WO2017212007A1 - Cationic carriers for nucleic acid delivery - Google Patents

Cationic carriers for nucleic acid delivery Download PDF

Info

Publication number
WO2017212007A1
WO2017212007A1 PCT/EP2017/064057 EP2017064057W WO2017212007A1 WO 2017212007 A1 WO2017212007 A1 WO 2017212007A1 EP 2017064057 W EP2017064057 W EP 2017064057W WO 2017212007 A1 WO2017212007 A1 WO 2017212007A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
rna
composition
sequence
diseases
Prior art date
Application number
PCT/EP2017/064057
Other languages
French (fr)
Inventor
Patrick Baumhof
Carolin THIELE
Original Assignee
Curevac Ag
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 Ag filed Critical Curevac Ag
Priority to EP17730740.2A priority Critical patent/EP3468609A1/en
Priority to US16/308,595 priority patent/US20190336608A1/en
Publication of WO2017212007A1 publication Critical patent/WO2017212007A1/en

Links

Classifications

    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/02Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C215/04Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated
    • C07C215/06Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and acyclic
    • C07C215/14Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and acyclic the nitrogen atom of the amino group being further bound to hydrocarbon groups substituted by amino groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/02Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C215/40Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton with quaternised nitrogen atoms bound to carbon atoms of the carbon skeleton
    • 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
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle

Definitions

  • the present invention is in the fields of medical therapy, disease prevention and drug delivery. It relates in particular to carriers that are useful for delivering certain types of active ingredients to subjects in need thereof. More specifically, the invention relates to the delivery of such active ingredients which represent bioactive compounds that are challenging to deliver across biological barriers to their targets within a living organism, such as to target organs, tissues, or cells. Examples of such bioactive compounds that are of great therapeutic value and at the same time difficult to deliver to their biological targets include nucleic acid-based vaccines and therapeutics.
  • gene therapeutic approaches have been developed as a specific form of such treatments which require the transfection of cells or tissues with genes and their insertion into the DNA of the cells, e.g. in the case of hereditary diseases in which a defective mutant allele is replaced with a functional one.
  • Transfer or insertion of nucleic acids or genes into an individual's cells still represents a major challenge today, even though it is absolutely necessary for achieving a significant therapeutic effect of the gene therapy.
  • nucleic acids or genes into an individual's cells
  • a number of different hurdles have to be passed.
  • the transport of nucleic acids typically occurs via association of the nucleic acid with the cell membrane and subsequent uptake by the endosomes.
  • the introduced nucleic acids are separated from the cytosol. As expression occurs in the cytosol, these nucleic acids have to depart the endosome. If the nucleic acids do not leave the endosome before the endosome fuses with a lysosome, they will suffer the usual fate of the content of the endosome and become degraded. Alternatively, the endosome may fuse with the cell membrane, leading to the return of its content into the extracellular medium. For efficient transfer of nucleic acids, the endosomal escape thus appears to be one of the most important steps additionally to the efficiency of transfection itself. Until now, there are different approaches addressing these issues. However, no approach has been entirely successful in all aspects so far.
  • Transfection agents used in the art today typically include various types of peptides, polymers, lipids, as well as other carrier compounds, which may be assembled into nano- or microparticles (see e.g. Gao, X., K. S. Kim, et al. (2007), AAPS J 9(1): E92-104). Most of these transfection agents have been successfully used only in in vitro reactions. When transfecting cells of a living animal with nucleic acids, further requirements have to be fulfilled. As an example, the complex of the nucleic acid and the carrier has to be stable in physiological salt solutions with respect to agglomeration. Furthermore, it must not interact with parts of the complement system of the host.
  • the complex must protect the nucleic acid from early extracellular degradation by ubiquitously occurring nucleases.
  • the carrier is not recognized by the adaptive immune system (immunogenicity) and does not stimulate an unspecific cytokine storm (acute immune response) (see Gao, Kim et al, (2007, supra); Martin, M. E. and K. G. Rice (2007), AAPS J 9(1): E18- 29; and Foerg and Merkle, (2008, supra)).
  • Foerg and Merkle discuss the therapeutic potential of peptide-, protein and nucleic acid-based drugs. According to their analysis, the full therapeutic potential of these drugs is frequently compromised by their limited ability to cross the plasma membrane of mammalian cells, resulting in poor cellular access and inadequate therapeutic efficacy. Today this hurdle represents a major challenge for the biomedical development and commercial success of many biopharmaceuticals.
  • Gao et al. (Gao et al. The AAPS Journal 2007; 9(1) Article 9) see the primary challenge for gene therapy in the development of a method that delivers a therapeutic gene to selected cells where proper gene expression can be achieved.
  • Gene delivery and particularly successful introduction of nucleic acids into cells or tissue is, however, not simple and typically dependent on many factors.
  • For successful delivery e.g., delivery of nucleic acids or genes into cells or tissue, many barriers must be overcome.
  • an ideal gene delivery method needs to meet 3 major criteria: (1) it should protect the transgene against degradation by nucleases in intercellular matrices, (2) it should bring the transgene across the plasma membrane and (3) it should have no detrimental effects.
  • viral or non-viral vectors or carriers typically, these viral or non-viral vectors must be able to overcome the above-mentioned barriers.
  • viral vectors such as adenoviruses, adeno-associated viruses, retroviruses, and herpes viruses.
  • Viral vectors are able to mediate gene transfer with high efficiency and the possibility of long-term gene expression, and satisfy 2 out of 3 criteria.
  • non-viral vectors are not as efficient as viral vectors, many non-viral vectors have been developed to provide safer alternatives in gene therapy.
  • Methods of non-viral gene delivery have been explored using physical (carrier-free gene delivery) and chemical approaches (synthetic vector-based gene delivery).
  • Physical approaches usually include simple injection using injection needles, electroporation, gene gun, ultrasound, and hydrodynamic delivery. Some of these approaches employ a physical force that permeates the cell membrane and facilitates intracellular gene transfer.
  • the chemical approaches typically use synthetic or naturally occurring compounds, e.g. cationic lipids or cationic polymers, as carriers to deliver the transgene into cells.
  • CPPs cell penetrating peptides
  • PTDs protein-transduction domains
  • CPP mediated drug delivery a major obstacle to CPP mediated drug delivery is thought to consist in the often rapid metabolic clearance of the peptides when in contact or passing the enzymatic barriers of epithelia and endothelia. Consequently, the metabolic stability of CPPs represents an important
  • peptide ligands can be short sequences taken from larger proteins that represent the essential amino acids needed for receptor recognition, such as EGF peptide used to target cancer cells.
  • Other peptide ligands have been identified including the ligands used to target the lectin-like oxidized LDL receptor (LOX-1). Up-regulation of LOX-1 in endothelial cells is associated with dysfunctional states such as hypertension and atherosclerosis.
  • LOX-1 lectin-like oxidized LDL receptor
  • Such peptide ligands are not suitable for many gene therapeutic approaches, as they cannot be linked to their cargo molecules by complexation or adhesion but require covalent bonds, e.g. crosslinkers, which typically exhibit cytotoxic effects in the cell.
  • Synthetic vectors may also be used for delivering cargo molecules into cells, e.g., for the purpose of gene therapy.
  • one main disadvantage of many synthetic vectors is their poor transfection efficiency compared to viral vectors and significant improvements are required to enable further clinical development.
  • Several barriers that limit nucleic acid transfer both in vitro and in vivo have been identified, and include poor intracellular delivery, toxicity and instability of vectors in physiological conditions (see. e.g. Read, M. L., K. H. Bremner, et al. (2003) : Vectors based on reducible polycations facilitate intracellular release of nucleic acids. J Gene Med 5(3): 232-45).
  • cationic or cationisable lipids show excellent transfection activity in cell culture, most do not perform well in the presence of serum, and only a few are active in vivo.
  • a dramatic change in size, surface charge, and lipid composition occurs when lipoplexes are exposed to the overwhelming amount of negatively charged and often amphipathic proteins and polysaccharides that are present in blood, mucus epithelial lining fluid, or tissue matrix.
  • lipoplexes tend to interact with negatively charged blood components and form large aggregates that could be absorbed onto the surface of circulating red blood cells, trapped in a thick mucus layer or embolized in microvasculatures, preventing them from reaching the intended target cells in the distal location. Furthermore, toxicity related to lipoplexes has been observed. Symptomes include inter alia induction of inflammatory cyokines. In humans, various degrees of adverse inflammatory reactions, including flu-like symptoms were noted among subjects who received lipoplexes. Accordingly, it appears questionable as to whether lipoplexes can be safely used in humans, in particular when repeated administration is required. One further approach in gene therapy utilizes cationic or cationisable polymers.
  • polyethylenimine PEI
  • polyamidoamine and polypropylamine dendrimers polyallylamine
  • cationic dextran chitosan
  • various proteins and peptides various proteins and peptides.
  • cationic or cationisable polymers share the function of condensing DNA into small particles and facilitating cellular uptake via endocytosis through charge-charge interaction with anionic sites on cell surfaces, their transfection activity and toxicity differ dramatically.
  • cationic or cationisable polymers exhibit better transfection efficiency with rising molecular weight due to stronger complexation of the negatively charged nucleic acid cargo.
  • a rising molecular weight also leads to a rising toxicity of the polymer.
  • PEI is perhaps the most active and most studied polymer for gene delivery, but its main drawback as a transfection reagent relates to its nonbiodegradable nature and toxicity. Furthermore, even though polyplexes formed by high molecular weight polymers exhibit improved stability under physiological conditions, data have indicated that such polymers can hinder vector unpacking. For example, poly(L-lysine) (PLL) of 19 and 36 amino acid residues was shown to dissociate from DNA more rapidly than PLL of 180 residues resulting in significantly enhanced short-term gene expression. A minimum length of six to eight cationic amino acids is required to compact DNA into structures active in receptor- mediated gene delivery. However, polyplexes formed with short polycations are unstable under physiological conditions and typically aggregate rapidly in physiological salt solutions.
  • Nucleic Acids Res 33(9): e86) developed a new type of synthetic vector based on a linear reducible polycation (RPC) prepared by oxidative polycondensation of the peptide Cys-Lysio-Cys that can be cleaved by the intracellular environment to facilitate release of nucleic acids. They could show that polyplexes formed by RPC are destabilised by reducing conditions enabling efficient release of DNA and mRNA. Cleavage of the RPC also reduced toxicity of the polycation to levels comparable with low molecular weight peptides. The disadvantage of this approach of Read et al. (2003, supra) was that the
  • the present prior art as exemplified above suffers from various disadvantages.
  • One particular disadvantage of the self-crosslinking peptides as described by Read et al. (2003, supra) or McKenzie et al. (2000 I and II, supra and US 6,770,740 Bl) concerns the high positive charge on the surface of the particles formed. Due to this charge, the particles exhibit a high instability towards agglomeration when subjecting these particles in vivo to raised salt concentrations. Such salt concentrations, however, typically occur in vivo in cells or extracellular media. Furthermore, complexes with a high positive charge show a strong tendency of opsonization.
  • a reversible derivatization of carriers with a stealthing agent being advantageous for in vivo gene delivery was only possible for peptide monomers but not for self-crosslinking peptides or rather for a polymeric carrier with a defined polymer chain length.
  • a reversible derivatization was not possible at the terminal ends of the crosslinked cationic peptide carrier.
  • high-molecular polymers with long polymer chains or with an undefined polymer chain length consisting of self-crosslinking peptides were described, which unfortunately compact their cargo to such an extent that cargo release in the cell is limited..
  • the extremely undefined polymer chain length is further problematic regarding regulatory approvement of a medicament based on RPC.
  • One precondition for such approvement is that every preparation of the medicament has the same composition, the same structure and the same properties. This cannot be ensured for complexes based on RPC's from the prior art.
  • the RPC-based polymers or complexes provided in the prior art are difficult to characterize due to their undefined structure or polymer chain length.
  • the object underlying the present invention is therefore to provide a carrier, particularly for the delivery of nucleic acids for therapeutic or prophylactic applications, which is capable of compacting the nucleic acids and which allows their efficient introduction into different cell lines in vitro but also enables transfection in vivo. As uptake by cells occurs via the endosomal route, such a carrier or a complexing agent should also allow or provide for efficient release of the nucleic acid from endosomes.
  • a further object is to provide a carrier that upon complexation with a nucleic acid exhibits resistance to agglomeration.
  • a yet further object is to provide enhanced stability to the nucleic acid cargo with respect to serum containing media. Another object is to enable efficient in vivo activity without a strong acute immune reaction.
  • a further object is to provide a composition that is particularly suitable for local delivery of a nucleic acid compound. Another object is to provide a means for administering a nucleic acid compound to a specific biological target while avoiding or reducing systemic exposure. A further object is to overcome any of the disadvantages or limitations of the known carriers for nucleic acid delivery as described e.g. herein-above. Further objects that are addressed by the present invention will become clear on the basis of the following description, the examples and the patent claims.
  • the invention provides a composition comprising a cationisable or permanently cationic lipid or lipidoid and a nucleic acid compound, wherein the lipid and the nucleic acid compound are non-covalently associated, and wherein the ratio of the lipid to the nucleic acid compound is not higher than about 2 nmol lipid per ⁇ g nucleic acid compound.
  • the composition comprises a permanently cationic lipid or lipidoid.
  • the nucleic acid compound may, for example, be any chemically modified or unmodified DNA or RNA.
  • the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound may form a complex.
  • Such complex may not be soluble in an aqueous evironment, so that the composition would typically comprise the complex in the form of a nanoparticle or a plurality of nanoparticles.
  • the nanoparticles comprise a permanently cationic lipid or lipidoid.
  • the invention is directed to such nanoparticles.
  • the nanoparticle - and thus the composition - may comprise further one or more other constituents, such as a targeting agent, a cell penetrating agent, and/or a stealth agent.
  • the invention provides a kit for the preparation of the composition or of the nanoparticles.
  • the kit may comprise a first kit component comprising the cationisable or permanently cationic lipid or lipidoid and a second kit component comprising the nucleic acid compound.
  • a yet further aspect of the invention is the medical use of the composition, the
  • compositions, nanoparticles or kits may be used for the prophylaxis, treatment and/or amelioration of diseases selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e.
  • (hereditary) diseases or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency.
  • One of the particularly preferred uses involves the extravascular administration of the composition and/or of the nanoparticles to a subject, such as by injection, infusion or
  • intradermal in particular intradermal, subcutaneous, intramuscular, interstitial, locoregional, intravitreal, periocular, intratumoural, intralymphatic, intranodal, intra-articular, intrasynovial, periarticular, intraperitoneal, intra-abdominal, intracardial, intrapericardial, intraventricular, intrapleural, perineural, intrathoracic, epidural, intradural, peridural, intrathecal, intramedullary intracerebral, intracavernous, intracorporus cavernosum, intraprostatic, intratesticular, intracartilaginous, intraosseous, intradiscal, intraspinal, intracaudal, intrabursal, intragingival, intraovarian, intrauterine, periodontal, retrobulbar, subarachnoid, subconjunctival or
  • intralesional injection, infusion or implantation or by topical administration to the skin or a mucosa, in particular dermal or cutaneous, nasal, buccal, sublingual, otic or auricular, ophthalmic, conjunctival, vaginal, rectal, intracervical, endosinusial, laryngeal, oropharyngeal, ureteral, or urethral administration; or by administration to the respiratory system by inhalation, in particular by aerosol administration to the lungs, bronchi, bronchioli, alveoli, or paranasal sinuses; or by transdermal or percutaneous administration.
  • nucleic acid compounds may be effectively delivered to biological targets by carriers based on a cationisable or permanently cationic lipid or lipidoid, such as a lipid or lipidoid having a quaternised ammonium group bearing a positive charge at any pH value of its environment, in particular if the respective nucleic acid compound and the cationisable or permanently cationic lipid or lipidoid are incorporated in a composition which is locally administered to a subject, rather than by systemic injection via the intravenous or intraarterial route.
  • a cationisable or permanently cationic lipid or lipidoid such as a lipid or lipidoid having a quaternised ammonium group bearing a positive charge at any pH value of its environment
  • Figures 1A to ID show the effect of the inventive polymer-lipid or polymer-lipidoid formulations on transfection efficiency of in HepG2 cells in vitro. All depicted transfection experiments were performed in triplicates, using GpLuc mRNA (SEQ ID NO: 14; also labelled as R2851 herein) as the cargo. Moreover, negative controls (buffer, passive pulsing) have been included.
  • (A) to (D) show the GpLuc levels obtained using the indicated transfection reagents.
  • polymer In addition to the inventive polymer-lipid(oid) transfection reagent, polymer only has been used for comparison as well as the pure, 'naked' GpLuc mRNA without the use of transfection reagents. For further details, see Example 2.
  • figures 2 A to 2 E show the effect of the inventive polymer-lipid or polymer- lipidoid formulations on transfection efficiency on Sol8 muscle cells in vitro.
  • GpLuc mRNA SEQ ID NO: 14 / R2851
  • negative controls buffer, passive pulsing
  • positive controls polymer only and pure, 'naked' GpLuc mRNA without transfection reagents.
  • Figures 3A and 3B show the in vitro release of tumor necrosis factor alpha (TNFa; 3 A) cytokines interferon alpha (IFNa; 3B) and in human peripheral blood mononuclear cells
  • Figure 4 shows the scanning laser ophthalmoscopy (SLO) analysis results of the subretinal injection of PpLuc mRNA (SEQ ID NO: 15) into rat eyes, 24 h after subretinal injection of the inventive polymer-lipid or polymer-lipidoid formulations, expressed as relative light units (RLU).
  • SLO scanning laser ophthalmoscopy
  • Example 6 shows the scanning laser ophthalmoscopy
  • Figure 6A shows the in vivo tissue distribution of an exemplary inventive PpLuc mRNA (SEQ ID NO: 15) polymer-lipid formulation of the permanently cationic lipid MC3-cat for the tissues liver, lung and spleen (mean value of four mice depicted) and figure 6B shows the in vivo lung distribution of an exemplary inventive polymer-lipid formulation.
  • SEQ ID NO: 15 the polymer-lipid formulation of the permanently cationic lipid MC3-cat for the tissues liver, lung and spleen
  • figure 6B shows the in vivo lung distribution of an exemplary inventive polymer-lipid formulation.
  • Each bar in figure 6A indicates the value of an individual mouse.
  • Each dot in figure 6B represents an individual animal and the horizontal lines represent median values.
  • Figures 7-A/B show HI titers 21 days after prime vaccination with different formulated HA-mRNA (Figure 7-A) and 14 days after boost vaccination ( Figure 7-B).
  • the dashed line indicates the conventionally defined protective HI titer of 1:40.
  • Figures 8-A-D show results of ELISA assays 21 days after prime vaccination and 14 days after boost vaccination with different formulated HA-mRNA and 14 days after boost vaccination (IgGl subtypes day 21 post-prime are shown in Figure 8-A, IgGl subtypes day 14 post-boost are shown in Figure 8-B, IgG2a subtypes day 21 post-prime are shown in Figure 8-C, IgG2a subtypes day 14 post-boost are shown in Figure 8-D).
  • Figure 9 shows T cell immune responses measured by IFNy production using Elispot.
  • Figure 10 shows GpLuc protein expression in A549 cells transfected with the mRNA construct R2851 in a formulation comprising 3-C12-OH.
  • Figure 11 shows GpLuc protein expression in A549 cells transfected with the mRNA construct R2851 in a formulation comprising DDAB.
  • the invention provides a composition comprising a cationisable or permanently cationic lipid or lipidoid and a nucleic acid compound, wherein the lipid or lipidoid and the nucleic acid compound are non-covalently associated, and wherein the ratio of the lipid to the nucleic acid compound is not higher than about 2 nmol lipid or lipidoid per ⁇ g nucleic acid compound.
  • the cationisable or permanently cationic lipid or lipidoid is a permanently cationic lipid.
  • composition refers to any type of composition in which the specified ingredients may be incorporated, optionally along with any further constituents.
  • the composition may be a dry composition such as a powder or granules, or a solid unit such as a lyophilised form or a tablet.
  • the composition may be in liquid form, and each constituent may be independently incorporated in dissolved or dispersed (e.g. suspended or emulsified) form.
  • the composition is formulated as a sterile solid composition, such as a powder or lyophilised form for reconstitution with an aqueous liquid carrier.
  • a cationisable or permanently cationic lipid or lipidoid means a cationisable or permanently cationic lipid or or a cationisable or permanently cationic lipidoid.
  • a “compound” means a chemical substance, which is a material consisting of molecules having essentially the same chemical structure and properties.
  • the molecules are typically identical with respect to their atomic composition and structural configuration.
  • the molecules of a compound are highly similar but not all of them are necessarily identical.
  • a segment of a polypeptide that is designated to consist of 50 amino acids may also contain individual molecules with e.g. 48 or 53 amino acids.
  • cationic means that the respective structure bears a positive charge, either permanently, or not permanently but in response to certain conditions such as pH.
  • cationic (in the absence of a modifying terms such as “permanently” covers both “permanently cationic” and “cationisable”.
  • permanently cationic means that the respective compound, or group or atom, is positively charged at any pH value or hydrogen ion activity of its environment. In many cases, the positive charge is results from the presence of a quaternary nitrogen atom. Where a compound carries a plurality of such positive charges, it may be referred to as permanently polycationic, which is a subcategory of permanently cationic.
  • poly- refers to a plurality of atoms or groups having the respective property in a compound. If put in parenthesis, the presence of a plurality is optional.
  • (poly) cationic means cationic and/or polycationic. However, the absence of the prefix should not be interpreted such as to exclude a plurality.
  • a polycationic compound is also a cationic compound and may be referred to as such.
  • “Cationisable” means that a compound, or group or atom, is positively charged at a lower pH and uncharged at a higher pH of its environment. Also in non-aqueous environments where no pH value can be determined, a cationisable compound, group or atom is positively charged at a high hydrogen ion concentration and uncharged at a low concentration or activity of hydrogen ions. It depends on the individual properties of the cationisable or polycationisable compound, in particular the pKa of the respective cationisable group or atom, at which pH or hydrogen ion concentration it is charged or uncharged. In diluted aqueous environments, the fraction of cationisable compounds, groups or atoms bearing a positive charge may be estimated using the so-called Henderson-Hasselbalch equation which is well-known to a person skilled in the art.
  • a moiety is cationisable, it is preferred that it is positively charged at a pH value of about 1 to 8, preferably 4 to 8, 5 to 8 or even 6 to 8, more preferably of a pH value of or below 8, of or below 7, most preferably at physiological pH values, e.g. about 7.3 to 7.4, i.e. under physiological conditions, particularly under physiological salt conditions of the cell in vivo.
  • cationised typically means that a cationisable structure is in a state where it actually bears a positively charge, as for example in the case of a basic amino acid such as arginine in a neutral physiological environment.
  • nucleic acid compounds may be effectively delivered to biological targets by carriers based on a cationisable or permanently cationic lipid or lipidoid, such as a lipid or lipidoid having a quaternised ammonium group bearing a positive charge at any pH value of its environment, in particular if the respective nucleic acid compound and the cationisable or permanently cationic lipid or lipidoid are incorporated in a composition which is locally administered to a subject, rather than by systemic injection via the intravenous or intraarterial route.
  • a cationisable or permanently cationic lipid or lipidoid such as a lipid or lipidoid having a quaternised ammonium group bearing a positive charge at any pH value of its environment
  • cationisable lipids i.e. which are predominantly cationic, i.e. positively charged, only at a pH which is lower than their pka (the logarithmic acid dissociation constant of the respective lipid).
  • pka the logarithmic acid dissociation constant of the respective lipid
  • compositions comprising a nucleic acid compound and a cationisable or permanently cationic lipid or lipidoid
  • a composition comprising a nucleic acid compound and a cationisable or permanently cationic lipid or lipidoid
  • the presence of such lipid or lipidoid leads to an unexpected effectiveness in the delivery of the nucleic acid compound to target cells, and at the same time is associated with an unexpectedly high level of tolerability, and a low degree of undesirable side effects or toxicity.
  • the cationisable or permanently cationic lipid or lipidoid may be any lipid, lipidoid or lipid- like compound that is generally known in the art which comprises a group or moiety which is cationisable or permanently cationic.
  • useful lipids that are permanently cationic are compounds with a quaternary ammonium function, such as lipids comprising a
  • lipid means any compound understood or classified as a lipid in the relevant technical field, which is in this case the field of nucleic acid formulation and delivery.
  • a lipid is characterised in that it is lipophilic or hydrophobic, or it comprises a lipophilic, or hydrophobic, domain.
  • This lipophilic domain may consist of one or more functional groups or moieties, such as one or more hydrocarbon chains or cyclic hydrocarbon groups.
  • the cationisable or permanently cationic lipid may further comprise a linking group which links the cationic group, which is substantially hydrophilic, with the lipophilic domain of the lipid.
  • a lipidoid also referred to as lipidoid compound, is a lipid-like compound, i.e. an
  • the lipidoid compound is preferably a compound which comprises two or more cationic nitrogen atoms and at least two lipophilic tails.
  • the lipidoid compound may be free of a hydrolysable linking group, in particular linking groups comprising hydrolysable ester, amide or carbamate groups.
  • the cationic nitrogen atoms of the lipidoid may be cationisable or cationisable or permanently cationic, or both types of cationic nitrogens may be present in the compound. Examples for potentially suitable lipids that are permanently cationic include, without limitation, the following compounds:
  • DHDEAB N,N-di-n-hexadecyl-N,N-dihydroxyethyl ammonium bromide
  • DHMHAC N,N-di-n-hexadecyl-N-methyl-N-(2-hydroxyethyl) ammonium chloride
  • DMHMAC N,N-myristyl-N-(l-hydroxyprop-2-yl)-N-methylammonium chloride
  • MOOHAC N-methyl-N-n-octadecyl-N-oleyl-N-hydroxyethyl ammonium chloride
  • DSD MA N,N-distearyl-N,N-dimethylammonium
  • DDAC N,N-distearyl-N,N-dimethylammonium chloride
  • DODAB N,N-dioctadecyl-N,N-dimethylammonium bromide
  • N,N-dioleyl-N,N-dimethylammonium and its salts e.g. N,N-dioleyl-N,N-dimethylammonium chloride ("DODAC");
  • DODAC N,N-dioleyl-N,N-dimethylammonium chloride
  • TODMAC3 N,N,N',N'-tetraoleyl-N,N'-dimethyl-l,3-propanediammonium chloride
  • DOTMA N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTMA also known as 1,2- dioleyloxy-3-trimethylaminopropane chloride
  • DOTAP N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTAP N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTAP.Cl also known as l,2-dioleoyloxy-3-trimethylaminopropane chloride
  • l,2-dioleoyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide endeavorDORI N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DMRIE l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DIMRI l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DPRIE l,2-dimpalmityloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DSRIE l,2-distearyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DOSPA N-[l-(2,3-dioleyloxy)propyl]-N-2-(sperminecarboxamido)ethyl)-N,N-dimethyl-ammonium trifluoracetate
  • DOSPA also referred to as 2,3-dioleyloxy-[2(sperminecarboxamido)ethyl]-N,N- dimethyl-l-propanaminiumtrifluoroacetate
  • SAINTs monomeric and dimeric pyridinium amphiphiles
  • SAINT-1 N-methyl-4-(dipalmityl)-methylpyridinium chloride
  • SAINT-2 N-methyl-4-(dioleyl)-methylpyridinium chloride
  • SAINT-8 N-methyl-4-(stearyl) (oleyl)-methylpyridinium chloride
  • DMPC l,2-dioleoyl-sn-glycero-3-phosphocholine (also dioleoylphosphatidylcholine; "DOPC”); l,2-dimyristoyl-sn-glycero-3-phosphocholine ("DMPC");
  • DPPC l,2-dipalmitoyl-sn-glycero-3-phosphocholine
  • DEPC l,2-dierucoyl-sn-glycero-3-phosphocholine
  • GIPC l-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine
  • AzPC l-palmitoyl-2-azelaoyl-sn-glycero-3-phosphocholine
  • DMEPC l,2-dimyristoyl-sn-glycero-3-ethylphosphocholine
  • DPePC 1,2- dipalmitoyl-sn-glycero-3-ethylphosphocholine
  • DC-6-14 0,0-ditetradecanoyl-N-(a-trimethylammonioacetyl)diethanolamine chloride
  • DC-6-14 (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4-(trimethylamino)butanoate and its salts
  • DLin-MC3-TMA also referred to as "MC3-cationized”
  • TC-Chol 3-beta-[N-(N',N',N'-trimethylaminoethane)carbamoyl]cholesterol iodide
  • TC-Chol 3-beta-[N-(N',N',N'-trimethylaminoethane)carbamoyl]cholesterol iodide
  • Lipofectin® (including DOTMA and DOPE, available from GIBCO/BRL);
  • Lipofectamin ® (comprising DOSPA and DOPE, available from GIBCO/BRL);
  • the cationisable or permanently cationic lipid is a compound according to one of the formulas
  • X represents a hydrophilic head group comprising a cationisable or permanently cationic nitrogen
  • Y, Y 1 and Y 2 are linking groups, each comprising an ether, ester, amide, urethane, thioether, disulphide, orthoester, or phosphoramide bond
  • Z, Z 1 , Z 2 , and Z 3 are independently selected and represent hydrophobic groups each comprising a linear or branched hydrocarbon chain or a cyclic hydrocarbon group, such as a steroid residue.
  • the number of carbon atoms in the linear or branched hydrocarbon chain is 6 or higher for Z; and 4 or higher for Z 1 or Z 2 or Z 3 , provided that, for compounds of formula lb, Z 1 and Z 2 together have at least 12 carbon atoms in their hydrocarbon chains, and for a compound of formula Ic, Z 1 , Z 2 and Z 3 together have at least 12 carbon atoms in their hydrocarbon chains.
  • the lipid does not comprise any group that exists in an anionised form at approximately neutral or physiological pH conditions, unless it also has more than one cationisable or permanently cationic groups whose positive charges dominate over the negative charge of the anionised group.
  • the hydrophilic headgroup X is permanently cationic, and thus renders the lipid also to be permanently cationic.
  • the hydrophilic headgroup typically is or comprises a quaternary ammonium group.
  • a quaternary ammonium group refers to a structure in which all four hydrogens of the ammonium cation (NH 4 + ) have been replaced by substituents.
  • the quaternary ammonium group is also sometimes referred to as quaternary amine group.
  • the quaternary ammonium group may, for example, be an N-substituted pyridinium moiety, or a quaternary ammonium group with two or three methyl, hydroxyxethyl or hydroxypropyl groups, such as a trimethylamino group. Again, the group may also be part of a larger group, such as a trialkylaminoalkyl group.
  • Some of the preferred quaternary ammonium groups are trialkylaminoalkyl groups selected from the following structures:
  • alkyl is selected from methyl, hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
  • trialkyl-N-CH2- wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2- hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
  • alkyl is selected from methyl, hydroxymethyl, ethyl, 2- hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
  • alkyl is selected from methyl, hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
  • trialkylaminoalkyl groups are trimethylaminomethyl, trimethylaminoethyl, trimethylaminopropyl, and trimethylaminobutyl.
  • the three optionally substituted alkyl groups in the trialkylaminoalkyl groups exhibited above may be selected to be different from each other, as for example in N,N-dimethyl- N-ethylaminoalkyl groups with alkyl being in particular linear alkyl chains with 1 to 6 carbon atoms; or N,N-dimethyl-N-hydroxyethylaminoalkyl groups, N,N-dimethyl-N-propylaminoalkyl groups, N,N-diethyl-N-hydroxyethylaminoalkyl groups, or N-methyl-N-ethyl-N- hydroxyethylaminoalkyl groups, or similar groups with different combinations of optionally substituted methyl-, ethyl, or propyl groups attached to the nitrogen atom of the aminoalkyl structure, again with the alkyl being preferably selected from linear alkyl chains with 1 to 6 carbon atoms.
  • the hydrophilic headgroup X comprises two or more ammonium groups which are optionally separated by a spacer.
  • Suitable spacers include, for example, flexible hydrophilic spacers such as oxyethylene-type spacers, flexible hydrophobic spacers such as alkylenes, or rigid hydrophobic spacers such as aromatic structures.
  • a trialkylaminoalkyl group as described above is attached to a further nitrogen atom, which may, for example, represent a tertiary amino group.
  • headgroups include in particular trimethylaminoalkylamino groups wherein the alkyl group between the two nitrogen atoms is preferably selected from linear alkyls with 1 to 6 carbon atoms.
  • the headgroup X comprises such further amino group, that group may be connected with the linking group Y via a spacer, such as an alkyl chain.
  • linking groups Y, or Y 1 and Y 2 link the hydrophilic headgroup X with the hydrophobic group Z, or with the hydrophobic groups Z 1 and Z 2 , and with Z 3 , if present.
  • Each linking group represents or comprises an ether, ester, amide, urethane, thioether, disulphide, orthoester, or phosphoramide bond, including any combinations of any of these.
  • ester groups and ether groups include dioxolane groups.
  • a dioxolane may also be understood as a cyclic acetal.
  • the linking groups Y, Y 1 and Y 2 are degradable under physiological conditions.
  • the expression "degradable under physiological conditions" which refers to a type of biodegradability, should be understood in the context of nucleic acid delivery. In this context, degradability requires some appreciable degree of degradation occurring within minutes, hours and/or days (rather than years) in order to be meaningful for in vivo applications. Preferably, this biodegradability is ensured by a chemical bond which is hydrolysable under physiological conditions, such as an ester, amide or acetal bond.
  • ester group this may be linked to the hydrophilic headgroup X via its carbonyl group or via the ester oxygen, for example according to the following formulas which are specific versions of formula la:
  • a suitable linking group Y, Y 1 and/or Y 2 , based on an ester group may further comprise a carbon atom or alkyl (or similar) spacer as in the following subscopes of formulas lb, Ic, and Id:
  • Linking groups Y 1 and Y 2 may be identical or different from each other. In one of the preferred embodiments, they are identical.
  • a linking alkyl (or similar) group with a spacer function may also be used between the ester group and the hydrophilic headgroup X, as in the following exemplary formulas:
  • Such linking alkyl group may be considered as part of the overall linking group Y, Y 1 or Y 2 , respectively.
  • the carbon atom in position 4 of the dioxolane ring may be connected to the headgroup X, and the carbon atom in position 2 may be linked to one or two hydrophobic groups, i.e. to Z or Z 1 and Z 2 .
  • the linking group may also comprise a dioxolane ring, but in this case the linking group should comprise a further carbon atom for linkage with Z 1 , Z 2 , and Z 3 .
  • Such further carbon atom may be connected directly to e.g. the carbon atom in position 2 of the dioxolane ring, or via an alkyl spacer.
  • Z, Z 1 , Z 2 , and Z 3 are independently selected and represent hydrophobic groups. Each comprises a linear or branched hydrocarbon chain or a cyclic hydrocarbon group, such as a steroid residue. Moreover, the number of carbon atoms in the linear or branched hydrocarbon chain is 6 or higher for Z; and 4 or higher for Z 1 or Z 2 or Z 3 , provided that, for compounds of formula lb or Id, Z 1 and Z 2 together have at least 12 carbon atoms in their hydrocarbon chains, and for a compound of formula Ic, Z 1 , Z 2 , and Z 3 together have at least 12 carbon atoms in their hydrocarbon chains.
  • Z, Z 1 , Z 2 , and/or Z 3 may be derived from fatty acids, glycerophospholipids, sphingolipids, glycerolipids, sterols, prenols, polyketides and the like.
  • the number of carbon atoms is at least 6, and preferably at least 8, or at least 10, or at least 12 carbon atoms, respectively.
  • Other preferred ranges for the number of carbon atoms in the hydrocarbon chain are from 8 to 24, from 10 to 22, or from 12 to 20, respectively, such as about 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • the steroid is cholesteryl or a derivative thereof.
  • the lipid is a compound of formula lb, Ic or Id, i.e. such as to exhibit more than one hydrophobic group.
  • the hydrophobic groups Z 1 and Z 2 may be identical or different; in a preferred embodiment, they are identical.
  • the groups Z 1 , Z 2 and Z 3 may be the same or different, and in a preferred embodiment, these are also identical.
  • the lipid is a compound of formula lb with the hydrophobic groups Z 1 and Z 2 being identical, wherein each of Z 1 and Z 2 represents a linear hydrocarbon chain with a length of 14 to 22 carbon atoms, either saturated, such as
  • - octadecyl also referred to as stearyl
  • - eicosyl also referred to as arachidyl
  • lipids examples include 2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane ("DLin-KC2-TMA"), (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4- (triimethylamino)butanoate ("DLin-MC3-TMA").
  • DLin-KC2-TMA 2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane
  • DLin-MC3-TMA 6-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane
  • DLin-MC3-TMA 6-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane
  • the lipid is a compound of formula Ic with the hydrophobic groups Z 1 , Z 2 and Z 3 being identical, wherein each of the groups Z 1 , Z 2 and represents a linear hydrocarbon chain with a length of 14 to 22 carbon atoms, either saturated or unsaturated, and preferably selected from those listed in the preceding paragraph.
  • the lipid is a compound of formula Id with the hydrophobic groups Z 1 and Z 2 being identical, and wherein each of the groups Z 1 and Z 2 and represents a linear hydrocarbon chain with a length of 14 to 22 carbon atoms, either saturated or unsaturated, and preferably selected as described above in the context of the linear hydrocarbon chains for compounds according to formula lb.
  • each of the groups Z 1 and Z 2 represents a branched or two-tailed hydrocarbon residue with a total number of 10 to 22 carbon atoms (per hydrophobic group Z 1 or Z 2 ).
  • Such branched or two-tailed hydrocarbon residue may be saturated or unsaturated.
  • a two-tailed structure may, for example, comprise two linear chains which may have different lengths and which are both connected to a carbon atom of the linking group Y 1 or Y 2 .
  • the permanently cationic lipid is a compound according to formula la, lb, Ic or Id wherein
  • X is selected from a quaternary ammonium group, in particular a trimethylammonium group
  • Y, Y 1 and/or Y 2 is are selected from linking groups comprising an ester or amide bond or a dioxolane ring; and/or Z is a steroid residue; and/or
  • Z 1 , Z 2 , and/or Z 3 are selected from saturated or unsaturated hydrocarbon chains with 14 to 22 carbon atoms.
  • a trimethylammonium group means the group -N + (CI3 ⁇ 4).
  • the lipid is a cationisable or permanently cationic compound, it may require an anion, unless it is a zwitterionic compound with at least as many anionic groups as permanently cationic groups.
  • the anion may be selected independently for each compound of interest. In principle, any biocompatible and - in particular if an in vivo use is contemplated - physiologically acceptable anion may be used. Particularly preferred anions include chloride, bromide, malonate, citrate, acetate, maleate, fumarate, succinate, lactate, tartrate, pamoate, hydrogen phosphate, in particular chloride.
  • anions may be selected from commonly known lists of pharmaceutical salts, such as the anions listed by Stahl et al., Handbook of Pharmaceutical Salts, Wiley- VCH (2002), as salts of classes I, II or III, from which salts of classes I and II are preferred as class I ions are physiologically ubiquitous or occur as intermediate metabolites in biochemical pathways, and class II salts, while not naturally occurring, have been used in pharmaceuticals and have shown low toxicity and good tolerability.
  • the lipid is cationisable or permanently cationic and a compound according to formula la, lb, Ic or Id which is not zwitterionic under substantially neutral or physiological conditions, and is selected from the following compounds:
  • DOTMA N-[l-(2,3-dioleyloxy)propyl] -N,N,N-trimethylammonium chloride
  • DOTMA N-[l-(2,3-dioleyloxy)propyl] -N,N,N-trimethylammonium chloride
  • DOTAP N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTAP N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTAP N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOTAP N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DMRIE l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DIMRI l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DPRIE l,2-dimpalmityloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DSRIE l,2-distearyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide
  • DOSPA N-[l-(2,3-dioleyloxy)propyl]-N-2-(sperminecarboxamido)ethyl)-N,N-dimethyl-ammonium trifluoracetate
  • DOSPA also referred to as 2,3-dioleyloxy-[2(sperminecarboxamido)ethyl]-N,N- dimethyl-l-propanaminiumtrifluoroacetate
  • C9-C17-C3 cat l-(2-octylcyclopropyl)heptadec-8-yl-4-(trimethylammonium)butanoate
  • C9-C17-P cat l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3-pyrrolidiniumcarboxylate
  • the cationisable or permanently cationic lipid is selected from (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4- (trimethylamino)butanoate and its salts ("DLin-MC3-TMA”), 2,2-dilinoleyl-4-(2- trimethylaminoethyl)-[l,3]-dioxolane (“DLin-KC2-TMA”), l-(2-octylcyclopropyl)heptadec-8-yl-4- (trimethylammonium)butanoate (“C9-C17-C3 cat”), l-(2-octylcyclopropyl)heptadec-8-yl-l,l- dimethyl-3-pyrrolidiniumcarboxylate (“C9-C17-P cat”), including any salts of these compounds.
  • DLin-MC3-TMA 2,2-dilin
  • the permeanently cationic lipid is (6Z,9Z,28Z,31Z)-heptatriaconta- 6,9,28,31-tetraen-19-yl-4-(trimethylamino)butanoate or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof.
  • the permeanently cationic lipid is 2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof.
  • the permeanently cationic lipid is l-(2-octylcyclopropyl)heptadec-8-yl-4- (trimethylammonium)butanoate ("C9-C17-C3 cat") or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof.
  • the cationisable or permanently cationic lipid is l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3- pyrrolidiniumcarboxylate ("C9-C17-P cat"), or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof.
  • the composition may comprise a cationisable or permanently cationic lipidoid compound, instead of or in addition to the cationisable or permanently cationic lipid.
  • a lipidoid is a lipid-like compound, i.e. an amphiphilic compound with lipid-like physical properties.
  • the lipidoid is a compound comprising at least two cationisable or permanently cationic nitrogen atoms and at least two lipophilic tails.
  • a "tail" is a substructure of a molecule representing a chain or chain-like structure, such as an optionally substituted hydrocarbyl, acyl or acyloxylalkyl chain of at least four, and more preferably at least six, carbon atoms.
  • the optionally substituted hydrocarbyl, acyl or acyloxyalkyl chain representing the lipophilic tail may be directly connected with a cationic nitrogen atom.
  • the lipidoid is a compound comprising two identical lipophilic tails, each of which is directly connected with a cationisable or permanently cationic nitrogen atom.
  • the lipidoid is a compound comprising three identical lipophilic tails, each tail being directly connected with a cationisable or permanently cationic nitrogen atom.
  • the lipidoid is a compound comprising four or more identical lipophilic tails, each tail being directly connected with a cationisable or
  • the lipidoid may optionally comprise a further nitrogen atom (which may be cationisable or permanently cationic) to which no lipophilic tail is connected.
  • lipidoid may also be understood as a compound having a cationic backbone derived from an oligoamine with the lipophilic tails being attached to the, or some of the, cationic nitrogens of the oligoamine, and wherein the cationic nitrogens, or at least some of them, have been substituted (e.g. alkylated) such as to render them permanently cationic.
  • the lipidoid is a compound comprising two identical lipophilic tails, each of which is directly connected with a permanently cationic nitrogen atom.
  • the lipidoid is a compound comprising three identical lipophilic tails, each tail being directly connected with a permanently cationic nitrogen atom.
  • the lipidoid is a compound comprising four or more identical lipophilic tails, each tail being directly connected with a permanently cationic nitrogen atom.
  • the lipidoid may optionally comprise a further nitrogen atom (which may be cationisable or permanently cationic) to which no lipophilic tail is connected.
  • Such lipidoid may also be understood as a compound having a cationic backbone derived from an oligoamine with the lipophilic tails being attached to the, or some of the, cationic nitrogens of the oligoamine, and wherein the cationic nitrogens, or at least some of them, have been substituted (e.g. alkylated) such as to render them permanently cationic.
  • the substituent may, for example, be a methyl or a hydroxyl.
  • the optionally substituted hydrocarbyl chain may be saturated, such as to resemble an alkyl, or it may be unsaturated, i.e. an alkenyl or alkynyl, each optionally having one, two, three or more carbon-carbon double bonds and/or triple bonds.
  • tail structures representing or including an acyl or acyloxyalkyl group, these may also comprise one, two or more carbon- carbon double bonds and/or triple bonds in the hydrocarbon segment of the tail.
  • the lipidoid compound is free of hydrolysable linking groups, such as ester, amide or carbamate groups.
  • a linking group is a group which links the lipophilic tails of the lipidoid molecule to the hydrophilic region comprising the cationic nitrogen atoms.
  • Conventional cationic lipids that have been proposed as carriers or agents to deliver nucleic acids to cells and enhance transfection often - if not typically - exhibit such linkers or linking groups, which are most often hydrolysable and/or enzymatically cleavable.
  • linkers with ester groups have been proposed, but also linkers with amide groups or carbamate groups, all of which are susceptible to hydrolytic and/or enzymatic cleavage in vivo.
  • hydrolysable means that an appreciable degree of hydrolysis occurs in a physiological fluid (such as interstitial fluid) under in vivo conditions within seconds, minutes, hours, or days; preferably, the respective compound or group is hydrolysed to at least 50% after not more than 7 days, or even after not more than 2 days.
  • physiological fluid such as interstitial fluid
  • the lipidoid compound is cationisable or permanently cationic.
  • at least one of the cationic nitrogen atoms of the lipidoid is cationisable or permanently cationic.
  • the lipidoid comprises two cationisable or permanently cationic nitrogen atoms, three cationisable or permanently cationic nitrogen atoms, or even four or more cationisable or permanently cationic nitrogen atoms.
  • the lipidoid compound is permanently cationic.
  • at least one of the cationic nitrogen atoms of the lipidoid is permanently cationic.
  • the lipidoid comprises two permanently cationic nitrogen atoms, three permanently cationic nitrogen atoms, or even four or more permanently cationic nitrogen atoms
  • the lipidoid compound is a cationisable or permanently cationic derivative of a compound according to formula II
  • each occurrence of RA is independently unsubstituted, cyclic or acyclic, branched or unbranched Ci-20 aliphatic; substituted or unsubstituted, cyclic or acyclic, branched or unbranched Ci-20 heteroaliphatic; substituted or unsubstituted aryl; substituted or
  • each occurrence of R5 is independently unsubstituted, cyclic or acyclic, branched or unbranched e-u aliphatic; substituted or unsubstituted aryl; or substituted or unsubstituted heteroaryl.
  • each occurrence of x is an integer from 1 to 10
  • each occurrence of y is an integer from 1 to 10.
  • R5 is Ce-Ci6 alkyl for at least one occurrence, or even at each occurrence.
  • At least one x is selected from 1 or 2, and optionally all occurrences of x are 1 or 2.
  • at least one y is selected from 1 or 2, and optionally all occurrences of y are 1 or 2.
  • all occurrences of R5 are Ce-C i6 alkyl
  • all occurrences of x are 1 or 2
  • all occurrences of y are 1 or 2.
  • such lipidoids may be prepared by reacting an oligoamine and an epoxide-terminated aliphatic compound at elevated temperatures, such as at 80 to 95 °C, in the absence of a solvent, followed by the quaternisation of one or more nitrogen atoms in the lipidoid.
  • Such compound includes a hydrophilic portion resulting from the opening of the epoxide by the amine and a hydrophobic aliphatic tail.
  • the oligoamine comprises from 2 to 5 nitrogen atoms.
  • the preferred oligoamines for preparing such lipidoids are, without limitation:
  • epoxide-terminated aliphatic compound are, without limitation, 2- alkyloxiranes wherein alkyl is butyl, hexyl, octyl, decyl, dodecyl, tetradecyl or octadecyl.
  • the alkyl group may further exhibit an alkyl side chain, such as a methyl, ethyl, propyl, or isopropyl side chain.
  • the alkyl group of the 2-alkyloxirane may also comprise one or more heteroatoms such as oxygen.
  • the epoxide-terminated aliphatic compound may comprise one or more carbon-carbon double or triple bonds.
  • the cationisable or permanently cationic lipidoid comprises at least one moiety of formula III:
  • R3 is hydrogen or hydroxyl
  • R4 is selected from linear or branched, saturated or unsaturated C6-C16 hydrocarbyl chain.
  • Ri and R2 may be selected independently of each other and independently for each occurrence. In one embodiment, Ri and/or R2 are the same for each occurrence, in particular when Ri and/or R2 is methyl. R3 may also be the same for each occurrence; for example, all instances of R3 may be hydroxyl. R4 may also be the same for each occurrence; for example, all instances of R4 may be a C6-C16 hydrocarbyl chain.
  • the cationisable or permanently cationic lipidoid comprises three identical moieties of formula III, wherein Ri and R2 are methyl; R3 is hydroxyl, and R4 is a linear or branched C6-C16 alkyl chain.
  • the lipidoid compound is a compound comprising three identical moieties of formula III, wherein Ri is methyl, R2 is methyl; R3 is hydroxyl, and R4 is a linear or branched C6-C16 alkyl chain.
  • a compound according to formula III may be based on the oligoamine backbone
  • R4 is a linear or branched C6-C16 alkyl, in particular a linear e, Ce, w or C12 alkyl.
  • the cationisable or permanently cationic lipidoid is a compound comprising the cation depicted in the formula IV: and further optionally an anion, preferably an anion as described above.
  • the cationisable or permanently cationic lipidoid is compound of formula IVa:
  • the compounds may be prepared by first reacting the oligoamine of the formula:
  • the cationisable or permanently cationic lipidoid is a cationisable or permanently cationic derivative of a com ound according to formula V:
  • Ri and R2 are each independently selected from the group consisting of hydrogen, an optionally substituted, saturated or unsaturated C1-C20 hydrocarbyl, and an optionally substituted, saturated or unsaturated C6-C20 acyl.
  • Ri and R2 of the lipidoid of formula V are both C1-C20 alkyl, more preferably Ci-C6 alkyl, in particular methyl, ethyl, propyl or isopropyl.
  • Ri and R2 may be methyl.
  • n is preferably not higher than about 5, in particular not higher than about 2, such as 1.
  • o is preferably selected from 0 or 1
  • m is preferably selected from the range from 1 to 6, such as 1, 2, 3, 4, 5 or 6.
  • Li and L2 are each independently selected from unsaturated Ce- C22 hydrocarbyls, in particular from C10-C22 hydrocarbyls.
  • unsaturated Ce- C22 hydrocarbyls in particular from C10-C22 hydrocarbyls.
  • preferred hydrocarbyls are linear omega-6 and omega-9 unsaturated hydrocarbon chains with 14, 16, 18, 20, or 22 carbon atoms.
  • lipidoids of formula V wherein Ri and R2 are both methyl, m is 3 or 4, n is 1, o is 0 or 1, and Li and L2 are identical linear omega-6 and omega-9 unsaturated hydrocarbon chains with 16 or 18 carbon atoms.
  • the lipidoid is a compound according to formula V as defined above except that n is 0.
  • the composition comprises two or more cationisable or permanently cationic lipidoids, each being independently selected as described above; or it may comprise a
  • the composition is substantially free of lipids other than the lipidoid defined above; or is substantially free of lipids other than those defined in one of the claims.
  • the composition is free of neutral or zwitterionic lipids; or that it is free of steroids such as cholesterol.
  • the following permanently cationic lipid and lipidoid structures have been used in the present invention:
  • the biologically active cargo material comprised in the composition or in the
  • nanoparticle(s) of the invention is preferably a nucleic acid compound or complex.
  • the nucleic acid compound is selected from chemically modified or unmodified DNA, single stranded or double stranded DNA, coding or non-coding DNA, optionally selected from a plasmid, (short) oligodesoxynucleotide (i.e. a (short) DNA oligonucleotide), genomic DNA, DNA primers, DNA probes, immunostimulatory DNA, aptamer, or any combination thereof.
  • a nucleic acid molecule may be selected e.g. from any PNA (peptide nucleic acid).
  • the nucleic acid is selected from chemically modified or unmodified RNA, single-stranded or double-stranded RNA, coding or non-coding RNA, optionally selected from messenger RNA (mRNA), (short) oligoribonucleotide (i.e. a (short) RNA
  • the nucleic acid molecule of the complex is an RNA. More preferably, the nucleic acid molecule of the complex is a (linear) single-stranded RNA, even more preferably an mRNA or an immunostimulatory RNA.
  • the biologically active cargo material is a combination of more than one nucleic acid compounds.
  • the nucleic acid may be a single- or a double-stranded nucleic acid compound or complex.
  • a double-stranded nucleic acid could also be considered as a combination of two nucleic acid compounds (i.e. the two antiparallel strands) which form a nucleic acid complex due to their association by non-covalent bonds.
  • a double-stranded nucleic acid may also be described as one compound or molecule.
  • the nucleic acid may also be a partially double-stranded or partially single stranded nucleic acid, comprising two strands which are at least partially self- complementary.
  • Such 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 molecule and both thus form a double-stranded nucleic acid molecule in this region, i.e. a partially double-stranded or partially single stranded nucleic acid (molecule).
  • the nucleic acid compound is a single-stranded nucleic acid.
  • the nucleic acid compound may be a circular or linear nucleic acid, preferably a linear nucleic acid.
  • the nucleic acid may be an artificial nucleic acid.
  • An "artificial nucleic acid molecule” or “artificial nucleic acid” may typically be understood to be a nucleic acid molecule, e.g. a DNA or an RNA, that does not occur naturally.
  • an artificial nucleic acid molecule may be understood as a non-natural nucleic acid molecule.
  • Such nucleic acid molecule may be non-natural due to its individual sequence (which does not occur naturally) and/or due to other modifications, e.g. structural modifications of nucleotides which do not occur naturally.
  • An artificial nucleic acid molecule may be a DNA molecule, an RNA molecule or a hybrid-molecule comprising DNA and RNA portions.
  • artificial nucleic acid molecules may be designed and/or generated by genetic engineering methods to correspond to a desired artificial sequence of nucleotides (heterologous sequence).
  • an artificial sequence is usually a sequence that may not occur naturally, i.e. it differs from the wild type sequence by at least one nucleotide.
  • wild type may be understood as a sequence occurring in nature.
  • artificial nucleic acid molecule is not restricted to mean “one single molecule” but is, typically, understood to comprise an ensemble of identical molecules. Accordingly, it may relate to a plurality of identical molecules contained in an aliquot.
  • sequences (protein, or respectively nucleic acid) which are defined in the present invention comprise or consist of a sequence (protein, or respectively nucleic acid) having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% to said sequence (protein, or respectively nucleic acid).
  • a combination of two or more different nucleic acids may be useful, for example, in the case of a composition comprising a nucleic acid (such as an RNA) encoding the heavy chain of an antibody as well as a nucleic acid encoding the light chain of the same antibody.
  • a nucleic acid such as an RNA
  • Another example is the combination of two or more nucleic acids to affect the part of an organism's immune system referred to as the CRISPR/Cas system (CRISPR: clustered regularly interspaced short palindromic repeats; Cas: CRISPR associated protein).
  • a yet further example is the combination of a guide RNA (gRNA) with an encoding nucleic acid within the composition or nanoparticle of the invention. Coding nucleic acids
  • the nucleic acid may encode a protein or a peptide, which may be selected, without being restricted thereto, e.g. from therapeutically active proteins or peptides, selected e.g. from antigens, e.g. tumour antigens, pathogenic antigens (e.g.
  • the coding nucleic acid may be transported into a cell, a tissue or an organism and the protein may be expressed subsequently in this cell, tissue or organism.
  • a bicistronic or multicistronic nucleic acid or RNA is typically a nucleic acid or an RNA, preferably an mRNA, that typically may have two (bicistronic) or more (multicistronic) coding regions.
  • a coding region in this context is a sequence of codons that is translatable into a peptide or protein.
  • the nucleic acid is mono-, bi-, or multicistronic, preferably as defined herein.
  • the coding sequences in a bi- or multicistronic nucleic acid molecule preferably encode distinct proteins or peptides as defined herein or a fragment or variant thereof.
  • the coding sequences encoding two or more proteins or peptides may be separated in the bi- or multicistronic nucleic acid by at least one IRES (internal ribosomal entry site) sequence, as defined below.
  • IRES internal ribosomal entry site
  • the bi- or even multicistronic nucleic acid may encode, for example, at least two, three, four, five, six or more (preferably different) proteins or peptides as defined herein or their fragments or variants as defined herein.
  • IRES internal ribosomal entry site sequence as defined above can function as a sole ribosome binding site, but it can also serve to provide a bi- or even
  • IRES sequences which can be used according to the invention, are those from picornaviruses (e.g. FMDV), pestiviruses (CFFV), polioviruses (PV), encephalomyocarditis viruses (ECMV), foot and mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), mouse leukoma virus (MLV), simian immunodeficiency viruses (SIV) or cricket paralysis viruses (CrPV).
  • picornaviruses e.g. FMDV
  • CFFV pestiviruses
  • PV polioviruses
  • ECMV encephalomyocarditis viruses
  • FMDV foot and mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MMV mouse leukoma virus
  • SIV simian immunodeficiency viruses
  • CrPV cricket paralysis viruses
  • the at least one coding sequence of the nucleic acid sequence according to the invention may encode at least two, three, four, five, six, seven, eight and more proteins or peptides (or fragments and derivatives thereof) as defined herein linked with or without an amino acid linker sequence, wherein said linker sequence can comprise rigid linkers, flexible linkers, cleavable linkers (e.g., self-cleaving peptides) or a combination thereof.
  • the proteins or peptides may be identical or different or a combination thereof.
  • proteins or peptides combinations can be encoded by said nucleic acid encoding at least two proteins or peptides as explained herein (also referred to herein as 'multi-antigen- constructs/nucleic acid'). It has to be noted that in the context of the invention, certain combinations of coding sequences (e.g., comprising at least two different proteins) may be generated by any combination of mono-, bi-, and multicistronic nucleic acids and/or multi-antigen-constructs/nucleic acid to obtain a poly- or even multivalent nucleic acid mixture.
  • the encoded peptides or proteins are selected from human, viral, bacterial, protozoan proteins or peptides. a) Therapeutically active proteins
  • therapeutically active proteins or peptides may be encoded by the nucleic acid comprised in the nanoparticle of the invention.
  • Therapeutically active proteins are defined herein as proteins which have an effect on healing, prevent prophylactically or treat therapeutically a disease, preferably as defined herein, or are proteins of which an individual is in need of, e.g. a native or modified native protein which individual's organism does not produce, or only produces in insufficient quantities. These may be selected from any naturally occurring or synthetically designed recombinant or isolated protein known to a skilled person.
  • therapeutically active proteins may comprise proteins capable of stimulating or inhibiting the signal transduction in the cell, e.g.
  • the nucleic acid may alternatively encode an antigen.
  • the term "antigen" refers to a substance which is recognised by the immune system and is capable of triggering an antigen-specific immune response, e.g. by formation of antibodies or antigen- specific T-cells as part of an adaptive immune response.
  • an antigenic epitope, fragment or peptide of a protein means particularly B cell and T cell epitopes which may be recognized by B cells, antibodies or T cells, respectively.
  • the antigen encoded by the nucleic acid typically represent any antigen, antigenic epitope or antigenic peptide falling under the above definition, and is preferably a protein and peptide antigen, e.g.
  • the antigen may be one derived from another organism that the host organism (e.g. a human subject) itself, such as a viral antigen, a bacterial antigen, a fungal antigen, a protozooal antigen, an animal antigen, an allergenic antigen etc.
  • Allergenic antigens also referred to as allergy antigens or allergens, are typically antigens which may cause an allergy in a human subject.
  • the antigen as encoded by the nucleic acid may be derived from the host itself.
  • antigens include tumour antigens, self-antigens or auto-antigens, such as auto- immune self-antigens, but also (non-self) antigens as defined herein which have originally been derived from cells outside the host organism, but which have been fragmented or degraded inside the host organism, tissue or cell, e.g. by protease degradation or other types of metabolism.
  • tumour antigens are those that are located on the surface of a tumour cell.
  • Tumour antigens may also represent proteins which are overexpressed in tumour cells compared to a normal cell.
  • tumour antigens also include antigens expressed in cells which are not, or which were originally not, themselves tumour cells but associated with a tumour.
  • antigens which are connected with formation or reformation of tumour- supplying blood vessels in particular those which are associated with neovascularisation, such growth factors like VEGF or bFGF, are also of interest.
  • Antigens associated with a tumour also include antigens from cells or tissues typically embedding the tumour.
  • tumour antigens proteins or peptides may be (over)expressed and occur in increased concentrations in the body fluids of patients that have developed a tumour.
  • tumour antigens also referred to as tumour antigens or tumour-associated antigens even though they are, strictly speaking, not antigens in that they do not induce an immune response.
  • Tumour antigens may be divided further into tumour-specific antigens (TSAs) and tumour- associated antigens (TAAs).
  • TSAs can only be presented by tumour cells and not by healthy cells. They typically result from a tumour-specific mutation.
  • TAAs which are more common, are usually produced by both tumour and healthy cells.
  • TAAs which are more common, are usually produced by both tumour and healthy cells.
  • tumour antigens are recognised by the immune system and the antigen-presenting cell can be destroyed by cytotoxic T cells.
  • tumour antigens can also occur on the surface of the tumour in the form of, e.g., a mutated receptor. In this case, they can also be recognised by antibodies.
  • the encoded antigen is an allergen
  • such antigen may be selected from antigens of any source, such as from animals, plants, molds, fungi, bacteria etc. Plant-derived allergens may, for example, be allergens from pollen.
  • the nucleic acid incorporated in the nanoparticle may encode the native antigen or a fragment or epitope thereof.
  • the nucleic acid compound encodes an antibody or an antibody fragment. The antibody or a fragment thereof is selected from the group consisting of (i) a single-chain antibody, (ii) a single-chain antibody fragment, (iii) a multiple-chain antibody, and (iv) a multiple-chain antibody fragment.
  • 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, C L .
  • 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 I, C H 2 und C H 3.
  • the antibody is selected from full-length antibodies.
  • an antibody may be any recombinantly produced or naturally occurring antibody, in particular an antibody suitable for therapeutic, diagnostic or scientific purposes, or an antibody which is associated with a disease, such as an immunological disease or cancer.
  • antibody is used in its broadest sense and specifically covers monoclonal and polyclonal antibodies (including agonist, antagonist, and blocking or neutralising antibodies) and antibody species with polyepitopic specificity.
  • the antibody may belong to any class of antibodies, such as IgM, IgD, IgG, IgA and IgE antibodies.
  • the antibody may resemble an antibody generated by immunisation in a host organism, or a recombinantly engineered version thereof, a chimeric antibody, a human antibody, a humanised antibody, a bispecific antibody, an intrabody.
  • the nucleic acid compound may also encode an antibody fragment, variant, adduct or derivative of an antibody, such as single-chain variable fragment, a diabody or a triabody.
  • the antibody fragment is preferably selected from Fab, Fab', F(ab') 2, Fc, Facb, pFc', Fd and Fv fragments of the aforementioned types of antibodies.
  • antibody fragments are known in the art.
  • a Fab fragment, antigen binding
  • a Fab fragment, antigen binding
  • 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 15-25 glycine, proline and/or serine residues.
  • the biologically active cargo material comprises a combination of at least two distinct RNAs, wherein one RNA encodes a heavy chain of an antibody or a fragment thereof and another RNA encodes the corresponding light chain of the antibody or a fragment thereof.
  • the biologically active cargo material comprises a combination of at least two distinct RNAs, wherein one RNA encodes a heavy chain variable region of an antibody or a fragment thereof and another RNA encodes the corresponding light chain variable region of the antibody or a fragment thereof.
  • the different chains of the antibody or antibody fragment are encoded by a multicistronic nucleic acid, also referred to as polycistronic nucleic acid.
  • the different strains of the antibody or antibody fragment are encoded by several monocistronic nucleic acids.
  • these nucleic acids may be used as cargo in combination within one composition, or nanoparticle, according to the invention.
  • the present invention comprises the use of at least one nucleic acid molecule for the preparation of a biologically active cargo material. If more than one nucleic acid molecule is used, the complexed nucleic acid molecules may be different, i.e. thereby forming a mixture of at least two distinct (complexed) nucleic acid molecules.
  • the biologically active cargo material comprises (i) a nucleic acid molecule encoding a CRISPR related protein; and/or
  • CRISPR related protein includes but is not limited to CAS9 (CRISPR-Associated Protein 9), CSY4, dCAS9, and dCAS9-effector domain (activator and/or inhibitor domain) fusion proteins.
  • the CRISPR related protein can be from any number of species including but not limited to Streptococcus pyogenes, Listeria innocua, and Streptococcus thermophilus.
  • gRNA guide RNA
  • artificial guide RNA also referred to as “artificial guide RNA”, “single guide RNA”, “small guide RNA” or “sgRNA”
  • sgRNA describes an RNA including a typically 20-25 nucleotides long sequence that is complementary to one strand of the 5'UTR of the gene of interest upstream of the transcription start site.
  • a description of sgRNA design can be found in e.g. Mali et al., 2013, Science 339:823-826.
  • the artificial sgRNA targets a gene of interest, directing the CRISPR related protein encoded by the artificial polynucleotide to interact with the gene of interest, e.g., a gene where modulation of transcription is desired.
  • the gene of interest is selected depending on the application.
  • a single nucleic acid molecule of the invention comprised in the composition or in the nanoparticle(s) of the invention comprises a single nucleic acid molecule encoding said CRISPR related protein and simultaneously said guide RNA(s).
  • the biologically active cargo material comprises a combination of more than one nucleic acid molecule.
  • more than one nucleic acid molecules of the invention comprise said nucleic acid molecule encoding a CRISPR related protein and said guide RNA(s).
  • the biologically active cargo material comprises two distinct RNA which express both a Cas9 protein and the target-specific gRNA.
  • the nucleic acid compound incorporated in the composition or nanoparticle of the invention is in the form of dsRNA, preferably siRNA.
  • dsRNA preferably siRNA
  • a dsRNA, or a siRNA is of interest particularly in connection with the phenomenon of RNA interference.
  • RNAi RNA interference
  • the in vitro technique of RNA interference (RNAi) is based on double-stranded RNA molecules (dsRNA) which trigger the sequence-specific suppression of gene expression (Zamore (2001) Nat. Struct. Biol. 9: 746-750; Sharp (2001) Genes Dev. 5:485-490: Hannon (2002) Nature 41: 244- 251).
  • the nucleic acid may, for example, be a double-stranded RNA (dsRNA) having a length from about 17 to about 29 base pairs, and preferably from about 19 to about 25 base pairs.
  • the dsRNA is preferably at least 90 %, more preferably at least 95 %, such as 100 %, (regarding the nucleotides of a dsRNA) complementary to a section of the nucleic acid sequence of a
  • therapeutically relevant protein or antigen as described hereinbefore either a coding or a non- coding section, preferably a coding section.
  • 90 % complementary means that, with a length of a dsRNA of, for example, 20 nucleotides, this contains not more than 2 nucleotides without complementarity with the corresponding section of the mRNA encoding the respective protein.
  • a double-stranded RNA whose sequence is wholly complementary with a section of the nucleic acid of a therapeutically relevant protein or antigen described hereinbefore.
  • the dsRNA has the general structure 5'-(Ni 7 -29)-3', and preferably the general structure 5'-(Nig-25)-3', or 5'-(Nig-24)-3', or 5'-(N 2 i-23)-3', respectively, wherein each N is a nucleotide, and wherein the nucleotide sequence is complementary to a section of the mRNA that corresponds to a therapeutically relevant protein or antigen described hereinbefore.
  • all the sections having a length of from 17 to 29, preferably from 19 to 25, base pairs that occur in the coding region of the mRNA can serve as target sequence for a dsRNA herein.
  • dsRNAs used as nucleic acid 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 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
  • the target sequence of a dsRNA used as nucleic acid 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.
  • Immunostimulatory nucleic acids a) Immunostimulatory CpG nucleic acids
  • the nucleic acid incorporated in the composition or nanoparticle of the invention is an immunostimulatory CpG nucleic acid, in particular a CpG-RNA or a CpG-DNA, which preferably induces an innate immune response.
  • immunostimulatory CpG nucleic acids include, without limitation, single-stranded CpG- DNA (ss CpG-DNA), double-stranded CpG-DNA (dsDNA), single-stranded CpG-RNA (ss CpG-RNA), and double-stranded CpG-RNA (ds CpG-RNA).
  • the CpG nucleic acid is a CpG-RNA, in particular a single-stranded CpG-RNA (ss CpG-RNA). That preferred length of the CpG nucleic acid in terms of nucleotides or base pairs is similar to that preferred for siRNA, as described above.
  • the CpG motifs are unmethylated.
  • the nucleic acid incorporated as biologically active cargo material in the composition or nanoparticle of the invention may be in the form of a of an immunostimulatory RNA (isRNA), which preferably elicits an innate immune response.
  • isRNA immunostimulatory RNA
  • Such isRNA may be a double-stranded RNA, a single-stranded RNA, or a partially double- stranded RNA, or a short RNA oligonucleotide. In one of the preferred embodiments, it is a single- stranded RNA.
  • the isRNA may be circular or linear.
  • a linear isRNA is used, such as a linear single-stranded RNA, or a long single-stranded RNA.
  • the isRNA may be a coding or non-coding RNA.
  • a non-coding RNA is used as isRNA, such as a non-coding single-stranded RNA, a non-coding linear RNA, a non-coding linear single-stranded RNA, or a non-coding long linear single-stranded RNA.
  • the isRNA carries a triphosphate at its 5'- end, as is the case for in vitro transcribed RNA. This preference applies to all aforementioned types of linear isRNA.
  • the isRNA used as biologically active cargo material according to the invention may be selected from any type or class of RNA, whether naturally occurring or synthetic, which is capable of inducing an innate immune response, and/or which is capable of enhancing or supporting an adaptive immune response induced by an antigen.
  • an immune response may occur in various ways.
  • a substantial factor for a suitable adaptive immune response is the stimulation of certain T-cell sub-populations.
  • T- lymphocytes are typically divided into two sub-populations, the T-helper 1 (Thl) cells and the T- helper 2 (Th2) cells, with which the immune system is capable of destroying intracellular (Thl) and extracellular (Th2) pathogens, such as antigens.
  • the two Th cell populations differ in the pattern of the effector proteins (cytokines) produced by them.
  • Thl cells assist the cellular immune response by activation of macrophages and cytotoxic T-cells.
  • Th2 cells on the other hand, promote the humoral immune response by stimulation of B-cells for conversion into plasma cells and by formation of antibodies (e.g. against antigens).
  • the Thl/Th2 ratio is therefore of great importance in the induction and maintenance of an adaptive immune response.
  • the Thl/Th2 ratio of the adaptive immune response is shifted towards the cellular response (Thl response), i.e. a cellular immune response is induced or enhanced.
  • the innate immune system which may support an adaptive immune response may be activated by ligands of toll-like receptors (TLRs).
  • TLRs are a family of highly conserved pattern recognition receptor (PRR) polypeptides that recognise pathogen-associated molecular patterns (PAMPs) and play a critical role in innate immunity in mammals.
  • PRR pattern recognition receptor
  • TLR1 TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 and TLR13.
  • TLR9 unmethylated bacterial DNA and synthetic analogs thereof (CpG DNA) are ligands for TLR9 (Hemmi H et al. (2000) Nature 408:740-5; Bauer S et al. (2001) Proc NatlAcadSci USA 98, 9237- 42).
  • 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 stimulate TLR3, TLR7, or TLR8, or intracellular receptors such as RIG-I, MDA-5 and others.
  • Lipford et al. determined certain G,U-containing oligoribonucleotides as immunostimulatory by acting via TLR7 and TLR8 (see WO 03/086280).
  • oligoribonucleotides described by Lipford et al. were believed to be derivable from RNA sources including ribosomal RNA, transfer RNA, messenger RNA, and viral RNA.
  • the isRNA used in the context of the invention may thus comprise any RNA sequence known to be immunostimulatory, including, without being limited thereto, RNA sequences representing and/or encoding ligands of TLRs, such as the murine family members TLR1 to TLR13, or more preferably selected from human family members TLR1 to TLR10, in particular TLR7 or TLR8; or ligands for intracellular receptors for RNA such as RIG-I or MDA-5 (see e.g. Meylan, E., Tschopp, J. (2006) : Toll-like receptors and RNA helicases: Two parallel ways to trigger antiviral responses. Mol. Cell 22, 561-569).
  • the isRNA may include ribosomal RNA (rRNA), transfer RNA (tRNA), messenger RNA (mRNA), and viral RNA (vRNA). It may comprise up to about 5000 nucleotides, such as from about 5 to about 5000 nucleotides, or from about 5 to about 1000, or from about 500 to about 5000, or from about 5 to about 500, or from about 5 to about 250, or from about 5 to about 100, or from about 5 to about 50 or or from about 5 to about 30
  • rRNA ribosomal RNA
  • tRNA transfer RNA
  • mRNA messenger RNA
  • vRNA viral RNA
  • the isRNA comprises or consists of a nucleic acid of formula (III) or (IV) :
  • G is guanosine (guanine), uridine (uracil) or an analogue of guanosine (guanine) or uridine (uracil), preferably guanosine (guanine) or an analogue thereof;
  • X is guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine), or an analogue of these nucleotides (nucleosides), preferably uridine (uracil) or an analogue thereof;
  • N is a nucleic acid sequence having a length of about 4 to 50, preferably of about 4 to 40, more preferably of about 4 to 30 or 4 to 20 nucleic acids, each N independently being selected from guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of these nucleotides (nucleosides);
  • a is an integer from 1 to 20, preferably from 1 to 15, most preferably from 1 to 10;
  • 1 is an integer from 1 to 40
  • G is guanosine (guanine) or an analogue thereof, and if 1 > 1, at least 50 % of these nucleotides (nucleosides) are guanosine (guanine) or an analogue thereof;
  • nucleic acid molecule of formula (V) has a length of at least 50 nucleotides, preferably of at least 100 nucleotides, more preferably of at least 150 nucleotides, even more preferably of at least 200 nucleotides and most preferably of at least 250 nucleotides;
  • C is cytidine (cytosine), uridine (uracil) or an analogue of cytidine (cytosine) or uridine (uracil), preferably cytidine (cytosine) or an analogue thereof;
  • X is guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of the above-mentioned nucleotides (nucleosides), preferably uridine (uracil) or an analogue thereof;
  • N is each a nucleic acid sequence having a length of about 4 to 50, preferably of about 4 to 40, more preferably of about 4 to 30 or 4 to 20 nucleic acids, each N being independently selected from guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of these nucleotides (nucleosides);
  • a is an integer from 1 to 20, preferably from 1 to 15, most preferably from 1 to 10;
  • nucleic acid molecule of formula (IV) has a length of at least 50 nucleotides, preferably of at least 100 nucleotides, more preferably of at least 150 nucleotides, even more preferably of at least 200 nucleotides and most preferably of at least 250 nucleotides.
  • N i.e. N u and N v
  • X preferably of any of the definitions given above for elements N (i.e. N u and N v ) and X
  • nucleic acid molecule may be selected from e.g. any of the following sequences:
  • the nucleic acid compound used as biologically active cargo material according to the present invention is in the form of a chemically modified nucleic acid, or is a stabilised nucleic acid, preferably a stabilised RNA or DNA, such as a RNA that is essentially resistant to in vivo degradation by an exo- or endonuclease.
  • modification(s) may refer to chemical modifications comprising backbone modifications as well as sugar modifications or base modifications.
  • the respective product of the modification may, for example, be termed a "modified nucleic acid” or a
  • a backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides contained in a nucleic acid compound, preferably an mRNA, are chemically modified.
  • a sugar modification is a chemical modification of the sugar of the nucleotides of the nucleic acid.
  • a base modification in connection with the present invention is a chemical modification of the base moiety of the nucleotides of the artificial nucleic acid, preferably an mRNA.
  • nucleotide analogues or modifications are preferably selected from those nucleotide analogues which are applicable for transcription and/or translation.
  • nucleosides and nucleotides can be modified in the sugar moiety.
  • the 2'-hydroxyl group (OH) can be modified or replaced with a number of different "oxy" or “deoxy” substituents.
  • “Deoxy” modifications include hydrogen, amino (e.g. N3 ⁇ 4; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); or the amino group can be attached to the sugar through a linker, wherein the linker comprises one or more of the atoms C, N, and 0.
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • an artificial nucleic acid preferably an mRNA, can include nucleotides containing, for instance, arabinose as the sugar.
  • the phosphate groups of the backbone of the nucleic acid compound can be modified by replacing one or more of the oxygen atoms with a different substituent.
  • the modified nucleosides and nucleotides can include the full replacement of an unmodified phosphate moiety with a modified phosphate as described herein.
  • modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters.
