WO2011069529A1 - Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques - Google Patents

Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques Download PDF

Info

Publication number
WO2011069529A1
WO2011069529A1 PCT/EP2009/008804 EP2009008804W WO2011069529A1 WO 2011069529 A1 WO2011069529 A1 WO 2011069529A1 EP 2009008804 W EP2009008804 W EP 2009008804W WO 2011069529 A1 WO2011069529 A1 WO 2011069529A1
Authority
WO
WIPO (PCT)
Prior art keywords
mage
nucleic acid
sequence
lyophilization
transfection
Prior art date
Application number
PCT/EP2009/008804
Other languages
English (en)
Inventor
Thorsten Mutzke
Original Assignee
Curevac Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curevac Gmbh filed Critical Curevac Gmbh
Priority to PCT/EP2009/008804 priority Critical patent/WO2011069529A1/fr
Priority to PCT/EP2010/006788 priority patent/WO2011069586A1/fr
Priority to US13/509,564 priority patent/US20120258046A1/en
Priority to EP10779700.3A priority patent/EP2510100B1/fr
Publication of WO2011069529A1 publication Critical patent/WO2011069529A1/fr
Priority to US14/492,334 priority patent/US9616084B2/en
Priority to US15/451,675 priority patent/US20170182081A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination

Definitions

  • the present invention is directed to (the use of) a solution containing at least one nucleic acid (sequence) and free mannose for lyophilization, transfection and/or injection, particularly of RNA and mRNA.
  • the inventive solution exhibits a positive effect on stabilization of the nucleic acid (sequence) during lyophilization and storage but also leads to a considerable increase of the transfection efficiency of a nucleic acid. It thus also increases in vivo expression of a protein encoded by such a nucleic acid upon increased transfection rate.
  • the present invention is furthermore directed to a method of lyophilization using the mannose-containing solution, to pharmaceutical compositions, vaccines, kits, first and second medical indications applying such a mannose-containing solution and/or a nucleic acid (sequence) lyophilized or resuspended with such a solution"
  • nucleic acids In gene therapy and many other therapeutically relevant biochemical and biotechnological applications the use of nucleic acids for therapeutic and diagnostic purposes is of major importance. As an example, rapid progress has occurred in recent years in the field of gene therapy and promising results have been achieved. Nucleic acids are therefore regarded as important tools for gene therapy and prophylactic and therapeutic vaccination against infectious and malignant diseases. Nucleic acids, both DNA and RNA, have been used widely in gene therapy, either in naked or in complexed form. In this context, the application of nucleic acids and particularly of RNA for therapeutic vaccination is revised permanently. On the one hand, nucleic acids and particularly RNA or mRNA molecules can be optimized for a more efficient transcription rate.
  • the 5' Cap structure the untranslated and translated regions are typically modified to stabilize the molecule or to change its characteristics to enhance its translation properties (see e.g. Pascolo, S. (2008), Handb Exp Pharmacol (183): 221 -35).
  • different formulations of nucleic acids and particularly of mRNA molecules or different delivery routes are investigated to achieve improved expression levels.
  • the encapsulation into cationic liposomes or cationic polymers see e.g. Hoerr, I., R. Obst, et a/. (2000), Eur J Immunol 30(1 ): 1 -7. ; Hess, P. R., D. Boczkowski, et al.
  • RNA pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med 1 84(2): 465-72; Boczkowski, et al, 1 996, supra), and the direct injection of naked RNA (see Hoerrr et al, 2000, supra). Despite all progress achieved regarding gene delivery it is very important to improve further transfection efficiency to make nucleic acids especially RNA applicable for all imaginable therapeutic purposes. Application of RNA thus represents a favored tool in modern molecular medicine. It also exhibits some superior properties over DNA cell transfection. As generally known, transfection of DNA molecules may lead to serious problems. E.g. application of DNA molecules bears the risk that the DNA integrates into the host genome.
  • RNA particularly mRNA
  • An advantage of using RNA rather than DNA is that no virus-derived promoter element has to be administered in vivo and no integration into the genome may occur.
  • RNA has not to overcome the barrier to the nucleus.
  • a main disadvantage resulting from the use of RNA is due to its huge instability.
  • DNA e.g., naked DNA
  • DNA introduced into a patient' circulatory system is typically not stable and therefore may have little chance of affecting most disease processes (see e.g. Poxon et al, Pharmaceutical development and Technology, 5(1 ), 1 1 5-122 (2000)) the problem of stability is even more evident in the case of RNA.
  • the physico chemical stability of RNAs in solution is extremely low. RNA is very susceptible to hydrolysis by ubiquitous ribonucleases and is typically completely degraded already after a few hours or days in solution. This even occurs in the absence of RNases, e.g. when stored a few hours or days in solution at room temperature.
  • RNA is typically stored at -20°C or even -80°C and RNAse free conditions to prevent a prior degradation of the RNA.
  • This method does not prevent a loss of function effectively and additionally is very cost-intensive for shipping when these temperatures have to be guaranteed.
  • One further method for stabilization comprises lyophilization or freeze-drying of the RNA. Lyophilization is a worldwide known and recognized method in the art to enhance storage stability of temperature sensitive biomolecules, such as nucleic acids. During lyophilization, typically water is removed from a frozen sample containing nucleic acids via sublimation. The process of lyophilization is usually characterized by a primary and a secondary drying step. During the primary drying step, free, i.e.
  • the sample containing nucleic acids is initially cooled below the freezing point of the solution and accordingly of the water contained therein. As a result, the water freezes. Dependent on temperature, rate of cooling down (freezing rate), and the time for freezing, the crystal structure of water is changed. This exhibits physical stress on the nucleic acid (sequence) and other components of the solution, which may lead to a damage of the nucleic acid, e.g. breakage of strands, loss of supercoiling, etc. Furthermore, due to the decrease of volume and loss of the hydration sphere, autocatalytic degradation processes are favored e.g. by traces of transition metals. Additionally, significant changes of pH are possible by concentration of traces of acids and bases.
  • Lyophilization involves two stresses, freezing and drying. Both are known to damage nucleic acids, such as non-viral vectors or plasmid DNA. In the literature, a number of cryoprotectants and lyoprotectants are discussed for lyophilization purposes to prevent these damages. In this context, cryoprotectants are understood as excipients, which allow influencing the structure of the ice and/or the eutectical temperature of the mixture. Lyoprotectants are typically excipients, which partially or totally replace the hydration sphere around a molecule and thus prevent catalytic and hydrolytic processes.
  • lyophilization causes the removal of the hydration sphere around the DNA, wherein it appears that there are approximately 20 water molecules per nucleotide pair bound most tightly to DNA. These water molecules do not form an ice-like structure upon low-temperature cooling. Upon DNA dehydration over hygroscopic salts at 0% relative humidity, only five or six water molecules remain (see e.g. Tao et a!, Biopolymers, 28, 1019-1030 (1989)). Lyophilization may increase the stability of DNA under long-term storage, but may also cause some damage upon the initial lyophilization process, potentially through changes in the DNA secondary structure, breaks of the nucleic acid chain(s) or the concentration of reactive elements such as contaminating metals.
  • Lyophilization can also cause damage upon the initial lyophilization process in other nucleic acid, e.g. RNA.
  • Agents that can substitute for non-freezable water, such as some carbohydrates, can demonstrate cryoprotective properties for DNA and other molecules during lyophilization of intact bacteria (see e.g. Israeli et al, Cryobiology, 30, 519-523 (1993); or Rudolph et al, Arch. Biochem. Biophys., 245, 134-143 (1986)).
  • specific carbohydrates are utilized in the art as Iyoprotective substances for enhancing stability of the nucleic acid (sequence) during lyophilization. They exhibit an effect on storage stability after lyophilisation of pure nucleic acids or nucleic acid (sequence) complexes (see e.g. Maitani, Y., Y. Aso, et al. (2008), Int J Pharm 356(1 -2): 69- 75 ; Quaak, S. C, J. H. van den Berg, et al. (2008), Eur J Pharm Biopharm 70(2): 429-38 ; Jones, K. L., D.
  • Lyoprotective properties are particularly described for sucrose, glucose, and trehalose. They allow to restore at least in part the transfection efficiency which is otherwise lost in many cases after lyophilisation (see Maitani et al, 2008, supra; Yadava, P., M. Gibbs, et al. (2008).
  • Sugars are able to prevent loss in activity due to the lyophilization process mainly by preventing particle fusion/aggregation especially in the case of liposome complexed nucleic acids (see Yadava et al, 2008, supra; Katas, H., S. Chen, etal. (2008), J Microencapsul: 1 -8; Molina etal, supra, 2001 ).
  • Poxon et al. (2000, supra) investigated the effect of lyophilization on plasmid DNA activity.
  • Poxon et al. (2000, supra) hypothetized, that a change in the DNA conformation from supercoiled to open circular and linear form would be indicative of damage of the plasmid DNA.
  • the percentage of supercoiled DNA did not change after lyophilization and subsequent DMED treatment, suggesting that other effects drew responsible for the loss of transfection efficiency.
  • Li et al. (see Li, B., S. Li, et al. (2000), J Pharm Sci 89(3): 355-64) furthermore showed that disaccharides are superior to monosaccharides using them as a cryoprotectant for lyophilization of lipid based gene delivery systems due to the prevention of aggregation. They noted that it is very important to prevent the particle size of the complexes during lyophilization. Unfortunately, in a specific example of lipid based gene delivery systems, lyophilization with mannose led to an increase in particle size, which was regarded as negative for transfection efficiency. Additionally Li et al. (2000, supra) showed that lipid delivery systems can be stored at room temperature without loss of transfection efficiency when lyophilized in 10% sucrose.
  • Li et al. (2000, supra) did not examine the stabilization due to the presence of mannose as a lyoprotectant. More importantly, they did not observe an increase in the expression of the encoded protein due to the presence of sugar (sucrose and trehalose) in the injection buffer.
  • Jones et a/ (2007, supra) is one rare document, which examines the effect of sugars on long term stability of mRNA. It describes the possibility to prevent storage depending loss of transfection activity in vitro. Jones et al (2007 , supra) uses trehalose as a lyoprotectant and shows a preventive effect on the loss of transfection activity at a storage temperature of 4°C for a period of 6 months. Integrity of the mRNA was only measured by loss of weight after recovering. At elevated temperatures (room temperature and higher) degradation and a dramatic loss of transfection efficiency took place. Additionally; transfection efficiency could not be improved using trehalose as lyoprotectant.
  • specific carbohydrates may also be utilized to improve biological activity and/or transfection efficiency, which is, at least at a first glance, independent from stability issues.
  • specific carbohydrates e.g. of mannose may be attributed to the interaction of these carbohydrates with specific receptors in the cell.
  • mannose may involve the mannose receptor targeted transfer.
  • the mannose receptor (MR) is primarily present on dendritic cells (DCs) and macrophages.
  • the carbohydrate recognition domains of the MR recognizes carbohydrates (e.g. mannose, fucose, glucose, N-Acetylglucosamine, maltose) on the cell walls of infectious agents (mainly bacteria and yeast) which leads to rapid internalization and phagocytosis.
  • mannose was covalently bound to the vector to ensure a combined uptake due to binding to the mannose receptor.
  • the expression of the mannose receptor is restricted to a few cell types (especially dendritic cells) which are not excessively present in the dermis and therefore it appeared unlikely that free mannose improves the expression of the encoded protein due to an increased uptake in mannose receptor expressing cells.
  • sucrose is a common trigger for endocytosis in animal cells and therefore the ODN internalizes into endosomes together with the sucrose.
  • Sun et al. (2007) only examined in vitro transfection assays which are very difficult to transfer to the in vivo situation due to the dilution effect. In tissues it thus appeared very unlikely that the nucleic acid and the sugar molecule enter the cell at the same time.
  • RNA is very cost-intensive to ensure temperatures at -20°C and below during shipment.
  • the problem underlying the present invention is solved by (the use of) a solution containing at least one nucleic acid (sequence) and free mannose for lyophilization, transfection and/or injection.
  • the inventive solution containing at least one nucleic acid (sequence) and mannose stabilizes the at least one nucleic acid (sequence) contained in the inventive solution during lyophilization and/or improves biological activity of the nucleic acid (sequence). This is particularly preferable true, if a protein is encoded by the at least one nucleic acid (sequence), as expression of an encoded protein may be increased thereby.
  • the present invention thus provides (the use of) a solution containing at least one nucleic acid (sequence) and free mannose for lyophilization, transfection and/or injection.
  • free mannose is preferably understood as a mannose, which is not covalently bound to the nucleic acid (sequence).
  • mannose is preferably a sugar monomer of the aldohexose series of carbohydrates.
  • Mannose as defined herein typically has the molecular formula C 6 H 12 C 6 , is also known under its lUPAC nomenclature as (2S,3S,4R,5R)- Pentahydroxyhexanal, (2A',3A , ,4 ,55)-Pentahydroxyhexanal. It is preferably identified under CAS number 31 103-86-3 and typically exhibits the following general structure:
  • Mannose is typically formed by the oxidation of mannitol. It can also be formed from D- glucose in the Lobry-de Bruyn-van Ekenstein transformation. Mannose as defined herein typically occurs in two diastereomeric isoforms, D-Mannose and L-Mannose (CAS numbers 3458-28-4 for D-mannose and 10030-80-5 for L-mannose). D-mannose is sold as a naturopathic remedy for urinary tract infections, and it is claimed to work through the disruption of adherence of bacteria in the urinary tract. D-Mannose and L-Mannose can be illustrated as the D and L straight-chain forms of mannose using Fischer projections according to the following structures:
  • mannose as used herein is a D-Mannose.
  • D-Mannose may be depicted according to at least one of the D-Mannose isomers a-D- Mannofuranose, ⁇ -D-Mannofuranose, a-D-Mannopyranose and ⁇ -D-Mannopyranose as represented by following Haworth-structures:
  • D- Mannose forms anomers, wherein ⁇ -D-Mannofuranose ocurrs in a concentration/frequency of less than 1 %, ⁇ -D-Mannofuranose in a concentration/frequency of less than 1 %, a-D- Mannopyranose in a concentration/frequency of about 67 % and ⁇ -D-Mannopyranose in a concentration/frequency of about 33 %.
  • D-Mannose may be selected more preferably from at least one, two, three or four of the anomers a-D-Mannofuranose, ⁇ -D- Mannofuranose, a-D-Mannopyranose and/or ⁇ -D-Mannopyranose. Most preferably, upon solubilization in an aqueous solution mannose typically forms the above anomers in an equilibrity reaction, typically in the above concentrations.
  • mannose as used herein is selected from an anomeric mixture of D-Mannose, preferably an anomeric mixture comprising a-D- Mannofuranose, ⁇ -D-Mannofuranose, a-D-Mannopyranose and ⁇ -D-Mannopyranose, more preferably in the above concentrations/frequencies.
  • mannose as used herein may be selected from L-mannose or a racemic mixture of D- Mannose and/or L-Mannose, wherein D-mannose preferably as described above. Such mixtures may be obtained e.g. by a non-selective synthesis of mannose, e.g.
  • mannose as used herein is typically present in the inventive solution for lyophilization, transfection and/or injection in a concentration of about 0.01 to about 10% (w/w), preferably in a concentration of about 0.01 to about 10% (w/w), more preferably in a concentration of about 0.1 to about 7.5% (w/w), even more preferably in a concentration of about 0.5 to about 5% (w/w), and most preferably in a concentration of about 1 to about 4% (w/w), e.g.
  • a concentration of about 1 % (w/w) mannose corresponds to a concentration of about 55,506 mM mannose. Any of the above and herein mentioned values and concentrations for mannose in % (w/w) may thus be calculated in mM on the above basis.
  • the present invention provides (use of) a solution containing at least one nucleic acid sequence and free mannose for lyophilization, transfection and/or injection of the at least one nucleic acid (sequence).
  • a lyophilized nucleic acid may be any suitable nucleic acid, selected e.g. from any (double-stranded or single-stranded) DNA, preferably, without being limited thereto, e.g.
  • genomic DNA single-stranded DNA molecules, double-stranded DNA molecules, coding DNA, DNA primers, DNA probes, immunostimulatory DNA, a (short) DNA oligonucleotide ((short) oligodesoxyribonucleotides), or may be selected e.g. from any PNA (peptide nucleic acid) or may be selected e.g.
  • RNA from any (double-stranded or single-stranded) RNA, preferably, without being limited thereto, a (short) RNA oligonucleotide ((short) oligoribonucleotide), a coding RNA, a messenger RNA (mRNA), an immunostimulatory RNA, a siRNA, an antisense RNA, a micro RNA or riboswitches, ribozymes or aptamers; etc.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be a ribosomal RNA (rRNA), a transfer RNA (tRNA), a messenger RNA (mRNA), or a viral RNA (vRNA).
  • rRNA ribosomal RNA
  • tRNA transfer RNA
  • mRNA messenger RNA
  • vRNA viral RNA
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is an RNA. More preferably, the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a (linear) single-stranded RNA, even more preferably an mRNA.
  • 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 or peptides.
  • Such coding sequences in di-, or even multicistronic mRNA may be separated by at least one IRES sequence, e.g. as defined herein.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a single- or a double-stranded nucleic acid (molecule) (which may also be regarded as a nucleic acid (molecule) due to non-covalent association of two single-stranded nucleic acid(s) (molecules)) or a partially double-stranded or partially single stranded nucleic acid, which are at least partially self complementary (both of these partially double-stranded or partially single stranded nucleic acid molecules are typically formed by a longer and a shorter single-stranded nucleic acid molecule or by two single stranded nucleic acid molecules, which are about equal in length, wherein one single- stranded nucleic acid molecule is in part complementary to the other single-stranded nucleic acid molecules molecule and both thus form a double-stranded nucleic acid molecules molecule in this region, i.e.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a single-stranded nucleic acid molecule.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a circular or linear nucleic acid molecule, preferably a linear nucleic acid molecule.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a coding nucleic acid, e.g. a DNA or RNA.
  • a coding DNA or RNA may be any DNA or RNA as defined above.
