa, polychondritis, polyglandular syndromes, polymyalgia rheumatica, primary agammaglobulinemia, primary biliary cirrhosis, psoriasis, Raynaud's phenomena, Reiter's syndrome, rheumatoid arthritis (RA), Sjorgen's syndrome, sarcoidosis, stiff- man syndrome, systemic lupus erythematosus (SLE), Takayasu arthritis, temporal arteritis/giant cell arteritis, uveitis, vasculitis, and vitiligo.
26. Use according to claim 22, wherein the allergic disorders or diseases are caused by allergens including grasses, pollens, molds, drugs, and numerous environmental triggers, and/or includes allergic disorders or diseases selected from allergic asthma, allergic conjunctivitis, allergic rhinitis ("hay fever"), anaphylaxis, angiodema, atopic dermatitis (eczema), urticaria (hives), eosinophilia, respiratory, allergies to insect stings, skin allergies (leading to and including various rashes, such as eczema, hives (urticaria) and (contact) dermatitis), food allergies, and allergies to medicine.
27. Use according to claim 22, wherein the genetic diseases include 3-beta- hydroxysteroid dehydrogenase deficiency (type II); 3-ketothiolase deficiency; 6- mercaptopurine sensitivity; Aarskog-Scott syndrome; Abetalipoproteinemia; Acatalasemia; Achondrogenesis; Achondrogenesis-hypochondrogenesis;
Achondroplasia; Achromatopsia; Acromesomelic dysplasia (Hunter-Thompson type); ACTH deficiency; Acyl-CoA dehydrogenase deficiency (short-chain, medium chain, long chain); Adenomatous polyposis coli; Adenosin-deaminase deficiency;
Adenylosuccinase deficiency; Adhalinopathy; Adrenal hyperplasia, congenital (due to 1 1 -beta-hydroxylase deficiency; due to 17-alpha-hydroxylase deficiency; due to 21 -hydroxylase deficiency); Adrenal hypoplasia, congenital, with hypogonadotropic hypogonadism; Adrenogenital syndrom; Adrenoleukodystrophy; Adrenomyeloneuropathy; Afibrinogenemia; Agammaglobulinemia; Alagille syndrome; Albinism (brown, ocular, oculocutaneous, rufous); Alcohol intolerance, acute; Aldolase A deficiency; Aldosteronism, glucocorticoid-remediable; Alexander disease; Alkaptonuria; Alopecia universalis; Alpha-1 -antichymotrypsin deficiency; Alpha-methylacyl-CoA racemase deficiency; Alpha-thalassemia/mental retardation syndrome; Alport syndrome; Alzheimer disease-1 (APP-related); Alzheimer disease- 3; Alzheimer disease-4; Amelogenesis imperfecta; Amyloid neuropathy (familial, several allelic types); Amyloidosis (Dutch type; Finnish type; hereditary renal; renal; senile systemic); Amytrophic lateral sclerosis; Analbuminemia; Androgen insensitivity; Anemia (Diamond-Blackfan); Anemia (hemolytic, due to PK deficiency); Anemia (hemolytic, Rh-null, suppressor type); Anemia (neonatal hemolytic, fatal and nearfatal); Anemia (sideroblastic, with ataxia); Anemia (sideroblastic/hypochromic); Anemia due to G6PD deficiency; Aneurysm (familial arterial); Angelman syndrome; Angioedema; Aniridia; Anterior segment anomalies and cataract; Anterior segment mesenchymal dysgenesis; Anterior segment mesenchymal dysgenesis and cataract; Antithrombin III deficiency; Anxiety-related personality traits; Apert syndrome; Apnea (postanesthetic); ApoA-l and apoC-lll deficiency (combined); Apolipoprotein A-Il deficiency; Apolipoprotein B-100 (ligand-defective); Apparent mineralocorticoid excess (hypertension due to); Argininemia; Argininosuccinicaciduria; Arthropathy (progressive pseudorheumatoid, of childhood); Aspartylglucosaminuria; Ataxia (episodic); Ataxia with isolated vitamin E deficiency; Ataxia-telangiectasia; Atelosteogenesis II; ATP-dependent DNA ligase I deficiency; Atrial septal defect with atrioventricular conduction defects; Atrichia with papular lesions; Autism (succinylpurinemic); Autoimmune polyglandular disease, type I; Autonomic nervous system dysfunction; Axenfeld anomaly; Azoospermia; Bamforth-Lazarus syndrome; Bannayan-Zonana syndrome; Barthsyndrome; Bartter syndrome (type 2 or type 3); Basal cell carcinoma ; Basal cell nevus syndrome; BCG infection; Beare-Stevenson cutis gyrata syndrome; Becker muscular dystrophy; Beckwith-Wiedemann syndrome; Bernard-Soulier syndrome (type B; type C); Bethlem myopathy; Bile acid malabsorption, primary ; Biotinidase deficiency; Bladder cancer; Bleeding disorder due to defective thromboxane A2 receptor; Bloom syndrome; Brachydactyly (type B1 or type C); Branchiootic syndrome; Branchiootorenal syndrome; Breast cancer (invasive intraductal; lobular; male, with Reifenstein syndrome; sporadic); Breast cancer-1 (early onset); Breast cancer-2 (early onset); Brody myopathy; Brugada syndrome; Brunner syndrome;
Burkitt lymphoma; Butterfly dystrophy (retinal); C1 q deficiency (type A; type B; type C ); C1 r/C1 s deficiency; C1 s deficiency, isolated; C2 deficiency ; C3 deficiency; C3b inactivator deficiency; C4 deficiency; C8 deficiency, type II; C9 deficiency; Campomelic dysplasia with autosomal sex reversal; Camptodactyly-arthropathy- coxa varapericarditis syndrome; Canavan disease; Carbamoylphosphate synthetase I deficiency; Carbohydrate-deficient glycoprotein syndrome (type I; type Ib; type II); Carcinoid tumor of lung; Cardioencephalomyopathy (fatal infantile, due to cytochrome c oxidase deficiency); Cardiomyopathy (dilated; X-linked dilated; familial hypertrophic; hypertrophic); Carnitine deficiency (systemic primary); Carnitine-acylcarnitine translocase deficiency; Carpal tunnel syndrome (familial); Cataract (cerulean; congenital; crystalline aculeiform; juvenile-onset; polymorphic and lamellar; punctate; zonular pulverulent); Cataract, Coppock-like; CD59 deficiency; Central core disease; Cerebellar ataxia; Cerebral amyloid angiopathy;
Cerebral arteriopathy with subcortical infarcts and leukoencephalopathy; Cerebral cavernous malformations-1; Cerebrooculofacioskeletal syndrome; Cerebrotendinous xanthomatosis; Cerebrovascular disease; Ceroid lipofuscinosis (neuronal, variant juvenile type, with granular osmiophilic deposits); Ceroid lipofuscinosis (neuronal-!, infantile); Ceroid-lipofuscinosis (neuronal-3, juvenile); Char syndrome; Charcot-
Marie-Tooth disease; Charcot-Marie-Tooth neuropathy; Charlevoix-Saguenay type; Chediak-Higashi syndrome; Chloride diarrhea (Finnish type); Cholestasis (benign recurrent intrahepatic); Cholestasis (familial intrahepatic); Cholestasis (progressive familial intrahepatic); Cholesteryl ester storage disease; Chondrodysplasia punctata (brachytelephalangic; rhizomelic; X-linked dominant; X-linked recessive; Grebe type); Chondrosarcoma; Choroideremia; Chronic granulomatous disease (autosomal, due to deficiency of CYBA); Chronic granulomatous disease (X-linked); Chronic granulomatous disease due to deficiency of NCF-1 ; Chronic granulomatous disease due to deficiency of NCF-2; Chylomicronemia syndrome, familial; Citrullinemia; classical Cockayne syndrome-1 ; Cleft lip, cleft jaw, cleft palate; Cleft lip/palate ectodermal dysplasia syndrome; Cleidocranial dysplasia; CMO Il deficiency; Coats disease; Cockayne syndrome-2, type B; Coffin-Lowry syndrome; Colchicine resistance; Colon adenocarcinoma; Colon cancer; Colorblindness (deutan; protan; tritan); Colorectal cancer; Combined factor V and VIII deficiency; Combined hyperlipemia (familial); Combined immunodeficiency (X-linked, moderate);
Complex I deficiency; Complex neurologic disorder; Cone dystrophy-3; Cone-rod dystrophy 3; Cone-rod dystrophy 6; Cone-rod retinal dystrophy-2; Congenital bilateral absence of vas deferens; Conjunctivitis, ligneous; Contractural arachnodactyly; Coproporphyria; Cornea plana congenita; Corneal clouding; Corneal dystrophy (Avellino type; gelatinous drop-like; Groenouw type I; lattice type I; Reis-Bucklers type); Cortisol resistance; Coumarin resistance; Cowden disease; CPT deficiency, hepatic (type I; type II); Cramps (familial, potassium-aggravated); Craniofacial-deafness-hand syndrome; Craniosynostosis (type 2); Cretinism;