  • Phosphorodithioates have both non-linking oxygens replaced by sulfur.
  • the phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylene-phosphonates).
  • the modification may relate to a nucleobase moiety of the nucleic acid compound.
  • nucleobases found in a nucleic acid such as RNA include, but are not limited to, adenine, guanine, cytosine and uracil.
  • the nucleosides and nucleotides described herein can be chemically modified on the major groove face.
  • the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
  • the base modifications are selected from 2-amino-6-chloropurineriboside-5'-triphosphate, 2-aminopurine-riboside-5'- triphosphate; 2-aminoadenosine-5'-triphosphate, 2'-amino-2'-deoxycytidine-triphosphate, 2- thiocytidine-5'-triphosphate, 2-thiouridine-5'-triphosphate, 2'-fluorothymidine-5'-triphosphate, 2'-0-methyl inosine-5'-triphosphate 4-thiouridine-5'-triphosphate, 5-aminoallylcytidine-5'- triphosphate, 5-aminoallyluridine-5'-triphosphate, 5-bromocytidine-5'-triphosphate, 5- bromouridine-5 '-triphosphate, 5-bromo-2'-deoxycytidine-5'-triphosphate, 5-bromo-2'- deoxyuridine-5'-triphosphat
  • 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.
  • modified nucleosides include pyridin-4-one ribonucleoside, 5-aza- uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5- hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5- propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl- pseudouridine, 5-taurinomethyl-2-thio-uridine, l-taurinomethyl-4-thio-uridine, 5-methyl-uridine,
  • modified nucleosides include 5-aza-cytidine, pseudoisocytidine, 3- methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5- hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-l- methyl-pseudoisocytidine, 4-thio-l-methyl-l-deaza-pseudoisocytidine, 1-methyl-l-deaza- pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2- thio-zebula
  • modified nucleosides include 2-aminopurine, 2, 6-diaminopurine, 7- deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7- deaza-2,6-diaminopurine, 7-deaza-8-aza-2, 6-diaminopurine, 1-methyladenosine, N6- methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2- methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6- di
  • modified nucleosides include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza- guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7- methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2- dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2- methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
  • the nucleotide can be modified on the major groove face and can include replacing hydrogen on C-5 of uracil with a methyl group or a halo group.
  • a modified nucleoside is 5'-0-(l-thiophosphate)-adenosine, 5'-0- (l-thiophosphate)-cytidine, 5'-0-(l-thiophosphate)-guanosine, 5'-0-(l-thiophosphate)-uridine or 5'-0-(l-thiophosphate)-pseudouridine.
  • a modified nucleic acid compound may comprise nucleoside modifications selected from 6-aza-cytidine, 2-thio-cytidine, a-thio- cytidine, pseudo-iso-cytidine, 5-aminoallyl-uridine, 5-iodo-uridine, Nl-methyl-pseudouridine, 5,6-dihydrouridine, a-thio-uridine, 4-thio-uridine, 6-aza-uridine, 5-hydroxy-uridine, deoxy- thymidine, 5-methyl-uridine, pyrrolo-cytidine, inosine, a-thio-guanosine, 6-methyl-guanosine, 5- methyl-cytdine, 8-oxo-guanosine, 7-deaza-guanosine, Nl-methyl-adenosine, 2-amino-6-chloro- purine, N6-methyl-2-amino-purine
  • the nucleic acid exhibits a lipid modification.
  • a lipid-modified nucleic acid or RNA as defined herein typically further comprises at least one linker covalently linked with that nucleic acid or RNA, and at least one lipid covalently linked with the respective linker.
  • the lipid-modified nucleic acid comprises 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 an nucleic acid molecule as defined herein, at least one linker covalently linked with that RNA, 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 modification is present at the terminal ends of a linear nucleic acid sequence.
  • a modified nucleic acid sequence as defined herein, particularly a modified RNA as defined herein can be modified by the addition of a so-called '5' cap' structure, which preferably stabilizes the nucleic acid as described herein.
  • a 5'-cap is an entity, typically a modified nucleotide entity, which generally "caps" the 5'- end of a mature RNA.
  • a 5 '-cap may typically be formed by a modified nucleotide, particularly by a derivative of a guanine nucleotide.
  • the 5'-cap is linked to the 5'-terminus via a 5'-5'- triphosphate linkage.
  • a 5'-cap may be methylated, e.g.
  • m7GpppN wherein N is the terminal 5' nucleotide of the nucleic acid carrying the 5'-cap, typically the 5'-end of an RNA.
  • m7GpppN is the 5'-cap structure, which naturally occurs in RNA transcribed by polymerase II and is therefore preferably not considered as modification comprised in a modified RNA in this context.
  • a modified RNA sequence of the present invention may comprise a m7GpppN as 5'- cap, but additionally the modified RNA sequence typically comprises at least one further modification as defined herein.
  • 5 'cap structures include glyceryl, inverted deoxy abasic residue (moiety), 4',5' methylene nucleotide, l-(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide, 1,5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4- dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide, 3'-3'-inverted nucleotide moiety, 3'-3'-inverted abasic moiety, 3'-2'-inverted nucleotide moiety, 3'-2'-inverted acetate,
  • modified 5'-cap structures are capl (methylation of the ribose of the adjacent nucleotide of m7G), cap2 (additional methylation of the ribose of the 2 nd nucleotide downstream of the m7G), cap3 (additional methylation of the ribose of the 3 rd nucleotide downstream of the m7G), cap4 (methylation of the ribose of the 4 th nucleotide downstream of the m7G), ARCA (anti-reverse cap analogue, modified ARCA (e.g.
  • the RNA according to the invention preferably comprises a 5'-cap structure.
  • the 5'-cap structure is added co-transcriptionally using cap- analogues as defined herein in an RNA in vitro transcription reaction as defined herein.
  • the 5'-cap structure is added via enzymatic capping using capping enzymes (e.g. vaccinia virus capping enzymes).
  • a nucleic acid may be selected which represents an mRNA that is essentially resistant to in vivo degradation by an exo- or endonucleases.
  • stabilisation can be effected, for example, by chemically modifying the phosphates of the backbone. Sugar or base modifications may be additionally used.
  • mRNA may also be stabilised against degradation by RNases by the addition of a so-called "5' cap” structure. Particular preference is given in this connection to an an G(5')ppp(5')G or a m7G(5')ppp(5')N as the 5'cap structures (N being A, G, C, or U).
  • the mRNA may exhibit a poly-A tail on the 3' terminus of typically about 10 to about 200 adenosine nucleotides, preferably of about 10 to about 100 adenosine nucleotides, or about 20 to about 100 adenosine nucleotides or even about 40 to about 80 adenosine nucleotides.
  • the mRNA may have a poly-C tail on the 3' terminus of typically about 10 to about 200 cytosine nucleotides, preferably about 10 to about 100 cytosine nucleotides, or about 20 to about 70 cytosine nucleotides, or about 20 to about 60 or even about 10 to about 40 cytosine nucleotides.
  • the nucleic acid sequence of the present invention may be modified, and thus stabilized, by modifying the guanosine/cytosine (G/C) content of the nucleic acid sequence.
  • G/C guanosine/cytosine
  • the G/C content of the coding sequence of the nucleic acid sequence of the present invention is modified, particularly increased, compared to the G/C content of the coding sequence of the respective wild-type nucleic acid sequence, i.e. the unmodified nucleic acid.
  • the amino acid sequence encoded by the nucleic acid is preferably not modified as compared to the amino acid sequence encoded by the respective wild-type nucleic acid.
  • This modification of the nucleic acid sequence of the present invention is based on the fact that the sequence of any nucleic acid region, particularly the sequence of any RNA region to be translated is important for efficient translation of that nucleic acid, particularly of that RNA.
  • the composition of the nucleic acid 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
  • the codons of the nucleic acid are therefore varied compared to the respective wild-type nucleic acid, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides.
  • the most favourable codons for the stability can be determined (so-called alternative codon usage).
  • the amino acid to be encoded by the nucleic acid there are various possibilities for modification of the nucleic acid sequence, compared to its wild-type sequence.
  • RNA molecules may also be transferrable to DNA molecules:
  • amino acids which are encoded by codons, containing exclusively G or C nucleotides
  • the codons for Pro CCC or CCG
  • Arg CGC or CGG
  • Ala GCC or GCG
  • GGC or GGG Gly
  • 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.
  • 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 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 Phe can be modified from UUU to UUC; 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 He can be modified from AUU or AUA to AUC; the 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;
  • 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 peptide or protein as defined herein or a fragment or variant thereof or the whole sequence of the wild type RNA sequence are substituted, thereby increasing the G/C content of said sequence.
  • RNA sequence of the present invention is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the corresponding modified RNA sequence is translated to a significantly poorer degree than in the case where codons coding for relatively "frequent" tRNAs are present.
  • the region which codes for a peptide or protein as defined herein or a fragment or variant thereof is modified compared to the corresponding region of the wild-type RNA sequence such that at least one codon of the wild-type sequence, which codes for a tRNA which is relatively rare in the cell, is exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA.
  • the sequence of the RNA of the present invention is modified such that codons for which frequently occurring tRNAs are available are inserted.
  • codons of the wild-type sequence which code for a tRNA which is relatively rare in the cell, can in each case be exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA.
  • Which tRNAs occur relatively frequently in the cell and which, in contrast, occur relatively rarely is known to a person skilled in the art; cf. e.g. Akashi, Curr. Opin. Genet. Dev. 2001, 11(6): 660-666.
  • the codons, which use for the particular amino acid the tRNA which occurs the most frequently e.g.
  • the Gly codon, which uses the tRNA, which occurs the most frequently in the (human) cell are particularly preferred.
  • This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) RNA sequence of the present invention.
  • the determination of a modified RNA sequence of the present invention as described above can be carried out using the computer program explained in WO 02/098443 - the disclosure content of which is included in its full scope in the present invention.
  • the nucleotide sequence of any desired RNA sequence can be modified with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence coded by the modified RNA sequence preferably not being modified compared to the non-modified sequence.
  • the source code in Visual Basic 6.0 development environment used: Microsoft Visual Studio
  • the A/U content in the environment of the ribosome binding site of the RNA sequence of the present invention is increased compared to the A/U content in the environment of the ribosome binding site of its respective wild-type RNA.
  • This modification increases the efficiency of ribosome binding to the RNA.
  • An effective binding of the ribosomes to the ribosome binding site e.g. to the Kozak sequence in turn has the effect of an efficient translation of the RNA.
  • the RNA sequence of the present invention may be modified with respect to potentially destabilizing sequence elements.
  • the coding sequence and/or the 5' and/or 3' untranslated region of this RNA sequence may be modified compared to the respective wild-type RNA such that it contains no destabilizing sequence elements, the encoded amino acid sequence of the modified RNA sequence preferably not being modified compared to its respective wild-type RNA.
  • DSE destabilizing sequence elements
  • RNA sequence for further stabilization of the modified RNA sequence, optionally in the region which encodes at least one peptide or protein as defined herein or a fragment or variant thereof, one or more such modifications compared to the corresponding region of the wild-type RNA can therefore be carried out, so that no or substantially no destabilizing sequence elements are contained there.
  • DSE present in the untranslated regions (3'- and/or 5'-UTR) can also be eliminated from the RNA sequence of the present invention by such modifications.
  • destabilizing sequences are e.g. AU-rich sequences (AURES), which occur in 3'-UTR sections of numerous unstable RNAs (Caput et al., Proc. Natl. Acad. Sci.
  • RNA sequence of the present invention is therefore preferably modified compared to the respective wild-type RNA such that the RNA sequence of the present invention contains no such destabilizing sequences.
  • sequence motifs which are recognized by possible endonucleases, e.g. the sequence GAACAAG, which is contained in the 3'-UTR segment of the gene encoding the transferrin receptor (Binder et al., EMBO J. 1994, 13: 1969 to 1980). These sequence motifs are also preferably removed in the RNA sequence of the present invention.
  • the mRNA used in the context of the invention has a modified the G/C content, preferably in its coding region, which means that the G/C content is modified, particularly increased, compared to the G/C content of the coding region of its corresponding wild-type mRNA, preferably without changing the encoded amino acid sequence.
  • the G/C content of the coding region may be increased by at least 7 %, or by at least 15 %, or by at least 20 %, compared to that of the wild-type mRNA which codes for an antigen, antigenic protein or antigenic peptide as described herein, or a fragment or variant thereof.
  • At least 5 %, 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, 70 %, or 80 %, such as 90 % or more, 95 % or more, or even 100 % of the substitutable codons in the coding region or in the whole sequence are substituted to increase the G/C content.
  • 100 % substitution means that essentially all substitutable codons of the coding region are substituted, which is one of the preferred embodiments of the invention.
  • an mRNA is used wherein the coding region is modified such that at least one codon of the wild-type sequence which codes for a tRNA which is relatively rare in the cell is exchanged for a codon which codes for a tRNA which is relatively frequent in the cell but encodes the same amino acid as the relatively rare tRNA.
  • tRNAs that occur relatively rarely or frequently in the cell are known to a person skilled in the art; cf. e.g. Akashi, Curr. Opin. Genet. Dev. 2001, 11 (6) : 660- 666. The most frequently occuring tRNAs for a particular amino acid are particularly preferred.
  • the nucleic acid sequence of the present invention may be modified, and thus stabilized, by adapting the sequences to the human codon usage.
  • a further preferred modification of the nucleic acid sequence of the present invention is based on the finding that codons encoding the same amino acid typically occur at different frequencies.
  • the coding sequence as defined herein is preferably modified compared to the corresponding coding sequence of the respective wild-type nucleic acid such that the frequency of the codons encoding the same amino acid corresponds to the naturally occurring frequency of that codon according to the human codon usage as e.g. shown in Table 1.
  • the wild type coding sequence is preferably adapted in a way that the codon "GCC” is used with a frequency of 0.40, the codon “GCT” is used with a frequency of 0.28, the codon “GCA” is used with a frequency of 0.22 and the codon “GCG” is used with a frequency of 0.10 etc. (see Table 1).
  • the nucleic acid sequence of the present invention comprises at least one coding sequence, wherein the coding sequence/sequence is codon-optimized as described herein. More preferably, the codon adaptation index (CAI) of the at least one coding sequence is at least 0.5, at least 0.8, at least 0.9 or at least 0.95. Most preferably, the codon adaptation index (CAI) of the at least one coding sequence is 1.
  • the wild type coding sequence is adapted in a way that the most frequent human codon "GCC” is always used for said amino acid, or for the amino acid Cysteine (Cys), the wild type sequence is adapted in a way that the most frequent human codon "TGC” is always used for said amino acid etc.
  • the nucleic acid sequence of the present invention may be modified by modifying, preferably increasing, the cytosine (C) content of the nucleic acid sequence, preferably of the coding sequence of the nucleic acid sequence, more preferably the coding sequence of the RNA sequence.
  • C cytosine
  • the C content of the coding sequence of the nucleic acid sequence of the present invention is modified, preferably increased, compared to the C content of the coding sequence of the respective wild-type nucleic acid, i.e. the unmodified nucleic acid.
  • the amino acid sequence encoded by the at least one coding sequence of the nucleic acid sequence of the present invention is preferably not modified as compared to the amino acid sequence encoded by the respective wild-type nucleic acid.
  • the modified nucleic acid, particularly the modified RNA sequence is modified such that at least 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, 70 % or 80 %, or at least 90 % of the theoretically possible maximum cytosine-content or even a maximum cytosine-content is achieved.
  • At least 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, 70 %, 80 %, 90 % or even 100 % of the codons of the target nucleic acid, particularly the modified RNA wild type sequence, which are "cytosine content optimizable" are replaced by codons having a higher cytosine-content than the ones present in the wild type sequence.
  • some of the codons of the wild type coding sequence may additionally be modified such that a codon for a relatively rare tRNA in the cell is exchanged by a codon for a relatively frequent tRNA in the cell, provided that the substituted codon for a relatively frequent tRNA carries the same amino acid as the relatively rare tRNA of the original wild type codon.
  • all of the codons for a relatively rare tRNA are replaced by a codon for a relatively frequent tRNA in the cell, except codons encoding amino acids, which are exclusively encoded by codons not containing any cytosine, or except for glutamine (Gin), which is encoded by two codons each containing the same number of cytosines.
  • the modified target nucleic acid preferably the RNA is modified such that at least 80 %, or at least 90 % of the theoretically possible maximum cytosine-content or even a maximum cytosine-content is achieved by means of codons, which code for relatively frequent tRNAs in the cell, wherein the amino acid sequence remains unchanged.
  • more than one codon may encode a particular amino acid. Accordingly, 18 out of 20 naturally occurring amino acids are encoded by more than one codon (with Tryp and Met being an exception), e.g. by two codons (e.g. Cys, Asp, Glu), by three codons (e.g. He), by four codons (e.g. Al, Gly, Pro) or by six codons (e.g. Leu, Arg, Ser).
  • two codons e.g. Cys, Asp, Glu
  • three codons e.g. He
  • four codons e.g. Al, Gly, Pro
  • six codons e.g. Leu, Arg, Ser
  • cytosine content-optimizable codon' refers to codons, which exhibit a lower content of cytosines than other codons encoding the same amino acid. Accordingly, any wild type codon, which may be replaced by another codon encoding the same amino acid and exhibiting a higher number of cytosines within that codon, is considered to be cytosine-optimizable (C-optimizable). Any such substitution of a C-optimizable wild type codon by the specific C-optimized codon within a wild type coding sequence increases its overall C-content and reflects a C-enriched modified nucleic acid sequence.
  • the nucleic acid sequence, particularly the RNA sequence of the present invention preferably the at least one coding sequence of the nucleic acid sequence of the present invention comprises or consists of a C-maximized RNA sequence containing C-optimized codons for all potentially C-optimizable codons. Accordingly, 100% or all of the theoretically replaceable C-optimizable codons are preferably replaced by C-optimized codons over the entire length of the coding sequence.
  • cytosine-content optimizable codons are codons, which contain a lower number of cytosines than other codons coding for the same amino acid.
  • any of the codons GCG, GCA, GCU codes for the amino acid Ala, which may be exchanged by the codon GCC encoding the same amino acid, and/or the codon UGU that codes for Cys may be exchanged by the codon UGC encoding the same amino acid, and/or the codon GAU which codes for Asp may be exchanged by the codon GAC encoding the same amino acid, and/or the codon that UUU that codes for Phe may be exchanged for the codon UUC encoding the same amino acid, and/or any of the codons GGG, GGA, GGU that code Gly may be exchanged by the codon GGC encoding the same amino acid, and/or the codon CAU that codes for His may be exchanged by the codon CAC encoding the same amino acid, and/or any of the codons AUA, AUU that code for He may be exchanged by the codon AUC, and/or any of the codons UUG, UUA, CUG, C
  • the number of cytosines is increased by 1 per exchanged codon.
  • Exchange of all non C-optimized codons (corresponding to C-optimizable codons) of the coding sequence results in a C-maximized coding sequence.
  • at least 70 %, preferably at least 80 %, more preferably at least 90 %, of the non C-optimized codons within the at least one coding sequence of the RNA sequence according to the invention are replaced by C-optimized codons.
  • the percentage of C-optimizable codons replaced by C-optimized codons is less than 70 %, while for other amino acids the percentage of replaced codons is higher than 70 % to meet the overall percentage of C-optimization of at least 70 % of all C-optimizable wild type codons of the coding sequence.
  • any modified C-enriched RNA sequence preferably contains at least 50 % C-optimized codons at C-optimizable wild type codon positions encoding any one of the above mentioned amino acids Ala, Cys, Asp, Phe, Gly, His, He, Leu, Asn, Pro, Arg, Ser, Thr, Val and Tyr, preferably at least 60 %.
  • codons encoding amino acids which are not cytosine content-optimizable and which are, however, encoded by at least two codons, may be used without any further selection process.
  • the codon of the wild type sequence that codes for a relatively rare tRNA in the cell e.g. a human cell
  • the relatively rare codon GAA coding for Glu may be exchanged by the relative frequent codon GAG coding for the same amino acid
  • the relatively rare codon AAA coding for Lys may be exchanged by the relative frequent codon AAG coding for the same amino acid
  • the relatively rare codon CAA coding for Gin may be exchanged for the relative frequent codon CAG encoding the same amino acid.
  • the at least one coding sequence as defined herein may be changed compared to the coding sequence of the respective wild type nucleic acid in such a way that an amino acid encoded by at least two or more codons, of which one comprises one additional cytosine, such a codon may be exchanged by the C-optimized codon comprising one additional cytosine, wherein the amino acid is preferably unaltered compared to the wild type sequence.
  • the nucleic acid sequence, particularly the RNA sequence of the present invention may contain 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 40 to 80 adenosine nucleotides or even more preferably about 50 to 70 adenosine nucleotides.
  • the poly(A) sequence in the RNA sequence of the present invention is derived from a DNA template by RNA in vitro transcription.
  • the poly(A) sequence may also be obtained in vitro by common methods of chemical-synthesis without being necessarily transcribed from a DNA-progenitor.
  • poly(A) sequences, or poly(A) tails may be generated by enzymatic polyadenylation of the RNA according to the present invention using commercially available polyadenylation kits and corresponding protocols known in the art.
  • the RNA as described herein optionally comprises a polyadenylation signal, which is defined herein as a signal, which conveys polyadenylation to a (transcribed) RNA by specific protein factors (e.g.
  • a consensus polyadenylation signal comprising the NN(U/T)ANA consensus sequence.
  • the polyadenylation signal comprises one of the following sequences: AA(U/T)AAA or A(U/T)(U/T)AAA (wherein uridine is usually present in RNA and thymidine is usually present in DNA).
  • the nucleic acid sequence, particularly the RNA sequence of the present invention may contain 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 poly(C) sequence in the RNA sequence of the present invention is derived from a DNA template by RNA in vitro transcription.
  • the nucleic acid sequence, particularly the RNA sequence according to the invention comprises at least one 5'- or 3'-UTR element.
  • an UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'- or 3'-UTR of any naturally occurring gene or which is derived from a fragment, a homolog or a variant of the 5'- or 3'-UTR of a gene.
  • the 5'- or 3'-UTR element used according to the present invention is heterologous to the at least one coding sequence of the RNA sequence of the invention. Even if 5'- or 3'-UTR elements derived from naturally occurring genes are preferred, also synthetically engineered UTR elements may be used in the context of the present invention.'
  • '3'UTR element' typically refers to a nucleic acid sequence, which comprises or consists of a nucleic acid sequence that is derived from a 3'UTR or from a variant of a 3'UTR.
  • a 3'UTR element in the sense of the present invention may represent the 3'UTR of a nucleic acid molecule, particularly of an RNA or DNA, preferably an mRNA.
  • a 3'UTR element may be the 3'UTR of an RNA, preferably of an mRNA, or it may be the transcription template for a 3'UTR of an RNA.
  • a 3'UTR element preferably is a nucleic acid sequence which corresponds to the 3'UTR of an RNA, preferably to the 3'UTR of an mRNA, such as an mRNA obtained by transcription of a genetically engineered vector construct.
  • the 3'UTR element fulfils the function of a 3'UTR or encodes a sequence which fulfils the function of a 3'UTR.
  • the at least one 3'UTR element comprises or consists of a nucleic acid sequence derived from the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene, or from a variant of the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene.
  • the nucleic acid sequence, particularly the RNA sequence of the present invention comprises a 3'UTR element, which may be derivable from a gene that relates to an RNA with an enhanced half-life (that provides a stable RNA), for example a 3'UTR element as defined and described below.
  • the 3' UTR element is a nucleic acid sequence derived from a 3' UTR of a gene, which preferably encodes a stable RNA, or from a homolog, a fragment or a variant of said gene
  • the 3'UTR element comprises or consists of a nucleic acid sequence, which is derived from a 3'UTR of a gene selected from the group consisting of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene, or from a variant of a 3'UTR of a gene selected from the group consisting of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene according to SEQ ID NOs: 1369-1390 of the patent application WO 2013/143700, whose disclosure is incorporated herein by reference, or from a homolog, a fragment or a variant thereof.
  • a collagen alpha gene such as a collagen alpha 1(1)
  • the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an albumin gene, preferably a vertebrate albumin gene, more preferably a mammalian albumin gene, most preferably a human albumin gene.
  • the RNA sequence according to the invention comprises a 3'-UTR element comprising a corresponding RNA sequence derived from the nucleic acids according to SEQ ID NOs: 1369-1390 of the patent application WO 2013/143700 or a fragment, homolog or variant thereof.
  • the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an alpha-or beta-globin gene, preferably a vertebrate alpha-or beta-globin gene, more preferably a mammalian alpha-or beta-globin gene, most preferably a human alpha-or beta-globin gene.
  • nucleic acid sequence which is derived from the 3'UTR of a noted gene' preferably refers to a nucleic acid sequence which is based on the 3'UTR sequence of a noted gene or on a part thereof, such as on the 3'UTR of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene or on a part thereof.
  • This term includes sequences corresponding to the entire 3'UTR sequence, i.e.
  • the full length 3'UTR sequence of a gene and sequences corresponding to a fragment of the 3'UTR sequence of a gene, such as an albumin gene, alpha-globin gene, beta-globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene.
  • a gene such as an albumin gene, alpha-globin gene, beta-globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene.
  • nucleic acid sequence which is derived from a variant of the 3'UTR of a noted gene' preferably refers to a nucleic acid sequence, which is based on a variant of the 3'UTR sequence of a gene, such as on a variant of the 3'UTR of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1(1) gene, or on a part thereof as described above.
  • This term includes sequences corresponding to the entire sequence of the variant of the 3'UTR of a gene, i.e.
  • a fragment in this context preferably consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length variant 3'UTR, which represents at least 20 %, preferably at least 30 %, more preferably at least 40 %, more preferably at least 50 %, even more preferably at least 60 %, even more preferably at least 70 %, even more preferably at least 80 %, and most preferably at least 90 % of the full- length variant 3'UTR.
  • the nucleic acid sequence particularly the RNA sequence according to the invention comprises a 5'-cap structure and/or at least one 3'- untranslated region element (3'-UTR element), preferably as defined herein. More preferably, the RNA further comprises a 5'-UTR element as defined herein.
  • the RNA sequence comprises, preferably in 5'- to 3'- direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) at least one coding sequence encoding at least one antigenic peptide or protein derived from a protein of interest or peptide of interest or a fragment or variant thereof, or a fragment or variant thereof, c.) a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from an alpha globin gene, a homolog, a fragment or a variant thereof; d.) optionally, a poly(A) sequence, preferably comprising 64 adenosines; e.) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and
  • the at least one nucleic acid sequence in particular, the RNA sequence comprises at least one 5'-untranslated region element (5'-UTR element).
  • the at least one 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a TOP gene or which is derived from a fragment, homolog or variant of the 5'-UTR of a TOP gene.
  • the 5'-UTR element does not comprise a TOP-motif or a 5'- TOP, as defined above.
  • the nucleic acid sequence of the 5'-UTR element which is derived from a 5'-UTR of a TOP gene, terminates at its 3'-end with a nucleotide located at position 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 upstream of the start codon (e.g. A(U/T)G) of the gene or RNA it is derived from.
  • the 5'-UTR element does not comprise any part of the protein coding sequence.
  • the only protein coding part of the at least one nucleic acid sequence, particularly of the RNA sequence is provided by the coding sequence.
  • the nucleic acid sequence derived from the 5'-UTR of a TOP gene is preferably derived from a eukaryotic TOP gene, preferably a plant or animal TOP gene, more preferably a chordate TOP gene, even more preferably a vertebrate TOP gene, most preferably a mammalian TOP gene, such as a human TOP gene.
  • the 5'-UTR element is preferably selected from 5'-UTR elements comprising or consisting of a nucleic acid sequence, which is derived from a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, whose disclosure is incorporated herein by reference, from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, from a variant thereof, or preferably from a corresponding RNA sequence.
  • the 5'-UTR element of the nucleic acid sequence comprises or consists of a nucleic acid sequence, which is derived from a nucleic acid sequence extending from nucleotide position 5 (i.e. the nucleotide that is located at position 5 in the sequence) to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position
  • nucleic acid sequence selected from SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application
  • WO2013/143700 from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO2013/143700 from a variant thereof, or a corresponding RNA sequence.
  • the 5' UTR element is derived from a nucleic acid sequence extending from the nucleotide position immediately 3' to the 5'-TOP to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g.
  • nucleotide position immediately 5' to the ATG sequence of a nucleic acid sequence selected from SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO2013/143700, from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO2013/143700, from a variant thereof, or a corresponding RNA sequence.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a TOP gene encoding a ribosomal protein or from a variant of a 5'-UTR of a TOP gene encoding a ribosomal protein.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 170, 193, 244, 259, 554, 650, 675, 700, 721, 913, 1016, 1063, 1120, 1138, and 1284-1360 of the patent application WO2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'-TOP motif.
  • the sequence extending from position 5 to the nucleotide immediately 5' to the ATG corresponds to the 5'-UTR of said sequences.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a TOP gene encoding a ribosomal Large protein (RPL) or from a homolog or variant of a 5'-UTR of a TOP gene encoding a ribosomal Large protein (RPL).
  • RPL ribosomal Large protein
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 259, 1284-1318, 1344, 1346, 1348-1354, 1357, 1358, 1421 and 1422 of the patent application WO2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'-TOP motif.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, or from a variant of the 5'UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, wherein preferably the 5'-UTR element does not comprise the 5'- TOP of said gene.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID NO: 17
  • SEQ ID NO: 17 5'-UTR of human ribosomal protein Large 32 lacking the 5' terminal oligopyrimidine tract: GGCGCTGCCTACGGAGGTGGCAGCCATCTCCTTCTCGGCATC; corresponding to SEQ ID No.
  • the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence of the above described sequences, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'UTR.
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the RNA sequence according to the invention comprises a 5'-UTR element, which comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a vertebrate TOP gene, such as a mammalian, e.g.
  • a human TOP gene selected from RPSA, RPS2, RPS3, RPS3A, RPS4, RPS5, RPS6, RPS7, RPS8, RPS9, RPS10, RPS11, RPS12, RPS13, RPS14, RPS15, RPS15A, RPS16, RPS17, RPS18, RPS19, RPS20, RPS21, RPS23, RPS24, RPS25, RPS26, RPS27, RPS27A, RPS28, RPS29, RPS30, RPL3, RPL4, RPL5, RPL6, RPL7, RPL7A, RPL8, RPL9, RPL10, RPL10A, RPL11, RPL12, RPL13, RPL13A, RPL14, RPL15, RPL17, RPL18, RPL18A, RPL19, RPL21, RPL22, RPL23, RPL23A, RPL24, RPL26, RPL27, RPL27A, RPL28, RPL29, R
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL21), an ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit 1, cardiac muscle (ATP5A1) gene, an hydroxysteroid (17-beta) dehydrogenase 4 gene (HSD17B4), an androgen- induced 1 gene (AIGl), cytochrome c oxidase subunit Vic gene (COX6C), or a N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAHl) or from a variant thereof, preferably from a vertebrate ribosomal protein Large 32 gene (RPL32), a vertebrate ribosomal protein Large 35
  • RPL21
  • HSD17B4 a mammalian androgen-induced 1 gene (AIG1), a mammalian cyto-chrome c oxidase subunit Vic gene (COX6C), or a mammalian N-acylsphingosine ami-dohydrolase (acid
  • ceramidase) 1 gene or from a variant thereof, most preferably from a human ribosomal protein Large 32 gene (RPL32), a human ribosomal protein Large 35 gene (RPL35), a human ribosomal protein Large 21 gene (RPL21), a human ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit 1, cardiac muscle (ATP5A1) gene, a human hydroxysteroid (17-beta) dehydrogenase 4 gene (HSD17B4), a human androgen-induced 1 gene (AIG1), a human cytochrome c oxidase subunit Vic gene (COX6C), or a human N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1) or from a variant thereof, wherein preferably the 5'UTR element does not comprise the 5 OP of said gene.
  • RPL32 human ribosomal protein Large 32 gene
  • RPL35 human
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence according to SEQ ID NOs: 1412-1420 of the patent application WO2013/143700, or a
  • the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence according SEQ ID NOs: 1412-1420 of the patent application WO 2013/143700, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20 % etc.
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence according to SEQ ID NO: 18 (5'-UTR of ATP5A1 lacking the 5' terminal oligopyrimidine tract) : GCGGCTCGGCCATTTTGTCCCAGTCAGTCCGGAGGCTGCGGCTGCAGAAGTACCGCCTGCGGAGTAACTG CAAAG; corresponding to SEQ ID NO: 1414 of the patent application WO 2013/143700) or preferably to a corresponding RNA sequence, or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucle
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the at least one 5'-UTR element and the at least one 3'UTR element act synergistically to increase protein production from the at least one RNA sequence as described above.
  • the RNA sequence according to the invention comprises, preferably in 5'- to 3'-direction: a. ) a 5'-cap structure, preferably m7GpppN; b. ) a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, a homolog, a fragment or a variant thereof; c. ) at least one coding sequence encoding at least one antigenic peptide or protein derived from a protein of interest or peptide of interest or a fragment or variant thereof, d.
  • a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from a gene providing a stable RNA, a homolog, a fragment or a variant thereof; e. ) optionally, a poly(A) sequence preferably comprising 64 adenosines; and f. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines.
  • the nucleic acid sequence, particularity the RNA sequence used according to the invention comprises a histone stem-loop sequence/structure.
  • histone stem-loop sequences are preferably selected from histone stem-loop sequences as disclosed in WO 2012/019780, the disclosure of which is incorporated herewith by reference.
  • a histone stem-loop sequence suitable to be used within the present invention, is preferably selected from at least one of the following formulae V or VI: formula V (stem-loop sequence without stem bordering elements) :
  • steml loop stem2 formula VI (stem-loop sequence with stem bordering elements) :
  • steml or stem2 bordering elements Ni-6 is a consecutive sequence of 1 to 6, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof;
  • steml [N0-2GN3-5] is reverse complementary or partially reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1, more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A
  • non-Watson-Crick base pairing e.g. wobble base pairing, reverse Watson-Crick base pairing, Hoogsteen base pairing, reverse Hoogsteen base pairing or are capable of base pairing with each other forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between steml and stem2, on the basis that one or more bases in one stem do not have a complementary base in the reverse complementary sequence of the other stem.
  • the nucleic acid sequence, particularly the RNA sequence may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae Va or Via: formula Va (stem-loop sequence without stem bordering elements):
  • steml loop stem2 formula Via (stem-loop sequence with stem bordering elements):
  • the at least one nucleic acid preferably the at least one RNA may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae Vb or VIb:
  • a particularly preferred histone stem-loop sequence is the sequence
  • CAAAGGCTCTTTTCAGAGCCACCA (according to SEQ ID NO: 19) or more preferably the corresponding RNA sequence CAAAGGCUCUUUUCAGAGCCACCA (according to SEQ ID NO: 20).
  • nucleic acid sequence in particular, to the DNA and/or RNA sequence of the present invention, and further to any DNA or RNA as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the respective nucleic acid sequence.
  • the nucleic acid sequence according to the invention particularly the RNA sequence according to the present invention which comprises at least one coding sequence as defined herein, may preferably comprise a 5' UTR and/or a 3' UTR preferably containing at least one histone stem-loop.
  • the 3' UTR of the RNA sequence according to the invention preferably comprises also a poly(A) and/or a poly(C) sequence as defined herein.
  • the single elements of the 3' UTR may occur therein in any order from 5' to 3' along the sequence of the RNA sequence of the present invention.
  • further elements as described herein may also be contained, such as a stabilizing sequence as defined herein (e.g. derived from the UTR of a globin gene), IRES sequences, etc.
  • Each of the elements may also be repeated in the RNA sequence according to the invention at least once (particularly in di- or multicistronic constructs), preferably twice or more.
  • the single elements may be present in the nucleic acid sequence, particularly in the RNA sequence according to the invention in the following order:
  • the nucleic acid sequence used in the present invention preferably comprises at least one of the following structural elements: a 5'- and/or 3'- untranslated region element (UTR element), particularly a 5'- UTR element, which preferably comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene or from a fragment, homolog or a variant thereof, or a 5'- and/or 3'-UTR element which may preferably be derivable from a gene that provides a stable RNA or from a homolog, fragment or variant thereof; a histone-stem-loop structure, preferably a histone- stem-loop in its 3' untranslated region; a 5'-cap structure; a poly-A tail; or a poly(C) sequence.
  • UTR element 5'- and/or 3'- untranslated region element
  • a 5'- UTR element which preferably comprises or consists of a nucleic acid sequence which is derived from the 5'-
  • the nucleic acid sequence comprises, preferably in 5'- to 3'-direction: a.) a 5'-CAP structure, preferably m7GpppN; b.) at least one coding sequence encoding at least one antigenic peptide of interest or protein of interest or a fragment or variant thereof, c.) a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from an alpha globin gene, a homolog, a fragment or a variant thereof; d.) optionally, a poly(A) sequence, preferably comprising 64 adenosines; e.) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and f.) optionally, a histone stem-loop, preferably comprising the RNA sequence according to SEQ ID NO: 20.
  • the nucleic acid sequence in particular, the RNA sequence used according to the invention comprises, preferably in 5'- to 3'- direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, a homolog, a fragment or a variant thereof; c. ) at least one coding sequence encoding at least one antigenic peptide of interest or protein of interest or a fragment or variant thereof, d.
  • a 5'-CAP structure preferably m7GpppN
  • a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, a homolog, a fragment or a variant thereof
  • c. at least one coding sequence encoding at least one antigenic peptide of interest or protein of interest or a fragment
  • a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from a gene providing a stable RNA; e. ) optionally, a poly(A) sequence preferably comprising 64 adenosines; f. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and optionally, a histone stem-loop, preferably comprising the RNA sequence according to SEQ ID NO: 20.
  • Nucleic acids used according to the present invention may be prepared by any method known in the art, including synthetic methods such as e.g. solid phase synthesis, as well as in vitro methods, such as in vitro transcription reactions or in vivo reactions, such as in vivo propagation of DNA plasmids in bacteria.
  • the nucleic acid is in the form of a coding nucleic acid, preferably an mRNA, which additionally or alternatively encodes a secretory signal peptide.
  • a secretory signal peptide typically exhibit a length of about 15 to 30 amino acids and are preferably located at the N-terminus of the encoded peptide, without being limited thereto.
  • Signal peptides as defined herein, preferably allow the transport of the encoded protein or peptide to a specific cell region or into a specific cellular compartment, such as to the cell surface, the endoplasmic reticulum (ER) or the endosomal-lysosomal compartment.
  • Proteins or peptides encoded by the nucleic acid may represent fragments or variants of naturally occurring proteins. Such fragments or variants may typically comprise a sequence having a sequence identity with one of the above mentioned proteins or peptides or sequences of their encoding nucleic acid sequences of at least 5 %, 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, preferably at least 70 %, more preferably at least 80 %, equally more preferably at least 85 %, even more preferably at least 90 % and most preferably at least 95 % or even 97 %, to the entire wild-type sequence, either on nucleic acid level or on amino acid level.
  • Proteins or peptides in the context of the present invention may comprise a sequence of an protein or peptide as defined herein, which is, with regard to its amino acid sequence or its encoded nucleic acid sequence, N-terminally, C-terminally and/or
  • nucleic acids intrasequentially truncated compared to the amino acid sequence of the native protein or its encoded nucleic acid sequence. Such truncation may occur either on the amino acid level or on the nucleic acid level.
  • a sequence identity with respect to such a fragment may therefore refer to the entire protein or peptide or to the entire coding nucleic acid sequence. The same applies accordingly to nucleic acids.
  • Such fragments of proteins or peptides may comprise a sequence of about 6 to about 20 or more amino acids, which includes fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 11, or 12 amino acids), as well as fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g. 13, 14, 15, 16, 17, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognised in their native form.
  • the fragments of proteins or peptides may also comprise epitopes of those proteins or peptides.
  • Epitopes also called "antigen determinants"
  • epitopes are typically fragments located on the outer surface of native proteins or peptides, preferably having 5 to 15 amino acids, more preferably having 5 to 12 amino acids, 6 to 9 amino acids, which may be recognised by antibodies or B-cell receptors in their native form.
  • Such epitopes may furthermore be selected from any of the herein mentioned variants of such proteins or peptides.
  • antigenic determinants can be conformational or discontinous epitopes which are composed of segments of the proteins or peptides that are discontinuous in the amino acid sequence of the proteins or peptides, but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain.
  • "Variants" of proteins or peptides as defined herein may be encoded by the nucleic acid, wherein nucleotides encoding the protein or peptide are replaced such that the encoded amino acid sequence is changed. Thereby a protein or peptide with one or more mutations is generated, such as with one or more substituted, inserted and/or deleted amino acids.
  • these fragments and/or variants have the same biological function or specific activity compared to the full-length native protein, e.g. its specific antigenic property.
  • the composition of the invention comprises the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound at a ratio of not higher than about 2 nmol lipid per ⁇ g nucleic acid compound, i.e. at a relatively low amount of cationic lipid. It has been unexpected that such low amounts of cationisable or permanently cationic lipid or lipidoid are highly effective in delivering the nucleic acid compound to cells, in particular when the composition is administered by local or locoregional administration as described in more detail below rather than by intravenous or intraarterial injection.
  • the composition comprises the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound at a ratio of not higher than about 1 nmol lipid or lipidoid per ⁇ g nucleic acid compound. In other specific embodiments, this ratio is in the range from about 0.05 to about 2 nmol/ ⁇ g, or from about 0.1 to about 1.5 nmol/ ⁇ g, or from about 0.25 to about 1.0 nmol ⁇ g, or from about 0.3 to about 0.8 nmol ⁇ g, such as about 0.4 nmol ⁇ g, respectively.
  • a “complex”, as used herein, is an association of molecules into larger units held together by forces that are weaker than covalent chemical bonds. Such complex may also be referred to as an association complex.
  • the forces by which a complex is held together are often hydrogen bonds, also known as hydrogen bridges, London forces, and/or dipolar attraction.
  • a complex involving a lipid and a nucleic acid is often referred to as a lipoplex, and a complex between a polymer and a nucleic acid is known as a polyplex.
  • the complex formed in the composition of the invention by the cationisable or permanently cationic lipid and the nucleic acid compound may be referred to as a lipoplex.
  • further constituents may participate in such complex.
  • the complex of the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound may not be fully soluble in an aqueous environment, but may exist in the form of a colloid, such as a nanoparticle, or a plurality of nanoparticles.
  • the essential constituents, i.e. the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound are incorporated in one or more nanoparticles.
  • the composition may comprise one or more nanoparticles comprising the cationisable or permanently cationic lipid or lipidoid and nucleic acid compound as described herein-above.
  • nanoparticles may be formed spontaneously when the cationisable or permanently cationic lipid and the nucleic acid cargo (or compound) are combined, e.g. in an aqueous environment.
  • nanoparticle is a submicron particle having any structure or morphology.
  • Submicron particles may also be referred to as colloids, or colloidal.
  • a nanoparticle may be classified, for example, as a nanocapsule, a vesicle, a liposome, a lipid nanoparticle, a micelle, a crosslinked micelle, a lipoplex, a polyplex, a mixed or hybrid complex, to mention only a few of the possible designations of specific types of nanoparticles.
  • the invention is also directed to the above-defined nanoparticle as such, as well as to a composition, in particular a pharmaceutical composition, comprising a plurality of such nanoparticles, in particular to a plurality of the preferred nanoparticles as described in more detail below.
  • a composition in particular a pharmaceutical composition
  • Any references to the "nanoparticle” of the invention should also be understood as referring to the plurality of such nanoparticles as typically comprised in a composition, and vice versa.
  • the nanoparticles may comprise a further biologically active cargo material in addition to the nucleic acid compound, or they may comprise any other compound having a carrier function or an auxiliary function.
  • a "biologically active cargo material” generically refers to a compound, or mixture or combination of compounds, which is intended to be delivered to a subject, or to an organ, tissue, or cell of a subject, by means of a formulation, carrier, vector or vehicle, in order to achieve a desired biologic effect, such as a pharmacological effect, including any type of prophylactic, therapeutic, diagnostic, or ameliorating effect.
  • the delivery of biologically active cargo material is the purpose of administering a product comprising such material, whereas the formulation, or carrier, vector or vehicle, which may in some cases also be considered as biologically active, are primarily the means for delivering the cargo material.
  • a preferred type of cargo is a nucleic acid, or a nucleic acid-based material.
  • a “carrier”, or “vehicle”, as used herein, may generically mean any compound, construct or material being part of a formulation which aids, enables, or improves the delivery of the biologically active compound or material. It may be biologically substantially inert, or it may be biologically active in that it interacts substantially with tissues, cells or subcellular components of the subject and, for example, enhance the uptake of the biologically active cargo material. In the context of the invention, the terms may be applied to the cationisable or permanently cationic lipid.
  • a “formulation”, with respect to a biologically active compound that is incorporated in it and administered by means of the formulation, is any product which is pharmaceutically acceptable in terms of its composition and manufacturing method which comprises at least one biologically active compound and one excipient, carrier, vehicle or other auxiliary material.
  • the nanoparticle of the invention substantially consists of the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound, or of the cargo-carrier complex as defined above.
  • the expression “substantially consists of should not be understood such as to exclude the presence of minor amounts of auxiliary materials in the nanoparticles such as solvents, cosolvents, surfactants, isotonising agents and the like.
  • the nanoparticle further comprises one or more compounds independently selected from targeting agents, cell penetrating agents, and stealth agents.
  • targeting agents cell penetrating agents, and stealth agents.
  • a targeting agent is a compound that has affinity to a target, such as a target located on or at the surface of a target cell, or an intracellular target.
  • the targeting agent may represent an antibody, an antibody fragment, or a small molecular agent having affinity to a target of interest.
  • Cell penetrating agents include cell-penetrating peptides (CPPs), as well as any other compounds with a similar biological or biomimetic function, i.e. to facilitate the uptake of cargo into cells.
  • a stealth agent in the context of the invention, means a compound or material which, when incorporated in the nanoparticle comprising the cationisable or permanently cationic lipid and the nucleic acid cargo, leads to a longer circulation time of the nanoparticle in the bloodstream of a subject to which the nanoparticle is injected, e.g. by intravenous injection or infusion.
  • An example for a stealth agent is a pegylated lipid whose lipid domain is capable of functioning as an anchor to the nanoparticle by e.g.
  • the nanoparticle shows decreased interaction with a subject's immune system while circulating in the bloodstream, which is typically associated with a prolonged elimination half life of the nanoparticle from the blood, as well as reduced immunogenicity and antigenicity.
  • PEG polyethylene glycol
  • pegylated lipids examples include l-(monomethoxy-polyethyleneglycol)-2,3- dimyristoylglycerol (PEG-DMG), N-[(methoxy poly(ethylene glycol) 2ooo)carbamoyl]-l,2- dimyristyloxypropyl-3-amine (PEG-C-DMA), or l,2-diacyal-sn-glycero-3-phosphoethanolamine- N-[methoxy(polyethylene glycol)]; in case of the latter, acyl may mean e.g. myristoyl, palmitoyl, stearoyl, or oleoyl, and the polyethylene glycol is typically polyethylene glycol-350 to
  • polyethylene glycol-5000 in particular polyethylene glycol-750, polyethylene glycol-1000, polyethylene glycol-2000, and polyethylene glycol-3000.
  • compositions and the nanoparticles of the invention are designed for local or locoregional administration, the incorporation of a stealth agent may not normally be required, unless for the purpose of reducing the immunogenicity of the composition.
  • the nanoparticle may optionally comprise a lipid which is not a permanently cationic lipid.
  • lipids which could potentially be useful in certain cases include cationisable lipids, uncharged lipids, in particular steroids such as cholesterol, and pegylated lipids such as pegylated phospholipids, which may also function as a stealth agent.
  • the inventors believe that in many cases the incorporation of any other lipids which are not cationisable or permanently cationic is not required, in particular not for cases in which the nucleic acid compound is to be delivered to cell by using a local or locoregional route of administration, as described in more detail above.
  • the nanoparticle is substantially free of non-cationic lipids or lipidoids.
  • the composition of the invention is also substantially free of non-cationic or lipids or lipidoids.
  • the nanoparticle or the composition does not contain any pharmaceutically relevant amounts of a non-cationic lipid nor of a non-cationic lipidoid.
  • the nanoparticle and/or composition is free of lipids or lipidoids which are not permanently cationic.
  • the nanoparticle essentially consists of one or more cationisable or permanently cationic lipids or lipidoids, one or more nucleic acid compounds, and optionally one or more compounds independently selected from targeting agents, cell penetrating agent, and stealth agents.
  • At least about 50 wt.-% of the nanoparticles in the composition of the invention consist of the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound, or at least 60 wt.-% thereof, at least 70 wt.-% thereof, at least 80 wt.-% thereof, at least 85 wt.-% thereof, at least 90 wt.-% thereof, or at least 95 wt.-% thereof, respectively.
  • the nanoparticles preferably and typically comprise at least a cationisable or permanently cationic lipid or lipidoid and a nucleic acid compound.
  • lipid and the nucleic acid all options and preferences as described above in the context of the composition in general also apply to the nanoparticles.
  • the nucleic acid compound comprised in the nanoparticles may be any chemically modified or unmodified DNA, single stranded or double stranded DNA, coding or non-coding DNA, optionally selected from plasmid, (short)
  • oligodesoxynucleotide i.e. a (short) DNA oligonucleotide
  • genomic DNA DNA primers
  • DNA probes DNA probes
  • immunostimulatory DNA aptamer
  • such a nucleic acid molecule may be selected e.g. from any PNA (peptide nucleic acid).
  • PNA peptide nucleic acid
  • the nucleic acid is selected from chemically modified or unmodified RNA, single-stranded or double-stranded RNA, coding or non-coding RNA, optionally selected from messenger RNA (mRNA), (short) oligoribonucleotide (i.e.
  • RNA oligonucleotide a (short) RNA oligonucleotide), viral RNA (vRNA), replicon RNA, transfer RNA (tRNA),ribosomal RNA (rRNA), immunostimulatory RNA (isRNA), microRNA, small interfering RNA (siRNA), small nuclear RNA (snRNA), small-hairpin RNMA (shRNA) or a riboswitch, an RNA aptamer, an RNA decoy, an antisense RNA, a ribozyme, or any combination thereof, as described herein-above.
  • the nucleic acid molecule of the complex is an RNA. More preferably, the nucleic acid molecule of the complex is a (linear) single- stranded RNA, even more preferably an mRNA or an immunostimulatory RNA.
  • the biologically active cargo material is a combination of more than one nucleic acid compounds.
  • the nanoparticles preferably have a hydrodynamic diameter as determined by dynamic laser scattering of not more than about 1,000 nm. More preferably, their hydrodynamic diameter is not higher than about 800 nm, such as in the range from about 30 nm to about 800 nm. In other preferred embodiments, the hydrodynamic diameter is in the range from about 50 nm to about 300 nm, or from about 60 nm to about 250 nm, from about 60 nm to about 150 nm, or from about 60 nm to about 120 nm, respectively. While these are preferred diameters of individual nanoparticles, this does not exclude the presence of nanoparticles of other diameters in the composition of the invention. However, the invention is preferably practised with compositions in which many - or even most - of the nanoparticles exhibit such diameters.
  • composition according to the invention which comprises a plurality of such nanoparticles may also be characterised by the mean hydrodynamic diameter as determined by dynamic laser scattering, which is also preferably not higher than 800 nm, such as in the range from about 30 nm to about 800 nm.
  • the "mean” should be understood as the Z-average, also known as the cumulants mean.
  • the measurement by dynamic laser scattering must also be conducted with an appropriate dispersant and at an appropriate dilution, following the recommendations of the manufacturer of the analytic equipment that is used.
  • a mean hydrodynamic diameter in the range from about 50 nm to about 300 nm, or from about 60 nm to about 250 nm, from about 60 nm to about 150 nm, or from about 60 nm to about 120 nm, respectively.
  • the nanoparticles may further be characterised by their electrokinetic potential, which may be expressed by means of the zeta potential.
  • the zeta potential is in the range from about 0 mV to about +50 mV, or from about 0 mV to about +10 mV, respectively.
  • the zeta potential is positive, i.e. higher than 0 mV, but not higher than +50 mV, or +40 mV, or +30 mV, or +20 mV, or +10 mV, respectively.
  • the zeta potential is in the range from about 0 mV to about -50 mV, or from about 0 mV to about -10 mV, respectively. In other embodiments, the zeta potential is negative, i.e. lower than 0 mV, but not lower than -50 mV, or -40 mV, or -30 mV, or - 20 mV, or -10 mV, respectively.
  • the zeta potential is in the range of 0 mV to -50 mV and the particles have an N/P ratio of 1 or less. It has been found by the inventors that such nanoparticles are particularly suitable for local administration.
  • the amount of the cationisable or permanently cationic lipid or lipidoid should also be selected taking the amount of the nucleic acid cargo into account. In one embodiment, these amounts are selected such as to result in an N/P ratio of the nanoparticle(s) or of the composition in the range from about 0.1 to about 20.
  • the N/P ratio is defined as the mole ratio of the nitrogen atoms ("N") of the basic nitrogen-containing groups of the lipid or lipidoid to the phosphate groups ("P") of the nucleic acid which is used as cargo.
  • the N/P ratio may be calculated on the basis that, for example, 1 ⁇ g RNA typically contains about 3 nmol phosphate residues, provided that the RNA exhibits a statistical distribution of bases.
  • the "N"-value of the lipid or lipidoid may be calculated on the basis of its molecular weight and the relative content of cationisable or permanently cationic and - if present - cationisable groups.
  • the N/P ratio of the nanoparticles or the composition is not higher than about 10. In another preferred embodiment, the N/P ratio is not higher than about 6, or not higher than about 3, respectively. In a preferred embodiment, the N/P ratio of the nanoparticles or the composition is not higher than about 1. For example, the N/P ratio may be in the range from about 0.01 to about 1, or from about 0.1 to about 0.99. In further preferred embodiments, the N/P ratio of the
  • nanoparticles or the composition is in the range from about 0.2 to about 0.9, such as about 0.3 ⁇ 0.2, 0.5 ⁇ 0.2, or 0.7 ⁇ 0.2.
  • Such low N/P ratios are commonly believed to be detrimental to the performance and in vivo efficacy of such carrier-cargo complexes, or nucleic-acid loaded nanoparticles.
  • the inventors found that such N/P ratios are indeed useful in the context of the present invention, in particular when the local or extravascular administration of the nanoparticles is intended.
  • the respectively nanoparticles have been found to be efficacious and at the same time well- tolerated.
  • the amount of cationic lipid in the composition of the invention as well as in the nanoparticle(s) is typically much lower than in conventional lipid-based carriers for nucleic acids as cargo.
  • the present invention may be practised with as little as about 0.1 to about 10% of the typical amount of lipids or lipidoids used in lipoplexes or lipid nanoparticles that have been proposed for the delivery of e.g. RNA and the transfection of cells.
  • the inventors assume that such low amount of lipid or lipidoid has been pivotal in achieving the high tolerability of the composition of the invention.
  • the nanoparticles may be prepared by a method comprising the step of combining one or more cationisable or permanently cationic lipids and/or lipidoids, optionally dissolved in an appropriate solvent (e.g. ethanol, DMSO), and one or more nucleic acid compounds in the presence of an aqueous liquid such as to allow the formation of a nanoparticle or a plurality of nanoparticles.
  • an appropriate solvent e.g. ethanol, DMSO
  • the mixing can be conducted by an suitable mixing device (e.g. laminar flow combination utilizing a T or Y valve; microfluidic devices or simple addition to a stirred solution).
  • composition of the invention which comprises the cationisable or permanently cationic lipid or lipidoid, the nucleic acid compound as cargo and optionally one or more further ingredients, and in particular the composition which comprises the complex and/or the nanoparticles as described above, is preferably formulated and processed such as to be suitable for administration to a subject, in particular to an animal or to a human subject.
  • the composition is sterile.
  • the composition may also be referred to as a pharmaceutical composition.
  • a pharmaceutical composition as defined herein where the nucleic acid compound is a coding nucleic acid which encodes at least one peptide or protein.
  • the coding nucleic acid may encode a therapeutically active protein or an antigen.
  • the invention is further directed to a vaccine comprising such
  • the vaccine may consist of the pharmaceutical composition, or it may comprise it along with other constituents.
  • inventive pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques.
  • the pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may be administered by parenteral injection, more preferably by subcutaneous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, or intraperitoneal, injection or infusion. Particularly preferred is intradermal and intramuscular injection.
  • parenteral injection more preferably by subcutaneous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, or intraperitoneal, injection or infusion.
  • intradermal and intramuscular injection is particularly preferred.
  • compositions may be aqueous. Alternatively, they may be oily suspensions. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • compositions, nanoparticle or composition comprising said nanoparticle as defined herein may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • the pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including diseases of the skin or of any other accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs.
  • the inventive pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may be formulated in a suitable formulation such as a liquid or an ointment, containing the nucleic acid suspended or dissolved in one or more carriers.
  • the pharmaceutical composition, nanoparticle or composition comprising said nanoparticle typically comprises a "safe and effective amount" of the components, particularly of the nucleic acid.
  • a "safe and effective amount” means an amount of nucleic acid is sufficient to significantly induce a prevention, or positive modification of a disease or disorder as defined herein. At the same time, however, a "safe and effective amount” is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk.
  • the composition, nanoparticle or composition comprising said nanoparticle may be designed as a ready-to-use injectable formulation.
  • it may be formulated as a sterile liquid suitable for injection.
  • it may be provided as a sterile aqueous solution, or a sterile aqueous suspension of nanoparticles, preferably with a pH in the range from about 4 to about 9, or more preferably from about 4.5 to about 8.5.
  • the osmolality of such liquid composition is preferably from about 150 to about 500 mOsmol/kg, and more preferably from about 200 to about 400 mOsmol/kg.
  • the pH may also be in the range from about 4.5 to about 8, or from about 5 to about 7.5; and the osmolality may in this case preferably be selected in the range from about 220 to about 350 mOsmol/kg, or from about 250 to about 330 mOsmol/kg, respectively.
  • the composition, nanoparticle or composition comprising said nanoparticle may be formulated as a concentrated form which requires dilution or even reconsitution before use.
  • it may be in the form of a liquid concentrate, which could be an aqueous and/or organic liquid formulation which requires dilution with an aqueous solvent or diluent.
  • the liquid concentrate comprises an organic solvent, such solvent is preferably selected from water- miscible organic solvents with relatively low toxicity such as ethanol or propylene glycol.
  • the composition, nanoparticle or composition comprising said nanoparticle of the invention is provided as a dry formulation for reconstitution with a liquid carrier such as to generate a liquid formulation suitable for injection.
  • a liquid carrier such as to generate a liquid formulation suitable for injection.
  • the dry formulation may be a sterile powder or lyophilised form for reconstitution with an aqueous liquid carrier.
  • composition, nanoparticle or composition comprising said nanoparticle may optionally comprise pharmaceutical excipients as required or useful.
  • pharmaceutical excipients include acids, bases, osmotic agents, antioxidants, stabilisers, surfactants, synergists, colouring agents, thickening agents, bulking agents, and - if required - preservatives.
  • the vaccine according to the invention is based on the same components as the
  • the vaccine according to the invention comprises at least one nucleic acid and a pharmaceutically acceptable carrier.
  • the vaccine may be provided in physically separate form and may be administered by separate administration steps.
  • the vaccine according to the invention may correspond to the (pharmaceutical) composition as described herein, especially where the mRNA sequences are provided by one single composition. However, the inventive vaccine may also be provided physically separated.
  • these RNA species may be provided such that, for example, two, three, four, five or six separate compositions, which may contain at least one mRNA species/sequence each (e.g. three distinct mRNA species/sequences), each encoding distinct antigenic peptides or proteins, are provided, which may or may not be combined.
  • the inventive vaccine may be a combination of at least two distinct compositions, each composition comprising at least one mRNA encoding at least one of the antigenic peptides or proteins defined herein.
  • the vaccine may be provided as a combination of at least one mRNA, preferably at least two, three, four, five, six or more mRNAs, each encoding one of the antigenic peptides or proteins defined herein.
  • the vaccine may be combined to provide one single composition prior to its use or it may be used such that more than one administration is required to administer the distinct mRNA sequences/species encoding any of the antigenic peptides or proteins as defined herein.
  • the vaccine contains at least one mRNA sequence, typically at least two mRNA sequences, encoding the antigen combinations defined herein, it may e.g. be administered by one single administration (combining all mRNA species/sequences), by at least two separate administrations.
  • the entities of the vaccine may be provided in liquid and or in dry (e.g. lyophilized) form. They may contain further components, in particular further components allowing for its pharmaceutical use.
  • the vaccine or the (pharmaceutical) composition may, e.g., additionally contain a pharmaceutically acceptable carrier and/or further auxiliary substances and additives.
  • the vaccine or (pharmaceutical) composition typically comprises a safe and effective amount of the nucleic acid, particularly mRNA according to the invention as defined herein, encoding an antigenic peptide or protein as defined herein or a fragment or variant thereof or a combination of antigens, preferably as defined herein.
  • safety and effective amount means an amount of the mRNA that is sufficient to significantly induce a positive modification of cancer or a disease or disorder related to cancer. At the same time, however, a “safe and effective amount” is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • the expression “safe and effective amount” preferably means an amount of the mRNA (and thus of the encoded antigen) that is suitable for stimulating the adaptive immune system in such a manner that no excessive or damaging immune reactions are achieved but, preferably, also no such immune reactions below a measurable level.
  • Such a "safe and effective amount" of the mRNA of the (pharmaceutical) composition or vaccine as defined herein may furthermore be selected in dependence of the type of mRNA, e.g.
  • a "safe and effective amount" of the mRNA of the (pharmaceutical) composition or vaccine as defined above will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying physician.
  • the vaccine or composition according to the invention can be used according to the invention for human and also for veterinary medical purposes, as a pharmaceutical composition or as a vaccine.
  • the nucleic acid particularly the mRNA of the (pharmaceutical) composition, vaccine or kit of parts according to the invention is provided in lyophilized form.
  • the lyophilized mRNA is reconstituted in a suitable buffer, advantageously based on an aqueous carrier, prior to administration, e.g. Ringer-Lactate solution, saline, or a phosphate buffer solution.
  • the (pharmaceutical) composition, the vaccine or the kit of parts according to the invention contains at least one, two, three, four, five, six or more mRNAs, preferably mRNAs which are provided separately in lyophilized form (optionally together with at least one further additive) and which are preferably reconstituted separately in a suitable buffer (such as Ringer-Lactate solution or phosphate buffer) prior to their use so as to allow individual administration of each of the (monocistronic) mRNAs.
  • the vaccine or (pharmaceutical) composition according to the invention may typically contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier as used herein preferably includes the liquid or non-liquid basis of the inventive vaccine.
  • the carrier will be water, typically pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate, citrate etc. buffered solutions.
  • water or preferably a buffer, more preferably an aqueous buffer may be used, containing a sodium salt, preferably at least 50 mM of a sodium salt, a calcium salt, preferably at least 0,01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt.
  • a sodium salt preferably at least 50 mM of a sodium salt
  • a calcium salt preferably at least 0,01 mM of a calcium salt
  • optionally a potassium salt preferably at least 3 mM of a potassium salt.
  • the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc.
  • examples of sodium salts include e.g. NaCl, Nal, NaBr, Na2C03, NaHC03, Na2SC>4
  • examples of the optional potassium salts include e.g. KC1, KI, KBr, K2CO3, KHCO3, K2SO4
  • examples of calcium salts include e.g. CaC , Cah, CaBr2, CaC03, CaSC , Ca(OH)2.
  • the buffer suitable for injection purposes as defined above may contain salts selected from sodium chloride (NaCl), calcium chloride (CaC ) and optionally potassium chloride (KC1), wherein further anions may be present additional to the chlorides. CaC can also be replaced by another salt like KC1.
  • the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCl), at least 3 mM potassium chloride (KC1) and at least 0,01 mM calcium chloride (CaC ).
  • the injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e.
  • the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects.
  • Reference media are e.g. in "in vivo” methods occurring liquids such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in “in vitro” methods, such as common buffers or liquids.
  • Such common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis.
  • compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person.
  • the term "compatible” as used herein means that the constituents of the inventive vaccine are capable of being mixed with the nucleic acid, particularly the mRNA according to the invention as defined herein, in such a manner that no interaction occurs, which would substantially reduce the pharmaceutical effectiveness of the inventive vaccine under typical use conditions.
  • Pharmaceutically acceptable carriers, fillers and diluents must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated.
  • Some examples of compounds which can be used as pharmaceutically acceptable carriers, fillers or constituents thereof are sugars, such as, for example, lactose, glucose, trehalose and sucrose; starches, such as, for example, corn starch or potato starch; dextrose; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulfate; vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol,
  • composition or vaccine can be administered, for example, systemically or locally.
  • routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes.
  • Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial,
  • composition or vaccines according to the present invention may be administered by an intradermal, subcutaneous, intramuscular or otherwise local or locoregional route, preferably by injection, which may be needle-free and/or needle injection.
  • Compositions/vaccines are therefore preferably formulated in liquid or solid form.
  • the suitable amount of the vaccine or composition according to the invention to be administered can be determined by routine experiments, e.g. by using animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non-human primate models.
  • Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4.
  • Suitable carriers for injection include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices.
  • Suitable pharmaceutically acceptable carriers for topical application include those which are suitable for use in lotions, creams, gels and the like. If the inventive composition or vaccine is to be administered perorally, tablets, capsules and the like are the preferred unit dose form.
  • the pharmaceutically acceptable carriers for the preparation of unit dose forms which can be used for oral administration are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art.
  • the inventive vaccine or composition can additionally contain one or more auxiliary substances in order to further increase the immunogenicity.
  • a synergistic action of the nucleic acid contained in the inventive composition and of an auxiliary substance, which may be optionally be co-formulated (or separately formulated) with the inventive vaccine or composition as described above, is preferably achieved thereby.
  • various mechanisms may play a role in this respect.
  • emulsifiers such as, for example, Tween
  • wetting agents such as, for example, sodium lauryl sulfate
  • colouring agents such as, for example, sodium lauryl sulfate
  • taste-imparting agents pharmaceutical carriers
  • tablet-forming agents such as, for example, stabilizers; antioxidants; preservatives.
  • the inventive vaccine or composition can also additionally contain any further compound, which is known to be immune-stimulating due to its binding affinity (as ligands) to human Tolllike receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 or TLR13.
  • any further compound which is known to be immune-stimulating due to its binding affinity (as ligands) to human Tolllike receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 or TLR13.
  • CpG nucleic acids in particular CpG-RNA or CpG-DNA.
  • a CpG-RNA or CpG- DNA 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 is preferably in the form of CpG-RNA, more preferably in the form of single-stranded CpG-RNA (ss CpG-RNA).
  • the CpG nucleic acid preferably contains at least one or more
  • CpG motif(s) (mitogenic) cytosine/guanine dinucleotide sequence(s) (CpG motif(s)).
  • CpG motif(s) (mitogenic) cytosine/guanine dinucleotide sequence(s) (CpG motif(s)).
  • at least one CpG motif contained in these sequences that is to say the C (cytosine) and the G (guanine) of the CpG motif, is unmethylated. All further cytosines or guanines optionally contained in these sequences can be either methylated or unmethylated.
  • the C (cytosine) and the G (guanine) of the CpG motif can also be present in methylated form.
  • the term 'inventive composition' may refer to the inventive composition comprising at least one artificial nucleic acid.
  • the term 'inventive vaccine' may refer to an inventive vaccine, which is based on the artificial nucleic acid, i.e. which comprises at least one artificial nucleic acid or which comprises the inventive composition comprising said artificial nucleic acid.
  • the composition or vaccine may be designed as a ready-to-use injectable formulation. In this case, it is preferably provided as a sterile aqueous solution, or suspension of nanoparticles, preferably with a pH in the range from about 4 to about 9, or more preferably from about 4.5 to about 8.5.
  • the osmolality of such liquid composition is preferably from about 150 to about 500 mOsmol/kg, and more preferably from about 200 to about 400 mOsmol/kg.
  • the pH may also be in the range from about 4.5 to about 8, or from about 5 to about 7.5; and the osmolality may in this case preferably be selected in the range from about 220 to about 350 mOsmol/kg, or from about 250 to about 330 mOsmol/kg, respectively.
  • the composition may be formulated as a concentrated form which requires dilution or even reconsitution before use.
  • it may be in the form of a liquid concentrate, which could be an aqueous and/or organic liquid formulation which requires dilution with an aqueous solvent or diluent.
  • the liquid concentrate comprises an organic solvent, such solvent is preferably selected from water-miscible organic solvents with relatively low toxicity such as ethanol or propylene glycol.
  • the composition of the invention is provided as a dry formulation for reconstitution with a liquid carrier such as to generate a liquid formulation suitable for injection.
  • a liquid carrier such as to generate a liquid formulation suitable for injection.
  • the dry formulation may be a powder, or a lyophilised form.
  • composition may optionally comprise pharmaceutical excipients as required or useful.
  • pharmaceutical excipients include acids, bases, osmotic agents, antioxidants, stabilisers, surfactants, synergists, colouring agents, thickening agents, bulking agents, and - if required - preservatives.
  • kits of parts comprising the constituents of the composition of the invention as defined herein.
  • the composition of the invention may be accommodated in one or different parts of the kit.
  • the invention provides a kit for preparing any such composition as defined herein, the kit comprising a first kit component comprising the cationisable or permanently cationic lipid or lipidoid, and a second kit component comprising the nucleic acid compound.
  • the first kit component may be provided as a sterile solid composition, such as a lyophilised form or powder, or as a sterile liquid composition.
  • the first kit component may comprise one or more inactive ingredients as described above.
  • the second kit component may be formulated, for example, as a sterile solid or liquid composition and also contain one or more additional inactive ingredients in addition to the nucleic acid compound.
  • the composition of the invention is obtained by combining and optionally mixing the content of the two components.
  • kits which comprises a first kit component comprising at least one cationisable or permanently cationic lipid or lipidoid and at least one nucleic acid compound, formulated e.g. as a sterile solid or liquid formulation, and a second kit component comprising a liquid carrier for dissolving or dispersing the content of the first kit component such as to obtain a composition of the invention as described above.
  • the kit components are preferably provided in sterile form, whether solid or liquid, and each of them may comprise one or more additional excipient, or inactive ingredient.
  • one component of the kit can comprise only one, several or all nucleic acids comprised in the kit.
  • each nucleic acid is comprised in a different component of the kit such that each component forms a part of the kit.
  • more than one nucleic acid may be comprised in a first component as part of the kit, whereas one or more other (second, third etc.) components (providing one or more other parts of the kit) may either contain one or more than one nucleic acids, which may be identical or partially identical or different from the first component.
  • any of the kit components described above comprising the cationisable or permanently cationic lipid and/or the nucleic acid compound are formulated to represent concentrates, whether in solid or liquid form, and may be designed to be diluted by a
  • biocompatible or physiologically tolerable liquid carrier which may optionally not part of the kit, such as sterile saline solution, sterile buffer, or other solutions that are frequently used as liquid diluents for injectable drugs.
  • liquid carrier typically means a well-tolerated aqueous injectable liquid composition having a physiologically acceptable composition, pH and osmolality.
  • kit or kit of parts may furthermore contain technical instructions with information on the administration and dosage of the nucleic acid sequence, the inventive pharmaceutical composition or of any of its components or parts, e.g. if the kit is prepared as a kit of parts.
  • nanoparticles, the kit and the composition as described above are particularly useful to deliver nucleic acid cargo to living cells such as to transfect the cells with the nucleic acid. This may serve a scientific research purpose, a diagnostic application or a therapy.
  • the nanoparticle(s) or the composition or the kit is used as a
  • a “medicament” means any compound, material, composition or formulation which is useful for the prophylaxis, prevention, treatment, cure, palliative treatment, amelioration, management, improvement, delay, stabilisation, or the prevention or delay of reoccurrence or spreading of a disease or condition, including the prevention, treatment or amelioration of any symptom of a disease or condition.
  • the nanoparticle or composition or kit as described above may be used in the prophylaxis, treatment and/or amelioration of diseases selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e.
  • infectious diseases preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e.
  • (hereditary) diseases or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency.
  • Such use may also be described as a method of treating a subject, in particular a human subject, having developed or being at risk of developing a disease selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e.
  • infectious diseases preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e.
  • (hereditary) diseases or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency, by administering an effective amount of the composition and/or the nanoparticles of the invention.
  • the administration of the composition and/or the nanoparticles may optionally include a reconstitution or dilution step, for example if the composition is provided as a concentrate or as a dry solid composition for reconstitution.
  • references to the composition in this respect should be understood as referring also to a composition obtained by reconstituting or diluting the solid or concentrated composition.
  • the actual administration of the composition may be effected by any known mode and route of administration, i.e. injectable or parenteral, or non-invasive, systemic or non-systemic.
  • systemic exposure may be achieved by injection or infusion of the composition into the bloodstream of a subject, in particular by intravenous or intraarterial injection or infusion.
  • the composition is administered to a subject by extravascular administration.
  • extravascular administration includes any mode and route of administration which avoids the direct injection or infusion into the bloodstream.
  • the extravascular injection is nevertheless performed by injecting, infusing or implanting the composition, even though not into a blood vessel, but to another site of administration, which site may be referred to as a local or locoregional site of administration.
  • locoregional means restricted to a region of a body.
  • the effect of local or locoregional administration is that the composition may not be rapidly diluted and distributed throughout the body of a subject by means of its systemic circulation, but that it may be retained locally or locoregionally at or near the site of administration, which is associated with an increased likelihood of local interaction with target structures or cell and a decreased systemic exposure.
  • the composition and/or nanoparticle of the invention is administered by extravascular, local or locoregional injection, infusion or implantation, in particular intradermal, subcutaneous, intramuscular, interstitial, locoregional, intravitreal, periocular, intratumoural, intralymphatic, intranodal, intra-articular, intrasynovial, periarticular, intraperitoneal, intra-abdominal, intracardial, intrapericardial, intraventricular, intrapleural, perineural, intrathoracic, epidural, intradural, peridural, intrathecal, intramedullary intracerebral, intracavernous, intracorporus cavernosum, intraprostatic, intratesticular, intracartilaginous, intraosseous, intradiscal, intraspinal, intracaudal, intrabursal, intragingival, intraovarian, intrauterine, periodontal, retrobulbar, subarachnoid, subconjunctival
  • the composition is administered topically to the skin or mucosa of a subject, in particular by dermal or cutaneous, nasal, buccal, sublingual, otic or auricular, ophthalmic, conjunctival, vaginal, rectal, intracervical, endosinusial, laryngeal, oropharyngeal, ureteral, or urethral administration.
  • the composition may be administered to the respiratory system by inhalation, in particular by aerosol administration to the lungs, bronchi, bronchioli, alveoli, or paranasal sinuses.
  • the composition may also be administered to a subject by transdermal or percutaneous administration.
  • the present invention furthermore provides several applications and uses of the composition or nanoparticle or kit or vaccine.
  • the (pharmaceutical) composition(s) or the vaccine may be used for human or for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine.
  • the invention provides the artificial nucleic acid, the inventive composition comprising at least one artificial nucleic acid, inventive composition comprising at least one polypeptide, the inventive vaccine or the inventive kit or kit of parts for use in a method of prophylactic (pre-exposure prophylaxis or post-exposure prophylaxis) and/or therapeutic treatment of e.g. virus infections.
  • the present invention is directed to the first medical use of the artificial nucleic acid, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts as defined herein as a medicament.
  • the invention provides the use of an artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprising at least one e.g. virus protein or peptide as defined herein, or a fragment or variant thereof as described herein for the preparation of a medicament.
  • the present invention is directed to the second medical use of the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts for the treatment of an infection with e.g. a virus or a disease or disorders related to an infection.
  • an infection e.g. a virus or a disease or disorders related to an infection.
  • inventive composition or the inventive vaccine in particular the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein or the inventive composition comprising at least one inventive polypeptide, can be administered, for example, systemically or locally.
  • Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes.
  • Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial, intrapulmonal, intracardial, and sublingual injections. More preferably, vaccines may be administered by an intradermal, subcutaneous, or
  • inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid) form.
  • inventive vaccine may be administered by conventional needle injection or needle-free jet injection.
  • inventive vaccine or composition may be administered by jet injection as defined herein, preferably intramuscularly or intradermally, more preferably intradermally.
  • a single dose of the artificial nucleic acid, composition or vaccine comprises a specific amount of the artificial nucleic acid as disclosed herein.