  • such a coding DNA or RNA may be a single- or a double-stranded DNA or RNA, more preferably a single-stranded DNA or RNA, and/or a circular or linear DNA or RNA, more preferably a linear DNA or RNA.
  • the coding DNA or RNA may be a (linear) single-stranded DNA or RNA.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a ((linear) single- stranded) messenger RNA (mRNA).
  • mRNA messenger RNA
  • Such 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 or peptides.
  • Such coding sequences in di-, or even multicistronic mRNA may be separated by at least one IRES sequence, e.g. as defined herein.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may encode a protein or a peptide, which may be selected, without being restricted thereto, e.g. from therapeutically active proteins or peptides, from antigens, e.g. tumor antigens, pathogenic antigens (e.g.
  • therapeutically active proteins may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • therapeutically active proteins may comprise proteins, capable of stimulating or inhibiting the signal transduction in the cell, e.g. cytokines, antibodies, etc.
  • Therapeutically active proteins may thus comprise cytokines of class I of the family of cytokines, having 4 positionally conserved cysteine residues (CCCC) and comprising a conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS), wherein X is a non-conserved amino acid.
  • Cytokines of class I of the family of cytokines comprise the GM-CSF subfamily, e.g.
  • Therapeutically active proteins may also comprise cytokines of class II of the family of cytokines, which also comprise 4 positionally conserved cystein residues (CCCC), but no conserved sequence motif Trp-Ser-X-Trp-Ser (WSXWS). Cytokines of class II of the family of cytokines comprise e.g.
  • Therapeutically active proteins may additionally comprise cytokines of the family of tumor necrose factors, e.g. TNF-alpha, TNF-beta, etc., or cytokines of the family of chemokines, which comprise 7 transmembrane helices and interact with G-protein, e.g. IL-8, MIP-1 , RANTES, CCR5, CXR4, etc., or cytokine specific receptors, such as TNF-RI, TNF-RII, CD40, OX40 (CD134), Fas, etc.
  • cytokines of the family of tumor necrose factors e.g. TNF-alpha, TNF-beta, etc.
  • chemokines which comprise 7 transmembrane helices and interact with G-protein, e.g. IL-8, MIP-1 , RANTES, CCR5, CXR4, etc.
  • cytokine specific receptors such as TNF-RI, TNF-RII, CD40, OX40 (CD134
  • Therapeutically active proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be selected from any of the proteins given in the following: 0ATL3, 0FC3, 0PA3, 0PD2, 4- 1 BBL, 5T4, 6Ckine, 707-AP, 9D7, A2M, AA, AAAS, AACT, AASS, ABAT, ABCA1 , ABCA4, ABCB1 , ABCB1 1 , 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,
  • Therapeutically active proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may further 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- x L , Bcl-x s , bik, CAD, Calpain, Caspase e.g.
  • AIF Apaf e.g. Apaf-1 , Apaf-2, Apaf-3, oder APO-2 (L), APO-3 (L)
  • Apopain Bad, Bak, Bax, Bcl-2, Bcl- x L , Bcl-x s , bik, CAD, Calpain, Caspase e.g.
  • a therapeutically active protein which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection can also be an adjuvant protein.
  • an adjuvant protein is preferably to be understood as any protein, which is capable to elicit an innate immune response as defined herein.
  • an innate immune response comprises activation of a pattern recognition receptor, such as e.g. a receptor selected from the Toll-like receptor (TLR) familiy, including e.g. a Toll like receptor selected from human TLR1 to TLR10 or from murine Toll like receptors TLR1 to TLR13.
  • TLR Toll-like receptor
  • an innate immune response is elicited in a mammal as defined above.
  • the adjuvant protein is selected from human adjuvant proteins or from pathogenic adjuvant proteins, in particular from bacterial adjuvant proteins.
  • mRNA encoding human proteins involved in adjuvant effects may be used as well.
  • Human adjuvant proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection typically comprise any human protein, which is capable of eliciting an innate immune response (in a mammal), e.g. as a reaction of the binding of an exogenous TLR ligand to a TLR.
  • human adjuvant proteins encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be selected from the group consisting of, without being limited thereto, cytokines which induce or enhance an innate immune response, including IL-2, IL-12, IL-15, IL-18, IL-21 CCL21 , GM-CSF and TNF-alpha; cytokines which are released from macrophages, including IL-1 , IL-6, IL-8, IL- 12 and TNF-alpha; from components of the complement system including C1 q, MBL, C1 r, C1 s, C2b, Bb, D, MASP-1 , MASP-2, C4b, C3b, C5a, C3a, C4a, C5b, C6, C7, C8, C9, CR1 , CR2, CR3, CR4, Cl qR, C1 INH, C4bp, MCP, DAF
  • NF- ⁇ , c-Fos, c-Jun, c-Myc e.g. IL-1 alpha, IL-1 beta, Beta-Defensin, IL-6, IFN gamma, IFN alpha and IFN beta; from costimulatory molecules, including CD28 or CD40-ligand or PD1 ; protein domains, including LAMP; cell surface proteins; or human adjuvant proteins including CD80, CD81 , CD86, trif, flt-3 ligand, thymopentin, Gp96 or fibronectin, etc., or any species homolog of any of the above human adjuvant proteins.
  • Pathogenic adjuvant proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection typically comprise any pathogenic (adjuvant) protein, which is capable of eliciting an innate immune response (in a mammal), more preferably selected from pathogenic (adjuvant) proteins derived from bacteria, protozoa, viruses, or fungi, animals, etc., and even more preferably from pathogenic adjuvant proteins selected from the group consisting of, without being limited thereto, bacterial proteins, protozoan proteins (e.g. profilin - like protein of Toxoplasma gondii), viral proteins, or fungal proteins, animal proteins, etc.
  • pathogenic (adjuvant) protein which is capable of eliciting an innate immune response (in a mammal)
  • pathogenic (adjuvant) proteins derived from bacteria, protozoa, viruses, or fungi, animals, etc. and even more preferably from pathogenic adjuvant
  • bacterial (adjuvant) proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may comprise any bacterial protein, which is capable of eliciting an innate immune response (preferably in a mammal) or shows an adjuvant character.
  • such bacterial (adjuvant) proteins are selected from the group consisting of bacterial heat shock proteins or chaperons, including Hsp60, Hsp70, Hsp90, Hspl OO; OmpA (Outer membrane protein) from gram-negative bacteria; bacterial porins, including OmpF; bacterial toxins, including pertussis toxin (PT) from Bordetella pertussis, pertussis adenylate cyclase toxin CyaA and CyaC from Bordetella pertussis, PT-9K/129G mutant from pertussis toxin, pertussis adenylate cyclase toxin CyaA and CyaC from Bordetella pertussis, tetanus toxin, cholera toxin (CT), cholera toxin B-subunit, CTK63 mutant from cholera toxin, CTE1 12K mutant from CT, Escherichia coli heat-labile
  • Bacterial (adjuvant) proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be selected from bacterial adjuvant proteins, even more preferably selected from the group consisting of, without being limited thereto, bacterial flagellins, including flagellins from organisms including Agrobacterium, Aquifex, Azospirillum, Bacillus, Bartonella, Bordetella, Borrelia, Burkholderia, Campylobacter, Caulobacte, Clostridium, Escherichia, Helicobacter, Lachnospiraceae, Legionella, Listeria, Proteus, Pseudomonas, Rhizobium, Rhodobacter, Roseburia, Salmonella, Serpulina, Serratia, Shigella, Treponema, Vibrio, Wolinella, Yersinia, more preferably flagellins from the species, without being limited thereto
  • Bacterial flagellins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection even more preferably comprise a sequence selected from the group comprising any of the following sequences as referred to their accession numbers: organism species gene name accession No Gl No
  • Rhizobium Rhizobium meliloti flaA M24526 Gl:1 52220 flaB
  • Protozoan proteins which may also be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be selected from any protozoan protein showing adjuvant character, more preferably, from the group consisting of, without being limited thereto, Tc52 from Trypanosoma cruzi, PFTG from Trypanosoma gondii, Protozoan heat shock proteins, LeIF from Leishmania spp., profi linlike protein from Toxoplasma gondii, etc.
  • Viral proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be selected from any viral protein showing adjuvant character, more preferably, from the group consisting of, without being limited thereto, Respiratory Syncytial Virus fusion glycoprotein (F-protein), envelope protein from MMT virus, mouse leukemia virus protein, Hemagglutinin protein of wild type measles virus, etc.
  • F-protein Respiratory Syncytial Virus fusion glycoprotein
  • Fungal proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be selected from any fungal protein showing adjuvant character, more preferably, from the group consisting of, without being limited thereto, fungal immunomodulatory protein (FIP; LZ-8), etc.
  • pathogenic adjuvant proteins which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may finally be selected from any further pathogenic protein showing adjuvant character, more preferably, from the group consisting of, without being limited thereto, Keyhole limpet hemocyanin (KLH), OspA, etc.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may alternatively encode an antigen.
  • 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.
  • the first step of an adaptive immune response is the activation of naive antigen-specific T cells by antigen-presenting cells. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing.
  • the three cell types that can serve as antigen- presenting cells are dendritic cells, macrophages, and B cells.
  • Tissue dendritic cells take up antigens by phagocytosis and macropinocytosis and are stimulated by infection to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells. Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents to express MHC class II molecules. The unique ability of B cells to bind and internalize soluble protein antigens via their receptors may be important to induce T cells. By presenting the antigen on MHC molecules leads to activation of T cells which induces their proliferation and differentiation into armed effector T cells.
  • effector T cells The most important function of effector T cells is the killing of infected cells by CD8 + cytotoxic T cells and the activation of macrophages by TH1 cells which together make up cell-mediated immunity, and the activation of B cells by both TH2 and TH1 cells to produce different classes of antibody, thus driving the humoral immune response.
  • T cells recognize an antigen by their T cell receptors which does not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogens' protein antigens, which are bound to MHC molecules on the surfaces of other cells.
  • T cells fall into two major classes that have different effector functions. The two classes are distinguished by the expression of the cell-surface proteins CD4 and CD8.
  • T cells differ in the class of MHC molecule that they recognize.
  • MHC class I and MHC class II- differ in their structure and expression pattern on tissues of the body.
  • CD4 + T cells bind to the MHC class II molecule and CD8 + T cells to the MHC class I molecule.
  • MHC class I and MHC class II have distinct distributions among cells that reflect the different effector functions of the T cells that recognize them.
  • MHC class I molecules present peptides from pathogens, commonly viruses to CD8 + T cells, which differentiate into cytotoxic T cells that are specialized to kill any cell that they specifically recognize. Almost all cells express MHC class I molecules, although the level of constitutive expression varies from one cell type to the next.
  • MHC class I molecules bind peptides from proteins degraded in the cytosol and transported in the endoplasmic reticulum. Thereby MHC class I molecules on the surface of cells infected with viruses or other cytosolic pathogens display peptides from these pathogen.
  • the CD8 + T cells that recognize MHC class hpeptide complexes are specialized to kill any cells displaying foreign peptides and so rid the body of cells infected with viruses and other cytosolic pathogens.
  • CD4 + T cells CD4 + helper T cells
  • MHC class II molecules are normally found on B lymphocytes, dendritic cells, and macrophages, cells that participate in immune responses, but not on other tissue cells. Macrophages, for example, are activated to kill the intravesicular pathogens they harbour, and B cells to secrete immunoglobulins against foreign molecules. MHC class II molecules are prevented from binding to peptides in the endoplasmic reticulum and thus MHC class II molecules bind peptides from proteins which are degraded in endosomes.
  • TH1 cells can capture peptides from pathogens that have entered the vesicular system of macrophages, or from antigens internalized by immature dendritic cells or the immunoglobulin receptors of B cells.
  • Pathogens that accumulate in large numbers inside macrophage and dendritic cell vesicles tend to stimulate the differentiation of TH1 cells, whereas extracellular antigens tend to stimulate the production of TH2 cells.
  • TH1 cells activate the microbicidal properties of macrophages and induce B cells to make IgG antibodies that are very effective of opsonising extracellular pathogens for ingestion by phagocytic cells
  • TH2 cells initiate the humoral response by activating naive B cells to secrete IgM, and induce the production of weakly opsonising antibodes such as IgCl and lgG3 (mouse) and lgG2 and lgG4 (human) as well as IgA and IgE (mouse and human).
  • antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection typically comprise any antigen, falling under the above definition, more preferably protein and peptide antigens, e.g. tumor antigens, allergy antigens, auto-immune self- antigens, pathogens, etc.
  • antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be antigens generated outside the cell, more typically antigens not derived from the host organism (e.g. a human) itself (i.e.
  • non-self antigens but rather derived from host cells outside the host organism, e.g. viral antigens, bacterial antigens, fungal antigens, protozoological antigens, animal antigens (preferably selected from animals or organisms as disclosed herein), allergy antigens, etc.
  • Allergy antigens are typically antigens, which cause an allergy in a human and may be derived from either a human or other sources.
  • Antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be furthermore antigens generated inside the cell, the tissue or the body, e.g. by secretion of proteins, their degradation, metabolism, etc.
  • antigens include antigens derived from the host organism (e.g. a human) itself, e.g. tumor antigens, self-antigens or auto-antigens, such as auto-immune self-antigens, etc., but also (non-self) antigens as defined above, which have been originally been derived from host cells outside the host organism, but which are fragmented or degraded inside the body, tissue or cell, e.g. by (protease) degradation, metabolism, etc.
  • host organism e.g. a human
  • tumor antigens e.g. tumor antigens, self-antigens or auto-antigens, such as auto-immune self-antigens, etc.
  • non-self antigens as defined above, which have been originally been derived from host cells outside the host organism, but which are fragmented or degraded inside the body, tissue or cell, e.g. by (protease) degradation, metabolism, etc.
  • Tumor antigens are preferably located on the surface of the (tumor) cell. Tumor antigens may also be selected from proteins, which are overexpressed in tumor cells compared to a normal cell. Furthermore, tumor antigens also includes antigens expressed in cells which are (were) not themselves (or originally not themselves) degenerated but are associated with the supposed tumor. Antigens which are connected with tumor-supplying vessels or (re)formation thereof, in particular those antigens which are associated with neovascularization, e.g.
  • Antigens connected with a tumor furthermore include antigens from cells or tissues, typically embedding the tumor. Further, some substances (usually proteins or peptides) are expressed in patients suffering (knowingly or not-knowingly) from a cancer disease and they occur in increased concentrations in the body fluids of said patients. These substances are also referred to as “tumor antigens", however they are not antigens in the stringent meaning of an immune response inducing substance.
  • the class of tumor antigens can be divided further into tumor-specific antigens (TSAs) and tumor-associated- antigens (TAAs). TSAs can only be presented by tumor cells and never by normal "healthy" cells.
  • 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.
  • tumor antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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.
  • colorectal cancer gastric cancer
  • lung cancer head and neck cancer
  • leukemia esophageal cancer
  • gastric cancer cervical cancer
  • ovarian adenocarcinoma antigen cancer breast cancer
  • bladder cancer head and neck cancer, lung cancer, melanoma,
  • gastric cancer pancreatic cancer
  • liver cancer breast cancer
  • gallbladder cancer colon cancer
  • ovarian cancer colorectal cancer, gastric cancer, liver cancer, pancreatic cancer, uterus cancer, cervix carcinoma, colon cancer,
  • gut carcinoma colorectal cancer, colon cancer, hepatocellular cancer, lung cancer, breast cancer, thyroid cancer, pancreatic cancer, liver cancer cervix cancer, bladder
  • bladder cancer lung cancer, T-cell cyp-B cyclophilin B leukemia, squamous cell carcinoma,
  • DAM-10/MAGE- differentiation antigen melanoma melanoma skin tumors, ovarian
  • DAM-6/MAGE- differentiation antigen melanoma melanoma skin tumors, ovarian
  • lung cancer ovarian cancer, head and neck cancer, colon cancer
  • EGFR/Her1 pancreatic cancer breast cancer lung cancer, breast cancer, bladder tumor cell-associated extracellular cancer, ovarian cancer, brain
  • EpCam epithelial cell adhesion molecule cancer EpCam epithelial cell adhesion molecule cancer, lung cancer
  • EZH2 (enhancer of Zeste homolog 2) prostate cancer, breast cancer
  • Fra-1 Fos-related antigen- 1 renal cell carcinoma thyroid cancer leukemia, renal cell carcinoma, head and neck cancer, colon cancer,
  • G250/CAIX glycoprotein 250 ovarian cancer, cervical cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • bladder cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • bladder cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • bladder cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • bladder cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • bladder cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • bladder cancer bladder cancer, lung cancer, sarcoma, melanoma, head and neck
  • breast cancer bladder cancer, human epidermal receptor- melanoma, ovarian cancer, pancreas
  • Her2/neu/ErbB2 2/neurological cancer gastric cancer
  • breast cancer melanoma
  • lung cancer ovarian cancer
  • sarcoma human telomerase reverse Non-Hodgkin-lymphoma
  • acute hTERT transcriptase leukemia acute hTERT transcriptase leukemia
  • tongue cancer hepatocellular carcinomas, melanoma, gastric cancer, esophageal, colon cancer,
  • bladder cancer bladder cancer, head and neck cancer, melanoma, colon cancer,
  • bladder cancer bladder cancer, head and neck cancer, melanoma, colon cancer,
  • MAGE-A2 melanoma antigen-A2 lung cancer sarcoma, leukemia bladder cancer, head and neck cancer, melanoma, colon cancer,
  • MAGE-A4 melanoma antigen-A4 lung cancer sarcoma, leukemia bladder cancer, head and neck cancer, melanoma, colon cancer,
  • MAGE-A6 melanoma antigen-A6 lung cancer sarcoma, leukemia bladder cancer, head and neck cancer, melanoma, colon cancer,
  • melanoma lung cancer, leukemia, preferentially expressed antigen of head and neck cancer, renal cell
  • bladder cancer bladder cancer, melanoma, renal
  • bladder cancer bladder cancer, melanoma, sarcoma, brain tumor, esophagel cancer, renal
  • SART-1 1 cancer lung cancer, uterine cancer head and neck cancer, lung cancer, squamous antigen rejecting tumor renal cell carcinoma, melanoma,