Creutzfeldt-Jakob disease ; Crigler-Najjar syndrome; Crouzon syndrome; Currarino syndrome; Cutis laxa; Cyclic hematopoiesis; Cyclic ichthyosis; Cylindromatosis; Cystic fibrosis; Cystinosis (nephropathies Cystinuria (type II; type III); Daltonism; Darier disease; D-bifunctional protein deficiency; Deafness, autosomal dominant 1; Deafness, autosomal dominant 1 1; Deafness, autosomal dominant 12; Deafness, autosomal dominant 15; Deafness, autosomal dominant 2; Deafness, autosomal dominant 3; Deafness, autosomal dominant 5; Deafness, autosomal dominant 8; Deafness, autosomal dominant 9; Deafness, autosomal recessive 1; Deafness, autosomal recessive 2; Deafness, autosomal recessive 21 ; Deafness, autosomal recessive 3; Deafness, autosomal recessive 4; Deafness, autosomal recessive 9;
Deafness, nonsyndromic sensorineural 13; Deafness, X-linked 1 ; Deafness, X-linked 3; Debrisoquine sensitivity; Dejerine-Sottas disease; Dementia (familial Danish); Dementia (frontotemporal, with parkinsonism); Dent disease; Dental anomalies; Dentatorubro-pallidoluysian atrophy; Denys-Drash syndrome; Dermatofibrosarcoma protuberans; Desmoid disease; Diabetes insipidus (nephrogenic ); Diabetes insipidus
(neurohypophyseal); Diabetes mellitus (insulin-resistant); Diabetes mellitus (rare form); Diabetes mellitus (type II); Diastrophic dysplasia; Dihydropyrimidinuria; Dosage-sensitive sex reversal; Doyne honeycomb degeneration of retina; Dubin- Johnson syndrome; Duchenne muscular dystrophy; Dyserythropoietic anemia with thrombocytopenia; Dysfibrinogenemia (alpha type; beta type; gamma type);
Dyskeratosis congenita-1 ; Dysprothrombinemia; Dystonia (DOPAresponsive); Dystonia (myoclonic); Dystonia-1 (torsion); Ectodermal dysplasia; Ectopia lentis; Ectopia pupillae; Ectrodactyly (ectodermal dysplasia, and cleft lip/palate syndrome 3); Ehlers-Danlos syndrome (progeroid form); Ehlers-Danlos syndrome (type I; type II; type III; type IV; type Vl; type VII); Elastin Supravalvar aortic stenosis;
Elliptocytosis-1 ; Elliptocytosis-2; Elliptocytosis-3; Ellis-van Creveld syndrome; Emery-Dreifuss muscular dystrophy; Emphysema; Encephalopathy; Endocardial fibroelastosis-2; Endometrial carcinoma; Endplate acetylcholinesterase deficiency; Enhanced S-cone syndrome; Enlarged vestibular aqueduct; Epidermolysis bullosa; Epidermolysis bullosa dystrophica (dominant or recessive); Epidermolysis bullosa simplex; Epidermolytic hyperkeratosis; Epidermolytic palmoplanar keratoderma; Epilepsy (generalize; juvenile; myoclonic; nocturnal frontal lobe; progressive myoclonic); Epilepsy, benign, neonatal (typei or type2); Epiphyseal dysplasia
(multiple); Episodic ataxia (type 2); Episodic ataxia/myokymia syndrome; Erythremias (alpha-; dysplasia); Erythrocytosis; Erythrokeratoderma; Estrogen resistance; Exertional myoglobinuria due to deficiency of LDH-A; Exostoses, multiple (type 1; type 2); Exudative vitreoretinopathy, X-linked; Fabry disease; Factor H deficiency; Factor VII deficiency; Factor X deficiency; Factor Xl deficiency; Factor
XII deficiency; Factor XIIIA deficiency; Factor XIIIB deficiency; Familial Mediterranean fever; Fanconi anemia; Fanconi-Bickel syndrome; Farber lipogranulomatosis; Fatty liver (acute); Favism; Fish-eye disease; Foveal hypoplasia; Fragile X syndrome; Frasier syndrome; Friedreich ataxia; fructose-bisphosphatase Fructose intolerance; Fucosidosis; Fumarase deficiency; Fundus albipunctatus;
Fundus flavimaculatus; G6PD deficiency; GABA-transaminase deficiency; Galactokinase deficiency with cataracts; Galactose epimerase deficiency; Galactosemia; Galactosialidosis; GAMT deficiency; Gardner syndrome; Gastric cancer; Gaucher disease; Generalized epilepsy with febrile seizures plus; Germ cell tumors; Gerstmann-Straussler disease; Giant cell hepatitis (neonatal); Giant platelet disorder; Giant-cell fibroblastoma; Gitelman syndrome; Glanzmann thrombasthenia (type A; type B); Glaucoma 1 A; Glaucoma 3A; Glioblastoma multiforme; Glomerulosclerosis (focal segmental); Glucose transport defect (blood-brain barrier); Glucose/galactose malabsorption; Glucosidase I deficiency; Glutaricaciduria (type I; type MB; type MC); Gluthation synthetase deficiency; Glycerol kinase deficiency;
Glycine receptor (alpha-1 polypeptide); Glycogen storage disease I; Glycogen storage disease II; Glycogen storage disease III; Glycogen storage disease IV; Glycogen storage disease Vl; Glycogen storage disease VII; Glycogenosis (hepatic, autosomal); Glycogenosis (X-linked hepatic); GM1 -gangliosidosis; GM2- gangliosidosis; Goiter (adolescent multinodular); Goiter (congenital); Goiter
(nonendemic, simple); Gonadal dysgenesis (XY type); Granulomatosis, septic; Graves disease; Greig cephalopolysyndactyly syndrome; Griscelli syndrome; Growth hormone deficient dwarfism; Growth retardation with deafness and mental retardation; Gynecomastia (familial, due to increased aromatase activity); Gyrate atrophy of choroid and retina with ornithinemia (B6 responsive or unresponsive);
Hailey-Hailey disease; Haim-Munk syndrome; Hand-foot-uterus syndrome;
Harderoporphyrinuria; HDL deficiency (familial); Heart block (nonprogressive or progressive); Heinz body anemia; HELLP syndrome; Hematuria (familial benign);
Heme oxygenase-1 deficiency; Hemiplegic migraine; Hemochromotosis;
Hemoglobin H disease; Hemolytic anemia due to ADA excess; Hemolytic anemia due to adenylate kinase deficiency; Hemolytic anemia due to band 3 defect;
Hemolytic anemia due to glucosephosphate isomerase deficiency; Hemolytic anemia due to glutathione synthetase deficiency; Hemolytic anemia due to hexokinase deficiency; Hemolytic anemia due to PGK deficiency; Hemolytic-uremic syndrome; Hemophagocytic lymphohistiocytosis; Hemophilia A; Hemophilia B;
Hemorrhagic diathesis due to factor V deficiency; Hemosiderosis (systemic, due to aceruloplasminemia); Hepatic lipase deficiency; Hepatoblastoma; Hepatocellular carcinoma; Hereditary hemorrhagic telangiectasia-1; Hereditary hemorrhagic telangiectasia-2; Hermansky-Pudlak syndrome; Heterotaxy (X-linked visceral);
Heterotopia (periventricular); Hippel-Lindau syndrom; Hirschsprung disease;
Histidine-rich glycoprotein Thrombophilia due to HRG deficiency; HMG-CoA lyase deficiency; Holoprosencephaly-2; Holoprosencephaly-3; Holoprosencephaly-4; Holoprosencephaly-5; Holt-Oram syndrome; Homocystinuria; Hoyeraal-
Hreidarsson; HPFH (deletion type or nondeletion type); HPRT-related gout;
Huntington disease; Hydrocephalus due to aqueductal stenosis; Hydrops fetalis;
Hyperbetalipoproteinemia; Hypercholesterolemia, familial; Hyperferritinemia- cataract syndrome; Hyperglycerolemia; Hyperglycinemia; Hyperimmunoglobulinemia D and periodic fever syndrome; Hyperinsulinism;
Hyperinsulinism-hyperammonemia syndrome; Hyperkalemic periodic paralysis;
Hyperlipoproteinemia; Hyperlysinemia; Hypermethioninemia (persistent, autosomal, dominant, due to methionine, adenosyltransferase I/Ill deficiency);
Hyperornithinemia-hyperammonemiahomocitrullinemia syndrome; Hyperoxaluria; Hyperparathyroidism; Hyperphenylalaninemia due to pterin-4acarbinolamine dehydratase deficiency; Hyperproinsulinemia; Hyperprolinemia; Hypertension;
Hyperthroidism (congenital); Hypertriglyceridemia; Hypoalphalipoproteinemia;