  • the artificial nucleic acid is provided in an amount of at least 40 ⁇ g per dose, preferably in an amount of from 40 to 700 ⁇ g per dose, more preferably in an amount of from 80 to 400 ⁇ g per dose.
  • the amount of the inventive artificial nucleic acid comprised in a single dose is typically at least 200 ⁇ g, preferably from 200 ⁇ g to 1.000 ⁇ g, more preferably from 300 ⁇ g to 850 ⁇ g, even more preferably from 300 ⁇ g to 700 ⁇ g.
  • the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 ⁇ g, preferably from 80 ⁇ g to 700 ⁇ g, more preferably from 80 ⁇ g to 400 ⁇ g.
  • the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 ⁇ g, preferably from 80 ⁇ g to 1.000 ⁇ g, more preferably from 80 ⁇ g to 850 ⁇ g, even more preferably from 80 ⁇ g to 700 ⁇ g.
  • the immunization protocol for the treatment or prophylaxis of e.g. a virus infection typically comprises a series of single doses or dosages of the inventive composition or the inventive vaccine.
  • a single dosage refers to the initial/first dose, a second dose or any further doses, respectively, which are preferably administered in order to "boost" the immune reaction.
  • the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts is provided for use in treatment or prophylaxis, preferably treatment or prophylaxis of e.g. a virus infection or a related disorder or disease, wherein the treatment or prophylaxis comprises the administration of a further active pharmaceutical ingredient. More preferably, in the case of the inventive vaccine or composition, which is based on the inventive artificial nucleic acid, a polypeptide may be coadministered as a further active pharmaceutical ingredient. For example, at least one e.g.
  • virus protein or peptide as described herein, or a fragment or variant thereof may be co-administered in order to induce or enhance an immune response.
  • an artificial nucleic acid as described herein may be co-administered as a further active pharmaceutical ingredient.
  • an artificial nucleic acid as described herein encoding at least one polypeptide as described herein may be co-administered in order to induce or enhance an immune response.
  • a further component of the inventive vaccine or composition may be an
  • immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably
  • immunoglobulins directed against e.g. a virus may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA.
  • a further immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the inventive artificial nucleic acid or by the inventive polypeptide.
  • the invention provides a method of treating or preventing a disorder, wherein the disorder is preferably an infection with e.g. a virus or a disorder related to an infection with e.g. a virus, wherein the method comprises administering to a subject in need thereof the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts.
  • the inventive composition comprising at least one inventive nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts.
  • such a method may preferably comprise the steps of: a) providing the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts; b) applying or administering the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts to a tissue or an organism; c) optionally administering immunoglobuline (IgGs) against e.g. the virus.
  • IgGs immunoglobuline
  • the present invention also provides a method for expression of at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, wherein the method preferably comprises the following steps: a) providing the inventive artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, preferably as defined herein, or a composition comprising said artificial nucleic acid; and b) applying or administering the inventive artificial nucleic acid or the inventive composition comprising said artificial nucleic acid to an expression system, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • an expression system e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • the method may be applied for laboratory, for research, for diagnostic, for commercial production of peptides or proteins and/or for therapeutic purposes.
  • inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, e.g. in naked or complexed form or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • the method may be carried out in vitro, in vivo or ex vivo.
  • the method may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of infectious diseases, or a related disorder.
  • the present invention also provides the use of the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, preferably for diagnostic or therapeutic purposes, for expression of e.g. an encoded virus antigenic peptide or protein, e.g. by applying or administering the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • the use may be applied for a (diagnostic) laboratory, for research, for diagnostics, for commercial production of peptides or proteins and/or for therapeutic purposes.
  • inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, preferably in naked form or complexed form, or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • a cell e.g. an expression host cell or a somatic cell
  • tissue or an organism preferably in naked form or complexed form
  • a composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • the use may be carried out in vitro, in vivo or ex vivo.
  • the use may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of e.g. a virus infection or a related disorder.
  • the invention provides the artificial nucleic acid, the inventive composition or the inventive vaccine for use as defined herein, preferably for use as a medicament, for use in treatment or prophylaxis, preferably treatment or prophylaxis of a e.g. a virus infection or a related disorder, or for use as a vaccine.
  • the composition or vaccine may be administered by conventional needle injection or needle-free jet injection, e.g. into, adjacent to and/or in close proximity to tumor tissue.
  • inventive composition or the inventive pharmaceutical composition is administered by jet injection.
  • Jet injection refers to a needle-free injection method, wherein a fluid comprising the inventive composition and, optionally, further suitable excipients is forced through an orifice, thus generating an ultra-fine liquid stream of high pressure that is capable of penetrating mammalian skin.
  • the liquid stream forms a hole in the skin, through which the liquid stream is pushed into the target tissue, e.g. tumor tissue.
  • jet injection may be used e.g. for intratumoral application of the inventive composition.
  • inventive composition or the inventive pharmaceutical composition may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intranodal, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques.
  • administration routes are intradermal and intramuscular injection.
  • inventive pharmaceutical composition may comprise further components for facilitating administration and uptake of components of the pharmaceutical composition.
  • further components may be an appropriate carrier or vehicle, antibacterial and/or antiviral agents.
  • a further component of the inventive pharmaceutical composition may be an
  • immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc.
  • a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA.
  • the inventive pharmaceutical composition typically comprises a "safe and effective amount" of the components of the inventive pharmaceutical composition, particularly of the RNA molecule(s) as defined herein.
  • a "safe and effective amount” means an amount of the RNA molecule(s) as defined herein as such that is sufficient to significantly induce a positive modification of e.g. a tumor or cancer disease.
  • a "safe and effective amount” is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • the inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general.
  • the present invention furthermore provides several applications and uses of the nucleic acid sequence as defined herein, of the inventive composition comprising a plurality of nucleic acid sequences as defined herein, of the inventive pharmaceutical composition, comprising the nucleic acid sequence as defined herein or of kits comprising same.
  • the present invention is directed to the first medical use of the nucleic acid sequence as defined herein or of the inventive composition comprising a plurality of nucleic acid sequences as defined herein as a medicament, particularly in gene therapy, preferably for the treatment of diseases as defined herein.
  • the present invention is directed to the second medical use of the nucleic acid sequence as defined herein or of the inventive composition comprising a plurality of nucleic acid sequences as defined herein, for the treatment of diseases as defined herein, preferably to the use of the nucleic acid sequence as defined herein, of the inventive composition comprising a plurality of nucleic acid sequences as defined herein, of a pharmaceutical composition comprising same or of kits comprising same for the preparation of a medicament for the prophylaxis, treatment and/or amelioration of diseases as defined herein.
  • the pharmaceutical composition is used or to be administered to a patient in need thereof for this purpose.
  • diseases as mentioned herein are preferably selected from infectious diseases, neoplasms (e.g. cancer or tumor diseases), diseases of the blood and blood-forming organs, endocrine, nutritional and metabolic diseases, diseases of the nervous system, diseases of the circulatory system, diseases of the respiratory system, diseases of the digestive system, diseases of the skin and subcutaneous tissue, diseases of the musculoskeletal system and connective tissue, and diseases of the genitourinary system.
  • inherited diseases selected from: lp36 deletion syndrome; 18p deletion syndrome; 21-hydroxylase deficiency; 45,X (Turner syndrome);
  • Acrocephalosyndactyly (Apert syndrome); acrocephalosyndactyly, type V (Pfeiffer syndrome); Acrocephaly (Apert syndrome); Acute cerebral Gaucher's disease (Gaucher disease type 2); acute intermittent porphyria; ACY2 deficiency (Canavan disease); AD (Alzheimer's disease); Sydney-type craniosynostosis (Muenke syndrome); Adenomatous Polyposis Coli (familial adenomatous polyposis); Adenomatous Polyposis of the Colon (familial adenomatous polyposis); ADP (ALA dehydratase deficiency); adenylosuccinate lyase deficiency; Adrenal gland disorders (21-hydroxylase deficiency); Adrenogenital syndrome (21-hydroxylase deficiency); Adrenoleukodystrophy; AIP (acute intermittent porphyria); AIS (acute intermittent por
  • alkaptonuria Alexander disease; alkaptonuria; Alkaptonuric ochronosis (alkaptonuria); alpha-1 antitrypsin deficiency; alpha-1 proteinase inhibitor (alpha-1 antitrypsin deficiency); alpha-1 related emphysema (alpha-1 antitrypsin deficiency); Alpha-galactosidase A deficiency (Fabry disease); ALS (amyotrophic lateral sclerosis); Alstrom syndrome; ALX (Alexander disease);
  • Alzheimer disease Alzheimer disease; Amelogenesis Imperfecta; Amino levulinic acid dehydratase deficiency (ALA dehydratase deficiency); Aminoacylase 2 deficiency (Canavan disease); amyotrophic lateral sclerosis; Anderson-Fabry disease (Fabry disease); androgen insensitivity syndrome; Anemia; Anemia, hereditary sideroblastic (X-linked sideroblastic anemia); Anemia, sex-linked
  • hypochromic sideroblastic X-linked sideroblastic anemia
  • Anemia splenic, familial (Gaucher disease); Angelman syndrome; Angiokeratoma Corporis Diffusum (Fabry's disease);
  • Angiokeratoma diffuse Fabry's disease
  • Angiomatosis retinae von Hippel-Lindau disease
  • ANH1 X-linked sideroblastic anemia
  • Leiden type factor V Leiden
  • thrombophilia Apert syndrome; AR deficiency (androgen insensitivity syndrome); AR-CMT2 ee (Charcot-Mare-Tooth disease, type 2); Arachnodactyly (Marfan syndrome); ARNSHL (Nonsyndromic deafness autosomal recessive); Arthro-ophthalmopathy, hereditary progressive (Stickler syndrome COL2A1); Arthrochalasis multiplex congenita (Ehlers-Danlos syndrome arthrochalasia type); AS (Angelman syndrome); Asp deficiency (Canavan disease); Aspa deficiency (Canavan disease); Aspartoacylase deficiency (Canavan disease); ataxia-telangiectasia; Autism-Dementia-Ataxia-Loss of Purposeful Hand Use syndrome (Rett syndrome); autosomal dominant juvenile ALS (amyotrophic lateral sclerosis, type 4); Autosomal dominant opitz G
  • Nonsyndromic deafness autosomal recessive Autosomal Recessive Sensorineural Hearing Impairment and Goiter (Pendred syndrome); AxD (Alexander disease); Ayerza syndrome (primary pulmonary hypertension); B variant of the Hexosaminidase GM2 gangliosidosis (Sandhoff disease); BANF (neurofibromatosis 2); Beare-Stevenson cutis gyrata syndrome; Benign paroxysmal peritonitis (Mediterranean fever, familial); Benjamin syndrome; beta thalassemia; BH4 Deficiency (tetrahydrobiopterin deficiency); Bilateral Acoustic Neurofibromatosis
  • Neurofibromatosis 2 neurofibromatosis 2
  • biotinidase deficiency bladder cancer
  • Bleeding disorders factor V Leiden thrombophilia
  • Bloch-Sulzberger syndrome incontinentia pigmenti
  • Bloom syndrome Bone diseases; Bone marrow diseases (X-linked sideroblastic anemia); Bonnevie-Ullrich syndrome (Turner syndrome); Bourneville disease (tuberous sclerosis); Bourneville phakomatosis
  • Chondrogenesis imperfecta achondrogenesis, type II
  • Choreoathetosis self-mutilation hyperuricemia syndrome Lesch-Nyhan syndrome
  • Classic Galactosemia galactosemia
  • Phenylketonuria phenylketonuria
  • Cleft lip and palate Cloverleaf skull with thanatophoric dwarfism (Thanatophoric dysplasia type 2); CLS (Coffin-Lowry syndrome); CMT (Charcot- Marie-Tooth disease); Cockayne syndrome; Coffin-Lowry syndrome; collagenopathy, types II and XI; Colon Cancer, familial Nonpolyposis (hereditary nonpolyposis colorectal cancer); Colon cancer, familial (familial adenomatous polyposis); Colorectal Cancer; Complete HPRT deficiency (Lesch-Nyhan syndrome); Complete hypoxanthine-guanine phosphoribosy transferase deficiency (Lesch-Nyhan syndrome); Compression neuropathy (hereditary neuropathy with liability to pressure palsies); Congenital adrenal hyperplasia (21-hydroxylase deficiency);
  • congenital bilateral absence of vas deferens Congenital absence of the vas deferens
  • Congenital erythropoietic porphyria Congenital heart disease
  • Congenital hypomyelination (Charcot-Marie- Tooth disease Type 1/Charcot-Marie-Tooth disease Type 4); Congenital hypothyroidism;
  • methemoglobinemia (Methemoglobinemia Congenital methaemoglobinaemia);
  • Congenital osteosclerosis achondroplasia
  • Congenital sideroblastic anaemia X-linked sideroblastic anemia
  • Connective tissue disease Conotruncal anomaly face syndrome (22qll.2 deletion syndrome); Cooley's Anemia (beta thalassemia); Copper storage disease (Wilson disease); Copper transport disease (Menkes disease); Coproporphyria, hereditary (hereditary coproporphyria); Coproporphyrinogen oxidase deficiency (hereditary coproporphyria); Cowden syndrome; CPO deficiency (hereditary coproporphyria); CPRO deficiency (hereditary
  • coproporphyria coproporphyria
  • CPX deficiency hereeditary coproporphyria
  • Craniofacial dysarthrosis Crouzon syndrome
  • Craniofacial Dysostosis Crouzon syndrome
  • Cretinism congenital hypothyroidism
  • Creutzfeldt-Jakob disease prion disease
  • Cri du chat Crohn's disease, fibrostenosing
  • Crouzon syndrome Crouzon syndrome with acanthosis nigricans
  • Crouzonodermoskeletal syndrome Crouzonodermoskeletal syndrome
  • CS Clarkayne syndrome
  • Curschmann- Batten-Steinert syndrome myotonic dystrophy
  • cutis gyrata syndrome of Beare-Stevenson Beare-Stevenson cutis gyrata syndrome
  • Disorder Mutation Chromosome D-glycerate dehydrogenase deficiency (hyperoxaluria, primary); Dappled metaphysis syndrome
  • Dejerine-Sottas syndrome (Charcot-Marie-Tooth disease); Delta-aminolevulinate dehydratase deficiency porphyria (ALA dehydratase deficiency); Dementia (CADASIL);
  • Ehlers-Danlos syndrome dermatosparaxis type Dermatosparaxis
  • developmental disabilities dHMN (Amyotrophic lateral sclerosis type 4 ); DHMN-V (distal spinal muscular atrophy, type V); DHTR deficiency (androgen insensitivity syndrome); Diffuse Globoid Body Sclerosis (Krabbe disease); DiGeorge syndrome; Dihydrotestosterone receptor deficiency (androgen insensitivity syndrome); distal spinal muscular atrophy, type V; DM1 (Myotonic dystrophy typel); DM2 (Myotonic dystrophy type2); Down syndrome; DSMAV (distal spinal muscular atrophy, type V); DSN (Charcot-Marie-Tooth disease type 4); DSS (Charcot-Marie-Tooth
  • achondroplasia Dwarf, thanatophoric (thanatophoric dysplasia); Dwarfism; Dwarfism-retinal atrophy-deafness syndrome (Cockayne syndrome); dysmyelinogenic leukodystrophy (Alexander disease); Dystrophia myotonica (myotonic dystrophy); dystrophia retinae pigmentosa-dysostosis syndrome (Usher syndrome); Early-Onset familial alzheimer disease (EOFAD) (Alzheimer disease); EDS (Ehlers-Danlos syndrome); Ehlers-Danlos syndrome; Ekman-Lobstein disease (osteogenesis imperfecta); Entrapment neuropathy (hereditary neuropathy with liability to pressure palsies); Epiloia (tuberous sclerosis); EPP (erythropoietic protoporphyria);
  • Erythroblastic anemia (beta thalassemia); Erythrohepatic protoporphyria (erythropoietic protoporphyria); Erythroid 5-aminolevulinate synthetase deficiency (X-linked sideroblastic anemia); Erythropoietic porphyria (congenital erythropoietic porphyria); Erythropoietic protoporphyria; Erythropoietic uroporphyria (congenital erythropoietic porphyria); Eye cancer (retinoblastoma FA - Friedreich ataxia); Fabry disease; Facial injuries and disorders; Factor V Leiden thrombophilia; FALS (amyotrophic lateral sclerosis); familial acoustic neuroma
  • neurofibromatosis type II familial adenomatous polyposis
  • familial Alzheimer disease (FAD) Alzheimer disease
  • familial amyotrophic lateral sclerosis familial amyotrophic lateral sclerosis
  • familial dysautonomia familial fat-induced hypertriglyceridemia (lipoprotein lipase deficiency, familial); familial hemochromatosis (hemochromatosis); familial LPL deficiency (lipoprotein lipase deficiency, familial); familial nonpolyposis colon cancer (hereditary nonpolyposis colorectal cancer); familial paroxysmal polyserositis (Mediterranean fever, familial); familial PCT (porphyria cutanea tarda); familial pressure sensitive neuropathy (hereditary neuropathy with liability to pressure palsies); familial primary pulmonary hypertension (FPPH) (primary pulmonary hypertension); Familial Turner syndrome (Noonan syndrome); familial vascular leukoencephalopathy (CADASIL); FAP (familial adenomatous polyposis); FD (familial dysautonomia); Female pseudo-Turner syndrome (Noonan syndrome); Ferrochelatase defic
  • Galactokinase deficiency disease (galactosemia); Galactose- 1-phosphate uridyl-transferase deficiency disease (galactosemia); galactosemia; Galactosylceramidase deficiency disease (Krabbe disease); Galactosylceramide lipidosis (Krabbe disease); galactosylcerebrosidase deficiency (Krabbe disease); galactosylsphingosine lipidosis (Krabbe disease); GALC deficiency (Krabbe disease); GALT deficiency (galactosemia); Gaucher disease; Gaucher-like disease (pseudo- Gaucher disease); GBA deficiency (Gaucher disease type 1); GD (Gaucher's disease); Genetic brain disorders; genetic emphysema (alpha-1 antitrypsin deficiency); genetic hemochromatosis (hemochromatosis); Gi
  • Glucosylceramide lipidosis (Gaucher disease); Glyceric aciduria (hyperoxaluria, primary); Glycine encephalopathy (Nonketotic hyperglycinemia); Glycolic aciduria (hyperoxaluria, primary); GM2 gangliosidosis, type 1 (Tay-Sachs disease); Goiter-deafness syndrome (Pendred syndrome);
  • Graefe-Usher syndrome Usher syndrome
  • Gronblad-Strandberg syndrome pseudoxanthoma elasticum
  • Guenther porphyria congenital erythropoietic porphyria
  • Gunther disease Usher syndrome
  • Gronblad-Strandberg syndrome pseudoxanthoma elasticum
  • Guenther porphyria congenital erythropoietic porphyria
  • HEF2A hemochromatosis type 2
  • HEF2B hemochromatosis type 2
  • Hematoporphyria porphyria
  • erythropoietic protoporphyria Hemochromatoses (hemochromatosis); hemochromatosis; hemoglobin M disease (methemoglobinemia beta-globin type); Hemoglobin S disease (sickle cell anemia); hemophilia; HEP (hepatoerythropoietic porphyria); hepatic AGT deficiency
  • Hereditary spinal sclerosis (Friedreich ataxia); Herrick's anemia (sickle cell anemia);
  • OSMED Weissenbacher-Zweymuller syndrome
  • otospondylomegaepiphyseal dysplasia Weissenbacher-Zweymuller syndrome
  • HexA deficiency Tay-Sachs disease
  • Hexosaminidase A deficiency T ay-Sachs disease
  • Hexosaminidase alpha- subunit deficiency variant B
  • Tay-Sachs disease HFE-associated hemochromatosis
  • HGPS hemochromatosis
  • HGPS Progeria
  • Hippel-Lindau disease von Hippel-Lindau disease
  • HLAH hemochromatosis
  • HMN V distal spinal muscular atrophy, type V
  • HMSN Charge-Marie- Tooth disease
  • HNPCC hereditary nonpolyposis colorectal cancer
  • HNPP hereditary neuropathy with liability to pressure palsies
  • homocystinuria Homogentisic acid oxidase deficiency (alkaptonuria); Homogentisic acidura (alkaptonuria); Homozygous porphyria cutanea tarda (hepatoerythropoietic porphyria); HP1 (hyperoxaluria, primary); HP2 (hyperoxaluria, primary); HPA (hyperphenylalaninemia); HPRT - Hypoxanthine-guanine
  • HSAN type III familial dysautonomia
  • HSAN3 familial dysautonomia
  • HSN-III familial dysautonomia
  • Human dermatosparaxis Ehlers-Danlos syndrome dermatosparaxis type
  • Huntington's disease Huntington's disease
  • Hutchinson-Gilford progeria syndrome progeria
  • Hyperandrogenism nonclassic type, due to 21 -hydroxylase deficiency (21 -hydroxylase deficiency); Hyperchylomicronemia, familial (lipoprotein lipase deficiency, familial); hyperglycinemia with ketoacidosis and leukopenia (propionic acidemia); Hyperlipoproteinemia type I (lipoprotein lipase deficiency, familial); hyperoxaluria, primary; hyperphenylalaninaemia (hyperphenylalaninemia);
  • hypophenylalaninemia Hypochondrodysplasia (hypochondroplasia); hypochondrogenesis; hypochondroplasia; Hypochromic anemia (X-linked sideroblastic anemia); Hypocupremia, congenital; Menkes syndrome); hypoxanthine phosphoribosyltransferse (HPRT) deficiency (Lesch-Nyhan syndrome); IAHSP (infantile-onset ascending hereditary spastic paralysis);
  • idiopathic hemochromatosis hemochromatosis, type 3
  • Idiopathic neonatal hemochromatosis hemochromatosis, neonatal
  • Idiopathic pulmonary hypertension primary pulmonary hypertension
  • Immune system disorders X-linked severe combined immunodeficiency
  • Incontinentia Pigmenti Infantile cerebral Gaucher's disease (Gaucher disease type 2); Infantile Gaucher disease (Gaucher disease type 2); infantile-onset ascending hereditary spastic paralysis; Infertility; inherited emphysema (alpha-1 antitrypsin deficiency); Inherited human transmissible spongiform encephalopathies (prion disease); inherited tendency to pressure palsies (hereditary neuropathy with liability to pressure palsies); Insley-Astley syndrome
  • juvenile amyotrophic lateral sclerosis (Amyotrophic lateral sclerosis type 2); Juvenile gout, choreoathetosis, mental retardation syndrome (Lesch-Nyhan syndrome); juvenile hyperuricemia syndrome (Lesch-Nyhan syndrome); JWS (Jackson-Weiss syndrome); KD (X-linked spinal-bulbar muscle atrophy); Kennedy disease (X-linked spinal-bulbar muscle atrophy); Kennedy spinal and bulbar muscular atrophy (X-linked spinal-bulbar muscle atrophy); Kerasin histiocytosis (Gaucher disease); Kerasin lipoidosis (Gaucher disease); Kerasin thesaurismosis (Gaucher disease); ketotic glycinemia (propionic acidemia); ketotic hyperglycinemia (propionic acidemia); Kidney diseases (hyperoxaluria, primary); Klinefelter syndrome; Klinefelter's syndrome; Kniest dysplasia; Krabbe disease; Lacunar dementia (CADASIL);
  • Lentiginosis perioral (Peutz-Jeghers syndrome); Lesch-Nyhan syndrome; Leukodystrophies; leukodystrophy with Rosenthal fibers (Alexander disease); Leukodystrophy, spongiform
  • Canavan disease LFS (Li-Fraumeni syndrome); Li-Fraumeni syndrome; Lipase D deficiency (lipoprotein lipase deficiency, familial); LIPD deficiency (lipoprotein lipase deficiency, familial); Lipidosis, cerebroside (Gaucher disease); Lipidosis, ganglioside, infantile (Tay-Sachs disease); Lipoid histiocytosis (kerasin type) (Gaucher disease); lipoprotein lipase deficiency, familial; Liver diseases (galactosemia); Lou Gehrig disease (amyotrophic lateral sclerosis); Louis-Bar syndrome (ataxia-telangiectasia); Lynch syndrome (hereditary nonpolyposis colorectal cancer); Lysyl- hydroxylase deficiency (Ehlers-Danlos syndrome kyphoscoliosis type); Machado-Joseph disease (Spinocer
  • Neurodegeneration Neill-Dingwall syndrome (Cockayne syndrome); Neuroblastoma, retinal (retinoblastoma); Neurodegeneration with brain iron accumulation type 1 (pantothenate kinase- associated neurodegeneration); Neurofibromatosis type I; Neurofibromatosis type II; Neurologic diseases; Neuromuscular disorders; neuronopathy, distal hereditary motor, type V (Distal spinal muscular atrophy type V); neuronopathy, distal hereditary motor, with pyramidal features (Amyotrophic lateral sclerosis type 4); NF (neurofibromatosis); Niemann-Pick (Niemann-Pick disease); Noack syndrome (Pfeiffer syndrome); Nonketotic hyperglycinemia (Glycine
  • Non-neuronopathic Gaucher disease Gaucher disease type 1
  • Non-phenylketonuric hyperphenylalaninemia tetrahydrobiopterin deficiency
  • nonsyndromic deafness Noonan syndrome
  • Norrbottnian Gaucher disease Gaucher disease type 3
  • Ochronosis alkaptonuria
  • Ochronotic arthritis alkaptonuria
  • 01 osteogenesis imperfecta
  • OSMED otospondylomegaepiphyseal dysplasia
  • osteogenesis imperfecta Osteopsathyrosis
  • osteogenesis imperfecta osteogenesis imperfecta
  • Osteosclerosis congenita achondroplasia
  • Oto-spondylo- megaepiphyseal dysplasia otospondylomegaepiphyseal dysplasia
  • otospondylomegaepiphyseal dysplasia Oxalosis (hyperoxaluria, primary); Oxaluria, primary (hyperoxaluria, primary);
  • pantothenate kinase-associated neurodegeneration Patau Syndrome (Trisomy 13); PBGD deficiency (acute intermittent porphyria); PCC deficiency (propionic acidemia); PCT (porphyria cutanea tarda); PDM (Myotonic dystrophy type 2); Pendred syndrome; Periodic disease
  • hydroxylase deficiency disease phenylketonuria
  • phenylketonuria phenylketonuria
  • Pheochromocytoma von Hippel-Lindau disease
  • Pierre Robin syndrome with fetal chondrodysplasia Weissenbacher- Zweymuller syndrome
  • Pigmentary cirrhosis hemochromatosis
  • PJS Puleutz-Jeghers syndrome
  • PKAN pantothenate kinase-associated neurodegeneration
  • PKU phenylketonuria
  • Plumboporphyria (ALA deficiency porphyria); PMA (Charcot-Marie-tooth disease); polyostotic fibrous dysplasia (McCune-Alb right syndrome); polyposis coli (familial adenomatous polyposis); polyposis, hamartomatous intestinal (Peutz-Jeghers syndrome); polyposis, intestinal, II (Peutz- Jeghers syndrome); polyps-and-spots syndrome (Peutz-Jeghers syndrome); Porphobilinogen synthase deficiency (ALA deficiency porphyria); porphyria; porphyrin disorder (porphyria); PPH (primary pulmonary hypertension); PPOX deficiency (variegate porphyria); Prader-Labhart- Willi syndrome (Prader-Willi syndrome); Prader-Willi syndrome; presenile and senile dementia (Alzheimer disease); primary hemochromatosis (hem
  • protoporphyrinogen oxidase deficiency variant porphyria
  • proximal myotonic dystrophy Myotonic dystrophy type 2
  • proximal myotonic myopathy Myotonic dystrophy type 2
  • pseudo- Gaucher disease pseudo-Ullrich-Turner syndrome (Noonan syndrome); pseudoxanthoma elasticum; psychosine lipidosis (Krabbe disease); pulmonary arterial hypertension (primary pulmonary hypertension); pulmonary hypertension (primary pulmonary hypertension); PWS (Prader-Willi syndrome); PXE - pseudoxanthoma elasticum (pseudoxanthoma elasticum); Rb (retinoblastoma); Recklinghausen disease, nerve (neurofibromatosis 1); Recurrent polyserositis (Mediterranean fever, familial); Retinal disorders; Retinitis pigmentosa-deafness syndrome (Usher syndrome); Retinoblasto
  • Roussy-Levy syndrome (Charcot-Marie-Tooth disease); RSTS (Rubinstein-Taybi syndrome); RTS (Rett syndrome) (Rubinstein-Taybi syndrome); RTT (Rett syndrome);
  • Rubinstein-Taybi syndrome Sack-Barabas syndrome (Ehlers-Danlos syndrome, vascular type); SADDAN; sarcoma family syndrome of Li and Fraumeni (Li-Fraumeni syndrome); sarcoma, breast, leukemia, and adrenal gland (SBLA) syndrome (Li-Fraumeni syndrome); SBLA syndrome (Li-Fraumeni syndrome); SBMA (X-linked spinal-bulbar muscle atrophy); SCD (sickle cell anemia); Schwannoma, acoustic, bilateral (neurofibromatosis 2); SCIDX1 (X-linked severe combined immunodeficiency); sclerosis tuberosa (tuberous sclerosis); SDAT (Alzheimer disease); SED congenita (spondyloepiphyseal dysplasia congenita); SED Strudwick
  • SEDc pondyloepiphyseal dysplasia congenita
  • SEMD Strudwick type (spondyloepimetaphyseal dysplasia, Strudwick type); senile dementia (Alzheimer disease type 2); severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN); Shprintzen syndrome (22qll.2 deletion syndrome); sickle cell anemia; skeleton-skin-brain syndrome (SADDAN); Skin pigmentation disorders; SMA (spinal muscular atrophy); SMED, Strudwick type (spondyloepimetaphyseal dysplasia, Strudwick type); SMED, type I (spondyloepimetaphyseal dysplasia, Strudwick type); Smith Lemli Opitz Syndrome; South-African genetic porphyria (vari
  • nucleic acid sequence as defined herein or the inventive composition comprising a plurality of nucleic acid sequences as defined herein may be used for the preparation of a pharmaceutical composition, particularly for purposes as defined herein, preferably for the use in gene therapy in the treatment of diseases as defined herein.
  • inventive pharmaceutical composition may furthermore be used in gene therapy particularly in the treatment of a disease or a disorder, preferably as defined herein.
  • inventive pharmaceutical composition furthermore provides several applications and uses of the inventive
  • the composition or the pharmaceutical composition or the kit or kit of parts may be used as a medicament, namely for treatment of tumor or cancer diseases.
  • the treatment is preferably done by intratumoral application, especially by injection into tumor tissue.
  • the present invention is directed to the second medical use of the RNA containing composition or the pharmaceutical composition, or the vaccine, or the kit or kit of parts as described above, wherein these subject matters are used for preparation of a medicament particularly for intratumoral application (administration) for treatment of tumor or cancer diseases.
  • diseases as mentioned herein are selected from tumor or cancer diseases which preferably include e.g.
  • Acute lymphoblastic leukemia Acute myeloid leukemia, Adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, Anal cancer, Appendix cancer, Astrocytoma, Basal cell carcinoma, Bile duct cancer, Bladder cancer, Bone cancer,
  • Osteosarcoma/Malignant fibrous histiocytoma Brainstem glioma, Brain tumor, cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic glioma, Breast cancer, Bronchial adenomas/carcinoids, Burkitt lymphoma, childhood Carcinoid tumor, gastrointestinal Carcinoid tumor, Carcinoma of unknown primary, primary Central nervous system lymphoma, childhood Cerebellar astrocytoma, childhood Cerebral astrocytoma/Malignant glioma, Cervical cancer, Childhood cancers, Chronic lymphocytic leukemia, Chronic myelogenous leukemia, Chronic myeloproliferative disorders, Colon Cancer, Cutaneous T-cell lymphoma, Desmoplastic small
  • Extragonadal Germ cell tumor Extrahepatic bile duct cancer, Intraocular melanoma,
  • Retinoblastoma Gallbladder cancer, Gastric (Stomach) cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal stromal tumor (GIST), extracranial, extragonadal, or ovarian Germ cell tumor, Gestational trophoblastic tumor, Glioma of the brain stem, Childhood Cerebral Astrocytoma, Childhood Visual Pathway and Hypothalamic Glioma, Gastric carcinoid, Hairy cell leukemia, Head and neck cancer, Heart cancer, Hepatocellular (liver) cancer, Hodgkin lymphoma,
  • hypopharyngeal cancer childhood Hypothalamic and visual pathway glioma, Intraocular Melanoma, Islet Cell Carcinoma (Endocrine Pancreas), Kaposi sarcoma, Kidney cancer (renal cell cancer), Laryngeal Cancer, Leukemias, acute lymphoblastic Leukemia, acute myeloid Leukemia, chronic lymphocytic Leukemia, chronic myelogenous Leukemia, hairy cell Leukemia, Lip and Oral Cavity Cancer, Liposarcoma, Liver Cancer, Non-Small Cell Lung Cancer, Small Cell Lung Cancer,
  • Lymphomas AIDS-related Lymphoma, Burkitt Lymphoma, cutaneous T-Cell Lymphoma, Hodgkin Lymphoma, Non-Hodgkin Lymphomas, Primary Central Nervous System Lymphoma,
  • Myelodysplastic/Myeloproliferative Diseases Chronic Myelogenous Leukemia, Adult Acute Myeloid Leukemia, Childhood Acute Myeloid Leukemia, Multiple Myeloma (Cancer of the Bone- Marrow), Chronic Myeloproliferative Disorders, Nasal cavity and paranasal sinus cancer, Nasopharyngeal carcinoma, Neuroblastoma, Oral Cancer, Oropharyngeal cancer,
  • Osteosarcoma/malignant fibrous histiocytoma of bone Ovarian cancer, Ovarian epithelial cancer (Surface epithelial-stromal tumor), Ovarian germ cell tumor, Ovarian low malignant potential tumor, Pancreatic cancer, islet cell Pancreatic cancer, Paranasal sinus and nasal cavity cancer, Parathyroid cancer, Penile cancer, Pharyngeal cancer, Pheochromocytoma, Pineal astrocytoma, Pineal germinoma, childhood Pineoblastoma and supratentorial primitive neuroectodermal tumors, Pituitary adenoma, Plasma cell neoplasia/Multiple myeloma, Pleuropulmonary blastoma, Primary central nervous system lymphoma, Prostate cancer, Rectal cancer, Renal cell carcinoma (kidney cancer), Cancer of the Renal pelvis and ureter, Retinoblastoma, childhood
  • Sarcoma soft tissue Sarcoma, uterine Sarcoma, Sezary syndrome, Skin cancer (nonmelanoma), Skin cancer (melanoma), Merkel cell Skin carcinoma, Small intestine cancer, Squamous cell carcinoma, metastatic Squamous neck cancer with occult primary, childhood Supratentorial primitive neuroectodermal tumor, Testicular cancer, Throat cancer, childhood Thymoma, Thymoma and Thymic carcinoma, Thyroid cancer, childhood Thyroid cancer, Transitional cell cancer of the renal pelvis and ureter, gestational Trophoblastic tumor, Urethral cancer, endometrial Uterine cancer, Uterine sarcoma, Vaginal cancer, childhood Visual pathway and hypothalamic glioma, Vulvar cancer, Waldenstrom macroglobulinemia, and childhood Wilms tumor (kidney cancer).
  • the medicament may be administered to the patient as a single dose or as several doses.
  • the medicament may be administered to a patient as a single dose followed by a second dose later and optionally even a third, fourth (or more) dose subsequent thereto et cetera.
  • the inventive composition is provided in an amount of at least 40 ⁇ g RNA per dose. More specifically, the amount of the mRNA comprised in a single dose is typically at least 200 ⁇ g, preferably from 200 ⁇ g to 1.000 ⁇ g, more preferably from 300 ⁇ g to 850 ⁇ g, even more preferably from 300 ⁇ g to 700 ⁇ g. In the case of intradermal injection, which is preferably carried out via jet injection (e.g. using a Tropis device), the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 ⁇ g, preferably from 80 ⁇ g to 700 ⁇ g, more preferably from 80 ⁇ g to 400 ⁇ g.
  • the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 ⁇ g, preferably from 80 ⁇ g to 1.000 ⁇ g, more preferably from 80 ⁇ g to 850 ⁇ g, even more preferably from 80 ⁇ g to 700 ⁇ g.
  • the immunization protocol for the treatment or prophylaxis of e.g. a virus infection typically comprises a series of single doses or dosages of the inventive composition or the inventive vaccine.
  • a single dosage refers to the initial/first dose, a second dose or any further doses, respectively, which are preferably administered in order to "boost" the immune reaction.
  • the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts is provided for use in treatment or prophylaxis, preferably treatment or prophylaxis of e.g. a virus infection or a related disorder or disease, wherein the treatment or prophylaxis comprises the administration of a further active pharmaceutical ingredient. More preferably, in the case of the inventive vaccine or composition, which is based on the inventive artificial nucleic acid, a polypeptide may be coadministered as a further active pharmaceutical ingredient. For example, at least one e.g.
  • virus protein or peptide as described herein, or a fragment or variant thereof may be co-administered in order to induce or enhance an immune response.
  • an artificial nucleic acid as described herein may be co-administered as a further active pharmaceutical ingredient.
  • an artificial nucleic acid as described herein encoding at least one polypeptide as described herein may be co-administered in order to induce or enhance an immune response.
  • a further component of the inventive vaccine or composition may be an
  • immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably
  • immunoglobulins directed against e.g. a virus may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA.
  • a further immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the inventive artificial nucleic acid or by the inventive polypeptide.
  • the invention provides a method of treating or preventing a disorder, wherein the disorder is preferably an infection with e.g. a virus or a disorder related to an infection with e.g. a virus, wherein the method comprises administering to a subject in need thereof the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts.
  • the inventive composition comprising at least one inventive nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts.
  • such a method may preferably comprise the steps of: a) providing the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts; b) applying or administering the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts to a tissue or an organism; c) optionally administering immunoglobuline (IgGs) against e.g. the virus.
  • IgGs immunoglobuline
  • the present invention also provides a method for expression of at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, wherein the method preferably comprises the following steps: a) providing the inventive artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, preferably as defined herein, or a composition comprising said artificial nucleic acid; and b) applying or administering the inventive artificial nucleic acid or the inventive composition comprising said artificial nucleic acid to an expression system, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • an expression system e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • the method may be applied for laboratory, for research, for diagnostic, for commercial production of peptides or proteins and/or for therapeutic purposes.
  • inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, e.g. in naked or complexed form or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • the method may be carried out in vitro, in vivo or ex vivo.
  • the method may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of infectious diseases, or a related disorder.
  • the present invention also provides the use of the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, preferably for diagnostic or therapeutic purposes, for expression of e.g. an encoded virus antigenic peptide or protein, e.g. by applying or administering the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
  • the use may be applied for a (diagnostic) laboratory, for research, for diagnostics, for commercial production of peptides or proteins and/or for therapeutic purposes.
  • inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, preferably in naked form or complexed form, or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • a cell e.g. an expression host cell or a somatic cell
  • tissue or an organism preferably in naked form or complexed form
  • a composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein.
  • the use may be carried out in vitro, in vivo or ex vivo.
  • the use may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of e.g. a virus infection or a related disorder.
  • the invention provides the artificial nucleic acid, the inventive composition or the inventive vaccine for use as defined herein, preferably for use as a medicament, for use in treatment or prophylaxis, preferably treatment or prophylaxis of a e.g. a virus infection or a related disorder, or for use as a vaccine.
  • the nucleotide acid molecule of the inventive composition encodes at least one epitope of at least one antigen.
  • the at least one antigen is selected from the group consisting of an antigen from a pathogen associated with infectious diseases, an antigen associated with allergies, an antigen associated with autoimmune diseases, and an antigen associated with cancer or tumor diseases, or a fragment, variant and/or derivative of said antigen.
  • the at least one antigen is derived from a pathogen, preferably a viral, bacterial, fungal or protozoan pathogen, preferably selected from the list consisting of: Rabies virus, Ebolavirus, Marburgvirus, Hepatitis B virus, human Papilloma virus (hPV), Bacillus anthracis, Respiratory syncytial virus (RSV), Herpes simplex virus (HSV), Dengue virus, Rotavirus, Influenza virus, human immunodeficiency virus (HIV), Yellow Fever virus, Mycobacterium tuberculosis, Plasmodium, Staphylococcus aureus, Chlamydia trachomatis, Cytomegalovirus (CMV) and
  • a pathogen preferably a viral, bacterial, fungal or protozoan pathogen, preferably selected from the list consisting of: Rabies virus, Ebolavirus, Marburgvirus, Hepatitis B virus, human Papilloma virus (hPV), Bac
  • the mRNA of the inventive composition may encode for a protein or a peptide, which comprises at least one epitope of a pathogenic antigen or a fragment, variant or derivative thereof.
  • pathogenic antigens are derived from pathogenic organisms, in particular bacterial, viral or protozoological (multicellular) pathogenic organisms, which evoke an immunological reaction by subject, in particular a mammalian subject, more particularly a human. More specifically, pathogenic antigens are preferably surface antigens, e.g. proteins (or fragments of proteins, e.g. the exterior portion of a surface antigen) located at the surface of the virus or the bacterial or protozoological organism.
  • Pathogenic antigens are peptide or protein antigens preferably derived from a pathogen associated with infectious disease which are preferably selected from antigens derived from the pathogens Acinetobacter baumannii, Anaplasma genus, Anaplasma phagocytophilum, Ancylostoma braziliense, Ancylostoma duodenale, Arcanobacte um haemolyticum, Asca s lumb coides, Aspergillus genus, Astroviridae, Babesia genus, Bacillus anthracis, Bacillus cereus, Bartonella henselae, BK virus, Blastocystis hominis, Blastomyces dermatitidis, Bordetella pertussis, Borrelia burgdorferi, Borrelia genus, Borrelia spp, Brucella genus, Brugia malayi, Bunyaviridae family, Burkholderia cepacia and
  • Cryptosporidium genus Cytomegalovirus (CMV), Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN-4), Dientamoeba fragilis, Ebolavirus (EBOVJ, Echinococcus genus, Ehrlichia chaffeensis, Ehrlichia ewingii, Ehrlichia genus, Entamoeba histolytica, Enterococcus genus, Enterovirus genus, Enteroviruses, mainly Coxsackie A virus and Enterovirus 71 (EV71), Epidermophyton spp,
  • Epstein-Barr Virus EBV
  • Escherichia coli 0157:H7 Escherichia coli 0157:H7
  • Olll and O104:H4 Fasciola hepatica and Fasciola gigantica
  • FFI prion Fasciola hepatica
  • Filarioidea superfamily Flaviviruses
  • Francisella tularensis
  • Hepatitis D Virus Hepatitis E Virus
  • Herpes simplex virus 1 and 2 Herpes simplex virus 1 and 2 (HSV-1 and HSV-2), Histoplasma capsulatum, HIV (Human immunodeficiency virus), Hortaea wasneckii, Human bocavirus (HBoV), Human herpesvirus 6 (HHV-6) and Human herpesvirus 7 (HHV-7), Human metapneumovirus (hMPV), Human papillomavirus (HPV), Human parainfluenza viruses (HPIV), Japanese
  • HBV Human bocavirus
  • HHV-6 Human herpesvirus 6
  • HHV-7 Human metapneumovirus
  • HPV Human papillomavirus
  • HPIV Human parainfluenza viruses
  • encephalitis virus JC virus, Junin virus, Kingella kingae, Klebsiella granulomatis, Kuru prion, Lassa virus, Legionella pneumophila, Leishmania genus, Leptospira genus, Listeria monocytogenes, Lymphocytic choriomeningitis virus (LCMV), Machupo virus, Malassezia spp, Marburg virus, Measles virus, Metagonimus yokagawai, Microspo dia phylum, Molluscum contagiosum virus (MCV), Mumps virus, Mycobacterium leprae and Mycobacterium lepromatosis, Mycobacterium tuberculosis, Mycobacterium ulcerans, Mycoplasma pneumoniae, Naegleria fowleri, Necator americanus, Neisseria gonorrhoeae, Neisseria meningitidis, Nocardia asteroides, Nocardia spp, Oncho
  • the pathogenic antigen may be preferably selected from the following antigens: Outer membrane protein A OmpA, biofilm associated protein Bap, transport protein MucK (Acinetobacter baumannii, Acinetobacter infections)); variable surface glycoprotein VSG, microtubule-associated protein MAPP15, trans-sialidase TSA (Trypanosoma brucei, African sleeping sickness (African trypanosomiasis)); HIV p24 antigen, HIV envelope proteins (Gpl20, Gp41, Gpl60), polyprotein GAG, negative factor protein Nef, trans-activator of transcription Tat (HIV (Human
  • envelope glycoproteins (gB, gC, gE, gH, gl, gK, gL) (Varicella zoster virus (VZV), Chickenpox); major outer membrane protein MOMP, probable outer membrane protein PMPC, outer membrane complex protein B OmcB, heat shock proteins Hsp60 HSP10, protein IncA, proteins from the type III secretion system, ribonucleotide reductase small chain protein NrdB, plasmid protein Pgp3, chlamydial outer protein N CopN, antigen CT521, antigen CT425, antigen CT043, antigen TC0052, antigen TC0189, antigen TC0582, antigen TC0660, antigen TC0726, antigen TC0816, antigen TC0828 [Chlamydia trachomatis, Chlamydia); low calcium response protein E LCrE, chlamydial outer protein N CopN, serine
  • CP40 surface protein CP60, surface protein CP15, surface-associated glycopeptides gp40, surface- associated glycopeptides gpl5, oocyst wall protein AB, profilin PRF, apyrase (Cryptosporidium genus, Cryptosporidiosis); fatty acid and retinol binding protein-1 FAR-1, tissue inhibitor of metalloproteinase TIMP (TMP), cysteine proteinase ACEY-1, cysteine proteinase ACCP-1, surface antigen Ac-16, secreted protein 2 ASP-2, metalloprotease 1 MTP-1, aspartyl protease inhibitor API-1, surface-associated antigen SAA-1, adult-specific secreted factor Xa serine protease inhibitor anticoagulant AP, cathepsin D-like aspartic protease ARR-1 (usually Ancylostoma braziliense; multiple other parasites, Cutaneous larva migrans (CLM)); cathe
  • dehydrogenase A LDHA, lactate dehydrogenase B LDHB (Taenia solium, Cysticercosis); pp65 antigen, membrane protein ppl5, capsid-proximal tegument protein ppl50, protein M45, DNA polymerase UL54, helicase UL105, glycoprotein gM, glycoprotein gN, glcoprotein H, glycoprotein B gB, protein UL83, protein UL94, protein UL99 (Cytomegalovirus (CMV), Cytomegalovirus infection); capsid protein C, premembrane protein prM, membrane protein M, envelope protein E (domain I, domain II, domain II), protein NS1, protein NS2A, protein NS2B, protein NS3, protein NS4A, protein 2K, protein NS4B, protein NS5 (Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN- 4)-Flaviviruses, Dengue fever); 39
  • diphtheria toxin precursor Tox diphteria toxin precursor Tox
  • diphteria toxin DT pilin-specific sortase SrtA
  • shaft pilin protein SpaA shaft pilin protein SpaA
  • tip pilin protein SpaC minor pilin protein SpaB
  • allergen Tri r 2 heat shock protein 60 Hsp60, fungal actin Act
  • antigen Tri r2 antigen Tri r4, antigen Tri tl, protein IV, glycerol-3-phosphate dehydrogenase Gpdl, osmosensor HwSholA, osmosensor HwSholB, histidine kinase HwHhk7B
  • infectiosum (Fifth disease)); pp65 antigen, glycoprotein 105, major capsid protein, envelope glycoprotein H, protein U51 (Human herpesvirus 6 (HHV-6) and Human herpesvirus 7 (HHV-7), Exanthem subitum); thioredoxin-glutathione reductase TGR, cathepsins LI and L2, Kunitz-type protein KTM, leucine aminopeptidase LAP, cysteine proteinase Fas2, saposin-like protein-2 SAP- 2, thioredoxin peroxidases TPx, Prx-1, Prx-2, cathepsin 1 cysteine proteinase CL3, protease cathepsin L CL1, phosphoglycerate kinase PGK, 27-kDa secretory protein, 60 kDa protein
  • HSP35alpha glutathione transferase GST, 28.5 kDa tegumental antigen 28.5 kDa TA, cathepsin B3 protease CatB3, Type I cystatin stefin-1, cathepsin L5, cathepsin Llg and cathepsin B, fatty acid binding protein FABP, leucine aminopeptidases LAP (Fasciola hepatica and Fasciola gigantica,
  • Fasciolosis prion protein (FFI prion, Fatal familial insomnia (FFI)); venom allergen homolog-like protein VAL-1, abundant larval transcript ALT-1, abundant larval transcript ALT-2, thioredoxin peroxidase TPX, vespid allergen homologue VAH, thiordoxin peroxidase 2 TPX-2, antigenic protein SXP (peptides N, Nl, N2, and N3), activation associated protein- 1 ASP-1, Thioredoxin TRX, transglutaminase BmTGA, glutathione-S-transferases GST, myosin, vespid allergen homologue VAH, 175 kDa collagenase, glyceraldehyde-3-phosphate dehydrogenase GAPDH, cuticular collagen Col-4, secreted larval acidic proteins SLAPs, chitinase CHI-1, maltose binding protein MBP, glyco
  • leukotoxin lktA leukotoxin lktA
  • adhesion FadA outer membrane protein RadD
  • high-molecular weight arginine-binding protein Fusobacterium genus, Fusobacterium infection
  • phospholipase C PLC heat-labile enterotoxin B
  • Iota toxin component lb protein CPE1281
  • pyruvate ferredoxin oxidoreductase elongation factor G EF-G
  • perfringolysin O Pfo glyceraldehyde-3-phosphate dehydrogenase GapC
  • fructose-bisphosphate aldolase Alf2 Clostridium perfringens enterotoxin
  • CPE alpha toxin AT, alpha toxoid ATd, epsilon-toxoid ETd, protein HP, large cytotoxin TpeL, endo- beta-N-acetylglucosaminidase Naglu, phosphoglyceromutase Pgm (usually Clostridium
  • fibronectin/fibrinogen-binding protein FBP54 fibronectin-binding protein FbaA, M protein type 1 Emml, M protein type 6 Emm6, immunoglobulin-binding protein 35 Sib35, Surface protein R28 Spr28, superoxide dismutase SOD, C5a peptidase ScpA, antigen I/I I Agl/II, adhesin AspA, G- related alpha2-macroglobulin-binding protein GRAB, surface fibrillar protein M5 (Streptococcus pyogenes, Group A streptococcal infection); C protein ⁇ antigen, arginine deiminase proteins, adhesin BibA, 105 kDA protein BPS, surface antigens c, surface antigens R, surface antigens X, trypsin-resistant protein Rl, trypsin-resistant protein R3, trypsin-resistant protein R4, surface immunogenic protein Sip, surface protein Rib
  • RNA polymerase L protein L, glycoprotein Gn, glycoprotein Gc, nucleocapsid protein S, envelope glycoprotein Gl, nucleoprotein NP, protein N, polyprotein M (Sin Nombre virus, Hantavirus, Hantavirus Pulmonary Syndrome (HPS)); heat shock protein HspA, heat shock protein HspB, citrate synthase GltA, protein UreB, heat shock protein Hsp60, neutrophil-activating protein NAP, catalase KatA, vacuolating cytotoxin VacA, urease alpha UreA, urease beta U
  • Helicobacter pylori infection integral membrane proteins, aggregation-prone proteins, O- antigen, toxin-antigens Stx2B, toxin-antigen StxlB, adhesion-antigen fragment Int28, protein EspA, protein EspB, Intimin, protein Tir, protein IntC300, protein Eae (Escherichia coli 0157:H7, Olll and O104:H4, Hemolytic-uremic syndrome (HUS)); RNA polymerase L, protein L, glycoprotein Gn, glycoprotein Gc, nucleocapsid protein S, envelope glycoprotein Gl,
  • nucleoprotein NP protein N
  • polyprotein M Bounyaviridae family, Hemorrhagic fever with renal syndrome (HFRS)
  • glycoprotein G matrix protein M
  • nucleoprotein N fusion protein F
  • polymerase L protein W
  • proteinC proteinC
  • phosphoprotein p non-structural protein V
  • polyprotein glycoproten Gp2, hepatitis A surface antigen HBAg, protein 2A, virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4, protein PIB, protein P2A, protein P3AB, protein P3D (Hepatitis A Virus, Hepatitis A); hepatitis B surface antigen HBsAg, Hepatitis B core antigen HbcAg, polymerase, protein Hbx, preS2 middle surface protein, surface protein L, large S protein, virus protein VP1, virus protein VP2, virus protein VP3, virus protein
  • transcriptional regulation protein IE63 (ICP27, UL54), protein UL55, protein UL56, viral replication protein ICP22 (IE68, US1), protein US2, serine/threonine-protein kinase US3, glycoprotein G (US4), glycoprotein J (US5), glycoprotein D (US6), glycoprotein I (US7), glycoprotein E (US8), tegument protein US9, capsid/tegument protein US10, Vmw21 protein (US11), ICP47 protein (IE12, US12), major transcriptional activator ICP4 (IE175, RSI), E3 ubiquitin ligase ICP0 (IE110), latency-related protein 1 LRP1, latency-related protein 2 LRP2, neurovirulence factor RL1 (ICP34.5), latency-associated transcript LAT (Herpes simplex virus 1 and 2 (HSV-1 and HSV-2), Herpes simplex); heat shock protein Hsp60, cell surface protein H1C, dipeptidyl peptidas
  • fatty acid and retinol binding protein- 1 FAR-1 tissue inhibitor of metalloproteinase TIMP (TMP), cysteine proteinase ACEY-1, cysteine proteinase ACCP-1, surface antigen Ac-16, secreted protein 2 ASP-2, metalloprotease 1 MTP-1, aspartyl protease inhibitor API-1, surface-associated antigen SAA-1, surface-associated antigen SAA-2, adult-specific secreted factor Xa, serine protease inhibitor anticoagulant AP, cathepsin D-like aspartic protease ARR-1, glutathione S-transferase GST, aspartic protease APR-1,
  • acetylcholinesterase AChE (Ancylostoma duodenale and Necator americanus, Hookworm infection); protein NS1, protein NP1, protein VP1, protein VP2, protein VP3 (Human bocavirus (HBoV), Human bocavirus infection); major surface protein 2 MSP2, major surface protein 4 MSP4, MSP variant SGV1, MSP variant SGV2, outer membrane protein OMP, outer membrande protein 19 OMP-19, major antigenic protein MAPI, major antigenic protein MAP1-2, major antigenic protein MAP1B, major antigenic protein MAPl-3, Erum2510 coding protein, protein GroEL, protein GroES, 30-kDA major outer membrane proteins, GE 100-kDa protein, GE 130-kDa protein, GE 160-kDa protein (Ehrlichia ewingii, Human ewingii ehrlichiosis); major surface proteins 1-5 (MSPla, MSPlb, MSP2, MSP3, MSP4, MSP5), type
  • outer membrane protein LipL32 outer membrane protein LipL32, membrane protein LIC10258, membrane protein LP30, membrane protein LIC12238, Ompa-like protein Lsa66, surface protein LigA, surface protein LigB, major outer membrane protein OmpLl, outer membrane protein LipL41, protein LigAni, surface protein LcpA, adhesion protein LipL53, outer membrane protein UpL32, surface protein Lsa63, flagellin FlaBl, membran lipoprotein LipL21, membrane protein pL40, leptospiral surface adhesin Lsa27, outer membrane protein OmpL36, outer membrane protein OmpL37, outer membrane protein OmpL47, outer membrane protein OmpL54, acyltransferase LpxA (Leptospira genus, Leptospirosis); listeriolysin O precursor Hly (LLO), invasion-associated protein lap (P60), Listeriolysin regulatory protein PrfA, Zinc
  • metalloproteinase Mpl Phosphatidylinositol- specific phospholipase C PLC (PlcA, PlcB), O- acetyltransferase Oat, ABC-transporter permease Im.G_1771, adhesion protein LAP, LAP receptor Hsp60, adhesin LapB, haemolysin listeriolysin O LLO, protein ActA, Internalin A InlA, protein InlB (Listeria monocytogenes, Listeriosis); outer surface protein A OspA, outer surface protein OspB, outer surface protein OspC, decorin binding protein A DbpA, decorin binding protein B DbpB, flagellar filament 41 kDa core protein Fla, basic membrane protein A BmpA (Immunodominant antigen P39), outer surface 22 kDa lipoprotein precursor (antigen IPLA7), variable surface lipoprotein vlsE (usually Borrelia burgdorferi
  • Pasteurella multocida toxin PMT adhesive protein Cp39 (Pasteurella genus, Pasteurellosis); "filamentous hemagglutinin FhaB, adenylate cyclase CyaA, pertussis toxin subunit 4 precursor PtxD, pertactin precursor Prn, toxin subunit 1 PtxA, protein Cpn60, protein brkA, pertussis toxin subunit 2 precursor PtxB, pertussis toxin subunit 3 precursor PtxC, pertussis toxin subunit 5 precursor PtxE, pertactin Prn, protein Fim2, protein Fim3; " (Bordetella pertussis, Pertussis (Whooping cough)); "Fl capsule antigen, virulence-associated V antigen, secreted effector protein LcrV, V antigen, outer membrane protease Pla,secreted effector protein YopD, putative secreted protein-t
  • dehydrogenase Gnd iron-binding protein PiaA
  • Murein hydrolase LytB proteon LytC
  • protease Al Streptococcus pneumoniae, Pneumococcal infection
  • major surface protein B kexin-like protease KEX1, protein A12, 55 kDa antigen P55, major surface glycoprotein Msg (Pneumocystis jirovecii, Pneumocystis pneumonia (PCP)); genome polyprotein, polymerase 3D, viral capsid protein VP1, viral capsid protein VP2, viral capsid protein VP3, viral capsid protein VP4, protease 2A, protease 3C (Poliovirus, Poliomyelitis); protein Nfal, exendin-3, secretory lipase, cathepsin B- like protease, cysteine protease, cathepsin, peroxiredoxin, protein CrylAc (usually Nae
  • intracytoplasmic protein D (Rickettsia akari, Rickettsialpox); envelope glycoprotein GP, polymerase L, nucleoprotein N, non-structural protein NSS (Rift Valley fever virus, Rift Valley fever (RVF)); outer membrane proteins OM, cell surface antigen OmpA, cell surface antigen OmpB (sca5), cell surface protein SCA4, cell surface protein SCAl, intracytoplasmic protein D (Rickettsia rickettsii, Rocky mountain spotted fever (RMSF)); "non-structural protein 6 NS6, non-structural protein 2 NS2, intermediate capsid protein VP6, inner capsid protein VP2, non-structural protein 3 NS3, RNA-directed RNA polymerase L, protein VP3, non-structural protein 1 NS1, nonstructural protein 5 NS5, outer capsid glycoprotein VP7, non-structural glycoprotein 4 NS4, outer capsid protein VP4" (Rotavirus, Rotavirus
  • nucleoprotein N non-structural protein NS3b, non-structural protein NS6, protein 7a, nonstructural protein NS8b, membrane protein M, envelope small membrane protein EsM, replicase polyprotein la, spike glycoprotein S, replicase polyprotein lab; (SARS coronavirus, SARS (Severe Acute Respiratory Syndrome)); serin protease, Atypical Sarcoptes Antigen 1 ASA1, glutathione S- transferases GST, cystein protease, serine protease, apolipoprotein (Sarcoptes scabiei, Scabies); glutathione S-transferases GST, paramyosin, hemoglbinase SM32, major egg antigen, 14 kDa fatty acid-binding protein Sml4, major larval surface antigen P37, 22,6 kDa tegumental antigen, calpain CANP, triphospate isomerase Tim, surface protein 9B, outer capsid protein VP
  • antigen Ss-IR antigen Ss-IR
  • antigen NIE strongylastacin
  • Na+-K+ ATPase Sseat-6 tropomysin SsTmy-1, protein LEC-5, 41 kDa aantigen P5, 41-kDa larval protein, 31-kDa larval protein, 28-kDa larval protein (Strongyloides stercoralis, Strongyloidiasis); glycerophosphodiester phosphodiesterase GlpQ (Gpd), outer membrane protein TmpB, protein Tp92, antigen TpFl, repeat protein Tpr, repeat protein F TprF, repeat protein G TprG, repeat protein I Tprl, repeat protein J TprJ, repeat protein K TprK, treponemal membrane protein A TmpA, lipoprotein, 15 kDa Tppl5, 47 kDa membrane antigen, miniferritin TpFl, adhesin Tp
  • SRl surface antigen P22, major antigen p24, major surface antigen p30, dense granule proteins (GRA1, GRA2, GRA3, GRA4, GRA5, GRA6, GRA7, GRA8, GRA9, GRA10), 28 kDa antigen, surface antigen SAG1, SAG2 related antigen, nucleoside-triphosphatase 1, nucleoside-triphosphatase 2, protein Stt3, HesB-like domain-containing protein, rhomboid-like protease 5, toxomepsin 1 (Toxoplasma gondii, Toxoplasmosis); 43 kDa secreted glycoprotein, 53 kDa secreted
  • Mtb72F protein Apa, immunogenic protein MPT63, periplasmic phosphate-binding lipoprotein PSTS1 (PBP-1), molecular chaperone DnaK, cell surface lipoprotein Mpt83, lipoprotein P23, phosphate transport system permease protein pstA, 14 kDa antigen, fibronectin-binding protein C FbpCl, Alanine dehydrogenase TB43, Glutamine synthetase 1, ESX-1 protein, protein CFP10, TB10.4 protein, protein MPT83, protein MTB12, protein MTB8, Rpf-like proteins, protein MTB32, protein MTB39, crystallin, heat-shock protein HSP65, protein PST-S (usually Mycobacterium tuberculosis, Tuberculosis); outer membrane protein FobA, outer membrane protein FobB, intracellular growth locus lglCl, intracellular growth locus IglC2, aminotransferase W
  • the pathogenic antigen is selected from a) HIV p24 antigen, HIV envelope proteins (Gpl20, Gp41, Gpl60), polyprotein GAG, negative factor protein Nef, trans-activator of transcription Tat if the infectious disease is HIV, preferably an infection with Human immunodeficiency virus, b) major outer membrane protein MOMP, probable outer membrane protein PMPC, outer membrane complex protein B OmcB, heat shock proteins Hsp60 HSP10, protein IncA, proteins from the type III secretion system, ribonucleotide reductase small chain protein NrdB, plasmid protein Pgp3, chlamydial outer protein N CopN, antigen CT521, antigen CT425, antigen CT043, antigen TC0052, antigen TC0189, antigen TC0582, antigen TC0660, antigen TC0726, antigen TC0816, antigen TC0828 if the infectious disease is an infenction with Chlamydia tracho
  • parainfluenza virus infection preferably an infection with Human parainfluenza viruses (HPIV); h) Hemagglutinin (HA), Neuraminidase (NA), Nucleoprotein (NP), Ml protein, M2 protein, NS1 protein, NS2 protein (NEP protein: nuclear export protein), PA protein, PB1 protein
  • nucleoprotein N large structural protein L, phophoprotein P, matrix protein M, glycoprotein G if the infectious disease is Rabies, preferably an infection with Rabies virus; j) fusionprotein F, nucleoprotein N, matrix protein M, matrix protein M2-1, matrix protein M2-2, phophoprotein P, small hydrophobic protein SH, major surface glycoprotein G, polymerase L, non-structural protein 1 NS1, non-structural protein 2 NS2 if the infectious disease is
  • Respiratory syncytial virus infection preferably an infection with Respiratory syncytial virus (RSV); k) secretory antigen SssA (Staphylococcus genus, Staphylococcal food poisoning); secretory antigen SssA (Staphylococcus genus e.g.
  • molecular chaperone DnaK cell surface lipoprotein Mpt83, lipoprotein P23, phosphate transport system permease protein pstA, 14 kDa antigen, fibronectin-binding protein C FbpCl, Alanine dehydrogenase TB43, Glutamine synthetase 1, ESX-1 protein, protein CFP10, TB10.4 protein, protein MPT83, protein MTB12, protein MTB8, Rpf-like proteins, protein MTB32, protein MTB39, crystallin, heat-shock protein HSP65, protein PST-S if the infectious disease is Tuberculosis, preferably an infection with Mycobacterium tuberculosis; or genome polyprotein, protein E, protein M, capsid protein C, protease NS3, protein NS1, protein NS2A, protein AS2B, protein NS4A, protein NS4B, protein NS5 if the infectious disease
  • GpLuc Gaussia princeps luciferase
  • PpLuc Photinus pyralis luciferase
  • MmEpo Mus musculus erythropoietin
  • GpLuc amino acid sequence (SEQ ID NO: 11) :
  • PpLuc amino acid sequence (SEQ ID NO: 12) :
  • GpLuc mRNA sequence (SEQ ID NO: 14), also labelled R2851 herein:
  • PpLuc mRNA sequence (SEQ ID NO: 15) :
  • MmEpo mRNA sequence (SEQ ID NO: 16) : GGGUCCCGCAGUCGGCGUCCAGCGGCUCUGCUUGUUCGUGUGUGUCGUUGCAGGCCUUAUUC AAGCUUACCAUGGGCGUGCCCGAGCGGCCGACCCUGCUCCUGCUGCUCAGCCUGCUGCUCAUCCCCCUG GGGCUGCCCGUCCUCUGCGCCCCCCCGCGCCUGAUCUGCGACUCCCGGGUGCUGGAGCGCUACAUCCUC GAGGCCAAGGAGGCGGAGAACGUGACCAUGGGCUGCGCCGAGGGGCCCCGGCUGAGCGAGAACAUCACG GUCCCCGACACCAAGGUGAACUUCUACGCCUGGAAGCGCAUGGAGGUGGAGGAGCAGGCCAUCGAGGU CUGGCAGGGCCUGUCCCUCCUGAGCGAGGCCAUCCUGCAGGCGCAGGCCCUCCUGGCCAACUCCAGCCA GCCCCCGGAGACACUGCAGCUCCACAUCGACAAGGCCAUCUCCGGGCUGCGGAGCC
  • the DNA sequence encoding Gaussia princeps luciferase was prepared by modifying the wild type encoding DNA sequence by introducing a GC-optimized sequence for stabilization. Sequences were introduced into a derived pUC19 vector and modified to comprise stabilizing UTR sequences derived from 32L4-5'-UTR ribosomal 5'TOP UTR (32L4) and 3'UTR derived from albumin 7, a histone stem-loop sequence, a stretch of 64x adenosine at the 3'-terminal end (poly- A-tail) and a stretch of 30x cytosine at the 3'- terminal end (poly-C-tail) were introduced 3' of the coding sequence.
  • the sequence contains following sequence elements: the coding sequence encoding Gaussia luciferase; stabilizing sequences derived from 32L4-5'-UTR ribosomal 5'TOP UTR (32L4); 64x adenosine at the 3'-terminal end (poly- A- tail); 5 nucleotides, 30 x cytosine at the 3'- terminal end (poly-C-tail) and 5 additional nucleotides.
  • R2851 as mentioned in the present context resembles a GC-enriched mRNA sequence encoding for a Gaussia princeps luciferase having a poly (A) -sequence with 64 adenylates, followed by 5 nucleotides, followed by a poly(C)-sequence with 30 cytidylates and a histone stem- loop sequence followed by another 5 nucleotides.
  • the respective DNA plasmids were enzymatically linearized and transcribed in vitro using DNA dependent T7 RNA polymerase in the presence of a nucleotide mixture under respective buffer conditions.
  • GpLuc mRNA SEQ ID NO: 14
  • MmEpo mRNA SEQ ID NO: 16
  • m7GpppG cap analog
  • PpLuc in vitro transcribed mRNA (SEQ ID NO: 15) was enzymatically capped using a Script- Cap capping system (Cellscript).
  • PpLuc mRNA (SEQ ID NO: 15) and MmEpo mRNA (SEQ ID NO: 16) were enzymatically polyadenylated using a commercial polyadenylation kit.
  • DOTAP Permanently cationic lipids: - l,2-dioleoyl-3-trimethylammonium-propane
  • DOTMA - N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride
  • C9-C17-C3- cat - l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3-pyrrolidiniumcarboxylate
  • LPFA Lipofectamin ®
  • DOSPA DOPE
  • DOPE DOSPA-DOPE
  • Cationisable or permanently cationic lipidoids - 3-C12 (see formula below)
  • polyethylene glycol/peptide polymers (HO-PEG 5000-S-(S-CHHHHHHRRRRHHHHHHC-S-) 7 -S-PEG 5000-OH) were used for further formulation experiments, and are hereinafter referred to as PB83.
  • 3-C12-OH were prepared.
  • Lipidoid 3-C12 may be obtained by acylation of tris(2-aminoethyl)amine with an activated lauric (C12) acid derivative, followed by reduction of the amide. Alternatively, it may be prepared by reductive amination with the corresponding aldehyde.
  • Lipidoid 3-C12-OH is prepared by addition of the terminal C12 alkyl epoxide with the same oligoamine according to Love et al., pp. 1864-1869, PNAS, vol. 107 (2010), no. 5 (cf. compound C12-110).
  • This example describes the preparation of nanoparticles of polymer-lipid or polymer- lipidoid complexed mRNA, which were subsequently used for further in vitro and in vivo experiments.
  • the respective mRNA was prepared as described above.
  • glycol/peptide polymers (PB83) were prepared as described above.
  • ringer lactate buffer (RiLa; alternatively e.g. saline (NaCl) or PBS buffer may be used
  • respective amounts of lipid(oid) and respective amounts of a polymer (PB83) were mixed to prepare compositions comprising a lipid(oid) and a peptide or polymer.
  • the carrier compositions were used to assemble nanoparticles with the mRNA by mixing the mRNA with respective amounts of polymer-lipid carrier and allowing an incubation period of 10 minutes at room temperature such as to enable the formation of a complex between the lipid, polymer and mRNA.
  • the nanoparticles were then used for further in in vitro and in vivo experiments.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compositions for nucleic acid delivery are provided which comprise a relatively low amount a permanently cationic lipid or lipidoid, such as a lipid comprising a quaternary ammonium group. The compositions are suitable for the delivery of chemically modified or unmodified DNA or RNA. Moreover, the compositions are suitable for local administration, such as by extravascular injection.

Description

CATIONIC CARRIERS FOR NUCLEIC ACID DELIVERY
Background of the Invention
The present invention is in the fields of medical therapy, disease prevention and drug delivery. It relates in particular to carriers that are useful for delivering certain types of active ingredients to subjects in need thereof. More specifically, the invention relates to the delivery of such active ingredients which represent bioactive compounds that are challenging to deliver across biological barriers to their targets within a living organism, such as to target organs, tissues, or cells. Examples of such bioactive compounds that are of great therapeutic value and at the same time difficult to deliver to their biological targets include nucleic acid-based vaccines and therapeutics.
Various diseases today require a treatment which involves administration of peptide-, protein-, and nucleic acid-based drugs, particularly the transfection of nucleic acids into cells or tissues. The full therapeutic potential of peptide-, protein-, and nucleic acid-based drugs is frequently compromised by their limited ability to cross the plasma membrane of mammalian cells due to their size and electric charge, resulting in poor cellular access and inadequate therapeutic efficacy. Today this hurdle represents a major challenge for the biomedical development and commercial success of many biopharmaceuticals (see e.g. Foerg and Merkle, Journal of Pharmaceutical Sciences, published online at www.interscience.wiley.com, 2008, 97(1): 144-62). For some diseases or disorders, gene therapeutic approaches have been developed as a specific form of such treatments which require the transfection of cells or tissues with genes and their insertion into the DNA of the cells, e.g. in the case of hereditary diseases in which a defective mutant allele is replaced with a functional one. Transfer or insertion of nucleic acids or genes into an individual's cells, however, still represents a major challenge today, even though it is absolutely necessary for achieving a significant therapeutic effect of the gene therapy.
To achieve successful transfer of nucleic acids or genes into an individual's cells, a number of different hurdles have to be passed. The transport of nucleic acids typically occurs via association of the nucleic acid with the cell membrane and subsequent uptake by the endosomes. In the endosomes, the introduced nucleic acids are separated from the cytosol. As expression occurs in the cytosol, these nucleic acids have to depart the endosome. If the nucleic acids do not leave the endosome before the endosome fuses with a lysosome, they will suffer the usual fate of the content of the endosome and become degraded. Alternatively, the endosome may fuse with the cell membrane, leading to the return of its content into the extracellular medium. For efficient transfer of nucleic acids, the endosomal escape thus appears to be one of the most important steps additionally to the efficiency of transfection itself. Until now, there are different approaches addressing these issues. However, no approach has been entirely successful in all aspects so far.
Transfection agents used in the art today typically include various types of peptides, polymers, lipids, as well as other carrier compounds, which may be assembled into nano- or microparticles (see e.g. Gao, X., K. S. Kim, et al. (2007), AAPS J 9(1): E92-104). Most of these transfection agents have been successfully used only in in vitro reactions. When transfecting cells of a living animal with nucleic acids, further requirements have to be fulfilled. As an example, the complex of the nucleic acid and the carrier has to be stable in physiological salt solutions with respect to agglomeration. Furthermore, it must not interact with parts of the complement system of the host. Additionally, the complex must protect the nucleic acid from early extracellular degradation by ubiquitously occurring nucleases. For gene therapeutic applications, it is furthermore of great importance that the carrier is not recognized by the adaptive immune system (immunogenicity) and does not stimulate an unspecific cytokine storm (acute immune response) (see Gao, Kim et al, (2007, supra); Martin, M. E. and K. G. Rice (2007), AAPS J 9(1): E18- 29; and Foerg and Merkle, (2008, supra)).
Foerg and Merkle (2008, supra) discuss the therapeutic potential of peptide-, protein and nucleic acid-based drugs. According to their analysis, the full therapeutic potential of these drugs is frequently compromised by their limited ability to cross the plasma membrane of mammalian cells, resulting in poor cellular access and inadequate therapeutic efficacy. Today this hurdle represents a major challenge for the biomedical development and commercial success of many biopharmaceuticals.
In this context, Gao et al. (Gao et al. The AAPS Journal 2007; 9(1) Article 9) see the primary challenge for gene therapy in the development of a method that delivers a therapeutic gene to selected cells where proper gene expression can be achieved. Gene delivery and particularly successful introduction of nucleic acids into cells or tissue is, however, not simple and typically dependent on many factors. For successful delivery, e.g., delivery of nucleic acids or genes into cells or tissue, many barriers must be overcome. According to Gao et al. (2007) an ideal gene delivery method needs to meet 3 major criteria: (1) it should protect the transgene against degradation by nucleases in intercellular matrices, (2) it should bring the transgene across the plasma membrane and (3) it should have no detrimental effects.
Typically, the transfection of cells with nucleic acids is carried out using viral or non-viral vectors or carriers. For successful delivery, these viral or non-viral vectors must be able to overcome the above-mentioned barriers. The most successful gene therapy strategies available today rely on the use of viral vectors, such as adenoviruses, adeno-associated viruses, retroviruses, and herpes viruses. Viral vectors are able to mediate gene transfer with high efficiency and the possibility of long-term gene expression, and satisfy 2 out of 3 criteria.
However, the acute immune response, immunogenicity, and insertion mutagenesis uncovered in gene therapy clinical trials have raised serious safety concerns about some commonly used viral vectors.
A solution to this problem may be found in the use of non-viral vectors. Although non-viral vectors are not as efficient as viral vectors, many non-viral vectors have been developed to provide safer alternatives in gene therapy. Methods of non-viral gene delivery have been explored using physical (carrier-free gene delivery) and chemical approaches (synthetic vector-based gene delivery). Physical approaches usually include simple injection using injection needles, electroporation, gene gun, ultrasound, and hydrodynamic delivery. Some of these approaches employ a physical force that permeates the cell membrane and facilitates intracellular gene transfer. The chemical approaches typically use synthetic or naturally occurring compounds, e.g. cationic lipids or cationic polymers, as carriers to deliver the transgene into cells. Although significant progress has been made in the basic science and applications of various non-viral gene delivery systems, the majority of non-viral approaches is still less efficient than viral vectors, especially for in vivo gene delivery (see e.g. Gao et al. The AAPS Journal 2007; 9(1) Article 9).