  • gastric cancer colon cancer
  • lung cancer breast cancer
  • breast cancer ovarian
  • the tumor antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection are selected from the group consisting of 5T4, 707-AP, 9D7, AFP, AlbZIP HPG1 , 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-catenin/m, BING-4, BRCA1/m, BRCA2/m, CA 1 5-3/CA 27-29, CA 1 9-9, CA72-4, CA125, calreticulin, CAMEL, CASP-8/m, cathepsin B, cathepsin L, CD19, CD20, CD22, CD25, CDE30, CD33, CD4, CD52
  • the tumor antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection are selected from the group consisting of MAGE-A1 (e.g. MAGE-A1 according to accession number M77481 ), MAGE-A2, MAGE-A3, MAGE-A6 (e.g. MAGE-A6 according to accession number NM_005363), MAGE-C1 , MAGE-C2, melan-A (e.g. melan-A according to accession number NM_00551 1 ), GP100 (e.g. GP100 according to accession number M77348), tyrosinase (e.g.
  • tyrosinase according to accession number NM_000372
  • survivin e.g. survivin according to accession number AF077350
  • CEA e.g. CEA according to accession number NM_004363
  • Her-2/neu e.g. Her-2/neu according to accession number M1 1 730
  • WTI e.g. WT1 according to accession number NM_000378
  • PRAME e.g. PRAME according to accession number NM_0061 15
  • EGFRI epidermal growth factor receptor 1
  • EGFRI epidermal growth factor receptor 1
  • mucin-1 e.g.
  • mucin-1 according to accession number NM_002456
  • SEC61 G e.g. SEC61 G according to accession number NM_014302
  • hTERT e.g. hTERT accession number NM_198253
  • 5T4 e.g. 5T4 according to accession number NM_006670
  • NY-Eso-1 e.g. NY-Eso1 according to accession number NM_001327)
  • TRP-2 e.g. TRP-2 according to accession number NM_001922
  • STEAP PCA
  • PSA PSMA
  • PSMA etc.
  • the tumor antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may form a cocktail of antigens, e.g. in an active (immunostimulatory) composition or a kit of parts (wherein preferably each antigen is contained in one part of the kit), preferably for eliciting an (adaptive) immune response for the treatment of prostate cancer (PCa), preferably of neoadjuvant and/or hormone-refractory prostate cancers, and diseases or disorders related thereto.
  • a cocktail of antigens e.g. in an active (immunostimulatory) composition or a kit of parts (wherein preferably each antigen is contained in one part of the kit), preferably for eliciting an (adaptive) immune response for the treatment of prostate cancer (PCa), preferably of neoadjuvant and/or hormone-refractory prostate cancers, and diseases or disorders related thereto.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is preferably at least one RNA, more preferably at least one mRNA, which may encode at least one, preferably two, three or even four (preferably different) antigens of the following group of antigens:
  • PSA Prostate-Specific Antigen
  • KLK3 Kallikrein-3
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be at least one RNA, more preferably at least one mRNA, which may encode at least two, three or four (preferably different) antigens of the following combinations of antigens:
  • PSA and PSCA are PSA and PSCA, or
  • PSMA and PSCA or • PSMA and STEAP, or
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be at least one RNA, more preferably at least one mRNA, which may encode at least two, three or four (preferably different) antigens:
  • At least one antigen is selected from:
  • PSA Prostate-Specific Antigen
  • PSMA Prostate-Specific Membrane Antigen
  • PSCA Prostate Stem Cell Antigen
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be at least one RNA, more preferably at least one mRNA, encoding four (preferably different) antigens selected from PSA, PSMA, PSCA and STEAP.
  • the tumor antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may form a cocktail of antigens, e.g. in an active (immunostimulatory) composition or a kit of parts (wherein preferably each antigen is contained in one part of the kit), preferably for eliciting an (adaptive) immune response for the treatment of non- small cell lung cancers (NSCLC), preferably selected from the three main sub-types squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, or of disorders related thereto.
  • NSCLC non- small cell lung cancers
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is preferably at least one RNA, more preferably at least one mRNA, which may encode at least one, preferably two, three, four, five, six, seven, eight, nine, ten eleven or twelve (preferably different) antigens of the following group of antigens:
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be at least one RNA, more preferably at least one mRNA, which may encode at least two, three, five or six (preferably different) antigens of the following combinations of antigens:
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be at least one RNA, more preferably at least one mRNA, which may encode at least one, preferably two, three, four, five, six, seven, eight, nine, ten eleven or twelve (preferably different) antigens of the following combinations of antigens:
  • VVT1 , 5T4, NY-ESO-1 , and Survivin or • VVTl , 5T4, NY-ESO-1 , and MAGE-C2, or
  • VVT1 VVT1 , 5T4, Survivin, and MAGE-C2, or
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also be at least one RNA, more preferably at least one mRNA, which may encode at least two (preferably different) antigens exclusively selected from any of the antigens of the above mentioned group(s) or subgroup(s) comprising (at least) any one of the following combinations of antigens:
  • the tumor antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may form a cocktail of antigens, e.g. in an active (immunostimulatory) composition or a kit of parts (wherein preferably each antigen is contained in one part of the kit), preferably for eliciting an (adaptive) immune response for the treatment of non- small cell lung cancers (NSCLC), preferably selected from the three main sub-types squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, or of disorders related thereto.
  • NSCLC non- small cell lung cancers
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is preferably at least one RNA, more preferably at least one mRNA, which may encode at least two (preferably different) antigens,
  • At least one, preferably at least two, three, four, five or even six, of these at least two antigens is (are) selected from:
  • the further antigen(s) is (are) selected from at least one antigen as defined herein, preferably in any of the herein mentioned combinations, groups or subgroups of antigens, e.g. the further antigen(s) is (are) selected from, e.g.:
  • the at least one antigen(s) according to a) is (are) selected from:
  • the at least one antigen(s) according to a) is (are) selected from:
  • the at least one antigen(s) according to b) is (are) selected from an antigen (antigens) as defined in one of the following combinations:
  • VVT1 VVT1 , MAGE-A2 and 5T4;
  • the at least one antigen(s) according to b) is (are) selected from the following combination:
  • each of the at least two (preferably different) antigens as defined herein may be encoded by one (monocistronic) RNA, preferably one (monocistronic) mRNA.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may comprise at least two (monocistronic) RNAs, preferably mRNAs, wherein each of these at least two (monocistronic) RNAs, preferably mRNAs, may encode just one (preferably different) antigen, preferably selected from one of the above mentioned combinations.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may comprise (at least) one bi- or even multicistronic RNA, preferably mRNA, i.e. (at least) one RNA which carries two or even more of the coding sequences of at the least two (preferably different) antigens, preferably selected from one of the above mentioned combinations.
  • Such coding sequences of the at least two (preferably different) antigens of the (at least) one bi- or even multicistronic RNA may be separated by at least one IRES (internal ribosomal entry site) sequence, as defined below.
  • the term "encoding at least two (preferably different) antigens” may mean, without being limited thereto, that the (at least) one (bi- or even multicistronic) RNA, preferably a mRNA, may encode e.g. at least two, three, four, five, six, seven, eight, nine, ten, eleven or twelve (preferably different) antigens of the above mentioned group(s) of antigens or their fragments or variants. More preferably, without being limited thereto, the (at least) one (bi- or even multicistronic) RNA, preferably mRNA, may encode e.g.
  • IRES internal ribosomal entry site
  • IRES sequences can function as a sole ribosome binding site, but it can also serve to provide a bi- or even multicistronic RNA as defined above which codes for 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 nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may comprise a mixture of at least one monocistronic RNA, preferably mRNA, as defined above, and at least one bi- or even multicistronic RNA, preferably mRNA, as defined above.
  • the at least one monocistronic RNA and/or the at least one bi- or even multicistronic RNA preferably encode different antigens or their fragments or variants, the antigens preferably being selected from one of the above mentioned groups or subgroups of antigens, more preferably in one of the above mentioned combinations.
  • the at least one monocistronic RNA and the at least one bi- or even multicistronic RNA may preferably also encode (in part) identical antigens selected from one of the above mentioned groups or subgroups of antigens, preferably in one of the above mentioned combinations, provided that the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection as a whole provides at least two (preferably different) antigens as defined above.
  • Such an embodiment may be advantageous e.g. for a staggered, e.g. time dependent, administration of the inventive solution for lyophilization, transfection and/or injection, e.g.
  • RNAs encoding the at least two (preferably different) antigens 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 pharmaceutical compositions, vaccines, lyophilized nucleic acids, etc.
  • one further class of antigens as encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection comprises allergy antigens.
  • Such allergy antigens may be selected from antigens derived from different sources, e.g. from animals, plants, fungi, bacteria, etc. Allergens in this context include e.g. grasses, pollens, molds, drugs, or numerous environmental triggers, etc. Allergy antigens typically belong to different classes of compounds, such as nucleic acids and their fragments, proteins or peptides and their fragments, carbohydrates, polysaccharides, sugars, lipids, phospholipids, etc. Of particular interest in the context of the present invention are antigens, which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection, i.e. protein or peptide antigens and their fragments or epitopes, or nucleic acids and their fragments, particularly nucleic acids and their fragments, encoding such protein or peptide antigens and their fragments or epitopes.
  • antigens derived from animals which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may include antigens derived from, without being limited thereto, insects, such as mite (e.g. house dust mites), mosquito, bee (e.g. honey bee, bumble bee), cockroache, tick, moth (e.g.
  • mite e.g. house dust mites
  • mosquito e.g. honey bee, bumble bee
  • cockroache e.g.
  • Antigens derived from plants, which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may include antigens derived from, without being limited thereto, fruits, such as kiwi, pineapple, jackfruit,papaya, lemon, orange, mandarin, melon, sharon fruit, strawberry, lychee, apple, cherry compassion apple, mango, passion fruit, plum, apricot, nectarine, pear, passion fruit, raspberry, grape, from vegetables, such as garlic, onion, leek, soya bean, celery, cauliflower, turnip, paprika, chickpea, fennel, zucchini, cucumber, carrot, yam, bean, pea, olive, tomato, potato, lentil, lettuce, avocado, parsley, horseradish, chirimoya, beet, pumkin, spinach, from spices, such as mustard, coriander, saffron, pepper, aniseed, from crop,
  • Antigens derived from fungi which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may include antigens derived from, without being limited thereto, e.g. Alternia sp., Aspergillus sp., Beauveria sp., Candida sp., Cladosporium sp., Endothia sp., Curcularia sp., Embellisia sp., Epicoccum sp., Fusarium sp., Malassezia sp., Penicillum sp., Pleospora sp., Saccharomyces sp., etc.
  • Antigens derived from bacteria which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may include antigens derived from, without being limited thereto, e.g. Bacillus tetani, Staphylococcus aureus, Streptomyces griseus, etc. Antibodies
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 cancer 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.
  • “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 lyophilized nucleic acid according to the present invention as well.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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.
  • each lyophilized nucleic acid encoding a specific (monoclonal) antibody being directed to specific antigens or immunogens or epitopes of a protein.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 determinant on the antigen.
  • 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. 4,81 6,567.
  • “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.
  • 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 into separate wells of microtiter plates and grown to produce monoclonal antibodies. The monoclonal antibodies are tested to determine which of them are suitable for detecting the antigen of interest. After being selected, the monoclonal antibodies can be grown in cell cultures or by injecting the hybridomas into mice.
  • the peptide sequences of these monoclonal antibodies have to be sequenced and the at least one nucleic acid (sequence) encoding these antibodies can be present as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • non-human monoclonal or polyclonal antibodies such as murine antibodies may also be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • Chimeric antibodies which may be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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.
  • Swiss Humanized antibodies which may be also encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may encode human antibodies, i.e. antibodies having only human sequences.
  • human antibodies can be isolated from human tissues or from immunized non-human host organisms which are transgene for the human IgG gene locus, and at least one nucleic acid (sequence) may be prepared according to procedures well known in the art. Additionally, human antibodies can be provided by the use of a phage display.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 by two different F ⁇ -domains, 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 by two different F ⁇ -domains, 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 nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also encode intrabodies, wherein these intrabodies may be antibodies as defined above. Since these antibodies are intracellular expressed antibodies, i.e. antibodies which may be 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 nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 also be encoded by the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may also encode antibody fragments selected from Fab, Fab', F(ab') 2 , Fc, Facb, pFc', Fd and Fv fragments of the aforementioned (full-length) antibodies.
  • antibody fragments are known in the art.
  • 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 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 1 5-25 glycine, proline and/or serine residues.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be in the form of dsRNA, preferably siRNA.
  • a dsRNA, or a siRNA is of interest particularly in connection with the phenomenon of RNA interference.
  • RNA interference 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 ).
  • dsRNA double-stranded RNA molecules
  • R and RnaseL protein kinase
  • siRNA small interfering RNA
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may thus be a double-stranded RNA (dsRNA) having a length of from 1 7 to 29, preferably from 19 to 25, and preferably being at least 90%, more preferably 95% and especially 00% (of the nucleotides of a dsRNA) complementary to a section of the nucleic acid (sequence) of a (therapeutically relevant) protein or antigen described (as active ingredient) hereinbefore, either a coding or a non-coding section, preferably a coding section.
  • dsRNA double-stranded RNA
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be a dsRNA having the general structure 5'-(N 17 .
  • each N is a (preferably different) nucleotide of a section of the mRNA of a therapeutically relevant protein or antigen described hereinbefore, preferably being selected from a continuous number of 1 7 to 29 nucleotides of the mRNA of a therapeutically relevant protein or antigen and being present in the general structure 5'-(N 17 _ 29 )-3 ' in their natural order.
  • dsRNAs used as nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection can also be directed against nucleotide sequences of a (therapeutically relevant) protein or antigen described (as active ingredient) hereinbefore that do not lie in the coding region, in particular in the 5' non-coding region of the mRNA, for example, therefore, against non-coding regions of the mRNA having a regulatory function.
  • the target sequence of the dsRNA used as nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection can therefore lie in the translated and untranslated region of the mRNA and/or in the region of the control elements of a protein or antigen described hereinbefore.
  • the target sequence of a dsRNA used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 mRNA.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be in the form of a CpG nucleic acid, in particular CpG-RNA or CpG-DNA.
  • a CpG-RNA or CpG-DNA used according to the invention can be a single-stranded CpG-DNA (ss CpG-DNA), a double-stranded CpG-DNA (dsDNA), a single-stranded CpG-RNA (ss CpG-RNA) or a double-stranded CpG-RNA (ds CpG-RNA).
  • the CpG nucleic acid used according to the invention is preferably in the form of CpG-RNA, more preferably in the form of single-stranded CpG-RNA (ss CpG-RNA). Also preferably, such CpG nucleic acids have a length as described above. Preferably the CpG motifs are unmethylated.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be in the form of an immunostimulatory RNA.
  • an immunostimulatory RNA used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be any (double-stranded or single-stranded) RNA, e.g. a coding RNA, as defined above.
  • the immunostimulatory RNA may be a single-stranded, a double-stranded or a partially double-stranded RNA, more preferably a single-stranded RNA, and/or a circular or linear RNA, more preferably a linear RNA. More preferably, the immunostimulatory RNA may be a (linear) single-stranded RNA. Even more preferably, the immunostimulatory RNA may be a ((linear) single-stranded) messenger RNA (mRNA). An immunostimulatory RNA may also occur as a short RNA oligonucleotide as defined above.
  • An immunostimulatory RNA as used herein may furthermore be selected from any class of RNA molecules, found in nature or being prepared synthetically, and which can induce an immune response.
  • an immune response may occur in various ways.
  • a substantial factor for a suitable immune response is the stimulation of different T-cell sub-populations.
  • T- lymphocytes are typically divided into two sub-populations, the T-helper 1 (Th1 ) cells and the T-helper 2 (Th2) cells, with which the immune system is capable of destroying intracellular (Th1 ) and extracellular (Th2) pathogens (e.g. antigens).
  • the two Th cell populations differ in the pattern of the effector proteins (cytokines) produced by them.
  • Th1 cells assist the cellular immune response by activation of macrophages and cytotoxic T- cells.
  • Th2 cells promote the humoral immune response by stimulation of the B-cells for conversion into plasma cells and by formation of antibodies (e.g. against antigens).