Hypobetalipoproteinemia; Hypocalcemia; Hypochondroplasia; Hypochromic microcytic anemia; Hypodontia; Hypofibrinogenemia; Hypoglobulinemia and absent B cells; Hypogonadism (hypergonadotropic); Hypogonadotropic (hypogonadism); Hypokalemic periodic paralysis; Hypomagnesemia; Hypomyelination (congenital); Hypoparathyroidism; Hypophosphatasia (adult; childhood; infantile; hereditary); Hypoprothrombinemia; Hypothyroidism
(congenital; hereditary congenital; nongoitrous); lchthyosiform erythroderma ; Ichthyosis ; Ichthyosis bullosa of Siemens ; lgG2 deficiency; lmmotile cilia syndrome-1; Immunodeficiency (T-cell receptor/CD3 complex); Immunodeficiency (X-linked, with hyper-lgM); Immunodeficiency due to defect in CD3-gamma; Immunodeficiency-centromeric i nstabi I ityfaci al anomalies syndrome; Incontinentia pigmenti; Insensitivity to pain (congenital, with anhidrosis); Insomnia (fatal familial); lnterleukin-2 receptor deficiency (alpha chain); Intervertebral disc disease; Iridogoniodysgenesis; Isolated growth hormone deficiency (lllig type with absent GH and Kowarski type with bioinactive GH); Isovalericacidemia ; Jackson-Weiss sydnrome; Jensen syndrome; Jervell and Lange-Nielsen syndrome; Joubert syndrom;
Juberg-Marsidi syndrome; Kallmann syndrome; Kanzaki disease; Keratitis; Keratoderma (palmoplanar); Keratosis palmoplantaris striata I; Keratosis palmoplantaris striata II; Ketoacidosis due to SCOT deficiency; Keutel syndrome; Klippel-Trenaumay syndrom; Kniest dysplasia; Kostmann neutropenia; Krabbe disease; Kurzripp-Polydaktylie syndrom; Lacticacidemia due to PDX1 deficiency;
Langer mesomelic dysplasia; Laron dwarfism; Laurence-Moon-Biedl-Bardet syndrom; LCHAD deficiency; Leber congenital amaurosis; Left-right axis malformation; Leigh syndrome; Leiomyomatosis (diffuse, with Alport syndrome); Leprechaunism; Leri- Weill dyschondrosteosis; Lesch-Nyhan syndrome; Leukemia (acute myeloid; acute promyelocyte; acute T-cell lymphoblastic; chronic myeloid; juvenile myelomonocytic; Leukemia-1 (T-cell acute lymphocytic); Leukocyte adhesion deficiency; Leydig cell adenoma; Lhermitte-Duclos syndrome; Liddle syndrome; Li-Fraumeni syndrome; Lipoamide dehydrogenase deficiency; Lipodystrophy; Lipoid adrenal hyperplasia; Lipoprotein lipase deficiency; Lissencephaly (X-linked); Lissencephaly-1 ; liver Glycogen storage disease (type 0);
Long QT syndrome-1; Long QT syndrome-2; Long QT syndrome-3; Long QT syndrome-5; Long QT syndrome-6; Lowe syndrome; Lung cancer; Lung cancer (nonsmall cell); Lung cancer (small cell); Lymphedema; Lymphoma (B-cell non- Hodgkin); Lymphoma (diffuse large cell); Lymphoma (follicular); Lymphoma (MALT); Lymphoma (mantel cell); Lymphoproliferative syndrome (X-linked); Lysinuric protein intolerance; Machado-Joseph disease; Macrocytic anemia refractory (of 5q syndrome); Macular dystrophy; Malignant mesothelioma; Malonyl- CoA decarboxylase deficiency ; Mannosidosis, (alpha- or beta- ); Maple syrup urine disease (type Ia; type Ib; type II); Marfan syndrome; Maroteaux-Lamy syndrome; Marshall syndrome; MASA syndrome; Mast cell leukemia; Mastocytosis with associated hematologic disorder; McArdle disease; McCune-Albright polyostotic fibrous dysplasia; McKusick-Kaufman syndrome; McLeod phenotype ; Medullary thyroid carcinoma; Medulloblastoma; Meesmann corneal dystrophy; Megaloblastic anemia-1; Melanoma; Membroproliferative glomerulonephritis ; Meniere disease; Meningioma (NF2-related; SIS-related); Menkes disease; Mental retardation (X- linked); Mephenytoin poor metabolizer; Mesothelioma; Metachromatic leukodystrophy; Metaphyseal chondrodysplasia (Murk Jansen type; Schmid type); Methemoglobinemia; Methionine adenosyltransferase deficiency (autosomal recessive); Methylcobalamin deficiency (cbl G type); Methylmalonicaciduria (mutase deficiency type); Mevalonicaciduria; MHC class Il deficiency; Microphthalmia (cataracts, and iris abnormalities); Miyoshi myopathy; MODY; Mohr-Tranebjaerg syndrome; Molybdenum cofactor deficiency (type A or type B); Monilethrix; Morbus Fabry; Morbus Gaucher; Mucopolysaccharidosis;
Mucoviscidosis; Muencke syndrome; Muir-Torre syndrome; Mulibrey nanism; Multiple carboxylase deficiency (biotinresponsive); Multiple endocrine neoplasia; Muscle glycogenosis; Muscular dystrophy (congenital merosindeficient); Muscular dystrophy (Fukuyama congenital); Muscular dystrophy (limb-girdle); Muscular dystrophy )Duchenne-like); Muscular dystrophy with epidermolysis bullosa simplex;
Myasthenic syndrome (slow-channel congenital); Mycobacterial infection (atypical, familial disseminated); Myelodysplastic syndrome; Myelogenous leukemia; Myeloid malignancy; Myeloperoxidase deficiency; Myoadenylate deaminase deficiency; Myoglobinuria/hemolysis due to PGK deficiency; Myoneurogastrointestinal encephalomyopathy syndrome; Myopathy (actin; congenital; desmin-related; cardioskeletal; distal; nemaline); Myopathy due to CPT Il deficiency; Myopathy due to phosphoglycerate mutase deficiency; Myotonia congenita; Myotonia levior; Myotonic dystrophy; Myxoid liposarcoma; NAGA deficiency; Nailpatella syndrome; Nemaline myopathy 1 (autosomal dominant); Nemaline myopathy 2 (autosomal recessive); Neonatal hyperparathyroidism; Nephrolithiasis; Nephronophthisis (juvenile); Nephropathy (chronic hypocomplementemic); Nephrosis-1; Nephrotic syndrome; Netherton syndrome; Neuroblastoma; Neurofibromatosis (type 1 or type 2); Neurolemmomatosis; neuronal-5 Ceroid-lipofuscinosis; Neuropathy;
Neutropenia (alloimmune neonatal); Niemann-Pick disease (type A; type B; type C1 ; type D); Night blindness (congenital stationary); Nijmegen breakage syndrome; Noncompaction of left ventricular myocardium; Nonepidermolytic palmoplantar keratoderma; Norrie disease; Norum disease; Nucleoside phosphorylase deficiency; Obesity; Occipital hornsyndrome; Ocular albinism (Nettleship-Falls type);
Oculopharyngeal muscular dystorphy; Oguchi disease; Oligodontia; Omenn syndrome; Opitz G syndrome; Optic nerve coloboma with renal disease; Ornithine transcarbamylase deficiency; Oroticaciduria; Orthostatic intolerance; OSMED syndrome; Ossification of posterior longitudinal ligament of spine; Osteoarthrosis; Osteogenesis imperfecta; Osteolysis; Osteopetrosis (recessive or idiopathic);
Osteosarcoma; Ovarian carcinoma; Ovarian dysgenesis; Pachyonychia congenita (Jackson-Lawler type or Jadassohn-Lewandowsky type); Paget disease of bone; Pallister-Hall syndrome; Pancreatic agenesis; Pancreatic cancer; Pancreatitis; Papillon-Lefevre syndrome; Paragangliomas ; Paramyotonia congenita; Parietal foramina; Parkinson disease (familial or juvenile); Paroxysmal nocturnal hemoglobinuria; Pelizaeus-Merzbacher disease; Pendred syndrome; Perineal hypospadias; Periodic fever; Peroxisomal biogenesis disorder; Persistent hyperinsulinemic hypoglycemia of infancy; Persistent Mullerian duct syndrome (type II); Peters anomaly; Peutz-Jeghers syndrome ; Pfeiffer syndrome; Phenylketonuria; Phosphoribosyl pyrophosphate synthetaserelated gout;
Phosphorylase kinase deficiency of liver and muscle; Piebaldism; Pilomatricoma; Pinealoma with bilateral retinoblastoma; Pituitary ACTH secreting adenoma; Pituitary hormone deficiency; Pituitary tumor; Placental steroid sulfatase deficiency; Plasmin inhibitor deficiency; Plasminogen deficiency (types I and II); Plasminogen Tochigi disease; Platelet disorder; Platelet glycoprotein IV deficiency; Platelet- activating factor acetylhydrolase deficiency; Polycystic kidney disease; Polycystic lipomembranous osteodysplasia with sclerosing leukenencephalophathy; Polydactyly, postaxial; Polyposis; Popliteal pterygium syndrome; Porphyria (acute hepatic or acute intermittent or congenital erythropoietic); Porphyria cutanea tarda;