Over the past decade, attractive prospects for a substantial improvement in the cellular delivery of nucleic acids have been announced that were supposed to result from their physical assembly or chemical ligation to so-called cell penetrating peptides (CPPs), also denoted as protein-transduction domains (PTDs) (see Foerg and Merkle, (2008, supra)). CPPs represent short peptide sequences of 10 to about 30 amino acids which can cross the plasma membrane of mammalian cells and may thus offer unprecedented opportunities for cellular drug delivery. Nearly all of these peptides comprise a series of cationic amino acids in combination with a sequence, which forms an a-helix at low pH. As the pH is continuously lowered in vivo by proton pumps, a conformational change of the peptide is usually initiated rapidly. This helix motif mediates an insertion into the membrane of the endosome leading to a release of its content into the cytoplasm (see Foerg and Merkle, (2008, supra); and Vives, E., P. Brodin, et al. (1997); A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem 272(25) : 16010-7). Despite these advantages, a major obstacle to CPP mediated drug delivery is thought to consist in the often rapid metabolic clearance of the peptides when in contact or passing the enzymatic barriers of epithelia and endothelia. Consequently, the metabolic stability of CPPs represents an important
biopharmaceutical factor for their cellular bioavailability. However, there are no CPPs available in the art which are on the one hand side stable enough to carry their cargo to the target before they are metabolically cleaved, and which on the other hand side can be cleared from the tissue before they can accumulate and reach toxic levels.
One further approach in the art for delivering cargo molecules into cells, e.g. for gene therapy, comprises the use of other types of peptide ligands (see Martin and Rice (see Martin and Rice, The AAPS Journal 2007; 9 (1) Article 3)). Such peptide ligands can be short sequences taken from larger proteins that represent the essential amino acids needed for receptor recognition, such as EGF peptide used to target cancer cells. Other peptide ligands have been identified including the ligands used to target the lectin-like oxidized LDL receptor (LOX-1). Up-regulation of LOX-1 in endothelial cells is associated with dysfunctional states such as hypertension and atherosclerosis. Such peptide ligands, however, are not suitable for many gene therapeutic approaches, as they cannot be linked to their cargo molecules by complexation or adhesion but require covalent bonds, e.g. crosslinkers, which typically exhibit cytotoxic effects in the cell.
Synthetic vectors may also be used for delivering cargo molecules into cells, e.g., for the purpose of gene therapy. However, one main disadvantage of many synthetic vectors is their poor transfection efficiency compared to viral vectors and significant improvements are required to enable further clinical development. Several barriers that limit nucleic acid transfer both in vitro and in vivo have been identified, and include poor intracellular delivery, toxicity and instability of vectors in physiological conditions (see. e.g. Read, M. L., K. H. Bremner, et al. (2003) : Vectors based on reducible polycations facilitate intracellular release of nucleic acids. J Gene Med 5(3): 232-45).
One specific approach in gene therapy uses cationic or cationisable lipids. However, although many cationic or cationisable lipids show excellent transfection activity in cell culture, most do not perform well in the presence of serum, and only a few are active in vivo. A dramatic change in size, surface charge, and lipid composition occurs when lipoplexes are exposed to the overwhelming amount of negatively charged and often amphipathic proteins and polysaccharides that are present in blood, mucus epithelial lining fluid, or tissue matrix. Once administered in vivo, lipoplexes tend to interact with negatively charged blood components and form large aggregates that could be absorbed onto the surface of circulating red blood cells, trapped in a thick mucus layer or embolized in microvasculatures, preventing them from reaching the intended target cells in the distal location. Furthermore, toxicity related to lipoplexes has been observed. Symptomes include inter alia induction of inflammatory cyokines. In humans, various degrees of adverse inflammatory reactions, including flu-like symptoms were noted among subjects who received lipoplexes. Accordingly, it appears questionable as to whether lipoplexes can be safely used in humans, in particular when repeated administration is required. One further approach in gene therapy utilizes cationic or cationisable polymers. Such polymers turned out to be efficient in the delivery of nucleic acids, as they can tightly complex and condense a negatively charged nucleic acid. Thus, a number of cationic or cationisable polymers have been explored as carriers for in vitro and in vivo gene delivery. These include
polyethylenimine (PEI), polyamidoamine and polypropylamine dendrimers, polyallylamine, cationic dextran, chitosan, various proteins and peptides. Although most cationic or cationisable polymers share the function of condensing DNA into small particles and facilitating cellular uptake via endocytosis through charge-charge interaction with anionic sites on cell surfaces, their transfection activity and toxicity differ dramatically. Interestingly, cationic or cationisable polymers exhibit better transfection efficiency with rising molecular weight due to stronger complexation of the negatively charged nucleic acid cargo. However, a rising molecular weight also leads to a rising toxicity of the polymer. PEI is perhaps the most active and most studied polymer for gene delivery, but its main drawback as a transfection reagent relates to its nonbiodegradable nature and toxicity. Furthermore, even though polyplexes formed by high molecular weight polymers exhibit improved stability under physiological conditions, data have indicated that such polymers can hinder vector unpacking. For example, poly(L-lysine) (PLL) of 19 and 36 amino acid residues was shown to dissociate from DNA more rapidly than PLL of 180 residues resulting in significantly enhanced short-term gene expression. A minimum length of six to eight cationic amino acids is required to compact DNA into structures active in receptor- mediated gene delivery. However, polyplexes formed with short polycations are unstable under physiological conditions and typically aggregate rapidly in physiological salt solutions. To overcome this negative impact, Read et al. (see Read, M. L., K. H. Bremner, et al. (2003) : Vectors based on reducible polycations facilitate intracellular release of nucleic acids. J Gene Med 5(3): 232-45; and Read, M. L., S. Singh, et al. (2005): A versatile reducible polycation-based system for efficient delivery of a broad range of nucleic acids. Nucleic Acids Res 33(9): e86) developed a new type of synthetic vector based on a linear reducible polycation (RPC) prepared by oxidative polycondensation of the peptide Cys-Lysio-Cys that can be cleaved by the intracellular environment to facilitate release of nucleic acids. They could show that polyplexes formed by RPC are destabilised by reducing conditions enabling efficient release of DNA and mRNA. Cleavage of the RPC also reduced toxicity of the polycation to levels comparable with low molecular weight peptides. The disadvantage of this approach of Read et al. (2003, supra) was that the
endosomolytic agent chloroquine or the cationic lipid DOTAP was additionally necessary to enhance transfection efficiency to adequate levels. As a consequence Read et al. (2005, supra) included histidine residues in the RPCs which have a known endosomal buffering capacity. They could show that histidine-rich RPCs can be cleaved by the intracellular reducing environment enabling efficient cytoplasmic delivery of a broad range of nucleic acids, including plasmid DNA, mRNA and siRNA molecules without the requirement for the endosomolytic agent chloroquine.
Read et al. (2005, supra) did not assess whether histidine-rich RPCs can be directly used for in vivo applications. In their study, transfections were performed in the absence of serum to avoid masking the ability of histidine residues to enhance gene transfer that may have arisen from binding of serum proteins to polyplexes restricting cellular uptake. Preliminary experiments indicate that the transfection properties of histidine-rich RPC polyplexes can be affected by the presence of serum proteins with a 50% decrease in GFP-positive cells observed in 10% FCS (fetal calf serum). For in vivo application they propose modifications with the hydrophilic polymer poly- [N-(2hydroxy-propyl)methacrylamide]. Thus, Read et al. (2005, supra) did not achieve the prevention of aggregation of polyplexes and binding of polycationic proteins to serum proteins. Furthermore, due to the large excess of polymer, which is characterized by the high N/P ratio, strong complexes are formed when complexing the nucleic acid, which are only of limited use in vivo due to their strong tendency of salt induced agglomeration and interactions with serum contents (opsonization). Additionally, these complexes may excite an acute immune response, when used for purposes of gene therapy. Neither did Read et al. (2003, supra) provide in vivo data for the RPC based complexes shown in the publication. It has turned out that these strong RPC based complexes are completely inactive subsequent to local administration into the dermis. Furthermore Read et al. (2005, supra) used stringent oxidation conditions (30% DMSO) to induce the generation of high molecular polymers with as long as possible chain lengths ("step-growth polymerization") to ensure complete complexation of the nucleic acid cargo.
In an approach similar to Read et al, McKenzie et al. (McKenzie, D. L., K. Y. Kwok, et al. (2000), J Biol Chem 275(14) : 9970-7., McKenzie, D. L., E. Smiley, et al. (2000), Bioconjug Chem 11(6): 901-9, and US 6,770,740 Bl) developed self-crosslinking peptides as gene delivery agents by inserting multiple cysteines into short synthetic peptides for the purpose of decreasing toxicity as observed with high-molecular polycations. For complexation of DNA they mixed the self- crosslinking peptides with DNA to induce interpeptide disulfide bonds concurrently to complexation of the DNA cargo. For in vivo gene delivery approaches they propose the
derivatization of the self-crosslinking peptides with a stealthing (e.g. polyethylene glycol) or targeting agent operatively attached to the peptide at a site distal from each terminus.. In a further approach the same authors developed for the purpose of masking DNA peptide condensates and thereby reducing interaction with blood components, the derivatization of the non crosslinking peptide CWKie with polyethylene glycol by reducible or non-reducible linkages (Kwok, K. Y., D. L. McKenzie, et al. (1999). "Formulation of highly soluble poly(ethylene glycol)- peptide DNA condensates." J Pharm Sci 88(10): 996-1003.). Summarizing the above, the present prior art as exemplified above suffers from various disadvantages. One particular disadvantage of the self-crosslinking peptides as described by Read et al. (2003, supra) or McKenzie et al. (2000 I and II, supra and US 6,770,740 Bl) concerns the high positive charge on the surface of the particles formed. Due to this charge, the particles exhibit a high instability towards agglomeration when subjecting these particles in vivo to raised salt concentrations. Such salt concentrations, however, typically occur in vivo in cells or extracellular media. Furthermore, complexes with a high positive charge show a strong tendency of opsonization. This leads to an enhanced uptake by macrophages and to a fast inactivation of the complex due to degradation. Particularly the uptake of these complexes by cells of the immune system in general leads to a downstream stimulation of different cytokines. This unspecific activation of the innate immune system, however, represents a severe disadvantage of these systems and should be avoided, particularly for the purpose of several aspects of gene therapy, where an acute immune response (cytokine storm) is strictly to be avoided. Additionally, in biological systems positively charged complexes can easily be bound or immobilized by negatively charged components of the extracellular matrix or the serum. Also, the nucleic acids in the complex may be released too early, leading to reduced efficiency of the transfer and half life of the complexes in vivo. Furthermore, a reversible derivatization of carriers with a stealthing agent being advantageous for in vivo gene delivery, such as polyethylene glycol (PEG), was only possible for peptide monomers but not for self-crosslinking peptides or rather for a polymeric carrier with a defined polymer chain length. In particular, such a reversible derivatization was not possible at the terminal ends of the crosslinked cationic peptide carrier. Additionally, in the prior art only high-molecular polymers with long polymer chains or with an undefined polymer chain length consisting of self-crosslinking peptides were described, which unfortunately compact their cargo to such an extent that cargo release in the cell is limited.. The extremely undefined polymer chain length is further problematic regarding regulatory approvement of a medicament based on RPC. One precondition for such approvement is that every preparation of the medicament has the same composition, the same structure and the same properties. This cannot be ensured for complexes based on RPC's from the prior art. Furthermore, the RPC-based polymers or complexes provided in the prior art are difficult to characterize due to their undefined structure or polymer chain length.
In consequence, no generally applicable method or carrier have been presented until today which allows both compacting and stabilizing a nucleic acid for the purposes of gene therapy and other therapeutic applications, and which show a good transfection activity in combination with a good release of the nucleic acid cargo, particularly in vivo and low or even no toxicity, e.g. due to the combination of a reversible stealthing and a reversible complexation of the nucleic acid by self-crosslinking polymers. Accordingly, there is still a need in the art to provide improved carriers for the purpose of gene transfer which are both stable enough to carry their cargo to the target before they being metabolically cleaved and which are nevertheless cleared from the tissue before they can accumulate and reach toxic levels.
The object underlying the present invention is therefore to provide a carrier, particularly for the delivery of nucleic acids for therapeutic or prophylactic applications, which is capable of compacting the nucleic acids and which allows their efficient introduction into different cell lines in vitro but also enables transfection in vivo. As uptake by cells occurs via the endosomal route, such a carrier or a complexing agent should also allow or provide for efficient release of the nucleic acid from endosomes. A further object is to provide a carrier that upon complexation with a nucleic acid exhibits resistance to agglomeration. A yet further object is to provide enhanced stability to the nucleic acid cargo with respect to serum containing media. Another object is to enable efficient in vivo activity without a strong acute immune reaction. A further object is to provide a composition that is particularly suitable for local delivery of a nucleic acid compound. Another object is to provide a means for administering a nucleic acid compound to a specific biological target while avoiding or reducing systemic exposure. A further object is to overcome any of the disadvantages or limitations of the known carriers for nucleic acid delivery as described e.g. herein-above. Further objects that are addressed by the present invention will become clear on the basis of the following description, the examples and the patent claims.
The objects are solved by the subject matter of the present invention as set forth in the patent claims.
Summary of the Invention
According to a first aspect, the invention provides a composition comprising a cationisable or permanently cationic lipid or lipidoid and a nucleic acid compound, wherein the lipid and the nucleic acid compound are non-covalently associated, and wherein the ratio of the lipid to the nucleic acid compound is not higher than about 2 nmol lipid per μg nucleic acid compound. Preferably, the composition comprises a permanently cationic lipid or lipidoid.
The nucleic acid compound may, for example, be any chemically modified or unmodified DNA or RNA.
In the composition of the invention, the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound may form a complex. Such complex may not be soluble in an aqueous evironment, so that the composition would typically comprise the complex in the form of a nanoparticle or a plurality of nanoparticles. Preferably, the nanoparticles comprise a permanently cationic lipid or lipidoid. According to a further aspect, the invention is directed to such nanoparticles. In addition to the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound, the nanoparticle - and thus the composition - may comprise further one or more other constituents, such as a targeting agent, a cell penetrating agent, and/or a stealth agent. According to a further aspect, the invention provides a kit for the preparation of the composition or of the nanoparticles. The kit may comprise a first kit component comprising the cationisable or permanently cationic lipid or lipidoid and a second kit component comprising the nucleic acid compound.
A yet further aspect of the invention is the medical use of the composition, the
nanoparticles, or the kits. For example, the composition, nanoparticles or kits may be used for the prophylaxis, treatment and/or amelioration of diseases selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e. (hereditary) diseases, or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency. One of the particularly preferred uses involves the extravascular administration of the composition and/or of the nanoparticles to a subject, such as by injection, infusion or
implantation, in particular intradermal, subcutaneous, intramuscular, interstitial, locoregional, intravitreal, periocular, intratumoural, intralymphatic, intranodal, intra-articular, intrasynovial, periarticular, intraperitoneal, intra-abdominal, intracardial, intrapericardial, intraventricular, intrapleural, perineural, intrathoracic, epidural, intradural, peridural, intrathecal, intramedullary intracerebral, intracavernous, intracorporus cavernosum, intraprostatic, intratesticular, intracartilaginous, intraosseous, intradiscal, intraspinal, intracaudal, intrabursal, intragingival, intraovarian, intrauterine, periodontal, retrobulbar, subarachnoid, subconjunctival or
intralesional injection, infusion or implantation; or by topical administration to the skin or a mucosa, in particular dermal or cutaneous, nasal, buccal, sublingual, otic or auricular, ophthalmic, conjunctival, vaginal, rectal, intracervical, endosinusial, laryngeal, oropharyngeal, ureteral, or urethral administration; or by administration to the respiratory system by inhalation, in particular by aerosol administration to the lungs, bronchi, bronchioli, alveoli, or paranasal sinuses; or by transdermal or percutaneous administration. The invention is based on the discovery that nucleic acid compounds may be effectively delivered to biological targets by carriers based on a cationisable or permanently cationic lipid or lipidoid, such as a lipid or lipidoid having a quaternised ammonium group bearing a positive charge at any pH value of its environment, in particular if the respective nucleic acid compound and the cationisable or permanently cationic lipid or lipidoid are incorporated in a composition which is locally administered to a subject, rather than by systemic injection via the intravenous or intraarterial route.
Further objects, aspects, useful embodiments, applications, beneficial effects and advantages of the invention will become apparent on the basis of the detailed description, the examples and claims below.
Brief Description of the Figures
Figures 1A to ID show the effect of the inventive polymer-lipid or polymer-lipidoid formulations on transfection efficiency of in HepG2 cells in vitro. All depicted transfection experiments were performed in triplicates, using GpLuc mRNA (SEQ ID NO: 14; also labelled as R2851 herein) as the cargo. Moreover, negative controls (buffer, passive pulsing) have been included. (A) to (D) show the GpLuc levels obtained using the indicated transfection reagents. In addition to the inventive polymer-lipid(oid) transfection reagent, polymer only has been used for comparison as well as the pure, 'naked' GpLuc mRNA without the use of transfection reagents. For further details, see Example 2.
Similarly, figures 2 A to 2 E show the effect of the inventive polymer-lipid or polymer- lipidoid formulations on transfection efficiency on Sol8 muscle cells in vitro. Again, all depicted transfection experiments were performed in triplicates, using GpLuc mRNA (SEQ ID NO: 14 / R2851) as the cargo; including negative controls (buffer, passive pulsing) and positive controls (polymer only and pure, 'naked' GpLuc mRNA without transfection reagents). For further details, see Example 3.
Figures 3A and 3B show the in vitro release of tumor necrosis factor alpha (TNFa; 3 A) cytokines interferon alpha (IFNa; 3B) and in human peripheral blood mononuclear cells
(hPBMCs) after treatment with different polymer-lipid or polymer-lipidoid complexed
GpLuc mRNA. For further details, see Example 4.
Figure 4 shows the scanning laser ophthalmoscopy (SLO) analysis results of the subretinal injection of PpLuc mRNA (SEQ ID NO: 15) into rat eyes, 24 h after subretinal injection of the inventive polymer-lipid or polymer-lipidoid formulations, expressed as relative light units (RLU). For the injection regimen and further details, see Example 5. Figure 5 shows the titers of antibodies against HA protein (hemagglutinin) as induced after intramuscular vaccination of Balb/c mice (n=8) with HA-mRNA (R2564, SEQ ID NO: 21) using an inventive polymer-lipidoid formulation of HA-mRNA or the 'naked' HA-mRNA alone. Each dot represents an individual animal and the horizontal lines represent median values. For further details, see Example 6.
Figure 6A shows the in vivo tissue distribution of an exemplary inventive PpLuc mRNA (SEQ ID NO: 15) polymer-lipid formulation of the permanently cationic lipid MC3-cat for the tissues liver, lung and spleen (mean value of four mice depicted) and figure 6B shows the in vivo lung distribution of an exemplary inventive polymer-lipid formulation. Each bar in figure 6A indicates the value of an individual mouse. Each dot in figure 6B represents an individual animal and the horizontal lines represent median values. For the injection regimen and further details, see Example 7.
Figures 7-A/B: show HI titers 21 days after prime vaccination with different formulated HA-mRNA (Figure 7-A) and 14 days after boost vaccination (Figure 7-B). The dashed line indicates the conventionally defined protective HI titer of 1:40.
Figures 8-A-D: show results of ELISA assays 21 days after prime vaccination and 14 days after boost vaccination with different formulated HA-mRNA and 14 days after boost vaccination (IgGl subtypes day 21 post-prime are shown in Figure 8-A, IgGl subtypes day 14 post-boost are shown in Figure 8-B, IgG2a subtypes day 21 post-prime are shown in Figure 8-C, IgG2a subtypes day 14 post-boost are shown in Figure 8-D).
Figure 9: shows T cell immune responses measured by IFNy production using Elispot.
Figure 10: shows GpLuc protein expression in A549 cells transfected with the mRNA construct R2851 in a formulation comprising 3-C12-OH.
Figure 11: shows GpLuc protein expression in A549 cells transfected with the mRNA construct R2851 in a formulation comprising DDAB.
Detailed Description of the Invention
Unless defined otherwise, or unless the specific context requires otherwise, all technical terms used herein have the same meaning as is commonly understood by a person skilled in the relevant technical field. Unless the context indicates or requires otherwise, the words "comprise", "comprises" and "comprising" and similar expressions are to be construed in an open and inclusive sense, as "including, but not limited to" in this description and in the claims.
The expressions, "one embodiment", "an embodiment", "a specific embodiment" and the like mean that a particular feature, property or characteristic, or a particular group or combination of features, properties or characteristics, as referred to in combination with the respective expression, is present in at least one of the embodiments of the invention. The occurrence of these expressions in various places throughout this description do not necessarily refer to the same embodiment. Moreover, the particular features, properties or characteristics may be combined in any suitable manner in one or more embodiments.
The singular forms "a", "an" and "the" should be understood as to include plural references unless the context clearly dictates otherwise.
Percentages in the context of numbers should be understood as relative to the total number of the respective items. In other cases, and unless the context dictates otherwise, percentages should be understood as percentages by weight (wt.-%).
In a first aspect, the invention provides a composition comprising a cationisable or permanently cationic lipid or lipidoid and a nucleic acid compound, wherein the lipid or lipidoid and the nucleic acid compound are non-covalently associated, and wherein the ratio of the lipid to the nucleic acid compound is not higher than about 2 nmol lipid or lipidoid per μg nucleic acid compound. Preferably, the cationisable or permanently cationic lipid or lipidoid is a permanently cationic lipid.
In the context of the invention, a "composition" refers to any type of composition in which the specified ingredients may be incorporated, optionally along with any further constituents. Thus, the composition may be a dry composition such as a powder or granules, or a solid unit such as a lyophilised form or a tablet. Alternatively, the composition may be in liquid form, and each constituent may be independently incorporated in dissolved or dispersed (e.g. suspended or emulsified) form. In one of the preferred embodiments, the composition is formulated as a sterile solid composition, such as a powder or lyophilised form for reconstitution with an aqueous liquid carrier. Such formulation is also preferred for those versions of the composition which comprise a nucleic acid compound which is not stable in an unfrozen aqueous composition for at least about 18 or preferably 24 months. As used herein, the expression "a cationisable or permanently cationic lipid or lipidoid" means a cationisable or permanently cationic lipid or or a cationisable or permanently cationic lipidoid.
A "compound" means a chemical substance, which is a material consisting of molecules having essentially the same chemical structure and properties. For a small molecular compound, the molecules are typically identical with respect to their atomic composition and structural configuration. For a macromolecular or polymeric compound, the molecules of a compound are highly similar but not all of them are necessarily identical. For example, a segment of a polypeptide that is designated to consist of 50 amino acids may also contain individual molecules with e.g. 48 or 53 amino acids.
Unless a different meaning is clear from the specific context, the term "cationic" as such means that the respective structure bears a positive charge, either permanently, or not permanently but in response to certain conditions such as pH. Thus, the term "cationic" (in the absence of a modifying terms such as "permanently" covers both "permanently cationic" and "cationisable".
As used herein, "permanently cationic" means that the respective compound, or group or atom, is positively charged at any pH value or hydrogen ion activity of its environment. In many cases, the positive charge is results from the presence of a quaternary nitrogen atom. Where a compound carries a plurality of such positive charges, it may be referred to as permanently polycationic, which is a subcategory of permanently cationic.
In this context, the prefix "poly-" refers to a plurality of atoms or groups having the respective property in a compound. If put in parenthesis, the presence of a plurality is optional. For example, (poly) cationic means cationic and/or polycationic. However, the absence of the prefix should not be interpreted such as to exclude a plurality. For example, a polycationic compound is also a cationic compound and may be referred to as such.
"Cationisable" means that a compound, or group or atom, is positively charged at a lower pH and uncharged at a higher pH of its environment. Also in non-aqueous environments where no pH value can be determined, a cationisable compound, group or atom is positively charged at a high hydrogen ion concentration and uncharged at a low concentration or activity of hydrogen ions. It depends on the individual properties of the cationisable or polycationisable compound, in particular the pKa of the respective cationisable group or atom, at which pH or hydrogen ion concentration it is charged or uncharged. In diluted aqueous environments, the fraction of cationisable compounds, groups or atoms bearing a positive charge may be estimated using the so-called Henderson-Hasselbalch equation which is well-known to a person skilled in the art.
For example, if a moiety is cationisable, it is preferred that it is positively charged at a pH value of about 1 to 8, preferably 4 to 8, 5 to 8 or even 6 to 8, more preferably of a pH value of or below 8, of or below 7, most preferably at physiological pH values, e.g. about 7.3 to 7.4, i.e. under physiological conditions, particularly under physiological salt conditions of the cell in vivo.
Unless a different meaning is clear from the specific context, "cationised" typically means that a cationisable structure is in a state where it actually bears a positively charge, as for example in the case of a basic amino acid such as arginine in a neutral physiological environment. The invention is based on the discovery that nucleic acid compounds may be effectively delivered to biological targets by carriers based on a cationisable or permanently cationic lipid or lipidoid, such as a lipid or lipidoid having a quaternised ammonium group bearing a positive charge at any pH value of its environment, in particular if the respective nucleic acid compound and the cationisable or permanently cationic lipid or lipidoid are incorporated in a composition which is locally administered to a subject, rather than by systemic injection via the intravenous or intraarterial route.
This is in contrast to the current state of the art in the field of lipid-mediated nucleic acid delivery where there is a clear preference for the use of cationisable lipids, i.e. which are predominantly cationic, i.e. positively charged, only at a pH which is lower than their pka (the logarithmic acid dissociation constant of the respective lipid). In this context, "predominantly cationic" means that more than 50% of the molecules of the compound are cationised.
Moreover, it has been found by the inventors that when a composition comprising a nucleic acid compound and a cationisable or permanently cationic lipid or lipidoid is administered e.g. by local or locoregional injection, the presence of such lipid or lipidoid leads to an unexpected effectiveness in the delivery of the nucleic acid compound to target cells, and at the same time is associated with an unexpectedly high level of tolerability, and a low degree of undesirable side effects or toxicity.
The cationisable or permanently cationic lipid or lipidoid may be any lipid, lipidoid or lipid- like compound that is generally known in the art which comprises a group or moiety which is cationisable or permanently cationic. Examples of useful lipids that are permanently cationic are compounds with a quaternary ammonium function, such as lipids comprising a
trimethylammonium moiety. As used herein, a "lipid" means any compound understood or classified as a lipid in the relevant technical field, which is in this case the field of nucleic acid formulation and delivery. Typically, a lipid is characterised in that it is lipophilic or hydrophobic, or it comprises a lipophilic, or hydrophobic, domain. This lipophilic domain may consist of one or more functional groups or moieties, such as one or more hydrocarbon chains or cyclic hydrocarbon groups.
The cationisable or permanently cationic lipid may further comprise a linking group which links the cationic group, which is substantially hydrophilic, with the lipophilic domain of the lipid.
A lipidoid, also referred to as lipidoid compound, is a lipid-like compound, i.e. an
amphiphilic compound with lipid-like physical properties. The lipidoid compound is preferably a compound which comprises two or more cationic nitrogen atoms and at least two lipophilic tails. In contrast to many conventional cationic lipids, the lipidoid compound may be free of a hydrolysable linking group, in particular linking groups comprising hydrolysable ester, amide or carbamate groups. The cationic nitrogen atoms of the lipidoid may be cationisable or cationisable or permanently cationic, or both types of cationic nitrogens may be present in the compound. Examples for potentially suitable lipids that are permanently cationic include, without limitation, the following compounds:
N,N-di-n-hexadecyl-N,N-dihydroxyethyl ammonium bromide ("DHDEAB");
N,N-di-n-hexadecyl-N-methyl-N-(2-hydroxyethyl) ammonium chloride ("DHMHAC");
N,N-myristyl-N-(l-hydroxyprop-2-yl)-N-methylammonium chloride ("DMHMAC");
N,N-di[(0-hexadecanoyl)hydroxyethyl]-N-hydroxyethyl-N-methyl ammonium bromide
("DOHEMAB");
N-methyl-N-n-octadecyl-N-oleyl-N-hydroxyethyl ammonium chloride ("MOOHAC");
N,N-di-n-octadecyl-N-methyl-N-dihydroxyethyl ammonium chloride ("DOMHAC");
N,N-distearyl-N,N-dimethylammonium ("DSD MA"; also known as N,N-dioctadecyl-N,N- dimethylammonium) and its salts, e.g. N,N-distearyl-N,N-dimethylammonium chloride ("DDAC" or DSDMAC") or N,N-dioctadecyl-N,N-dimethylammonium bromide ("DODAB" or "DDAB");
N,N-dioleyl-N,N-dimethylammonium and its salts, e.g. N,N-dioleyl-N,N-dimethylammonium chloride ("DODAC");
N,N-dioctadecyl-N,N-dimethylammonium and its salts;
N,N,N',N'-tetraoleyl-N,N'-dimethyl-l,3-propanediammonium chloride ("TODMAC3");
N,N,N',N'-tetraoleyl-N,N'-dimethyl-l,6-hexanediammonium chloride („TODMAC6");
N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTMA"; also known as 1,2- dioleyloxy-3-trimethylaminopropane chloride);
N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTAP" or "DOTAP.Cl", also known as l,2-dioleoyloxy-3-trimethylaminopropane chloride); l,2-dioleoyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide („DORI");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide („DORIE");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxypropyl ammonium bromide („DORIE-HP");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxybutyl ammonium bromide („DORIE-HB");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxypentyl ammonium bromide („DORIE-HPe");
l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE" or "DIMRI");
l,2-dimpalmityloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DPRIE");
l,2-distearyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DSRIE");
l,2-dilinoleyloxy-3-trimethylaminopropane chloride ("DLin-TMA.G");
l,2-dilinoleoyl-3-trimethylaminopropane chloride ("DLin-TAP.G");
rac-[(2,3-dioctadecyloxypropyl) (2-hydroxyethyl)]-dimethylammonium chloride ("CLIPl"); rac-[2(2,3-dihexadecyloxypropyl-oxymethyloxy)ethyl]trimethylammonium ("CLIP6");
rac- [2 (2, 3-dihexadecyloxypropyl-oxysuccinyloxy) ethyl] -trimethylammonium ("C LI P9 ") ;
N-[l-(2,3-dioleyloxy)propyl]-N-2-(sperminecarboxamido)ethyl)-N,N-dimethyl-ammonium trifluoracetate ("DOSPA"; also referred to as 2,3-dioleyloxy-[2(sperminecarboxamido)ethyl]-N,N- dimethyl-l-propanaminiumtrifluoroacetate);
monomeric and dimeric pyridinium amphiphiles (so called SAINTs), such as:
N-methyl-4-(dipalmityl)-methylpyridinium chloride ("SAINT-1");
N-methyl-4-(dioleyl)-methylpyridinium chloride ("SAINT-2");
N-methyl-4-(distearyl)-methylpyridinium chloride ("SAINT-5"); or
N-methyl-4-(stearyl) (oleyl)-methylpyridinium chloride ("SAINT-8");
synthetic phosphatidylcholines, such as:
l,2-dioleoyl-sn-glycero-3-phosphocholine (also dioleoylphosphatidylcholine; "DOPC"); l,2-dimyristoyl-sn-glycero-3-phosphocholine ("DMPC");
l,2-dipalmitoyl-sn-glycero-3-phosphocholine ("DPPC");
l,2-dierucoyl-sn-glycero-3-phosphocholine ("DEPC");
l-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine ("GIPC");
l-palmitoyl-2-azelaoyl-sn-glycero-3-phosphocholine ("AzPC");
l-palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (16:0-05:0 (CHO) PC);
l-palmitoyl-2-(9'-oxo-nonanoyl)-sn-glycero-3-phosphocholine;
l,2-dimyristoyl-sn-glycero-3-ethylphosphocholine ("DMEPC");
1,2- dipalmitoyl-sn-glycero-3-ethylphosphocholine ("DPePC");
0,0-ditetradecanoyl-N-(a-trimethylammonioacetyl)diethanolamine chloride ("DC-6-14"); (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4-(trimethylamino)butanoate and its salts ("DLin-MC3-TMA", also referred to as "MC3-cationized"); 3-beta-[N-(N',N',N'-trimethylaminoethane)carbamoyl]cholesterol iodide ("TC-Chol");
Lipofectin® (including DOTMA and DOPE, available from GIBCO/BRL);
Lipofectamin® (comprising DOSPA and DOPE, available from GIBCO/BRL);
l-(2-octylcyclopropyl)heptadec-8-yl-4-(trimethylammonium)butanoate ("C9-C17-C3 cat");
l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3-pyrrolidiniumcarboxylate ("C9-C17-P cat").
According to another one of the preferred embodiments, the cationisable or permanently cationic lipid is a compound according to one of the formulas
X-Y-Z (formula la)
X-Y(Zi)-Z2 (formula lb)
X-Y(Zi)(Z2)-Z3 (formula ic)
Ζΐ-γΐ-χ-γ2-Ζ2 (formula Id) wherein X represents a hydrophilic head group comprising a cationisable or permanently cationic nitrogen; Y, Y1 and Y2 are linking groups, each comprising an ether, ester, amide, urethane, thioether, disulphide, orthoester, or phosphoramide bond; and Z, Z1, Z2, and Z3 are independently selected and represent hydrophobic groups each comprising a linear or branched hydrocarbon chain or a cyclic hydrocarbon group, such as a steroid residue. Moreover, the number of carbon atoms in the linear or branched hydrocarbon chain is 6 or higher for Z; and 4 or higher for Z1 or Z2 or Z3, provided that, for compounds of formula lb, Z1 and Z2 together have at least 12 carbon atoms in their hydrocarbon chains, and for a compound of formula Ic, Z1, Z2 and Z3 together have at least 12 carbon atoms in their hydrocarbon chains.
In one of the preferred embodiments, the lipid does not comprise any group that exists in an anionised form at approximately neutral or physiological pH conditions, unless it also has more than one cationisable or permanently cationic groups whose positive charges dominate over the negative charge of the anionised group.
Preferably, the hydrophilic headgroup X is permanently cationic, and thus renders the lipid also to be permanently cationic. The hydrophilic headgroup typically is or comprises a quaternary ammonium group. As used herein, a quaternary ammonium group refers to a structure in which all four hydrogens of the ammonium cation (NH4 +) have been replaced by substituents. The quaternary ammonium group is also sometimes referred to as quaternary amine group.
The quaternary ammonium group may, for example, be an N-substituted pyridinium moiety, or a quaternary ammonium group with two or three methyl, hydroxyxethyl or hydroxypropyl groups, such as a trimethylamino group. Again, the group may also be part of a larger group, such as a trialkylaminoalkyl group. Some of the preferred quaternary ammonium groups are trialkylaminoalkyl groups selected from the following structures:
(i) trialkyl-N-, wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
(ii) trialkyl-N-CH2-, wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2- hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
(iii) trialkyl-N-CH2-CH2-, wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2- hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
(iv) trialkyl-N-C H2-CH2-CH2-, wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
(v) trialkyl-N-CH2-CH2-CH2-CH2-, wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3-hydroxypropyl;
(vi) trialkyl-N-C H2-CH2-CH2-CH2-CH2-, wherein alkyl is selected from methyl,
hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3- hydroxypropyl; or
(vii) trialkyl-N-CH2-CH2-CH2-CH2-CH2-CH2-, wherein alkyl is selected from methyl, hydroxymethyl, ethyl, 2-hydroxyethyl, n-propyl, isopropyl, 2-hydroxypropyl, and 3- hydroxypropyl.
Among the particularly preferred trialkylaminoalkyl groups are trimethylaminomethyl, trimethylaminoethyl, trimethylaminopropyl, and trimethylaminobutyl.
Alternatively, the three optionally substituted alkyl groups in the trialkylaminoalkyl groups exhibited above may be selected to be different from each other, as for example in N,N-dimethyl- N-ethylaminoalkyl groups with alkyl being in particular linear alkyl chains with 1 to 6 carbon atoms; or N,N-dimethyl-N-hydroxyethylaminoalkyl groups, N,N-dimethyl-N-propylaminoalkyl groups, N,N-diethyl-N-hydroxyethylaminoalkyl groups, or N-methyl-N-ethyl-N- hydroxyethylaminoalkyl groups, or similar groups with different combinations of optionally substituted methyl-, ethyl, or propyl groups attached to the nitrogen atom of the aminoalkyl structure, again with the alkyl being preferably selected from linear alkyl chains with 1 to 6 carbon atoms. In a further embodiment, the hydrophilic headgroup X comprises two or more ammonium groups which are optionally separated by a spacer. Suitable spacers include, for example, flexible hydrophilic spacers such as oxyethylene-type spacers, flexible hydrophobic spacers such as alkylenes, or rigid hydrophobic spacers such as aromatic structures. In some of the preferred embodiments, a trialkylaminoalkyl group as described above is attached to a further nitrogen atom, which may, for example, represent a tertiary amino group. Examples for such headgroups include in particular trimethylaminoalkylamino groups wherein the alkyl group between the two nitrogen atoms is preferably selected from linear alkyls with 1 to 6 carbon atoms. In case the headgroup X comprises such further amino group, that group may be connected with the linking group Y via a spacer, such as an alkyl chain.
As mentioned, the linking groups Y, or Y1 and Y2, link the hydrophilic headgroup X with the hydrophobic group Z, or with the hydrophobic groups Z1 and Z2, and with Z3, if present. Each linking group represents or comprises an ether, ester, amide, urethane, thioether, disulphide, orthoester, or phosphoramide bond, including any combinations of any of these. Among the preferred linking groups are ester groups and ether groups, and in the case of ethers, these include dioxolane groups. A dioxolane may also be understood as a cyclic acetal.
In a preferred embodiment, the linking groups Y, Y1 and Y2, are degradable under physiological conditions. As used herein, the expression "degradable under physiological conditions", which refers to a type of biodegradability, should be understood in the context of nucleic acid delivery. In this context, degradability requires some appreciable degree of degradation occurring within minutes, hours and/or days (rather than years) in order to be meaningful for in vivo applications. Preferably, this biodegradability is ensured by a chemical bond which is hydrolysable under physiological conditions, such as an ester, amide or acetal bond.
In the case of the ester group, this may be linked to the hydrophilic headgroup X via its carbonyl group or via the ester oxygen, for example according to the following formulas which are specific versions of formula la:
X-fCO)0-Z
x-orco)-z
In the case of lipid compounds according to formulas lb, Ic and Id which comprise more than one hydrophobic group, a suitable linking group Y, Y1 and/or Y2, based on an ester group may further comprise a carbon atom or alkyl (or similar) spacer as in the following subscopes of formulas lb, Ic, and Id:
X-(CO)0-fCH2)k-CH-(Zi)-Z2
X-0(CO)-(CH2)k-CH-(Zi)-Z2
X-(CO)0-(CH2)k-C-(Zi) (Z2)-Z3
X-0(CO)-(CH2)k-C-(Zi) (Z2) -Z3
X-0(CO)-(CH2)k-CH-(Zi)-Z2
Zi-CH-(CH2)k-0(CO)-X-(CO)0-(CH2)k-CH-Z2
Zi-CH-(CH2)k-(CO)0-X-0(CO)-(CH2)k-CH-Z2 wherein k is from 0 to about 10, and preferably selected from 0 and 1. The same principle applies to linking groups based on other functional groups, such as amides.
Linking groups Y1 and Y2 may be identical or different from each other. In one of the preferred embodiments, they are identical.
As mentioned, a linking alkyl (or similar) group with a spacer function may also be used between the ester group and the hydrophilic headgroup X, as in the following exemplary formulas:
Figure imgf000021_0001
X-(CH2)k-(CO)0-(CH2)k-CH-(Zi)-Z2
X-(CH2)k-0(CO)-(CH2)k-CH-(Zi)-Z2
X-(CH2)k-(CO)0-(CH2)k-C-(Zi) (Z2)-Z3
X-(CH2)k-0(CO)-(CH2)k-C-(Zi) (Z2)-Z3
Zi-CH-(CH2)k-0(CO)-(CH2)k-X-(CH2)k-(CO)0-(CH2)k-CH-Z2
Zi-CH-(CH2)k-(CO)0-(CH2)k-X-(CH2)k-0(CO)-(CH2)k-CH-Z2 wherein k is as previously defined. Such linking alkyl group may be considered as part of the overall linking group Y, Y1 or Y2, respectively.
In the case of a dioxolane linker, this is particularly useful in lipids of formulas la and lb. For example, the carbon atom in position 4 of the dioxolane ring may be connected to the headgroup X, and the carbon atom in position 2 may be linked to one or two hydrophobic groups, i.e. to Z or Z1 and Z2. In the case of a lipid according to formula Ic, the linking group may also comprise a dioxolane ring, but in this case the linking group should comprise a further carbon atom for linkage with Z1, Z2, and Z3. Such further carbon atom may be connected directly to e.g. the carbon atom in position 2 of the dioxolane ring, or via an alkyl spacer.
The skilled person will understand that it may not always be possible to draw a sharp line between the hydrophilic headgroup X and the linking group Y, Y1 or Y2. In some borderline cases, an atom or group may be considered as being part of either of the components, without being technically unreasonable. The same is true for the interface between the linking group (s) and the hydrophobic groups Z, Z1, Z2, and Z3.
As defined above, Z, Z1, Z2, and Z3 are independently selected and represent hydrophobic groups. Each comprises a linear or branched hydrocarbon chain or a cyclic hydrocarbon group, such as a steroid residue. Moreover, the number of carbon atoms in the linear or branched hydrocarbon chain is 6 or higher for Z; and 4 or higher for Z1 or Z2 or Z3, provided that, for compounds of formula lb or Id, Z1 and Z2 together have at least 12 carbon atoms in their hydrocarbon chains, and for a compound of formula Ic, Z1, Z2, and Z3 together have at least 12 carbon atoms in their hydrocarbon chains. For example, Z, Z1, Z2, and/or Z3 may be derived from fatty acids, glycerophospholipids, sphingolipids, glycerolipids, sterols, prenols, polyketides and the like.
In the case of Z representing a linear or branched hydrocarbon chain, the number of carbon atoms is at least 6, and preferably at least 8, or at least 10, or at least 12 carbon atoms, respectively. Other preferred ranges for the number of carbon atoms in the hydrocarbon chain are from 8 to 24, from 10 to 22, or from 12 to 20, respectively, such as about 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms. In the case of Z representing a steroid, it is further preferred that the steroid is cholesteryl or a derivative thereof.
In one of the preferred embodiments, the lipid is a compound of formula lb, Ic or Id, i.e. such as to exhibit more than one hydrophobic group. In the case of the compound of formula lb or Id, the hydrophobic groups Z1 and Z2 may be identical or different; in a preferred embodiment, they are identical. In the case of a compound of formula Ic, the groups Z1, Z2 and Z3 may be the same or different, and in a preferred embodiment, these are also identical.
In a further preferred embodiment, the lipid is a compound of formula lb with the hydrophobic groups Z1 and Z2 being identical, wherein each of Z1 and Z2 represents a linear hydrocarbon chain with a length of 14 to 22 carbon atoms, either saturated, such as
- tetradecyl (also referred to as myristyl),
- hexadecyl (also referred to as cetyl or palmityl),
- octadecyl (also referred to as stearyl), - eicosyl (also referred to as arachidyl), or
- docosyl (also referred to as behenyl),
or unsaturated, such as
- myristoleyl,
- palmitoleyl,
- oleyl,
- elaidyl,
- linoleyl,
- linolelaidyl,
- a-linolenyl, or
- arachidonyl.
Examples of such lipids include 2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane ("DLin-KC2-TMA"), (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4- (triimethylamino)butanoate ("DLin-MC3-TMA"). Obviously, the cationic lipids also require the presence of an anion, which should be selected from physiologically acceptable cations, such as chloride.
In a further preferred embodiment, the lipid is a compound of formula Ic with the hydrophobic groups Z1, Z2 and Z3 being identical, wherein each of the groups Z1, Z2 and represents a linear hydrocarbon chain with a length of 14 to 22 carbon atoms, either saturated or unsaturated, and preferably selected from those listed in the preceding paragraph.
In a further preferred embodiment, the lipid is a compound of formula Id with the hydrophobic groups Z1 and Z2 being identical, and wherein each of the groups Z1 and Z2 and represents a linear hydrocarbon chain with a length of 14 to 22 carbon atoms, either saturated or unsaturated, and preferably selected as described above in the context of the linear hydrocarbon chains for compounds according to formula lb.
Alternatively, and in accordance with another preferred embodiment for a lipid that is a compound of formula Id with the hydrophobic groups Z! and Z2 being identical, each of the groups Z1 and Z2 represents a branched or two-tailed hydrocarbon residue with a total number of 10 to 22 carbon atoms (per hydrophobic group Z1 or Z2). Such branched or two-tailed hydrocarbon residue may be saturated or unsaturated. A two-tailed structure may, for example, comprise two linear chains which may have different lengths and which are both connected to a carbon atom of the linking group Y1 or Y2. As mentioned previously, in such a case it may also be reasonable to consider that carbon atom to which both linear chains are connected as part of the hydrophobic group rather than the linking group. This would be more in line with common terminology according to which such hydrophobic group would be termed, for example, "9-nonadecyl" rather than separately naming the two tails (octyl and decyl) and the linking Ci member.
In a further preferred embodiment, the permanently cationic lipid is a compound according to formula la, lb, Ic or Id wherein
X is selected from a quaternary ammonium group, in particular a trimethylammonium group; and/or
Y, Y1 and/or Y2 is are selected from linking groups comprising an ester or amide bond or a dioxolane ring; and/or Z is a steroid residue; and/or
Z1, Z2, and/or Z3 are selected from saturated or unsaturated hydrocarbon chains with 14 to 22 carbon atoms.
For the avoidance of doubt, in the context of a permanently cationic lipid or lipidoid, a trimethylammonium group means the group -N+(CI¾).
Since the lipid (or lipidoid, as described below) is a cationisable or permanently cationic compound, it may require an anion, unless it is a zwitterionic compound with at least as many anionic groups as permanently cationic groups. The anion may be selected independently for each compound of interest. In principle, any biocompatible and - in particular if an in vivo use is contemplated - physiologically acceptable anion may be used. Particularly preferred anions include chloride, bromide, malonate, citrate, acetate, maleate, fumarate, succinate, lactate, tartrate, pamoate, hydrogen phosphate, in particular chloride. Further optional anions may be selected from commonly known lists of pharmaceutical salts, such as the anions listed by Stahl et al., Handbook of Pharmaceutical Salts, Wiley- VCH (2002), as salts of classes I, II or III, from which salts of classes I and II are preferred as class I ions are physiologically ubiquitous or occur as intermediate metabolites in biochemical pathways, and class II salts, while not naturally occurring, have been used in pharmaceuticals and have shown low toxicity and good tolerability.
In some of the preferred embodiments, the lipid is cationisable or permanently cationic and a compound according to formula la, lb, Ic or Id which is not zwitterionic under substantially neutral or physiological conditions, and is selected from the following compounds:
N,N-di[(0-hexadecanoyl)hydroxyethyl]-N-hydroxyethyl-N-methyl ammonium bromide
("DOHEMAB");
N-[l-(2,3-dioleyloxy)propyl] -N,N,N-trimethylammonium chloride ("DOTMA"; also known as 1,2- dioleyloxy-3-trimethylaminopropane chloride);
N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTAP" or "DOTAP.Cl", also known as l,2-dioleoyloxy-3-trimethylaminopropane chloride);
l,2-dioleoyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide („DORI");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide („DORIE");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxypropyl ammonium bromide („DORIE-HP");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxybutyl ammonium bromide („DORIE-HB");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxypentyl ammonium bromide („DORIE-HPe");
l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE" or "DIMRI")
l,2-dimpalmityloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DPRIE");
l,2-distearyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DSRIE");
l,2-dilinoleyloxy-3-trimethylaminopropane chloride ("DLin-TMA.G");
l,2-dilinoleoyl-3-trimethylaminopropane chloride ("DLin-TAP.G");
rac-[(2,3-dioctadecyloxypropyl)(2-hydroxyethyl)]-dimethylammonium chloride ("CLIPl");
rac-[2(2,3-dihexadecyloxypropyl-oxymethyloxy)ethyl]trimethylammonium ("CLIP6");
rac- [2 (2, 3-dihexadecyloxypropyl-oxysuccinyloxy) ethyl] -trimethylammonium ("C LI P9 ") ;
N-[l-(2,3-dioleyloxy)propyl]-N-2-(sperminecarboxamido)ethyl)-N,N-dimethyl-ammonium trifluoracetate ("DOSPA"; also referred to as 2,3-dioleyloxy-[2(sperminecarboxamido)ethyl]-N,N- dimethyl-l-propanaminiumtrifluoroacetate);
0,0-ditetradecanoyl-N-(a-trimethylammonioacetyl)diethanolamine chloride ("DC-6-14");
(6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4-(trimethylamino)butanoate and its salts ("DLin-MC3-TMA", also referred to as "MC3-cationized");
2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane ("DLin-KC2-TMA", also referred to as "KC2 cationised");
3-beta-[N-(N',N',N'-trimethylaminoethane)carbamoyl]cholesterol iodide ("TC-Chol");
l-(2-octylcyclopropyl)heptadec-8-yl-4-(trimethylammonium)butanoate ("C9-C17-C3 cat"); l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3-pyrrolidiniumcarboxylate ("C9-C17-P cat").
In one of the particularly preferred embodiments, the cationisable or permanently cationic lipid is selected from (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4- (trimethylamino)butanoate and its salts ("DLin-MC3-TMA"), 2,2-dilinoleyl-4-(2- trimethylaminoethyl)-[l,3]-dioxolane ("DLin-KC2-TMA"), l-(2-octylcyclopropyl)heptadec-8-yl-4- (trimethylammonium)butanoate ("C9-C17-C3 cat"), l-(2-octylcyclopropyl)heptadec-8-yl-l,l- dimethyl-3-pyrrolidiniumcarboxylate ("C9-C17-P cat"), including any salts of these compounds. In one preferred embodiment, the permeanently cationic lipid is (6Z,9Z,28Z,31Z)-heptatriaconta- 6,9,28,31-tetraen-19-yl-4-(trimethylamino)butanoate or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof. In abother preferred embodiment, the permeanently cationic lipid is 2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof. In another preferred embodiment, the permeanently cationic lipid is l-(2-octylcyclopropyl)heptadec-8-yl-4- (trimethylammonium)butanoate ("C9-C17-C3 cat") or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof. In another preferred embodiment, the cationisable or permanently cationic lipid is l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3- pyrrolidiniumcarboxylate ("C9-C17-P cat"), or a salt thereof, in particular a halogen salt, such as the chloride or bromide salt thereof.
In the list above, as in the other lists of specific lipids in this description, some commonly used abbreviations are mentioned which are used inconsistently in the technical literature for different compounds (e.g. DSDMA, DODMA etc.). The compounds in the list are therefore disclosed primarily by their chemical names, and the abbreviations should be disregarded if inconsistent.
As mentioned, the composition may comprise a cationisable or permanently cationic lipidoid compound, instead of or in addition to the cationisable or permanently cationic lipid. As described, a lipidoid is a lipid-like compound, i.e. an amphiphilic compound with lipid-like physical properties.
In one embodiment, the lipidoid is a compound comprising at least two cationisable or permanently cationic nitrogen atoms and at least two lipophilic tails. As used herein, a "tail" is a substructure of a molecule representing a chain or chain-like structure, such as an optionally substituted hydrocarbyl, acyl or acyloxylalkyl chain of at least four, and more preferably at least six, carbon atoms. The optionally substituted hydrocarbyl, acyl or acyloxyalkyl chain representing the lipophilic tail may be directly connected with a cationic nitrogen atom.
In one specific embodiment, the lipidoid is a compound comprising two identical lipophilic tails, each of which is directly connected with a cationisable or permanently cationic nitrogen atom. In another specific embodiment, the lipidoid is a compound comprising three identical lipophilic tails, each tail being directly connected with a cationisable or permanently cationic nitrogen atom. In a further specific embodiment, the lipidoid is a compound comprising four or more identical lipophilic tails, each tail being directly connected with a cationisable or
permanently cationic nitrogen atom. In each of these embodiments, the lipidoid may optionally comprise a further nitrogen atom (which may be cationisable or permanently cationic) to which no lipophilic tail is connected. Such lipidoid may also be understood as a compound having a cationic backbone derived from an oligoamine with the lipophilic tails being attached to the, or some of the, cationic nitrogens of the oligoamine, and wherein the cationic nitrogens, or at least some of them, have been substituted (e.g. alkylated) such as to render them permanently cationic. In a preferred specific embodiment, the lipidoid is a compound comprising two identical lipophilic tails, each of which is directly connected with a permanently cationic nitrogen atom. In another specific embodiment, the lipidoid is a compound comprising three identical lipophilic tails, each tail being directly connected with a permanently cationic nitrogen atom. In a further specific embodiment, the lipidoid is a compound comprising four or more identical lipophilic tails, each tail being directly connected with a permanently cationic nitrogen atom. In each of these embodiments, the lipidoid may optionally comprise a further nitrogen atom (which may be cationisable or permanently cationic) to which no lipophilic tail is connected. Such lipidoid may also be understood as a compound having a cationic backbone derived from an oligoamine with the lipophilic tails being attached to the, or some of the, cationic nitrogens of the oligoamine, and wherein the cationic nitrogens, or at least some of them, have been substituted (e.g. alkylated) such as to render them permanently cationic.
If the lipophilic tails are substituted hydrocarbyl (e.g. alkyl) chains, the substituent may, for example, be a methyl or a hydroxyl.
The optionally substituted hydrocarbyl chain may be saturated, such as to resemble an alkyl, or it may be unsaturated, i.e. an alkenyl or alkynyl, each optionally having one, two, three or more carbon-carbon double bonds and/or triple bonds. In the case of tail structures representing or including an acyl or acyloxyalkyl group, these may also comprise one, two or more carbon- carbon double bonds and/or triple bonds in the hydrocarbon segment of the tail.
In one embodiment, the lipidoid compound is free of hydrolysable linking groups, such as ester, amide or carbamate groups. As used herein, a linking group is a group which links the lipophilic tails of the lipidoid molecule to the hydrophilic region comprising the cationic nitrogen atoms. Conventional cationic lipids that have been proposed as carriers or agents to deliver nucleic acids to cells and enhance transfection often - if not typically - exhibit such linkers or linking groups, which are most often hydrolysable and/or enzymatically cleavable. In particular, linkers with ester groups have been proposed, but also linkers with amide groups or carbamate groups, all of which are susceptible to hydrolytic and/or enzymatic cleavage in vivo.
As used herein, hydrolysable means that an appreciable degree of hydrolysis occurs in a physiological fluid (such as interstitial fluid) under in vivo conditions within seconds, minutes, hours, or days; preferably, the respective compound or group is hydrolysed to at least 50% after not more than 7 days, or even after not more than 2 days.
As said, the lipidoid compound is cationisable or permanently cationic. In one embodiment, at least one of the cationic nitrogen atoms of the lipidoid is cationisable or permanently cationic. Optionally, the lipidoid comprises two cationisable or permanently cationic nitrogen atoms, three cationisable or permanently cationic nitrogen atoms, or even four or more cationisable or permanently cationic nitrogen atoms.
Preferably, the lipidoid compound is permanently cationic. In one embodiment, at least one of the cationic nitrogen atoms of the lipidoid is permanently cationic. Optionally, the lipidoid comprises two permanently cationic nitrogen atoms, three permanently cationic nitrogen atoms, or even four or more permanently cationic nitrogen atoms
In a further embodiment, the lipidoid compound is a cationisable or permanently cationic derivative of a compound according to formula II
Figure imgf000028_0001
(formula II)
or a pharmaceutically acceptable salt thereof.
In formula II, each occurrence of RA is independently unsubstituted, cyclic or acyclic, branched or unbranched Ci-20 aliphatic; substituted or unsubstituted, cyclic or acyclic, branched or unbranched Ci-20 heteroaliphatic; substituted or unsubstituted aryl; substituted or
unsubstituted heteroaryl;
Figure imgf000028_0002
wherein at least one RA is
Figure imgf000028_0003
Moreover, each occurrence of R5 is independently unsubstituted, cyclic or acyclic, branched or unbranched e-u aliphatic; substituted or unsubstituted aryl; or substituted or unsubstituted heteroaryl. Furthermore, each occurrence of x is an integer from 1 to 10, and each occurrence of y is an integer from 1 to 10. Optionally, R5 is Ce-Ci6 alkyl for at least one occurrence, or even at each occurrence.
According to one of the preferred embodiments, at least one x is selected from 1 or 2, and optionally all occurrences of x are 1 or 2. Moreover, at least one y is selected from 1 or 2, and optionally all occurrences of y are 1 or 2. In a further embodiment, all occurrences of R5 are Ce-C i6 alkyl, all occurrences of x are 1 or 2, and all occurrences of y are 1 or 2. In some embodiments, such lipidoids may be prepared by reacting an oligoamine and an epoxide-terminated aliphatic compound at elevated temperatures, such as at 80 to 95 °C, in the absence of a solvent, followed by the quaternisation of one or more nitrogen atoms in the lipidoid. Such compound includes a hydrophilic portion resulting from the opening of the epoxide by the amine and a hydrophobic aliphatic tail. Preferably, the oligoamine comprises from 2 to 5 nitrogen atoms. Among the preferred oligoamines for preparing such lipidoids are, without limitation:
Figure imgf000029_0001
H3C-NH-CH2-CH2-CH2-NH2
H3C-NH-CH2-CH2-CH2-NH-CH3
H2N-CH2-CH2-NH-CH2-CH2-OH
H2N-CH2-CH2-NH-CH2-CH2-NH2
H2N-CH2-CH2-CH2-NH-CH2-CH2-NH2
H2N-CH2-CH2-NH(CH3)-CH2-CH2-NH2
HO-CH2-CH2-NH-CH2-CH2-NH-CH2-CH2-OH
H2N-CH2-CH2-NH-CH2-CH2-NH-CH2-CH2-NH2
H2N-CH2-CH2-N(CH2-CH2-NH2) -CH2-CH2-NH2
H2N-CH2-CH2-NH-CH2-CH2-NH-CH2-CH2-NH-CH2-CH2-NH2
Among the preferred epoxide-terminated aliphatic compound are, without limitation, 2- alkyloxiranes wherein alkyl is butyl, hexyl, octyl, decyl, dodecyl, tetradecyl or octadecyl.
Optionally, the alkyl group may further exhibit an alkyl side chain, such as a methyl, ethyl, propyl, or isopropyl side chain. Moreover, the alkyl group of the 2-alkyloxirane may also comprise one or more heteroatoms such as oxygen. Furthermore, the epoxide-terminated aliphatic compound may comprise one or more carbon-carbon double or triple bonds. For further guidance regarding the preparation of such lipidoid compounds which may further be quaternised, reference is made to US8969353, the disclosure of which is incorporated herein in its entirety.
According to a further embodiment, the cationisable or permanently cationic lipidoid comprises at least one moiety of formula III:
-N+(Ri) (R2)-CH2-CH(R3)-R4 (formula III) wherein independently for each individual moiety of formula III, Ri and R2 are
independently from each other selected from Ci-C4-alkyl; R3 is hydrogen or hydroxyl; and R4 is selected from linear or branched, saturated or unsaturated C6-C16 hydrocarbyl chain.
Ri and R2 may be selected independently of each other and independently for each occurrence. In one embodiment, Ri and/or R2 are the same for each occurrence, in particular when Ri and/or R2 is methyl. R3 may also be the same for each occurrence; for example, all instances of R3 may be hydroxyl. R4 may also be the same for each occurrence; for example, all instances of R4 may be a C6-C16 hydrocarbyl chain.
In a further embodiment, the cationisable or permanently cationic lipidoid comprises three identical moieties of formula III, wherein Ri and R2 are methyl; R3 is hydroxyl, and R4 is a linear or branched C6-C16 alkyl chain.
In yet a further embodiment, the lipidoid compound is a compound comprising three identical moieties of formula III, wherein Ri is methyl, R2 is methyl; R3 is hydroxyl, and R4 is a linear or branched C6-C16 alkyl chain.
For example, a compound according to formula III may be based on the oligoamine backbone
H2N-CH2-CH2-N(CH2-CH2-NH2)-CH2-CH2-NH2 wherein for each of the three primary amino groups, one of the hydrogen atoms is substituted with
-CH2-CH(OH)-R4 wherein R4 is a linear or branched C6-C16 alkyl, in particular a linear e, Ce, w or C12 alkyl.
In one specific embodiment, the cationisable or permanently cationic lipidoid is a compound comprising the cation depicted in the formula IV:
Figure imgf000031_0001
and further optionally an anion, preferably an anion as described above.
In a further specific embodiment, the cationisable or permanently cationic lipidoid is compound of formula IVa:
Figure imgf000031_0002
The compounds may be prepared by first reacting the oligoamine of the formula:
H2N-CH2-CH2-N(CH2-CH2-NH2) -CH2-CH2-NH2 with an unbranched, saturated terminal C12 alkyl epoxide, optionally followed by quaternisation with activated methyl such as methyl iodide. One aspect of the invention is directed to this compound as such, including any salts thereof.
In a further embodiment, the cationisable or permanently cationic lipidoid is a cationisable or permanently cationic derivative of a com ound according to formula V:
Figure imgf000031_0003
(formula V) wherein Ri and R2 are each independently selected from the group consisting of hydrogen, an optionally substituted, saturated or unsaturated C1-C20 hydrocarbyl, and an optionally substituted, saturated or unsaturated C6-C20 acyl. Moreover, Li and L2 are each independently selected from optionally substituted, saturated or unsaturated C1-C30 hydrocarbyls; m and o are each independently selected from the group consisting of zero and any positive integer; and n is any positive integer.
In one of the preferred embodiments, Ri and R2 of the lipidoid of formula V are both C1-C20 alkyl, more preferably Ci-C6 alkyl, in particular methyl, ethyl, propyl or isopropyl. For example, both of Ri and R2 may be methyl. Moreover, n is preferably not higher than about 5, in particular not higher than about 2, such as 1. Furthermore, o is preferably selected from 0 or 1, and m is preferably selected from the range from 1 to 6, such as 1, 2, 3, 4, 5 or 6.
In a further embodiment, Li and L2 are each independently selected from unsaturated Ce- C22 hydrocarbyls, in particular from C10-C22 hydrocarbyls. Among the preferred hydrocarbyls are linear omega-6 and omega-9 unsaturated hydrocarbon chains with 14, 16, 18, 20, or 22 carbon atoms.
Also preferred are lipidoids of formula V wherein Ri and R2 are both methyl, m is 3 or 4, n is 1, o is 0 or 1, and Li and L2 are identical linear omega-6 and omega-9 unsaturated hydrocarbon chains with 16 or 18 carbon atoms.
In a further embodiment, the lipidoid is a compound according to formula V as defined above except that n is 0.
Optionally, the composition comprises two or more cationisable or permanently cationic lipidoids, each being independently selected as described above; or it may comprise a
combination of a cationisable or permanently cationic lipidoid with another lipidoid or cationic lipid.
In one embodiment, the composition is substantially free of lipids other than the lipidoid defined above; or is substantially free of lipids other than those defined in one of the claims. In fact, it is one of the particular advantages of the present invention that it does benefit from the properties and advantageous effects of the lipidoid in terms of effective delivery of the nucleic acid but without requiring the presence of those other lipids which are not cationic as defined above and which are often used to prepare lipoplexes or lipid nanoparticles, such as zwitterionic phospholipids or steroids such as cholesterol, which are sometimes referred to as helper lipids. Accordingly, it is one of the preferred embodiments of the invention that the composition is free of neutral or zwitterionic lipids; or that it is free of steroids such as cholesterol. Without being restricted thereto, the following permanently cationic lipid and lipidoid structures have been used in the present invention:
MC3 cat
DOTAP
Figure imgf000033_0001
DOTMA
C9-C17-C3 O CHj cat
C9-C17-P o
cat
Figure imgf000034_0001
The biologically active cargo material comprised in the composition or in the
nanoparticle(s) of the invention is preferably a nucleic acid compound or complex. In some of the preferred embodiments, the nucleic acid compound is selected from chemically modified or unmodified DNA, single stranded or double stranded DNA, coding or non-coding DNA, optionally selected from a plasmid, (short) oligodesoxynucleotide (i.e. a (short) DNA oligonucleotide), genomic DNA, DNA primers, DNA probes, immunostimulatory DNA, aptamer, or any combination thereof. Alternatively, or in addition, such a nucleic acid molecule may be selected e.g. from any PNA (peptide nucleic acid). Further alternatively, or in addition, and also according to a particularly preferred embodiment, the nucleic acid is selected from chemically modified or unmodified RNA, single-stranded or double-stranded RNA, coding or non-coding RNA, optionally selected from messenger RNA (mRNA), (short) oligoribonucleotide (i.e. a (short) RNA
oligonucleotide), viral RNA (vRNA), replicon RNA, transfer RNA (tRNA), ribosomal RNA (rRNA), immunostimulatory RNA (isRNA), micro RNA, small interfering RNA (siRNA), small nuclear RNA (snRNA), small-hairpin RNA (shRNA) or a riboswitch, an RNA aptamer, an RNA decoy, an antisense RNA, a ribozyme, or any combination thereof. Preferably, the nucleic acid molecule of the complex is an RNA. More preferably, the nucleic acid molecule of the complex is a (linear) single-stranded RNA, even more preferably an mRNA or an immunostimulatory RNA.
Optionally, the biologically active cargo material is a combination of more than one nucleic acid compounds.
Described from a different angle, the nucleic acid may be a single- or a double-stranded nucleic acid compound or complex. Strictly speaking, a double-stranded nucleic acid could also be considered as a combination of two nucleic acid compounds (i.e. the two antiparallel strands) which form a nucleic acid complex due to their association by non-covalent bonds. However, like in common technical language, a double-stranded nucleic acid may also be described as one compound or molecule. The nucleic acid may also be a partially double-stranded or partially single stranded nucleic acid, comprising two strands which are at least partially self- complementary. Such 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 molecule and both thus form a double-stranded nucleic acid molecule in this region, i.e. a partially double-stranded or partially single stranded nucleic acid (molecule). Preferably, the nucleic acid compound is a single-stranded nucleic acid. Furthermore, the nucleic acid compound may be a circular or linear nucleic acid, preferably a linear nucleic acid.
Optionally, the nucleic acid may be an artificial nucleic acid. An "artificial nucleic acid molecule" or "artificial nucleic acid" may typically be understood to be a nucleic acid molecule, e.g. a DNA or an RNA, that does not occur naturally. In other words, an artificial nucleic acid molecule may be understood as a non-natural nucleic acid molecule. Such nucleic acid molecule may be non-natural due to its individual sequence (which does not occur naturally) and/or due to other modifications, e.g. structural modifications of nucleotides which do not occur naturally. An artificial nucleic acid molecule may be a DNA molecule, an RNA molecule or a hybrid-molecule comprising DNA and RNA portions. Typically, artificial nucleic acid molecules may be designed and/or generated by genetic engineering methods to correspond to a desired artificial sequence of nucleotides (heterologous sequence). In this context an artificial sequence is usually a sequence that may not occur naturally, i.e. it differs from the wild type sequence by at least one nucleotide. The term "wild type" may be understood as a sequence occurring in nature. Further, the term "artificial nucleic acid molecule" is not restricted to mean "one single molecule" but is, typically, understood to comprise an ensemble of identical molecules. Accordingly, it may relate to a plurality of identical molecules contained in an aliquot.
In a further embodiment, the sequences (protein, or respectively nucleic acid) which are defined in the present invention comprise or consist of a sequence (protein, or respectively nucleic acid) having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% to said sequence (protein, or respectively nucleic acid).
A combination of two or more different nucleic acids may be useful, for example, in the case of a composition comprising a nucleic acid (such as an RNA) encoding the heavy chain of an antibody as well as a nucleic acid encoding the light chain of the same antibody. Another example is the combination of two or more nucleic acids to affect the part of an organism's immune system referred to as the CRISPR/Cas system (CRISPR: clustered regularly interspaced short palindromic repeats; Cas: CRISPR associated protein).
A yet further example is the combination of a guide RNA (gRNA) with an encoding nucleic acid within the composition or nanoparticle of the invention. Coding nucleic acids
The nucleic acid may encode a protein or a peptide, which may be selected, without being restricted thereto, e.g. from therapeutically active proteins or peptides, selected e.g. from antigens, e.g. tumour antigens, pathogenic antigens (e.g. selected, from animal antigens, from viral antigens, from protozoal antigens, from bacterial antigens), allergenic antigens, autoimmune antigens, or further antigens, from allergens, from antibodies, from immunostimulatory proteins or peptides, from antigen-specific T-cell receptors, or from any other protein or peptide suitable for a specific (therapeutic) application, wherein the coding nucleic acid may be transported into a cell, a tissue or an organism and the protein may be expressed subsequently in this cell, tissue or organism. Bicistronic nucleic acid or RNA and multicistronic nucleic acid or RNA: A bicistronic or multicistronic nucleic acid or RNA is typically a nucleic acid or an RNA, preferably an mRNA, that typically may have two (bicistronic) or more (multicistronic) coding regions. A coding region in this context is a sequence of codons that is translatable into a peptide or protein.
According to certain embodiments of the present invention, the nucleic acid is mono-, bi-, or multicistronic, preferably as defined herein. The coding sequences in a bi- or multicistronic nucleic acid molecule preferably encode distinct proteins or peptides as defined herein or a fragment or variant thereof. Preferably, the coding sequences encoding two or more proteins or peptides may be separated in the bi- or multicistronic nucleic acid by at least one IRES (internal ribosomal entry site) sequence, as defined below. Thus, the term "encoding two or more proteins or peptides" may mean, without being limited thereto, that the bi- or even multicistronic nucleic acid, may encode e.g. at least two, three, four, five, six or more (preferably different) proteins or peptides and/or proteins or peptides or their fragments or variants within the definitions provided herein. More preferably, without being limited thereto, the bi- or even multicistronic nucleic acid, may encode, for example, at least two, three, four, five, six or more (preferably different) proteins or peptides as defined herein or their fragments or variants as defined herein. In this context, a so-called IRES (internal ribosomal entry site) sequence as defined above can function as a sole ribosome binding site, but it can also serve to provide a bi- or even
multicistronic nucleic acid as defined above, which encodes several proteins or peptides which are to be translated by the ribosomes independently of one another. Examples of IRES sequences, which can be used according to the invention, are those from picornaviruses (e.g. FMDV), pestiviruses (CFFV), polioviruses (PV), encephalomyocarditis viruses (ECMV), foot and mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), mouse leukoma virus (MLV), simian immunodeficiency viruses (SIV) or cricket paralysis viruses (CrPV).
According to a further embodiment, the at least one coding sequence of the nucleic acid sequence according to the invention may encode at least two, three, four, five, six, seven, eight and more proteins or peptides (or fragments and derivatives thereof) as defined herein linked with or without an amino acid linker sequence, wherein said linker sequence can comprise rigid linkers, flexible linkers, cleavable linkers (e.g., self-cleaving peptides) or a combination thereof. Therein, the proteins or peptides may be identical or different or a combination thereof. Particular proteins or peptides combinations can be encoded by said nucleic acid encoding at least two proteins or peptides as explained herein (also referred to herein as 'multi-antigen- constructs/nucleic acid'). It has to be noted that in the context of the invention, certain combinations of coding sequences (e.g., comprising at least two different proteins) may be generated by any combination of mono-, bi-, and multicistronic nucleic acids and/or multi-antigen-constructs/nucleic acid to obtain a poly- or even multivalent nucleic acid mixture.
In particular preferred aspects, the encoded peptides or proteins are selected from human, viral, bacterial, protozoan proteins or peptides. a) Therapeutically active proteins
In the context of the present invention, therapeutically active proteins or peptides may be encoded by the nucleic acid comprised in the nanoparticle of the invention. Therapeutically active proteins are defined herein as proteins which have an effect on healing, prevent prophylactically or treat therapeutically a disease, preferably as defined herein, or are proteins of which an individual is in need of, e.g. a native or modified native protein which individual's organism does not produce, or only produces in insufficient quantities. These may be selected from any naturally occurring or synthetically designed recombinant or isolated protein known to a skilled person. Without being restricted thereto, therapeutically active proteins may comprise proteins capable of stimulating or inhibiting the signal transduction in the cell, e.g. cytokines, lymphokines, monokines, growth factors, receptors, signal transduction molecules, transcription factors, etc.; anticoagulants; antithrombins; antiallergic proteins; apoptotic factors or apoptosis related proteins, therapeutic active enzymes and any protein connected with any acquired disease or any hereditary disease. b) Antigens
The nucleic acid may alternatively encode an antigen. According to the present invention, the term "antigen" refers to a substance which is recognised by the immune system and is capable of triggering an antigen-specific immune response, e.g. by formation of antibodies or antigen- specific T-cells as part of an adaptive immune response. In this context, an antigenic epitope, fragment or peptide of a protein means particularly B cell and T cell epitopes which may be recognized by B cells, antibodies or T cells, respectively. In the context of the present invention, the antigen encoded by the nucleic acid typically represent any antigen, antigenic epitope or antigenic peptide falling under the above definition, and is preferably a protein and peptide antigen, e.g. a tumour antigen, allergenic antigen, autoimmune self-antigen, pathogenic antigen, etc. In particular, the antigen may be one derived from another organism that the host organism (e.g. a human subject) itself, such as a viral antigen, a bacterial antigen, a fungal antigen, a protozooal antigen, an animal antigen, an allergenic antigen etc. Allergenic antigens, also referred to as allergy antigens or allergens, are typically antigens which may cause an allergy in a human subject.
Alternatively, the antigen as encoded by the nucleic acid may be derived from the host itself. Examples for such antigens include tumour antigens, self-antigens or auto-antigens, such as auto- immune self-antigens, but also (non-self) antigens as defined herein which have originally been derived from cells outside the host organism, but which have been fragmented or degraded inside the host organism, tissue or cell, e.g. by protease degradation or other types of metabolism.
One class of antigens also preferred in the context of the present invention is that of tumour antigens. Among the preferred tumour antigens are those that are located on the surface of a tumour cell. Tumour antigens may also represent proteins which are overexpressed in tumour cells compared to a normal cell. Furthermore, tumour antigens also include antigens expressed in cells which are not, or which were originally not, themselves tumour cells but associated with a tumour. For example, antigens which are connected with formation or reformation of tumour- supplying blood vessels, in particular those which are associated with neovascularisation, such growth factors like VEGF or bFGF, are also of interest. Antigens associated with a tumour also include antigens from cells or tissues typically embedding the tumour. Furthermore, certain other proteins or peptides may be (over)expressed and occur in increased concentrations in the body fluids of patients that have developed a tumour. These substances are also referred to as tumour antigens or tumour-associated antigens even though they are, strictly speaking, not antigens in that they do not induce an immune response.
Tumour antigens may be divided further into tumour-specific antigens (TSAs) and tumour- associated antigens (TAAs). TSAs can only be presented by tumour cells and not by healthy cells. They typically result from a tumour-specific mutation. TAAs, which are more common, are usually produced by both tumour and healthy cells. These antigens are recognised by the immune system and the antigen-presenting cell can be destroyed by cytotoxic T cells. Additionally, tumour antigens can also occur on the surface of the tumour in the form of, e.g., a mutated receptor. In this case, they can also be recognised by antibodies. If the encoded antigen is an allergen, such antigen may be selected from antigens of any source, such as from animals, plants, molds, fungi, bacteria etc. Plant-derived allergens may, for example, be allergens from pollen. Again, the nucleic acid incorporated in the nanoparticle may encode the native antigen or a fragment or epitope thereof. c) Antibodies According to a further embodiment, the nucleic acid compound encodes an antibody or an antibody fragment. The antibody or a fragment thereof is selected from the group consisting of (i) a single-chain antibody, (ii) a single-chain antibody fragment, (iii) a multiple-chain antibody, and (iv) a multiple-chain antibody fragment.
In general, an antibody consists of a light chain and a heavy chain both having variable and constant domains. The light chain consists of an N-terminal variable domain, VL, and a C-terminal constant domain, CL. In contrast, the heavy chain of the IgG antibody, for example, is comprised of an N-terminal variable domain, VH, and three constant domains, CHI, CH2 und CH3.