  • the Th1/Th2 ratio is therefore of great importance in the immune response.
  • the Th1 Th2 ratio of the immune response is preferably shifted in the direction towards the cellular response (Th1 response) and a cellular immune response is thereby induced.
  • the immune system may be activated by ligands of Toll-like receptors (TLRs).
  • TLRs are a family of highly conserved pattern recognition receptor (PRR) polypeptides that recognize pathogen- associated molecular patterns (PAMPs) and play a critical role in innate immunity in mammals.
  • PRR pattern recognition receptor
  • TLR1 - TLR13 Toll-like receptors: TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR13
  • TLR1 - TLR13 Toll-like receptors: TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR13
  • CpG DNA unmethylated bacterial DNA and synthetic analogs thereof
  • ligands for certain TLRs include certain nucleic acid molecules and that certain types of RNA are immunostimulatory in a sequence-independent or sequence-dependent manner, wherein these various immunostimulatory RNAs may e.g. stimulate TLR3, TLR7, or TLR8, or intracellular receptors such as RIG-I, MDA-5, etc.
  • these various immunostimulatory RNAs may e.g. stimulate TLR3, TLR7, or TLR8, or intracellular receptors such as RIG-I, MDA-5, etc.
  • Lipford et a/ determined certain G,U- containing oligoribonucleotides as immunostimulatory by acting via TLR7 and TLR8 (see WO 03/086280).
  • RNA sources including ribosomal RNA, transfer RNA, messenger RNA, and viral RNA.
  • any RNA molecule as e.g. defined above (irrespective of its specific length, strandedness, modification and/or nucleotide sequence) may have immunostimulatory properties, i.e. enhance the immune response.
  • An RNA as defined above and being the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may thus be used to enhance (unspecific) immunostimulation, if suitable and desired for a specific treatment.
  • the at least one (immunostimulatory) RNA (molecule) used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may thus comprise any RNA sequence known to be immunostimulatory, including, without being limited thereto, RNA sequences representing and/or encoding ligands of TLRs, preferably selected from family members TLR1 - TLR13, more preferably from TLR7 and TLR8, ligands for intracellular receptors for RNA (such as RIG-I or MAD-5, etc.) (see e.g. Meylan, E., Tschopp, J. (2006). Toll-like receptors and RNA helicases: two parallel ways to trigger antiviral responses. Mol.
  • immunostimulatory RNA molecules used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may include any other RNA capable of eliciting an immune response.
  • an immunostimulatory RNA may include ribosomal RNA (rRNA), transfer RNA (tRNA), messenger RNA (mRNA), and viral RNA (vRNA).
  • Such further (classes of) immunostimulatory RNA molecules which may be used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection, without being limited thereto, may comprise e.g. an RNA molecule of formula (I):
  • G is guanosine, uracil or an analogue of guanosine or uracil;
  • X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above- mentioned nucleotides
  • n is an integer and is at least 3;
  • n is an integer from 1 to 40
  • n > 1 at least 50% of the nucleotides are guanosine or an analogue thereof.
  • C is cytosine, uracil or an analogue of cytosine or uracil;
  • X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above- mentioned nucleotides
  • I is an integer from 1 to 40
  • n is an integer and is at least 3;
  • n is an integer from 1 to 40
  • the immunostimulatory RNA molecules used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may comprise a length as defined above in general for RNA molecules of the RNA of the present invention, more preferably a length of 5 to 5000, of 500 to 5000 or, more preferably, of 1000 to 5000 or, alternatively, of 5 to 1000, 5 to 500, 5 to 250, of 5 to 100, of 5 to 50 or, more preferably, of 5 to 30 nucleotides.
  • the immunostimulatory RNA used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be furthermore modified, preferably "chemically modified” in order to enhance the immunostimulatory properties of said RNA.
  • chemical modification means that the immuostimulatory RNA is modified by replacement, insertion or removal of individual or several atoms or atomic groups compared with naturally occurring RNA species.
  • the chemical modification of the immunostimulatory RNA comprises at least one analogue of naturally occurring nucleotides.
  • examples which may be mentioned for nucleotide analogues and which may be used herein for modification are analogues of guanosine, uracil, adenosine, thymidine, cytosine.
  • the modifications may refer to modifications of the base, the ribose moiety and/or the phosphate backbone moiety.
  • analogues of guanosine, uracil, adenosine, and cytosine include, without implying any limitation, any naturally occurring or non-naturally occurring guanosine, uracil, adenosine, thymidine or cytosine that has been altered chemically, for example by acetylation, methylation, hydroxylation, etc., including 1 - methyl-adenosine, 1 -methyl-guanosine, 1 -methyl-inosine, 2,2-dimethyl-guanosine, 2,6- diaminopurine, 2'-Amino-2'-deoxyadenosine, 2'-Amino-2'-deoxycytidine, 2'-Amino-2'- deoxyguanosine, 2'-Amino-2'-deoxyuridine, 2-Amino-6-chloropurineriboside, 2- Aminopurine-riboside, 2'-Araaden
  • analogue as described above, particular preference is given according to the invention to those analogues that increase the immunogenicity of the immunostimulatory RNA sequence used as the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection and/or do not interfere with a further modification that has been introduced into said immunostimulatory RNA.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection as defined above may also occur in the form of a modified nucleic acid, wherein any modification, as defined herein, may be introduced into the nucleic acid prior to lyophilization, transfection and/or injection. Modifications as defined herein preferably lead to a further stabilized nucleic acid as defined herein.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may thus be provided as a "stabilized nucleic acid", preferably as a stabilized RNA, more preferably as an RNA that is essentially resistant to in vivo degradation (e.g. by an exo- or endo-nuclease).
  • stabilization can be effected, for example, by a modified phosphate backbone of the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • a backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides contained in the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection are chemically modified.
  • Nucleotides that may be preferably used in this connection contain e.g. a phosphorothioate- modified phosphate backbone, preferably at least one of the phosphate oxygens contained in the phosphate backbone being replaced by a sulfur atom.
  • Stabilized at least one nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may further include, for example: non-ionic phosphate analogues, such as, for example, alkyl and aryl phosphonates, in which the charged phosphonate oxygen is replaced by an alkyl or aryl group, or phosphodiesters and alkylphosphotriesters, in which the charged oxygen residue is present in alkylated form.
  • Such backbone modifications typically include, without implying any limitation, modifications from the group consisting of methylphosphonates, phosphoramidates and phosphorothioates (e.g. cytidine-5'-0-(1 - thiophosphate)).
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may additionally or alternatively also contain sugar modifications.
  • a sugar modification in connection with the present invention is a chemical modification of the sugar of the nucleotides of the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection typically includes, without implying any limitation, sugar modifications selected from the group consisting of 2'-deoxy-2'-fluoro- oligoribonucleotide (2 '-fluoro-2'-deoxycytidine-5 '-triphosphate, 2'-fluoro-2'-deoxyuridine- 5'-triphosphate), 2'-deoxy-2'-deamine oligoribonucleotide (2'-amino-2'-deoxycytidine-5'- triphosphate, 2'-amino-2'-deoxyuridine-5'-triphosphate), 2'-0-alkyl oligoribonucleotide,
  • Significant in this case means an increase in the expression of the protein compared with the expression of the native nucleic acid (sequence) by at least 20%, preferably at least 30%, 40%, 50% or 60%, more preferably by at least 70%, 80%, 90% or even 100% and most preferably by at least 150%, 200% or even 300% or more.
  • a nucleotide having such a base modification is preferably selected from the group of the base-modified nucleotides consisting of 2-amino- 6-chloropurineriboside-5'-triphosphate, 2-aminoadenosine-5'-triphosphate, 2-thiocytidine- 5'-triphosphate, 2-thiouridine-5'-triphosphate, 4-thiouridine-5'-triphosphate, 5- aminoallylcytidine-5'-triphosphate, 5-aminoallyluridine-5'-triphosphate, 5-bromocytidine- 5 '-triphosphate, 5-bromouridine-5'-triphosphate, 5-iodocytidine-5'-triphosphate, 5- iodouridine-5'-triphosphate, 5-methylcytidine-5'-triphosphate, 5-methyluridine-5'- triphosphate, 6-azacytidine-5 '-triphosphate, 6-azauridine-5'-triphosphate, 6- chloropur
  • nucleotides for base modifications selected from the group of base-modified nucleotides consisting of 5- methylcytidine-5'-triphosphate, 7-deazaguanosine-5'-triphosphate, 5-bromocytidine-5'- triphosphate, and pseudouridine-5'-triphosphate.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection can likewise be modified (and preferably stabilized) by introducing further modified nucleotides containing modifications of their ribose or base moieties.
  • nucleotide analogues are defined as non-natively occurring variants of naturally occurring nucleotides.
  • analogues are chemically derivatized nucleotides with non-natively occurring functional groups, which are preferably added to or deleted from the naturally occurring nucleotide or which substitute the naturally occurring functional groups of a nucleotide. Accordingly, each component of the naturally occurring nucleotide may be modified, namely the base component, the sugar (ribose) component and/or the phosphate component forming the backbone (see above) of the nucleic acid sequence.
  • Exemplary analogues of guanosine, uracil, adenosine, and cytosine include, without implying any limitation, any naturally occurring or non-naturally occurring guanosine, uracil, adenosine, thymidine or cytosine that has been altered chemically, for example by acetylation, methylation, hydroxylation, etc., including 1 -methyl-adenosine, 1 -methyl-guanosine, 1 - methyl-inosine, 2,2-dimethyl-guanosine, 2,6-diaminopurine, 2'-Amino-2'-deoxyadenosine, 2'- Amino-2'-deoxycytidine, 2'-Amino-2'-deoxyguanosine, 2'-Amino-2'-deoxyuridine, 2-Amino-6- chloropurineriboside, 2-Aminopurine-riboside, 2'-Araadeno
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection can contain a lipid modification.
  • a lipid-modified nucleic acid typically comprises a nucleic acid as defined herein.
  • Such a lipid-modified nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection typically further comprises at least one linker covalently linked with that nucleic acid, and at least one lipid covalently linked with the respective linker.
  • the lipid-modified nucleic acid comprises an at least one nucleic acid as defined herein and at least one (bifunctional) lipid covalently linked (without a linker) with that nucleic acid.
  • the lipid-modified nucleic acid comprises a nucleic acid RNA as defined herein, at least one linker covalently linked with that nucleic acid, and at least one lipid covalently linked with the respective linker, and also at least one (bifunctional) lipid covalently linked (without a linker) with that nucleic acid.
  • the lipid which may be contained in the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection (complexed or covalently bound thereto) is typically a lipid or a lipophilic residue that preferably is itself biologically active.
  • lipids preferably include natural substances or compounds such as, for example, vitamins, e.g. alpha-tocopherol (vitamin E), including RRR-alpha-tocopherol (formerly D-alpha- tocopherol), L-alpha-tocopherol, the racemate D,L-alpha-tocopherol, vitamin E succinate (VES), or vitamin A and its derivatives, e.g.
  • retinoic acid retinol
  • vitamin D and its derivatives e.g. vitamin D and also the ergosterol precursors thereof
  • vitamin E and its derivatives vitamin K and its derivatives, e.g. vitamin K and related quinone or phytol compounds, or steroids, such as bile acids, for example cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxygenin, testosterone, cholesterol or thiocholesterol.
  • bile acids for example cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxygenin, testosterone, cholesterol or thiocholesterol.
  • Further lipids or lipophilic residues within the scope of the present invention include, without implying any limitation, polyalkylene glycols (Oberhauser et a/., Nucl.
  • Acids Res., 1 992, 20, 533) aliphatic groups such as, for example, C1 -C20-alkanes, C1 -C20-alkenes or C1 -C20-alkanol compounds, etc., such as, for example, dodecanediol, hexadecanol or undecyl residues (Saison-Behmoaras et al., EMBO J, 1 991 , 1 0, 1 1 1 ; Kabanov et a/., FEBS Lett., 1990, 259, 327; Svinarchuk et a/., Biochimie, 1993, 75, 49), phospholipids such as, for example, phosphatidylglycerol, diacylphosphatidylglycerol, phosphatidylcholine, dipalmitoylphosphatidylcholine, distearoylphosphatidylcholine, phosphatidylserine, phosphat
  • polyamines or polyalkylene glycols such as, for example, polyethylene glycol (PEG) (Manoharan et a/., Nucleosides & Nucleotides, 1 995, 14, 969), hexaethylene glycol (HEG), palmitin or palmityl residues (Mishra et a/., Biochim. Biophys. Acta, 1995, 1 264, 229), octadecylamines or hexylamino-carbonyl-oxycholesterol residues (Crooke et a/., J. Pharmacol. Exp. Ther., 1996, 277, 923), and also waxes, terpenes, alicyclic hydrocarbons, saturated and mono- or poly-unsaturated fatty acid residues, etc.
  • PEG polyethylene glycol
  • HEG hexaethylene glycol
  • HOG hexaethylene glycol
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may likewise be stabi lized in order to prevent degradation of the nucleic acid by various approaches, particularly, when RNA or mRNA is used as a nucleic acid for the inventive purpose.
  • RNAases ribonucleases
  • the natural degradation of mRNA in the cytoplasm of cells is very finely regulated and RNase contaminations may be generally removed by special treatment prior to use of said compositions, in particular with diethyl pyrocarbonate (DEPC).
  • DEPC diethyl pyrocarbonate
  • a number of mechanisms of natural degradation are known in this connection in the prior art, which may be utilized as well.
  • the terminal structure is typically of critical importance for an mRNA.
  • cap structure a modified guanosine nucleotide
  • the so-called poly-A tail is typically a sequence of up to 200 adenosine nucleotides
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection can therefore be stabilized against degradation by RNases by the addition of a so-called "5' cap” structure.
  • m7G(5')ppp 5'(A,G(5')ppp(5')A or G(5')ppp(5')G as the 5' cap" structure.
  • such a modification is introduced only if a modification, for example a lipid modification, has not already been introduced at the 5' end of the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection, if provided as a mRNA or if the modification does not interfere with the immunogenic properties of the (unmodified or chemically modified) nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection.
  • a modification for example a lipid modification
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may contain, especially if the nucleic acid is in the form of a mRNA, a poly-A tail on the 3' terminus of typically about 10 to 200 adenosine nucleotides, preferably about 10 to 100 adenosine nucleotides, more preferably about 20 to 100 adenosine nucleotides or even more preferably about 40 to 80 adenosine nucleotides.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may contain, especially if the nucleic acid is in the form of a mRNA, a poly-C tail on the 3' terminus of typically about 10 to 200 cytosine nucleotides, preferably about 10 to 100 cytosine nucleotides, more preferably about 20 to 70 cytosine nucleotides or even more preferably about 20 to 60 or even 10 to 40 cytosine nucleotides.
  • the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection may be modified, and thus stabilized, especially if the nucleic acid is in the form of a mRNA, by modifying the G/C content of the nucleic acid, particularly an mRNA, preferably of the coding region thereof.
  • the G/C content of the coding region of the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is modified, particularly increased, compared to the G/C content of the coding region of its particular wild type mRNA, i.e. the unmodified mRNA.
  • the encoded amino acid sequence of the at least one mRNA is preferably not modified compared to the coded amino acid sequence of the particular wild type mRNA.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is based on the fact that the sequence of any mRNA region to be translated is important for efficient translation of that mRNA.
  • composition and the sequence of various nucleotides are 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 mRNA are therefore varied compared to its wild type mRNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides.
  • the most favorable codons for the stabi lity can be determined (so-called alternative codon usage).
  • nucleic acid sequence
  • transfection and/or injection especially if the nucleic acid is in the form of a mRNA
  • nucleic acid is in the form of a mRNA
  • amino acids which are encoded by codons which contain exclusively G or C nucleotides no modification of the codon is necessary.
  • the codons for Pro (CCC or CCG), Arg (CGC or CGG), Ala (GCC or GCG) and Gly (GGC or GGG) require no modification, since no A or U is present.
  • codons which contain A and/or U nucleotides can be 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 modified from CCU or CCA to CCC or CCG;
  • the codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG; the codons for Ala can be modified from GCU or GCA to GCC or GCG; the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • the codons for Leu can be modified from UUA, UUG, CUU or CUA to CUC or CUG;
  • the codons for Ser can be modified from UCU or UCA or AGU to UCC, UCG or AGC; the codon for Tyr can be modified from UAU to UAC;
  • the codon for Cys can be modified from UGU to UGC;
  • the codon for His can be modified from CAU to CAC;
  • the codon for Gin can be modified from CAA to CAG;
  • the codons for lie can be modified from AUU or AUA to AUC;
  • codons for Thr can be modified from ACU or ACA to ACC or ACG;
  • the codon for Asn can be modified from AAU to AAC;
  • the codon for Lys can be modified from AAA to AAG;
  • the codons for Val can be modified from GUU or GUA to GUC or GUG;
  • the codon for Asp can be modified from GAU to GAC;
  • the codon for Glu can be modified from GAA to GAG;
  • the stop codon UAA can be modified to UAG or UGA.
  • substitutions listed above can be used either individually or in all possible combinations to increase the G/C content of the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection, especially if the nucleic acid is in the form of a mRNA, compared to its particular wild type mRNA (i.e. the original sequence).
  • all codons for Thr occurring in the wild type sequence can be modified to ACC (or ACG).
  • substitution possibilities substitution of all codons coding for Thr in the original sequence (wild type m NA) to ACC
  • the G/C content of the coding region of the nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection 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 coded region of the wild type mRNA.
  • At least 5%, 10%, 20%, 30%, 40%, 50%, 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 peptide as defined herein or its fragment or variant thereof or the whole sequence of the wild type mRNA sequence are substituted, thereby increasing the GC/content of said sequence.
  • nucleic acid (sequence) of the inventive solution for lyophilization, transfection and/or injection is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.