Porphyria hepatoerythropoietic ; Porphyria variegata; Prader-Willi syndrome;
Precocious puberty; Premature ovarian failure; Progeria Typ I; Progeria Typ II;
Progressive external ophthalmoplegia; Progressive intrahepatic cholestasis-2; Prolactinoma (hyperparathyroidism, carcinoid syndrome); Prolidase deficiency;
Propionicacidemia; Prostate cancer; Protein S deficiency; Proteinuria;
Protoporphyria (erythropoietic); Pseudoachondroplasia; Pseudohermaphroditism;
Pseudohypoaldosteronism; Pseudohypoparathyroidism; Pseudovaginal perineoscrotal hypospadias; Pseudovitamin D deficiency rickets; Pseudoxanthoma elasticum (autosomal dominant; autosomal recessive); Pulmonary alveolar proteinosis; Pulmonary hypertension; Purpura fulminans; Pycnodysostosis;
Pyropoikilocytosis; Pyruvate carboxylase deficiency; Pyruvate dehydrogenase deficiency; Rabson-Mendenhall syndrome; Refsum disease; Renal cell carcinoma;
Renal tubular acidosis; Renal tubular acidosis with deafness; Renal tubular acidosis- osteopetrosis syndrome; Reticulosis (familial histiocytic); Retinal degeneration;
Retinal dystrophy; Retinitis pigmentosa; Retinitis punctata albescens;
Retinoblastoma; Retinol binding protein deficiency; Retinoschisis; Rett syndrome;
Rh(mod) syndrome; Rhabdoid predisposition syndrome; Rhabdoid tumors ;
Rhabdomyosarcoma; Rhabdomyosarcoma (alveolar); Rhizomelic chondrodysplasia punctata; Ribbing-Syndrom; Rickets (vitamin D-resistant); Rieger anomaly; Robinow syndrome; Rothmund-Thomson syndrome; Rubenstein-Taybi syndrome;
Saccharopinuria; Saethre-Chotzen syndrome; SaIIa disease; Sandhoff disease
(infantile, juvenile, and adult forms); Sanfilippo syndrome (type A or type B);
Schindler disease; Schizencephaly; Schizophrenia (chronic); Schwannoma (sporadic); SCID (autosomal recessive, T-negative/Bpositive type); Secretory pathway w/TMD; SED congenita; Segawa syndrome; Selective T-cell defect; SEMD (Pakistani type); SEMD (Strudwick type); Septooptic dysplasia; Severe combined immunodeficiency (B cellnegative); Severe combined immunodeficiency (T-cell negative, B-cell/natural killer cell-positive type); Severe combined immunodeficiency (Xlinked); Severe combined immunodeficiency due to ADA deficiency; Sex reversal (XY, with adrenal failure); Sezary syndrome; Shah-
Waardenburg syndrome; Short stature; Shprintzen-Goldberg syndrome; Sialic acid storage disorder; Sialidosis (type I or type II); Sialuria; Sickle cell anemia; Simpson- Golabi-Behmel syndrome; Situs ambiguus; Sjogren-Larsson syndrome; Smith- Fineman-Myers syndrome; Smith-Lemli-Opitz syndrome (type I or type II); Somatotroph i noma; Sorsby fundus dystrophy; Spastic paraplegia; Spherocytosis; Spherocytosis-1 ; Spherocytosis-2; Spinal and bulbar muscular atrophy of Kennedy; Spinal muscular atrophy; Spinocerebellar ataxia; Spondylocostal dysostosis;
Spondyloepiphyseal dysplasia tarda; Spondylometaphyseal dysplasia (Japanese type); Stargardt disease-1 ; Steatocystoma multiplex; Stickler syndrome; Sturge- Weber syndrom; Subcortical laminal heteropia; Subcortical laminar heterotopia; Succinic semialdehyde dehydrogenase deficiency; Sucrose intolerance; Sutherland- Haan syndrome; Sweat chloride elevation without CF; Symphalangism; Synostoses syndrome; Synpolydactyly; Tangier disease; Tay-Sachs disease; T-cell acute lymphoblastic leukemia; T-cell immunodeficiency; T-cell prolymphocyte leukemia; Thalassemia (alpha- or delta-); Thalassemia due to Hb Lepore; Thanatophoric dysplasia (types I or II); Thiamine-responsive megaloblastic anemia syndrome; Thrombocythemia; Thrombophilia (dysplasminogenemic); Thrombophilia due to heparin cofactor Il deficiency; Thrombophilia due to protein C deficiency; Thrombophilia due to thrombomodulin defect; Thyroid adenoma; Thyroid hormone resistance; Thyroid iodine peroxidase deficiency; Tietz syndrome; Tolbutamide poor metabolizer; Townes-Brocks syndrome; Transcobalamin Il deficiency; Treacher Collins mandibulofacial dysostosis; Trichodontoosseous syndrome;
Trichorhinophalangeal syndrome; Trichothiodystrophy; Trifunctional protein deficiency (type I or type II); Trypsinogen deficiency; Tuberous sclerosis-1; Tuberous sclerosis-2; Turcot syndrome; Tyrosine phosphatase; Tyrosinemia; Ulnar-mammary syndrome; Urolithiasis (2,8-dihydroxyadenine); Usher syndrome (type I B or type 2A); Venous malformations; Ventricular tachycardia; Virilization; Vitamin K- dependent coagulation defect; VLCAD deficiency; Vohwinkel syndrome; von Hippel-Lindau syndrome; von Willebrand disease; Waardenburg syndrome; Waardenburg syndrome/ocular albinism; Waardenburg-Shah neurologic variant; Waardenburg-Shah syndrome; Wagner syndrome; Warfarin sensitivity; Watson syndrome; Weissenbacher-Zweymuller syndrome; Werner syndrome; Weyers acrodental dysostosis; White sponge nevus; Williams-Beuren syndrome; Wilms tumor (typei ); Wilson disease; Wiskott-Aldrich syndrome; Wolcott-Rallison syndrome; Wolfram syndrome; Wolman disease; Xanthinuria (type I); Xeroderma pigmentosum; X-SCID; Yemenite deaf-blind hypopigmentation syndrome; ypocalciuric hypercalcemia (type I); Zellweger syndrome; and Zlotogora-Ogur syndrome.
28. Use according to claim 22, wherein the genetic diseases include autosomal- recessive inherited diseases including adenosine deaminase deficiency, familial hypercholesterolemia, Canavan's syndrome, Gaucher's disease, Fanconi anaemia, neuronal ceroid lipofuscinoses, mucoviscidosis (cystic fibrosis), sickle cell anaemia, phenylketonuria, alcaptonuria, albinism, hypothyreosis, galactosaemia, alpha-1 - anti-trypsin deficiency, Xeroderma pigmentosum, Ribbing's syndrome, mucopolysaccharidoses, cleft lip, jaw, palate, Laurence Moon Biedl Bardet sydrome, short rib polydactylia syndrome, cretinism, Joubert's syndrome, type Il progeria, brachydactylia, adrenogenital syndrome, and X-chromosome inherited diseases, including colour blindness, red/green blindness, fragile X syndrome, muscular dystrophy (Duchenne and Becker-Kiener type), haemophilia A and B,
G6PD deficiency, Fabry's disease, mucopolysaccharidosis, Nome's syndrome, Retinitis pigmentosa, septic granulomatosis, X-SCID, ornithine transcarbamylase deficiency, Lesch-Nyhan syndrome, or autosomal-dominant inherited diseases, including for example, hereditary angiooedema, Marfan syndrome, neurofibromatosis, type I progeria, Osteogenesis imperfecta, Klippel-Trenaurnay syndrome, Sturge-Weber syndrome, Hippel-Lindau syndrome and tuberosis sclerosis.
29. Use according to claim 22, wherein the treatment of a patient in need of a specific therapeutically active protein includes the treatment of dialysis patients.
30. Kit comprising the modified (m)RNA as defined in any of claims 1 to 18, and/or the immunosuppressive composition according to any of claims 1 to 18, and/or a pharmaceutical composition according to any of claims 19 to 21 , and optionally technical instructions with information on the administration and dosage of the
Immunosuppressive composition and/or the pharmaceutical composition.
31 . Modified (m)RNA containing the at least one modified (m)RNA as defined by claims 1 to 18 (as component of the immunosuppressive composition).
PCT/EP2008/003033 2008-04-16 2008-04-16 MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION WO2009127230A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/EP2008/003033 WO2009127230A1 (en) 2008-04-16 2008-04-16 MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2008/003033 WO2009127230A1 (en) 2008-04-16 2008-04-16 MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION

Publications (1)

Publication Number Publication Date
WO2009127230A1 true WO2009127230A1 (en) 2009-10-22

Family

ID=39739915

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/003033 WO2009127230A1 (en) 2008-04-16 2008-04-16 MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION

Country Status (1)

Country Link
WO (1) WO2009127230A1 (en)

Cited By (171)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011012316A3 (en) * 2009-07-31 2011-10-13 Ludwig-Maximilians-Universität Rna with a combination of unmodified and modified nucleotides for protein expression
WO2012045075A1 (en) * 2010-10-01 2012-04-05 Jason Schrum Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP2558571A2 (en) * 2010-04-16 2013-02-20 Immune Disease Institute Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof
CN102947450A (en) * 2009-12-07 2013-02-27 宾夕法尼亚州大学信托人 Rna preparations comprising purified modified rna for reprogramming cells
WO2013112053A1 (en) 2012-01-27 2013-08-01 Prosensa Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
WO2013151668A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of secreted proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013182683A1 (en) * 2012-06-08 2013-12-12 Ethris Gmbh Pulmonary delivery of messenger rna
EP2694660A1 (en) * 2011-04-03 2014-02-12 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
WO2014100714A1 (en) 2012-12-20 2014-06-26 Sarepta Therapeutics, Inc. Improved exon skipping compositions for treating muscular dystrophy
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2014153240A2 (en) 2013-03-14 2014-09-25 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
WO2014153220A2 (en) 2013-03-14 2014-09-25 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
WO2014154844A1 (en) * 2013-03-28 2014-10-02 Eberhard Karls Universitaet Tuebingen Medizinische Fakultaet Stabilized polyribonucleotide coding for an elastic fibrous protein
US8853377B2 (en) 2010-11-30 2014-10-07 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
US8871737B2 (en) 2010-09-22 2014-10-28 Alios Biopharma, Inc. Substituted nucleotide analogs
US8916538B2 (en) 2012-03-21 2014-12-23 Vertex Pharmaceuticals Incorporated Solid forms of a thiophosphoramidate nucleotide prodrug
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US8980865B2 (en) 2011-12-22 2015-03-17 Alios Biopharma, Inc. Substituted nucleotide analogs
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP1685844B1 (en) 2002-07-03 2015-03-18 CureVac GmbH Immunostimulation by chemically modified RNA
US9012427B2 (en) 2012-03-22 2015-04-21 Alios Biopharma, Inc. Pharmaceutical combinations comprising a thionucleotide analog
WO2015062738A1 (en) * 2013-11-01 2015-05-07 Curevac Gmbh Modified rna with decreased immunostimulatory properties
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9163213B2 (en) 2005-08-23 2015-10-20 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
EP2590626B1 (en) 2010-07-06 2015-10-28 GlaxoSmithKline Biologicals SA Liposomes with lipids having an advantageous pka-value for rna delivery
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
EP2931319A4 (en) * 2012-12-13 2016-05-04 Moderna Therapeutics Inc Modified nucleic acid molecules and uses thereof
US9371511B2 (en) 2005-08-23 2016-06-21 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US9376669B2 (en) 2012-11-01 2016-06-28 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US9422577B2 (en) 2011-12-05 2016-08-23 Factor Bioscience Inc. Methods and products for transfecting cells
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US9555122B2 (en) 2009-12-21 2017-01-31 Ethris Gmbh Conjugate with target-finding ligand and use thereof
WO2017022650A1 (en) * 2015-07-31 2017-02-09 株式会社ジーンケア研究所 MODIFIED siRNA, AND PHARMACEUTICAL COMPOSITION CONTAINING SAME
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2017049245A2 (en) 2015-09-17 2017-03-23 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2017066781A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified phosphate linkage
WO2017066791A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Sugar substituted mrna cap analogs
WO2017066793A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs and methods of mrna capping
WO2017066789A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified sugar
WO2017066782A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Hydrophobic mrna cap analogs
US9629804B2 (en) 2013-10-22 2017-04-25 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger RNA
WO2017112865A1 (en) 2015-12-22 2017-06-29 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US9770489B2 (en) 2014-01-31 2017-09-26 Factor Bioscience Inc. Methods and products for nucleic acid production and delivery
WO2017180587A2 (en) 2016-04-11 2017-10-19 Obsidian Therapeutics, Inc. Regulated biocircuit systems
WO2017218704A1 (en) 2016-06-14 2017-12-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
WO2018005805A1 (en) 2016-06-30 2018-01-04 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
AU2015215938B2 (en) * 2009-12-07 2018-01-18 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
WO2017191274A3 (en) * 2016-05-04 2018-02-08 Curevac Ag Rna encoding a therapeutic protein
US20180086816A1 (en) * 2013-03-13 2018-03-29 Modernatx, Inc. Long-lived polynucleotide molecules
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018118627A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2018118599A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2018118662A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US10022455B2 (en) 2014-05-30 2018-07-17 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10023626B2 (en) 2013-09-30 2018-07-17 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2018170336A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Lipid nanoparticle formulation
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
WO2018208980A1 (en) * 2017-05-10 2018-11-15 Oric Pharmaceuticals, Inc. Cd73 inhibitors
US10138507B2 (en) 2013-03-15 2018-11-27 Modernatx, Inc. Manufacturing methods for production of RNA transcripts
US10138213B2 (en) 2014-06-24 2018-11-27 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
US10137206B2 (en) 2016-08-17 2018-11-27 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10143758B2 (en) 2009-12-01 2018-12-04 Translate Bio, Inc. Liver specific delivery of messenger RNA
WO2018232120A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
US10201620B2 (en) 2011-12-30 2019-02-12 Cellscript, Llc Making and using in vitro-synthesized ssRNA for introducing into mammalian cells to induce a biological or biochemical effect
WO2019036638A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods of preparing modified rna
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
WO2019059973A1 (en) 2017-09-22 2019-03-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2019067979A1 (en) 2017-09-28 2019-04-04 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
WO2019067975A1 (en) 2017-09-28 2019-04-04 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
WO2019067981A1 (en) 2017-09-28 2019-04-04 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US10266843B2 (en) * 2016-04-08 2019-04-23 Translate Bio, Inc. Multimeric coding nucleic acid and uses thereof
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10385088B2 (en) 2013-10-02 2019-08-20 Modernatx, Inc. Polynucleotide molecules and uses thereof
WO2019158583A1 (en) * 2018-02-13 2019-08-22 Ethris Gmbh Polyribonucleotide containing deuterated nucleotides
US10407683B2 (en) 2014-07-16 2019-09-10 Modernatx, Inc. Circular polynucleotides
EP3542802A1 (en) * 2013-11-01 2019-09-25 CureVac AG Modified rna with decreased immunostimulatory properties
CN110327982A (en) * 2019-07-03 2019-10-15 燕山大学 It is a kind of using PEI- Ciprofloxacin micella as Pd nano particle of template and preparation method thereof
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
WO2019241315A1 (en) 2018-06-12 2019-12-19 Obsidian Therapeutics, Inc. Pde5 derived regulatory constructs and methods of use in immunotherapy
WO2020023688A1 (en) 2018-07-27 2020-01-30 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
US10590161B2 (en) 2013-03-15 2020-03-17 Modernatx, Inc. Ion exchange purification of mRNA
WO2020061457A1 (en) 2018-09-20 2020-03-26 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof
WO2020061367A1 (en) 2018-09-19 2020-03-26 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2020086742A1 (en) 2018-10-24 2020-04-30 Obsidian Therapeutics, Inc. Er tunable protein regulation
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
WO2020123574A1 (en) 2018-12-13 2020-06-18 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US10730924B2 (en) 2016-05-18 2020-08-04 Modernatx, Inc. Polynucleotides encoding relaxin
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
US10758629B2 (en) 2018-05-29 2020-09-01 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2020214763A1 (en) 2019-04-18 2020-10-22 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US10849920B2 (en) 2015-10-05 2020-12-01 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
WO2021079002A2 (en) 2019-10-24 2021-04-29 Novago Therapeutics Ag Novel anti-nogo-a antibodies
US11027025B2 (en) 2013-07-11 2021-06-08 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
US11028120B2 (en) 2019-10-30 2021-06-08 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
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
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
WO2021245184A1 (en) 2020-06-02 2021-12-09 Neurimmune Ag HUMAN ANTIBODIES AGAINST SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS-2 (SARS-CoV-2)
WO2022002040A1 (en) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
CN113995833A (en) * 2021-11-10 2022-02-01 南京理工大学 Application of adenosine deaminase and modifier thereof in preparation of diabetic wound repair drugs
US11241505B2 (en) 2015-02-13 2022-02-08 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
WO2022037652A1 (en) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11325938B2 (en) 2018-04-30 2022-05-10 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
US11377469B2 (en) 2017-11-03 2022-07-05 Oric Pharmaceuticals, Inc. CD73 inhibitors
WO2022152109A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022152141A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Polymer conjugated lipid compounds and lipid nanoparticle compositions
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
US11459563B2 (en) 2017-02-03 2022-10-04 The University Of Western Australia Treatment for NEAT1 associated disease
WO2022232478A1 (en) 2021-04-30 2022-11-03 Sarepta Therapeutics, Inc. Treatment methods for muscular dystrophy
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
WO2022247755A1 (en) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
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
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
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
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11667910B2 (en) 2015-05-29 2023-06-06 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
WO2023122752A1 (en) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Constrained lipids and methods of use thereof
EP4219717A2 (en) 2018-06-13 2023-08-02 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
US11739125B2 (en) 2013-08-21 2023-08-29 Cure Vac SE Respiratory syncytial virus (RSV) vaccine
US11761009B2 (en) 2014-12-12 2023-09-19 CureVac SE Artificial nucleic acid molecules for improved protein expression
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
WO2023178230A1 (en) 2022-03-17 2023-09-21 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer conjugates
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
US11786590B2 (en) 2015-11-09 2023-10-17 CureVac SE Rotavirus vaccines
US11859215B2 (en) 2017-11-22 2024-01-02 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
US11866754B2 (en) 2015-10-16 2024-01-09 Modernatx, Inc. Trinucleotide mRNA cap analogs
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
WO2024037578A1 (en) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition of lipid nanoparticles
US11939601B2 (en) 2017-11-22 2024-03-26 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
US11951179B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1604688A1 (en) * 2001-06-05 2005-12-14 CureVac GmbH Stabilised mRNA with increased G/C-content and optimised codon usage for gene therapy
WO2007024708A2 (en) * 2005-08-23 2007-03-01 The Trustees Of The University Of Pennsylvania Rna containing modified nucleosides and methods of use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1604688A1 (en) * 2001-06-05 2005-12-14 CureVac GmbH Stabilised mRNA with increased G/C-content and optimised codon usage for gene therapy
WO2007024708A2 (en) * 2005-08-23 2007-03-01 The Trustees Of The University Of Pennsylvania Rna containing modified nucleosides and methods of use thereof