In one of the preferred embodiments, the antibody is selected from full-length antibodies. Such an antibody may be any recombinantly produced or naturally occurring antibody, in particular an antibody suitable for therapeutic, diagnostic or scientific purposes, or an antibody which is associated with a disease, such as an immunological disease or cancer. The term
"antibody" is used in its broadest sense and specifically covers monoclonal and polyclonal antibodies (including agonist, antagonist, and blocking or neutralising antibodies) and antibody species with polyepitopic specificity. The antibody may belong to any class of antibodies, such as IgM, IgD, IgG, IgA and IgE antibodies. Moreover, the antibody may resemble an antibody generated by immunisation in a host organism, or a recombinantly engineered version thereof, a chimeric antibody, a human antibody, a humanised antibody, a bispecific antibody, an intrabody. Moreover, the nucleic acid compound may also encode an antibody fragment, variant, adduct or derivative of an antibody, such as single-chain variable fragment, a diabody or a triabody. The antibody fragment is preferably selected from Fab, Fab', F(ab') 2, Fc, Facb, pFc', Fd and Fv fragments of the aforementioned types of antibodies. In general, antibody fragments are known in the art. For example, a Fab ("fragment, antigen binding") fragment is composed of one constant and one variable domain of each of the heavy and the light chain. The two variable domains bind the epitope on specific antigens. The two chains are connected via a disulfide linkage. A scFv ("single chain variable fragment") fragment, for example, typically consists of the variable domains of the light and heavy chains. The domains are linked by an artificial linkage, in general a polypeptide linkage such as a peptide composed of 15-25 glycine, proline and/or serine residues.
In one embodiment, the biologically active cargo material comprises a combination of at least two distinct RNAs, wherein one RNA encodes a heavy chain of an antibody or a fragment thereof and another RNA encodes the corresponding light chain of the antibody or a fragment thereof.
In a further embodiment, the biologically active cargo material comprises a combination of at least two distinct RNAs, wherein one RNA encodes a heavy chain variable region of an antibody or a fragment thereof and another RNA encodes the corresponding light chain variable region of the antibody or a fragment thereof. Moreover, it is preferred that the different chains of the antibody or antibody fragment are encoded by a multicistronic nucleic acid, also referred to as polycistronic nucleic acid.
Alternatively, the different strains of the antibody or antibody fragment are encoded by several monocistronic nucleic acids. As mentioned, these nucleic acids may be used as cargo in combination within one composition, or nanoparticle, according to the invention. According to a further embodiment, the present invention comprises the use of at least one nucleic acid molecule for the preparation of a biologically active cargo material. If more than one nucleic acid molecule is used, the complexed nucleic acid molecules may be different, i.e. thereby forming a mixture of at least two distinct (complexed) nucleic acid molecules.
In one embodiment, the biologically active cargo material comprises (i) a nucleic acid molecule encoding a CRISPR related protein; and/or
(ii) one or more guide RNA(s) sequence(s). The term "CRISPR related protein" includes but is not limited to CAS9 (CRISPR-Associated Protein 9), CSY4, dCAS9, and dCAS9-effector domain (activator and/or inhibitor domain) fusion proteins. The CRISPR related protein can be from any number of species including but not limited to Streptococcus pyogenes, Listeria innocua, and Streptococcus thermophilus.
The term "guide RNA (gRNA)", also referred to as "artificial guide RNA", "single guide RNA", "small guide RNA" or "sgRNA", describes an RNA including a typically 20-25 nucleotides long sequence that is complementary to one strand of the 5'UTR of the gene of interest upstream of the transcription start site. A description of sgRNA design can be found in e.g. Mali et al., 2013, Science 339:823-826. The artificial sgRNA targets a gene of interest, directing the CRISPR related protein encoded by the artificial polynucleotide to interact with the gene of interest, e.g., a gene where modulation of transcription is desired. The gene of interest is selected depending on the application.
In one embodiment, a single nucleic acid molecule of the invention comprised in the composition or in the nanoparticle(s) of the invention comprises a single nucleic acid molecule encoding said CRISPR related protein and simultaneously said guide RNA(s).
In a further embodiment, the biologically active cargo material comprises a combination of more than one nucleic acid molecule. In another embodiment, more than one nucleic acid molecules of the invention comprise said nucleic acid molecule encoding a CRISPR related protein and said guide RNA(s). In this case, the biologically active cargo material comprises two distinct RNA which express both a Cas9 protein and the target-specific gRNA. siRNA
In a further preferred embodiment, the nucleic acid compound incorporated in the composition or nanoparticle of the invention is in the form of dsRNA, preferably siRNA. A dsRNA, or a siRNA, is of interest particularly in connection with the phenomenon of RNA interference. The in vitro technique of RNA interference (RNAi) is based on double-stranded RNA molecules (dsRNA) which trigger the sequence-specific suppression of gene expression (Zamore (2001) Nat. Struct. Biol. 9: 746-750; Sharp (2001) Genes Dev. 5:485-490: Hannon (2002) Nature 41: 244- 251). In the transfection of mammalian cells with long dsRNA, the activation of protein kinase R and RnaseL brings about unspecific effects, such as, for example, an interferon response (Stark et al. (1998) Annu. Rev. Biochem. 67: 227-264; He and Katze (2002) Viral Immunol. 15: 95-119). These unspecific effects are avoided when shorter, for example 21- to 23-mer, so-called siRNA (small interfering RNA), is used, because unspecific effects are not triggered by siRNA that is shorter than 30 bp (Elbashir et al. (2001) Nature 411: 494-498). The nucleic acid may, for example, be a double-stranded RNA (dsRNA) having a length from about 17 to about 29 base pairs, and preferably from about 19 to about 25 base pairs. The dsRNA is preferably at least 90 %, more preferably at least 95 %, such as 100 %, (regarding the nucleotides of a dsRNA) complementary to a section of the nucleic acid sequence of a
therapeutically relevant protein or antigen as described hereinbefore, either a coding or a non- coding section, preferably a coding section. 90 % complementary means that, with a length of a dsRNA of, for example, 20 nucleotides, this contains not more than 2 nucleotides without complementarity with the corresponding section of the mRNA encoding the respective protein. Also preferred is a double-stranded RNA whose sequence is wholly complementary with a section of the nucleic acid of a therapeutically relevant protein or antigen described hereinbefore.
In one embodiment, the dsRNA has the general structure 5'-(Ni7-29)-3', and preferably the general structure 5'-(Nig-25)-3', or 5'-(Nig-24)-3', or 5'-(N2i-23)-3', respectively, wherein each N is a nucleotide, and wherein the nucleotide sequence is complementary to a section of the mRNA that corresponds to a therapeutically relevant protein or antigen described hereinbefore. In principle, all the sections having a length of from 17 to 29, preferably from 19 to 25, base pairs that occur in the coding region of the mRNA can serve as target sequence for a dsRNA herein. Equally, dsRNAs used as nucleic acid 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 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 nucleic acid 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.
Immunostimulatory nucleic acids a) Immunostimulatory CpG nucleic acids
According to another embodiment, the nucleic acid incorporated in the composition or nanoparticle of the invention is an immunostimulatory CpG nucleic acid, in particular a CpG-RNA or a CpG-DNA, which preferably induces an innate immune response. Examples of potentially suitable immunostimulatory CpG nucleic acids include, without limitation, single-stranded CpG- DNA (ss CpG-DNA), double-stranded CpG-DNA (dsDNA), single-stranded CpG-RNA (ss CpG-RNA), and double-stranded CpG-RNA (ds CpG-RNA). Preferably, the CpG nucleic acid is a CpG-RNA, in particular a single-stranded CpG-RNA (ss CpG-RNA). That preferred length of the CpG nucleic acid in terms of nucleotides or base pairs is similar to that preferred for siRNA, as described above. Preferably, the CpG motifs are unmethylated. b) Immunostimulatory RNA (isRNA)
According to a further alternative, the nucleic acid incorporated as biologically active cargo material in the composition or nanoparticle of the invention may be in the form of a of an immunostimulatory RNA (isRNA), which preferably elicits an innate immune response.
Such isRNA may be a double-stranded RNA, a single-stranded RNA, or a partially double- stranded RNA, or a short RNA oligonucleotide. In one of the preferred embodiments, it is a single- stranded RNA.
Moreover, the isRNA may be circular or linear. In one of the preferred embodiments, a linear isRNA is used, such as a linear single-stranded RNA, or a long single-stranded RNA.
Moreover, the isRNA may be a coding or non-coding RNA. According to one of the preferred embodiments, a non-coding RNA is used as isRNA, such as a non-coding single-stranded RNA, a non-coding linear RNA, a non-coding linear single-stranded RNA, or a non-coding long linear single-stranded RNA.
According to one further preferred embodiment, the isRNA carries a triphosphate at its 5'- end, as is the case for in vitro transcribed RNA. This preference applies to all aforementioned types of linear isRNA.
Again, the isRNA used as biologically active cargo material according to the invention may be selected from any type or class of RNA, whether naturally occurring or synthetic, which is capable of inducing an innate immune response, and/or which is capable of enhancing or supporting an adaptive immune response induced by an antigen.
In this context, an immune response may occur in various ways. A substantial factor for a suitable adaptive immune response is the stimulation of certain T-cell sub-populations. T- lymphocytes are typically divided into two sub-populations, the T-helper 1 (Thl) cells and the T- helper 2 (Th2) cells, with which the immune system is capable of destroying intracellular (Thl) and extracellular (Th2) pathogens, such as antigens. The two Th cell populations differ in the pattern of the effector proteins (cytokines) produced by them. Thus, Thl cells assist the cellular immune response by activation of macrophages and cytotoxic T-cells. Th2 cells, on the other hand, promote the humoral immune response by stimulation of B-cells for conversion into plasma cells and by formation of antibodies (e.g. against antigens). The Thl/Th2 ratio is therefore of great importance in the induction and maintenance of an adaptive immune response.
In the context of the present invention, it is preferred that the Thl/Th2 ratio of the adaptive immune response is shifted towards the cellular response (Thl response), i.e. a cellular immune response is induced or enhanced. For example, the innate immune system which may support an adaptive immune response may be activated by ligands of toll-like receptors (TLRs). TLRs are a family of highly conserved pattern recognition receptor (PRR) polypeptides that recognise pathogen-associated molecular patterns (PAMPs) and play a critical role in innate immunity in mammals. Currently, at least thirteen family members have been identified and designated as toll- like receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 and TLR13. Furthermore, a number of specific TLR ligands have been identified. It was found that unmethylated bacterial DNA and synthetic analogs thereof (CpG DNA) are ligands for TLR9 (Hemmi H et al. (2000) Nature 408:740-5; Bauer S et al. (2001) Proc NatlAcadSci USA 98, 9237- 42). Furthermore, it has been reported that 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 stimulate TLR3, TLR7, or TLR8, or intracellular receptors such as RIG-I, MDA-5 and others. Lipford et al. determined certain G,U-containing oligoribonucleotides as immunostimulatory by acting via TLR7 and TLR8 (see WO 03/086280). The immunostimulatory G,U-containing
oligoribonucleotides described by Lipford et al. were believed to be derivable from RNA sources including ribosomal RNA, transfer RNA, messenger RNA, and viral RNA.
The isRNA used in the context of the invention may thus comprise any RNA sequence known to be immunostimulatory, including, without being limited thereto, RNA sequences representing and/or encoding ligands of TLRs, such as the murine family members TLR1 to TLR13, or more preferably selected from human family members TLR1 to TLR10, in particular TLR7 or TLR8; or ligands for intracellular receptors for RNA such as RIG-I or MDA-5 (see e.g. Meylan, E., Tschopp, J. (2006) : Toll-like receptors and RNA helicases: Two parallel ways to trigger antiviral responses. Mol. Cell 22, 561-569).
Without being limited thereto, the isRNA may include ribosomal RNA (rRNA), transfer RNA (tRNA), messenger RNA (mRNA), and viral RNA (vRNA). It may comprise up to about 5000 nucleotides, such as from about 5 to about 5000 nucleotides, or from about 5 to about 1000, or from about 500 to about 5000, or from about 5 to about 500, or from about 5 to about 250, or from about 5 to about 100, or from about 5 to about 50 or or from about 5 to about 30
nucleotides, respectively. According to a further preferred aspect of this embodiment, the isRNA comprises or consists of a nucleic acid of formula (III) or (IV) :
(NuGiXmGnNv)a (formula III) wherein:
G is guanosine (guanine), uridine (uracil) or an analogue of guanosine (guanine) or uridine (uracil), preferably guanosine (guanine) or an analogue thereof;
X is guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine), or an analogue of these nucleotides (nucleosides), preferably uridine (uracil) or an analogue thereof;
N is a nucleic acid sequence having a length of about 4 to 50, preferably of about 4 to 40, more preferably of about 4 to 30 or 4 to 20 nucleic acids, each N independently being selected from guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of these nucleotides (nucleosides);
a is an integer from 1 to 20, preferably from 1 to 15, most preferably from 1 to 10;
1 is an integer from 1 to 40,
wherein if 1 = 1, G is guanosine (guanine) or an analogue thereof, and if 1 > 1, at least 50 % of these nucleotides (nucleosides) are guanosine (guanine) or an analogue thereof;
m is an integer and is at least 3; wherein if m = 3, X is uridine (uracil) or an analogue thereof, and if m > 3, at least 3 successive uridines (uracils) or analogues of uridine (uracil) occur; n is an integer from 1 to 40, wherein if n = 1, G is guanosine (guanine) or an analogue thereof, and if n > 1, at least 50 % of these nucleotides (nucleosides) are guanosine (guanine) or an analogue thereof;
u, v are independently from each other an integer from 0 to 50, wherein preferably if u = 0, v > 1, or ifv = 0, u > 1;
wherein the nucleic acid molecule of formula (V) has a length of at least 50 nucleotides, preferably of at least 100 nucleotides, more preferably of at least 150 nucleotides, even more preferably of at least 200 nucleotides and most preferably of at least 250 nucleotides;
(NuCiXmC„Nv)a (formula IV) wherein:
C is cytidine (cytosine), uridine (uracil) or an analogue of cytidine (cytosine) or uridine (uracil), preferably cytidine (cytosine) or an analogue thereof;
X is guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of the above-mentioned nucleotides (nucleosides), preferably uridine (uracil) or an analogue thereof; N is each a nucleic acid sequence having a length of about 4 to 50, preferably of about 4 to 40, more preferably of about 4 to 30 or 4 to 20 nucleic acids, each N being independently selected from guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of these nucleotides (nucleosides);
a is an integer from 1 to 20, preferably from 1 to 15, most preferably from 1 to 10;
1 is an integer from 1 to 40, wherein if 1 = 1, C is cytidine (cytosine) or an analogue thereof, and if 1 > 1, at least 50 % of these nucleotides (nucleosides) are cytidine (cytosine) or an analogue thereof;
m is an integer and is at least 3; wherein if m = 3, X is uridine (uracil) or an analogue thereof, and if m > 3, at least 3 successive uridines (uracils) or analogues of uridine (uracil) occur; n is an integer from 1 to 40, wherein if n = 1, C is cytidine (cytosine) or an analogue thereof, and if n > 1, at least 50 % of these nucleotides (nucleosides) are cytidine (cytosine) or an analogue thereof;
u, v are independently from each other an integer from 0 to 50, wherein preferably if u = 0, v > l, or ifv = 0, u > l;
wherein the nucleic acid molecule of formula (IV) according to the invention has a length of at least 50 nucleotides, preferably of at least 100 nucleotides, more preferably of at least 150 nucleotides, even more preferably of at least 200 nucleotides and most preferably of at least 250 nucleotides. For formula (IV), any of the definitions given above for elements N (i.e. Nu and Nv) and X
(Xm), particularly the core structure as defined above, as well as for integers a, 1, m, n, u and v, similarly apply to elements of formula (III) correspondingly, wherein in formula (IV) the core structure is defined by CiXmCn. The definition of bordering elements Nu and Nv is identical to the definitions given above for Nu and Nv. According to a very particularly preferred aspect of this embodiment, the nucleic acid molecule according to formula (III) may be selected from e.g. any of the following sequences:
UAGCGAAGCUCUUGGACCUAGGUUUUUUUUUUUUUUUGGGUGCGUUCCUAGAAGUACACG
(SEQ ID NO: 1)
UAGCGAAGCUCUUGGACCUAGGUUUUUUUUUUUUUUUGGGUGCGUUCCUAGAAGUACACGAUC GCUUCGAGAACCUGGAUCCAAAAAAAAAAAAAAACCCACGCAAGGAUCUUCAUGUGC (SEQ ID NO: 2)
GGGAGAAAGCUCAAGCUUGGAGCAAUGCCCGCACAUUGAGGAAACCGAGUUGCAUAUCUCAGAG UAUUGGCCCCCGUGUAGGUUAUUCUUGACAGACAGUGGAGCUUAUUCACUCCCAGGAUCCGAGUCGCA UACUACGGUACUGGUGACAGACCUAGGUCGUCAGUUGACCAGUCCGCCACUAGACGUGAGUCCGUCAA AGCAGUUAGAUGUUACACUCUAUUAGAUC (SEQ ID NO: 3)
GGGAGAAAGCUCAAGCUUGGAGCAAUGCCCGCACAUUGAGGAAACCGAGUUGCAUAUCUCAGAG UAUUGGCCCCCGUGUAGGUUAUUCUUGACAGACAGUGGAGCUUAUUCACUCCCAGGAUCCGAGUCGCA UACUACGGUACUGGUGACAGACCUAGGUCGUCAGUUGACCAGUCCGCCACUAGACGUGAGUCCGUCAA AGCAGUUAGAUGUUACACUCUAUUAGAUCUCGGAUUACAGCUGGAAGGAGCAGGAGUAGUGUUCUUG CUCUAAGUACCGAGUGUGCCCAAUACCCGAUCAGCUUAUUAACGAACGGCUCCUCCUCUUAGACUGCA GCGUAAGUGCGGAAUCUGGGGAUCAAAUUACUGACUGCCUGGAUUACCCUCGGACAUAUAACCUUGUA GCACGCUGUUGCUGUAUAGGUGACCAACGCCCACUCGAGUAGACCAGCUCUCUUAGUCCGGACAAUGA UAGGAGGCGCGGUCAAUCUACUUCUGGCUAGUUAAGAAUAGGCUGCACCGACCUCUAUAAGUAGCGUG UCCUCUAG (SEQ ID NO: 4)
GGGAGAAAGCUCAAGCUUGGAGCAAUGCCCGCACAUUGAGGAAACCGAGUUGCAUAUCUCAGAG UAUUGGCCCCCGUGUAGGUUAUUCUUGACAGACAGUGGAGCUUAUUCACUCCCAGGAUCCGAGUCGCA UACUACGGUACUGGUGACAGACCUAGGUCGUCAGUUGACCAGUCCGCCACUAGACGUGAGUCCGUCAA AGCAGUUAGAUGUUACACUCUAUUAGAUCUCGGAUUACAGCUGGAAGGAGCAGGAGUAGUGUUCUUG CUCUAAGUACCGAGUGUGCCCAAUACCCGAUCAGCUUAUUAACGAACGGCUCCUCCUCUUAGACUGCA GCGUAAGUGCGGAAUCUGGGGAUCAAAUUACUGACUGCCUGGAUUACCCUCGGACAUAUAACCUUGUA GCACGCUGUUGCUGUAUAGGUGACCAACGCCCACUCGAGUAGACCAGCUCUCUUAGUCCGGACAAUGA UAGGAGGCGCGGUCAAUCUACUUCUGGCUAGUUAAGAAUAGGCUGCACCGACCUCUAUAAGUAGCGUG UCCUCUAGAGCUACGCAGGUUCGCAAUAAAAGCGUUGAUUAGUGUGCAUAGAACAGACCUCUUAUUC GGUGAAACGCCAGAAUGCUAAAUUCCAAUAACUCUUCCCAAAACGCGUACGGCCGAAGACGCGCGCUU AUCUUGUGUACGUUCUCGCACAUGGAAGAAUCAGCGGGCAUGGUGGUAGGGCAAUAGGGGAGCUGGG UAGCAGCGAAAAAGGGCCCCUGCGCACGUAGCUUCGCUGUUCGUCUGAAACAACCCGGCAUCCGUUGU AGCGAUCCCGUUAUCAGUGUUAUUCUUGUGCGCACUAAGAUUCAUGGUGUAGUCGACAAUAACAGCG UCUUGGCAGAUUCUGGUCACGUGCCCUAUGCCCGGGCUUGUGCCUCUCAGGUGCACAGCGAUACUUAA AGCCUUCAAGGUACUCGACGUGGGUACCGAUUCGUGACACUUCCUAAGAUUAUUCCACUGUGUUAGCC CCGCACCGCCGACCUAAACUGGUCCAAUGUAUACGCAUUCGCUGAGCGGAUCGAUAAUAAAAGCUUGA AUU (SEQ ID NO: 5)
GGGAGAAAGCUCAAGCUUAUCCAAGUAGGCUGGUCACCUGUACAACGUAGCCGGUAUUUUUUU UUUUUUUUUUUUUUUGACCGUCUCAAGGUCCAAGUUAGUCUGCCUAUAAAGGUGCGGAUCCACAGCU GAUGAAAGACUUGUGCGGUACGGUUAAUCUCCCCUUUUUUUUUUUUUUUUUUUUUAGUAAAUGCGU CUACUGAAUCCAGCGAUGAUGCUGGCCCAGAUC (SEQ ID NO: 6)
GGGAGAAAGCUCAAGCUUAUCCAAGUAGGCUGGUCACCUGUACAACGUAGCCGGUAUUUUUUU UUUUUUUUUUUUUUUGACCGUCUCAAGGUCCAAGUUAGUCUGCCUAUAAAGGUGCGGAUCCACAGCU GAUGAAAGACUUGUGCGGUACGGUUAAUCUCCCCUUUUUUUUUUUUUUUUUUUUUAGUAAAUGCGU CUACUGAAUCCAGCGAUGAUGCUGGCCCAGAUCUUCGACCACAAGUGCAUAUAGUAGUCAUCGAGGGU CGCCUUUUUUUUUUUUUUUUUUUUUUUGGCCCAGUUCUGAGACUUCGCUAGAGACUACAGUUACAGC UGCAGUAGUAACCACUGCGGCUAUUGCAGGAAAUCCCGUUCAGGUUUUUUUUUUUUUUUUUUUUUCC GCUCACUAUGAUUAAGAACCAGGUGGAGUGUCACUGCUCUCGAGGUCUCACGAGAGCGCUCGAUACAG UCCUUGGAAGAAUCUUUUUUUUUUUUUUUUUUUUUUGUGCGACGAUCACAGAGAACUUCUAUUCAU GCAGGUCUGCUCUAG (SEQ ID NO: 7)
GGGAGAAAGCUCAAGCUUAUCCAAGUAGGCUGGUCACCUGUACAACGUAGCCGGUAUUUUUUU UUUUUUUUUUUUUUUGACCGUCUCAAGGUCCAAGUUAGUCUGCCUAUAAAGGUGCGGAUCCACAGCU GAUGAAAGACUUGUGCGGUACGGUUAAUCUCCCCUUUUUUUUUUUUUUUUUUUUUAGUAAAUGCGU CUACUGAAUCCAGCGAUGAUGCUGGCCCAGAUCUUCGACCACAAGUGCAUAUAGUAGUCAUCGAGGGU CGCCUUUUUUUUUUUUUUUUUUUUUUUGGCCCAGUUCUGAGACUUCGCUAGAGACUACAGUUACAGC UGCAGUAGUAACCACUGCGGCUAUUGCAGGAAAUCCCGUUCAGGUUUUUUUUUUUUUUUUUUUUUCC GCUCACUAUGAUUAAGAACCAGGUGGAGUGUCACUGCUCUCGAGGUCUCACGAGAGCGCUCGAUACAG UCCUUGGAAGAAUCUUUUUUUUUUUUUUUUUUUUUUGUGCGACGAUCACAGAGAACUUCUAUUCAU GCAGGUCUGCUCUAGAACGAACUGACCUGACGCCUGAACUUAUGAGCGUGCGUAUUUUUUUUUUUUU UUUUUUUUUUCCUCCCAACAAAUGUCGAUCAAUAGCUGGGCUGUUGGAGACGCGUCAGCAAAUGCCG UGGCUCCAUAGGACGUGUAGACUUCUAUUUUUUUUUUUUUUUUUUUUUCCCGGGACCACAAAUAAUA UUCUUGCUUGGUUGGGCGCAAGGGCCCCGUAUCAGGUCAUAAACGGGUACAUGUUGCACAGGCUCCUU UUUUUUUUUUUUUUUUUUUUUCGCUGAGUUAUUCCGGUCUCAAAAGACGGCAGACGUCAGUCGACAA CACGGUCUAAAGCAGUGCUACAAUCUGCCGUGUUCGUGUUUUUUUUUUUUUUUUUUUUGUGAACCUA CACGGCGUGCACUGUAGUUCGCAAUUCAUAGGGUACCGGCUCAGAGUUAUGCCUUGGUUGAAAACUGC CCAGCAUACUUUUUUUUUUUUUUUUUUUUCAUAUUCCCAUGCUAAGCAAGGGAUGCCGCGAGUCAUG UUAAGCUUGAAUU (SEQ ID NO: 8) According to another very particularly preferred embodiment, the nucleic acid molecule according to formula (IV) may be selected from e.g. any of the following sequences:
UAGCGAAGCUCUUGGACCUACCUUUUUUUUUUUUUUCCCUGCGUUCCUAGAAGUACACG
(SEQ ID NO: 9) or UAGCGAAGCUCUUGGACCUACCUUUUUUUUUUUUUUUCCCUGCGUUCCUAGAAGUACACGAUC GCUUCGAGAACCUGGAUGGAAAAAAAAAAAAAAAGGGACGCAAGGAUCUUCAUGUGC (SEQ ID NO: 10) In a further embodiment, the nucleic acid compound used as biologically active cargo material according to the present invention is in the form of a chemically modified nucleic acid, or is a stabilised nucleic acid, preferably a stabilised RNA or DNA, such as a RNA that is essentially resistant to in vivo degradation by an exo- or endonuclease.
Chemical modifications:
The terms "modification(s)", "chemical modification(s)", "modified" and the like with respect to a nucleic acid, as used herein, may refer to chemical modifications comprising backbone modifications as well as sugar modifications or base modifications. The respective product of the modification may, for example, be termed a "modified nucleic acid" or a
"chemically modified nucleic acid".
A backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides contained in a nucleic acid compound, preferably an mRNA, are chemically modified. A sugar modification is a chemical modification of the sugar of the nucleotides of the nucleic acid. Furthermore, a base modification in connection with the present invention is a chemical modification of the base moiety of the nucleotides of the artificial nucleic acid, preferably an mRNA. In this context, nucleotide analogues or modifications are preferably selected from those nucleotide analogues which are applicable for transcription and/or translation.
Sugar Modifications:
As said, the nucleosides and nucleotides can be modified in the sugar moiety. For example, the 2'-hydroxyl group (OH) can be modified or replaced with a number of different "oxy" or "deoxy" substituents. Examples of "oxy" -2'-hydroxyl group modifications include, but are not limited to, alkoxy or aryloxy (-OR, e.g., R = H, alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar); polyethyleneglycols (PEG), -0(CH2CH20)nCH2CH2OR; "locked" nucleic acids (LNA) in which the 2'- hydroxyl is connected, e.g., by a methylene bridge, to the 4'-carbon of the same ribose sugar; and amino groups (-O-amino, wherein the amino group, e.g., NRR, can be alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, or diheteroaryl amino, ethylene diamine, polyamino) or aminoalkoxy.
"Deoxy" modifications include hydrogen, amino (e.g. N¾; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); or the amino group can be attached to the sugar through a linker, wherein the linker comprises one or more of the atoms C, N, and 0. The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, an artificial nucleic acid, preferably an mRNA, can include nucleotides containing, for instance, arabinose as the sugar. Backbone Modifications:
The phosphate groups of the backbone of the nucleic acid compound can be modified by replacing one or more of the oxygen atoms with a different substituent. Further, the modified nucleosides and nucleotides can include the full replacement of an unmodified phosphate moiety with a modified phosphate as described herein. Examples of modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters. Phosphorodithioates have both non-linking oxygens replaced by sulfur. The phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylene-phosphonates).
Base Modifications:
Optionally, the modification may relate to a nucleobase moiety of the nucleic acid compound. Examples of nucleobases found in a nucleic acid such as RNA include, but are not limited to, adenine, guanine, cytosine and uracil. For example, the nucleosides and nucleotides described herein can be chemically modified on the major groove face. In some embodiments, the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
In particularly preferred embodiments of the present invention, the base modifications are selected from 2-amino-6-chloropurineriboside-5'-triphosphate, 2-aminopurine-riboside-5'- triphosphate; 2-aminoadenosine-5'-triphosphate, 2'-amino-2'-deoxycytidine-triphosphate, 2- thiocytidine-5'-triphosphate, 2-thiouridine-5'-triphosphate, 2'-fluorothymidine-5'-triphosphate, 2'-0-methyl inosine-5'-triphosphate 4-thiouridine-5'-triphosphate, 5-aminoallylcytidine-5'- triphosphate, 5-aminoallyluridine-5'-triphosphate, 5-bromocytidine-5'-triphosphate, 5- bromouridine-5 '-triphosphate, 5-bromo-2'-deoxycytidine-5'-triphosphate, 5-bromo-2'- deoxyuridine-5'-triphosphate, 5-iodocytidine-5'-triphosphate, 5-iodo-2'-deoxycytidine-5'- triphosphate, 5-iodouridine-5 '-triphosphate, 5-iodo-2'-deoxyuridine-5'-triphosphate, 5- methylcytidine-5'-triphosphate, 5-methyluridine-5'-triphosphate, 5-propynyl-2'-deoxycytidine- 5'-triphosphate, 5-propynyl-2'-deoxyuridine-5'-triphosphate, 6-azacytidine-5'-triphosphate, 6- azauridine-5'-triphosphate, 6-chloropurineriboside-5'-triphosphate, 7-deazaadenosine-5'- triphosphate, 7-deazaguanosine-5'-triphosphate, 8-azaadenosine-5'-triphosphate, 8- azidoadenosine-5'-triphosphate, benzimidazole-riboside-5'-triphosphate, Nl-methyladenosine- 5'-triphosphate, Nl-methylguanosine-5'-triphosphate, N6-methyladenosine-5'-triphosphate, 06- methylguanosine-5'-triphosphate, pseudouridine-5'-triphosphate, or puromycin-5'-triphosphate, xanthosine-5'-triphosphate. Particular preference is given to 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.
In some embodiments, modified nucleosides include pyridin-4-one ribonucleoside, 5-aza- uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5- hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5- propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl- pseudouridine, 5-taurinomethyl-2-thio-uridine, l-taurinomethyl-4-thio-uridine, 5-methyl-uridine,
1- methyl-pseudouridine, 4-thio-l-methyl-pseudouridine, 2-thio-l-methyl-pseudouridine, 1- methyl-l-deaza-pseudouridine, 2-thio-l-methyl-l-deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine,
2- methoxy-4-thio-uridine, 4-methoxy-pseudouridine, and 4-methoxy-2-thio-pseudouridine.
In some embodiments, modified nucleosides include 5-aza-cytidine, pseudoisocytidine, 3- methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5- hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-l- methyl-pseudoisocytidine, 4-thio-l-methyl-l-deaza-pseudoisocytidine, 1-methyl-l-deaza- pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2- thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy- pseudoisocytidine, and 4-methoxy-l-methyl-pseudoisocytidine.
In other embodiments, modified nucleosides include 2-aminopurine, 2, 6-diaminopurine, 7- deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7- deaza-2,6-diaminopurine, 7-deaza-8-aza-2, 6-diaminopurine, 1-methyladenosine, N6- methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2- methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6- dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, and 2-methoxy-adenine. In other embodiments, modified nucleosides include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza- guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7- methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2- dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2- methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
In some embodiments, the nucleotide can be modified on the major groove face and can include replacing hydrogen on C-5 of uracil with a methyl group or a halo group.
In specific embodiments, a modified nucleoside is 5'-0-(l-thiophosphate)-adenosine, 5'-0- (l-thiophosphate)-cytidine, 5'-0-(l-thiophosphate)-guanosine, 5'-0-(l-thiophosphate)-uridine or 5'-0-(l-thiophosphate)-pseudouridine.
In further specific embodiments, a modified nucleic acid compound, preferably an mRNA, may comprise nucleoside modifications selected from 6-aza-cytidine, 2-thio-cytidine, a-thio- cytidine, pseudo-iso-cytidine, 5-aminoallyl-uridine, 5-iodo-uridine, Nl-methyl-pseudouridine, 5,6-dihydrouridine, a-thio-uridine, 4-thio-uridine, 6-aza-uridine, 5-hydroxy-uridine, deoxy- thymidine, 5-methyl-uridine, pyrrolo-cytidine, inosine, a-thio-guanosine, 6-methyl-guanosine, 5- methyl-cytdine, 8-oxo-guanosine, 7-deaza-guanosine, Nl-methyl-adenosine, 2-amino-6-chloro- purine, N6-methyl-2-amino-purine, pseudo-iso-cytidine, 6-chloro-purine, N6-methyl-adenosine, a-thio-adenosine, 8-azido-adenosine, 7-deaza-adenosine.
In one embodiment, the nucleic acid exhibits a lipid modification. Such a lipid-modified nucleic acid or RNA as defined herein typically further comprises at least one linker covalently linked with that nucleic acid or RNA, and at least one lipid covalently linked with the respective linker. Alternatively, the lipid-modified nucleic acid comprises at least one nucleic acid as defined herein and at least one (bifunctional) lipid covalently linked (without a linker) with that nucleic acid. According to a third alternative, the lipid-modified nucleic acid comprises an nucleic acid molecule as defined herein, at least one linker covalently linked with that RNA, 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. In this context, it is particularly preferred that the lipid modification is present at the terminal ends of a linear nucleic acid sequence.
According to another preferred embodiment of the invention, a modified nucleic acid sequence as defined herein, particularly a modified RNA as defined herein can be modified by the addition of a so-called '5' cap' structure, which preferably stabilizes the nucleic acid as described herein. A 5'-cap is an entity, typically a modified nucleotide entity, which generally "caps" the 5'- end of a mature RNA. A 5 '-cap may typically be formed by a modified nucleotide, particularly by a derivative of a guanine nucleotide. Preferably, the 5'-cap is linked to the 5'-terminus via a 5'-5'- triphosphate linkage. A 5'-cap may be methylated, e.g. m7GpppN, wherein N is the terminal 5' nucleotide of the nucleic acid carrying the 5'-cap, typically the 5'-end of an RNA. m7GpppN is the 5'-cap structure, which naturally occurs in RNA transcribed by polymerase II and is therefore preferably not considered as modification comprised in a modified RNA in this context.
Accordingly, a modified RNA sequence of the present invention may comprise a m7GpppN as 5'- cap, but additionally the modified RNA sequence typically comprises at least one further modification as defined herein.
Further examples of 5 'cap structures include glyceryl, inverted deoxy abasic residue (moiety), 4',5' methylene nucleotide, l-(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide, 1,5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4- dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide, 3'-3'-inverted nucleotide moiety, 3'-3'-inverted abasic moiety, 3'-2'-inverted nucleotide moiety, 3'-2'-inverted abasic moiety, 1,4-butanediol phosphate, 3'-phosphoramidate, hexylphosphate, aminohexyl phosphate, 3'-phosphate, 3'phosphorothioate, phosphorodithioate, or bridging or non-bridging
methylphosphonate moiety. These modified 5'-cap structures are regarded as at least one modification in this context.
Particularly preferred modified 5'-cap structures are capl (methylation of the ribose of the adjacent nucleotide of m7G), cap2 (additional methylation of the ribose of the 2nd nucleotide downstream of the m7G), cap3 (additional methylation of the ribose of the 3rd nucleotide downstream of the m7G), cap4 (methylation of the ribose of the 4th nucleotide downstream of the m7G), ARCA (anti-reverse cap analogue, modified ARCA (e.g. phosphothioate modified ARCA), inosine, Nl-methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2- amino-guanosine, LNA-guanosine, and 2-azido-guanosine. Accordingly, the RNA according to the invention preferably comprises a 5'-cap structure.
In a preferred embodiment, the 5'-cap structure is added co-transcriptionally using cap- analogues as defined herein in an RNA in vitro transcription reaction as defined herein. In another embodiment, the 5'-cap structure is added via enzymatic capping using capping enzymes (e.g. vaccinia virus capping enzymes).
Optionally, a nucleic acid may be selected which represents an mRNA that is essentially resistant to in vivo degradation by an exo- or endonucleases. Such stabilisation can be effected, for example, by chemically modifying the phosphates of the backbone. Sugar or base modifications may be additionally used. mRNA may also be stabilised against degradation by RNases by the addition of a so-called "5' cap" structure. Particular preference is given in this connection to an an G(5')ppp(5')G or a m7G(5')ppp(5')N as the 5'cap structures (N being A, G, C, or U). According to another example, the mRNA may exhibit a poly-A tail on the 3' terminus of typically about 10 to about 200 adenosine nucleotides, preferably of about 10 to about 100 adenosine nucleotides, or about 20 to about 100 adenosine nucleotides or even about 40 to about 80 adenosine nucleotides. According to a further example, the mRNA may have a poly-C tail on the 3' terminus of typically about 10 to about 200 cytosine nucleotides, preferably about 10 to about 100 cytosine nucleotides, or about 20 to about 70 cytosine nucleotides, or about 20 to about 60 or even about 10 to about 40 cytosine nucleotides.
According to another embodiment, the nucleic acid sequence of the present invention may be modified, and thus stabilized, by modifying the guanosine/cytosine (G/C) content of the nucleic acid sequence. In a particularly preferred embodiment of the present invention, the G/C content of the coding sequence of the nucleic acid sequence of the present invention is modified, particularly increased, compared to the G/C content of the coding sequence of the respective wild-type nucleic acid sequence, i.e. the unmodified nucleic acid. The amino acid sequence encoded by the nucleic acid is preferably not modified as compared to the amino acid sequence encoded by the respective wild-type nucleic acid. This modification of the nucleic acid sequence of the present invention is based on the fact that the sequence of any nucleic acid region, particularly the sequence of any RNA region to be translated is important for efficient translation of that nucleic acid, particularly of that RNA. Thus, the composition of the nucleic acid and the sequence of various nucleotides are important. In particular, in case of RNA, sequences having an increased G (guanosine)/C (cytosine) content are more stable than sequences having an increased A
(adenosine) /U (uracil) content. According to the invention, the codons of the nucleic acid are therefore varied compared to the respective wild-type nucleic acid, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides. In respect to the fact that several codons code for one and the same amino acid (so-called degeneration of the genetic code), the most favourable codons for the stability can be determined (so-called alternative codon usage). Depending on the amino acid to be encoded by the nucleic acid, there are various possibilities for modification of the nucleic acid sequence, compared to its wild-type sequence.
The following modifications may apply for RNA molecules, but may also be transferrable to DNA molecules: In the case of amino acids, which are encoded by codons, containing exclusively G or C nucleotides, no modification of the codon is necessary. Thus, 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. In contrast, 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. In other cases, although A or U nucleotides cannot be eliminated from the codons, it is however possible to decrease the A and U content by using codons which contain a lower content of A and/or U nucleotides. Examples of these are: the codons for Phe can be modified from UUU to UUC; 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 He can be modified from AUU or AUA to AUC; the 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. In the case of the codons for Met (AUG) and Trp (UGG), on the other hand, there is no possibility of sequence modification. The substitutions listed above can be used either individually or in all possible combinations to increase the G/C content of the RNA sequence of the present invention compared to its particular wild-type RNA (i.e. the original sequence). Thus, for example, all codons for Thr occurring in the wild-type sequence can be modified to ACC (or ACG). Preferably, however, for example, combinations of the above substitution possibilities are used:
- substitution of all codons coding for Thr in the original sequence (wild-type RNA) to ACC (or ACG) and
- substitution of all codons originally coding for Ser to UCC (or UCG or AGC); substitution of all codons coding for He in the original sequence to AUC and - substitution of all codons originally coding for Lys to AAG and
- substitution of all codons originally coding for Tyr to UAC; substitution of all codons coding for Val in the original sequence to GUC (or GUG) and
- substitution of all codons originally coding for Glu to GAG and - substitution of all codons originally coding for Ala to GCC (or GCG) and
- substitution of all codons originally coding for Arg to CGC (or CGG); substitution of all codons coding for Val in the original sequence to GUC (or GUG) and
- substitution of all codons originally coding for Glu to GAG and - substitution of all codons originally coding for Ala to GCC (or GCG) and
- substitution of all codons originally coding for Gly to GGC (or GGG) and
- substitution of all codons originally coding for Asn to AAC; substitution of all codons coding for Val in the original sequence to GUC (or GUG) and
- substitution of all codons originally coding for Phe to UUC and - substitution of all codons originally coding for Cys to UGC and
- substitution of all codons originally coding for Leu to CUG (or CUC) and
- substitution of all codons originally coding for Gin to CAG and
- substitution of all codons originally coding for Pro to CCC (or CCG); etc.
According to a specific embodiment 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 peptide or protein as defined herein or a fragment or variant thereof or the whole sequence of the wild type RNA sequence are substituted, thereby increasing the G/C content of said sequence. In this context, it is particularly preferable to increase the G/C content of the RNA sequence of the present invention, preferably of the at least one coding sequence of the RNA sequence according to the invention, to the maximum (i.e. 100 % of the substitutable codons) as compared to the wild-type sequence. According to the invention, a further preferred modification of the RNA sequence of the present invention is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells. Thus, if so-called "rare codons" are present in the RNA sequence of the present invention to an increased extent, the corresponding modified RNA sequence is translated to a significantly poorer degree than in the case where codons coding for relatively "frequent" tRNAs are present. According to the invention, in the modified RNA sequence of the present invention, the region which codes for a peptide or protein as defined herein or a fragment or variant thereof is modified compared to the corresponding region of the wild-type RNA sequence such that at least one codon of the wild-type sequence, which codes for a tRNA which is relatively rare in the cell, is exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA. By this modification, the sequence of the RNA of the present invention is modified such that codons for which frequently occurring tRNAs are available are inserted. In other words, according to the invention, by this modification all codons of the wild-type sequence, which code for a tRNA which is relatively rare in the cell, can in each case be exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA. Which tRNAs occur relatively frequently in the cell and which, in contrast, occur relatively rarely is known to a person skilled in the art; cf. e.g. Akashi, Curr. Opin. Genet. Dev. 2001, 11(6): 660-666. The codons, which use for the particular amino acid the tRNA which occurs the most frequently, e.g. the Gly codon, which uses the tRNA, which occurs the most frequently in the (human) cell, are particularly preferred. According to the invention, it is particularly preferable to link the sequential G/C content which is increased, in particular maximized, in the modified RNA sequence of the present invention, with the "frequent" codons without modifying the amino acid sequence of the protein encoded by the coding sequence of the RNA sequence. This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) RNA sequence of the present invention. The determination of a modified RNA sequence of the present invention as described above (increased G/C content; exchange of tRNAs) can be carried out using the computer program explained in WO 02/098443 - the disclosure content of which is included in its full scope in the present invention. Using this computer program, the nucleotide sequence of any desired RNA sequence can be modified with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence coded by the modified RNA sequence preferably not being modified compared to the non-modified sequence. Alternatively, it is also possible to modify only the G/C content or only the codon usage compared to the original sequence. The source code in Visual Basic 6.0 (development environment used: Microsoft Visual Studio
Enterprise 6.0 with Servicepack 3) is also described in WO 02/098443. In a further preferred embodiment of the present invention, the A/U content in the environment of the ribosome binding site of the RNA sequence of the present invention is increased compared to the A/U content in the environment of the ribosome binding site of its respective wild-type RNA. This modification (an increased A/U content around the ribosome binding site) increases the efficiency of ribosome binding to the RNA. An effective binding of the ribosomes to the ribosome binding site (e.g. to the Kozak sequence) in turn has the effect of an efficient translation of the RNA. According to a further embodiment of the present invention, the RNA sequence of the present invention may be modified with respect to potentially destabilizing sequence elements. Particularly, the coding sequence and/or the 5' and/or 3' untranslated region of this RNA sequence may be modified compared to the respective wild-type RNA such that it contains no destabilizing sequence elements, the encoded amino acid sequence of the modified RNA sequence preferably not being modified compared to its respective wild-type RNA. It is known that, for example in sequences of eukaryotic RNAs, destabilizing sequence elements (DSE) occur, to which signal proteins bind and regulate enzymatic degradation of RNA in vivo. For further stabilization of the modified RNA sequence, optionally in the region which encodes at least one peptide or protein as defined herein or a fragment or variant thereof, one or more such modifications compared to the corresponding region of the wild-type RNA can therefore be carried out, so that no or substantially no destabilizing sequence elements are contained there. According to the invention, DSE present in the untranslated regions (3'- and/or 5'-UTR) can also be eliminated from the RNA sequence of the present invention by such modifications. Such destabilizing sequences are e.g. AU-rich sequences (AURES), which occur in 3'-UTR sections of numerous unstable RNAs (Caput et al., Proc. Natl. Acad. Sci. USA 1986, 83: 1670 to 1674). The RNA sequence of the present invention is therefore preferably modified compared to the respective wild-type RNA such that the RNA sequence of the present invention contains no such destabilizing sequences. This also applies to those sequence motifs which are recognized by possible endonucleases, e.g. the sequence GAACAAG, which is contained in the 3'-UTR segment of the gene encoding the transferrin receptor (Binder et al., EMBO J. 1994, 13: 1969 to 1980). These sequence motifs are also preferably removed in the RNA sequence of the present invention.
According to another preferred embodiment, the mRNA used in the context of the invention has a modified the G/C content, preferably in its coding region, which means that the G/C content is modified, particularly increased, compared to the G/C content of the coding region of its corresponding wild-type mRNA, preferably without changing the encoded amino acid sequence. For example, the G/C content of the coding region may be increased by at least 7 %, or by at least 15 %, or by at least 20 %, compared to that of the wild-type mRNA which codes for an antigen, antigenic protein or antigenic peptide as described herein, or a fragment or variant thereof. According to a specific embodiment, at least 5 %, 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, 70 %, or 80 %, such as 90 % or more, 95 % or more, or even 100 % of the substitutable codons in the coding region or in the whole sequence are substituted to increase the G/C content. In this context, 100 % substitution means that essentially all substitutable codons of the coding region are substituted, which is one of the preferred embodiments of the invention. In another preferred embodiment, an mRNA is used wherein the coding region is modified such that at least one codon of the wild-type sequence which codes for a tRNA which is relatively rare in the cell is exchanged for a codon which codes for a tRNA which is relatively frequent in the cell but encodes the same amino acid as the relatively rare tRNA. tRNAs that occur relatively rarely or frequently in the cell are known to a person skilled in the art; cf. e.g. Akashi, Curr. Opin. Genet. Dev. 2001, 11 (6) : 660- 666. The most frequently occuring tRNAs for a particular amino acid are particularly preferred.
According to another embodiment, the nucleic acid sequence of the present invention, may be modified, and thus stabilized, by adapting the sequences to the human codon usage. According to the invention, a further preferred modification of the nucleic acid sequence of the present invention is based on the finding that codons encoding the same amino acid typically occur at different frequencies. According to the invention, in the modified nucleic acid sequence of the present invention, the coding sequence as defined herein is preferably modified compared to the corresponding coding sequence of the respective wild-type nucleic acid such that the frequency of the codons encoding the same amino acid corresponds to the naturally occurring frequency of that codon according to the human codon usage as e.g. shown in Table 1.
For example, in the case of the amino acid alanine (Ala) present in an amino acid sequence encoded by the at least one coding sequence of the a nucleic acid sequence according to the invention, the wild type coding sequence is preferably adapted in a way that the codon "GCC" is used with a frequency of 0.40, the codon "GCT" is used with a frequency of 0.28, the codon "GCA" is used with a frequency of 0.22 and the codon "GCG" is used with a frequency of 0.10 etc. (see Table 1).
Table 1: Human codon usage table
Amino acid Codon Fraction /1000
Ala GCG 0.10 7.4
Ala GCA 0.22 15.8
Ala GCT 0.28 18.5
Ala GCC* 0.40 27.7
Cys TGT 0.42 10.6
Cys TGC* 0.58 12.6
Asp GAT 0.44 21.8
Asp GAC* 0.56 25.1
Glu GAG* 0.59 39.6
Glu GAA 0.41 29.0 Amino acid Codon Fraction /1000
Phe TTT 0.43 17.6
Phe TTC* 0.57 20.3
Gly GGG 0.23 16.5
Gly GGA 0.26 16.5
Gly GGT 0.18 10.8
Gly GGC* 0.33 22.2
His CAT 0.41 10.9
His CAC* 0.59 15.1
He ATA 0.14 7.5
He ATT 0.35 16.0
He ATC* 0.52 20.8
Lys AAG* 0.60 31.9
Lys AAA 0.40 24.4
Leu TTG 0.12 12.9
Leu TTA 0.06 7.7
Leu CTG* 0.43 39.6
Leu CTA 0.07 7.2
Leu CTT 0.12 13.2
Leu CTC 0.20 19.6
Met ATG* 1 22.0
Asn AAT 0.44 17.0
Asn AAC* 0.56 19.1
Pro CCG 0.11 6.9
Pro CCA 0.27 16.9
Pro CCT 0.29 17.5
Pro CCC* 0.33 19.8 Amino acid Codon Fraction /1000
Gin CAG* 0.73 34.2
Gin CAA 0.27 12.3
Arg AGG 0.22 12.0
Arg AGA* 0.21 12.1
Arg CGG 0.19 11.4
Arg CGA 0.10 6.2
Arg CGT 0.09 4.5
Arg CGC 0.19 10.4
Ser AGT 0.14 12.1
Ser AGC* 0.25 19.5
Ser TCG 0.06 4.4
Ser TCA 0.15 12.2
Ser TCT 0.18 15.2
Ser TCC 0.23 17.7
Thr ACG 0.12 6.1
Thr ACA 0.27 15.1
Thr ACT 0.23 13.1
Thr ACC* 0.38 18.9
Val GTG* 0.48 28.1
Val GTA 0.10 7.1
Val GTT 0.17 11.0
Val GTC 0.25 14.5
Trp TGG* 1 13.2
Tyr TAT 0.42 12.2
Tyr TAC* 0.58 15.3
Stop TGA* 0.61 1.6 Amino acid Codon Fraction /1000
Stop TAG 0.17 0.8
Stop TAA 0.22 1.0
*: most frequent codon
As described above it is preferred according to the invention, that all codons of the wild- type sequence which code for a tRNA, which is relatively rare in the cell, are exchanged for a codon which codes for a tRNA, which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA. Therefore it is particularly preferred that the most frequent codons are used for each encoded amino acid (see Table■ 1, most frequent codons are marked with asterisks). Such an optimization procedure increases the codon adaptation index (CAI) and ultimately maximises the CAI. In the context of the invention, sequences with increased or maximized CAI are typically referred to as "codon-optimized" sequences and/or CAI increased and/or maximized sequences. According to a preferred embodiment, the nucleic acid sequence of the present invention comprises at least one coding sequence, wherein the coding sequence/sequence is codon-optimized as described herein. More preferably, the codon adaptation index (CAI) of the at least one coding sequence is at least 0.5, at least 0.8, at least 0.9 or at least 0.95. Most preferably, the codon adaptation index (CAI) of the at least one coding sequence is 1.
For example, in the case of the amino acid alanine (Ala) present in the amino acid sequence encoded by the at least one coding sequence of the nucleic acid sequence according to the invention, the wild type coding sequence is adapted in a way that the most frequent human codon "GCC" is always used for said amino acid, or for the amino acid Cysteine (Cys), the wild type sequence is adapted in a way that the most frequent human codon "TGC" is always used for said amino acid etc.
According to another embodiment, the nucleic acid sequence of the present invention may be modified by modifying, preferably increasing, the cytosine (C) content of the nucleic acid sequence, preferably of the coding sequence of the nucleic acid sequence, more preferably the coding sequence of the RNA sequence.
In a particularly preferred embodiment of the present invention, the C content of the coding sequence of the nucleic acid sequence of the present invention is modified, preferably increased, compared to the C content of the coding sequence of the respective wild-type nucleic acid, i.e. the unmodified nucleic acid. The amino acid sequence encoded by the at least one coding sequence of the nucleic acid sequence of the present invention is preferably not modified as compared to the amino acid sequence encoded by the respective wild-type nucleic acid.
In a preferred embodiment of the present invention, the modified nucleic acid, particularly the modified RNA sequence is modified such that at least 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, 70 % or 80 %, or at least 90 % of the theoretically possible maximum cytosine-content or even a maximum cytosine-content is achieved.
In further preferred embodiments, at least 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, 70 %, 80 %, 90 % or even 100 % of the codons of the target nucleic acid, particularly the modified RNA wild type sequence, which are "cytosine content optimizable" are replaced by codons having a higher cytosine-content than the ones present in the wild type sequence.
In a further preferred embodiment, some of the codons of the wild type coding sequence may additionally be modified such that a codon for a relatively rare tRNA in the cell is exchanged by a codon for a relatively frequent tRNA in the cell, provided that the substituted codon for a relatively frequent tRNA carries the same amino acid as the relatively rare tRNA of the original wild type codon. Preferably, all of the codons for a relatively rare tRNA are replaced by a codon for a relatively frequent tRNA in the cell, except codons encoding amino acids, which are exclusively encoded by codons not containing any cytosine, or except for glutamine (Gin), which is encoded by two codons each containing the same number of cytosines.
In a further preferred embodiment of the present invention, the modified target nucleic acid, preferably the RNA is modified such that at least 80 %, or at least 90 % of the theoretically possible maximum cytosine-content or even a maximum cytosine-content is achieved by means of codons, which code for relatively frequent tRNAs in the cell, wherein the amino acid sequence remains unchanged.
Due to the naturally occurring degeneracy of the genetic code, more than one codon may encode a particular amino acid. Accordingly, 18 out of 20 naturally occurring amino acids are encoded by more than one codon (with Tryp and Met being an exception), e.g. by two codons (e.g. Cys, Asp, Glu), by three codons (e.g. He), by four codons (e.g. Al, Gly, Pro) or by six codons (e.g. Leu, Arg, Ser). However, not all codons encoding the same amino acid are utilized with the same frequency under in vivo conditions. Depending on each single organism, a typical codon usage profile is established.
The term 'cytosine content-optimizable codon' as used within the context of the present invention refers to codons, which exhibit a lower content of cytosines than other codons encoding the same amino acid. Accordingly, any wild type codon, which may be replaced by another codon encoding the same amino acid and exhibiting a higher number of cytosines within that codon, is considered to be cytosine-optimizable (C-optimizable). Any such substitution of a C-optimizable wild type codon by the specific C-optimized codon within a wild type coding sequence increases its overall C-content and reflects a C-enriched modified nucleic acid sequence. According to a preferred embodiment, the nucleic acid sequence, particularly the RNA sequence of the present invention, preferably the at least one coding sequence of the nucleic acid sequence of the present invention comprises or consists of a C-maximized RNA sequence containing C-optimized codons for all potentially C-optimizable codons. Accordingly, 100% or all of the theoretically replaceable C-optimizable codons are preferably replaced by C-optimized codons over the entire length of the coding sequence.
In this context, cytosine-content optimizable codons are codons, which contain a lower number of cytosines than other codons coding for the same amino acid.
Any of the codons GCG, GCA, GCU codes for the amino acid Ala, which may be exchanged by the codon GCC encoding the same amino acid, and/or the codon UGU that codes for Cys may be exchanged by the codon UGC encoding the same amino acid, and/or the codon GAU which codes for Asp may be exchanged by the codon GAC encoding the same amino acid, and/or the codon that UUU that codes for Phe may be exchanged for the codon UUC encoding the same amino acid, and/or any of the codons GGG, GGA, GGU that code Gly may be exchanged by the codon GGC encoding the same amino acid, and/or the codon CAU that codes for His may be exchanged by the codon CAC encoding the same amino acid, and/or any of the codons AUA, AUU that code for He may be exchanged by the codon AUC, and/or any of the codons UUG, UUA, CUG, CUA, CUU coding for Leu may be exchanged by the codon CUC encoding the same amino acid, and/or the codon AAU that codes for Asn may be exchanged by the codon AAC encoding the same amino acid, and/or any of the codons CCG, CCA, CCU coding for Pro may be exchanged by the codon CCC encoding the same amino acid, and/or any of the codons AGG, AGA, CGG, CGA, CGU coding for Arg may be exchanged by the codon CGC encoding the same amino acid, and/or any of the codons AGU, AGC, UCG, UCA, UCU coding for Ser may be exchanged by the codon UCC encoding the same amino acid, and/or any of the codons ACG, ACA, ACU coding for Thr may be exchanged by the codon ACC encoding the same amino acid, and/or any of the codons GUG, GUA, GUU coding for Val may be exchanged by the codon GUC encoding the same amino acid, and/or the codon UAU coding for Tyr may be exchanged by the codon UAC encoding the same amino acid.
In any of the above instances, the number of cytosines is increased by 1 per exchanged codon. Exchange of all non C-optimized codons (corresponding to C-optimizable codons) of the coding sequence results in a C-maximized coding sequence. In the context of the invention, at least 70 %, preferably at least 80 %, more preferably at least 90 %, of the non C-optimized codons within the at least one coding sequence of the RNA sequence according to the invention are replaced by C-optimized codons.
It may be preferred that for some amino acids the percentage of C-optimizable codons replaced by C-optimized codons is less than 70 %, while for other amino acids the percentage of replaced codons is higher than 70 % to meet the overall percentage of C-optimization of at least 70 % of all C-optimizable wild type codons of the coding sequence.
Preferably, in a C-optimized RNA sequence of the invention, at least 50 % of the C- optimizable wild type codons for any given amino acid are replaced by C-optimized codons, e.g. any modified C-enriched RNA sequence preferably contains at least 50 % C-optimized codons at C-optimizable wild type codon positions encoding any one of the above mentioned amino acids Ala, Cys, Asp, Phe, Gly, His, He, Leu, Asn, Pro, Arg, Ser, Thr, Val and Tyr, preferably at least 60 %.
In this context, codons encoding amino acids, which are not cytosine content-optimizable and which are, however, encoded by at least two codons, may be used without any further selection process. However, the codon of the wild type sequence that codes for a relatively rare tRNA in the cell, e.g. a human cell, may be exchanged for a codon that codes for a relatively frequent tRNA in the cell, wherein both code for the same amino acid. Accordingly, the relatively rare codon GAA coding for Glu may be exchanged by the relative frequent codon GAG coding for the same amino acid, and/or the relatively rare codon AAA coding for Lys may be exchanged by the relative frequent codon AAG coding for the same amino acid, and/or the relatively rare codon CAA coding for Gin may be exchanged for the relative frequent codon CAG encoding the same amino acid.
In this context, the amino acids Met (AUG) and Trp (UGG), which are encoded by only one codon each, remain unchanged. Stop codons are not cytosine-content optimized, however, the relatively rare stop codons amber, ochre (UAA, UAG) may be exchanged by the relatively frequent stop codon opal (UGA).
The single substitutions listed above may be used individually as well as in all possible combinations in order to optimize the cytosine-content of the modified nucleic acid sequence compared to the wild type nucleic acid sequence.
Accordingly, the at least one coding sequence as defined herein may be changed compared to the coding sequence of the respective wild type nucleic acid in such a way that an amino acid encoded by at least two or more codons, of which one comprises one additional cytosine, such a codon may be exchanged by the C-optimized codon comprising one additional cytosine, wherein the amino acid is preferably unaltered compared to the wild type sequence.
According to a further preferred embodiment, the nucleic acid sequence, particularly the RNA sequence of the present invention may contain 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 40 to 80 adenosine nucleotides or even more preferably about 50 to 70 adenosine nucleotides.
Preferably, the poly(A) sequence in the RNA sequence of the present invention is derived from a DNA template by RNA in vitro transcription. Alternatively, the poly(A) sequence may also be obtained in vitro by common methods of chemical-synthesis without being necessarily transcribed from a DNA-progenitor. Moreover, poly(A) sequences, or poly(A) tails may be generated by enzymatic polyadenylation of the RNA according to the present invention using commercially available polyadenylation kits and corresponding protocols known in the art. Alternatively, the RNA as described herein optionally comprises a polyadenylation signal, which is defined herein as a signal, which conveys polyadenylation to a (transcribed) RNA by specific protein factors (e.g. cleavage and polyadenylation specificity factor (CPSF), cleavage stimulation factor (CstF), cleavage factors I and II (CF I and CF II), poly(A) polymerase (PAP)). In this context, a consensus polyadenylation signal is preferred comprising the NN(U/T)ANA consensus sequence. In a particularly preferred aspect, the polyadenylation signal comprises one of the following sequences: AA(U/T)AAA or A(U/T)(U/T)AAA (wherein uridine is usually present in RNA and thymidine is usually present in DNA).
According to a further preferred embodiment, the nucleic acid sequence, particularly the RNA sequence of the present invention may contain 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. Preferably, the poly(C) sequence in the RNA sequence of the present invention is derived from a DNA template by RNA in vitro transcription. In a preferred embodiment, the nucleic acid sequence, particularly the RNA sequence according to the invention comprises at least one 5'- or 3'-UTR element. In this context, an UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'- or 3'-UTR of any naturally occurring gene or which is derived from a fragment, a homolog or a variant of the 5'- or 3'-UTR of a gene. Preferably, the 5'- or 3'-UTR element used according to the present invention is heterologous to the at least one coding sequence of the RNA sequence of the invention. Even if 5'- or 3'-UTR elements derived from naturally occurring genes are preferred, also synthetically engineered UTR elements may be used in the context of the present invention.'
The term '3'UTR element' typically refers to a nucleic acid sequence, which comprises or consists of a nucleic acid sequence that is derived from a 3'UTR or from a variant of a 3'UTR. A 3'UTR element in the sense of the present invention may represent the 3'UTR of a nucleic acid molecule, particularly of an RNA or DNA, preferably an mRNA. Thus, in the sense of the present invention, preferably, a 3'UTR element may be the 3'UTR of an RNA, preferably of an mRNA, or it may be the transcription template for a 3'UTR of an RNA. Thus, a 3'UTR element preferably is a nucleic acid sequence which corresponds to the 3'UTR of an RNA, preferably to the 3'UTR of an mRNA, such as an mRNA obtained by transcription of a genetically engineered vector construct. Preferably, the 3'UTR element fulfils the function of a 3'UTR or encodes a sequence which fulfils the function of a 3'UTR.
Preferably, the at least one 3'UTR element comprises or consists of a nucleic acid sequence derived from the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene, or from a variant of the 3'UTR of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene.
Preferably, the nucleic acid sequence, particularly the RNA sequence of the present invention comprises a 3'UTR element, which may be derivable from a gene that relates to an RNA with an enhanced half-life (that provides a stable RNA), for example a 3'UTR element as defined and described below. Preferably, the 3' UTR element is a nucleic acid sequence derived from a 3' UTR of a gene, which preferably encodes a stable RNA, or from a homolog, a fragment or a variant of said gene
In a particularly preferred embodiment, the 3'UTR element comprises or consists of a nucleic acid sequence, which is derived from a 3'UTR of a gene selected from the group consisting of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene, or from a variant of a 3'UTR of a gene selected from the group consisting of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene according to SEQ ID NOs: 1369-1390 of the patent application WO 2013/143700, whose disclosure is incorporated herein by reference, or from a homolog, a fragment or a variant thereof. In a particularly preferred embodiment, the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an albumin gene, preferably a vertebrate albumin gene, more preferably a mammalian albumin gene, most preferably a human albumin gene.
In this context it is particularly preferred that the RNA sequence according to the invention comprises a 3'-UTR element comprising a corresponding RNA sequence derived from the nucleic acids according to SEQ ID NOs: 1369-1390 of the patent application WO 2013/143700 or a fragment, homolog or variant thereof.
In another particularly preferred embodiment, the 3'UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'UTR of an alpha-or beta-globin gene, preferably a vertebrate alpha-or beta-globin gene, more preferably a mammalian alpha-or beta-globin gene, most preferably a human alpha-or beta-globin gene. The term 'a nucleic acid sequence which is derived from the 3'UTR of a [...] gene' preferably refers to a nucleic acid sequence which is based on the 3'UTR sequence of a [...] gene or on a part thereof, such as on the 3'UTR of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene or on a part thereof. This term includes sequences corresponding to the entire 3'UTR sequence, i.e. the full length 3'UTR sequence of a gene, and sequences corresponding to a fragment of the 3'UTR sequence of a gene, such as an albumin gene, alpha-globin gene, beta-globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene.
The term 'a nucleic acid sequence which is derived from a variant of the 3'UTR of a [...] gene' preferably refers to a nucleic acid sequence, which is based on a variant of the 3'UTR sequence of a gene, such as on a variant of the 3'UTR of an albumin gene, an alpha-globin gene, a beta-globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1(1) gene, or on a part thereof as described above. This term includes sequences corresponding to the entire sequence of the variant of the 3'UTR of a gene, i.e. the full length variant 3'UTR sequence of a gene, and sequences corresponding to a fragment of the variant 3'UTR sequence of a gene. A fragment in this context preferably consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length variant 3'UTR, which represents at least 20 %, preferably at least 30 %, more preferably at least 40 %, more preferably at least 50 %, even more preferably at least 60 %, even more preferably at least 70 %, even more preferably at least 80 %, and most preferably at least 90 % of the full- length variant 3'UTR. Such a fragment of a variant, in the sense of the present invention, is preferably a functional fragment of a variant as described herein. According to a preferred embodiment, the nucleic acid sequence, particularly the RNA sequence according to the invention comprises a 5'-cap structure and/or at least one 3'- untranslated region element (3'-UTR element), preferably as defined herein. More preferably, the RNA further comprises a 5'-UTR element as defined herein.
In a particularly preferred embodiment the RNA sequence comprises, preferably in 5'- to 3'- direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) at least one coding sequence encoding at least one antigenic peptide or protein derived from a protein of interest or peptide of interest or a fragment or variant thereof, or a fragment or variant thereof, c.) a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from an alpha globin gene, a homolog, a fragment or a variant thereof; d.) optionally, a poly(A) sequence, preferably comprising 64 adenosines; e.) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and
In a particularly preferred embodiment, the at least one nucleic acid sequence, in particular, the RNA sequence comprises at least one 5'-untranslated region element (5'-UTR element).
Preferably, the at least one 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a TOP gene or which is derived from a fragment, homolog or variant of the 5'-UTR of a TOP gene.
It is particularly preferred that the 5'-UTR element does not comprise a TOP-motif or a 5'- TOP, as defined above.
In some embodiments, the nucleic acid sequence of the 5'-UTR element, which is derived from a 5'-UTR of a TOP gene, terminates at its 3'-end with a nucleotide located at position 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 upstream of the start codon (e.g. A(U/T)G) of the gene or RNA it is derived from. Thus, the 5'-UTR element does not comprise any part of the protein coding sequence. Thus, preferably, the only protein coding part of the at least one nucleic acid sequence, particularly of the RNA sequence, is provided by the coding sequence. The nucleic acid sequence derived from the 5'-UTR of a TOP gene is preferably derived from a eukaryotic TOP gene, preferably a plant or animal TOP gene, more preferably a chordate TOP gene, even more preferably a vertebrate TOP gene, most preferably a mammalian TOP gene, such as a human TOP gene.
For example, the 5'-UTR element is preferably selected from 5'-UTR elements comprising or consisting of a nucleic acid sequence, which is derived from a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, whose disclosure is incorporated herein by reference, from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, from a variant thereof, or preferably from a corresponding RNA sequence. The term "homologs of SEQ ID NOs: 1-1363, SEQ ID NO:
1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700" refers to sequences of other species than homo sapiens, which are homologous to the sequences according to SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO. 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700. In a preferred embodiment, the 5'-UTR element of the nucleic acid sequence, particularly of the RNA sequence according to the invention comprises or consists of a nucleic acid sequence, which is derived from a nucleic acid sequence extending from nucleotide position 5 (i.e. the nucleotide that is located at position 5 in the sequence) to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position
immediately 5' to the ATG sequence, of a nucleic acid sequence selected from SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application
WO2013/143700, from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO2013/143700 from a variant thereof, or a corresponding RNA sequence. It is particularly preferred that the 5' UTR element is derived from a nucleic acid sequence extending from the nucleotide position immediately 3' to the 5'-TOP to the nucleotide position immediately 5' to the start codon (located at the 3' end of the sequences), e.g. the nucleotide position immediately 5' to the ATG sequence, of a nucleic acid sequence selected from SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO2013/143700, from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO2013/143700, from a variant thereof, or a corresponding RNA sequence.
In a particularly preferred embodiment, the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a TOP gene encoding a ribosomal protein or from a variant of a 5'-UTR of a TOP gene encoding a ribosomal protein. For example, the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 170, 193, 244, 259, 554, 650, 675, 700, 721, 913, 1016, 1063, 1120, 1138, and 1284-1360 of the patent application WO2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'-TOP motif. As described above, the sequence extending from position 5 to the nucleotide immediately 5' to the ATG (which is located at the 3'end of the sequences) corresponds to the 5'-UTR of said sequences.
Preferably, the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a TOP gene encoding a ribosomal Large protein (RPL) or from a homolog or variant of a 5'-UTR of a TOP gene encoding a ribosomal Large protein (RPL). For example, the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a nucleic acid sequence according to any of SEQ ID NOs: 67, 259, 1284-1318, 1344, 1346, 1348-1354, 1357, 1358, 1421 and 1422 of the patent application WO2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'-TOP motif.
In a particularly preferred embodiment, the 5'-UTR element comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, or from a variant of the 5'UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, wherein preferably the 5'-UTR element does not comprise the 5'- TOP of said gene.
Accordingly, in a particularly preferred embodiment, the 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence according to SEQ ID NO: 17 (5'-UTR of human ribosomal protein Large 32 lacking the 5' terminal oligopyrimidine tract: GGCGCTGCCTACGGAGGTGGCAGCCATCTCCTTCTCGGCATC; corresponding to SEQ ID No. 1368 of the patent application WO2013/143700) or preferably to a corresponding RNA sequence, or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to the nucleic acid sequence of the above described sequences, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'UTR.
Preferably, the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more. Preferably, the fragment is a functional fragment as described herein.
In some embodiments, the RNA sequence according to the invention comprises a 5'-UTR element, which comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a vertebrate TOP gene, such as a mammalian, e.g. a human TOP gene, selected from RPSA, RPS2, RPS3, RPS3A, RPS4, RPS5, RPS6, RPS7, RPS8, RPS9, RPS10, RPS11, RPS12, RPS13, RPS14, RPS15, RPS15A, RPS16, RPS17, RPS18, RPS19, RPS20, RPS21, RPS23, RPS24, RPS25, RPS26, RPS27, RPS27A, RPS28, RPS29, RPS30, RPL3, RPL4, RPL5, RPL6, RPL7, RPL7A, RPL8, RPL9, RPL10, RPL10A, RPL11, RPL12, RPL13, RPL13A, RPL14, RPL15, RPL17, RPL18, RPL18A, RPL19, RPL21, RPL22, RPL23, RPL23A, RPL24, RPL26, RPL27, RPL27A, RPL28, RPL29, RPL30, RPL31, RPL32, RPL34, RPL35, RPL35A, RPL36, RPL36A, RPL37, RPL37A, RPL38, RPL39, RPL40, RPL41, RPLP0, RPLP1, RPLP2, RPLP3, RPLP0, RPLP1, RPLP2, EEF1A1, EEF1B2, EEF1D, EEF1G, EEF2, EIF3E, EIF3F, EIF3H, EIF2S3, EIF3C, EIF3K, EIF3EIP, EIF4A2, PABPC1, HNRNPA1, TPT1, TUBB1, UBA52, NPM1, ATP5G2, GNB2L1, NME2, UQCRB, or from a homolog or variant thereof, wherein preferably the 5'-UTR element does not comprise a TOP-motif or the 5'-TOP of said genes, and wherein optionally the 5'-UTR element starts at its 5'-end with a nucleotide located at position 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 downstream of the 5'-terminal oligopyrimidine tract (TOP) and wherein further optionally the 5'UTR element which is derived from a 5'-UTR of a TOP gene terminates at its 3'-end with a nucleotide located at position 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 upstream of the start codon (A(U/T)G) of the gene it is derived from.
In further particularly preferred embodiments, the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL21), an ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit 1, cardiac muscle (ATP5A1) gene, an hydroxysteroid (17-beta) dehydrogenase 4 gene (HSD17B4), an androgen- induced 1 gene (AIGl), cytochrome c oxidase subunit Vic gene (COX6C), or a N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAHl) or from a variant thereof, preferably from a vertebrate ribosomal protein Large 32 gene (RPL32), a vertebrate ribosomal protein Large 35 gene (RPL35), a vertebrate ribosomal protein Large 21 gene (RPL21), a vertebrate ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit 1, cardiac muscle (ATP5A1) gene, a vertebrate hydroxysteroid (17-beta) dehydrogenase 4 gene (HSD17B4), a vertebrate androgen- induced 1 gene (AIG1), a vertebrate cytochrome c oxidase subunit Vic gene (COX6C), or a vertebrate N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1) or from a variant thereof, more preferably from a mammalian ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL21), a mammalian ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit 1, cardiac muscle (ATP5A1) gene, a mammalian hydroxysteroid (17-beta) dehydrogenase 4 gene
(HSD17B4), a mammalian androgen-induced 1 gene (AIG1), a mammalian cyto-chrome c oxidase subunit Vic gene (COX6C), or a mammalian N-acylsphingosine ami-dohydrolase (acid
ceramidase) 1 gene (ASAH1) or from a variant thereof, most preferably from a human ribosomal protein Large 32 gene (RPL32), a human ribosomal protein Large 35 gene (RPL35), a human ribosomal protein Large 21 gene (RPL21), a human ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit 1, cardiac muscle (ATP5A1) gene, a human hydroxysteroid (17-beta) dehydrogenase 4 gene (HSD17B4), a human androgen-induced 1 gene (AIG1), a human cytochrome c oxidase subunit Vic gene (COX6C), or a human N-acylsphingosine amidohydrolase (acid ceramidase) 1 gene (ASAH1) or from a variant thereof, wherein preferably the 5'UTR element does not comprise the 5 OP of said gene.
Accordingly, in a particularly preferred embodiment, the 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence according to SEQ ID NOs: 1412-1420 of the patent application WO2013/143700, or a
corresponding RNA sequence or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence according SEQ ID NOs: 1412-1420 of the patent application WO 2013/143700, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20 % etc. of the full-length 5'UTR. Preferably, the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more. Preferably, the fragment is a functional fragment as described herein.
Accordingly, in a particularly preferred embodiment, the 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence according to SEQ ID NO: 18 (5'-UTR of ATP5A1 lacking the 5' terminal oligopyrimidine tract) : GCGGCTCGGCCATTTTGTCCCAGTCAGTCCGGAGGCTGCGGCTGCAGAAGTACCGCCTGCGGAGTAACTG CAAAG; corresponding to SEQ ID NO: 1414 of the patent application WO 2013/143700) or preferably to a corresponding RNA sequence, or wherein the at least one 5'UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40 %, preferably of at least about 50 %, preferably of at least about 60 %, preferably of at least about 70 %, more preferably of at least about 80 %, more preferably of at least about 90 %, even more preferably of at least about 95 %, even more preferably of at least about 99 % to the nucleic acid sequence as described above, wherein, preferably, the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20 % etc. of the full- length 5'UTR. Preferably, the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more. Preferably, the fragment is a functional fragment as described herein. Preferably, the at least one 5'-UTR element and the at least one 3'UTR element act synergistically to increase protein production from the at least one RNA sequence as described above.
According to a particularly preferred embodiment the RNA sequence according to the invention comprises, preferably in 5'- to 3'-direction: a. ) a 5'-cap structure, preferably m7GpppN; b. ) a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, a homolog, a fragment or a variant thereof; c. ) at least one coding sequence encoding at least one antigenic peptide or protein derived from a protein of interest or peptide of interest or a fragment or variant thereof, d. ) a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from a gene providing a stable RNA, a homolog, a fragment or a variant thereof; e. ) optionally, a poly(A) sequence preferably comprising 64 adenosines; and f. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines.
In a particularly preferred embodiment, the nucleic acid sequence, particularity the RNA sequence used according to the invention comprises a histone stem-loop sequence/structure. Such histone stem-loop sequences are preferably selected from histone stem-loop sequences as disclosed in WO 2012/019780, the disclosure of which is incorporated herewith by reference.