Abstract

La présente invention concerne (l'utilisation d') une solution contenant au moins un(e) (séquence d') acide(s) nucléique(s) et du mannose libre pour lyophilisation, transfection et/ou injection, en particulier d'ARN et d'ARNm. La solution de l'invention présente un effet positif sur la stabilisation de l'/la (séquence d') acide(s) nucléique(s) pendant la lyophilisation et le stockage mais conduit également à une augmentation considérable de l'efficacité de transfection d'un acide nucléique. Elle augmente donc également l'expression in vivo d'une protéine codée par un tel acide nucléique par l'intermédiaire d'un taux de transfection augmenté. La présente invention concerne en outre un procédé de lyophilisation utilisant la solution contenant du mannose, des compositions pharmaceutiques, des vaccins, des kits, des première et deuxième indications médicales appliquant une telle solution contenant du mannose et/ou un(e) tel(le) (séquence d') acide(s) nucléique(s) lyophilisé(e) ou remis(e) en suspension avec une telle solution.()
PCT/EP2009/008804 2009-12-09 2009-12-09 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques WO2011069529A1 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
PCT/EP2009/008804 WO2011069529A1 (fr) 2009-12-09 2009-12-09 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques
PCT/EP2010/006788 WO2011069586A1 (fr) 2009-12-09 2010-11-08 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques
US13/509,564 US20120258046A1 (en) 2009-12-09 2010-11-08 Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
EP10779700.3A EP2510100B1 (fr) 2009-12-09 2010-11-08 Solution contenant mannose pour la lyophilisation, transfection et/ou injection d'acides nucléiques
US14/492,334 US9616084B2 (en) 2009-12-09 2014-09-22 Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
US15/451,675 US20170182081A1 (en) 2009-12-09 2017-03-07 Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2009/008804 WO2011069529A1 (fr) 2009-12-09 2009-12-09 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques

Publications (1)

Publication Number Publication Date
WO2011069529A1 true WO2011069529A1 (fr) 2011-06-16

Family

ID=42126466

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2009/008804 WO2011069529A1 (fr) 2009-12-09 2009-12-09 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques
PCT/EP2010/006788 WO2011069586A1 (fr) 2009-12-09 2010-11-08 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/006788 WO2011069586A1 (fr) 2009-12-09 2010-11-08 Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques

Country Status (2)

Country Link
US (3) US20120258046A1 (fr)
WO (2) WO2011069529A1 (fr)

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013120500A1 (fr) * 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation en vue d'augmenter l'expression d'un antigène tumoral codé
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
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
WO2016000792A1 (fr) * 2014-07-04 2016-01-07 Biontech Ag Préparations d'arn stabilisées
US9234013B2 (en) 2010-08-13 2016-01-12 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded protein
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9447431B2 (en) 2012-02-15 2016-09-20 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2016184575A1 (fr) * 2015-05-20 2016-11-24 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longue
WO2016184576A3 (fr) * 2015-05-20 2016-12-29 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longue
WO2016184577A3 (fr) * 2015-05-20 2017-01-12 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longu
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
US9669089B2 (en) 2012-02-15 2017-06-06 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
EP3178488A1 (fr) * 2012-02-15 2017-06-14 CureVac AG Acide nucléique comprenant ou codant pour une tige-boucle d'histone et séquence poly(a) ou signal de polyadénylation pour augmenter l'expression d'une protéine codée
US9683233B2 (en) 2012-03-27 2017-06-20 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
WO2017214175A1 (fr) * 2016-06-07 2017-12-14 Modernatx, Inc. Arn modifié codant pour des polypeptides vegf-a, formulations et utilisations associées
CN108148870A (zh) * 2018-01-12 2018-06-12 中国人民解放军第四军医大学 一种提高壳聚糖体外转运siRNA效率的方法
US10047375B2 (en) 2013-12-30 2018-08-14 Curevac Ag Artificial nucleic acid molecules
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
US10080809B2 (en) 2012-03-27 2018-09-25 Curevac Ag Artificial nucleic acid molecules comprising a 5′TOP UTR
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
US10232024B2 (en) 2012-02-15 2019-03-19 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US10507183B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Cleavable lipids
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11254951B2 (en) 2014-12-30 2022-02-22 Curevac Ag Artificial nucleic acid molecules
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
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11446250B2 (en) 2015-04-17 2022-09-20 Curevac Real Estate Gmbh Lyophilization of RNA
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US11697816B2 (en) 2013-12-30 2023-07-11 CureVac SE Artificial nucleic acid molecules
US11696892B2 (en) 2017-10-31 2023-07-11 Modernatx, Inc. Lipid nanoparticles for delivering modified RNA encoding a VEGF-A polypeptide
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines

Families Citing this family (183)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009030254A1 (fr) 2007-09-04 2009-03-12 Curevac Gmbh Complexes d'arn et de peptides cationiques pour transfection et immunostimulation
ES2460899T3 (es) 2008-03-03 2014-05-14 The University Of Miami Inmunoterapia basada en células cancerosas alógenas
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
ES2759015T3 (es) 2010-02-10 2020-05-07 Mayo Found Medical Education & Res Métodos y materiales para tratar el cáncer
BR112013002298A2 (pt) 2010-07-30 2016-05-24 Curevac Gmbh complexação de ácidos nucleicos com componentes catiônicos reticulados com dissulfeto para transfecção e estimulação imunológica.
WO2012089225A1 (fr) 2010-12-29 2012-07-05 Curevac Gmbh Combinaison de vaccination et d'inhibition de la présentation des antigènes restreinte par le cmh de classe i
WO2012116715A1 (fr) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des nouveaux-nés et des enfants en bas âge
WO2012113413A1 (fr) 2011-02-21 2012-08-30 Curevac Gmbh Composition de vaccin comprenant des acides nucléiques immunostimulateurs complexés et des antigènes emballés avec des conjugués de polyéthylèneglycol/peptide à liaison disulfure
SI2717893T1 (sl) 2011-06-08 2019-10-30 Translate Bio Inc Sestavki lipidnih nanodelcev in postopki za dostavo mRNA
WO2013113326A1 (fr) 2012-01-31 2013-08-08 Curevac Gmbh Composition pharmaceutique comprenant un complexe support polymère - charge et au moins un antigène de protéine ou de peptide
US9517258B2 (en) 2012-03-15 2016-12-13 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
CN104220599A (zh) 2012-03-27 2014-12-17 库瑞瓦格有限责任公司 人工核酸分子
SG10201710472PA (en) 2013-02-22 2018-02-27 Curevac Ag Combination of vaccination and inhibition of the pd-1 pathway
WO2014159813A1 (fr) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Molécules polynucléotidiques à longue durée de vie
KR102311614B1 (ko) 2013-03-14 2021-10-08 샤이어 휴먼 지네틱 테라피즈 인크. Cftr mrna 조성물 및 관련 방법 및 사용
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
AU2014239184B2 (en) 2013-03-14 2018-11-08 Translate Bio, Inc. Methods and compositions for delivering mRNA coded antibodies
EP2972360B1 (fr) 2013-03-15 2018-03-07 Translate Bio, Inc. Stimulation synergique de l'administration d'acides nucléiques par le biais de formulations mélangées
CN112386604A (zh) 2013-04-29 2021-02-23 纪念斯隆-凯特琳癌症中心 cGAS的调节剂
CN110195072A (zh) 2013-08-21 2019-09-03 库瑞瓦格股份公司 狂犬病疫苗
RU2733424C2 (ru) 2013-08-21 2020-10-01 Куревак Аг Способ повышения экспрессии кодируемых рнк белков
RU2723328C2 (ru) 2013-08-21 2020-06-09 Куревак Аг Вакцина против респираторно-синцитиального вируса (рсв)
WO2015024669A1 (fr) 2013-08-21 2015-02-26 Curevac Gmbh Vaccin combiné
CN105408751A (zh) * 2013-10-31 2016-03-16 Sk电信有限公社 用于诊断胰腺癌的组合物以及使用该组合物诊断胰腺癌的方法
ES2806575T3 (es) 2013-11-01 2021-02-18 Curevac Ag ARN modificado con propiedades inmunoestimuladoras disminuidas
CN107075515B (zh) 2013-11-22 2020-10-30 米纳治疗有限公司 C/EBPα组合物和使用方法
SG10201805660WA (en) 2013-12-30 2018-08-30 Curevac Ag Methods for rna analysis
ES2754239T3 (es) 2014-03-12 2020-04-16 Curevac Ag Combinación de vacunación y agonistas de OX40
WO2015143221A1 (fr) 2014-03-19 2015-09-24 Mayo Foundation For Medical Education And Research Méthodes et matériels pour le traitement du cancer
WO2015143223A1 (fr) 2014-03-19 2015-09-24 Mayo Foundation For Medical Education And Research Procédés et matériel pour traiter le cancer
US10369216B2 (en) 2014-04-01 2019-08-06 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
EP3151866B1 (fr) 2014-06-09 2023-03-08 Voyager Therapeutics, Inc. Capsides chimériques
ES2727776T3 (es) 2014-06-10 2019-10-18 Curevac Ag Método para mejorar la producción de ARN
CA2955250A1 (fr) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Polynucleotides chimeriques
WO2016014846A1 (fr) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Polynucléotides modifiés destinés à la production d'anticorps intracellulaires
EP3215191A4 (fr) 2014-11-05 2018-08-01 Voyager Therapeutics, Inc. Polynucléotides codant pour la dopa décarboxylase et destinés au traitement de la maladie de parkinson
JP6863891B2 (ja) 2014-11-14 2021-04-21 ボイジャー セラピューティクス インコーポレイテッドVoyager Therapeutics,Inc. 調節性ポリヌクレオチド
CN114717264A (zh) 2014-11-14 2022-07-08 沃雅戈治疗公司 治疗肌萎缩性侧索硬化(als)的组合物和方法
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
WO2016091391A1 (fr) 2014-12-12 2016-06-16 Curevac Ag Molécules d'acides nucléiques artificielles destinées à améliorer l'expression de protéines
SG11201705844SA (en) 2015-02-06 2017-08-30 Heat Biologics Inc Vector co-expressing vaccine and costimulatory molecules
EP4353257A2 (fr) 2015-04-13 2024-04-17 CureVac Manufacturing GmbH Procédé de production de compositions d'arn
RU2749113C2 (ru) 2015-04-22 2021-06-04 Куревак Аг Содержащая рнк композиция для лечения опухолевых заболеваний
WO2016174271A1 (fr) 2015-04-30 2016-11-03 Curevac Ag Poly(n)polymérase immobilisée
DK3294885T3 (da) 2015-05-08 2020-08-10 Curevac Real Estate Gmbh Fremgangsmåde til at fremstille rna
MX2017014538A (es) 2015-05-15 2018-03-02 Curevac Ag Regimenes de cebado-refuerzo que implican la administracion de al menos un constructo de arnm.
US11608513B2 (en) 2015-05-29 2023-03-21 CureVac SE Method for adding cap structures to RNA using immobilized enzymes
DE202016009003U1 (de) 2015-05-29 2021-05-28 Curevac Real Estate Gmbh Zusammensetzung umfassend in vitro transkribierte RNA erhältlich durch ein Verfahren zur Herstellung und Reinigung von RNA mit mindestens einem Schritt mit einer tangentialen Flussfiltration
WO2017009376A1 (fr) 2015-07-13 2017-01-19 Curevac Ag Procédé de production d'arn à partir d'adn circulaire et adn matriciel correspondant
AU2016324310B2 (en) 2015-09-17 2021-04-08 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
CA3001003A1 (fr) 2015-10-05 2017-04-13 Modernatx, Inc. Procedes d'administration therapeutique de medicaments a base d'acide ribonucleique messager
EP3362576A1 (fr) 2015-10-12 2018-08-22 CureVac AG Procédé automatisé d'isolement, de sélection et/ou de détection de micro-organismes ou de cellules présents dans une solution
WO2017066782A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffes d'arnm hydrophobes
WO2017066781A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm à liaison phosphate modifié
WO2017066789A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm avec sucre modifié
WO2017066791A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm à substitution sucre
EP3362460A1 (fr) 2015-10-16 2018-08-22 Modernatx, Inc. Analogues de coiffes arnm et procédés de coiffage d'arnm
WO2017067592A1 (fr) * 2015-10-21 2017-04-27 Biontech Ag Particules immunostimulantes cytotoxiques et utilisation de ces dernières
WO2017081110A1 (fr) 2015-11-09 2017-05-18 Curevac Ag Vaccins contre les rotavirus
JP7114465B2 (ja) 2015-12-22 2022-08-08 モデルナティエックス インコーポレイテッド 薬剤の細胞内送達のための化合物および組成物
WO2017109134A1 (fr) 2015-12-22 2017-06-29 Curevac Ag Procédé de production de compositions de molécules d'arn
EP3394280A1 (fr) 2015-12-23 2018-10-31 CureVac AG Procédé de transcription in vitro d'arn utilisant un tampon contenant un acide dicarboxyliqlue ou un acide tricarboxylique ou un sel de celui-ci
EP3394093B1 (fr) 2015-12-23 2022-01-26 Modernatx, Inc. Procédés d'utilisation de polynucléotides codant pour un ligand ox40
WO2017120612A1 (fr) 2016-01-10 2017-07-13 Modernatx, Inc. Arnm thérapeutiques codant pour des anticorps anti-ctla-4
WO2017140905A1 (fr) 2016-02-17 2017-08-24 Curevac Ag Vaccin contre le virus zika
WO2017149139A1 (fr) 2016-03-03 2017-09-08 Curevac Ag Analyse d'arn par hydrolyse totale
EP3825400A1 (fr) 2016-04-08 2021-05-26 Translate Bio Ma, Inc. Acide nucléique codant multimère et ses utilisations
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
EP3448874A4 (fr) 2016-04-29 2020-04-22 Voyager Therapeutics, Inc. Compositions pour le traitement de maladies
EP3452101A2 (fr) 2016-05-04 2019-03-13 CureVac AG Arn codant pour une protéine thérapeutique
US11141474B2 (en) 2016-05-04 2021-10-12 Curevac Ag Artificial nucleic acid molecules encoding a norovirus antigen and uses thereof
WO2017201258A1 (fr) 2016-05-18 2017-11-23 Voyager Therapeutics, Inc. Compositions et méthodes de traitement de la maladie de huntington
BR112018073384A2 (pt) 2016-05-18 2019-03-06 Voyager Therapeutics, Inc. polinucleotídeos moduladores
WO2017212009A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour cargo d'acides nucléiques
CN109312313A (zh) 2016-06-13 2019-02-05 川斯勒佰尔公司 用于治疗鸟氨酸转氨甲酰酶缺乏症的信使rna疗法
WO2017218704A1 (fr) 2016-06-14 2017-12-21 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
WO2018010815A1 (fr) * 2016-07-15 2018-01-18 Biontech Rna Pharmaceuticals Gmbh Formule pour l'administration d'arn.
EP3506817A4 (fr) 2016-08-30 2020-07-22 The Regents of The University of California Procédés de ciblage et d'administration biomédicaux, et dispositifs et systèmes pour la mise en oeuvre de ceux-ci
WO2018071405A1 (fr) 2016-10-11 2018-04-19 University Of Miami Vecteurs et cellules de vaccin pour immunité contre le virus zika
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
US11279923B2 (en) 2016-11-28 2022-03-22 Curevac Ag Method for purifying RNA
WO2018104540A1 (fr) 2016-12-08 2018-06-14 Curevac Ag Arn pour la cicatrisation des plaies
US11464836B2 (en) 2016-12-08 2022-10-11 Curevac Ag RNA for treatment or prophylaxis of a liver disease
CN110494549A (zh) * 2016-12-22 2019-11-22 阿维塔斯有限公司 通过可逆性渗透对非粘附细胞的无载体细胞内递送
EP3558354A1 (fr) 2016-12-23 2019-10-30 CureVac AG Vaccin contre le virus de lassa
WO2018115507A2 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre l'hénipavirus
US11141476B2 (en) 2016-12-23 2021-10-12 Curevac Ag MERS coronavirus vaccine
MA47603A (fr) 2017-02-27 2020-01-01 Translate Bio Inc Nouvel arnm cftr à codons optimisés
CA3055653A1 (fr) 2017-03-15 2018-09-20 Modernatx, Inc. Formulation de nanoparticules lipidiques
AU2018234692B2 (en) 2017-03-15 2022-06-02 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
US20200085944A1 (en) 2017-03-17 2020-03-19 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
EP3601576A1 (fr) 2017-03-24 2020-02-05 CureVac AG Acides nucléiques codant pour des protéines associées à crispr et leurs utilisations
WO2018187260A1 (fr) 2017-04-04 2018-10-11 Heat Biologics, Inc. Vaccination intratumorale
WO2018191657A1 (fr) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipides pour administration d'agents actifs
US11752181B2 (en) 2017-05-05 2023-09-12 Voyager Therapeutics, Inc. Compositions and methods of treating Huntington's disease
WO2018204786A1 (fr) 2017-05-05 2018-11-08 Voyager Therapeutics, Inc. Compositions et méthodes de traitement de la sclérose latérale amyotrophique (sla)
IL270631B2 (en) 2017-05-16 2024-03-01 Translate Bio Inc Treatment of cystic fibrosis through the administration of mRNA with an optimal codon encoding ctfr
CA3063723A1 (fr) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides codant pour des polypeptides d'interleukine-12 (il12) ancres et leurs utilisations
WO2018232006A1 (fr) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucléotides codant pour le facteur viii de coagulation
EP3638678A1 (fr) 2017-06-14 2020-04-22 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents
JOP20190269A1 (ar) 2017-06-15 2019-11-20 Voyager Therapeutics Inc بولي نوكليوتيدات aadc لعلاج مرض باركنسون
BR112019028280A2 (pt) 2017-07-04 2020-07-14 Curevac Ag moléculas de ácido nucleico
JP7229989B2 (ja) 2017-07-17 2023-02-28 ボイジャー セラピューティクス インコーポレイテッド 軌道アレイガイドシステム
WO2019036638A1 (fr) 2017-08-18 2019-02-21 Modernatx, Inc. Procédés de préparation d'arn modifié
WO2019038332A1 (fr) 2017-08-22 2019-02-28 Curevac Ag Vaccin contre les bunyavirus
MX2020002348A (es) 2017-08-31 2020-10-08 Modernatx Inc Métodos de elaboración de nanopartículas lipídicas.
WO2019048645A1 (fr) 2017-09-08 2019-03-14 Mina Therapeutics Limited Compositions stabilisées de petits arn activateurs (parna) de cebpa et procédés d'utilisation
EP3679139B1 (fr) 2017-09-08 2022-11-02 MiNA Therapeutics Limited Compositions stabilisées de petits arn activateurs (parna) de hnf4a et procédés d'utilisation
US20200237799A1 (en) 2017-10-16 2020-07-30 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
WO2019079240A1 (fr) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Traitement de la sclérose latérale amyotrophique (sla)
JP2021502079A (ja) 2017-11-08 2021-01-28 キュアバック アーゲー RNA配列の適合(Adaptation)
WO2019104152A1 (fr) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucléotides codant pour l'ornithine transcarbamylase pour le traitement de troubles du cycle de l'urée
WO2019104195A1 (fr) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucléotides codant pour des sous-unités alpha et bêta de propionyl-coa carboxylase pour le traitement de l'acidémie propionique
JP7423521B2 (ja) 2017-11-22 2024-01-29 モダーナティエックス・インコーポレイテッド フェニルケトン尿症の治療用のフェニルアラニンヒドロキシラーゼをコードするポリヌクレオチド
US11931406B2 (en) 2017-12-13 2024-03-19 CureVac SE Flavivirus vaccine
MA51306A (fr) 2017-12-20 2020-10-28 Translate Bio Inc Compositions et procédés améliorés pour le traitement du déficit en ornithine transcarbamylase
US11525158B2 (en) 2017-12-21 2022-12-13 CureVac SE Linear double stranded DNA coupled to a single support or a tag and methods for producing said linear double stranded DNA
WO2019193183A2 (fr) 2018-04-05 2019-10-10 Curevac Ag Nouvelles molécules d'acide nucléique de fièvre jaune pour la vaccination
WO2019200171A1 (fr) 2018-04-11 2019-10-17 Modernatx, Inc. Arn messager comprenant des éléments d'arn fonctionnels
EP3775211B1 (fr) 2018-04-12 2023-04-05 MiNA Therapeutics Limited Compositions
US20210170017A1 (en) 2018-04-17 2021-06-10 Curevac Ag Novel rsv rna molecules and compositions for vaccination
JP2021527651A (ja) 2018-06-15 2021-10-14 ミナ セラピューティクス リミテッド C/EBPアルファsaRNAを含む併用療法
EP3813874A1 (fr) 2018-06-27 2021-05-05 CureVac AG Nouvelles molécules d'arn de virus lassa et compositions pour la vaccination
KR20210027368A (ko) 2018-06-28 2021-03-10 큐어백 아게 알엔에이 시험관 내 전사를 위한 바이오리액터
KR20210027493A (ko) * 2018-07-19 2021-03-10 주식회사 헬릭스미스 네이키드 dna 유전자 요법을 위한 동결 건조된 약제학적 조성물
WO2020023390A1 (fr) 2018-07-25 2020-01-30 Modernatx, Inc. Traitement enzymatique substitutif basé sur l'arnm combiné à un chaperon pharmacologique pour le traitement de troubles du stockage lysosomal
WO2020047201A1 (fr) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucléotides codant pour l'acyl-coa déshydrogénase à très longue chaîne pour le traitement de l'insuffisance en acyl-coa déshydrogénase à très longue chaîne
EP3849594A2 (fr) 2018-09-13 2021-07-21 Modernatx, Inc. Polynucléotides codant pour les sous-unités e1-alpha, e1-beta et e2 du complexe alpha-cétoacide déshydrogénase à chaîne ramifiée pour le traitement de la leucinose
US20230009009A1 (en) 2018-09-13 2023-01-12 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020056239A1 (fr) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucléotides codant pour le polypeptide a1, de la famille de l'uridine diphosphate glycosyltransférase 1, pour le traitement du syndrome de crigler-najjar
JP7410135B2 (ja) 2018-09-19 2024-01-09 モデルナティエックス インコーポレイテッド 治療薬の細胞内送達のための化合物及び組成物
WO2020061457A1 (fr) 2018-09-20 2020-03-26 Modernatx, Inc. Préparation de nanoparticules lipidiques et leurs méthodes d'administration
US20220152225A1 (en) 2018-09-27 2022-05-19 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
EP3897702A2 (fr) 2018-12-21 2021-10-27 CureVac AG Arn pour vaccins antipaludiques
CA3128215A1 (fr) 2019-01-31 2020-08-06 Modernatx, Inc. Procedes de preparation de nanoparticules lipidiques
SG11202108098QA (en) 2019-01-31 2021-08-30 Modernatx Inc Vortex mixers and associated methods, systems, and apparatuses thereof
CA3125511A1 (fr) 2019-02-08 2020-08-13 Curevac Ag Arn codant administre dans l'espace suprachoroidien pour le traitement de maladies ophtalmiques
US20220211740A1 (en) 2019-04-12 2022-07-07 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2020227642A1 (fr) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions pour peau et plaies et leurs méthodes d'utilisation
WO2020254535A1 (fr) 2019-06-18 2020-12-24 Curevac Ag Vaccin à arnm rotavirus
CN114585633A (zh) 2019-08-19 2022-06-03 米纳治疗有限公司 寡核苷酸缀合物组合物和使用方法
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
DE202021004130U1 (de) 2020-02-04 2022-10-26 Curevac Ag Coronavirus-Vakzine
EP4077272A1 (fr) 2020-04-09 2022-10-26 Suzhou Abogen Biosciences Co., Ltd. Compositions de nanoparticules lipidiques
TW202204622A (zh) 2020-04-09 2022-02-01 大陸商蘇州艾博生物科技有限公司 針對冠狀病毒之核酸疫苗
MX2022015132A (es) 2020-05-29 2023-03-08 CureVac SE Vacunas combinadas a base de acidos nucleicos.
CN111588859B (zh) * 2020-06-02 2021-09-03 成都可恩生物科技有限公司 一种冻干保护剂及其应用和冻干苗及其制备方法
KR20230030588A (ko) 2020-06-30 2023-03-06 쑤저우 아보젠 바이오사이언시스 컴퍼니 리미티드 지질 화합물 및 지질 나노입자 조성물
EP4172194A1 (fr) 2020-07-31 2023-05-03 CureVac SE Mélanges d'anticorps codés par des acides nucléiques
KR20230054672A (ko) 2020-08-20 2023-04-25 쑤저우 아보젠 바이오사이언시스 컴퍼니 리미티드 지질 화합물 및 지질 나노입자 조성물
WO2022043551A2 (fr) 2020-08-31 2022-03-03 Curevac Ag Vaccins contre le coronavirus à base d'acides nucléiques multivalents
EP4208286A1 (fr) 2020-09-01 2023-07-12 CureVac RNA Printer GmbH Dispositif de fabrication d'un produit pharmaceutique
US20230406895A1 (en) 2020-11-13 2023-12-21 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
CA3205569A1 (fr) 2020-12-22 2022-06-30 CureVac SE Vaccin a arn contre des variants sras-cov-2
CA3171051A1 (fr) 2020-12-22 2022-06-30 Curevac Ag Composition pharmaceutique comprenant des vecteurs lipidique encapsulant de l'arn pour une administration multidose
WO2022137133A1 (fr) 2020-12-22 2022-06-30 Curevac Ag Vaccin à arn contre des variants sras-cov-2
WO2022152109A2 (fr) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
CN116615472A (zh) 2021-01-14 2023-08-18 苏州艾博生物科技有限公司 聚合物缀合的脂质化合物和脂质纳米颗粒组合物
WO2022162027A2 (fr) 2021-01-27 2022-08-04 Curevac Ag Procédé de réduction des propriétés immunostimulatrices d'arn transcrit in vitro
CN112877294B (zh) * 2021-02-23 2021-08-24 赛浦生物科技(长春)有限公司 基因修饰的间充质干细胞外泌体的制备及其应用
AU2022246144A1 (en) 2021-03-26 2023-09-21 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
EP4313152A1 (fr) 2021-03-26 2024-02-07 GlaxoSmithKline Biologicals S.A. Compositions immunogènes
CA3171429A1 (fr) 2021-03-31 2022-09-30 Alexander SCHWENGER Seringues contenant des compositions pharmaceutiques comprenant de l'arn
EP4334446A1 (fr) 2021-05-03 2024-03-13 CureVac SE Séquence d'acide nucléique améliorée pour l'expression spécifique de type cellulaire
WO2022247755A1 (fr) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
CA3231523A1 (fr) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Lipides acycliques et leurs procedes d'utilisation
WO2023044333A1 (fr) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Lipides cycliques et leurs procédés d'utilisation
AR127312A1 (es) 2021-10-08 2024-01-10 Suzhou Abogen Biosciences Co Ltd Compuestos lipídicos ycomposiciones de nanopartículas lipídicas
CN116064598B (zh) 2021-10-08 2024-03-12 苏州艾博生物科技有限公司 冠状病毒的核酸疫苗
AU2022358824A1 (en) 2021-10-08 2024-04-11 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
WO2023099884A1 (fr) 2021-12-01 2023-06-08 Mina Therapeutics Limited Compositions d'arnsa de pax6 et procédés d'utilisation
CN114191379A (zh) * 2021-12-17 2022-03-18 西安交通大学医学院第一附属医院 以Pluronic F127为载体进行在体局部转基因的修复皮肤损伤药物及制备方法
WO2023122752A1 (fr) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Lipides contraints et procédés d'utilisation associés
WO2023144330A1 (fr) 2022-01-28 2023-08-03 CureVac SE Inhibiteurs de facteurs de transcription codés par un acide nucleique
WO2023170435A1 (fr) 2022-03-07 2023-09-14 Mina Therapeutics Limited Compositions de petits arn activateurs d'il10 et procédés d'utilisation
WO2023196931A1 (fr) 2022-04-07 2023-10-12 Renagade Therapeutics Management Inc. Lipides cycliques et nanoparticules lipidiques (npl) pour l'apport d'acides nucléiques ou de peptides destinés à être utilisés dans la vaccination contre des agents infectieux
WO2023218019A1 (fr) 2022-05-12 2023-11-16 Etherna Immunotherapies Nv Composition d'arn lyophilisée
WO2023227608A1 (fr) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Vaccin à base d'acide nucléique codant pour un polypeptide antigénique fimh d'escherichia coli
WO2024037578A1 (fr) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition de nanoparticules lipidiques
WO2024068545A1 (fr) 2022-09-26 2024-04-04 Glaxosmithkline Biologicals Sa Vaccins contre le virus de la grippe
US11833224B1 (en) 2023-02-08 2023-12-05 Leuvian Llc Lyoprotectant compositions and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009125986A2 (fr) * 2008-04-09 2009-10-15 Viromed Co., Ltd. Formulations lyophilisées d'adn pour une expression améliorée d'un adn plasmidique