Cited By (438)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1685844B1 (en) 2002-07-03 2015-03-18 CureVac GmbH Immunostimulation by chemically modified RNA
US9163213B2 (en) 2005-08-23 2015-10-20 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US9371511B2 (en) 2005-08-23 2016-06-21 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US10745698B2 (en) 2009-07-31 2020-08-18 Ethris Gmbh RNA with a combination of unmodified and modified nucleotides for protein expression
US20150290288A1 (en) * 2009-07-31 2015-10-15 Ethris Gmbh Rna with a combination of unmodified and modified nucleotides for protein expression
WO2011012316A3 (en) * 2009-07-31 2011-10-13 Ludwig-Maximilians-Universität Rna with a combination of unmodified and modified nucleotides for protein expression
EP3581197A1 (en) * 2009-07-31 2019-12-18 ethris GmbH Rna with a combination of unmodified and modified nucleotides for protein expression
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10143758B2 (en) 2009-12-01 2018-12-04 Translate Bio, Inc. Liver specific delivery of messenger RNA
JP2016171804A (en) * 2009-12-07 2016-09-29 ザ トラスティース オブ ザ ユニバーシティ オブ ペンシルベニア Rna preparations for cell reprogramming comprising purified modified rna
CN107090436A (en) * 2009-12-07 2017-08-25 宾夕法尼亚州大学信托人 RNA preparations for the RNA through modification comprising purifying of reprogrammed cell
AU2018202479B2 (en) * 2009-12-07 2020-09-10 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US11028370B2 (en) 2009-12-07 2021-06-08 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
AU2015215938B2 (en) * 2009-12-07 2018-01-18 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
CN107090436B (en) * 2009-12-07 2021-11-30 宾夕法尼亚州大学信托人 RNA preparation comprising purified modified RNA for reprogramming cells
JP2019162125A (en) * 2009-12-07 2019-09-26 ザ トラスティース オブ ザ ユニバーシティ オブ ペンシルベニア Rna preparations comprising purified modified rna for reprogramming cells
EP2510099A4 (en) * 2009-12-07 2014-06-11 Univ Pennsylvania Rna preparations comprising purified modified rna for reprogramming cells
US10006007B2 (en) 2009-12-07 2018-06-26 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
EP3287525A1 (en) * 2009-12-07 2018-02-28 The Trustees of The University of Pennsylvania Rna preparations comprising purified modified rna for reprogramming cells
CN102947450A (en) * 2009-12-07 2013-02-27 宾夕法尼亚州大学信托人 Rna preparations comprising purified modified rna for reprogramming cells
JP2013512690A (en) * 2009-12-07 2013-04-18 ザ トラスティース オブ ザ ユニバーシティ オブ ペンシルベニア RNA preparation comprising purified modified RNA for reprogramming cells
CN102947450B (en) * 2009-12-07 2016-11-23 宾夕法尼亚州大学信托人 RNA preparation for the modified RNA comprising purification of reprogrammed cell
EP3623474A1 (en) * 2009-12-07 2020-03-18 The Trustees of The University of Pennsylvania Rna preparations comprising purified modified rna for reprogramming cells
US9371544B2 (en) 2009-12-07 2016-06-21 The Trustees Of The University Of Pennsylvania Compositions and methods for reprogramming eukaryotic cells
CN114317612A (en) * 2009-12-07 2022-04-12 宾夕法尼亚州大学信托人 RNA preparations comprising purified modified RNA for reprogramming cells
EP3112467A1 (en) * 2009-12-07 2017-01-04 The Trustees of the University of Pennsylvania Rna preparations comprising purified modified rna for reprogramming cells
US11739300B2 (en) 2009-12-07 2023-08-29 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
US9555122B2 (en) 2009-12-21 2017-01-31 Ethris Gmbh Conjugate with target-finding ligand and use thereof
US10080804B2 (en) 2009-12-21 2018-09-25 Ethris Gmbh Conjugate with target-finding ligand and use thereof
US10583195B2 (en) 2009-12-21 2020-03-10 Ethris Gmbh Conjugate with target-finding ligand and use thereof
US10344265B2 (en) 2010-04-16 2019-07-09 Children's Medical Center Corporation Sustained polypeptide expression from synthetic, modified RNAs and uses thereof
EP2558571A4 (en) * 2010-04-16 2014-09-24 Immune Disease Inst Inc Sustained polypeptide expression from synthetic, modified rnas and uses thereof
US9803177B2 (en) 2010-04-16 2017-10-31 Children's Medical Center Corporation Induced pluripotent stem cells with synthetic modified RNAs
EP3072961A1 (en) * 2010-04-16 2016-09-28 Children's Medical Center Corporation Sustained polypeptide expression from synthetic, modified rnas and uses thereof
EP2558571A2 (en) * 2010-04-16 2013-02-20 Immune Disease Institute Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof
US11186829B2 (en) 2010-04-16 2021-11-30 Children's Medical Center Corporation Isolated mammalian somatic cells containing modified RNA encoding OCT4, SOX2, and KLF4
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11690861B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690864B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690863B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11865080B2 (en) 2010-07-06 2024-01-09 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638693B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising RNA encoding an immunogen and lipid formulations comprising mole percentage of lipids
EP2590626B1 (en) 2010-07-06 2015-10-28 GlaxoSmithKline Biologicals SA Liposomes with lipids having an advantageous pka-value for rna delivery
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11690862B1 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11730754B2 (en) 2010-07-06 2023-08-22 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11845925B2 (en) 2010-07-06 2023-12-19 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11324770B2 (en) 2010-07-06 2022-05-10 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11759475B2 (en) 2010-07-06 2023-09-19 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11850305B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Method of making lipid formulations with RNA encoding immunogens
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
US11851660B2 (en) 2010-07-06 2023-12-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US8871737B2 (en) 2010-09-22 2014-10-28 Alios Biopharma, Inc. Substituted nucleotide analogs
US9278990B2 (en) 2010-09-22 2016-03-08 Alios Biopharma, Inc. Substituted nucleotide analogs
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US20130203115A1 (en) * 2010-10-01 2013-08-08 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) * 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2012045075A1 (en) * 2010-10-01 2012-04-05 Jason Schrum Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US8853377B2 (en) 2010-11-30 2014-10-07 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
US9061021B2 (en) 2010-11-30 2015-06-23 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
US11135274B2 (en) 2010-11-30 2021-10-05 Translate Bio, Inc. MRNA for use in treatment of human genetic diseases
US9956271B2 (en) 2010-11-30 2018-05-01 Translate Bio, Inc. mRNA for use in treatment of human genetic diseases
US10898574B2 (en) 2011-03-31 2021-01-26 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US11911474B2 (en) 2011-03-31 2024-02-27 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
EP3460064A1 (en) * 2011-04-03 2019-03-27 The General Hospital Corporation d/b/a Massachusetts General Hospital Efficient protein expression in vivo using modified rna (mod-rna)
EP2694660A4 (en) * 2011-04-03 2014-11-19 Gen Hospital Corp Efficient protein expression in vivo using modified rna (mod-rna)
EP2694660A1 (en) * 2011-04-03 2014-02-12 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
US10086043B2 (en) 2011-04-03 2018-10-02 The General Hospital Corporation Efficient protein expression in vivo using modified RNA (MOD-RNA)
JP2019147811A (en) * 2011-04-03 2019-09-05 ザ ジェネラル ホスピタル コーポレーション ドゥーイング ビジネス アズ マサチューセッツ ジェネラル ホスピタル Efficient protein expression in vivo using modified rna (mod-rna)
US10413618B2 (en) 2011-06-08 2019-09-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10888626B2 (en) 2011-06-08 2021-01-12 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US9597413B2 (en) 2011-06-08 2017-03-21 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mRNA
US11185595B2 (en) 2011-06-08 2021-11-30 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US11291734B2 (en) 2011-06-08 2022-04-05 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11338044B2 (en) 2011-06-08 2022-05-24 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10507249B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10238754B2 (en) 2011-06-08 2019-03-26 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11052159B2 (en) 2011-06-08 2021-07-06 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10350303B1 (en) 2011-06-08 2019-07-16 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9605278B2 (en) 2011-12-05 2017-03-28 Factor Bioscience Inc. Methods and products for transfecting cells
US10662410B1 (en) 2011-12-05 2020-05-26 Factor Bioscience Inc. Methods and products for transfecting cells
US9422577B2 (en) 2011-12-05 2016-08-23 Factor Bioscience Inc. Methods and products for transfecting cells
US11466293B2 (en) 2011-12-05 2022-10-11 Factor Bioscience Inc. Methods and products for transfecting cells
US10472611B2 (en) 2011-12-05 2019-11-12 Factor Bioscience Inc. Methods and products for transfecting cells
US9605277B2 (en) 2011-12-05 2017-03-28 Factor Bioscience, Inc. Methods and products for transfecting cells
US11708586B2 (en) 2011-12-05 2023-07-25 Factor Bioscience Inc. Methods and products for transfecting cells
US10982229B2 (en) 2011-12-05 2021-04-20 Factor Bioscience Inc. Methods and products for transfecting cells
US10829738B2 (en) 2011-12-05 2020-11-10 Factor Bioscience Inc. Methods and products for transfecting cells
US11692203B2 (en) 2011-12-05 2023-07-04 Factor Bioscience Inc. Methods and products for transfecting cells
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
RU2649364C2 (en) * 2011-12-16 2018-04-02 Модерна Терапьютикс, Инк. Modified nucleoside, nucleotide and nucleic acid compositions
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8980865B2 (en) 2011-12-22 2015-03-17 Alios Biopharma, Inc. Substituted nucleotide analogs
US9605018B2 (en) 2011-12-22 2017-03-28 Alios Biopharma, Inc. Substituted nucleotide analogs
US10201620B2 (en) 2011-12-30 2019-02-12 Cellscript, Llc Making and using in vitro-synthesized ssRNA for introducing into mammalian cells to induce a biological or biochemical effect
US11135314B2 (en) 2011-12-30 2021-10-05 Cellscript, Llc Making and using in vitro-synthesized ssRNA for introducing into mammalian cells to induce a biological or biochemical effect
WO2013112053A1 (en) 2012-01-27 2013-08-01 Prosensa Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
EP4043039A1 (en) 2012-01-27 2022-08-17 BioMarin Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
US9394330B2 (en) 2012-03-21 2016-07-19 Alios Biopharma, Inc. Solid forms of a thiophosphoramidate nucleotide prodrug
US9856284B2 (en) 2012-03-21 2018-01-02 Alios Biopharma, Inc. Solid forms of a thiophosphoramidate nucleotide prodrug