A histone stem-loop sequence, suitable to be used within the present invention, is preferably selected from at least one of the following formulae V or VI: formula V (stem-loop sequence without stem bordering elements) :
Figure imgf000075_0001
steml loop stem2 formula VI (stem-loop sequence with stem bordering elements) :
steml steml loop stem2 stem2
bordering element bordering element wherein: steml or stem2 bordering elements Ni-6 is a consecutive sequence of 1 to 6, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof; steml [N0-2GN3-5] is reverse complementary or partially reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1, more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof, and wherein G is guanosine or an analogue thereof, and may be optionally replaced by a cytidine or an analogue thereof, provided that its complementary nucleotide cytidine in stem2 is replaced by guanosine; loop sequence [No-4(U/T)No-4] is located between elements steml and stem2, and is a consecutive sequence of 3 to 5 nucleotides, more preferably of 4 nucleotides; wherein each N0-4 is independent from another a consecutive sequence of 0 to 4, preferably of 1 to 3, more preferably of 1 to 2 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; and wherein U/T represents uridine, or optionally thymidine; stem2 [N3-5CN0-2] is reverse complementary or partially reverse complementary with element steml, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N0-2 is a consecutive sequence of 0 to 2, preferably of 0 to 1, more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G or C or a nucleotide analogue thereof; and wherein C is cytidine or an analogue thereof, and may be optionally replaced by a guanosine or an analogue thereof provided that its complementary nucleoside guanosine in steml is replaced by cytidine; wherein steml and stem2 are capable of base pairing with each other forming a reverse complementary sequence, wherein base pairing may occur between steml and stem2, e.g. by Watson-Crick base pairing of nucleotides A and U/T or G and C or by non-Watson-Crick base pairing e.g. wobble base pairing, reverse Watson-Crick base pairing, Hoogsteen base pairing, reverse Hoogsteen base pairing or are capable of base pairing with each other forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between steml and stem2, on the basis that one or more bases in one stem do not have a complementary base in the reverse complementary sequence of the other stem.
According to a further preferred embodiment, the nucleic acid sequence, particularly the RNA sequence may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae Va or Via: formula Va (stem-loop sequence without stem bordering elements):
Figure imgf000077_0001
steml loop stem2 formula Via (stem-loop sequence with stem bordering elements):
N2-5 [No-iGNs-s] [Ni-3(U/T) No-2] [N3-5CN0-i] N2.5 steml steml loop stem2 stem2
bordering element bordering element wherein N, C, G, T and U are as defined above.
According to a further more particularly preferred embodiment, the at least one nucleic acid, preferably the at least one RNA may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae Vb or VIb:
Formula Vb (stem-loop sequence without stem bordering elements):
[N1GN4] [N2(U/T)Ni] [N4CN1] formula VIb (stem-loop sequence with stem bordering elements):
Figure imgf000078_0001
steml steml loop stem2 stem2
bordering element bordering element wherein N, C, G, T and U are as defined above.
A particularly preferred histone stem-loop sequence is the sequence
CAAAGGCTCTTTTCAGAGCCACCA (according to SEQ ID NO: 19) or more preferably the corresponding RNA sequence CAAAGGCUCUUUUCAGAGCCACCA (according to SEQ ID NO: 20).
Any of the above modifications may be applied to the nucleic acid sequence, in particular, to the DNA and/or RNA sequence of the present invention, and further to any DNA or RNA as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the respective nucleic acid sequence. A person skilled in the art will be able to take his choice accordingly. The nucleic acid sequence according to the invention, particularly the RNA sequence according to the present invention which comprises at least one coding sequence as defined herein, may preferably comprise a 5' UTR and/or a 3' UTR preferably containing at least one histone stem-loop. The 3' UTR of the RNA sequence according to the invention preferably comprises also a poly(A) and/or a poly(C) sequence as defined herein. The single elements of the 3' UTR may occur therein in any order from 5' to 3' along the sequence of the RNA sequence of the present invention. In addition, further elements as described herein, may also be contained, such as a stabilizing sequence as defined herein (e.g. derived from the UTR of a globin gene), IRES sequences, etc. Each of the elements may also be repeated in the RNA sequence according to the invention at least once (particularly in di- or multicistronic constructs), preferably twice or more. As an example, the single elements may be present in the nucleic acid sequence, particularly in the RNA sequence according to the invention in the following order:
5' - coding sequence - histone stem-loop - poly(A)/(C) sequence - 3'; or
5' - coding sequence - poly(A)/(C) sequence - histone stem-loop - 3'; or
5' - coding sequence - histone stem-loop - polyadenylation signal - 3'; or
5' - coding sequence - polyadenylation signal- histone stem-loop - 3'; or 5' - coding sequence - histone stem-loop - histone stem-loop - poly(A)/(C) sequence - 3'; or
5' - coding sequence - histone stem-loop - histone stem-loop - polyadenylation signal- 3'; or
5' coding sequence - stabilizing sequence - poly(A)/(C) sequence - histone stem-loop -
3'; or
5' - coding sequence - stabilizing sequence - poly(A)/(C) sequence - poly(A)/(C) sequence - histone stem-loop - 3'; etc.
According to a further embodiment, the nucleic acid sequence used in the present invention, particularly the RNA sequence, preferably comprises at least one of the following structural elements: a 5'- and/or 3'- untranslated region element (UTR element), particularly a 5'- UTR element, which preferably comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene or from a fragment, homolog or a variant thereof, or a 5'- and/or 3'-UTR element which may preferably be derivable from a gene that provides a stable RNA or from a homolog, fragment or variant thereof; a histone-stem-loop structure, preferably a histone- stem-loop in its 3' untranslated region; a 5'-cap structure; a poly-A tail; or a poly(C) sequence.
In a particularly preferred embodiment the nucleic acid sequence, in particular, the RNA sequence comprises, preferably in 5'- to 3'-direction: a.) a 5'-CAP structure, preferably m7GpppN; b.) at least one coding sequence encoding at least one antigenic peptide of interest or protein of interest or a fragment or variant thereof, c.) a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from an alpha globin gene, a homolog, a fragment or a variant thereof; d.) optionally, a poly(A) sequence, preferably comprising 64 adenosines; e.) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and f.) optionally, a histone stem-loop, preferably comprising the RNA sequence according to SEQ ID NO: 20. According to another particularly preferred embodiment the nucleic acid sequence, in particular, the RNA sequence used according to the invention comprises, preferably in 5'- to 3'- direction: a. ) a 5'-CAP structure, preferably m7GpppN; b. ) a 5'-UTR element which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene, a homolog, a fragment or a variant thereof; c. ) at least one coding sequence encoding at least one antigenic peptide of interest or protein of interest or a fragment or variant thereof, d. ) a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from a gene providing a stable RNA; e. ) optionally, a poly(A) sequence preferably comprising 64 adenosines; f. ) optionally, a poly(C) sequence, preferably comprising 30 cytosines; and optionally, a histone stem-loop, preferably comprising the RNA sequence according to SEQ ID NO: 20.
Nucleic acids used according to the present invention may be prepared by any method known in the art, including synthetic methods such as e.g. solid phase synthesis, as well as in vitro methods, such as in vitro transcription reactions or in vivo reactions, such as in vivo propagation of DNA plasmids in bacteria.
According to another preferred embodiment, the nucleic acid is in the form of a coding nucleic acid, preferably an mRNA, which additionally or alternatively encodes a secretory signal peptide. Such signal peptides typically exhibit a length of about 15 to 30 amino acids and are preferably located at the N-terminus of the encoded peptide, without being limited thereto. Signal peptides, as defined herein, preferably allow the transport of the encoded protein or peptide to a specific cell region or into a specific cellular compartment, such as to the cell surface, the endoplasmic reticulum (ER) or the endosomal-lysosomal compartment.
Proteins or peptides encoded by the nucleic acid may represent fragments or variants of naturally occurring proteins. Such fragments or variants may typically comprise a sequence having a sequence identity with one of the above mentioned proteins or peptides or sequences of their encoding nucleic acid sequences of at least 5 %, 10 %, 20 %, 30 %, 40 %, 50 %, 60 %, preferably at least 70 %, more preferably at least 80 %, equally more preferably at least 85 %, even more preferably at least 90 % and most preferably at least 95 % or even 97 %, to the entire wild-type sequence, either on nucleic acid level or on amino acid level.
"Fragments" of proteins or peptides in the context of the present invention may comprise a sequence of an protein or peptide as defined herein, which is, with regard to its amino acid sequence or its encoded nucleic acid sequence, N-terminally, C-terminally and/or
intrasequentially truncated compared to the amino acid sequence of the native protein or its encoded nucleic acid sequence. Such truncation may occur either on the amino acid level or on the nucleic acid level. A sequence identity with respect to such a fragment may therefore refer to the entire protein or peptide or to the entire coding nucleic acid sequence. The same applies accordingly to nucleic acids.
Such fragments of proteins or peptides may comprise a sequence of about 6 to about 20 or more amino acids, which includes fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 11, or 12 amino acids), as well as fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g. 13, 14, 15, 16, 17, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence. These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognised in their native form. The fragments of proteins or peptides may also comprise epitopes of those proteins or peptides. Epitopes (also called "antigen determinants"), in the context of the present invention, are typically fragments located on the outer surface of native proteins or peptides, preferably having 5 to 15 amino acids, more preferably having 5 to 12 amino acids, 6 to 9 amino acids, which may be recognised by antibodies or B-cell receptors in their native form. Such epitopes may furthermore be selected from any of the herein mentioned variants of such proteins or peptides. In this context, antigenic determinants can be conformational or discontinous epitopes which are composed of segments of the proteins or peptides that are discontinuous in the amino acid sequence of the proteins or peptides, but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain. "Variants" of proteins or peptides as defined herein may be encoded by the nucleic acid, wherein nucleotides encoding the protein or peptide are replaced such that the encoded amino acid sequence is changed. Thereby a protein or peptide with one or more mutations is generated, such as with one or more substituted, inserted and/or deleted amino acids. Preferably, these fragments and/or variants have the same biological function or specific activity compared to the full-length native protein, e.g. its specific antigenic property.
As mentioned, the composition of the invention comprises the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound at a ratio of not higher than about 2 nmol lipid per μg nucleic acid compound, i.e. at a relatively low amount of cationic lipid. It has been unexpected that such low amounts of cationisable or permanently cationic lipid or lipidoid are highly effective in delivering the nucleic acid compound to cells, in particular when the composition is administered by local or locoregional administration as described in more detail below rather than by intravenous or intraarterial injection. In another preferred embodiment, the composition comprises the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound at a ratio of not higher than about 1 nmol lipid or lipidoid per μg nucleic acid compound. In other specific embodiments, this ratio is in the range from about 0.05 to about 2 nmol/ μg, or from about 0.1 to about 1.5 nmol/ μg, or from about 0.25 to about 1.0 nmol^g, or from about 0.3 to about 0.8 nmol^g, such as about 0.4 nmol^g, respectively.
In one of the preferred embodiments, the nucleic acid compound and the cationisable or permanently cationic lipid or lipidoid together form a complex.
A "complex", as used herein, is an association of molecules into larger units held together by forces that are weaker than covalent chemical bonds. Such complex may also be referred to as an association complex. The forces by which a complex is held together are often hydrogen bonds, also known as hydrogen bridges, London forces, and/or dipolar attraction. A complex involving a lipid and a nucleic acid is often referred to as a lipoplex, and a complex between a polymer and a nucleic acid is known as a polyplex. Accordingly, the complex formed in the composition of the invention by the cationisable or permanently cationic lipid and the nucleic acid compound may be referred to as a lipoplex. Optionally, further constituents may participate in such complex.
As is often the case with lipoplexes, the complex of the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound may not be fully soluble in an aqueous environment, but may exist in the form of a colloid, such as a nanoparticle, or a plurality of nanoparticles. In one embodiment of the composition of the invention, the essential constituents, i.e. the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound, are incorporated in one or more nanoparticles. In other words, the composition may comprise one or more nanoparticles comprising the cationisable or permanently cationic lipid or lipidoid and nucleic acid compound as described herein-above.
Typically such nanoparticles may be formed spontaneously when the cationisable or permanently cationic lipid and the nucleic acid cargo (or compound) are combined, e.g. in an aqueous environment.
A "nanoparticle", as used herein, is a submicron particle having any structure or morphology. Submicron particles may also be referred to as colloids, or colloidal. With respect to the material on which the nanoparticle is based, and to the structure or morphology, a nanoparticle may be classified, for example, as a nanocapsule, a vesicle, a liposome, a lipid nanoparticle, a micelle, a crosslinked micelle, a lipoplex, a polyplex, a mixed or hybrid complex, to mention only a few of the possible designations of specific types of nanoparticles.
According to this aspect, the invention is also directed to the above-defined nanoparticle as such, as well as to a composition, in particular a pharmaceutical composition, comprising a plurality of such nanoparticles, in particular to a plurality of the preferred nanoparticles as described in more detail below. Any references to the "nanoparticle" of the invention should also be understood as referring to the plurality of such nanoparticles as typically comprised in a composition, and vice versa.
Optionally, the nanoparticles may comprise a further biologically active cargo material in addition to the nucleic acid compound, or they may comprise any other compound having a carrier function or an auxiliary function.
In the context of the invention, a "biologically active cargo material" generically refers to a compound, or mixture or combination of compounds, which is intended to be delivered to a subject, or to an organ, tissue, or cell of a subject, by means of a formulation, carrier, vector or vehicle, in order to achieve a desired biologic effect, such as a pharmacological effect, including any type of prophylactic, therapeutic, diagnostic, or ameliorating effect. The delivery of biologically active cargo material is the purpose of administering a product comprising such material, whereas the formulation, or carrier, vector or vehicle, which may in some cases also be considered as biologically active, are primarily the means for delivering the cargo material. Unless different meanings are evident from the context, the expressions "biologically active cargo material", "biologically active compound", "cargo material", "cargo" and the like are used synonymously. As will be described in more detail below, a preferred type of cargo is a nucleic acid, or a nucleic acid-based material. A "carrier", or "vehicle", as used herein, may generically mean any compound, construct or material being part of a formulation which aids, enables, or improves the delivery of the biologically active compound or material. It may be biologically substantially inert, or it may be biologically active in that it interacts substantially with tissues, cells or subcellular components of the subject and, for example, enhance the uptake of the biologically active cargo material. In the context of the invention, the terms may be applied to the cationisable or permanently cationic lipid.
A "formulation", with respect to a biologically active compound that is incorporated in it and administered by means of the formulation, is any product which is pharmaceutically acceptable in terms of its composition and manufacturing method which comprises at least one biologically active compound and one excipient, carrier, vehicle or other auxiliary material.
In one of the embodiments, the nanoparticle of the invention substantially consists of the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound, or of the cargo-carrier complex as defined above. In this specific context, the expression "substantially consists of should not be understood such as to exclude the presence of minor amounts of auxiliary materials in the nanoparticles such as solvents, cosolvents, surfactants, isotonising agents and the like.
In another specific embodiment, the nanoparticle further comprises one or more compounds independently selected from targeting agents, cell penetrating agents, and stealth agents. Regarding the optional features and preferences relating to targeting agents, cell penetrating agents, and stealth agents.
As used herein, a targeting agent is a compound that has affinity to a target, such as a target located on or at the surface of a target cell, or an intracellular target. For example, the targeting agent may represent an antibody, an antibody fragment, or a small molecular agent having affinity to a target of interest. Cell penetrating agents include cell-penetrating peptides (CPPs), as well as any other compounds with a similar biological or biomimetic function, i.e. to facilitate the uptake of cargo into cells. A stealth agent, in the context of the invention, means a compound or material which, when incorporated in the nanoparticle comprising the cationisable or permanently cationic lipid and the nucleic acid cargo, leads to a longer circulation time of the nanoparticle in the bloodstream of a subject to which the nanoparticle is injected, e.g. by intravenous injection or infusion. An example for a stealth agent is a pegylated lipid whose lipid domain is capable of functioning as an anchor to the nanoparticle by e.g. interacting with a hydrophobic group of the cationisable or permanently cationic or cationisable lipid, whereas its polyethylene glycol (PEG) domain may impart "stealth" properties to the nanoparticle, which means that the nanoparticle shows decreased interaction with a subject's immune system while circulating in the bloodstream, which is typically associated with a prolonged elimination half life of the nanoparticle from the blood, as well as reduced immunogenicity and antigenicity.
Examples of useful pegylated lipids include l-(monomethoxy-polyethyleneglycol)-2,3- dimyristoylglycerol (PEG-DMG), N-[(methoxy poly(ethylene glycol) 2ooo)carbamoyl]-l,2- dimyristyloxypropyl-3-amine (PEG-C-DMA), or l,2-diacyal-sn-glycero-3-phosphoethanolamine- N-[methoxy(polyethylene glycol)]; in case of the latter, acyl may mean e.g. myristoyl, palmitoyl, stearoyl, or oleoyl, and the polyethylene glycol is typically polyethylene glycol-350 to
polyethylene glycol-5000, in particular polyethylene glycol-750, polyethylene glycol-1000, polyethylene glycol-2000, and polyethylene glycol-3000.
However, if the composition and the nanoparticles of the invention are designed for local or locoregional administration, the incorporation of a stealth agent may not normally be required, unless for the purpose of reducing the immunogenicity of the composition.
Accordingly, the nanoparticle may optionally comprise a lipid which is not a permanently cationic lipid. Examples of such lipids which could potentially be useful in certain cases include cationisable lipids, uncharged lipids, in particular steroids such as cholesterol, and pegylated lipids such as pegylated phospholipids, which may also function as a stealth agent. However, the inventors believe that in many cases the incorporation of any other lipids which are not cationisable or permanently cationic is not required, in particular not for cases in which the nucleic acid compound is to be delivered to cell by using a local or locoregional route of administration, as described in more detail above. It is therefore another preferred embodiment according to which the nanoparticle is substantially free of non-cationic lipids or lipidoids. In a related embodiment, the composition of the invention is also substantially free of non-cationic or lipids or lipidoids. In other words, according to these embodiments, the nanoparticle or the composition does not contain any pharmaceutically relevant amounts of a non-cationic lipid nor of a non-cationic lipidoid. In yet another embodiment, the nanoparticle and/or composition is free of lipids or lipidoids which are not permanently cationic.
In another specific embodiment, the nanoparticle essentially consists of one or more cationisable or permanently cationic lipids or lipidoids, one or more nucleic acid compounds, and optionally one or more compounds independently selected from targeting agents, cell penetrating agent, and stealth agents.
Alternatively, at least about 50 wt.-% of the nanoparticles in the composition of the invention consist of the cationisable or permanently cationic lipid or lipidoid and the nucleic acid compound, or at least 60 wt.-% thereof, at least 70 wt.-% thereof, at least 80 wt.-% thereof, at least 85 wt.-% thereof, at least 90 wt.-% thereof, or at least 95 wt.-% thereof, respectively.
In any case, the nanoparticles preferably and typically comprise at least a cationisable or permanently cationic lipid or lipidoid and a nucleic acid compound. With respect to the lipid and the nucleic acid, all options and preferences as described above in the context of the composition in general also apply to the nanoparticles. For example, the nucleic acid compound comprised in the nanoparticles may be any chemically modified or unmodified DNA, single stranded or double stranded DNA, coding or non-coding DNA, optionally selected from plasmid, (short)
oligodesoxynucleotide (i.e. a (short) DNA oligonucleotide), genomic DNA, DNA primers, DNA probes, immunostimulatory DNA, aptamer, or any combination thereof. Alternatively, or in addition, such a nucleic acid molecule may be selected e.g. from any PNA (peptide nucleic acid). Further alternatively, or in addition, and also according to a particularly preferred embodiment, the nucleic acid is selected from chemically modified or unmodified RNA, single-stranded or double-stranded RNA, coding or non-coding RNA, optionally selected from messenger RNA (mRNA), (short) oligoribonucleotide (i.e. a (short) RNA oligonucleotide), viral RNA (vRNA), replicon RNA, transfer RNA (tRNA),ribosomal RNA (rRNA), immunostimulatory RNA (isRNA), microRNA, small interfering RNA (siRNA), small nuclear RNA (snRNA), small-hairpin RNMA (shRNA) or a riboswitch, an RNA aptamer, an RNA decoy, an antisense RNA, a ribozyme, or any combination thereof, as described herein-above. Preferably, the nucleic acid molecule of the complex is an RNA. More preferably, the nucleic acid molecule of the complex is a (linear) single- stranded RNA, even more preferably an mRNA or an immunostimulatory RNA.
Optionally, the biologically active cargo material is a combination of more than one nucleic acid compounds.
The nanoparticles preferably have a hydrodynamic diameter as determined by dynamic laser scattering of not more than about 1,000 nm. More preferably, their hydrodynamic diameter is not higher than about 800 nm, such as in the range from about 30 nm to about 800 nm. In other preferred embodiments, the hydrodynamic diameter is in the range from about 50 nm to about 300 nm, or from about 60 nm to about 250 nm, from about 60 nm to about 150 nm, or from about 60 nm to about 120 nm, respectively. While these are preferred diameters of individual nanoparticles, this does not exclude the presence of nanoparticles of other diameters in the composition of the invention. However, the invention is preferably practised with compositions in which many - or even most - of the nanoparticles exhibit such diameters.
Moreover, the composition according to the invention which comprises a plurality of such nanoparticles may also be characterised by the mean hydrodynamic diameter as determined by dynamic laser scattering, which is also preferably not higher than 800 nm, such as in the range from about 30 nm to about 800 nm. In the context of the hydrodynamic diameter, the "mean" should be understood as the Z-average, also known as the cumulants mean. Obviously, the measurement by dynamic laser scattering must also be conducted with an appropriate dispersant and at an appropriate dilution, following the recommendations of the manufacturer of the analytic equipment that is used. Particularly preferred is a mean hydrodynamic diameter in the range from about 50 nm to about 300 nm, or from about 60 nm to about 250 nm, from about 60 nm to about 150 nm, or from about 60 nm to about 120 nm, respectively.
The nanoparticles may further be characterised by their electrokinetic potential, which may be expressed by means of the zeta potential. In some embodiments, the zeta potential is in the range from about 0 mV to about +50 mV, or from about 0 mV to about +10 mV, respectively. In other embodiments, the zeta potential is positive, i.e. higher than 0 mV, but not higher than +50 mV, or +40 mV, or +30 mV, or +20 mV, or +10 mV, respectively.
In some preferred embodiments, the zeta potential is in the range from about 0 mV to about -50 mV, or from about 0 mV to about -10 mV, respectively. In other embodiments, the zeta potential is negative, i.e. lower than 0 mV, but not lower than -50 mV, or -40 mV, or -30 mV, or - 20 mV, or -10 mV, respectively.
In another preferred embodiment, the zeta potential is in the range of 0 mV to -50 mV and the particles have an N/P ratio of 1 or less. It has been found by the inventors that such nanoparticles are particularly suitable for local administration.
The amount of the cationisable or permanently cationic lipid or lipidoid should also be selected taking the amount of the nucleic acid cargo into account. In one embodiment, these amounts are selected such as to result in an N/P ratio of the nanoparticle(s) or of the composition in the range from about 0.1 to about 20. In this context, the N/P ratio is defined as the mole ratio of the nitrogen atoms ("N") of the basic nitrogen-containing groups of the lipid or lipidoid to the phosphate groups ("P") of the nucleic acid which is used as cargo. The N/P ratio may be calculated on the basis that, for example, 1 μg RNA typically contains about 3 nmol phosphate residues, provided that the RNA exhibits a statistical distribution of bases. The "N"-value of the lipid or lipidoid may be calculated on the basis of its molecular weight and the relative content of cationisable or permanently cationic and - if present - cationisable groups.
In a preferred embodiment, the N/P ratio of the nanoparticles or the composition is not higher than about 10. In another preferred embodiment, the N/P ratio is not higher than about 6, or not higher than about 3, respectively. In a preferred embodiment, the N/P ratio of the nanoparticles or the composition is not higher than about 1. For example, the N/P ratio may be in the range from about 0.01 to about 1, or from about 0.1 to about 0.99. In further preferred embodiments, the N/P ratio of the
nanoparticles or the composition is in the range from about 0.2 to about 0.9, such as about 0.3±0.2, 0.5±0.2, or 0.7±0.2.
Such low N/P ratios are commonly believed to be detrimental to the performance and in vivo efficacy of such carrier-cargo complexes, or nucleic-acid loaded nanoparticles. However, the inventors found that such N/P ratios are indeed useful in the context of the present invention, in particular when the local or extravascular administration of the nanoparticles is intended. Here, the respectively nanoparticles have been found to be efficacious and at the same time well- tolerated.
As mentioned above, the amount of cationic lipid in the composition of the invention as well as in the nanoparticle(s) is typically much lower than in conventional lipid-based carriers for nucleic acids as cargo. The present invention may be practised with as little as about 0.1 to about 10% of the typical amount of lipids or lipidoids used in lipoplexes or lipid nanoparticles that have been proposed for the delivery of e.g. RNA and the transfection of cells. Without wishing to be bound by theory, the inventors assume that such low amount of lipid or lipidoid has been pivotal in achieving the high tolerability of the composition of the invention.
The nanoparticles may be prepared by a method comprising the step of combining one or more cationisable or permanently cationic lipids and/or lipidoids, optionally dissolved in an appropriate solvent (e.g. ethanol, DMSO), and one or more nucleic acid compounds in the presence of an aqueous liquid such as to allow the formation of a nanoparticle or a plurality of nanoparticles. The mixing can be conducted by an suitable mixing device (e.g. laminar flow combination utilizing a T or Y valve; microfluidic devices or simple addition to a stirred solution). The composition of the invention which comprises the cationisable or permanently cationic lipid or lipidoid, the nucleic acid compound as cargo and optionally one or more further ingredients, and in particular the composition which comprises the complex and/or the nanoparticles as described above, is preferably formulated and processed such as to be suitable for administration to a subject, in particular to an animal or to a human subject. Preferably, the composition is sterile.
In this respect, the composition may also be referred to as a pharmaceutical composition. This is a general preference which may be applied to any of the options and preferences described herein with respect to the constituents and other features of the composition or the nanoparticles. In other words, the invention is also directed e.g. to a pharmaceutical composition as defined herein where the nucleic acid compound is a coding nucleic acid which encodes at least one peptide or protein. For example , the coding nucleic acid may encode a therapeutically active protein or an antigen. The invention is further directed to a vaccine comprising such
pharmaceutical composition wherein the coding nucleic acid encodes at least one antigen. In this context, the vaccine may consist of the pharmaceutical composition, or it may comprise it along with other constituents.
The inventive pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term parenteral, as used herein, includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques.
Preferably, the pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may be administered by parenteral injection, more preferably by subcutaneous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, or intraperitoneal, injection or infusion. Particularly preferred is intradermal and intramuscular injection. Sterile injectable forms of the
pharmaceutical compositions may be aqueous. Alternatively, they may be oily suspensions. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
The pharmaceutical composition, nanoparticle or composition comprising said nanoparticle as defined herein may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
The pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including diseases of the skin or of any other accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs. For topical applications, the inventive pharmaceutical composition, nanoparticle or composition comprising said nanoparticle may be formulated in a suitable formulation such as a liquid or an ointment, containing the nucleic acid suspended or dissolved in one or more carriers. The pharmaceutical composition, nanoparticle or composition comprising said nanoparticle typically comprises a "safe and effective amount" of the components, particularly of the nucleic acid. As used herein, a "safe and effective amount" means an amount of nucleic acid is sufficient to significantly induce a prevention, or positive modification of a disease or disorder as defined herein. At the same time, however, a "safe and effective amount" is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk.
If injection is intended, the composition, nanoparticle or composition comprising said nanoparticle may be designed as a ready-to-use injectable formulation. For example, it may be formulated as a sterile liquid suitable for injection. In this case, it may be provided as a sterile aqueous solution, or a sterile aqueous suspension of nanoparticles, preferably with a pH in the range from about 4 to about 9, or more preferably from about 4.5 to about 8.5. The osmolality of such liquid composition is preferably from about 150 to about 500 mOsmol/kg, and more preferably from about 200 to about 400 mOsmol/kg. If the composition is to be injected intravenously, the pH may also be in the range from about 4.5 to about 8, or from about 5 to about 7.5; and the osmolality may in this case preferably be selected in the range from about 220 to about 350 mOsmol/kg, or from about 250 to about 330 mOsmol/kg, respectively.
Alternatively, the composition, nanoparticle or composition comprising said nanoparticle may be formulated as a concentrated form which requires dilution or even reconsitution before use. For example, it may be in the form of a liquid concentrate, which could be an aqueous and/or organic liquid formulation which requires dilution with an aqueous solvent or diluent. If the liquid concentrate comprises an organic solvent, such solvent is preferably selected from water- miscible organic solvents with relatively low toxicity such as ethanol or propylene glycol.
In one of the preferred embodiments, the composition, nanoparticle or composition comprising said nanoparticle of the invention is provided as a dry formulation for reconstitution with a liquid carrier such as to generate a liquid formulation suitable for injection. In particular, the dry formulation may be a sterile powder or lyophilised form for reconstitution with an aqueous liquid carrier.
In order to optimise its performance, stability or tolerability, the composition, nanoparticle or composition comprising said nanoparticle may optionally comprise pharmaceutical excipients as required or useful. Potentially useful excipients include acids, bases, osmotic agents, antioxidants, stabilisers, surfactants, synergists, colouring agents, thickening agents, bulking agents, and - if required - preservatives. The vaccine according to the invention is based on the same components as the
(pharmaceutical) composition described herein. Insofar, it may be referred to the description of the (pharmaceutical) composition as provided herein. Preferably, the vaccine according to the invention comprises at least one nucleic acid and a pharmaceutically acceptable carrier. In embodiments, where the vaccine comprises more than one nucleic acid, particularly more than one mRNA sequence (such as a plurality of RNA sequences according to the invention, wherein each preferably encodes a distinct antigenic peptide or protein), the vaccine may be provided in physically separate form and may be administered by separate administration steps. The vaccine according to the invention may correspond to the (pharmaceutical) composition as described herein, especially where the mRNA sequences are provided by one single composition. However, the inventive vaccine may also be provided physically separated. For instance, in embodiments, wherein the vaccine comprises more than one mRNA sequences/species, these RNA species may be provided such that, for example, two, three, four, five or six separate compositions, which may contain at least one mRNA species/sequence each (e.g. three distinct mRNA species/sequences), each encoding distinct antigenic peptides or proteins, are provided, which may or may not be combined. Also, the inventive vaccine may be a combination of at least two distinct compositions, each composition comprising at least one mRNA encoding at least one of the antigenic peptides or proteins defined herein. Alternatively, the vaccine may be provided as a combination of at least one mRNA, preferably at least two, three, four, five, six or more mRNAs, each encoding one of the antigenic peptides or proteins defined herein. The vaccine may be combined to provide one single composition prior to its use or it may be used such that more than one administration is required to administer the distinct mRNA sequences/species encoding any of the antigenic peptides or proteins as defined herein. If the vaccine contains at least one mRNA sequence, typically at least two mRNA sequences, encoding the antigen combinations defined herein, it may e.g. be administered by one single administration (combining all mRNA species/sequences), by at least two separate administrations. Accordingly; any combination of mono-, bi- or multicistronic mRNAs encoding the at least one antigenic peptide or protein or any combination of antigens as defined herein (and optionally further antigens), provided as separate entities (containing one mRNA species) or as combined entity (containing more than one mRNA species), is understood as a vaccine according to the present invention.
As with the (pharmaceutical) composition according to the present invention, the entities of the vaccine may be provided in liquid and or in dry (e.g. lyophilized) form. They may contain further components, in particular further components allowing for its pharmaceutical use. The vaccine or the (pharmaceutical) composition may, e.g., additionally contain a pharmaceutically acceptable carrier and/or further auxiliary substances and additives. The vaccine or (pharmaceutical) composition typically comprises a safe and effective amount of the nucleic acid, particularly mRNA according to the invention as defined herein, encoding an antigenic peptide or protein as defined herein or a fragment or variant thereof or a combination of antigens, preferably as defined herein. As used herein, "safe and effective amount" means an amount of the mRNA that is sufficient to significantly induce a positive modification of cancer or a disease or disorder related to cancer. At the same time, however, a "safe and effective amount" is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment. In relation to the vaccine or (pharmaceutical) composition of the present invention, the expression "safe and effective amount" preferably means an amount of the mRNA (and thus of the encoded antigen) that is suitable for stimulating the adaptive immune system in such a manner that no excessive or damaging immune reactions are achieved but, preferably, also no such immune reactions below a measurable level. Such a "safe and effective amount" of the mRNA of the (pharmaceutical) composition or vaccine as defined herein may furthermore be selected in dependence of the type of mRNA, e.g.
monocistronic, bi- or even multicistronic mRNA, since a bi- or even multicistronic mRNA may lead to a significantly higher expression of the encoded antigen(s) than the use of an equal amount of a monocistronic mRNA. A "safe and effective amount" of the mRNA of the (pharmaceutical) composition or vaccine as defined above will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying physician. The vaccine or composition according to the invention can be used according to the invention for human and also for veterinary medical purposes, as a pharmaceutical composition or as a vaccine.
In a preferred embodiment, the nucleic acid, particularly the mRNA of the (pharmaceutical) composition, vaccine or kit of parts according to the invention is provided in lyophilized form. Preferably, the lyophilized mRNA is reconstituted in a suitable buffer, advantageously based on an aqueous carrier, prior to administration, e.g. Ringer-Lactate solution, saline, or a phosphate buffer solution. In a preferred embodiment, the (pharmaceutical) composition, the vaccine or the kit of parts according to the invention contains at least one, two, three, four, five, six or more mRNAs, preferably mRNAs which are provided separately in lyophilized form (optionally together with at least one further additive) and which are preferably reconstituted separately in a suitable buffer (such as Ringer-Lactate solution or phosphate buffer) prior to their use so as to allow individual administration of each of the (monocistronic) mRNAs. The vaccine or (pharmaceutical) composition according to the invention may typically contain a pharmaceutically acceptable carrier. The expression "pharmaceutically acceptable carrier" as used herein preferably includes the liquid or non-liquid basis of the inventive vaccine. If the inventive vaccine is provided in liquid form, the carrier will be water, typically pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate, citrate etc. buffered solutions. Particularly for injection of the inventive vaccine, water or preferably a buffer, more preferably an aqueous buffer, may be used, containing a sodium salt, preferably at least 50 mM of a sodium salt, a calcium salt, preferably at least 0,01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt. According to a preferred
embodiment, the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc. Without being limited thereto, examples of sodium salts include e.g. NaCl, Nal, NaBr, Na2C03, NaHC03, Na2SC>4, examples of the optional potassium salts include e.g. KC1, KI, KBr, K2CO3, KHCO3, K2SO4, and examples of calcium salts include e.g. CaC , Cah, CaBr2, CaC03, CaSC , Ca(OH)2. Furthermore, organic anions of the aforementioned cations may be contained in the buffer. According to a more preferred embodiment, the buffer suitable for injection purposes as defined above, may contain salts selected from sodium chloride (NaCl), calcium chloride (CaC ) and optionally potassium chloride (KC1), wherein further anions may be present additional to the chlorides. CaC can also be replaced by another salt like KC1. Typically, the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCl), at least 3 mM potassium chloride (KC1) and at least 0,01 mM calcium chloride (CaC ). The injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e. the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects. Reference media are e.g. in "in vivo" methods occurring liquids such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in "in vitro" methods, such as common buffers or liquids. Such common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis. However, one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person. The term "compatible" as used herein means that the constituents of the inventive vaccine are capable of being mixed with the nucleic acid, particularly the mRNA according to the invention as defined herein, in such a manner that no interaction occurs, which would substantially reduce the pharmaceutical effectiveness of the inventive vaccine under typical use conditions.
Pharmaceutically acceptable carriers, fillers and diluents must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated. Some examples of compounds which can be used as pharmaceutically acceptable carriers, fillers or constituents thereof are sugars, such as, for example, lactose, glucose, trehalose and sucrose; starches, such as, for example, corn starch or potato starch; dextrose; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulfate; vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid. The choice of a pharmaceutically acceptable carrier is determined, in principle, by the manner, in which the pharmaceutical composition or vaccine according to the invention is administered. The composition or vaccine can be administered, for example, systemically or locally. Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes. Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial,
intrapulmonal, intracardial, and sublingual injections. More preferably, composition or vaccines according to the present invention may be administered by an intradermal, subcutaneous, intramuscular or otherwise local or locoregional route, preferably by injection, which may be needle-free and/or needle injection. Compositions/vaccines are therefore preferably formulated in liquid or solid form. The suitable amount of the vaccine or composition according to the invention to be administered can be determined by routine experiments, e.g. by using animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non-human primate models. Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4. Suitable carriers for injection include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices. Suitable pharmaceutically acceptable carriers for topical application include those which are suitable for use in lotions, creams, gels and the like. If the inventive composition or vaccine is to be administered perorally, tablets, capsules and the like are the preferred unit dose form. The pharmaceutically acceptable carriers for the preparation of unit dose forms which can be used for oral administration are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art. The inventive vaccine or composition can additionally contain one or more auxiliary substances in order to further increase the immunogenicity. A synergistic action of the nucleic acid contained in the inventive composition and of an auxiliary substance, which may be optionally be co-formulated (or separately formulated) with the inventive vaccine or composition as described above, is preferably achieved thereby. Depending on the various types of auxiliary substances, various mechanisms may play a role in this respect.
Further additives which may be included in the inventive vaccine or composition are emulsifiers, such as, for example, Tween; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet-forming agents; stabilizers; antioxidants; preservatives.
The inventive vaccine or composition can also additionally contain any further compound, which is known to be immune-stimulating due to its binding affinity (as ligands) to human Tolllike receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 or TLR13.
Another class of compounds, which may be added to an inventive vaccine or composition in this context, may be CpG nucleic acids, in particular CpG-RNA or CpG-DNA. A CpG-RNA or CpG- DNA 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 is preferably in the form of CpG-RNA, more preferably in the form of single-stranded CpG-RNA (ss CpG-RNA). The CpG nucleic acid preferably contains at least one or more
(mitogenic) cytosine/guanine dinucleotide sequence(s) (CpG motif(s)). According to a first preferred alternative, at least one CpG motif contained in these sequences, that is to say the C (cytosine) and the G (guanine) of the CpG motif, is unmethylated. All further cytosines or guanines optionally contained in these sequences can be either methylated or unmethylated. According to a further preferred alternative, however, the C (cytosine) and the G (guanine) of the CpG motif can also be present in methylated form.
As used herein, the term 'inventive composition' may refer to the inventive composition comprising at least one artificial nucleic acid. Likewise, the term 'inventive vaccine', as used in this context, may refer to an inventive vaccine, which is based on the artificial nucleic acid, i.e. which comprises at least one artificial nucleic acid or which comprises the inventive composition comprising said artificial nucleic acid. The composition or vaccine may be designed as a ready-to-use injectable formulation. In this case, it is preferably provided as a sterile aqueous solution, or suspension of nanoparticles, preferably with a pH in the range from about 4 to about 9, or more preferably from about 4.5 to about 8.5. The osmolality of such liquid composition is preferably from about 150 to about 500 mOsmol/kg, and more preferably from about 200 to about 400 mOsmol/kg. If the composition is to be injected intravenously, the pH may also be in the range from about 4.5 to about 8, or from about 5 to about 7.5; and the osmolality may in this case preferably be selected in the range from about 220 to about 350 mOsmol/kg, or from about 250 to about 330 mOsmol/kg, respectively.
Alternatively, the composition may be formulated as a concentrated form which requires dilution or even reconsitution before use. For example, it may be in the form of a liquid concentrate, which could be an aqueous and/or organic liquid formulation which requires dilution with an aqueous solvent or diluent. If the liquid concentrate comprises an organic solvent, such solvent is preferably selected from water-miscible organic solvents with relatively low toxicity such as ethanol or propylene glycol.
In one of the preferred embodiments, the composition of the invention is provided as a dry formulation for reconstitution with a liquid carrier such as to generate a liquid formulation suitable for injection. The dry formulation may be a powder, or a lyophilised form.
In order to optimise its performance, stability or tolerability, the composition may optionally comprise pharmaceutical excipients as required or useful. Potentially useful excipients include acids, bases, osmotic agents, antioxidants, stabilisers, surfactants, synergists, colouring agents, thickening agents, bulking agents, and - if required - preservatives.
The invention is also directed to a kit, particularly kits of parts, comprising the constituents of the composition of the invention as defined herein. The composition of the invention may be accommodated in one or different parts of the kit. In one embodiment, the invention provides a kit for preparing any such composition as defined herein, the kit comprising a first kit component comprising the cationisable or permanently cationic lipid or lipidoid, and a second kit component comprising the nucleic acid compound.
For example, the first kit component may be provided as a sterile solid composition, such as a lyophilised form or powder, or as a sterile liquid composition. In addition to the cationisable or permanently cationic lipid or lipidoid, the first kit component may comprise one or more inactive ingredients as described above. Similarly, the second kit component may be formulated, for example, as a sterile solid or liquid composition and also contain one or more additional inactive ingredients in addition to the nucleic acid compound. The composition of the invention is obtained by combining and optionally mixing the content of the two components.
Alternatively, but also within the scope of the invention, a kit is provided which comprises a first kit component comprising at least one cationisable or permanently cationic lipid or lipidoid and at least one nucleic acid compound, formulated e.g. as a sterile solid or liquid formulation, and a second kit component comprising a liquid carrier for dissolving or dispersing the content of the first kit component such as to obtain a composition of the invention as described above. Again, the kit components are preferably provided in sterile form, whether solid or liquid, and each of them may comprise one or more additional excipient, or inactive ingredient. In case the kit or kit of parts comprises a plurality of nucleic acids, one component of the kit can comprise only one, several or all nucleic acids comprised in the kit. In an alternative embodiment, each nucleic acid is comprised in a different component of the kit such that each component forms a part of the kit. Also, more than one nucleic acid may be comprised in a first component as part of the kit, whereas one or more other (second, third etc.) components (providing one or more other parts of the kit) may either contain one or more than one nucleic acids, which may be identical or partially identical or different from the first component.
Optionally, any of the kit components described above comprising the cationisable or permanently cationic lipid and/or the nucleic acid compound are formulated to represent concentrates, whether in solid or liquid form, and may be designed to be diluted by a
biocompatible or physiologically tolerable liquid carrier which may optionally not part of the kit, such as sterile saline solution, sterile buffer, or other solutions that are frequently used as liquid diluents for injectable drugs.
In this context of injectable formulations, the expression "liquid carrier" typically means a well-tolerated aqueous injectable liquid composition having a physiologically acceptable composition, pH and osmolality.
The kit or kit of parts may furthermore contain technical instructions with information on the administration and dosage of the nucleic acid sequence, the inventive pharmaceutical composition or of any of its components or parts, e.g. if the kit is prepared as a kit of parts.
The nanoparticles, the kit and the composition as described above are particularly useful to deliver nucleic acid cargo to living cells such as to transfect the cells with the nucleic acid. This may serve a scientific research purpose, a diagnostic application or a therapy. In one of the preferred embodiments, the nanoparticle(s) or the composition or the kit is used as a
medicament. As used herein, a "medicament" means any compound, material, composition or formulation which is useful for the prophylaxis, prevention, treatment, cure, palliative treatment, amelioration, management, improvement, delay, stabilisation, or the prevention or delay of reoccurrence or spreading of a disease or condition, including the prevention, treatment or amelioration of any symptom of a disease or condition.
More specifically, the nanoparticle or composition or kit as described above may be used in the prophylaxis, treatment and/or amelioration of diseases selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e. (hereditary) diseases, or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency.
Such use may also be described as a method of treating a subject, in particular a human subject, having developed or being at risk of developing a disease selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e. (hereditary) diseases, or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency, by administering an effective amount of the composition and/or the nanoparticles of the invention. In this context, the administration of the composition and/or the nanoparticles may optionally include a reconstitution or dilution step, for example if the composition is provided as a concentrate or as a dry solid composition for reconstitution. Moreover, references to the composition in this respect should be understood as referring also to a composition obtained by reconstituting or diluting the solid or concentrated composition.
The actual administration of the composition may be effected by any known mode and route of administration, i.e. injectable or parenteral, or non-invasive, systemic or non-systemic. For example, systemic exposure may be achieved by injection or infusion of the composition into the bloodstream of a subject, in particular by intravenous or intraarterial injection or infusion. Alternatively, and according to one of the particularly preferred embodiments, the composition is administered to a subject by extravascular administration. Such extravascular administration includes any mode and route of administration which avoids the direct injection or infusion into the bloodstream. Preferably, the extravascular injection is nevertheless performed by injecting, infusing or implanting the composition, even though not into a blood vessel, but to another site of administration, which site may be referred to as a local or locoregional site of administration. In this context, locoregional means restricted to a region of a body. The effect of local or locoregional administration is that the composition may not be rapidly diluted and distributed throughout the body of a subject by means of its systemic circulation, but that it may be retained locally or locoregionally at or near the site of administration, which is associated with an increased likelihood of local interaction with target structures or cell and a decreased systemic exposure.
In one of the particularly preferred embodiments, the composition and/or nanoparticle of the invention is administered by extravascular, local or locoregional injection, infusion or implantation, in particular intradermal, subcutaneous, intramuscular, interstitial, locoregional, intravitreal, periocular, intratumoural, intralymphatic, intranodal, intra-articular, intrasynovial, periarticular, intraperitoneal, intra-abdominal, intracardial, intrapericardial, intraventricular, intrapleural, perineural, intrathoracic, epidural, intradural, peridural, intrathecal, intramedullary intracerebral, intracavernous, intracorporus cavernosum, intraprostatic, intratesticular, intracartilaginous, intraosseous, intradiscal, intraspinal, intracaudal, intrabursal, intragingival, intraovarian, intrauterine, periodontal, retrobulbar, subarachnoid, subconjunctival or intralesional injection, infusion or implantation.
Alternatively, the composition is administered topically to the skin or mucosa of a subject, in particular by dermal or cutaneous, nasal, buccal, sublingual, otic or auricular, ophthalmic, conjunctival, vaginal, rectal, intracervical, endosinusial, laryngeal, oropharyngeal, ureteral, or urethral administration.
Optionally, the composition may be administered to the respiratory system by inhalation, in particular by aerosol administration to the lungs, bronchi, bronchioli, alveoli, or paranasal sinuses. Optionally, the composition may also be administered to a subject by transdermal or percutaneous administration.
The present invention furthermore provides several applications and uses of the composition or nanoparticle or kit or vaccine. In particular, the (pharmaceutical) composition(s) or the vaccine may be used for human or for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine. In a further aspect, the invention provides the artificial nucleic acid, the inventive composition comprising at least one artificial nucleic acid, inventive composition comprising at least one polypeptide, the inventive vaccine or the inventive kit or kit of parts for use in a method of prophylactic (pre-exposure prophylaxis or post-exposure prophylaxis) and/or therapeutic treatment of e.g. virus infections. Consequently, in a further aspect, the present invention is directed to the first medical use of the artificial nucleic acid, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts as defined herein as a medicament. Particularly, the invention provides the use of an artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprising at least one e.g. virus protein or peptide as defined herein, or a fragment or variant thereof as described herein for the preparation of a medicament.
According to another aspect, the present invention is directed to the second medical use of the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts for the treatment of an infection with e.g. a virus or a disease or disorders related to an infection.
The inventive composition or the inventive vaccine, in particular the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein or the inventive composition comprising at least one inventive polypeptide, can be administered, for example, systemically or locally. Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes. Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial, intrapulmonal, intracardial, and sublingual injections. More preferably, vaccines may be administered by an intradermal, subcutaneous, or
intramuscular route. Inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid) form. Preferably, the inventive vaccine may be administered by conventional needle injection or needle-free jet injection. In a preferred embodiment the inventive vaccine or composition may be administered by jet injection as defined herein, preferably intramuscularly or intradermally, more preferably intradermally.
In a preferred embodiment, a single dose of the artificial nucleic acid, composition or vaccine comprises a specific amount of the artificial nucleic acid as disclosed herein. Preferably, the artificial nucleic acid is provided in an amount of at least 40 μg per dose, preferably in an amount of from 40 to 700 μg per dose, more preferably in an amount of from 80 to 400 μg per dose. More specifically, in the case of intradermal injection, which is preferably carried out by using a conventional needle, the amount of the inventive artificial nucleic acid comprised in a single dose is typically at least 200 μg, preferably from 200 μg to 1.000 μg, more preferably from 300 μg to 850 μg, even more preferably from 300 μg to 700 μg. In the case of intradermal injection, which is preferably carried out via jet injection (e.g. using a Tropis device), the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 μg, preferably from 80 μg to 700 μg, more preferably from 80 μg to 400 μg. Moreover, in the case of intramuscular injection, which is preferably carried out by using a conventional needle or via jet injection, the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 μg, preferably from 80 μg to 1.000 μg, more preferably from 80 μg to 850 μg, even more preferably from 80 μg to 700 μg.
The immunization protocol for the treatment or prophylaxis of e.g. a virus infection, i.e the immunization of a subject against e.g. a virus, typically comprises a series of single doses or dosages of the inventive composition or the inventive vaccine. A single dosage, as used herein, refers to the initial/first dose, a second dose or any further doses, respectively, which are preferably administered in order to "boost" the immune reaction.
According to a preferred embodiment, the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts is provided for use in treatment or prophylaxis, preferably treatment or prophylaxis of e.g. a virus infection or a related disorder or disease, wherein the treatment or prophylaxis comprises the administration of a further active pharmaceutical ingredient. More preferably, in the case of the inventive vaccine or composition, which is based on the inventive artificial nucleic acid, a polypeptide may be coadministered as a further active pharmaceutical ingredient. For example, at least one e.g. virus protein or peptide as described herein, or a fragment or variant thereof, may be co-administered in order to induce or enhance an immune response. Likewise, in the case of the inventive vaccine or composition, which is based on the inventive polypeptide as described herein, an artificial nucleic acid as described herein may be co-administered as a further active pharmaceutical ingredient. For example, an artificial nucleic acid as described herein encoding at least one polypeptide as described herein may be co-administered in order to induce or enhance an immune response. A further component of the inventive vaccine or composition may be an
immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably
immunoglobulins directed against e.g. a virus. Preferably, such a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA. Such an immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the inventive artificial nucleic acid or by the inventive polypeptide.
In a further aspect the invention provides a method of treating or preventing a disorder, wherein the disorder is preferably an infection with e.g. a virus or a disorder related to an infection with e.g. a virus, wherein the method comprises administering to a subject in need thereof the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts.
In particular, such a method may preferably comprise the steps of: a) providing the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts; b) applying or administering the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts to a tissue or an organism; c) optionally administering immunoglobuline (IgGs) against e.g. the virus.
According to a further aspect, the present invention also provides a method for expression of at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, wherein the method preferably comprises the following steps: a) providing the inventive artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, preferably as defined herein, or a composition comprising said artificial nucleic acid; and b) applying or administering the inventive artificial nucleic acid or the inventive composition comprising said artificial nucleic acid to an expression system, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
The method may be applied for laboratory, for research, for diagnostic, for commercial production of peptides or proteins and/or for therapeutic purposes. In this context, typically after preparing the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, e.g. in naked or complexed form or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein. The method may be carried out in vitro, in vivo or ex vivo. The method may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of infectious diseases, or a related disorder.
In this context, in vitro is defined herein as transfection or transduction of the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein into cells in culture outside of an organism; in vivo is defined herein as transfection or transduction of the inventive artificial nucleic acid or of the inventive composition or vaccine into cells by application of the inventive mRNA or of the inventive composition to the whole organism or individual and ex vivo is defined herein as transfection or transduction of the inventive artificial nucleic acid or of the inventive composition or vaccine into cells outside of an organism or individual and subsequent application of the transfected cells to the organism or individual.
Likewise, according to another aspect, the present invention also provides the use of the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, preferably for diagnostic or therapeutic purposes, for expression of e.g. an encoded virus antigenic peptide or protein, e.g. by applying or administering the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism. The use may be applied for a (diagnostic) laboratory, for research, for diagnostics, for commercial production of peptides or proteins and/or for therapeutic purposes. In this context, typically after preparing the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, preferably in naked form or complexed form, or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein. The use may be carried out in vitro, in vivo or ex vivo. The use may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of e.g. a virus infection or a related disorder.
In a particularly preferred embodiment, the invention provides the artificial nucleic acid, the inventive composition or the inventive vaccine for use as defined herein, preferably for use as a medicament, for use in treatment or prophylaxis, preferably treatment or prophylaxis of a e.g. a virus infection or a related disorder, or for use as a vaccine.
The composition or vaccine may be administered by conventional needle injection or needle-free jet injection, e.g. into, adjacent to and/or in close proximity to tumor tissue. In a preferred embodiment, the inventive composition or the inventive pharmaceutical composition is administered by jet injection. Jet injection refers to a needle-free injection method, wherein a fluid comprising the inventive composition and, optionally, further suitable excipients is forced through an orifice, thus generating an ultra-fine liquid stream of high pressure that is capable of penetrating mammalian skin. In principle, the liquid stream forms a hole in the skin, through which the liquid stream is pushed into the target tissue, e.g. tumor tissue. Accordingly, jet injection may be used e.g. for intratumoral application of the inventive composition.
In other embodiments, the inventive composition or the inventive pharmaceutical composition may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intranodal, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques.
Further particularly preferred administration routes are intradermal and intramuscular injection.
Despite, the inventive pharmaceutical composition may comprise further components for facilitating administration and uptake of components of the pharmaceutical composition. Such further components may be an appropriate carrier or vehicle, antibacterial and/or antiviral agents.
A further component of the inventive pharmaceutical composition may be an
immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc. Preferably, such a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA. The inventive pharmaceutical composition typically comprises a "safe and effective amount" of the components of the inventive pharmaceutical composition, particularly of the RNA molecule(s) as defined herein. As used herein, a "safe and effective amount" means an amount of the RNA molecule(s) as defined herein as such that is sufficient to significantly induce a positive modification of e.g. a tumor or cancer disease. At the same time, however, a "safe and effective amount" is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
The inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general.
The present invention furthermore provides several applications and uses of the nucleic acid sequence as defined herein, of the inventive composition comprising a plurality of nucleic acid sequences as defined herein, of the inventive pharmaceutical composition, comprising the nucleic acid sequence as defined herein or of kits comprising same.
According to one specific aspect, the present invention is directed to the first medical use of the nucleic acid sequence as defined herein or of the inventive composition comprising a plurality of nucleic acid sequences as defined herein as a medicament, particularly in gene therapy, preferably for the treatment of diseases as defined herein.
According to another aspect, the present invention is directed to the second medical use of the nucleic acid sequence as defined herein or of the inventive composition comprising a plurality of nucleic acid sequences as defined herein, for the treatment of diseases as defined herein, preferably to the use of the nucleic acid sequence as defined herein, of the inventive composition comprising a plurality of nucleic acid sequences as defined herein, of a pharmaceutical composition comprising same or of kits comprising same for the preparation of a medicament for the prophylaxis, treatment and/or amelioration of diseases as defined herein. Preferably, the pharmaceutical composition is used or to be administered to a patient in need thereof for this purpose.
Preferably, diseases as mentioned herein are preferably selected from infectious diseases, neoplasms (e.g. cancer or tumor diseases), diseases of the blood and blood-forming organs, endocrine, nutritional and metabolic diseases, diseases of the nervous system, diseases of the circulatory system, diseases of the respiratory system, diseases of the digestive system, diseases of the skin and subcutaneous tissue, diseases of the musculoskeletal system and connective tissue, and diseases of the genitourinary system.
In this context particularly preferred are inherited diseases selected from: lp36 deletion syndrome; 18p deletion syndrome; 21-hydroxylase deficiency; 45,X (Turner syndrome);
47,XX,+21 (Down syndrome); 47,XXX (triple X syndrome); 47,XXY (Klinefelter syndrome);
47,XY,+21 (Down syndrome); 47,XYY syndrome; 5-ALA dehydratase-deficient porphyria (ALA dehydratase deficiency); 5-aminolaevulinic dehydratase deficiency porphyria (ALA dehydratase deficiency); 5p deletion syndrome (Cri du chat) 5p- syndrome (Cri du chat); A-T (ataxia- telangiectasia); AAT (alpha- 1 antitrypsin deficiency); Absence of vas deferens (congenital bilateral absence of vas deferens); Absent vasa (congenital bilateral absence of vas deferens); aceruloplasminemia; ACG2 (achondrogenesis type II); ACH (achondroplasia); Achondrogenesis type II; achondroplasia; Acid beta-glucosidase deficiency (Gaucher disease type 1);
Acrocephalosyndactyly (Apert) (Apert syndrome); acrocephalosyndactyly, type V (Pfeiffer syndrome); Acrocephaly (Apert syndrome); Acute cerebral Gaucher's disease (Gaucher disease type 2); acute intermittent porphyria; ACY2 deficiency (Canavan disease); AD (Alzheimer's disease); Adelaide-type craniosynostosis (Muenke syndrome); Adenomatous Polyposis Coli (familial adenomatous polyposis); Adenomatous Polyposis of the Colon (familial adenomatous polyposis); ADP (ALA dehydratase deficiency); adenylosuccinate lyase deficiency; Adrenal gland disorders (21-hydroxylase deficiency); Adrenogenital syndrome (21-hydroxylase deficiency); Adrenoleukodystrophy; AIP (acute intermittent porphyria); AIS (androgen insensitivity syndrome); AKU (alkaptonuria); ALA dehydratase porphyria (ALA dehydratase deficiency); ALA- D porphyria (ALA dehydratase deficiency); ALA dehydratase deficiency; Alcaptonuria
(alkaptonuria); Alexander disease; alkaptonuria; Alkaptonuric ochronosis (alkaptonuria); alpha-1 antitrypsin deficiency; alpha-1 proteinase inhibitor (alpha-1 antitrypsin deficiency); alpha-1 related emphysema (alpha-1 antitrypsin deficiency); Alpha-galactosidase A deficiency (Fabry disease); ALS (amyotrophic lateral sclerosis); Alstrom syndrome; ALX (Alexander disease);
Alzheimer disease; Amelogenesis Imperfecta; Amino levulinic acid dehydratase deficiency (ALA dehydratase deficiency); Aminoacylase 2 deficiency (Canavan disease); amyotrophic lateral sclerosis; Anderson-Fabry disease (Fabry disease); androgen insensitivity syndrome; Anemia; Anemia, hereditary sideroblastic (X-linked sideroblastic anemia); Anemia, sex-linked
hypochromic sideroblastic (X-linked sideroblastic anemia); Anemia, splenic, familial (Gaucher disease); Angelman syndrome; Angiokeratoma Corporis Diffusum (Fabry's disease);
Angiokeratoma diffuse (Fabry's disease); Angiomatosis retinae (von Hippel-Lindau disease); ANH1 (X-linked sideroblastic anemia); APC resistance, Leiden type (factor V Leiden
thrombophilia); Apert syndrome; AR deficiency (androgen insensitivity syndrome); AR-CMT2 ee (Charcot-Mare-Tooth disease, type 2); Arachnodactyly (Marfan syndrome); ARNSHL (Nonsyndromic deafness autosomal recessive); Arthro-ophthalmopathy, hereditary progressive (Stickler syndrome COL2A1); Arthrochalasis multiplex congenita (Ehlers-Danlos syndrome arthrochalasia type); AS (Angelman syndrome); Asp deficiency (Canavan disease); Aspa deficiency (Canavan disease); Aspartoacylase deficiency (Canavan disease); ataxia-telangiectasia; Autism-Dementia-Ataxia-Loss of Purposeful Hand Use syndrome (Rett syndrome); autosomal dominant juvenile ALS (amyotrophic lateral sclerosis, type 4); Autosomal dominant opitz G/BBB syndrome (22qll.2 deletion syndrome); autosomal recessive form of juvenile ALS type 3 (Amyotrophic lateral sclerosis type 2); Autosomal recessive nonsyndromic hearing loss
(Nonsyndromic deafness autosomal recessive); Autosomal Recessive Sensorineural Hearing Impairment and Goiter (Pendred syndrome); AxD (Alexander disease); Ayerza syndrome (primary pulmonary hypertension); B variant of the Hexosaminidase GM2 gangliosidosis (Sandhoff disease); BANF (neurofibromatosis 2); Beare-Stevenson cutis gyrata syndrome; Benign paroxysmal peritonitis (Mediterranean fever, familial); Benjamin syndrome; beta thalassemia; BH4 Deficiency (tetrahydrobiopterin deficiency); Bilateral Acoustic Neurofibromatosis
(neurofibromatosis 2); biotinidase deficiency; bladder cancer; Bleeding disorders (factor V Leiden thrombophilia); Bloch-Sulzberger syndrome (incontinentia pigmenti); Bloom syndrome; Bone diseases; Bone marrow diseases (X-linked sideroblastic anemia); Bonnevie-Ullrich syndrome (Turner syndrome); Bourneville disease (tuberous sclerosis); Bourneville phakomatosis
(tuberous sclerosis); Brain diseases (prion disease); breast cancer; Birt-Hogg-Dube syndrome; Brittle bone disease (osteogenesis imperfecta); Broad Thumb-Hallux syndrome (Rubinstein- Taybi syndrome); Bronze Diabetes (hemochromatosis); Bronzed cirrhosis (hemochromatosis); Bulbospinal muscular atrophy, X-linked (Kennedy disease); Burger-Grutz syndrome (lipoprotein lipase deficiency, familial); CADASIL; CGD Chronic Granulomatous Disorder; Camptomelic dysplasia; Canavan disease; Cancer; Cancer Family syndrome (hereditary nonpolyposis colorectal cancer); Cancer of breast (breast cancer); Cancer of the bladder (bladder cancer); Carboxylase Deficiency, Multiple, Late-Onset (biotinidase deficiency); Cardiomyopathy (Noonan syndrome); Cat cry syndrome (Cri du chat); CAVD (congenital bilateral absence of vas deferens); Caylor cardiofacial syndrome (22qll.2 deletion syndrome); CBAVD (congenital bilateral absence of vas deferens); Celiac Disease; CEP (congenital erythropoietic porphyria); Ceramide trihexosidase deficiency (Fabry disease); Cerebelloretinal Angiomatosis, familial (von Hippel-Lindau disease); Cerebral arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL); Cerebral autosomal dominant ateriopathy with subcortical infarcts and leukoencephalopathy (CADASIL); Cerebral sclerosis (tuberous sclerosis); Cerebroatrophic Hyperammonemia (Rett syndrome); Cerebroside Lipidosis syndrome (Gaucher disease); CF (cystic fibrosis); CH (congenital hypothyroidism); Charcot disease (amyotrophic lateral sclerosis); Charcot-Marie-Tooth disease; Chondrodystrophia (achondroplasia); Chondrodystrophy syndrome (achondroplasia); Chondrodystrophy with sensorineural deafness (otospondylomegaepiphyseal dysplasia);
Chondrogenesis imperfecta (achondrogenesis, type II); Choreoathetosis self-mutilation hyperuricemia syndrome (Lesch-Nyhan syndrome); Classic Galactosemia (galactosemia);
Classical Ehlers-Danlos syndrome (Ehlers-Danlos syndrome classical type) ; Classical
Phenylketonuria (phenylketonuria); Cleft lip and palate (Stickler syndrome); Cloverleaf skull with thanatophoric dwarfism (Thanatophoric dysplasia type 2); CLS (Coffin-Lowry syndrome); CMT (Charcot- Marie-Tooth disease); Cockayne syndrome; Coffin-Lowry syndrome; collagenopathy, types II and XI; Colon Cancer, familial Nonpolyposis (hereditary nonpolyposis colorectal cancer); Colon cancer, familial (familial adenomatous polyposis); Colorectal Cancer; Complete HPRT deficiency (Lesch-Nyhan syndrome); Complete hypoxanthine-guanine phosphoribosy transferase deficiency (Lesch-Nyhan syndrome); Compression neuropathy (hereditary neuropathy with liability to pressure palsies); Congenital adrenal hyperplasia (21-hydroxylase deficiency);
congenital bilateral absence of vas deferens (Congenital absence of the vas deferens); Congenital erythropoietic porphyria; Congenital heart disease; Congenital hypomyelination (Charcot-Marie- Tooth disease Type 1/Charcot-Marie-Tooth disease Type 4); Congenital hypothyroidism;
Congenital methemoglobinemia (Methemoglobinemia Congenital methaemoglobinaemia);
Congenital osteosclerosis (achondroplasia); Congenital sideroblastic anaemia (X-linked sideroblastic anemia); Connective tissue disease; Conotruncal anomaly face syndrome (22qll.2 deletion syndrome); Cooley's Anemia (beta thalassemia); Copper storage disease (Wilson disease); Copper transport disease (Menkes disease); Coproporphyria, hereditary (hereditary coproporphyria); Coproporphyrinogen oxidase deficiency (hereditary coproporphyria); Cowden syndrome; CPO deficiency (hereditary coproporphyria); CPRO deficiency (hereditary
coproporphyria); CPX deficiency (hereditary coproporphyria); Craniofacial dysarthrosis (Crouzon syndrome); Craniofacial Dysostosis (Crouzon syndrome); Cretinism (congenital hypothyroidism); Creutzfeldt-Jakob disease (prion disease); Cri du chat (Crohn's disease, fibrostenosing); Crouzon syndrome; Crouzon syndrome with acanthosis nigricans (Crouzonodermoskeletal syndrome); Crouzonodermoskeletal syndrome; CS (Cockayne syndrome) (Cowden syndrome); Curschmann- Batten-Steinert syndrome (myotonic dystrophy); cutis gyrata syndrome of Beare-Stevenson (Beare-Stevenson cutis gyrata syndrome); Disorder Mutation Chromosome; D-glycerate dehydrogenase deficiency (hyperoxaluria, primary); Dappled metaphysis syndrome
(spondyloepimetaphyseal dysplasia, Strudwick type); DAT - Dementia Alzheimer's type
(Alzheimer disease); Genetic hypercalciuria (Dent's disease); DBMD (muscular dystrophy, Duchenne and Becker types); Deafness with goiter (Pendred syndrome); Deafness-retinitis pigmentosa syndrome (Usher syndrome); Deficiency disease, Phenylalanine Hydroxylase (phenylketonuria); Degenerative nerve diseases; de Grouchy syndrome 1 (De Grouchy
Syndrome); Dejerine-Sottas syndrome (Charcot-Marie-Tooth disease); Delta-aminolevulinate dehydratase deficiency porphyria (ALA dehydratase deficiency); Dementia (CADASIL);
demyelinogenic leukodystrophy (Alexander disease); Dermatosparactic type of Ehlers-Danlos syndrome (Ehlers-Danlos syndrome dermatosparaxis type); Dermatosparaxis (Ehlers-Danlos syndrome dermatosparaxis type) ; developmental disabilities; dHMN (Amyotrophic lateral sclerosis type 4 ); DHMN-V (distal spinal muscular atrophy, type V); DHTR deficiency (androgen insensitivity syndrome); Diffuse Globoid Body Sclerosis (Krabbe disease); DiGeorge syndrome; Dihydrotestosterone receptor deficiency (androgen insensitivity syndrome); distal spinal muscular atrophy, type V; DM1 (Myotonic dystrophy typel); DM2 (Myotonic dystrophy type2); Down syndrome; DSMAV (distal spinal muscular atrophy, type V); DSN (Charcot-Marie-Tooth disease type 4); DSS (Charcot-Marie-Tooth disease, type 4); Duchenne/Becker muscular dystrophy (muscular dystrophy, Duchenne and Becker types); Dwarf, achondroplastic
(achondroplasia); Dwarf, thanatophoric (thanatophoric dysplasia); Dwarfism; Dwarfism-retinal atrophy-deafness syndrome (Cockayne syndrome); dysmyelinogenic leukodystrophy (Alexander disease); Dystrophia myotonica (myotonic dystrophy); dystrophia retinae pigmentosa-dysostosis syndrome (Usher syndrome); Early-Onset familial alzheimer disease (EOFAD) (Alzheimer disease); EDS (Ehlers-Danlos syndrome); Ehlers-Danlos syndrome; Ekman-Lobstein disease (osteogenesis imperfecta); Entrapment neuropathy (hereditary neuropathy with liability to pressure palsies); Epiloia (tuberous sclerosis); EPP (erythropoietic protoporphyria);
Erythroblastic anemia (beta thalassemia); Erythrohepatic protoporphyria (erythropoietic protoporphyria); Erythroid 5-aminolevulinate synthetase deficiency (X-linked sideroblastic anemia); Erythropoietic porphyria (congenital erythropoietic porphyria); Erythropoietic protoporphyria; Erythropoietic uroporphyria (congenital erythropoietic porphyria); Eye cancer (retinoblastoma FA - Friedreich ataxia); Fabry disease; Facial injuries and disorders; Factor V Leiden thrombophilia; FALS (amyotrophic lateral sclerosis); familial acoustic neuroma
(neurofibromatosis type II); familial adenomatous polyposis; familial Alzheimer disease (FAD) (Alzheimer disease); familial amyotrophic lateral sclerosis (amyotrophic lateral sclerosis);
familial dysautonomia; familial fat-induced hypertriglyceridemia (lipoprotein lipase deficiency, familial); familial hemochromatosis (hemochromatosis); familial LPL deficiency (lipoprotein lipase deficiency, familial); familial nonpolyposis colon cancer (hereditary nonpolyposis colorectal cancer); familial paroxysmal polyserositis (Mediterranean fever, familial); familial PCT (porphyria cutanea tarda); familial pressure sensitive neuropathy (hereditary neuropathy with liability to pressure palsies); familial primary pulmonary hypertension (FPPH) (primary pulmonary hypertension); Familial Turner syndrome (Noonan syndrome); familial vascular leukoencephalopathy (CADASIL); FAP (familial adenomatous polyposis); FD (familial dysautonomia); Female pseudo-Turner syndrome (Noonan syndrome); Ferrochelatase deficiency (erythropoietic protoporphyria); ferroportin disease (Haemochromatosis type 4); Fever (Mediterranean fever, familial); FG syndrome; FGFR3-associated coronal synostosis (Muenke syndrome); Fibrinoid degeneration of astrocytes (Alexander disease); Fibrocystic disease of the pancreas (cystic fibrosis); FMF (Mediterranean fever, familial); Foiling disease (phenylketonuria); fra(X) syndrome (fragile X syndrome); fragile X syndrome; Fragilitas ossium (osteogenesis imperfecta); FRAXA syndrome (fragile X syndrome); FRDA (Friedreich's ataxia); Friedreich ataxia (Friedreich's ataxia); Friedreich's ataxia; FXS (fragile X syndrome); G6PD deficiency;
Galactokinase deficiency disease (galactosemia); Galactose- 1-phosphate uridyl-transferase deficiency disease (galactosemia); galactosemia; Galactosylceramidase deficiency disease (Krabbe disease); Galactosylceramide lipidosis (Krabbe disease); galactosylcerebrosidase deficiency (Krabbe disease); galactosylsphingosine lipidosis (Krabbe disease); GALC deficiency (Krabbe disease); GALT deficiency (galactosemia); Gaucher disease; Gaucher-like disease (pseudo- Gaucher disease); GBA deficiency (Gaucher disease type 1); GD (Gaucher's disease); Genetic brain disorders; genetic emphysema (alpha-1 antitrypsin deficiency); genetic hemochromatosis (hemochromatosis); Giant cell hepatitis, neonatal (Neonatal hemochromatosis); GLA deficiency (Fabry disease); Glioblastoma, retinal (retinoblastoma); Glioma, retinal (retinoblastoma); globoid cell leukodystrophy (GCL, GLD) (Krabbe disease); globoid cell leukoencephalopathy (Krabbe disease); Glucocerebrosidase deficiency (Gaucher disease); Glucocerebrosidosis (Gaucher disease); Glucosyl cerebroside lipidosis (Gaucher disease) ; Glucosylceramidase deficiency (Gaucher disease); Glucosylceramide beta-glucosidase deficiency (Gaucher disease);
Glucosylceramide lipidosis (Gaucher disease); Glyceric aciduria (hyperoxaluria, primary); Glycine encephalopathy (Nonketotic hyperglycinemia); Glycolic aciduria (hyperoxaluria, primary); GM2 gangliosidosis, type 1 (Tay-Sachs disease); Goiter-deafness syndrome (Pendred syndrome);
Graefe-Usher syndrome (Usher syndrome); Gronblad-Strandberg syndrome (pseudoxanthoma elasticum); Guenther porphyria (congenital erythropoietic porphyria); Gunther disease
(congenital erythropoietic porphyria); Haemochromatosis (hemochromatosis); Hallgren syndrome (Usher syndrome); Harlequin Ichthyosis; Hb S disease (sickle cell anemia); HCH (hypochondroplasia); HCP (hereditary coproporphyria); Head and brain malformations; Hearing disorders and deafness; Hearing problems in children; HEF2A (hemochromatosis type 2); HEF2B (hemochromatosis type 2); Hematoporphyria (porphyria); Heme synthetase deficiency
(erythropoietic protoporphyria); Hemochromatoses (hemochromatosis); hemochromatosis; hemoglobin M disease (methemoglobinemia beta-globin type); Hemoglobin S disease (sickle cell anemia); hemophilia; HEP (hepatoerythropoietic porphyria); hepatic AGT deficiency
(hyperoxaluria, primary); hepatoerythropoietic porphyria; Hepatolenticular degeneration syndrome (Wilson disease); Hereditary arthro-ophthalmopathy (Stickler syndrome); Hereditary coproporphyria; Hereditary dystopic lipidosis (Fabry disease); Hereditary hemochromatosis (HHC) (hemochromatosis); Hereditary Inclusion Body Myopathy (skeletal muscle regeneration); Hereditary iron-loading anemia (X-linked sideroblastic anemia); Hereditary motor and sensory neuropathy (Charcot-Marie-Tooth disease); Hereditary motor neuronopathy (spinal muscular atrophy); Hereditary motor neuronopathy, type V (distal spinal muscular atrophy, type V); Hereditary Multiple Exostoses; Hereditary nonpolyposis colorectal cancer; Hereditary periodic fever syndrome (Mediterranean fever, familial); Hereditary Polyposis Coli (familial adenomatous polyposis); Hereditary pulmonary emphysema (alpha-1 antitrypsin deficiency); Hereditary resistance to activated protein C (factor V Leiden thrombophilia); Hereditary sensory and autonomic neuropathy type III (familial dysautonomia); Hereditary spastic paraplegia (infantile- onset ascending hereditary spastic paralysis); Hereditary spinal ataxia (Friedreich ataxia);
Hereditary spinal sclerosis (Friedreich ataxia); Herrick's anemia (sickle cell anemia);
Heterozygous OSMED (Weissenbacher-Zweymuller syndrome); Heterozygous
otospondylomegaepiphyseal dysplasia (Weissenbacher-Zweymuller syndrome); HexA deficiency (Tay-Sachs disease); Hexosaminidase A deficiency (T ay-Sachs disease); Hexosaminidase alpha- subunit deficiency (variant B) (Tay-Sachs disease); HFE-associated hemochromatosis
(hemochromatosis); HGPS (Progeria); Hippel-Lindau disease (von Hippel-Lindau disease); HLAH (hemochromatosis); HMN V (distal spinal muscular atrophy, type V); HMSN (Charcot-Marie- Tooth disease); HNPCC (hereditary nonpolyposis colorectal cancer); HNPP (hereditary neuropathy with liability to pressure palsies); homocystinuria; Homogentisic acid oxidase deficiency (alkaptonuria); Homogentisic acidura (alkaptonuria); Homozygous porphyria cutanea tarda (hepatoerythropoietic porphyria); HP1 (hyperoxaluria, primary); HP2 (hyperoxaluria, primary); HPA (hyperphenylalaninemia); HPRT - Hypoxanthine-guanine
phosphoribosyltransferase deficiency (Lesch-Nyhan syndrome); HSAN type III (familial dysautonomia) ; HSAN3 (familial dysautonomia); HSN-III (familial dysautonomia); Human dermatosparaxis (Ehlers-Danlos syndrome dermatosparaxis type); Huntington's disease;
Hutchinson-Gilford progeria syndrome (progeria); Hyperandrogenism, nonclassic type, due to 21 -hydroxylase deficiency (21 -hydroxylase deficiency); Hyperchylomicronemia, familial (lipoprotein lipase deficiency, familial); hyperglycinemia with ketoacidosis and leukopenia (propionic acidemia); Hyperlipoproteinemia type I (lipoprotein lipase deficiency, familial); hyperoxaluria, primary; hyperphenylalaninaemia (hyperphenylalaninemia);
hyperphenylalaninemia; Hypochondrodysplasia (hypochondroplasia); hypochondrogenesis; hypochondroplasia; Hypochromic anemia (X-linked sideroblastic anemia); Hypocupremia, congenital; Menkes syndrome); hypoxanthine phosphoribosyltransferse (HPRT) deficiency (Lesch-Nyhan syndrome); IAHSP (infantile-onset ascending hereditary spastic paralysis);
idiopathic hemochromatosis (hemochromatosis, type 3); Idiopathic neonatal hemochromatosis (hemochromatosis, neonatal); Idiopathic pulmonary hypertension (primary pulmonary hypertension); Immune system disorders (X-linked severe combined immunodeficiency); Incontinentia Pigmenti; Infantile cerebral Gaucher's disease (Gaucher disease type 2); Infantile Gaucher disease (Gaucher disease type 2); infantile-onset ascending hereditary spastic paralysis; Infertility; inherited emphysema (alpha-1 antitrypsin deficiency); Inherited human transmissible spongiform encephalopathies (prion disease); inherited tendency to pressure palsies (hereditary neuropathy with liability to pressure palsies); Insley-Astley syndrome
(otospondylomegaepiphyseal dysplasia); Intermittent acute porphyria syndrome (acute intermittent porphyria); Intestinal polyposis-cutaneous pigmentation syndrome (Peutz-Jeghers syndrome); IP (incontinentia pigmenti); Iron storage disorder (hemochromatosis); Isodicentric 15 (idicl5); Isolated deafness (nonsyndromic deafness); Jackson-Weiss syndrome; JH
(Haemochromatosis type 2); Joubert syndrome; JPLS (Juvenile Primary Lateral Sclerosis);
juvenile amyotrophic lateral sclerosis (Amyotrophic lateral sclerosis type 2); Juvenile gout, choreoathetosis, mental retardation syndrome (Lesch-Nyhan syndrome); juvenile hyperuricemia syndrome (Lesch-Nyhan syndrome); JWS (Jackson-Weiss syndrome); KD (X-linked spinal-bulbar muscle atrophy); Kennedy disease (X-linked spinal-bulbar muscle atrophy); Kennedy spinal and bulbar muscular atrophy (X-linked spinal-bulbar muscle atrophy); Kerasin histiocytosis (Gaucher disease); Kerasin lipoidosis (Gaucher disease); Kerasin thesaurismosis (Gaucher disease); ketotic glycinemia (propionic acidemia); ketotic hyperglycinemia (propionic acidemia); Kidney diseases (hyperoxaluria, primary); Klinefelter syndrome; Klinefelter's syndrome; Kniest dysplasia; Krabbe disease; Lacunar dementia (CADASIL); Langer-Saldino achondrogenesis (achondrogenesis, type II); Langer-Saldino dysplasia (achondrogenesis, type II); Late-onset Alzheimer disease (Alzheimer disease type 2); Late-onset familial Alzheimer disease (AD2) (Alzheimer disease type 2); late- onset Krabbe disease (LOKD) (Krabbe disease); Learning Disorders (Learning disability);
Lentiginosis, perioral (Peutz-Jeghers syndrome); Lesch-Nyhan syndrome; Leukodystrophies; leukodystrophy with Rosenthal fibers (Alexander disease); Leukodystrophy, spongiform
(Canavan disease); LFS (Li-Fraumeni syndrome); Li-Fraumeni syndrome; Lipase D deficiency (lipoprotein lipase deficiency, familial); LIPD deficiency (lipoprotein lipase deficiency, familial); Lipidosis, cerebroside (Gaucher disease); Lipidosis, ganglioside, infantile (Tay-Sachs disease); Lipoid histiocytosis (kerasin type) (Gaucher disease); lipoprotein lipase deficiency, familial; Liver diseases (galactosemia); Lou Gehrig disease (amyotrophic lateral sclerosis); Louis-Bar syndrome (ataxia-telangiectasia); Lynch syndrome (hereditary nonpolyposis colorectal cancer); Lysyl- hydroxylase deficiency (Ehlers-Danlos syndrome kyphoscoliosis type); Machado-Joseph disease (Spinocerebellar ataxia type 3); Male breast cancer (breast cancer); Male genital disorders; Male Turner syndrome (Noonan syndrome); Malignant neoplasm of breast (breast cancer); malignant tumor of breast (breast cancer); Malignant tumor of urinary bladder (bladder cancer); Mammary cancer (breast cancer); Marfan syndrome 15; Marker X syndrome (fragile X syndrome); Martin- Bell syndrome (fragile X syndrome); McCune- Albright syndrome; McLeod syndrome; MEDNIK; Mediterranean Anemia (beta thalassemia); Mediterranean fever, familial; Mega-epiphyseal dwarfism (otospondylomegaepiphyseal dysplasia); Menkea syndrome (Menkes syndrome); Menkes syndrome; Mental retardation with osteocartilaginous abnormalities (Coffin-Lowry syndrome); Metabolic disorders; Metatropic dwarfism, type II (Kniest dysplasia); Metatropic dysplasia type II (Kniest dysplasia); Methemoglobinemia beta-globin type; methylmalonic acidemia; MFS (Marfan syndrome); MHAM (Cowden syndrome); MK (Menkes syndrome); Micro syndrome; Microcephaly; MMA (methylmalonic acidemia); MNK (Menkes syndrome); Monosomy lp36 syndrome (lp36 deletion syndrome); monosomy X (Turner syndrome); Motor neuron disease, amyotrophic lateral sclerosis (amyotrophic lateral sclerosis); Movement disorders; Mowat-Wilson syndrome; Mucopolysaccharidosis (MPS I); Mucoviscidosis (cystic fibrosis);
Muenke syndrome; Multi-Infarct dementia (CADASIL); Multiple carboxylase deficiency, late-onset (biotinidase deficiency); Multiple hamartoma syndrome (Cowden syndrome); Multiple neurofibromatosis (neurofibromatosis); Muscular dystrophy; Muscular dystrophy, Duchenne and Becker type; Myotonia atrophica (myotonic dystrophy); Myotonia dystrophica (myotonic dystrophy); myotonic dystrophy; Myxedema, congenital (congenital hypothyroidism); Nance- Insley syndrome (otospondylomegaepiphyseal dysplasia); Nance-Sweeney chondrodysplasia (otospondylomegaepiphyseal dysplasia); NBIA1 (pantothenate kinase-associated
neurodegeneration); Neill-Dingwall syndrome (Cockayne syndrome); Neuroblastoma, retinal (retinoblastoma); Neurodegeneration with brain iron accumulation type 1 (pantothenate kinase- associated neurodegeneration); Neurofibromatosis type I; Neurofibromatosis type II; Neurologic diseases; Neuromuscular disorders; neuronopathy, distal hereditary motor, type V (Distal spinal muscular atrophy type V); neuronopathy, distal hereditary motor, with pyramidal features (Amyotrophic lateral sclerosis type 4); NF (neurofibromatosis); Niemann-Pick (Niemann-Pick disease); Noack syndrome (Pfeiffer syndrome); Nonketotic hyperglycinemia (Glycine
encephalopathy); Non-neuronopathic Gaucher disease (Gaucher disease type 1); Non- phenylketonuric hyperphenylalaninemia (tetrahydrobiopterin deficiency); nonsyndromic deafness; Noonan syndrome; Norrbottnian Gaucher disease (Gaucher disease type 3); Ochronosis (alkaptonuria); Ochronotic arthritis (alkaptonuria); 01 (osteogenesis imperfecta); OSMED (otospondylomegaepiphyseal dysplasia); osteogenesis imperfecta; Osteopsathyrosis
(osteogenesis imperfecta); Osteosclerosis congenita (achondroplasia); Oto-spondylo- megaepiphyseal dysplasia (otospondylomegaepiphyseal dysplasia); otospondylomegaepiphyseal dysplasia; Oxalosis (hyperoxaluria, primary); Oxaluria, primary (hyperoxaluria, primary);
pantothenate kinase-associated neurodegeneration; Patau Syndrome (Trisomy 13); PBGD deficiency (acute intermittent porphyria); PCC deficiency (propionic acidemia); PCT (porphyria cutanea tarda); PDM (Myotonic dystrophy type 2); Pendred syndrome; Periodic disease
(Mediterranean fever, familial); Periodic peritonitis (Mediterranean fever, familial); Periorificial lentiginosis syndrome (Peutz-Jeghers syndrome); Peripheral nerve disorders (familial dysautonomia); Peripheral neurofibromatosis (neurofibromatosis 1); Peroneal muscular atrophy (Charcot- Marie-Tooth disease); peroxisomal alanine:glyoxylate aminotransferase deficiency (hyperoxaluria, primary); Peutz-Jeghers syndrome; Pfeiffer syndrome; Phenylalanine
hydroxylase deficiency disease (phenylketonuria); phenylketonuria; Pheochromocytoma (von Hippel-Lindau disease); Pierre Robin syndrome with fetal chondrodysplasia (Weissenbacher- Zweymuller syndrome); Pigmentary cirrhosis (hemochromatosis); PJS (Peutz-Jeghers syndrome); PKAN (pantothenate kinase-associated neurodegeneration); PKU (phenylketonuria);
Plumboporphyria (ALA deficiency porphyria); PMA (Charcot-Marie-tooth disease); polyostotic fibrous dysplasia (McCune-Alb right syndrome); polyposis coli (familial adenomatous polyposis); polyposis, hamartomatous intestinal (Peutz-Jeghers syndrome); polyposis, intestinal, II (Peutz- Jeghers syndrome); polyps-and-spots syndrome (Peutz-Jeghers syndrome); Porphobilinogen synthase deficiency (ALA deficiency porphyria); porphyria; porphyrin disorder (porphyria); PPH (primary pulmonary hypertension); PPOX deficiency (variegate porphyria); Prader-Labhart- Willi syndrome (Prader-Willi syndrome); Prader-Willi syndrome; presenile and senile dementia (Alzheimer disease); primary hemochromatosis (hemochromatosis); primary hyperuricemia syndrome (Lesch-Nyhan syndrome); primary pulmonary hypertension; primary senile degenerative dementia (Alzheimer disease); prion disease; procollagen type EDS VII, mutant (Ehlers-Danlos syndrome arthrochalasia type); progeria (Hutchinson Gilford Progeria
Syndrome); Progeria-like syndrome (Cockayne syndrome); progeroid nanism (Cockayne syndrome); progressive chorea, chronic hereditary (Huntington) (Huntington's disease);
progressive muscular atrophy (spinal muscular atrophy); progressively deforming osteogenesis imperfecta with normal sclerae (Osteogenesis imperfecta type III); PROMM (Myotonic dystrophy type 2); propionic academia; propionyl-CoA carboxylase deficiency (propionic acidemia); protein C deficiency; protein S deficiency; protoporphyria (erythropoietic protoporphyria);
protoporphyrinogen oxidase deficiency (variegate porphyria); proximal myotonic dystrophy (Myotonic dystrophy type 2); proximal myotonic myopathy (Myotonic dystrophy type 2); pseudo- Gaucher disease; pseudo-Ullrich-Turner syndrome (Noonan syndrome); pseudoxanthoma elasticum; psychosine lipidosis (Krabbe disease); pulmonary arterial hypertension (primary pulmonary hypertension); pulmonary hypertension (primary pulmonary hypertension); PWS (Prader-Willi syndrome); PXE - pseudoxanthoma elasticum (pseudoxanthoma elasticum); Rb (retinoblastoma); Recklinghausen disease, nerve (neurofibromatosis 1); Recurrent polyserositis (Mediterranean fever, familial); Retinal disorders; Retinitis pigmentosa-deafness syndrome (Usher syndrome); Retinoblastoma; Rett syndrome; RFALS type 3 (Amyotrophic lateral sclerosis type 2); Ricker syndrome (Myotonic dystrophy type 2); Riley-Day syndrome (familial
dysautonomia); Roussy-Levy syndrome (Charcot-Marie-Tooth disease); RSTS (Rubinstein-Taybi syndrome); RTS (Rett syndrome) (Rubinstein-Taybi syndrome); RTT (Rett syndrome);
Rubinstein-Taybi syndrome; Sack-Barabas syndrome (Ehlers-Danlos syndrome, vascular type); SADDAN; sarcoma family syndrome of Li and Fraumeni (Li-Fraumeni syndrome); sarcoma, breast, leukemia, and adrenal gland (SBLA) syndrome (Li-Fraumeni syndrome); SBLA syndrome (Li-Fraumeni syndrome); SBMA (X-linked spinal-bulbar muscle atrophy); SCD (sickle cell anemia); Schwannoma, acoustic, bilateral (neurofibromatosis 2); SCIDX1 (X-linked severe combined immunodeficiency); sclerosis tuberosa (tuberous sclerosis); SDAT (Alzheimer disease); SED congenita (spondyloepiphyseal dysplasia congenita); SED Strudwick
(spondyloepimetaphyseal dysplasia, Strudwick type); SEDc (spondyloepiphyseal dysplasia congenita); SEMD, Strudwick type (spondyloepimetaphyseal dysplasia, Strudwick type); senile dementia (Alzheimer disease type 2); severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN); Shprintzen syndrome (22qll.2 deletion syndrome); sickle cell anemia; skeleton-skin-brain syndrome (SADDAN); Skin pigmentation disorders; SMA (spinal muscular atrophy); SMED, Strudwick type (spondyloepimetaphyseal dysplasia, Strudwick type); SMED, type I (spondyloepimetaphyseal dysplasia, Strudwick type); Smith Lemli Opitz Syndrome; South-African genetic porphyria (variegate porphyria); spastic paralysis, infantile onset ascending (infantile-onset ascending hereditary spastic paralysis); Speech and communication disorders; sphingolipidosis, Tay-Sachs (Tay-Sachs disease); spinal-bulbar muscular atrophy; spinal muscular atrophy; spinal muscular atrophy, distal type V (Distal spinal muscular atrophy type V); spinal muscular atrophy, distal, with upper limb predominance (Distal spinal muscular atrophy type V); spinocerebellar ataxia; spondyloepimetaphyseal dysplasia, Strudwick type; spondyloepiphyseal dysplasia congenital; spondyloepiphyseal dysplasia (collagenopathy, types II and XI); spondylometaepiphyseal dysplasia congenita, Strudwick type (spondyloepimetaphyseal dysplasia, Strudwick type); spondylometaphyseal dysplasia (SMD) (spondyloepimetaphyseal dysplasia, Strudwick type); spondylometaphyseal dysplasia, Strudwick type
(spondyloepimetaphyseal dysplasia, Strudwick type); spongy degeneration of central nervous system (Canavan disease); spongy degeneration of the brain (Canavan disease); spongy degeneration of white matter in infancy (Canavan disease); sporadic primary pulmonary hypertension (primary pulmonary hypertension); SSB syndrome (SADDAN); steely hair syndrome (Menkes syndrome); Steinert disease (myotonic dystrophy); Steinert myotonic dystrophy syndrome (myotonic dystrophy); Stickler syndrome; stroke (CADASIL); Strudwick syndrome (spondyloepimetaphyseal dysplasia, Strudwick type); subacute neuronopathic Gaucher disease (Gaucher disease type 3); Swedish genetic porphyria (acute intermittent porphyria); Swedish porphyria (acute intermittent porphyria); Swiss cheese cartilage dysplasia (Kniest dysplasia); Tay-Sachs disease; TD - thanatophoric dwarfism (thanatophoric dysplasia); TD with straight femurs and cloverleaf skull (thanatophoric dysplasia Type 2); Telangiectasia, cerebello- oculocutaneous (ataxia-telangiectasia); Testicular feminization syndrome (androgen insensitivity syndrome); tetrahydrobiopterin deficiency; TFM - testicular feminization syndrome (androgen insensitivity syndrome); thalassemia intermedia (beta thalassemia); Thalassemia Major (beta thalassemia); thanatophoric dysplasia; thiamine-responsive megaloblastic anemia with diabetes mellitus and sensorineural deafness; Thrombophilia due to deficiency of cofactor for activated protein C, Leiden type (factor V Leiden thrombophilia); Thyroid disease; Tomaculous neuropathy (hereditary neuropathy with liability to pressure palsies); Total HPRT deficiency (Lesch-Nyhan syndrome); Total hypoxanthine-guanine phosphoribosyl transferase deficiency (Lesch-Nyhan syndrome); Tourette's Syndrome; Transmissible dementias (prion disease); Transmissible spongiform encephalopathies (prion disease); Treacher Collins syndrome; Trias fragilitis ossium (osteogenesis imperfecta Type I); triple X syndrome; Triplo X syndrome (triple X syndrome); Trisomy 21 (Down syndrome); Trisomy X (triple X syndrome); Troisier-Hanot-Chauffard syndrome (hemochromatosis); TS (Turner syndrome); TSD (Tay-Sachs disease); TSEs (prion disease); tuberose sclerosis (tuberous sclerosis); tuberous sclerosis; Turner syndrome; Turner syndrome in female with X chromosome (Noonan syndrome); Turner's phenotype, karyotype normal (Noonan syndrome); Turner's syndrome (Turner syndrome); Turner-like syndrome (Noonan syndrome); Type 2 Gaucher disease (Gaucher disease type 2); Type 3 Gaucher disease (Gaucher disease type 3); UDP-galactose-4-epimerase deficiency disease (galactosemia); UDP glucose 4-epimerase deficiency disease (galactosemia); UDP glucose hexose-l-phosphate uridylyltransferase deficiency (galactosemia); Ullrich-Noonan syndrome (Noonan syndrome); Ullrich-Turner syndrome (Turner syndrome); Undifferentiated deafness (nonsyndromic deafness); UPS deficiency (acute intermittent porphyria); Urinary bladder cancer (bladder cancer); UROD deficiency (porphyria cutanea tarda); Uroporphyrinogen decarboxylase deficiency (porphyria cutanea tarda); Uroporphyrinogen synthase deficiency (acute intermittent porphyria); UROS deficiency (congenital erythropoietic porphyria); Usher syndrome; UTP hexose-l- phosphate uridylyltransferase deficiency (galactosemia); Van Bogaert-Bertrand syndrome (Canavan disease); Van der Hoeve syndrome (osteogenesis imperfecta Type I); variegate porphyria; Velocardiofacial syndrome (22qll.2 deletion syndrome); VHL syndrome (von Hippel- Lindau disease); Vision impairment and blindness (Alstrom syndrome); Von Bogaert-Bertrand disease (Canavan disease); von Hippel-Lindau disease; Von Recklenhausen-Applebaum disease (hemochromatosis); von Recklinghausen disease (neurofibromatosis 1); VP (variegate porphyria); Vrolik disease (osteogenesis imperfecta); Waardenburg syndrome; Warburg Sjo Fledelius Syndrome (Micro syndrome); WD (Wilson disease); Weissenbacher-Zweymuller syndrome; Wilson disease; Wilson's disease (Wilson disease); Wolf-Hirschhorn syndrome; Wolff Periodic disease (Mediterranean fever, familial); WZS (Weissenbacher-Zweymuller syndrome); Xeroderma Pigmentosum; X-linked mental retardation and macroorchidism (fragile X syndrome); X-linked primary hyperuricemia (Lesch-Nyhan syndrome); X-linked severe combined
immunodeficiency; X-linked sideroblastic anemia; X-linked spinal-bulbar muscle atrophy
(Kennedy disease); X-linked uric aciduria enzyme defect (Lesch-Nyhan syndrome); X-SCID relinked severe combined immunodeficiency); XLSA (X-linked sideroblastic anemia); XSCID (X- linked severe combined immunodeficiency); XXX syndrome (triple X syndrome); XXXX syndrome (48, XXXX); XXXXX syndrome (49, XXXXX); XXY syndrome (Klinefelter syndrome); XXY trisomy (Klinefelter syndrome); XYY karyotype (47,XYY syndrome); XYY syndrome (47,XYY syndrome); and YY syndrome (47,XYY syndrome).