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4401796A (en) 1981-04-30 1983-08-30 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5849473A (en) * 1991-02-15 1998-12-15 Cobe Laboratories, Inc. Method of lyophilization of mammalian sperm cells
US5955448A (en) * 1994-08-19 1999-09-21 Quadrant Holdings Cambridge Limited Method for stabilization of biological substances during drying and subsequent storage and compositions thereof
IL114848A0 (en) * 1994-08-16 1995-12-08 Chile Lab Sa New composition and subcompositions of same process for obtaining them and their molecular identification and their anti-inflammatory analgesic antipruritic and local antipyretic therapeutic effect in human beings and animals
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
EP1818409A1 (fr) 1999-09-09 2007-08-15 CureVac GmbH Transfer de mARN à l'aide de composés polycationiques
ES2340499T3 (es) 2001-06-05 2010-06-04 Curevac Gmbh Arnm de antigeno tumoral estabilizado con un contenido de g/c aumentado.
DE10162480A1 (de) 2001-12-19 2003-08-07 Ingmar Hoerr Die Applikation von mRNA für den Einsatz als Therapeutikum gegen Tumorerkrankungen
CA2480775C (fr) 2002-04-04 2016-05-31 Coley Pharmaceutical Gmbh Oligoribonucleotides immunostimulants contenant de la guanine et de l'uracile
DE10229872A1 (de) 2002-07-03 2004-01-29 Curevac Gmbh Immunstimulation durch chemisch modifizierte RNA
DE10335833A1 (de) 2003-08-05 2005-03-03 Curevac Gmbh Transfektion von Blutzellen mit mRNA zur Immunstimulation und Gentherapie
DE102004042546A1 (de) 2004-09-02 2006-03-09 Curevac Gmbh Kombinationstherapie zur Immunstimulation
DE102005023170A1 (de) 2005-05-19 2006-11-23 Curevac Gmbh Optimierte Formulierung für mRNA
JP2010507361A (ja) 2006-07-31 2010-03-11 キュアバック ゲーエムベーハー 具体的には免疫刺激剤/アジュバントとしての、一般式(I):GlXmGn、または一般式(II):ClXmCnで表される核酸
DE102006061015A1 (de) 2006-12-22 2008-06-26 Curevac Gmbh Verfahren zur Reinigung von RNA im präparativen Maßstab mittels HPLC
DE102007001370A1 (de) 2007-01-09 2008-07-10 Curevac Gmbh RNA-kodierte Antikörper
EP2125030A2 (fr) * 2007-01-29 2009-12-02 Medipol S.A. Particules colloïdales à base de chitosane pour la délivrance d'arn
WO2009030254A1 (fr) 2007-09-04 2009-03-12 Curevac Gmbh Complexes d'arn et de peptides cationiques pour transfection et immunostimulation
WO2009046739A1 (fr) 2007-10-09 2009-04-16 Curevac Gmbh Composition pour traiter le cancer de la prostate (pca)
WO2009046738A1 (fr) 2007-10-09 2009-04-16 Curevac Gmbh Composition pour traiter le cancer du poumon, notamment les cancers du poumon non à petites cellules (nsclc)
SG188104A1 (en) 2008-01-31 2013-03-28 Curevac Gmbh Nucleic acids comprising formula (nuglxmgnnv)a and derivatives thereof as an immunostimulating agents /adjuvants
WO2010037408A1 (fr) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprenant un arnm complexé et un arnm nu pour déclencher ou augmenter une réponse immunostimulante chez un mammifère et utilisations de ladite composition
JP5804453B2 (ja) 2009-05-14 2015-11-04 国立大学法人 東京大学 結晶性ポリオール微粒子及びその調製方法
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009125986A2 (fr) * 2008-04-09 2009-10-15 Viromed Co., Ltd. Formulations lyophilisées d'adn pour une expression améliorée d'un adn plasmidique

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
JONES K.L., DRANE D. & GOWANS E.J.: "Long-term storage of DNA-free RNA for use in vaccine studies", BIOTECHNIQUES, vol. 43, no. 5, 2007, pages 675 - 681, XP002581476 *
NAKAMURA K. ET AL.: "Enhanced gene transfection in macrophages by Histidine-conjugated mannosylated cationic liposomes", BIOL. PHARM. BULL., vol. 32, no. 9, September 2009 (2009-09-01), pages 1628 - 1631, XP002581475 *
POXON S W ET AL: "The effect of lyophilization on plasmid DNA activity", PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY, NEW YORK, NY, US, vol. 5, no. 1, 1 January 2000 (2000-01-01), pages 115 - 122, XP009131956, ISSN: 1083-7450 *
SMITH STEVE ET AL: "Optimal storage conditions for highly dilute DNA samples: a role for trehalose as a preserving agent", JOURNAL OF FORENSIC SCIENCES, CALLAGHAN AND CO, CHICAGO, IL, US LNKD- DOI:10.1520/JFS2004411, vol. 50, no. 5, 1 January 2005 (2005-01-01), pages 1101 - 1108, XP009097021, ISSN: 0022-1198 *

Cited By (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, 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
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, 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
US9839697B2 (en) 2010-08-13 2017-12-12 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
US10653799B2 (en) 2010-08-13 2020-05-19 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded protein
US9234013B2 (en) 2010-08-13 2016-01-12 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded protein
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, 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
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9950068B2 (en) 2011-03-31 2018-04-24 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
US10702478B2 (en) 2011-06-08 2020-07-07 Translate Bio, Inc. Cleavable lipids
US11234936B2 (en) 2011-06-08 2022-02-01 Translate Bio, Inc. Cleavable lipids
US10507183B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Cleavable lipids
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
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
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
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
US10682406B2 (en) 2012-02-15 2020-06-16 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
US9447431B2 (en) 2012-02-15 2016-09-20 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
EP3348644A1 (fr) * 2012-02-15 2018-07-18 CureVac AG Acide nucléique comprenant ou codant pour une tige-boucle d'histone et séquence poly(a) ou signal de polyadénylation pour augmenter l'expression d'une protéine codée
US10912826B2 (en) 2012-02-15 2021-02-09 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
US10898589B2 (en) 2012-02-15 2021-01-26 Cure Vac AG Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US9669089B2 (en) 2012-02-15 2017-06-06 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
US10010592B2 (en) 2012-02-15 2018-07-03 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
US11110156B2 (en) 2012-02-15 2021-09-07 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
US10111968B2 (en) 2012-02-15 2018-10-30 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US10166283B2 (en) 2012-02-15 2019-01-01 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
US10232024B2 (en) 2012-02-15 2019-03-19 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
CN108396033A (zh) * 2012-02-15 2018-08-14 库瑞瓦格股份公司 增加编码的肿瘤抗原表达的包含或编码组蛋白茎环和聚腺苷酸序列或聚腺苷酸化信号的核酸
US10799577B2 (en) 2012-02-15 2020-10-13 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
WO2013120500A1 (fr) * 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation en vue d'augmenter l'expression d'un antigène tumoral codé
WO2013120627A1 (fr) * 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'un antigène tumoral codé
EP3178488A1 (fr) * 2012-02-15 2017-06-14 CureVac AG Acide nucléique comprenant ou codant pour une tige-boucle d'histone et séquence poly(a) ou signal de polyadénylation pour augmenter l'expression d'une protéine codée
RU2650795C2 (ru) * 2012-02-15 2018-04-17 Кьюрвак Аг Нуклеиновая кислота, содержащая или кодирующая гистоновую структуру типа"стебель-петля" и поли(а)-последовательность или сигнал полиаденилирования, для увеличения экспрессии кодируемого опухолевого антигена
EP3527662A1 (fr) * 2012-02-15 2019-08-21 CureVac AG Acide nucléique comprenant ou codant pour une tige-boucle d'histone et séquence poly(a) ou signal de polyadénylation pour augmenter l'expression d'une protéine codée
CN104105792B (zh) * 2012-02-15 2018-03-09 库瑞瓦格股份公司 用于增加编码的肿瘤抗原表达的包含或编码组蛋白茎环和聚腺苷酸序列或聚腺苷酸化信号的核酸
US10610605B2 (en) 2012-02-15 2020-04-07 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
CN104105792A (zh) * 2012-02-15 2014-10-15 库瑞瓦格有限责任公司 用于增加编码的肿瘤抗原表达的包含或编码组蛋白茎环和聚腺苷酸序列或聚腺苷酸化信号的核酸
US10738306B2 (en) 2012-03-27 2020-08-11 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
US10080809B2 (en) 2012-03-27 2018-09-25 Curevac Ag Artificial nucleic acid molecules comprising a 5′TOP UTR
US9683233B2 (en) 2012-03-27 2017-06-20 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of 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
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
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and 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
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, 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
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10047375B2 (en) 2013-12-30 2018-08-14 Curevac Ag Artificial nucleic acid molecules
US11697816B2 (en) 2013-12-30 2023-07-11 CureVac SE Artificial nucleic acid molecules
WO2016000792A1 (fr) * 2014-07-04 2016-01-07 Biontech Ag Préparations d'arn stabilisées
US20170136129A1 (en) * 2014-07-04 2017-05-18 Biontech Ag Stabilised Formulations of RNA
US10626400B2 (en) 2014-07-04 2020-04-21 Biontech Ag Stabilised formulations of RNA
US11254951B2 (en) 2014-12-30 2022-02-22 Curevac Ag Artificial nucleic acid molecules
US11491112B2 (en) 2015-04-17 2022-11-08 CureVac Manufacturing GmbH Lyophilization of RNA
US11446250B2 (en) 2015-04-17 2022-09-20 Curevac Real Estate Gmbh Lyophilization of RNA
WO2016184575A1 (fr) * 2015-05-20 2016-11-24 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longue
WO2016184577A3 (fr) * 2015-05-20 2017-01-12 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longu
US10517827B2 (en) 2015-05-20 2019-12-31 Curevac Ag Dry powder composition comprising long-chain RNA
CN107530448A (zh) * 2015-05-20 2018-01-02 库瑞瓦格股份公司 包含长链rna的干粉组合物
US11179337B2 (en) 2015-05-20 2021-11-23 Curevac Ag Dry powder composition comprising long-chain RNA
EP3928800A3 (fr) * 2015-05-20 2022-03-23 CureVac AG Composition de poudre sèche comprenant de l'arn à longue chaîne
EP3916091A3 (fr) * 2015-05-20 2022-03-30 CureVac AG Composition de poudre sèche comprenant de l'arn à longue chaîne
EP3297682B1 (fr) 2015-05-20 2021-07-14 CureVac AG Composition de poudre sèche comprenant de l'arn à chaîne longue
US11433027B2 (en) 2015-05-20 2022-09-06 Curevac Ag Dry powder composition comprising long-chain RNA
WO2016184576A3 (fr) * 2015-05-20 2016-12-29 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longue
CN107635587A (zh) * 2015-05-20 2018-01-26 库瑞瓦格股份公司 包含长链rna的干燥粉末组合物
US11534405B2 (en) 2015-05-20 2022-12-27 Curevac Ag Dry powder composition comprising long-chain RNA
US10729654B2 (en) 2015-05-20 2020-08-04 Curevac Ag Dry powder composition comprising long-chain RNA
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US10702597B2 (en) 2015-07-21 2020-07-07 Modernatx, Inc. CHIKV RNA vaccines
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10517940B2 (en) 2015-10-22 2019-12-31 Modernatx, Inc. Zika virus RNA vaccines
US10238731B2 (en) 2015-10-22 2019-03-26 Modernatx, Inc. Chikagunya virus RNA vaccines
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10702600B1 (en) 2015-10-22 2020-07-07 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US10272150B2 (en) 2015-10-22 2019-04-30 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10675342B2 (en) 2015-10-22 2020-06-09 Modernatx, Inc. Chikungunya virus RNA vaccines
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10716846B2 (en) 2015-10-22 2020-07-21 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
US10702599B2 (en) 2015-10-22 2020-07-07 Modernatx, Inc. HPIV3 RNA vaccines
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US10543269B2 (en) 2015-10-22 2020-01-28 Modernatx, Inc. hMPV RNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
WO2017214175A1 (fr) * 2016-06-07 2017-12-14 Modernatx, Inc. Arn modifié codant pour des polypeptides vegf-a, formulations et utilisations associées
JP2021100961A (ja) * 2016-06-07 2021-07-08 モデルナティエックス インコーポレイテッドModernaTX,Inc. Vegf−aポリペプチドをコードする改変rna、製剤、およびそれに関連する使用
US11866475B2 (en) 2016-06-07 2024-01-09 Modernatx, Inc. Modified RNA encoding VEGF-A polypeptides, formulations, and uses relating thereto
JP7167226B2 (ja) 2016-06-07 2022-11-08 モデルナティエックス インコーポレイテッド Vegf-aポリペプチドをコードする改変rna、製剤、およびそれに関連する使用
RU2756313C2 (ru) * 2016-06-07 2021-09-29 Модернатикс, Инк. Модифицированная РНК, кодирующая полипептиды VEGF-A, составы, содержащие ее, и пути их применения
EP3971204A1 (fr) * 2016-06-07 2022-03-23 Modernatx, Inc. Arn modifié codant pour des polypeptides vegf-a, formulations et utilisations associées
TWI818375B (zh) * 2016-06-07 2023-10-11 美商瑪德那特克斯股份有限公司 編碼vegf-a多肽的經修飾rna、配製物及關於它們之用途
JP2019521976A (ja) * 2016-06-07 2019-08-08 モデルナティエックス インコーポレイテッドModernaTX,Inc. Vegf−aポリペプチドをコードする改変rna、製剤、およびそれに関連する使用
TWI752036B (zh) * 2016-06-07 2022-01-11 美商瑪德那特克斯股份有限公司 編碼vegf-a多肽的經修飾rna、配製物及關於它們之用途
CN109790207A (zh) * 2016-06-07 2019-05-21 摩登纳特斯有限公司 编码vegf-a多肽的经修饰的rna、配制品和关于它们的用途
CN109790207B (zh) * 2016-06-07 2023-06-20 摩登纳特斯有限公司 编码vegf-a多肽的经修饰的rna、配制品和关于它们的用途
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
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
US11696946B2 (en) 2016-11-11 2023-07-11 Modernatx, Inc. Influenza vaccine
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11918644B2 (en) 2017-03-15 2024-03-05 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
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
US11696892B2 (en) 2017-10-31 2023-07-11 Modernatx, Inc. Lipid nanoparticles for delivering modified RNA encoding a VEGF-A polypeptide
CN108148870A (zh) * 2018-01-12 2018-06-12 中国人民解放军第四军医大学 一种提高壳聚糖体外转运siRNA效率的方法
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine

Also Published As

Publication number Publication date
WO2011069586A1 (fr) 2011-06-16
US20150141498A1 (en) 2015-05-21
US20170182081A1 (en) 2017-06-29
US9616084B2 (en) 2017-04-11
US20120258046A1 (en) 2012-10-11

Similar Documents

Publication Publication Date Title
US20210060175A1 (en) Methods of immunostimulation with complexes of rna and cationic carriers
US9616084B2 (en) Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
US20110053829A1 (en) Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
WO2011069587A1 (fr) Lyophilisation d'acides nucléiques dans des solutions contenant du lactate
WO2010088927A1 (fr) Utilisation de pei pour l'amélioration de la libération endosomale et de l'expression d'acides nucléiques transfectés, complexés par des composés cationiques ou polycationiques
EP2510100B1 (fr) Solution contenant mannose pour la lyophilisation, transfection et/ou injection d'acides nucléiques

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: 09767986

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: 09767986

Country of ref document: EP

Kind code of ref document: A1