US8916538B2 (en) 2012-03-21 2014-12-23 Vertex Pharmaceuticals Incorporated Solid forms of a thiophosphoramidate nucleotide prodrug
US9012427B2 (en) 2012-03-22 2015-04-21 Alios Biopharma, Inc. Pharmaceutical combinations comprising a thionucleotide analog
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US10385106B2 (en) 2012-04-02 2019-08-20 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
EP3978030A1 (en) 2012-04-02 2022-04-06 ModernaTX, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US11564998B2 (en) 2012-04-02 2023-01-31 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US10772975B2 (en) 2012-04-02 2020-09-15 Modernatx, Inc. Modified Polynucleotides for the production of biologics and proteins associated with human disease
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
US10703789B2 (en) 2012-04-02 2020-07-07 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2013151665A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of proteins associated with human disease
US10577403B2 (en) 2012-04-02 2020-03-03 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
WO2013151668A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of secreted proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
EP3501550A1 (en) 2012-04-02 2019-06-26 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
WO2013182683A1 (en) * 2012-06-08 2013-12-12 Ethris Gmbh Pulmonary delivery of messenger rna
EA037448B1 (en) * 2012-06-08 2021-03-30 Этрис Гмбх METHOD FOR EXPRESSING A THERAPEUTIC mRNA WITH PEI IN LUNG AND PHARMACEUTICAL COMPOSITION
EP3800254A1 (en) * 2012-06-08 2021-04-07 Ethris GmbH Pulmonary delivery of messenger rna
AU2013273504B2 (en) * 2012-06-08 2017-12-07 Ethris Gmbh Pulmonary delivery of messenger RNA
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
JP2015518874A (en) * 2012-06-08 2015-07-06 エスリス ゲーエムベーハーethris GmbH Pulmonary delivery of messenger RNA
US9512456B2 (en) 2012-08-14 2016-12-06 Modernatx, Inc. Enzymes and polymerases for the synthesis of RNA
US9758797B2 (en) 2012-11-01 2017-09-12 Factor Bioscience, Inc. Methods and products for expressing proteins in cells
US9464285B2 (en) 2012-11-01 2016-10-11 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US10724053B2 (en) 2012-11-01 2020-07-28 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US9657282B2 (en) 2012-11-01 2017-05-23 Factor Bioscience, Inc. Methods and products for expressing proteins in cells
US10415060B2 (en) 2012-11-01 2019-09-17 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US10752917B2 (en) 2012-11-01 2020-08-25 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US10752918B2 (en) 2012-11-01 2020-08-25 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US10752919B2 (en) 2012-11-01 2020-08-25 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US10767195B2 (en) 2012-11-01 2020-09-08 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US10590437B2 (en) 2012-11-01 2020-03-17 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US11339409B2 (en) 2012-11-01 2022-05-24 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US11339410B2 (en) 2012-11-01 2022-05-24 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US9487768B2 (en) 2012-11-01 2016-11-08 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US11332758B2 (en) 2012-11-01 2022-05-17 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US11332759B2 (en) 2012-11-01 2022-05-17 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US9447395B2 (en) 2012-11-01 2016-09-20 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US9376669B2 (en) 2012-11-01 2016-06-28 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
EP2931319A4 (en) * 2012-12-13 2016-05-04 Moderna Therapeutics Inc Modified nucleic acid molecules and uses thereof
WO2014100714A1 (en) 2012-12-20 2014-06-26 Sarepta Therapeutics, Inc. Improved exon skipping compositions for treating muscular dystrophy
EP3885439A1 (en) 2012-12-20 2021-09-29 Sarepta Therapeutics, Inc. Improved exon skipping compositions for treating muscular dystrophy
US20180086816A1 (en) * 2013-03-13 2018-03-29 Modernatx, Inc. Long-lived polynucleotide molecules
US11603399B2 (en) * 2013-03-13 2023-03-14 Modernatx, Inc. Long-lived polynucleotide molecules
US10420791B2 (en) 2013-03-14 2019-09-24 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
US11820977B2 (en) 2013-03-14 2023-11-21 Translate Bio, Inc. Methods for purification of messenger RNA
EP3495485A2 (en) 2013-03-14 2019-06-12 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US11692189B2 (en) 2013-03-14 2023-07-04 Translate Bio, Inc. Methods for purification of messenger RNA
US11510937B2 (en) 2013-03-14 2022-11-29 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US9713626B2 (en) 2013-03-14 2017-07-25 Rana Therapeutics, Inc. CFTR mRNA compositions and related methods and uses
US10876104B2 (en) 2013-03-14 2020-12-29 Translate Bio, Inc. Methods for purification of messenger RNA
WO2014153220A2 (en) 2013-03-14 2014-09-25 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
EP3760720A1 (en) 2013-03-14 2021-01-06 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
EP3998339A1 (en) 2013-03-14 2022-05-18 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
WO2014153240A2 (en) 2013-03-14 2014-09-25 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
EP3633035A1 (en) 2013-03-14 2020-04-08 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US10138507B2 (en) 2013-03-15 2018-11-27 Modernatx, Inc. Manufacturing methods for production of RNA transcripts
US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
US10590161B2 (en) 2013-03-15 2020-03-17 Modernatx, Inc. Ion exchange purification of mRNA
US11845772B2 (en) 2013-03-15 2023-12-19 Modernatx, Inc. Ribonucleic acid purification
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US10858647B2 (en) 2013-03-15 2020-12-08 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2014154844A1 (en) * 2013-03-28 2014-10-02 Eberhard Karls Universitaet Tuebingen Medizinische Fakultaet Stabilized polyribonucleotide coding for an elastic fibrous protein
US11027025B2 (en) 2013-07-11 2021-06-08 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
US11739125B2 (en) 2013-08-21 2023-08-29 Cure Vac SE Respiratory syncytial virus (RSV) vaccine
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10023626B2 (en) 2013-09-30 2018-07-17 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10385088B2 (en) 2013-10-02 2019-08-20 Modernatx, Inc. Polynucleotide molecules and uses thereof
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US11377642B2 (en) 2013-10-22 2022-07-05 Translate Bio, Inc. mRNA therapy for phenylketonuria
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US10959953B2 (en) 2013-10-22 2021-03-30 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US11890377B2 (en) 2013-10-22 2024-02-06 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US10052284B2 (en) 2013-10-22 2018-08-21 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US10493031B2 (en) 2013-10-22 2019-12-03 Translate Bio, Inc. Lipid formulations for delivery of messenger RNA
US9629804B2 (en) 2013-10-22 2017-04-25 Shire Human Genetic Therapies, Inc. Lipid formulations for delivery of messenger RNA
US10208295B2 (en) 2013-10-22 2019-02-19 Translate Bio, Inc. MRNA therapy for phenylketonuria
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US20160235864A1 (en) * 2013-11-01 2016-08-18 Curevac Ag Modified rna with decreased immunostimulatory properties
WO2015062738A1 (en) * 2013-11-01 2015-05-07 Curevac Gmbh Modified rna with decreased immunostimulatory properties
EP3062798B1 (en) 2013-11-01 2020-05-06 CureVac AG Modified rna with decreased immunostimulatory properties
EP3542802A1 (en) * 2013-11-01 2019-09-25 CureVac AG Modified rna with decreased immunostimulatory properties
US10898584B2 (en) 2013-11-01 2021-01-26 Curevac Ag Modified RNA with decreased immunostimulatory properties
US10124042B2 (en) 2014-01-31 2018-11-13 Factor Bioscience Inc. Methods and products for nucleic acid production and delivery
US9770489B2 (en) 2014-01-31 2017-09-26 Factor Bioscience Inc. Methods and products for nucleic acid production and delivery
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
US11884692B2 (en) 2014-04-25 2024-01-30 Translate Bio, Inc. Methods for purification of messenger RNA
US11059841B2 (en) 2014-04-25 2021-07-13 Translate Bio, Inc. Methods for purification of messenger RNA
US10155785B2 (en) 2014-04-25 2018-12-18 Translate Bio, Inc. Methods for purification of messenger RNA
US10286082B2 (en) 2014-05-30 2019-05-14 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10493166B2 (en) 2014-05-30 2019-12-03 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10022455B2 (en) 2014-05-30 2018-07-17 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10286083B2 (en) 2014-05-30 2019-05-14 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10293057B2 (en) 2014-05-30 2019-05-21 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US11433144B2 (en) 2014-05-30 2022-09-06 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10912844B2 (en) 2014-05-30 2021-02-09 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10286086B2 (en) 2014-06-19 2019-05-14 Modernatx, Inc. Alternative nucleic acid molecules and uses thereof
US10138213B2 (en) 2014-06-24 2018-11-27 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
US11104652B2 (en) 2014-06-24 2021-08-31 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
US10407683B2 (en) 2014-07-16 2019-09-10 Modernatx, Inc. Circular polynucleotides
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
US11761009B2 (en) 2014-12-12 2023-09-19 CureVac SE Artificial nucleic acid molecules for improved protein expression
US11241505B2 (en) 2015-02-13 2022-02-08 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11667910B2 (en) 2015-05-29 2023-06-06 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US11760992B2 (en) 2015-05-29 2023-09-19 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of 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
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US10702597B2 (en) 2015-07-21 2020-07-07 Modernatx, Inc. CHIKV RNA vaccines
WO2017022650A1 (en) * 2015-07-31 2017-02-09 株式会社ジーンケア研究所 MODIFIED siRNA, AND PHARMACEUTICAL COMPOSITION CONTAINING SAME
US10548990B2 (en) 2015-07-31 2020-02-04 Genecare Research Institute Co., Ltd. Modified siRNA and pharmaceutical composition
US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
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
US11590157B2 (en) 2015-10-05 2023-02-28 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
US10849920B2 (en) 2015-10-05 2020-12-01 Modernatx, Inc. Methods for therapeutic administration of messenger ribonucleic acid drugs
WO2017066781A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs with modified phosphate linkage
US10570388B2 (en) 2015-10-16 2020-02-25 Modernatx, Inc. Phosphate replacement 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
US11866754B2 (en) 2015-10-16 2024-01-09 Modernatx, Inc. Trinucleotide mRNA cap analogs