In a further preferred aspect, the nucleic acid sequence as defined herein or the inventive composition comprising a plurality of nucleic acid sequences as defined herein may be used for the preparation of a pharmaceutical composition, particularly for purposes as defined herein, preferably for the use in gene therapy in the treatment of diseases as defined herein.
The inventive pharmaceutical composition may furthermore be used in gene therapy particularly in the treatment of a disease or a disorder, preferably as defined herein. The present invention furthermore provides several applications and uses of the inventive
RNA containing composition, or the pharmaceutical composition, or the vaccine, or the kit or kit of parts as defined herein. In one embodiment, the composition or the pharmaceutical composition or the kit or kit of parts may be used as a medicament, namely for treatment of tumor or cancer diseases. In this context the treatment is preferably done by intratumoral application, especially by injection into tumor tissue. According to another aspect, the present invention is directed to the second medical use of the RNA containing composition or the pharmaceutical composition, or the vaccine, or the kit or kit of parts as described above, wherein these subject matters are used for preparation of a medicament particularly for intratumoral application (administration) for treatment of tumor or cancer diseases. Preferably, diseases as mentioned herein are selected from tumor or cancer diseases which preferably include e.g. Acute lymphoblastic leukemia, Acute myeloid leukemia, Adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, Anal cancer, Appendix cancer, Astrocytoma, Basal cell carcinoma, Bile duct cancer, Bladder cancer, Bone cancer,
Osteosarcoma/Malignant fibrous histiocytoma, Brainstem glioma, Brain tumor, cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic glioma, Breast cancer, Bronchial adenomas/carcinoids, Burkitt lymphoma, childhood Carcinoid tumor, gastrointestinal Carcinoid tumor, Carcinoma of unknown primary, primary Central nervous system lymphoma, childhood Cerebellar astrocytoma, childhood Cerebral astrocytoma/Malignant glioma, Cervical cancer, Childhood cancers, Chronic lymphocytic leukemia, Chronic myelogenous leukemia, Chronic myeloproliferative disorders, Colon Cancer, Cutaneous T-cell lymphoma, Desmoplastic small round cell tumor, Endometrial cancer, Ependymoma, Esophageal cancer, Ewing's sarcoma in the Ewing family of tumors, Childhood Extracranial germ cell tumor,
Extragonadal Germ cell tumor, Extrahepatic bile duct cancer, Intraocular melanoma,
Retinoblastoma, Gallbladder cancer, Gastric (Stomach) cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal stromal tumor (GIST), extracranial, extragonadal, or ovarian Germ cell tumor, Gestational trophoblastic tumor, Glioma of the brain stem, Childhood Cerebral Astrocytoma, Childhood Visual Pathway and Hypothalamic Glioma, Gastric carcinoid, Hairy cell leukemia, Head and neck cancer, Heart cancer, Hepatocellular (liver) cancer, Hodgkin lymphoma,
Hypopharyngeal cancer, childhood Hypothalamic and visual pathway glioma, Intraocular Melanoma, Islet Cell Carcinoma (Endocrine Pancreas), Kaposi sarcoma, Kidney cancer (renal cell cancer), Laryngeal Cancer, Leukemias, acute lymphoblastic Leukemia, acute myeloid Leukemia, chronic lymphocytic Leukemia, chronic myelogenous Leukemia, hairy cell Leukemia, Lip and Oral Cavity Cancer, Liposarcoma, Liver Cancer, Non-Small Cell Lung Cancer, Small Cell Lung Cancer,
Lymphomas, AIDS-related Lymphoma, Burkitt Lymphoma, cutaneous T-Cell Lymphoma, Hodgkin Lymphoma, Non-Hodgkin Lymphomas, Primary Central Nervous System Lymphoma,
Waldenstrom Macroglobulinemia, Malignant Fibrous Histiocytoma of Bone/Osteosarcoma, Childhood Medulloblastoma, Melanoma, Intraocular (Eye) Melanoma, Merkel Cell Carcinoma, Adult Malignant Mesothelioma, Childhood Mesothelioma, Metastatic Squamous Neck Cancer with Occult Primary, Mouth Cancer, Childhood Multiple Endocrine Neoplasia Syndrome, Multiple Myeloma/Plasma Cell Neoplasm, Mycosis Fungoides, Myelodysplastic Syndromes,
Myelodysplastic/Myeloproliferative Diseases, Chronic Myelogenous Leukemia, Adult Acute Myeloid Leukemia, Childhood Acute Myeloid Leukemia, Multiple Myeloma (Cancer of the Bone- Marrow), Chronic Myeloproliferative Disorders, Nasal cavity and paranasal sinus cancer, Nasopharyngeal carcinoma, Neuroblastoma, Oral Cancer, Oropharyngeal cancer,
Osteosarcoma/malignant fibrous histiocytoma of bone, Ovarian cancer, Ovarian epithelial cancer (Surface epithelial-stromal tumor), Ovarian germ cell tumor, Ovarian low malignant potential tumor, Pancreatic cancer, islet cell Pancreatic cancer, Paranasal sinus and nasal cavity cancer, Parathyroid cancer, Penile cancer, Pharyngeal cancer, Pheochromocytoma, Pineal astrocytoma, Pineal germinoma, childhood Pineoblastoma and supratentorial primitive neuroectodermal tumors, Pituitary adenoma, Plasma cell neoplasia/Multiple myeloma, Pleuropulmonary blastoma, Primary central nervous system lymphoma, Prostate cancer, Rectal cancer, Renal cell carcinoma (kidney cancer), Cancer of the Renal pelvis and ureter, Retinoblastoma, childhood
Rhabdomyosarcoma, Salivary gland cancer, Sarcoma of the Ewing family of tumors, Kaposi
Sarcoma, soft tissue Sarcoma, uterine Sarcoma, Sezary syndrome, Skin cancer (nonmelanoma), Skin cancer (melanoma), Merkel cell Skin carcinoma, Small intestine cancer, Squamous cell carcinoma, metastatic Squamous neck cancer with occult primary, childhood Supratentorial primitive neuroectodermal tumor, Testicular cancer, Throat cancer, childhood Thymoma, Thymoma and Thymic carcinoma, Thyroid cancer, childhood Thyroid cancer, Transitional cell cancer of the renal pelvis and ureter, gestational Trophoblastic tumor, Urethral cancer, endometrial Uterine cancer, Uterine sarcoma, Vaginal cancer, childhood Visual pathway and hypothalamic glioma, Vulvar cancer, Waldenstrom macroglobulinemia, and childhood Wilms tumor (kidney cancer).
Especially preferred examples of tumors or cancers that are suitable for intratumoral administration are prostate cancer, lung cancer, breast cancer, brain cancer, head and neck cancer, thyroid cancer, colon cancer, stomach cancer, liver cancer, pancreas cancer, ovary cancer, skin cancer, urinary bladder, uterus and cervix.
According to a specific embodiment, the medicament may be administered to the patient as a single dose or as several doses. In certain embodiments, the medicament may be administered to a patient as a single dose followed by a second dose later and optionally even a third, fourth (or more) dose subsequent thereto et cetera.
Preferably, the inventive composition is provided in an amount of at least 40 μg RNA per dose. More specifically, the amount of the mRNA comprised in a single dose is typically at least 200 μg, preferably from 200 μg to 1.000 μg, more preferably from 300 μg to 850 μg, even more preferably from 300 μg to 700 μg. In the case of intradermal injection, which is preferably carried out via jet injection (e.g. using a Tropis device), the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 μg, preferably from 80 μg to 700 μg, more preferably from 80 μg to 400 μg. Moreover, in the case of intramuscular injection, which is preferably carried out by using a conventional needle or via jet injection, the amount of the artificial nucleic acid comprised in a single dose is typically at least 80 μg, preferably from 80 μg to 1.000 μg, more preferably from 80 μg to 850 μg, even more preferably from 80 μg to 700 μg.
The immunization protocol for the treatment or prophylaxis of e.g. a virus infection, i.e the immunization of a subject against e.g. a virus, typically comprises a series of single doses or dosages of the inventive composition or the inventive vaccine. A single dosage, as used herein, refers to the initial/first dose, a second dose or any further doses, respectively, which are preferably administered in order to "boost" the immune reaction.
According to a preferred embodiment, the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts is provided for use in treatment or prophylaxis, preferably treatment or prophylaxis of e.g. a virus infection or a related disorder or disease, wherein the treatment or prophylaxis comprises the administration of a further active pharmaceutical ingredient. More preferably, in the case of the inventive vaccine or composition, which is based on the inventive artificial nucleic acid, a polypeptide may be coadministered as a further active pharmaceutical ingredient. For example, at least one e.g. virus protein or peptide as described herein, or a fragment or variant thereof, may be co-administered in order to induce or enhance an immune response. Likewise, in the case of the inventive vaccine or composition, which is based on the inventive polypeptide as described herein, an artificial nucleic acid as described herein may be co-administered as a further active pharmaceutical ingredient. For example, an artificial nucleic acid as described herein encoding at least one polypeptide as described herein may be co-administered in order to induce or enhance an immune response. A further component of the inventive vaccine or composition may be an
immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably
immunoglobulins directed against e.g. a virus. Preferably, such a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA. Such an immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the inventive artificial nucleic acid or by the inventive polypeptide.
In a further aspect the invention provides a method of treating or preventing a disorder, wherein the disorder is preferably an infection with e.g. a virus or a disorder related to an infection with e.g. a virus, wherein the method comprises administering to a subject in need thereof the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts. In particular, such a method may preferably comprise the steps of: a) providing the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts; b) applying or administering the artificial nucleic acid as disclosed herein, the inventive composition comprising at least one artificial nucleic acid as disclosed herein, the inventive polypeptides as described herein, the inventive composition comprising at least one inventive polypeptide, the inventive vaccine or the inventive kit or kit of parts to a tissue or an organism; c) optionally administering immunoglobuline (IgGs) against e.g. the virus.
According to a further aspect, the present invention also provides a method for expression of at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, wherein the method preferably comprises the following steps: a) providing the inventive artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprising e.g. at least one virus, or a fragment or variant thereof, preferably as defined herein, or a composition comprising said artificial nucleic acid; and b) applying or administering the inventive artificial nucleic acid or the inventive composition comprising said artificial nucleic acid to an expression system, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism.
The method may be applied for laboratory, for research, for diagnostic, for commercial production of peptides or proteins and/or for therapeutic purposes. In this context, typically after preparing the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, e.g. in naked or complexed form or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein. The method may be carried out in vitro, in vivo or ex vivo. The method may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of infectious diseases, or a related disorder.
In this context, in vitro is defined herein as transfection or transduction of the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein into cells in culture outside of an organism; in vivo is defined herein as transfection or transduction of the inventive artificial nucleic acid or of the inventive composition or vaccine into cells by application of the inventive mRNA or of the inventive composition to the whole organism or individual and ex vivo is defined herein as transfection or transduction of the inventive artificial nucleic acid or of the inventive composition or vaccine into cells outside of an organism or individual and subsequent application of the transfected cells to the organism or individual. Likewise, according to another aspect, the present invention also provides the use of the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, preferably for diagnostic or therapeutic purposes, for expression of e.g. an encoded virus antigenic peptide or protein, e.g. by applying or administering the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, e.g. to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism. The use may be applied for a (diagnostic) laboratory, for research, for diagnostics, for commercial production of peptides or proteins and/or for therapeutic purposes. In this context, typically after preparing the inventive artificial nucleic acid as defined herein or of the inventive composition or vaccine as defined herein, it is typically applied or administered to a cell-free expression system, a cell (e.g. an expression host cell or a somatic cell), a tissue or an organism, preferably in naked form or complexed form, or as a (pharmaceutical) composition or vaccine as described herein, preferably via transfection or by using any of the administration modes as described herein. The use may be carried out in vitro, in vivo or ex vivo. The use may furthermore be carried out in the context of the treatment of a specific disease, particularly in the treatment of e.g. a virus infection or a related disorder.
In a particularly preferred embodiment, the invention provides the artificial nucleic acid, the inventive composition or the inventive vaccine for use as defined herein, preferably for use as a medicament, for use in treatment or prophylaxis, preferably treatment or prophylaxis of a e.g. a virus infection or a related disorder, or for use as a vaccine.
In another embodiment, the nucleotide acid molecule of the inventive composition, preferably the mRNA molecule, encodes at least one epitope of at least one antigen. In preferred embodiments of the invention the at least one antigen is selected from the group consisting of an antigen from a pathogen associated with infectious diseases, an antigen associated with allergies, an antigen associated with autoimmune diseases, and an antigen associated with cancer or tumor diseases, or a fragment, variant and/or derivative of said antigen.
Preferably the at least one antigen is derived from a pathogen, preferably a viral, bacterial, fungal or protozoan pathogen, preferably selected from the list consisting of: Rabies virus, Ebolavirus, Marburgvirus, Hepatitis B virus, human Papilloma virus (hPV), Bacillus anthracis, Respiratory syncytial virus (RSV), Herpes simplex virus (HSV), Dengue virus, Rotavirus, Influenza virus, human immunodeficiency virus (HIV), Yellow Fever virus, Mycobacterium tuberculosis, Plasmodium, Staphylococcus aureus, Chlamydia trachomatis, Cytomegalovirus (CMV) and
Hepatitis B virus (HBV). In this context the mRNA of the inventive composition may encode for a protein or a peptide, which comprises at least one epitope of a pathogenic antigen or a fragment, variant or derivative thereof. Such pathogenic antigens are derived from pathogenic organisms, in particular bacterial, viral or protozoological (multicellular) pathogenic organisms, which evoke an immunological reaction by subject, in particular a mammalian subject, more particularly a human. More specifically, pathogenic antigens are preferably surface antigens, e.g. proteins (or fragments of proteins, e.g. the exterior portion of a surface antigen) located at the surface of the virus or the bacterial or protozoological organism.
Pathogenic antigens are peptide or protein antigens preferably derived from a pathogen associated with infectious disease which are preferably selected from antigens derived from the pathogens Acinetobacter baumannii, Anaplasma genus, Anaplasma phagocytophilum, Ancylostoma braziliense, Ancylostoma duodenale, Arcanobacte um haemolyticum, Asca s lumb coides, Aspergillus genus, Astroviridae, Babesia genus, Bacillus anthracis, Bacillus cereus, Bartonella henselae, BK virus, Blastocystis hominis, Blastomyces dermatitidis, Bordetella pertussis, Borrelia burgdorferi, Borrelia genus, Borrelia spp, Brucella genus, Brugia malayi, Bunyaviridae family, Burkholderia cepacia and other Burkholderia species, Burkholderia mallei, Burkholderia pseudomallei, Caliciviridae family, Campylobacter genus, Candida albicans, Candida spp, Chlamydia trachomatis, Chlamydophila pneumoniae, Chlamydophila psittaci, CJD prion, Clonorchis sinensis, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium perfringens, Clostridium spp, Clostridium tetani, Coccidioides spp, coronaviruses, Corynebacterium diphtheriae, Coxiella burnetii, Crimean-Congo hemorrhagic fever virus, Cryptococcus neoformans,
Cryptosporidium genus, Cytomegalovirus (CMV), Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN-4), Dientamoeba fragilis, Ebolavirus (EBOVJ, Echinococcus genus, Ehrlichia chaffeensis, Ehrlichia ewingii, Ehrlichia genus, Entamoeba histolytica, Enterococcus genus, Enterovirus genus, Enteroviruses, mainly Coxsackie A virus and Enterovirus 71 (EV71), Epidermophyton spp,
Epstein-Barr Virus (EBV), Escherichia coli 0157:H7, Olll and O104:H4, Fasciola hepatica and Fasciola gigantica, FFI prion, Filarioidea superfamily, Flaviviruses, Francisella tularensis,
Fusobacterium genus, Geotrichum candidum, Giardia intestinalis, Gnathostoma spp, GSS prion, Guanarito virus, Haemophilus ducreyi, Haemophilus influenzae, Helicobacter pylori, Henipavirus (Hendra virus Nipah virus), Hepatitis A Virus, Hepatitis B Virus (HBV), Hepatitis C Virus (HCV),
Hepatitis D Virus, Hepatitis E Virus, Herpes simplex virus 1 and 2 (HSV-1 and HSV-2), Histoplasma capsulatum, HIV (Human immunodeficiency virus), Hortaea werneckii, Human bocavirus (HBoV), Human herpesvirus 6 (HHV-6) and Human herpesvirus 7 (HHV-7), Human metapneumovirus (hMPV), Human papillomavirus (HPV), Human parainfluenza viruses (HPIV), Japanese
encephalitis virus, JC virus, Junin virus, Kingella kingae, Klebsiella granulomatis, Kuru prion, Lassa virus, Legionella pneumophila, Leishmania genus, Leptospira genus, Listeria monocytogenes, Lymphocytic choriomeningitis virus (LCMV), Machupo virus, Malassezia spp, Marburg virus, Measles virus, Metagonimus yokagawai, Microspo dia phylum, Molluscum contagiosum virus (MCV), Mumps virus, Mycobacterium leprae and Mycobacterium lepromatosis, Mycobacterium tuberculosis, Mycobacterium ulcerans, Mycoplasma pneumoniae, Naegleria fowleri, Necator americanus, Neisseria gonorrhoeae, Neisseria meningitidis, Nocardia asteroides, Nocardia spp, Onchocerca volvulus, Orientia tsutsugamushi, Orthomyxoviridae family (Influenza),
Paracoccidioides brasiliensis, Paragonimus spp, Paragonimus westermani, Parvovirus B19, Pasteurella genus, Plasmodium genus, Pneumocystis jirovecii, Poliovirus, Rabies virus, Respiratory syncytial virus (RSV), Rhinovirus, rhinoviruses, Rickettsia akari, Rickettsia genus, Rickettsia prowazekii, Rickettsia rickettsii, Rickettsia typhi, Rift Valley fever virus, Rotavirus, Rubella virus, Sabia virus, Salmonella genus, Sarcoptes scabiei, SARS coronavirus, Schistosoma genus, Shigella genus, Sin Nombre virus, Hantavirus, Sporothrix schenckii, Staphylococcus genus, Staphylococcus genus, Streptococcus agalactiae, Streptococcus pneumoniae, Streptococcus pyogenes, Strongyloides stercoralis, Taenia genus, Taenia solium, Tick-borne encephalitis virus (TBEV), Toxocara canis or Toxocara cati, Toxoplasma gondii, Treponema pallidum, Trichinella spiralis, Trichomonas vaginalis, Trichophyton spp, Trichuris trichiura, Trypanosoma brucei, Trypanosoma cruzi, Ureaplasma urealyticum, Varicella zoster virus (VZV), Varicella zoster virus (VZV), Variola major or Variola minor, vCJD prion, Venezuelan equine encephalitis virus, Vibrio cholerae, West Nile virus, Western equine encephalitis virus, Wuchereria bancrofti, Yellow fever virus, Yersinia enterocolitica, Yersinia pestis, and Yersinia pseudotuberculosis.
Furthermore, the pathogenic antigen (antigen derived from a pathogen associated with infectious disease) may be preferably selected from the following antigens: Outer membrane protein A OmpA, biofilm associated protein Bap, transport protein MucK (Acinetobacter baumannii, Acinetobacter infections)); variable surface glycoprotein VSG, microtubule-associated protein MAPP15, trans-sialidase TSA (Trypanosoma brucei, African sleeping sickness (African trypanosomiasis)); HIV p24 antigen, HIV envelope proteins (Gpl20, Gp41, Gpl60), polyprotein GAG, negative factor protein Nef, trans-activator of transcription Tat (HIV (Human
immunodeficiency virus), AIDS (Acquired immunodeficiency syndrome)); galactose-inhibitable adherence protein GIAP, 29 kDa antigen Eh29, Gal/GalNAc lectin, protein CRT, 125 kDa immunodominant antigen, protein M17, adhesin ADH112, protein STIRP (Entamoeba histolytica, Amoebiasis); Major surface proteins 1-5 (MSPla, MSP■, MSP2, MSP3, MSP4, MSP5), type IV secreotion system proteins (VirB2, VirB7, VirBll, VirD4) (Anaplasma genus, Anaplasmosis); protective Antigen PA, edema factor EF, lethal facotor LF, the S-layer homology proteins SLH (Bacillus anthracis, Anthrax); acranolysin, phospholipase D, collagen-binding protein CbpA (Arcanobacterium haemolyticum, Arcanobacterium haemolyticum infection); nucleocapsid protein NP, glycoprotein precursor GPC, glycoprotein GP1, glycoprotein GP2 (Junin virus, Argentine hemorrhagic fever); chitin-protein layer proteins, 14 kDa suarface antigen A14, major sperm protein MSP, MSP polymerization-organizing protein MPOP, MSP fiber protein 2 MFP2, MSP polymerization-activating kinase MPAK, ABA-l-like protein ALB, protein ABA-1, cuticulin CUT-1 (Ascaris lumbricoides, Ascariasis); 41 kDa allergen Asp vl3, allergen Asp f3, major conidial surface protein rodlet A, protease Peplp, GPI-anchored protein Gellp, GPI-anchored protein Crflp (Aspergillus genus, Aspergillosis); family VP26 protein, VP29 protein (Astroviridae, Astrovirus infection); Rhoptry-associated protein 1 RAP-1, merozoite surface antigens MSA-1, MSA-2 (al, a2, b, c), 12D3, 11C5, 21B4, P29, variant erythrocyte surface antigen VESA1, Apical Membrane Antigen 1 AMA-1 (Babesia genus, Babesiosis); hemolysin, enterotoxin C, PXOl-51, glycolate oxidase, ABC-transporter, penicillin-bingdn protein, zinc transporter family protein, pseudouridine synthase Rsu, plasmid replication protein RepX, oligoendopeptidase F, prophage membrane protein, protein HemK, flagellar antigen H, 28.5-kDa cell surface antigen (Bacillus cereus, Bacillus cereus infection); large T antigen LT, small T antigen, capsid protein VP1, capsid protein VP2 (BK virus, BK virus infection); 29 kDa-protein, caspase-3-like antigens, glycoproteins (Blastocystis hominis, Blastocystis hominis infection); yeast surface adhesin WI-1 (Blastomyces dermatitidis, Blastomycosis); nucleoprotein N, polymerase L, matrix protein Z, glycoprotein GP (Machupo virus, Bolivian hemorrhagic fever); outer surface protein A OspA, outer surface protein OspB, outer surface protein OspC, decorin binding protein A DbpA, decorin binding protein B DbpB, flagellar filament 41 kDa core protein Fla, basic membrane protein A precursor BmpA (Immunodominant antigen P39), outer surface 22 kDa lipoprotein precursor (antigen IPLA7), variable surface lipoprotein vlsE (Borrelia genus, Borrelia infection); Botulinum neurotoxins BoNT/Al, BoNT/A2, BoNT/A3, BoNT/B, BoNT/C, BoNT/D, BoNT/E, BoNT/F, BoNT/G, recombinant botulinum toxin F He domain FHc (Clostridium botulinum, Botulism (and Infant botulism)); nucleocapsid, glycoprotein precursor (Sabia virus, Brazilian hemorrhagic fever); copper/Zinc superoxide dismutase SodC, bacterioferritin Bfr, 50S ribosomal protein RplL, OmpA- like transmembrane domain-containing protein Omp31, immunogenic 39-kDa protein M5 P39, zinc ABC transporter periplasmic zinc-bnding protein znuA, periplasmic immunogenic protein Bp26, 30S ribosomal protein S12 RpsL, glyceraldehyde-3-phosphate dehydrogenase Gap, 25 kDa outer-membrane immunogenic protein precursor Omp25, invasion protein B lalB, trigger factor Tig, molecular chaperone DnaK, putative peptidyl-prolyl cis-trans isomerase SurA, lipoprotein Ompl9, outer membrane protein MotY Ompl6, conserved outer membrane protein D15, malate dehydrogenase Mdh, component of the Type-IV secretion system (T4SS) VirJ, lipoprotein of unknown function BAB1_0187 (Brucella genus, Brucellosis); members of the ABC transporter family (LolC, OppA, and PotF), putative lipoprotein releasing system transmembrane protein LolC /E, flagellin FliC, Burkholderia intracellular motility A BimA, bacterial Elongation factor-Tu EF-Tu, 17 kDa OmpA-like protein, boaA coding protein, boaB coding protein (Burkholderia cepacia and other Burkholderia species, Burkholderia infection); mycolyl-transferase Ag85A, heat-shock protein Hsp65, protein TB10.4, 19 kDa antigen, protein PstS3, heat-shock protein Hsp70 (Mycobacterium ulcerans, Buruli ulcer); norovirus major and minor viral capsid proteins VP1 and VP2, genome polyprotein, Sapoviurus capsid protein VP1, protein Vp3, geome polyprotein (Caliciviridae family, Calicivirus infection (Norovirus and Sapovirus)); major outer membrane protein PorA, flagellin FlaA, surface antigen CjaA, fibronectin binding protein CadF, aspartate/glutamate-binding ABC transporter protein PeblA, protein FspAl, protein FspA2 (Campylobacter genus, Campylobacteriosis); glycolytic enzyme enolase, secreted aspartyl proteinases SAPl-10, glycophosphatidylinositol (GPI)-linked cell wall protein, protein Hyrl, complement receptor 3-related protein CR3-RP, adhesin Als3p, heat shock protein 90 kDa hsp90, cell surface hydrophobicity protein CSH (usually Candida albicans and other Candida species, Candidiasis); 17-kDa antigen, protein P26, trimeric autotransporter adhesins TAAs, Bartonella adhesin A BadA, variably expressed outer-membrane proteins Vomps, protein Pap3, protein HbpA, envelope-associated protease HtrA, protein OMP89, protein GroEL, protein LalB, protein OMP43, dihydrolipoamide succinyltransferase SucB (Bartonella henselae, Cat-scratch disease); amastigote surface protein-2, amastigote-specific surface protein SSP4, cruzipain, trans-sialidase TS, trypomastigote surface glycoprotein TSA-1, complement regulatory protein CRP-10, protein G4, protein G2, paraxonemal rod protein PAR2, paraflagellar rod component Pari, mucin- Associated Surface Proteins MPSP (Trypanosoma cruzi, Chagas Disease (American
trypanosomiasis)); envelope glycoproteins (gB, gC, gE, gH, gl, gK, gL) (Varicella zoster virus (VZV), Chickenpox); major outer membrane protein MOMP, probable outer membrane protein PMPC, outer membrane complex protein B OmcB, heat shock proteins Hsp60 HSP10, protein IncA, proteins from the type III secretion system, ribonucleotide reductase small chain protein NrdB, plasmid protein Pgp3, chlamydial outer protein N CopN, antigen CT521, antigen CT425, antigen CT043, antigen TC0052, antigen TC0189, antigen TC0582, antigen TC0660, antigen TC0726, antigen TC0816, antigen TC0828 [Chlamydia trachomatis, Chlamydia); low calcium response protein E LCrE, chlamydial outer protein N CopN, serine/threonine-protein kinase PknD, acyl- carrier-protein S-malonyltransferase FabD, single-stranded DNA-binding protein Ssb, major outer membrane protein MOMP, outer membrane protein 2 Omp2, polymorphic membrane protein family (Pmpl, Pmp2, Pmp3, Pmp4, Pmp5, Pmp6, Pmp7, Pmp8, Pmp9, PmplO, Pmpll, Pmpl2, Pmpl3, Pmpl4, Pmpl5, Pmpl6, Pmpl7, Pmpl8, Pmpl9, Pmp20, Pmp21) (Chlamydophila pneumoniae, Chlamydophila pneumoniae infection); cholera toxin B CTB, toxin coregulated pilin A TcpA, toxin coregulated pilin TcpF, toxin co-regulated pilus biosynthesis ptrotein F TcpF, cholera enterotoxin subunit A, cholera enterotoxin subunit B, Heat-stable enterotoxin ST, mannose-sensitive hemagglutinin MSHA, outer membrane protein U Porin ompU, Poring B protein, polymorphic membrane protein-D (Vibrio cholerae, Cholera); propionyl-CoA carboxylase PCC, 14-3-3 protein, prohibitin, cysteine proteases, glutathione transferases, gelsolin, cathepsin L proteinase CatL, Tegumental Protein 20.8 kDa TP20.8, tegumental protein 31.8 kDa TP31.8, lysophosphatidic acid phosphatase LPAP, (Clonorchis sinensis, Clonorchiasis); surface layer proteins SLPs, glutamate dehydrogenase antigen GDH, toxin A, toxin B, cysteine protease Cwp84, cysteine protease Cwpl3, cysteine protease Cwpl9, Cell Wall Protein CwpV, flagellar protein FliC, flagellar protein FliD (Clostridium difficile, Clostridium difficile infection); rhinoviruses: capsid proteins VP1, VP2, VP3, VP4; coronaviruses: sprike proteins S, envelope proteins E, membrane proteins M, nucleocapsid proteins N (usually rhinoviruses and coronaviruses, Common cold (Acute viral rhinopharyngitis; Acute coryza)); prion protein Prp (CJD prion, Creutzfeldt-Jakob disease (CJD)); envelope protein Gc, envelope protein Gn, nucleocapsid proteins (Crimean-Congo hemorrhagic fever virus, Crimean-Congo hemorrhagic fever (CCHF)); virulence-associated DEAD- box RNA helicase VAD1, galactoxylomannan-protein GalXM, glucuronoxylomannan GXM, mannoprotein MP (Cryptococcus neoformans, Cryptococcosis); acidic ribosomal protein P2 CpP2, mucin antigens Mucl, Muc2, Muc3 Muc4, Muc5, Muc6, Muc7, surface adherence protein CP20, surface adherence protein CP23, surface protein CP12, surface protein CP21, surface protein
CP40, surface protein CP60, surface protein CP15, surface-associated glycopeptides gp40, surface- associated glycopeptides gpl5, oocyst wall protein AB, profilin PRF, apyrase (Cryptosporidium genus, Cryptosporidiosis); fatty acid and retinol binding protein-1 FAR-1, tissue inhibitor of metalloproteinase TIMP (TMP), cysteine proteinase ACEY-1, cysteine proteinase ACCP-1, surface antigen Ac-16, secreted protein 2 ASP-2, metalloprotease 1 MTP-1, aspartyl protease inhibitor API-1, surface-associated antigen SAA-1, adult-specific secreted factor Xa serine protease inhibitor anticoagulant AP, cathepsin D-like aspartic protease ARR-1 (usually Ancylostoma braziliense; multiple other parasites, Cutaneous larva migrans (CLM)); cathepsin L-like proteases, 53/25-kDa antigen, 8kDa family members, cysticercus protein with a marginal trypsin-like activity TsAg5, oncosphere protein TSOL18, oncosphere protein TSOL45-1A, lactate
dehydrogenase A LDHA, lactate dehydrogenase B LDHB (Taenia solium, Cysticercosis); pp65 antigen, membrane protein ppl5, capsid-proximal tegument protein ppl50, protein M45, DNA polymerase UL54, helicase UL105, glycoprotein gM, glycoprotein gN, glcoprotein H, glycoprotein B gB, protein UL83, protein UL94, protein UL99 (Cytomegalovirus (CMV), Cytomegalovirus infection); capsid protein C, premembrane protein prM, membrane protein M, envelope protein E (domain I, domain II, domain II), protein NS1, protein NS2A, protein NS2B, protein NS3, protein NS4A, protein 2K, protein NS4B, protein NS5 (Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN- 4)-Flaviviruses, Dengue fever); 39 kDa protein (Dientamoeba fragilis, Dientamoebiasis);
diphtheria toxin precursor Tox, diphteria toxin DT, pilin-specific sortase SrtA, shaft pilin protein SpaA, tip pilin protein SpaC, minor pilin protein SpaB, surface-associated protein DIP1281
(Corynebacterium diphtheriae, Diphtheria); glycoprotein GP, nucleoprotein NP, minor matrix protein VP24, major matrix protein VP40, transcription activator VP30, polymerase cofactor VP35, RNA polymerase L (Ebolavirus (EBOV), Ebola hemorrhagic fever); prion protein (vCJD prion, Variant Creutzfeldt-Jakob disease (vCJD, nvCJD)); UvrABC system protein B, protein Flpl, protein Flp2, protein Flp3, protein TadA, hemoglobin receptor HgbA, outer membrane protein TdhA, protein CpsRA, regulator CpxR, protein Sap A, 18 kDa antigen, outer membrane protein NcaA, protein LspA, protein LspAl, protein LspA2, protein LspB, outer membrane component DsrA, lectin DltA, lipoprotein Hip, major outer membrane protein OMP, outer membrane protein OmpA2 (Haemophilus ducreyi, Chancroid); aspartyl protease 1 Pepl, phospholipase B PLB, alpha- mannosidase 1 AMN1, glucanosyltransferase GEL1, urease URE, peroxisomal matrix protein Pmpl, proline-rich antigen Pra, humal T-cell reative protein TcrP (Coccidioides immitis and
Coccidioides posadasii, Coccidioidomycosis); allergen Tri r 2, heat shock protein 60 Hsp60, fungal actin Act, antigen Tri r2, antigen Tri r4, antigen Tri tl, protein IV, glycerol-3-phosphate dehydrogenase Gpdl, osmosensor HwSholA, osmosensor HwSholB, histidine kinase HwHhk7B, allergen Mala s 1, allergen Mala s 11, thioredoxin Trx Mala s 13, allergen Mala f, allergen Mala s (usually Trichophyton spp, Epidermophyton spp., Malassezia spp., Hortaea werneckii,
Dermatophytosis); protein EG95, protein EG10, protein EG18, protein EgA31, protein EM18, antigen EPC1, antigen B, antigen 5, protein P29, protein 14-3-3, 8-kDa protein, myophilin, heat shock protein 20 HSP20, glycoprotein GP-89, fatty acid binding protein FAPB (Echinococcus genus, Echinococcosis); major surface protein 2 MSP2, major surface protein 4 MSP4, MSP variant SGV1, MSP variant SGV2, outer membrane protein OMP, outer membrande protein 19 OMP-19, major antigenic protein MAPI, major antigenic protein MAP1-2, major antigenic protein MAP1B, major antigenic protein MAP1-3, Erum2510 coding protein, protein GroEL, protein GroES, 30- kDA major outer membrane proteins, GE 100-kDa protein, GE 130-kDa protein, GE 160-kDa protein (Ehrlichia genus, Ehrlichiosis); secreted antigen SagA, sagA-like proteins SalA and SalB, collagen adhesin Scm, surface proteins Fmsl (EbpA(fm), Fms5 (EbpB(fm), Fms9 (EpbC(fm) and FmslO, protein EbpC(fm), 96 kDa immunoprotective glycoprotein Gl (Enterococcus genus, Enterococcus infection); genome polyprotein, polymerase 3D, viral capsid protein VP1, viral capsid protein VP2, viral capsid protein VP3, viral capsid protein VP4, protease 2A, protease 3C (Enterovirus genus, Enterovirus infection); outer membrane proteins OM, 60 kDa outer membrane protein, cell surface antigen OmpA, cell surface antigen OmpB (sca5), 134 kDa outer membrane protein, 31 kDa outer membrane protein, 29.5 kDa outer membrane protein, cell surface protein SCA4, cell surface protein Adrl (RP827), cell surface protein Adr2 (RP828), cell surface protein SCA1, Invasion protein invA, cell division protein fts, secretion proteins sec Ofamily, virulence proteins virB, tlyA, tlyC, parvulin-like protein Pip, preprotein translocase SecA, 120-kDa surface protein antigen SPA, 138 kD complex antigen, major 100-kD protein (protein I), intracytoplasmic protein D, protective surface protein antigen SPA (Rickettsia prowazekii, Epidemic typhus); Epstein-Barr nuclear antigens (EBNA-1, EBNA-2, EBNA-3A, EBNA-3B, EBNA- 3C, EBNA-leader protein (EBNA-LP)), latent membrane proteins (LMP-1, LMP-2A, LMP-2B), early antigen EBV-EA, membrane antigen EBV-MA, viral capsid antigen EBV-VCA, alkaline nuclease EBV-AN, glycoprotein H, glycoprotein gp350, glycoprotein gpllO, glycoprotein gp42, glycoprotein gHgL, glycoprotein gB (Epstein-Barr Virus (EBV), Epstein-Barr Virus Infectious Mononucleosis); cpasid protein VP2, capsid protein VP1, major protein NS1 (Parvovirus B19, Erythema
infectiosum (Fifth disease)); pp65 antigen, glycoprotein 105, major capsid protein, envelope glycoprotein H, protein U51 (Human herpesvirus 6 (HHV-6) and Human herpesvirus 7 (HHV-7), Exanthem subitum); thioredoxin-glutathione reductase TGR, cathepsins LI and L2, Kunitz-type protein KTM, leucine aminopeptidase LAP, cysteine proteinase Fas2, saposin-like protein-2 SAP- 2, thioredoxin peroxidases TPx, Prx-1, Prx-2, cathepsin 1 cysteine proteinase CL3, protease cathepsin L CL1, phosphoglycerate kinase PGK, 27-kDa secretory protein, 60 kDa protein
HSP35alpha, glutathione transferase GST, 28.5 kDa tegumental antigen 28.5 kDa TA, cathepsin B3 protease CatB3, Type I cystatin stefin-1, cathepsin L5, cathepsin Llg and cathepsin B, fatty acid binding protein FABP, leucine aminopeptidases LAP (Fasciola hepatica and Fasciola gigantica,
Fasciolosis); prion protein (FFI prion, Fatal familial insomnia (FFI)); venom allergen homolog-like protein VAL-1, abundant larval transcript ALT-1, abundant larval transcript ALT-2, thioredoxin peroxidase TPX, vespid allergen homologue VAH, thiordoxin peroxidase 2 TPX-2, antigenic protein SXP (peptides N, Nl, N2, and N3), activation associated protein- 1 ASP-1, Thioredoxin TRX, transglutaminase BmTGA, glutathione-S-transferases GST, myosin, vespid allergen homologue VAH, 175 kDa collagenase, glyceraldehyde-3-phosphate dehydrogenase GAPDH, cuticular collagen Col-4, secreted larval acidic proteins SLAPs, chitinase CHI-1, maltose binding protein MBP, glycolytic enzyme fructose-l,6-bisphosphate aldolase Fba, tropomyosin TMY-1, nematode specific gene product OvB20, onchocystatin CPI-2, Cox-2 (Filarioidea superfamily, Filariasis); phospholipase C PLC, heat-labile enterotoxin B, Iota toxin component lb, protein CPE1281 , pyruvate ferredoxin oxidoreductase, elongation factor G EF-G, perfringolysin O Pfo,
glyceraldehyde-3-phosphate dehydrogenase GapC, Fructose-bisphosphate aldolase Alf2,
Clostridium perfringens enterotoxin CPE, alpha toxin AT, alpha toxoid ATd, epsilon-toxoid ETd, protein HP, large cytotoxin TpeL, endo-beta-N-acetylglucosaminidase Naglu,
phosphoglyceromutase Pgm (Clostridium perfringens, Food poisoning by Clostridium
perfringens); leukotoxin lktA, adhesion FadA, outer membrane protein RadD, high-molecular weight arginine-binding protein (Fusobacterium genus, Fusobacterium infection); phospholipase C PLC, heat-labile enterotoxin B, Iota toxin component lb, protein CPE1281, pyruvate ferredoxin oxidoreductase, elongation factor G EF-G, perfringolysin O Pfo, glyceraldehyde-3-phosphate dehydrogenase GapC, fructose-bisphosphate aldolase Alf2, Clostridium perfringens enterotoxin
CPE, alpha toxin AT, alpha toxoid ATd, epsilon-toxoid ETd, protein HP, large cytotoxin TpeL, endo- beta-N-acetylglucosaminidase Naglu, phosphoglyceromutase Pgm (usually Clostridium
perfringens; other Clostridium species, Gas gangrene (Clostridial myonecrosis)); lipase A, lipase B, peroxidase Decl (Geotrichum candidum, Geotrichosis); prion protein (GSS prion, Gerstmann- Straussler-Scheinker syndrome (GSS)); cyst wall proteins CWP1, CWP2, CWP3, variant surface protein VSP, VSP1, VSP2, VSP3, VSP4, VSP5, VSP6, 56 kDa antigen, pyruvate ferredoxin oxidoreductase PFOR, alcohol dehydrogenase E ADHE, alpha-giardin, alpha8-giardin, alphal- guiardin, beta-giardin, cystein proteases, glutathione-S-transferase GST, arginine deiminase ADI, fructose- 1,6-bisphosphat aldolase FBA, Giardia trophozoite antigens GTA (GTAl, GTA2), ornithine carboxyl transferase OCT, striated fiber-asseblin-like protein SALP, uridine phosphoryl-like protein UPL, alpha-tubulin, beta-tubulin (Giardia intestinalis, Giardiasis); members of the ABC transporter family (LolC, OppA, and PotF), putative lipoprotein releasing system transmembrane protein LolC/E, flagellin FliC, Burkholderia intracellular motility A BimA, bacterial Elongation factor-Tu EF-Tu, 17 kDa OmpA-like protein, boaA coding protein (Burkholderia mallei, Glanders); cyclophilin CyP, 24 kDa third-stage larvae protien GS24, excretion-secretion products ESPs (40, 80, 120 and 208 kDa) (Gnathostoma spinigerum and Gnathostoma hispidum, Gnathostomiasis); pilin proteins, minor pilin-associated subunit pilC, major pilin subunit and variants pilE, pilS, phase variation protein porA, Porin B PorB, protein TraD, Neisserial outer membrane antigen H.8, 70kDa antigen, major outer membrane protein PI, outer membrane proteins P1A and P1B, W antigen, surface protein A NspA, transferrin binding protein TbpA, transferrin binding protein TbpB , PBP2, mtrR coding protein, ponA coding protein, membrane permease FbpBC, FbpABC protein system, LbpAB proteins, outer membrane protein Opa, outer membrane transporter FetA, iron-repressed regulator MpeR (Neisseria gonorrhoeae, Gonorrhea); outer membrane protein A OmpA, outer membrane protein C OmpC, outer membrane protein K17 OmpK17 (Klebsiella granulomatis, Granuloma inguinale (Donovanosis)); fibronectin-binding protein Sfb,
fibronectin/fibrinogen-binding protein FBP54, fibronectin-binding protein FbaA, M protein type 1 Emml, M protein type 6 Emm6, immunoglobulin-binding protein 35 Sib35, Surface protein R28 Spr28, superoxide dismutase SOD, C5a peptidase ScpA, antigen I/I I Agl/II, adhesin AspA, G- related alpha2-macroglobulin-binding protein GRAB, surface fibrillar protein M5 (Streptococcus pyogenes, Group A streptococcal infection); C protein β antigen, arginine deiminase proteins, adhesin BibA, 105 kDA protein BPS, surface antigens c, surface antigens R, surface antigens X, trypsin-resistant protein Rl, trypsin-resistant protein R3, trypsin-resistant protein R4, surface immunogenic protein Sip, surface protein Rib, Leucine-rich repeats protein LrrG, serine-rich repeat protein Srr-2, C protein alpha-antigen Bca, Beta antigen Bag, surface antigen Epsilon, alpha-like protein ALP1, alpha-like protein ALP5 surface antigen delta, alpha-like protein ALP2, alpha-like protein ALP3, alpha-like protein ALP4, Cbeta protein Bac (Streptococcus agalactiae, Group B streptococcal infection); transferrin-binding protein 2 Tbp2, phosphatase P4, outer membrane protein P6, peptidoglycan-associated lipoprotein Pal, protein D, protein E, adherence and penetration protein Hap, outer membrane protein 26 Omp26, outer membrane protein P5 (Fimbrin), outer membrane protein D15, outer membrane protein OmpP2, 5'-nucleotidase NucA, outer membrane protein PI, outer membrane protein P2, outer membrane lipoprotein Pep, Lipoprotein E, outer membrane protein P4, fuculokinase FucK, [Cu,Zn] -superoxide dismutase SodC, protease HtrA, protein 0145, alpha-galactosylceramide (Haemophilus influenzae,
Haemophilus influenzae infection); polymerase 3D, viral capsid protein VP1, viral capsid protein VP2, viral capsid protein VP3, viral capsid protein VP4, protease 2A, protease 3C (Enteroviruses, mainly Coxsackie A virus and Enterovirus 71 (EV71), Hand, foot and mouth disease (HFMD)); RNA polymerase L, protein L, glycoprotein Gn, glycoprotein Gc, nucleocapsid protein S, envelope glycoprotein Gl, nucleoprotein NP, protein N, polyprotein M (Sin Nombre virus, Hantavirus, Hantavirus Pulmonary Syndrome (HPS)); heat shock protein HspA, heat shock protein HspB, citrate synthase GltA, protein UreB, heat shock protein Hsp60, neutrophil-activating protein NAP, catalase KatA, vacuolating cytotoxin VacA, urease alpha UreA, urease beta Ureb, protein CpnlO, protein groES, heat shock protein HsplO, protein MopB, cytotoxicity-associated 10 kDa protein CAG, 36 kDa antigen, beta-lactamase HcpA, Beta-lactamase HcpB (Helicobacter pylori,
Helicobacter pylori infection); integral membrane proteins, aggregation-prone proteins, O- antigen, toxin-antigens Stx2B, toxin-antigen StxlB, adhesion-antigen fragment Int28, protein EspA, protein EspB, Intimin, protein Tir, protein IntC300, protein Eae (Escherichia coli 0157:H7, Olll and O104:H4, Hemolytic-uremic syndrome (HUS)); RNA polymerase L, protein L, glycoprotein Gn, glycoprotein Gc, nucleocapsid protein S, envelope glycoprotein Gl,
nucleoprotein NP, protein N, polyprotein M (Bunyaviridae family, Hemorrhagic fever with renal syndrome (HFRS)); glycoprotein G, matrix protein M, nucleoprotein N, fusion protein F, polymerase L, protein W, proteinC, phosphoprotein p, non-structural protein V (Henipavirus (Hendra virus Nipah virus), Henipavirus infections); polyprotein, glycoproten Gp2, hepatitis A surface antigen HBAg, protein 2A, virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4, protein PIB, protein P2A, protein P3AB, protein P3D (Hepatitis A Virus, Hepatitis A); hepatitis B surface antigen HBsAg, Hepatitis B core antigen HbcAg, polymerase, protein Hbx, preS2 middle surface protein, surface protein L, large S protein, virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4 (Hepatitis B Virus (HBV), Hepatitis B); envelope glycoprotein El gp32 gp35 , envelope glycoprotein E2 NS1 gp68 gp70, capsid protein C , core protein Core, polyprotein, virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4, antigen G, protein NS3, protein NS5A, (Hepatitis C Virus, Hepatitis C); virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4, large hepaptitis delta antigen, small hepaptitis delta antigen (Hepatitis D Virus, Hepatitis D); virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4, capsid protein E2 (Hepatitis E Virus, Hepatitis E); glycoprotein L UL1, uracil-DNA glycosylase UL2, protein UL3, protein UL4, DNA replication protein UL5, portal protein UL6, virion maturation protein UL7, DNA helicase UL8, replication origin-binding protein UL9, glycoprotein M UL10, protein UL11, alkaline exonuclease UL12, serine-threonine protein kinase UL13, tegument protein UL14, terminase UL15, tegument protein UL16, protein UL17, capsid protein VP23 UL18, major capsid protein VP5 UL19, membrane protein UL20, tegument protein UL21, Glycoprotein H (UL22), Thymidine Kinase UL23, protein UL24, protein UL25, capsid protein P40 (UL26, VP24, VP22A), glycoprotein B (UL27), ICP18.5 protein (UL28), major DNA-binding protein ICP8 (UL29), DNA polymerase UL30, nuclear matrix protein UL31, envelope glycoprotein UL32, protein UL33, inner nuclear membrane protein UL34, capsid protein VP26 (UL35), large tegument protein UL36, capsid assembly protein UL37, VP19C protein (UL38), ribonucleotide reductase (Large subunit) UL39, ribonucleotide reductase (Small subunit) UL40, tegument protein/virion host shutoff VHS protein (UL41), DNA polymerase processivity factor UL42, membrane protein UL43, glycoprotein C (UL44), membrane protein UL45, tegument proteins VPll/12 (UL46), tegument protein VP13/14 (UL47), virion maturation protein VP16 (UL48, Alpha-TIF), envelope protein UL49, dUTP diphosphatase UL50, tegument protein UL51, DNA helicase/primase complex protein UL52, glycoprotein K (UL53),
transcriptional regulation protein IE63 (ICP27, UL54), protein UL55, protein UL56, viral replication protein ICP22 (IE68, US1), protein US2, serine/threonine-protein kinase US3, glycoprotein G (US4), glycoprotein J (US5), glycoprotein D (US6), glycoprotein I (US7), glycoprotein E (US8), tegument protein US9, capsid/tegument protein US10, Vmw21 protein (US11), ICP47 protein (IE12, US12), major transcriptional activator ICP4 (IE175, RSI), E3 ubiquitin ligase ICP0 (IE110), latency-related protein 1 LRP1, latency-related protein 2 LRP2, neurovirulence factor RL1 (ICP34.5), latency-associated transcript LAT (Herpes simplex virus 1 and 2 (HSV-1 and HSV-2), Herpes simplex); heat shock protein Hsp60, cell surface protein H1C, dipeptidyl peptidase type IV DppIV, M antigen, 70 kDa protein, 17 kDa histone-like protein
(Histoplasma capsulatum, Histoplasmosis); fatty acid and retinol binding protein- 1 FAR-1, tissue inhibitor of metalloproteinase TIMP (TMP), cysteine proteinase ACEY-1, cysteine proteinase ACCP-1, surface antigen Ac-16, secreted protein 2 ASP-2, metalloprotease 1 MTP-1, aspartyl protease inhibitor API-1, surface-associated antigen SAA-1, surface-associated antigen SAA-2, adult-specific secreted factor Xa, serine protease inhibitor anticoagulant AP, cathepsin D-like aspartic protease ARR-1, glutathione S-transferase GST, aspartic protease APR-1,
acetylcholinesterase AChE (Ancylostoma duodenale and Necator americanus, Hookworm infection); protein NS1, protein NP1, protein VP1, protein VP2, protein VP3 (Human bocavirus (HBoV), Human bocavirus infection); major surface protein 2 MSP2, major surface protein 4 MSP4, MSP variant SGV1, MSP variant SGV2, outer membrane protein OMP, outer membrande protein 19 OMP-19, major antigenic protein MAPI, major antigenic protein MAP1-2, major antigenic protein MAP1B, major antigenic protein MAPl-3, Erum2510 coding protein, protein GroEL, protein GroES, 30-kDA major outer membrane proteins, GE 100-kDa protein, GE 130-kDa protein, GE 160-kDa protein (Ehrlichia ewingii, Human ewingii ehrlichiosis); major surface proteins 1-5 (MSPla, MSPlb, MSP2, MSP3, MSP4, MSP5), type IV secreotion system proteins VirB2, VirB7, VirBll, VirD4 (Anaplasma phagocytophilum, Human granulocytic anaplasmosis (HGA)); protein NSl, small hydrophobic protein NS2, SH protein, fusion protein F, glycoprotein G, matrix protein M, matrix protein M2-1, matrix protein M2-2, phosphoprotein P, nucleoprotein N, polymerase L (Human metapneumovirus (hMPV), Human metapneumovirus infection); major surface protein 2 MSP2, major surface protein 4 MSP4, MSP variant SGV1, MSP variant SGV2, outer membrane protein OMP, outer membrande protein 19 OMP-19, major antigenic protein MAPI, major antigenic protein MAP1-2, major antigenic protein MAP1B, major antigenic protein MAPl-3, Erum2510 coding protein, protein GroEL, protein GroES, 30-kDA major outer membrane proteins, GE 100-kDa protein, GE 130-kDa protein, GE 160-kDa protein (Ehrlichia chaffeensis, Human monocytic ehrlichiosis); replication protein El, regulatory protein E2, protein E3, protein E4, protein E5, protein E6, protein E7, protein E8, major capsid protein LI, minor capsid protein L2 (Human papillomavirus (HPV), Human papillomavirus (HPV) infection); fusion protein F, hemagglutinin-neuramidase HN, glycoprotein G, matrix protein M, phosphoprotein P, nucleoprotein N, polymerase L (Human parainfluenza viruses (HPIV), Human parainfluenza virus infection); Hemagglutinin (HA), Neuraminidase (NA), Nucleoprotein (NP), Ml protein, M2 protein, NSl protein, NS2 protein (NEP protein: nuclear export protein), PA protein, PB1 protein (polymerase basic 1 protein), PB1-F2 protein and PB2 protein (Orthomyxoviridae family, Influenza virus (flu)); genome polyprotein, protein E, protein M, capsid protein C (Japanese encephalitis virus, Japanese encephalitis); RTX toxin, type IV pili, major pilus subunit PilA, regulatory transcription factors PilS and PilR, protein sigma54, outer membrane proteins (Kingella kingae, Kingella kingae infection); prion protein (Kuru prion, Kuru); nucleoprotein N, polymerase L, matrix protein Z, glycoprotein GP (Lassa virus, Lassa fever); peptidoglycan- associated lipoprotein PAL, 60 kDa chaperonin Cpn60 (groEL, HspB), type IV pilin PilE, outer membrane protein MIP, major outer membrane protein MompS, zinc metalloproteinase MSP (Legionella pneumophila, Legionellosis (Legionnaires' disease, Pontiac fever)); P4 nuclease, protein WD, ribonucleotide reductase M2, surface membrane glycoprotein Pg46, cysteine proteinase CP, glucose-regulated protein 78 GRP-78, stage-specific S antigen-like protein A2, ATPase Fl, beta-tubulin, heat shock protein 70 Hsp70, KMP-11, glycoprotein GP63, protein BT1, nucleoside hydrolase NH, cell surface protein Bl, ribosomal protein Pl-like protein PI, sterol 24- c-methyltransferase SMT, LACK protein, histone HI, SPB1 protein, thiol specific antioxidant TSA, protein antigen STll, signal peptidase SP, histone H2B, suface antigen PSA-2, cystein proteinase b Cpb (Leishmania genus, Leishmaniasis); major membrane protein I, serine-rich antigen- 45 kDa, 10 kDa caperonin GroES, HSP kDa antigen, amino-oxononanoate synthase AONS, protein recombinase A RecA, Acetyl-/propionyl-coenzyme A carboxylase alpha, alanine racemase, 60 kDa chaperonin 2, ESAT-6-like protein EcxB (L-ESAT-6), protein Lsr2, protein ML0276, Heparin- binding hemagglutinin HBHA, heat-shock protein 65 Hsp65, mycPl or ML0041 coding protein , htrA2 or ML0176 coding protein , htrA4 or ML2659 coding protein, gcp or ML0379 coding protein, clpC or ML0235 coding protein (Mycobacterium leprae and Mycobacterium
lepromatosis, Leprosy); outer membrane protein LipL32, membrane protein LIC10258, membrane protein LP30, membrane protein LIC12238, Ompa-like protein Lsa66, surface protein LigA, surface protein LigB, major outer membrane protein OmpLl, outer membrane protein LipL41, protein LigAni, surface protein LcpA, adhesion protein LipL53, outer membrane protein UpL32, surface protein Lsa63, flagellin FlaBl, membran lipoprotein LipL21, membrane protein pL40, leptospiral surface adhesin Lsa27, outer membrane protein OmpL36, outer membrane protein OmpL37, outer membrane protein OmpL47, outer membrane protein OmpL54, acyltransferase LpxA (Leptospira genus, Leptospirosis); listeriolysin O precursor Hly (LLO), invasion-associated protein lap (P60), Listeriolysin regulatory protein PrfA, Zinc
metalloproteinase Mpl, Phosphatidylinositol- specific phospholipase C PLC (PlcA, PlcB), O- acetyltransferase Oat, ABC-transporter permease Im.G_1771, adhesion protein LAP, LAP receptor Hsp60, adhesin LapB, haemolysin listeriolysin O LLO, protein ActA, Internalin A InlA, protein InlB (Listeria monocytogenes, Listeriosis); outer surface protein A OspA, outer surface protein OspB, outer surface protein OspC, decorin binding protein A DbpA, decorin binding protein B DbpB, flagellar filament 41 kDa core protein Fla, basic membrane protein A BmpA (Immunodominant antigen P39), outer surface 22 kDa lipoprotein precursor (antigen IPLA7), variable surface lipoprotein vlsE (usually Borrelia burgdorferi and other Borrelia species, Lyme disease (Lyme borreliosis)); venom allergen homolog-like protein VAL-1, abundant larval transcript ALT- 1, abundant larval transcript ALT-2, thioredoxin peroxidase TPX, vespid allergen homologue VAH, thiordoxin peroxidase 2 TPX-2, antigenic protein SXP (peptides N, Nl, N2, and N3), activation associated protein-1 ASP-1, thioredoxin TRX, transglutaminase BmTGA, glutathione-S- transferases GST, myosin, vespid allergen homologue VAH, 175 kDa collagenase, glyceraldehyde- 3-phosphate dehydrogenase GAPDH, cuticular collagen Col-4, Secreted Larval Acidic Proteins SLAPs, chitinase CHI-1, maltose binding protein MBP, glycolytic enzyme fructose-1,6- bisphosphate aldolase Fba, tropomyosin TMY-1, nematode specific gene product OvB20, onchocystatin CPI-2, protein Cox-2 (Wuchereria bancrofti and Brugia malayi, Lymphatic filariasis (Elephantiasis)); glycoprotein GP, matrix protein Z, polymerase L, nucleoprotein N (Lymphocytic choriomeningitis virus (LCMV), Lymphocytic choriomeningitis); thrombospondin-related anonymous protein TRAP, SSP2 Sporozoite surface protein 2, apical membrane antigen 1 AMA1, rhoptry membrane antigen RMA1, acidic basic repeat antigen ABRA, cell-traversal protein PF, protein Pvs25, merozoite surface protein 1 MSP-1, merozoite surface protein 2 MSP-2, ring- infected erythrocyte surface antigen RESALiver stage antigen 3 LSA-3, protein Eba-175, serine repeat antigen 5 SERA-5, circumsporozoite protein CS, merozoite surface protein 3 MSP3, merozoite surface protein 8 MSP8, enolase PF10, hepatocyte erythrocyte protein 17 kDa HEP17, erythrocyte membrane protein 1 EMP1, protein Kbetamerozoite surface protein 4/5 MSP 4/5, heat shock protein Hsp90, glutamate-rich protein GLURP, merozoite surface protein 4 MSP-4, protein STARP, circumsporozoite protein-related antigen precursor CRA (Plasmodium genus, Malaria); nucleoprotein N, membrane-associated protein VP24, minor nucleoprotein VP30, polymerase cofactor VP35, polymerase L, matrix protein VP40, envelope glycoprotein GP (Marburg virus, Marburg hemorrhagic fever (MHF)); protein C, matrix protein M, phosphoprotein P, non-structural protein V, hemagglutinin glycoprotein H, polymerase L, nucleoprotein N, fusion protein F (Measles virus, Measles); members of the ABC transporter family (LolC, OppA, and PotF), putative lipoprotein releasing system transmembrane protein LolC/E, flagellin FliC, Burkholderia intracellular motility A BimA, bacterial Elongation factor-Tu EF-Tu, 17 kDa OmpA- like protein, boaA coding protein, boaB coding protein (Burkholderia pseudomallei, Melioidosis (Whitmore's disease)); pilin proteins, minor pilin-associated subunit pilC, major pilin subunit and variants pilE, pilS, phase variation protein porA, Porin B PorB, protein TraD, Neisserial outer membrane antigen H.8, 70kDa antigen, major outer membrane protein PI, outer membrane proteins P1A and P1B, W antigen, surface protein A NspA, transferrin binding protein TbpA, transferrin binding protein TbpB , PBP2, mtrR coding protein, ponA coding protein, membrane permease FbpBC, FbpABC protein system, LbpAB proteins, outer membrane protein Opa, outer membrane transporter FetA, iron-repressed regulator MpeR, factor H-binding protein fHbp, adhesin NadA, protein NhbA, repressor FarR (Neisseria meningitidis, Meningococcal disease); 66 kDa protein, 22 kDa protein (usually Metagonimus yokagawai, Metagonimiasis); polar tube proteins (34, 75, and 170 kDa in Glugea, 35, 55 and 150kDa in Encephalitozoon), kinesin-related protein, RNA polymerase II largest subunit, similar ot integral membrane protein YIPA, anti- silencing protein 1, heat shock transcription factor HSF, protein kinase, thymidine kinase, NOP-2 like nucleolar protein (Microsporidia phylum, Microsporidiosis); CASP8 and FADD-like apoptosis regulator, Glutathione peroxidase GPX1, RNA helicase NPH-II NPH2, Poly(A) polymerase catalytic subunit PAPL, Major envelope protein P43K, early transcription factor 70 kDa subunit VETFS, early transcription factor 82 kDa subunit VETFL, metalloendopeptidase Gl-type, nucleoside triphosphatase I NPHl, replication protein A28-like MC134L, RNA polymease 7 kDa subunit RP07 (Molluscum contagiosum virus (MCV), Molluscum contagiosum (MC)); matrix protein M, phosphoprotein P/V, small hydrophobic protein SH, nucleoprotein N, protein V, fusion glycoprotein F, hemagglutinin-neuraminidase HN, RNA polymerase L (Mumps virus, Mumps); Outer membrane proteins OM, cell surface antigen OmpA, cell surface antigen OmpB (sca5), cell surface protein SCA4, cell surface protein SCA1, intracytoplasmic protein D, crystalline surface layer protein SLP, protective surface protein antigen SPA (Rickettsia typhi, Murine typhus (Endemic typhus)); adhesin PI, adhesion P30, protein pll6, protein P40, cytoskeletal protein HMW1, cytoskeletal protein HMW2, cytoskeletal protein HMW3, MPN152 coding protein, MPN426 coding protein, MPN456 coding protein, MPN-500coding protein (Mycoplasma pneumoniae, Mycoplasma pneumonia); NocA, Iron dependent regulatory protein, VapA, VapD, VapF, VapG, caseinolytic protease, filament tip-associated 43-kDa protein, protein P24, protein P61, 15-kDa protein, 56-kDa protein (usually Nocardia asteroides and other Nocardia species, Nocardiosis); venom allergen homolog-like protein VAL-1, abundant larval transcript ALT- 1, abundant larval transcript ALT-2, thioredoxin peroxidase TPX, vespid allergen homologue VAH, thiordoxin peroxidase 2 TPX-2, antigenic protein SXP (peptides N, Nl, N2, and N3), activation associated protein-1 ASP-1, Thioredoxin TRX, transglutaminase BmTGA, glutathione-S- transferases GST, myosin, vespid allergen homologue VAH, 175 kDa collagenase, glyceraldehyde- 3-phosphate dehydrogenase GAPDH, cuticular collagen Col-4, Secreted Larval Acidic Proteins SLAPs, chitinase CHI-1, maltose binding protein MBP, glycolytic enzyme fructose-1,6- bisphosphate aldolase Fba, tropomyosin TMY-1, nematode specific gene product OvB20, onchocystatin CPI-2, Cox-2 (Onchocerca volvulus, Onchocerciasis (River blindness)); 43 kDa secreted glycoprotein, glycoprotein gpO, glycoprotein gp75, antigen Pb27, antigen Pb40, heat shock protein Hsp65, heat shock protein Hsp70, heat shock protein Hsp90, protein P10, triosephosphate isomerase TPI, N-acetyl-glucosamine-binding lectin Paracoccin, 28 kDa protein Pb28 (Paracoccidioides brasiliensis, Paracoccidioidomycosis (South American blastomycosis)); 28-kDa cruzipain-like cystein protease Pw28CCP (usually Paragonimus westermani and other Paragonimus species, Paragonimiasis); outer membrane protein OmpH, outer membrane protein Omp28, protein PM1539, protein PM0355, protein PM1417, repair protein MutL, protein BcbC, prtein PM0305, formate dehydrogenase-N, protein PM0698, protein PM1422, DNA gyrase, lipoprotein PlpE, adhesive protein Cp39, heme aquisition system receptor HasR, 39 kDa capsular protein, iron-regulated OMP IROMP, outer membrane protein OmpA87, fimbrial protein Ptf, fimbrial subunit protein PtfA, transferrin binding protein Tbpl, esterase enzyme MesA,