US10563195B2 (en) 2015-10-16 2020-02-18 Modernatx, Inc. Phosphate replacement mRNA cap analogs
US10428106B2 (en) 2015-10-16 2019-10-01 Modernatx, Inc. Phosphate replacement mRNA cap analogs
WO2017066791A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Sugar substituted mrna cap analogs
EP4086269A1 (en) 2015-10-16 2022-11-09 ModernaTX, Inc. Mrna cap analogs with modified phosphate linkage
WO2017066793A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Mrna cap analogs and methods of mrna capping
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10702599B2 (en) 2015-10-22 2020-07-07 Modernatx, Inc. HPIV3 RNA vaccines
US10702600B1 (en) 2015-10-22 2020-07-07 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10716846B2 (en) 2015-10-22 2020-07-21 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10543269B2 (en) 2015-10-22 2020-01-28 Modernatx, Inc. hMPV RNA vaccines
US10675342B2 (en) 2015-10-22 2020-06-09 Modernatx, Inc. Chikungunya virus RNA vaccines
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
EP3718565B1 (en) 2015-10-22 2022-04-27 ModernaTX, Inc. Respiratory virus vaccines
US10517940B2 (en) 2015-10-22 2019-12-31 Modernatx, Inc. Zika virus RNA vaccines
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10238731B2 (en) 2015-10-22 2019-03-26 Modernatx, Inc. Chikagunya virus RNA vaccines
US10272150B2 (en) 2015-10-22 2019-04-30 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US11786590B2 (en) 2015-11-09 2023-10-17 CureVac SE Rotavirus vaccines
WO2017112865A1 (en) 2015-12-22 2017-06-29 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
EP4036079A2 (en) 2015-12-22 2022-08-03 ModernaTX, Inc. Compounds and compositions for intracellular delivery of agents
US11124804B2 (en) 2016-04-08 2021-09-21 Translate Bio, Inc. Multimeric coding nucleic acid and uses thereof
US10428349B2 (en) 2016-04-08 2019-10-01 Translate Bio, Inc. Multimeric coding nucleic acid and uses thereof
US10266843B2 (en) * 2016-04-08 2019-04-23 Translate Bio, Inc. Multimeric coding nucleic acid and uses thereof
WO2017180587A2 (en) 2016-04-11 2017-10-19 Obsidian Therapeutics, Inc. Regulated biocircuit systems
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
WO2017191274A3 (en) * 2016-05-04 2018-02-08 Curevac Ag Rna encoding a therapeutic protein
US10730924B2 (en) 2016-05-18 2020-08-04 Modernatx, Inc. Polynucleotides encoding relaxin
WO2017218704A1 (en) 2016-06-14 2017-12-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
WO2018005805A1 (en) 2016-06-30 2018-01-04 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
US10363321B2 (en) 2016-08-17 2019-07-30 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10350304B2 (en) 2016-08-17 2019-07-16 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10576167B2 (en) 2016-08-17 2020-03-03 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10369233B2 (en) 2016-08-17 2019-08-06 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10894092B2 (en) 2016-08-17 2021-01-19 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US11904023B2 (en) 2016-08-17 2024-02-20 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10888627B2 (en) 2016-08-17 2021-01-12 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US10137206B2 (en) 2016-08-17 2018-11-27 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
US11541113B2 (en) 2016-10-21 2023-01-03 Modernatx, Inc. Human cytomegalovirus vaccine
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11642364B2 (en) 2016-12-19 2023-05-09 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US11395855B2 (en) 2016-12-19 2022-07-26 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US10888578B2 (en) 2016-12-19 2021-01-12 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US11000600B2 (en) 2016-12-19 2021-05-11 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US11382981B2 (en) 2016-12-19 2022-07-12 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2018118662A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
EP4115908A1 (en) 2016-12-19 2023-01-11 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
EP4122497A1 (en) 2016-12-19 2023-01-25 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2018118599A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2018118627A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
US11459563B2 (en) 2017-02-03 2022-10-04 The University Of Western Australia Treatment for NEAT1 associated disease
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
EP4186888A1 (en) 2017-03-15 2023-05-31 ModernaTX, Inc. Compound and compositions for intracellular delivery of therapeutic agents
WO2018170306A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
WO2018170336A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Lipid nanoparticle formulation
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
US11576922B2 (en) 2017-05-10 2023-02-14 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11129841B2 (en) 2017-05-10 2021-09-28 Oric Pharmaceuticals, Inc. CD73 inhibitors
WO2018208980A1 (en) * 2017-05-10 2018-11-15 Oric Pharmaceuticals, Inc. Cd73 inhibitors
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
WO2018232120A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
WO2019036638A1 (en) 2017-08-18 2019-02-21 Modernatx, Inc. Methods of preparing modified rna
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
WO2019059973A1 (en) 2017-09-22 2019-03-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2019067979A1 (en) 2017-09-28 2019-04-04 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
WO2019067981A1 (en) 2017-09-28 2019-04-04 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
WO2019067975A1 (en) 2017-09-28 2019-04-04 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
US11377469B2 (en) 2017-11-03 2022-07-05 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11859215B2 (en) 2017-11-22 2024-01-02 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
US11939601B2 (en) 2017-11-22 2024-03-26 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019158583A1 (en) * 2018-02-13 2019-08-22 Ethris Gmbh Polyribonucleotide containing deuterated nucleotides
US11325938B2 (en) 2018-04-30 2022-05-10 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11807658B2 (en) 2018-04-30 2023-11-07 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11338041B2 (en) 2018-05-29 2022-05-24 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US10765760B2 (en) 2018-05-29 2020-09-08 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US11491238B2 (en) 2018-05-29 2022-11-08 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
US10758629B2 (en) 2018-05-29 2020-09-01 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
WO2019241315A1 (en) 2018-06-12 2019-12-19 Obsidian Therapeutics, Inc. Pde5 derived regulatory constructs and methods of use in immunotherapy
EP4219717A2 (en) 2018-06-13 2023-08-02 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
WO2020023688A1 (en) 2018-07-27 2020-01-30 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
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
WO2020086742A1 (en) 2018-10-24 2020-04-30 Obsidian Therapeutics, Inc. Er tunable protein regulation
WO2020123574A1 (en) 2018-12-13 2020-06-18 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
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
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
WO2020214763A1 (en) 2019-04-18 2020-10-22 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
CN110327982A (en) * 2019-07-03 2019-10-15 燕山大学 It is a kind of using PEI- Ciprofloxacin micella as Pd nano particle of template and preparation method thereof
US11814333B2 (en) 2019-07-30 2023-11-14 Factor Bioscience Inc. Cationic lipids and transfection methods
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
US10611722B1 (en) 2019-07-30 2020-04-07 Factor Bioscience Inc. Cationic lipids and transfection methods
US11242311B2 (en) 2019-07-30 2022-02-08 Factor Bioscience Inc. Cationic lipids and transfection methods
US10752576B1 (en) 2019-07-30 2020-08-25 Factor Bioscience Inc. Cationic lipids and transfection methods
US10556855B1 (en) 2019-07-30 2020-02-11 Factor Bioscience Inc. Cationic lipids and transfection methods
WO2021079002A2 (en) 2019-10-24 2021-04-29 Novago Therapeutics Ag Novel anti-nogo-a antibodies
US11530236B2 (en) 2019-10-30 2022-12-20 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11130778B2 (en) 2019-10-30 2021-09-28 Oric Pharmaceuticals, Inc. CD73 inhibitors
US11028120B2 (en) 2019-10-30 2021-06-08 Oric Pharmaceuticals, Inc. CD73 inhibitors
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
WO2021245184A1 (en) 2020-06-02 2021-12-09 Neurimmune Ag HUMAN ANTIBODIES AGAINST SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS-2 (SARS-CoV-2)
WO2022002040A1 (en) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022037652A1 (en) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
WO2022152109A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2022152141A2 (en) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Polymer conjugated lipid compounds and lipid nanoparticle compositions
WO2022232478A1 (en) 2021-04-30 2022-11-03 Sarepta Therapeutics, Inc. Treatment methods for muscular dystrophy
WO2022247755A1 (en) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
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
CN113995833A (en) * 2021-11-10 2022-02-01 南京理工大学 Application of adenosine deaminase and modifier thereof in preparation of diabetic wound repair drugs
CN113995833B (en) * 2021-11-10 2023-12-26 南京南方元生物科技有限公司 Adenosine deaminase and application of modification thereof in preparation of diabetes wound repair drugs
WO2023122752A1 (en) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Constrained lipids and methods of use thereof
WO2023178230A1 (en) 2022-03-17 2023-09-21 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer conjugates
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
WO2024037578A1 (en) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition of lipid nanoparticles
US11951179B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951181B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951180B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery

Similar Documents

Publication Publication Date Title
US20210060175A1 (en) Methods of immunostimulation with complexes of rna and cationic carriers
WO2009127230A1 (en) MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
US20100047261A1 (en) Base-modified rna for increasing the expression of a protein
WO2010088927A1 (en) Use of pei for the improvement of endosomal release and expression of transfected nucleic acids, complexed with cationic or polycationic compounds
WO2011069528A1 (en) Lyophilization of nucleic acids in lactate-containing solutions

Legal Events

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

Ref document number: 08758342

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08758342

Country of ref document: EP

Kind code of ref document: A1