Pasteurella multocida toxin PMT, adhesive protein Cp39 (Pasteurella genus, Pasteurellosis); "filamentous hemagglutinin FhaB, adenylate cyclase CyaA, pertussis toxin subunit 4 precursor PtxD, pertactin precursor Prn, toxin subunit 1 PtxA, protein Cpn60, protein brkA, pertussis toxin subunit 2 precursor PtxB, pertussis toxin subunit 3 precursor PtxC, pertussis toxin subunit 5 precursor PtxE, pertactin Prn, protein Fim2, protein Fim3; " (Bordetella pertussis, Pertussis (Whooping cough)); "Fl capsule antigen, virulence-associated V antigen, secreted effector protein LcrV, V antigen, outer membrane protease Pla,secreted effector protein YopD, putative secreted protein-tyrosine phosphatase YopH, needle complex major subunit YscF, protein kinase YopO, putative autotransporter protein YapF, inner membrane ABC-transporter YbtQ (Irp7), putative sugar binding protein YPO0612, heat shock protein 90 HtpG, putative sulfatase protein YdeN, outer-membrane lipoprotein carrier protein LolA, secretion chaperone YerA, putative lipoprotein YPO0420, hemolysin activator protein HpmB, pesticin/yersiniabactin outer membrane receptor Psn, secreted effector protein YopE, secreted effector protein YopF, secreted effector protein YopK, outer membrane protein YopN , outer membrane protein YopM, Coagulase/fibrinolysin precursor Pla ; " (Yersinia pestis, Plague); protein PhpA, surface adhesin PsaA, pneumolysin Ply, ATP-dependent protease Clp, lipoate-protein ligase LplA, cell wall surface anchored protein psrP, sortase SrtA, glutamyl-tRNA synthetase GltX, choline binding protein A CbpA, pneumococcal surface protein A PspA, pneumococcal surface protein C PspC, 6-phosphogluconate
dehydrogenase Gnd, iron-binding protein PiaA, Murein hydrolase LytB, proteon LytC, protease Al (Streptococcus pneumoniae, Pneumococcal infection); major surface protein B, kexin-like protease KEX1, protein A12, 55 kDa antigen P55, major surface glycoprotein Msg (Pneumocystis jirovecii, Pneumocystis pneumonia (PCP)); genome polyprotein, polymerase 3D, viral capsid protein VP1, viral capsid protein VP2, viral capsid protein VP3, viral capsid protein VP4, protease 2A, protease 3C (Poliovirus, Poliomyelitis); protein Nfal, exendin-3, secretory lipase, cathepsin B- like protease, cysteine protease, cathepsin, peroxiredoxin, protein CrylAc (usually Naegleria fowleri, Primary amoebic meningoencephalitis (PAM)); agnoprotein, large T antigen, small T antigen, major capsid protein VP1, minor capsid protein Vp2 (JC virus, Progressive multifocal leukoencephalopathy); low calcium response protein E LCrE, chlamydial outer protein N CopN, serine/threonine-protein kinase PknD, acyl-carrier-protein S-malonyltransferase FabD, single- stranded DNA-binding protein Ssb, major outer membrane protein MOMP, outer membrane protein 2 Omp2, polymorphic membrane protein family (Pmpl, Pmp2, Pmp3, Pmp4, Pmp5, Pmp6, Pmp7, Pmp8, Pmp9, PmplO, Pmpll, Pmpl2, Pmpl3, Pmpl4, Pmpl5, Pmpl6, Pmpl7, Pmpl8, Pmpl9, Pmp20, Pmp21) (Chlamydophila psittaci, Psittacosis); outer membrane protein PI, heat shock protein B HspB, peptide ABC transporter, GTP-binding protein, protein IcmB, ribonuclease R, phosphatas SixA, protein DsbD, outer membrane protein TolC, DNA-binding protein PhoB, ATPase DotB, heat shock protein B HspB, membrane protein Coml, 28 kDa protein, DNA-3-methyladenine glycosidase I, pouter membrane protein OmpH, outer membrane protein AdaA, glycine cleavage system T-protein (Coxiella burnetii, Q fever); nucleoprotein N, large structural protein L, phophoprotein P, matrix protein M, glycoprotein G (Rabies virus, Rabies); fusionprotein F, nucleoprotein N, matrix protein M, matrix protein M2-1, matrix protein M2-2, phophoprotein P, small hydrophobic protein SH, major surface glycoprotein G, polymerase L, non-structural protein 1 NS1, non-structural protein 2 NS2 (Respiratory syncytial virus (RSV), Respiratory syncytial virus infection); genome polyprotein, polymerase 3D, viral capsid protein VP1, viral capsid protein VP2, viral capsid protein VP3, viral capsid protein VP4, protease 2A, protease 3C (Rhinovirus, Rhinovirus infection); outer membrane proteins OM, cell surface antigen OmpA, cell surface antigen OmpB (sca5), cell surface protein SCA4, cell surface protein SCAl, protein PS120, intracytoplasmic protein D, protective surface protein antigen SPA (Rickettsia genus, Rickettsial infection); outer membrane proteins OM, cell surface antigen OmpA, cell surface antigen OmpB (sca5), cell surface protein SCA4, cell surface protein SCAl,
intracytoplasmic protein D (Rickettsia akari, Rickettsialpox); envelope glycoprotein GP, polymerase L, nucleoprotein N, non-structural protein NSS (Rift Valley fever virus, Rift Valley fever (RVF)); outer membrane proteins OM, cell surface antigen OmpA, cell surface antigen OmpB (sca5), cell surface protein SCA4, cell surface protein SCAl, intracytoplasmic protein D (Rickettsia rickettsii, Rocky mountain spotted fever (RMSF)); "non-structural protein 6 NS6, non-structural protein 2 NS2, intermediate capsid protein VP6, inner capsid protein VP2, non-structural protein 3 NS3, RNA-directed RNA polymerase L, protein VP3, non-structural protein 1 NS1, nonstructural protein 5 NS5, outer capsid glycoprotein VP7, non-structural glycoprotein 4 NS4, outer capsid protein VP4" (Rotavirus, Rotavirus infection); polyprotein P200, glycoprotein El, glycoprotein E2, protein NS2, capsid protein C (Rubella virus, Rubella); chaperonin GroEL
(MopA), inositol phosphate phosphatase SopB, heat shock protein HslU, chaperone protein DnaJ, protein TviB, protein IroN, flagellin FliC, invasion protein SipC, glycoprotein gp43, outer membrane protein LamB, outer membrane protein PagC, outer membrane protein TolC, outer membrane protein NmpC, outer membrane protein FadL, transport protein SadA, transferase WgaP, effector proteins SifA, SteC, SseL, SseJ and SseF (Salmonella genus, Salmonellosis), protein 14, non-structural protein NS7b, non-structural protein NS8a, protein 9b, protein 3a,
nucleoprotein N, non-structural protein NS3b, non-structural protein NS6, protein 7a, nonstructural protein NS8b, membrane protein M, envelope small membrane protein EsM, replicase polyprotein la, spike glycoprotein S, replicase polyprotein lab; (SARS coronavirus, SARS (Severe Acute Respiratory Syndrome)); serin protease, Atypical Sarcoptes Antigen 1 ASA1, glutathione S- transferases GST, cystein protease, serine protease, apolipoprotein (Sarcoptes scabiei, Scabies); glutathione S-transferases GST, paramyosin, hemoglbinase SM32, major egg antigen, 14 kDa fatty acid-binding protein Sml4, major larval surface antigen P37, 22,6 kDa tegumental antigen, calpain CANP, triphospate isomerase Tim, surface protein 9B, outer capsid protein VP2, 23 kDa integral membrane protein Sm23, Cu/Zn-superoxide dismutase, glycoprotein Gp, myosin
(Schistosoma genus, Schistosomiasis (Bilharziosis)); 60 kDa chaperonin, 56 kDa type-specific antigen, pyruvate phosphate dikinase, 4-hydroxybenzoate octaprenyltransferase (Orientia tsutsugamushi, Scrub typhus); dehydrogenase GuaB, invasion protein Spa32, invasin IpaA, invasin IpaB, invasin IpaC, invasin IpaD, invasin IpaH, invasin IpaJ (Shigella genus, Shigellosis (Bacillary dysentery)); protein P53, virion protein US10 homolog, transcriptional regulator IE63, transcriptional transactivator IE62, protease P33, alpha trans-inducing factor 74 kDa protein, deoxyuridine 5'-triphosphate nucleotidohydrolase, transcriptional transactivator IE4, membrane protein UL43 homolog, nuclear phosphoprotein UL3 homolog, nuclear protein UL4 homolog, replication origin-binding protein, membrane protein 2, phosphoprotein 32, protein 57,DNA polymerase processivity factor, portal protein 54, DNA primase, tegument protein UL14 homolog, tegument protein UL21 homolog, tegument protein UL55 homolog,tripartite terminase subunit UL33 homolog,tripartite terminase subunit UL15 homolog, capsid-binding protein 44, virion- packaging protein 43 [Varicella zoster virus (VZV), Shingles (Herpes zoster)); truncated 3-beta hydroxy-5-ene steroid dehydrogenase homolog, virion membrane protein A13, protein A19, protein A31, truncated protein A35 homolog, protein A37.5 homolog, protein A47, protein A49, protein A51, semaphorin-like protein A43, serine proteinase inhibitor 1, serine proteinase inhibitor 2, serine proteinase inhibitor 3, protein A6, protein B15, protein CI, protein C5, protein C6, protein F7, protein F8, protein F9, protein Fll, protein F14, protein F15, protein F16 [Variola major or Variola minor, Smallpox [Variola)); adhesin/glycoprotein gp70, proteases (Sporothrix schenckii, Sporotrichosis); heme-iron binding protein IsdB, collagen adhesin Cna, clumping factor A ClfA, protein MecA, fibronectin-binding protein A FnbA, enterotoxin type A EntA, enterotoxin type B EntB, enterotoxin type C EntCl, enterotoxin type C EntC2, enterotoxin type D EntD, enterotoxin type E EntE, Toxic shock syndrome toxin-1 TSST-1, Staphylokinase, Penicillin binding protein 2a PBP2a (MecA), secretory antigen SssA (Staphylococcus genus, Staphylococcal food poisoning); heme-iron binding protein IsdB, collagen adhesin Cna, clumping factor A ClfA, protein MecA, fibronectin-binding protein A FnbA, enterotoxin type A EntA, enterotoxin type B EntB, enterotoxin type C EntCl, enterotoxin type C EntC2, enterotoxin type D EntD, enterotoxin type E EntE, Toxic shock syndrome toxin-1 TSST-1, Staphylokinase, Penicillin binding protein 2a PBP2a (MecA), secretory antigen SssA (Staphylococcus genus e.g. aureus, Staphylococcal infection); antigen Ss-IR, antigen NIE, strongylastacin, Na+-K+ ATPase Sseat-6, tropomysin SsTmy-1, protein LEC-5, 41 kDa aantigen P5, 41-kDa larval protein, 31-kDa larval protein, 28-kDa larval protein (Strongyloides stercoralis, Strongyloidiasis); glycerophosphodiester phosphodiesterase GlpQ (Gpd), outer membrane protein TmpB, protein Tp92, antigen TpFl, repeat protein Tpr, repeat protein F TprF, repeat protein G TprG, repeat protein I Tprl, repeat protein J TprJ, repeat protein K TprK, treponemal membrane protein A TmpA, lipoprotein, 15 kDa Tppl5, 47 kDa membrane antigen, miniferritin TpFl, adhesin Tp0751, lipoprotein TP0136, protein TpN17, protein TpN47, outer membrane protein TP0136, outer membrane protein TP0155, outer membrane protein TP0326, outer membrane protein TP0483, outer membrane protein TP0956 (Treponema pallidum, Syphilis); Cathepsin L-like proteases, 53/25-kDa antigen, 8kDa family members, cysticercus protein with a marginal trypsin-like activity TsAg5, oncosphere protein TSOL18, oncosphere protein TSOL45-1A, lactate dehydrogenase A LDHA, lactate dehydrogenase B LDHB (Taenia genus, Taeniasis); tetanus toxin TetX, tetanus toxin C TTC, 140 kDa S layer protein, flavoprotein beta-subunit CT3, phospholipase (lecithinase), phosphocarrier protein HPr
(Clostridium tetani, Tetanus (Lockjaw)); genome polyprotein, protein E, protein M, capsid protein C (Tick-borne encephalitis virus (TBEV), Tick-borne encephalitis); 58-kDa antigen, 68-kDa antigens, Toxocara larvae excretory-secretory antigen TES, 32-kDa glycoprotein, glycoprotein TES-70, glycoprotein GP31, excretory-secretory antigen TcES-57, perienteric fluid antigen Pe, soluble extract antigens Ex, excretory/secretory larval antigens ES, antigen TES-120, polyprotein allergen TBA-1, cathepsin L-like cysteine protease c-cpl-1, 26-kDa protein (Toxocara canis or Toxocara cati, Toxocariasis (Ocular Larva Migrans (OLM) and Visceral Larva Migrans (VLM))); microneme proteins ( MICl, MIC2, MIC3, MIC4, MIC5, MIC6, MIC7, MIC8), rhoptry protein Rop2, rhoptry proteins (Ropl, Rop2, Rop3, Rop4, Rop5, Rop6, Rop7, Ropl6, Rjopl7), protein
SRl,surface antigen P22, major antigen p24, major surface antigen p30, dense granule proteins (GRA1, GRA2, GRA3, GRA4, GRA5, GRA6, GRA7, GRA8, GRA9, GRA10), 28 kDa antigen, surface antigen SAG1, SAG2 related antigen, nucleoside-triphosphatase 1, nucleoside-triphosphatase 2, protein Stt3, HesB-like domain-containing protein, rhomboid-like protease 5, toxomepsin 1 (Toxoplasma gondii, Toxoplasmosis); 43 kDa secreted glycoprotein, 53 kDa secreted
glycoprotein, paramyosin, antigen Ts21, antigen Ts87, antigen p46000, TSL-1 antigens, caveolin-1 CAV-1, 49 kDa newborn larva antigen, prosaposin homologue, serine protease, serine proteinase inhibitor, 45 -kDa glycoprotein Gp45 (Trichinella spiralis, Trichinellosis); Myb-like transcriptional factors (Mybl, Myb2, Myb3), adhesion protein AP23, adhesion protein AP33, adhesin protein AP33-3, adhesins AP51, adhesin AP65, adhesion protein AP65-1, alpha-actinin, kinesin-associated protein, teneurin, 62 kDa proteinase, subtilisin-like serine protease SUB1, cysteine proteinase gene 3 CP3, alpha-enolase Enol, cysteine proteinase CP30, heat shock proteins (Hsp70, Hsp60) , immunogenic protein P270, (Trichomonas vaginalis, Trichomoniasis); beta-tubulin, 47-kDa protein, secretory leucocyte-like proteinase-1 SLP-1, 50-kDa protein TT50, 17 kDa antigen, 43/47 kDa protein (Trichuris trichiura, Trichuriasis (Whipworm infection)); protein ESAT-6 (EsxA), 10 kDa filtrate antigen EsxB, secreted antigen 85-B FBPB, fibronectin-binding protein A FbpA (Ag85A), serine protease PepA, PPE family protein PPE18, fibronectin-binding protein D FbpD, immunogenic protein MPT64, secreted protein MPT51, catalase-peroxidase-peroxynitritase T KATG, periplasmic phosphate-binding lipoprotein PSTS3 (PBP-3, Phos-1), iron-regulated heparin binding hemagglutinin Hbha, PPE family protein PPE14, PPE family protein PPE68, protein
Mtb72F, protein Apa, immunogenic protein MPT63, periplasmic phosphate-binding lipoprotein PSTS1 (PBP-1), molecular chaperone DnaK, cell surface lipoprotein Mpt83, lipoprotein P23, phosphate transport system permease protein pstA, 14 kDa antigen, fibronectin-binding protein C FbpCl, Alanine dehydrogenase TB43, Glutamine synthetase 1, ESX-1 protein, protein CFP10, TB10.4 protein, protein MPT83, protein MTB12, protein MTB8, Rpf-like proteins, protein MTB32, protein MTB39, crystallin, heat-shock protein HSP65, protein PST-S (usually Mycobacterium tuberculosis, Tuberculosis); outer membrane protein FobA, outer membrane protein FobB, intracellular growth locus lglCl, intracellular growth locus IglC2, aminotransferase Wbtl, chaperonin GroEL, 17 kDa major membrane protein TUL4, lipoprotein LpnA, chitinase family 18 protein, isocitrate dehydrogenase, Nif3 family protein, type IV pili glycosylation protein, outer membrane protein tolC, FAD binding family protein, type IV pilin multimeric outer membrane protein, two component sensor protein KdpD, chaperone protein DnaK, protein TolQ (Francisella tularensis, Tularemia); "MB antigen, urease, protein GyrA, protein GyrB, protein ParC, protein ParE, lipid associated membrane proteins LAMP, thymidine kinase TK, phospholipase PL-A1, phospholipase PL-A2, phospholipase PL-C, surface-expressed 96-kDa antigen; " (Ureaplasma urealyticum, Ureaplasma urealyticum infection); non-structural polyprotein, structural polyprotein, capsid protein CP, protein El, protein E2, protein E3, protease PI, protease P2, protease P3 [Venezuelan equine encephalitis virus, Venezuelan equine encephalitis); glycoprotein GP, matrix protein Z, polymerase L, nucleoprotein N (Guanarito virus, Venezuelan hemorrhagic fever); polyprotein, protein E, protein M, capsid protein C, protease NS3, protein NS1, protein NS2A, protein AS2B, brotein NS4A, protein NS4B, protein NS5 [West Nile virus, West Nile Fever); cpasid protein CP, protein El, protein E2, protein E3, protease P2 (Western equine encephalitis virus, Western equine encephalitis); genome polyprotein, protein E, protein M, capsid protein C, protease NS3, protein NS1, protein NS2A, protein AS2B, protein NS4A, protein NS4B, protein NS5 (Yellow fever virus, Yellow fever); putative Yop targeting protein YobB, effector protein YopD, effector protein YopE, protein YopH, effector protein YopJ, protein translocation protein YopK, effector protein YopT, protein YpkA, flagellar biosyntheses protein FlhA, peptidase M48, potassium efflux system KefA, transcriptional regulatoer RovA, adhesin Ifp, translocator portein LcrV, protein PcrV, invasin Inv, outer membrane protein OmpF-like porin, adhesin YadA, protein kinase C, phospholipase CI, protein PsaA, mannosyltransferase-like protein WbyK, protein YscU, antigen YPMa (Yersinia pseudotuberculosis, Yersinia pseudotuberculosis infection); effector protein YopB, 60 kDa chaperonin, protein WbcP, tyrosin-protein phosphatase YopH, protein YopQ, enterotoxin, Galactoside permease, reductaase NrdE, protein YasN, Invasin Inv, adhesin YadA, outer membrane porin F OmpF, protein UspAl, protein EibA, protein Hia, cell surface protein Ail, chaperone SycD, protein LcrD, protein LcrG, protein LcrV, protein SycE, protein YopE, regulator protein TyeA, protein YopM, protein YopN, protein YopO, protein YopT, protein YopD, protease ClpP, protein MyfA, protein FilA, and protein PsaA (Yersinia enterocolitica, Yersiniosis) (in brackets is the particular pathogen or the family of pathogens of which the antigen(s) is/are derived and the infectious disease with which the pathogen is associated).
In particularly preferred embodiments the pathogenic antigen is selected from a) HIV p24 antigen, HIV envelope proteins (Gpl20, Gp41, Gpl60), polyprotein GAG, negative factor protein Nef, trans-activator of transcription Tat if the infectious disease is HIV, preferably an infection with Human immunodeficiency virus, b) major outer membrane protein MOMP, probable outer membrane protein PMPC, outer membrane complex protein B OmcB, heat shock proteins Hsp60 HSP10, protein IncA, proteins from the type III secretion system, ribonucleotide reductase small chain protein NrdB, plasmid protein Pgp3, chlamydial outer protein N CopN, antigen CT521, antigen CT425, antigen CT043, antigen TC0052, antigen TC0189, antigen TC0582, antigen TC0660, antigen TC0726, antigen TC0816, antigen TC0828 if the infectious disease is an infenction with Chlamydia trachomatis, c) pp65 antigen, membrane protein ppl5, capsid-proximal tegument protein ppl50, protein M45, DNA polymerase UL54, helicase UL105, glycoprotein gM, glycoprotein gN, glcoprotein H, glycoprotein B gB, protein UL83, protein UL94, protein UL99 if the infectious disease is Cytomegalovirus infection, preferably an infection with Cytomegalovirus (CMV); d) capsid protein C, premembrane protein prM, membrane protein M, envelope protein E (domain I, domain II, domain II), protein NS1, protein NS2A, protein NS2B, protein NS3, protein NS4A, protein 2K, protein NS4B, protein NS5 if the infectious disease is Dengue fever, preferably an infection with Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN-4)-Flaviviruses; e) hepatitis B surface antigen HBsAg, Hepatitis B core antigen HbcAg, polymerase, protein Hbx, preS2 middle surface protein, surface protein L, large S protein, virus protein VP1, virus protein VP2, virus protein VP3, virus protein VP4 if the infectious disease is Hepatits B, preferably an infection with Hepatitis B Virus (HBV); f) replication protein El, regulatory protein E2, protein E3, protein E4, protein E5, protein E6, protein E7, protein E8, major capsid protein LI, minor capsid protein L2 if the infectious disease is Human papillomavirus (HPV) infection, preferably an infection with Human papillomavirus (HPV); g) fusion protein F, hemagglutinin-neuramidase HN, glycoprotein G, matrix protein M, phosphoprotein P, nucleoprotein N, polymerase L if the infectious disease is Human
parainfluenza virus infection, preferably an infection with Human parainfluenza viruses (HPIV); h) Hemagglutinin (HA), Neuraminidase (NA), Nucleoprotein (NP), Ml protein, M2 protein, NS1 protein, NS2 protein (NEP protein: nuclear export protein), PA protein, PB1 protein
(polymerase basic 1 protein), PB1-F2 protein and PB2 protein (Orthomyxoviridae family, Influenza virus (flu)); i) nucleoprotein N, large structural protein L, phophoprotein P, matrix protein M, glycoprotein G if the infectious disease is Rabies, preferably an infection with Rabies virus; j) fusionprotein F, nucleoprotein N, matrix protein M, matrix protein M2-1, matrix protein M2-2, phophoprotein P, small hydrophobic protein SH, major surface glycoprotein G, polymerase L, non-structural protein 1 NS1, non-structural protein 2 NS2 if the infectious disease is
Respiratory syncytial virus infection, preferably an infection with Respiratory syncytial virus (RSV); k) secretory antigen SssA (Staphylococcus genus, Staphylococcal food poisoning); secretory antigen SssA (Staphylococcus genus e.g. aureus, Staphylococcal infection); molecular chaperone DnaK, cell surface lipoprotein Mpt83, lipoprotein P23, phosphate transport system permease protein pstA, 14 kDa antigen, fibronectin-binding protein C FbpCl, Alanine dehydrogenase TB43, Glutamine synthetase 1, ESX-1 protein, protein CFP10, TB10.4 protein, protein MPT83, protein MTB12, protein MTB8, Rpf-like proteins, protein MTB32, protein MTB39, crystallin, heat-shock protein HSP65, protein PST-S if the infectious disease is Tuberculosis, preferably an infection with Mycobacterium tuberculosis; or genome polyprotein, protein E, protein M, capsid protein C, protease NS3, protein NS1, protein NS2A, protein AS2B, protein NS4A, protein NS4B, protein NS5 if the infectious disease is Yellow fever, perferably an infection with Yellow fever virus.
Examples
The following examples are intended to further illustrate the invention. They are merely illustrative and not intended to limit the scope of the subject matter of the invention.
Example 1: Preparation of DNA and mRNA constructs
For the present examples, a DNA sequence encoding Gaussia princeps luciferase (GpLuc), Photinus pyralis luciferase (PpLuc) and Mus musculus erythropoietin (MmEpo) was prepared and used for subsequent RNA in vitro transcription reactions. The obtained mRNA constructs were used for further in vitro and in vivo experiments. The respective amino acid sequences, the mRNA sequences of GpLuc, PpLuc and MmEpo as well as preparation step details are provided below.
GpLuc, amino acid sequence (SEQ ID NO: 11) :
MGVKVLFALICIAVAEAKPTENNEDFNIVAVASNFATTDLDADRGKLPGKKLPLEVLKEMEANAR KAGCTRGCLICLSHIKCTPKMKKFIPGRCHTYEGDKESAQGGIGEAIVDIPEIPGFKDLEPMEQFIAQVDLC VDCTTGCLKGLANVQCSDLLKKWLPQRCATFASKIQGQVDKIKGAGGD
PpLuc, amino acid sequence (SEQ ID NO: 12) :
MEDAKNIKKGPAPFYPLEDGTAGEQLHKAMKRYALVPGTIAFTDAHIEVDITYAEYFEMSVRLAE AMKRYGLNTNHRIWCSENSLQFFMPVLGALFIGVAVAPANDIYNERELLNSMGISQPTWFVSKKGLQKI LNVQKKLPIIQKIIIMDSKTDYQGFQSMYTFVTSHLPPGFNEYDFVPESFDRDKTIALIMNSSGSTGLPKGV ALPHRTACVRFSHARDPIFGNQIIPDTAILSWPFHHGFGMFTTLGYLICGFRWLMYRFEEELFLRSLQDY KIQSALLVPTLFSFFAKSTLIDKYDLSNLHEIASGGAPLSKEVGEAVAKRFHLPGIRQGYGLTETTSAILITPE GDDKPGAVGKWPFFEAKWDLDTGKTLGVNQRGELCVRGPMIMSGYVNNPEATNALIDKDGWLHSGDI AYWDEDEHFFIVDRLKSLIKYKGYQVAPAELESILLQHPNIFDAGVAGLPDDDAGELPAAVWLEHGKTM TEKEIVDYVASQVTTAKKLRGGWFVDEVPKGLTGKLDARKIREILIKAKKGGKIAV
MmEpo, amino acid sequence (SEQ ID NO: 13):
MGVPERPTLLLLLSLLLIPLGLPVLCAPPRLICDSRVLERYILEAKEAENVTMGCAEGPRLSENITVP DTKVNFYAWKRMEVEEQAIEVWQGLSLLSEAILQAQALLANSSQPPETLQLHIDKAISGLRSLTSLLRVLG AQKELMSPPDTTPPAPLRTLTVDTFCKLFRVYANFLRGKLKLYTGEVCRRGDR
GpLuc, mRNA sequence (SEQ ID NO: 14), also labelled R2851 herein:
GGGGCGCUGCCUACGGAGGUGGCAGCCAUCUCCUUCUCGGCAUCAAGCUUACCAUGGGCGUGAA GGUCCUGUUCGCCCUCAUCUGCAUCGCCGUGGCGGAGGCCAAGCCCACCGAGAACAACGAGGACUUCAA CAUCGUGGCCGUCGCCAGCAACUUCGCCACCACGGACCUGGACGCGGACCGGGGGAAGCUGCCGGGCAA GAAGCUCCCCCUGGAGGUGCUGAAGGAGAUGGAGGCCAACGCCCGCAAGGCCGGGUGCACCCGGGGCUG CCUCAUCUGCCUGUCCCACAUCAAGUGCACCCCCAAGAUGAAGAAGUUCAUCCCCGGGCGCUGCCACAC CUACGAGGGCGACAAGGAGAGCGCGCAGGGCGGGAUCGGCGAGGCCAUCGUGGACAUCCCGGAGAUCCC CGGGUUCAAGGACCUGGAGCCCAUGGAGCAGUUCAUCGCCCAGGUCGACCUCUGCGUGGACUGCACGAC CGGCUGCCUGAAGGGGCUGGCCAACGUGCAGUGCUCCGACCUCCUGAAGAAGUGGCUGCCCCAGCGGUG CGCCACCUUCGCGAGCAAGAUCCAGGGCCAGGUCGACAAGAUCAAGGGCGCCGGGGGCGACUGAGGACU AGUGCAUCACAUUUAAAAGCAUCUCAGCCUACCAUGAGAAUAAGAGAAAGAAAAUGAAGAUCAAUAG CUUAUUCAUCUCUUUUUCUUUUUCGUUGGUGUAAAGCCAACACCCUGUCUAAAAAACAUAAAUUUCU UUAAUCAUUUUGCCUCUUUUCUCUGUGCUUCAAUUAAUAAAAAAUGGAAAGAACCUAGAUCUAAAAA AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAUGCAUCCC CCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCUCUUUUCAGAGCCACCAGAAUU
PpLuc, mRNA sequence (SEQ ID NO: 15) :
GGGGCGCUGCCUACGGAGGUGGCAGCCAUCUCCUUCUCGGCAUCAAGCUUGAGGAUGGAGGACG CCAAGAACAUCAAGAAGGGCCCGGCGCCCUUCUACCCGCUGGAGGACGGGACCGCCGGCGAGCAGCUCC ACAAGGCCAUGAAGCGGUACGCCCUGGUGCCGGGCACGAUCGCCUUCACCGACGCCCACAUCGAGGUCG ACAUCACCUACGCGGAGUACUUCGAGAUGAGCGUGCGCCUGGCCGAGGCCAUGAAGCGGUACGGCCUG AACACCAACCACCGGAUCGUGGUGUGCUCGGAGAACAGCCUGCAGUUCUUCAUGCCGGUGCUGGGCGCC CUCUUCAUCGGCGUGGCCGUCGCCCCGGCGAACGACAUCUACAACGAGCGGGAGCUGCUGAACAGCAUG GGGAUCAGCCAGCCGACCGUGGUGUUCGUGAGCAAGAAGGGCCUGCAGAAGAUCCUGAACGUGCAGAA GAAGCUGCCCAUCAUCCAGAAGAUCAUCAUCAUGGACAGCAAGACCGACUACCAGGGCUUCCAGUCGA UGUACACGUUCGUGACCAGCCACCUCCCGCCGGGCUUCAACGAGUACGACUUCGUCCCGGAGAGCUUCG ACCGGGACAAGACCAUCGCCCUGAUCAUGAACAGCAGCGGCAGCACCGGCCUGCCGAAGGGGGUGGCCC UGCCGCACCGGACCGCCUGCGUGCGCUUCUCGCACGCCCGGGACCCCAUCUUCGGCAACCAGAUCAUCCC GGACACCGCCAUCCUGAGCGUGGUGCCGUUCCACCACGGCUUCGGCAUGUUCACGACCCUGGGCUACCU CAUCUGCGGCUUCCGGGUGGUCCUGAUGUACCGGUUCGAGGAGGAGCUGUUCCUGCGGAGCCUGCAGG ACUACAAGAUCCAGAGCGCGCUGCUCGUGCCGACCCUGUUCAGCUUCUUCGCCAAGAGCACCCUGAUCG ACAAGUACGACCUGUCGAACCUGCACGAGAUCGCCAGCGGGGGCGCCCCGCUGAGCAAGGAGGUGGGCG AGGCCGUGGCCAAGCGGUUCCACCUCCCGGGCAUCCGCCAGGGCUACGGCCUGACCGAGACCACGAGCG CGAUCCUGAUCACCCCCGAGGGGGACGACAAGCCGGGCGCCGUGGGCAAGGUGGUCCCGUUCUUCGAGG CCAAGGUGGUGGACCUGGACACCGGCAAGACCCUGGGCGUGAACCAGCGGGGCGAGCUGUGCGUGCGGG GGCCGAUGAUCAUGAGCGGCUACGUGAACAACCCGGAGGCCACCAACGCCCUCAUCGACAAGGACGGCU GGCUGCACAGCGGCGACAUCGCCUACUGGGACGAGGACGAGCACUUCUUCAUCGUCGACCGGCUGAAG UCGCUGAUCAAGUACAAGGGCUACCAGGUGGCGCCGGCCGAGCUGGAGAGCAUCCUGCUCCAGCACCCC AACAUCUUCGACGCCGGCGUGGCCGGGCUGCCGGACGACGACGCCGGCGAGCUGCCGGCCGCGGUGGUG GUGCUGGAGCACGGCAAGACCAUGACGGAGAAGGAGAUCGUCGACUACGUGGCCAGCCAGGUGACCAC CGCCAAGAAGCUGCGGGGCGGCGUGGUGUUCGUGGACGAGGUCCCGAAGGGCCUGACCGGGAAGCUCG ACGCCCGGAAGAUCCGCGAGAUCCUGAUCAAGGCCAAGAAGGGCGGCAAGAUCGCCGUGUAAGACUAG UGCAUCACAUUUAAAAGCAUCUCAGCCUACCAUGAGAAUAAGAGAAAGAAAAUGAAGAUCAAUAGCU UAUUCAUCUCUUUUUCUUUUUCGUUGGUGUAAAGCCAACACCCUGUCUAAAAAACAUAAAUUUCUUU AAUCAUUUUGCCUCUUUUCUCUGUGCUUCAAUUAAUAAAAAAUGGAAAGAACCUAGAUCUAAAAAAA AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAUGCAUCCCCC CCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCUCUUUUCAGAGCCACCAGAAUU
MmEpo mRNA sequence (SEQ ID NO: 16) : GGGUCCCGCAGUCGGCGUCCAGCGGCUCUGCUUGUUCGUGUGUGUGUCGUUGCAGGCCUUAUUC AAGCUUACCAUGGGCGUGCCCGAGCGGCCGACCCUGCUCCUGCUGCUCAGCCUGCUGCUCAUCCCCCUG GGGCUGCCCGUCCUCUGCGCCCCCCCGCGCCUGAUCUGCGACUCCCGGGUGCUGGAGCGCUACAUCCUC GAGGCCAAGGAGGCGGAGAACGUGACCAUGGGCUGCGCCGAGGGGCCCCGGCUGAGCGAGAACAUCACG GUCCCCGACACCAAGGUGAACUUCUACGCCUGGAAGCGCAUGGAGGUGGAGGAGCAGGCCAUCGAGGU CUGGCAGGGCCUGUCCCUCCUGAGCGAGGCCAUCCUGCAGGCGCAGGCCCUCCUGGCCAACUCCAGCCA GCCCCCGGAGACACUGCAGCUCCACAUCGACAAGGCCAUCUCCGGGCUGCGGAGCCUGACCUCCCUCCU GCGCGUGCUGGGCGCGCAGAAGGAGCUCAUGAGCCCGCCCGACACGACCCCCCCGGCCCCGCUGCGGACC CUGACCGUGGACACGUUCUGCAAGCUCUUCCGCGUCUACGCCAACUUCCUGCGGGGCAAGCUGAAGCUC UACACCGGGGAGGUGUGCCGCCGGGGCGACCGCUGACCACUAGUGCAUCACAUUUAAAAGCAUCUCAG CCUACCAUGAGAAUAAGAGAAAGAAAAUGAAGAUCAAUAGCUUAUUCAUCUCUUUUUCUUUUUCGUU GGUGUAAAGCCAACACCCUGUCUAAAAAACAUAAAUUUCUUUAAUCAUUUUGCCUCUUUUCUCUGUG CUUCAAUUAAUAAAAAAUGGAAAGAACCUAGAUCUAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAUGCAUCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAA AGGCUCUUUUCAGAGCCACCAGAAUU Preparation of DNA and mRNA constructs:
The DNA sequence encoding Gaussia princeps luciferase was prepared by modifying the wild type encoding DNA sequence by introducing a GC-optimized sequence for stabilization. Sequences were introduced into a derived pUC19 vector and modified to comprise stabilizing UTR sequences derived from 32L4-5'-UTR ribosomal 5'TOP UTR (32L4) and 3'UTR derived from albumin 7, a histone stem-loop sequence, a stretch of 64x adenosine at the 3'-terminal end (poly- A-tail) and a stretch of 30x cytosine at the 3'- terminal end (poly-C-tail) were introduced 3' of the coding sequence. The sequence contains following sequence elements: the coding sequence encoding Gaussia luciferase; stabilizing sequences derived from 32L4-5'-UTR ribosomal 5'TOP UTR (32L4); 64x adenosine at the 3'-terminal end (poly- A- tail); 5 nucleotides, 30 x cytosine at the 3'- terminal end (poly-C-tail) and 5 additional nucleotides.
R2851 as mentioned in the present context resembles a GC-enriched mRNA sequence encoding for a Gaussia princeps luciferase having a poly (A) -sequence with 64 adenylates, followed by 5 nucleotides, followed by a poly(C)-sequence with 30 cytidylates and a histone stem- loop sequence followed by another 5 nucleotides.
RNA in vitro transcription:
The respective DNA plasmids were enzymatically linearized and transcribed in vitro using DNA dependent T7 RNA polymerase in the presence of a nucleotide mixture under respective buffer conditions. GpLuc mRNA (SEQ ID NO: 14) and MmEpo mRNA (SEQ ID NO: 16) were co- transcriptionally capped by adding a cap analog (m7GpppG) to the nucleotide mixture.
Post-transcriptional capping and polyadenylation:
PpLuc in vitro transcribed mRNA (SEQ ID NO: 15) was enzymatically capped using a Script- Cap capping system (Cellscript). PpLuc mRNA (SEQ ID NO: 15) and MmEpo mRNA (SEQ ID NO: 16) were enzymatically polyadenylated using a commercial polyadenylation kit.
Purification of mRNA constructs:
Subsequently, the obtained mRNA constructs were purified using PureMessenger®
(CureVac, Tubingen, Germany; WO 2008/077592 Al) and used for the further experiments. For the in vitro and in vivo experiments described below, the following polymers, lipids and transfection agents were used:
Polymers:
- Polyethylene glycol/peptide polymers (PB83)
- JetPEI
Permanently cationic lipids: - l,2-dioleoyl-3-trimethylammonium-propane ("DOTAP")
- N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTMA")
- ((6Z,9Z, 28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl-4-(trimethylamino)butanoate ("DLin-MC3-TMA" or "MC3-cationised" or "MC3-cat")
- l-(2-octylcyclopropyl)heptyldec-8-yl-4-(trimethylammonium)butanoate ("C9-C17-C3- cat") - l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3-pyrrolidiniumcarboxylate ("C9-C17- P-cat")
- Lipofectamin® ("LPFA"; comprising DOSPA and DOPE) Cationisable or permanently cationic lipidoids: - 3-C12 (see formula below)
- 3-C12-OH (see formula below)
-3-C12-OH cat (see formula below)
Synthesis and purification of disulfide-linked polyethylene glycol/peptide polymer conjugates
An amount of 20 mg peptide (CHHHHHHRRRRHHHHHHC-NH2) TFA salt was dissolved in 2 mL borate buffer pH 8.5 and stirred at room temperature for approximately 18 hours. Then, 12.6 mg PEG-SH 5000 (Sunbright) dissolved in N-methylpyrrolidone was added to the peptide solution and filled up to 3 mL with borate buffer pH 8.5. After 18 hours incubation at room temperature, the reaction mixture was purified and concentrated by centricon procedure (MWCO lOkDa), washed against water and lyophilized. After lyophilisation, the lyophilisate was dissolved in ELGA water and the concentration was adjusted to 10 mg/mL. The obtained polyethylene glycol/peptide polymers (HO-PEG 5000-S-(S-CHHHHHHRRRRHHHHHHC-S-)7-S-PEG 5000-OH) were used for further formulation experiments, and are hereinafter referred to as PB83.
Preparation of lipidoids
The lipidoids
Figure imgf000148_0001
3-C12 and
Figure imgf000149_0001
(formula IVa)
3-C12-OH were prepared. Lipidoid 3-C12 may be obtained by acylation of tris(2-aminoethyl)amine with an activated lauric (C12) acid derivative, followed by reduction of the amide. Alternatively, it may be prepared by reductive amination with the corresponding aldehyde. Lipidoid 3-C12-OH is prepared by addition of the terminal C12 alkyl epoxide with the same oligoamine according to Love et al., pp. 1864-1869, PNAS, vol. 107 (2010), no. 5 (cf. compound C12-110).
The lipidoid 3-C12-OH-cat comprising the cation according to formula IV:
Figure imgf000149_0002
(formula IV) was prepared from 3-C12-OH by reaction with activated methyl group such as methyl iodide.
Formulation of polymer-lipid(oid) complexed mRNA
This example describes the preparation of nanoparticles of polymer-lipid or polymer- lipidoid complexed mRNA, which were subsequently used for further in vitro and in vivo experiments. The respective mRNA was prepared as described above. Polyethylene
glycol/peptide polymers (PB83) were prepared as described above.
First, ringer lactate buffer (RiLa; alternatively e.g. saline (NaCl) or PBS buffer may be used), respective amounts of lipid(oid), and respective amounts of a polymer (PB83) were mixed to prepare compositions comprising a lipid(oid) and a peptide or polymer. Then, the carrier compositions were used to assemble nanoparticles with the mRNA by mixing the mRNA with respective amounts of polymer-lipid carrier and allowing an incubation period of 10 minutes at room temperature such as to enable the formation of a complex between the lipid, polymer and mRNA. The nanoparticles were then used for further in in vitro and in vivo experiments. Relevant parameters in that context are the amount and kind of lipid, the amount and kind of polymer, and the N/P ratio. Using the above described procedure with several different polymers, lipids and cargo mRNAs, a wide variety of polymer-lipid mRNA complexes could be formulated (see Table 2 for their detailed characteristics).
Table 2: Characteristics of the polymer-lipid(oid)-mRNA complexes
Carrier description Polymer Polymer/ N/P Lipid Lipid/RNA RNA
RNA ratio ratio ratio SEQ (w/w) (nmol^g) ID NO
PB83 N/P 0.7, 0.4 DOTAP PB83 2 0.7 DOTAP 0.4 14
PB83 N/P 1.4, 0.14 DOTAP PB83 4 1.4 DOTAP 0.14 14
PB83 N/P 1.4, 1.4 DOTAP PB83 4 1.4 DOTAP 1.4 14
PB83 N/P 1.4, 1.4 DOTAP PB83 4 1.4 DOTAP 1.4 15
PB83 N/P 1.4, 4.2 DOTAP PB83 4 1.4 DOTAP 4.2 14
PB83 N/P 1.4, 7 DOTAP PB83 4 1.4 DOTAP 7 14
PB83 N/P 1.4, 14 DOTAP PB83 4 1.4 DOTAP 14 14
PB83 N/P 2.8, 0.14 DOTAP PB83 8 2.8 DOTAP 0.14 14
PB83 N/P 2.8, 1.4 DOTAP PB83 8 2.8 DOTAP 1.4 14
PB83 N/P 2.8, 1.4 DOTAP PB83 8 2.8 DOTAP 1.4 15
PB83 N/P 2.8, 4.2 DOTAP PB83 8 2.8 DOTAP 4.2 14
PB83 N/P 2.8, 7 DOTAP PB83 8 2.8 DOTAP 7 14
PB83 N/P 2.8, 14 DOTAP PB83 8 2.8 DOTAP 14 14
PB83 N/P 6, 0.4 DOTAP PB83 17 6 DOTAP 0.4 14
PB83 N/P 4.15, 1.4 DOTAP PB83 12 4.15 DOTAP 1.4 14
PB83 N/P 4.15, 1.4 DOTAP PB83 12 4.15 DOTAP 1.4 15
PB83 N/P 4.15, 7 DOTAP PB83 12 4.15 DOTAP 7 14
PB83 N/P 1.4, 0.05 DOTMA PB83 4 1.4 DOTMA 0.05 14
PB83 N/P 1.4, 0.25 DOTMA PB83 4 1.4 DOTMA 0.25 14
PB83 N/P 1.4, 0.5 DOTMA PB83 4 1.4 DOTMA 0.5 14
PB83 N/P 2.8, 0.05 DOTMA PB83 8 2.8 DOTMA 0.05 14 Carrier description Polymer Polymer/ N/P Lipid Lipid/RNA RNA
RNA ratio ratio ratio SEQ (w/w) (nmol^g) ID NO
PB83 N/P 2.8, 0.25 DOTMA PB83 8 2.8 DOTMA 0.25 14
PB83 N/P 2.8, 0.5 DOTMA PB83 8 2.8 DOTMA 0.5 14
PB83 N/P 4.15, 0.05 DOTMA PB83 12 4.15 DOTMA 0.05 14
PB83 N/P 4.15, 0.25 DOTMA PB83 12 4.15 DOTMA 0.25 14
PB83N/P 4.15, 0.5 DOTMA PB83 12 4.15 DOTMA 0.5 14
PB83N/P1.4,0.1MC3-cat PB83 4 1.4 MC3-cat 0.1 14
PB83N/P0.7,0.4MC3-cat PB83 2 0.7 MC3-cat 0.4 14
PB83N/P0.7,0.4MC3-cat PB83 2 0.7 MC3-cat 0.4 15
PB83N/P1.4,0.5MC3-cat PB83 4 1.4 MC3-cat 0.5 14
PB83 N/P 1.4, 1 MC3-cat PB83 4 1.4 MC3-cat 1 14
PB83N/P 2, 0.4MC3-cat PB83 6 2 MC3-cat 0.4 15
PB83N/P2.8,0.1MC3-cat PB83 8 2.8 MC3-cat 0.1 14
PB83N/P2.8,0.5MC3-cat PB83 8 2.8 MC3-cat 0.5 14
PB83N/P2.8, 1 MC3-cat PB83 8 2.8 MC3-cat 1 14
PB83N/P 4.15, 0.1 MC3-cat PB83 12 4.15 MC3-cat 0.1 14
PB83N/P 6, 0.4MC3-cat PB83 17 6 MC3-cat 0.4 14
PB83N/P 6, 0.4MC3-cat PB83 17 6 MC3-cat 0.4 15
PB83N/P 4.15, 0.4 MC3-cat PB83 12 4.15 MC3-cat 0.4 16
PB83N/P 4.15, 0.5 MC3-cat PB83 12 4.15 MC3-cat 0.5 14
PB83N/P 6.9,0.4MC3-cat PB83 20 6.9 MC3-cat 0.4 16
PB83N/P10,0.4MC3-cat PB83 28 10 MC3-cat 0.4 15
PB83N/P0.7,0.4LPFA PB83 2 0.7 LPFA 0.4 14
PB83N/P 6, 0.4LPFA PB83 17 6 LPFA 0.4 14
PB83 N/P 0.7, 0.053-C12 PB83 2 0.7 3-C12 0.05 14
PB83N/P0.7,0.13-C12 PB83 2 0.7 3-C12 0.1 14
PB83N/P0.7,0.43-C12 PB83 2 0.7 3-C12 0.4 14
PB83N/P0.7, 13-C12 PB83 2 0.7 3-C12 1 14 Carrier description Polymer Polymer/ N/P Lipid Lipid/RNA RNA
RNA ratio ratio ratio SEQ (w/w) (nmol^g) ID NO
PB83N/P2, 0.053-C12 PB83 6 2 3-C12 0.05 14
PB83N/P2, 0.13-C12 PB83 6 2 3-C12 0.1 14
PB83N/P2, 0.43-C12 PB83 6 2 3-C12 0.4 14
PB83N/P2, 13-C12 PB83 6 2 3-C12 1 14
PB83N/P6, 0.13-C12 PB83 17 6 3-C12 0.1 14
PB83N/P6, 0.43-C12 PB83 17 6 3-C12 0.4 14
PB83N/P6, 13-C12 PB83 17 6 3-C12 1 14
PB83 N/P 0.7, 0.053-C12-OH PB83 2 0.7 3-C12-OH 0.05 14
PB83N/P0.7,0.13-C12-OH PB83 2 0.7 3-C12-OH 0.1 14
PB83N/P0.7,0.43-C12-OH PB83 2 0.7 3-C12-OH 0.4 14
PB83N/P0.7,0.43-C12-OH PB83 2 0.7 3-C12-OH 0.4 15
PB83N/P0.7, 13-C12-OH PB83 2 0.7 3-C12-OH 1 14
PB83N/P2, 0.053-C12-OH PB83 6 2 3-C12-OH 0.05 14
PB83N/P2, 0.13-C12-OH PB83 6 2 3-C12-OH 0.1 14
PB83N/P2, 0.43-C12-OH PB83 6 2 3-C12-OH 0.4 14
PB83N/P2, 13-C12-OH PB83 6 2 3-C12-OH 1 14
PB83N/P6, 0.13-C12-OH PB83 17 6 3-C12-OH 0.1 14
PB83N/P6, 0.43-C12-OH PB83 17 6 3-C12-OH 0.4 14
PB83N/P6, 13-C12-OH PB83 17 6 3-C12-OH 1 14
PB83N/P6, 0.43-C12-OH-cat PB83 17 6 3-C12-OH-cat 0.4 14
PB83 N/P 0.7, 0.4 C9-C17-C3- PB83 17 0.7 C9-C17-C3-cat 0.4 14 cat
PB83 N/P 6, 0.4 C9-C17-C3-cat PB83 17 6 C9-C17-C3-cat 0.4 14
PB83 N/P 0.7, 0.4 C9-C17-P-cat PB83 2 0.7 C9-C17-P-cat 0.4 14
PB83 N/P 6, 0.4 C9-C17-P-cat PB83 17 6 C9-C17-P-cat 0.4 14
JetPEI N/P 6, 1.4 DOTAP JetPEI 17 6 DOTAP 1.4 14
JetPEI N/P 6, 7.2 DOTAP JetPEI 17 6 DOTAP 7.2 14
JetPEI N/P 6, 14.3 DOTAP JetPEI 17 6 DOTAP 14.3 14 In order to characterize the integrity of the obtained polymer-lipid(oid) complexed mRNA particles, RNA agarose gel shift assays were performed. In addition, size measurements were performed to evaluate whether the obtained nanoparticles have a uniform size profile.
For the RNA gel shift assay, a conventional RNA agarose gel was prepared and loaded with the respective polymer-lipid(oid) complexed mRNA particles. The gel bands were visualized using a bio imager. All tested polymer-lipid(oid) complexed mRNAs were analyzed and determined to be stable under the respective conditions (data not shown).
For determining the particle sizes, samples comprising polymer-lipid(oid) complexed mRNAs were diluted in ringer lactate (RiLa; alternatively saline (NaCl)) to a final volume of 50 μL· The size measurement was performed using a Zetasizer® device.
The results of the gel shift assay and the particle size analysis showed that the obtained polymer-lipid(oid)-mRNA complexes were stable and uniform over a broad range of polymer-to- lipid(oid) ratios.
Example 2: Effect of different polymer-lipid(oid) formulations on transfection efficiency of HepG2 cells in vitro
This example describes the evaluation of the effect of different polymer-lipid(oid) formulations (as described under Example■ 1) on transfection efficiency on HepG2 cells (human liver carcinoma cell line). As a read-out for transfection efficiency, GpLuc mRNA (SEQ ID NO: 14) was used as a cargo. Successful transfection with the cargo leads to the translation of the luciferase protein and to a secretion of Gp luciferase protein into the cell culture supernatant.
Transfection of HepG2 cells:
A volume of 0.2 mL HepG2 cells (10.000 cells) were seeded in a 96 well tissue culture plate. After removing the medium from each well, 100 μί, RPMI 1640 medium (with 1 % Penicillin and 1 % Streptomycin, 1 % L-Glutamin) was added to each well. Afterwards, HepG2 cells were transfected with 100 μί, transfection mix (in triplicates) of the above described polymer- lipid(oid) mRNA complexes and respective controls (in triplicates), and cells were incubated at 37 °C and 5 % CO2 for 90 minutes. After incubation, 150 μί, medium was exchanged with 150 μί, fresh RPMI 1640 medium supplemented with 10 % fetal calf serum. Twenty-four hours post transfection, 10 μί, of supernatant of each well was extracted and used for further luminescence analysis (see below).
Luminescence detection and analysis:
A volume of 10 μί, supernatant was transferred to a 96 well plate for GpLuc measurement. Then, coelenterazine working solution (100 μΜ) was prepared (1 mL coelenterazine stock solution (4.72 mM in EtOH) in 49 mL phosphate buffered saline supplemented with 5 mM NaCl, pH 7.2). A volume of 100 μΐ. coelenterazine working solution was used as a substrate for GpLuc and measured after 5 seconds in a commercially available microplate reader.
Results: As shown in Figure 1A for high-charge polymer-lipid complexes (N/P 2; N/P 4; N/P 6 ), the combination of polymer PB83 and cationic lipid DOTAP led to a strong increase in transfection efficiency (up to 3 log units), compared to transfections with polymer alone. The expression levels are increasing with increasing N/P ratio.
As shown in Figure IB, for high-charge polymer-lipid complexes (N/P 2; N/P 4; N/P 6), the combination of polymer PB83 and cationic lipid MC3-cat and DOTMA led to a strong increase in transfection efficiency (up to 3 log units) compared to transfections with polymer alone. The effect was less pronounced when MVL5 lipid was used (PB83 N/P 6 0.1 MVL5).
It was further found that the addition of even small amounts of lipidoid (3-C12; 3-C12-OH, or 3-C12-OH-cat) to the cationic polymer-peptide conjugate (PB83) led to a profound increase of transfection efficiency, as can be seen in figures 1C and ID.
To summarize the above, the results show that the polymer-lipid carrier of the present invention is capable over a broad range to increase transfection efficiency in vitro. This effect was demonstrated for a variety of complexes and a wide range of N/P ratios, and for both low-charge ratio and high-charge ratio complexes. Example 3: Effect of different polymer-lipid(oid) formulations on transfection efficiency of differentiated Sol8 muscle cells in vitro
The purpose of this example is to assess the effect of lipid additives such as permanently cationic lipids or lipidoids on transfection / expression efficiency in Sol8 cells. It shows that the positive effect of the inventive polymer-lipid(oid) formulations (as described under Example 1) on transfection efficiency observed in Example 2 also translates to other cell types such as Sol8 muscle cells. Sol8 is a myogenic cell line isolated by Daubas et al. from primary cultures of soleus muscle taken from the leg of a normal C3H mouse. As a read-out for transfection efficiency, GpLuc mRNA (SEQ ID NO: 14) was used as the cargo. A successful transfection with the cargo leads to the translation of the luciferase protein and to a secretion of Gp luciferase protein into the cell culture supernatant. For further details on the GpLuc mRNA cargo see Example 1.
Transfection of Sol8 cells:
For a subset on hydrogel, a volume of 0.2 mL Sol8 cells (20.000 cells) were seeded in 96 well glass bottom plates (Softwell Hydrogel coated with collagen (elasticity E = 12 kPa)) on day 1. For another subset without hydrogel, a volume of 0.2 mL with 10.000 Sol8 cells per well were seeded in a 96 well glass bottom plates.
After removing the medium from each well, 100 DMEM medium (with 2 % horse serum) was added to each well. Afterwards, on day 2, Sol8 cells were transfected with 100 transfection mix of polymer-lipid(oid) complexes (in triplicates without serum) and respective controls (also in triplicates), and cells were incubated at 37 °C and 5 % CO2 for 120 minutes (non-hydrogel subset) or respectively 90 minutes (hydrogel subset). After incubation, 150 μί, medium was exchanged with 150 μί, fresh DMEM medium supplemented with 10 % fetal calf serum.
Twenty-four hours post transfection, i.e. on day 3, 10 μί, of supernatant of each well was extracted and used for quantification of protein expression (measurement of GpLuc) via luminescence analysis as described above.
Results:
As shown in figure 2A (hydrogel subset) and figure 2B, 2C, 2D and 2E (non-hydrogel subset), the increase of transfection efficiency that has been observed in HepG2 cells was also shown for the in vitro transfection of S0I8 muscle cells, both for low-charge ratio complexes and high-charge ratio complexes and a broad range of permanently cationic lipids and lipidoids. Compared to low-charge ratio complexes, high-charge ratio complexes require an even smaller amount of lipid to achieve a marked increase of the expression level. In the case of DOTAP, very small amount of lipid were effective in combination with low- and high-charge ratio complexes to increase expression levels in S0I8 cells. It was further found that the addition of even small amounts of lipidoids (namely 3-C12 and 3-C12-OH) to a cationic polymer-peptide conjugate (PB83) leads to a profound increase of transfection efficiency. Example 4: In vitro cytokine stimulation in human PBMCs
In this example, the intrinsic stimulation of the immune system evoked by the nanoparticles of the invention was evaluated. To assess the impact of the inventive formulation on immune stimulation, the release of cytokines interferon alpha (IFNa) and tumor necrosis factor alpha (TNFa) in human peripheral blood mononuclear cells (hPBMCs) after treatment with different polymer-lipid(oid) complexed GpLuc mRNA (SEQ ID NO: 14) formulations was measured.
Preparation and stimulation of hPBMCs:
Human peripheral blood mononuclear cells (hPBMCs) from peripheral blood of healthy donors were isolated using a Ficoll gradient and washed subsequently with 1 x PBS (phosphate- buffered saline). Isolated cells were seeded on 96 well microtiter plates (2 x 105 cells/well). The hPBMCs were incubated for 24 h with 10 μί, of the respective polymer-lipid(oid) complexed mRNA particles or certain controls (e.g. 'naked' mRNA, Ringer Lactate buffer (RiLa), CpG2216, RNAdjuvant®) in X-VIVO 15 Medium (Lonza) in triplicates. The immunostimulatory effect upon hPBMC stimulation was measured by detecting the cytokine production using specific antibodies detecting human TNFa and human IFNa.
Cytokine ELISA:
ELISA microtiter plates (Nunc Maxisorb) were incubated overnight (o/n) with binding buffer (0.02 % NaN3, 15 mM Na2C03, 15 mM NaHC03, pH 9.7), additionally containing a specific cytokine antibody. Cells were then blocked with 1 x PBS, containing 1 % BSA (bovine serum albumin). The cell supernatant was added and incubated for 4 h at 37 °C. Subsequently, the microtiter plate was washed with 1 x PBS, containing 0.05 % Tween-20 and then incubated with a Biotin-labelled secondary antibody (BD Pharmingen, Heidelberg, Germany). Streptavidin- coupled horse radish peroxidase was added to the plate. Then, the plate was again washed with 1 x PBS, containing 0.05 % Tween-20, and ABTS (2,2'-azino-bis(3-ethyl-benzthiazoline-6-sulfonic acid) was added as a substrate. The amount of cytokine was determined by measuring the absorption at 405 nm (OD 405) using a standard curve with recombinant cytokines (BD
Pharmingen, Heidelberg, Germany) with the Sunrise ELISA-Reader from Tecan (Crailsheim, Germany). In parallel, the GpLuc concentration in the cell culture supernatant was also determined. Results:
In result, the tested compositions did not stimulate the secretion of cytokines in human PBMCs as can be seen in figures 3A (for TNFa) and 3B (for IFNa), indicating that the nanoparticles of the invention have no or only a very minor intrinsic potential to stimulate the immune system in the absence of a potent positive control such as CpG oligodeoxynucleotide.
Example 5: Scanning Laser Ophthalmoscopy (SLO) analysis of rat eyes 24 h after subretinal injection of luciferase-mRNA mRNA complexes were prepared by mixing PpLuc mRNA (SEQ ID NO: 15) and cationic polymer - lipid or lipidoid solutions (e.g. MC3-cat or 3-C12-OH) at different charge ratios. Animals treated with Ringer's buffer served as controls. Lyophilized formulations were rehydrated with Ringer's buffer. The final mRNA concentration was 2.5 μg/μL. 2 doses of 2 were injected into each eye. 4 eyes (2 rats) were treated per group. Twentyfour hours after the treatment, noninvasive Scanning Laser Ophthalmoscopy (SLO) was used to image the retina (data not shown).
Luciferin solution was injected into the tail vein of the rats and the eyes were analyzed after an incubation time of 2 minutes for at least 10 minutes. During this time the rats additionally received a luciferin solution to sniff. For a detailed analysis, the animals were then sacrificed, their eyes removed and frozen. Subsequently, eye samples were analyzed for the levels of transfection. To that end the eyes were mechanically disrupted in a TissueLyser and lysed.
Luciferase activity of each sample was assayed in a luminometer.
The results of the subretinal injection of luciferase-mRNA are shown in figure 4, expressed as relative light units (RLU).
Example 6: Induction of a humoral and cellular immune response after intramuscular vaccination of mice
Preparation of DNA and mRNA constructs
For the present example, a DNA sequence encoding the hemagglutinin (HA) protein of influenza A virus (A/Netherlands/602/2009(HlNl)) was prepared and used for subsequent in vitro transcription reactions. The respective mRNA sequence as well as further details on the vaccination regimen are provided below. G/C-enriched mRNA sequence R2564 coding for the hemagglutinin (HA) protein of influenza A virus (A/Netherlands/602/2009(HlNl)) (SEQ ID NO: 21):
GGGGCGCUGCCUACGGAGGUGGCAGCCAUCUCCUUCUCGGCAUCAAGCUUACCAUGAAGGCCAU CCUGGUGGUCCUCCUGUACACCUUCGCCACCGCGAACGCCGACACGCUGUGCAUCGGCUACCACGCCAA CAACAGCACCGACACCGUGGACACCGUGCUCGAGAAGAACGUCACGGUGACCCACUCCGUGAACCUGCU GGAGGACAAGCACAACGGGAAGCUCUGCAAGCUGCGGGGCGUCGCCCCGCUGCACCUCGGGAAGUGCAA CAUCGCCGGCUGGAUCCUGGGGAACCCGGAGUGCGAGAGCCUGUCCACCGCGAGCUCCUGGAGCUACAU CGUGGAGACCUCCAGCUCCGACAACGGCACGUGCUACCCCGGCGACUUCAUCGACUACGAGGAGCUCCG CGAGCAGCUGAGCUCCGUGAGCUCCUUCGAGCGGUUCGAGAUCUUCCCCAAGACCAGCUCCUGGCCCAA CCACGACAGCAACAAGGGGGUCACCGCCGCCUGCCCGCACGCCGGCGCGAAGUCCUUCUACAAGAACCU GAUCUGGCUCGUGAAGAAGGGGAACAGCUACCCCAAGCUGUCCAAGAGCUACAUCAACGACAAGGGCA AGGAGGUGCUGGUCCUCUGGGGGAUCCACCACCCCAGCACCUCCGCCGACCAGCAGAGCCUGUACCAGA ACGCCGACGCCUACGUGUUCGUGGGCUCCAGCCGCUACUCCAAGAAGUUCAAGCCCGAGAUCGCCAUCC GGCCGAAGGUCCGCGACCAGGAGGGCCGGAUGAACUACUACUGGACGCUGGUGGAGCCCGGGGACAAGA UCACCUUCGAGGCGACCGGCAACCUCGUGGUCCCCCGCUACGCCUUCGCCAUGGAGCGGAACGCCGGGA GCGGCAUCAUCAUCUCCGACACCCCCGUGCACGACUGCAACACGACCUGCCAGACCCCGAAGGGCGCCA UCAACACCAGCCUGCCCUUCCAGAACAUCCACCCCAUCACGAUCGGGAAGUGCCCCAAGUACGUGAAGU CCACCAAGCUGCGCCUCGCGACCGGCCUGCGGAACGUCCCGAGCAUCCAGUCCCGCGGGCUGUUCGGCG CCAUCGCCGGGUUCAUCGAGGGCGGCUGGACCGGGAUGGUGGACGGCUGGUACGGGUACCACCACCAGA ACGAGCAGGGCAGCGGGUACGCCGCCGACCUCAAGUCCACGCAGAACGCGAUCGACGAGAUCACCAACA AGGUGAACAGCGUCAUCGAGAAGAUGAACACCCAGUUCACCGCCGUGGGCAAGGAGUUCAACCACCUG GAGAAGCGGAUCGAGAACCUGAACAAGAAGGUCGACGACGGCUUCCUCGACAUCUGGACGUACAACGC CGAGCUGCUGGUGCUCCUGGAGAACGAGCGCACCCUGGACUACCACGACUCCAACGUGAAGAACCUCUA CGAGAAGGUCCGGAGCCAGCUGAAGAACAACGCCAAGGAGAUCGGGAACGGCUGCUUCGAGUUCUACC ACAAGUGCGACAACACCUGCAUGGAGUCCGUGAAGAACGGGACCUACGACUACCCCAAGUACAGCGAG GAGGCCAAGCUGAACCGCGAGGAGAUCGACGGCGUGAAGCUCGAGUCCACGCGGAUCUACCAGAUCCUG GCGAUCUACAGCACCGUCGCCAGCUCCCUGGUGCUCGUGGUCAGCCUGGGGGCCAUCIJCCUUCUGGAUG UGCAGCAACGGCUCCCUGCAGUGCCGCAUCUGCAUCUGACCACUAGUGCAUCACAUUUAAAAGCAUCU CAGCCUACCAUGAGAAUAAGAGAAAGAAAAUGAAGAUCAAUAGCUUAUUCAUCUCUUUUUCUUUUUC GUUGGUGUAAAGCCAACACCCUGUCUAAAAAACAUAAAUUUCUUUAAUCAUUUUGCCUCUUUUCUCU GUGCUUCAAUUAAUAAAAAAUGGAAAGAACCUAGAUCUAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAUGCAUCCCCCCCCCCCCCCCCCCCCCCCCCCCCC CCAAAGGCUCUUUUCAGAGCCACCAGAAUU
According to a first preparation, the DNA sequence coding for the above mentioned mRNA was prepared. The construct R2564 (SEQ ID NO: 17) was prepared by introducing a 5'-TOP-UTR derived from the ribosomal protein 32L, modifying the wild type coding sequence by introducing a GC-optimized sequence for stabilization, followed by a stabilizing sequence derived from the albumin-3'-UTR, a stretch of 64 adenosines (poly(A)-sequence), a stretch of 30 cytosines
(poly(C)-sequence), and a histone stem loop. Preparation of the vaccine
The 'naked' mRNA R2564 was administered in Ringer's Lactate solution (RiLa). The co- formulation of naked mRNA R2564 with PB83 and 3-C12-OH was generated by mixing all components directly before administration.
Immunization
Balb/c mice (n=8 per group) were vaccinated intramuscularly (left M. tibialis) on day 0 and boosted on day 25, either with 10 μg HA-mRNA (R2564, SEQ ID NO: 21, 'naked' HA-mRNA) alone or with 10 μg HA-mRNA co-formulated with PB83 N/P 0.7, 0.4 3-C12-OH; see table 3 below. Therein, the indicated amount in μg refers to the mass of the nucleic acid molecule per se.
Table 3: Experimental setup
Figure imgf000159_0001
All animals received boost injections on day 25. Induction of functional humoral responses was analysed on day 40 by collecting blood samples and determining the serum hemagglutination inhibition (HI) antibody titer (see table 2 below), which is generally used as a surrogate marker of immune protection against influenza virus infection. A HI titer of 1:40 or greater is typically considered to confer protection. Ringer lactate-buffer (RiLa) treated mice served as negative controls. Table 4: Vaccination schedule
Figure imgf000160_0001
Hemagglutination inhibition assay
For hemagglutination inhibition (HI) assay mouse sera were heat inactivated (56 °C, 30 min), incubated with kaolin, and pre-adsorbed to chicken red blood cells (CRBC) (both Labor Dr. Merck & Ko liege n, Ochsenhausen, Germany). For the HI assay, 50 of 2-fold dilutions of pre- treated sera were incubated for 45 minutes with 4 hemagglutination units (HAU) of inactivated A/California/5 7/2009 (NIBSC, Potters Bar, UK) and 50 μΐ 0.5 % CRBC were added.
Results: As can be seen in figure 5, all mice vaccinated with the PB83 N/P 0.7, 0.4 3-C12-OH- formulation developed Hl-titers≥1:40. Figure 5 further shows that the intramuscular vaccination with a formulation comprising HA-mRNA (R2564) and the polymer-lipidoid carrier based on PB83 and 3-C12-OH induces higher antibody titers against the HA protein compared to vaccination with the HA-mRNA (R2564) alone.
Example 7: Effect of different polymer-lipid formulations on transfection efficiency in vivo
In this example, the effect of exemplary polymer-lipid compositions on transfection efficiency in vivo was evaluated. For this purpose, mice were injected intravenously with the polymer-lipid complexed PpLuc mRNA (SEQ ID NO: 15) and PpLuc expression (intracellular enzyme location) was detected and quantified in organ lysates 5-6 hours after the application via luminometric measurement.
Injection to mice:
BALB/c mice were injected intravenously with 100 of the respective transfection solution containing the inventive polymer-lipid complexed PpLuc mRNA, LNP formulated PpLuc mRNA and RiLa control (see Table 5). Five to six hours after injection, kidney, pancreas, liver, heart, lung and spleen were isolated and flash frozen in liquid nitrogen and stored at -80 °C for later expression analysis.
Table 5: Injection regimen for indicated mice groups
Figure imgf000161_0001
Tissue lysis: Frozen tissue samples (kidney, pancreas, liver, heart, lung, and spleen) were lysed in a tissue lyser (QIAGEN) at full speed for 3 minutes. A volume of 800 μΐ., of lysis buffer (25 mM Tris- HC1, 2 mM EDTA, 10 % (V/V) glycerol, 1 % (V/V) Triton X-100, 2 mM DTT, 1 mM
phenylmethylsulfonyl fluoride) was added to the sample and lysed for additional 6 minutes at full speed. Following that, samples were centrifuged at 13.500 rpm at 4 °C for 10 minutes. The respective supernatants were collected and stored at -80 °C.
Luciferase measurement:
A volume of 20 μΐ., of obtained tissue lysates were transferred to white LIA plates (Greiner) on ice. After that, 50 μΐ., of Beetle-Juice buffer (PJK GmbH) containing D-luciferin and ATP was added. After 2 seconds, luminescence data was recorded in a Chameleon™ plate reader (Hidex). Results:
As shown for polymer-lipid complexed PpLuc mRNA (SEQ ID NO: 15) in figures 6A and 6B, the positive effect on transfection efficiency also translates to the in vivo situation. Moreover the PpLuc protein expression outperformed the expression observed for transfection by the polymer alone (data not shown). Example 7: Effect of further polymer-lipid formulations on transfection efficiency in vivo
This example describes an immune response analysis in female Balb/c mice upon vaccination with different RNA formulations, i.e. an induction of a humoral and cellular immune response after intramuscular vaccination. Preparation of DNA and mRNA constructs
For the present example, a DNA sequence encoding the hemagglutinin [HA) protein of influenza A virus (A/Netherlands/602/2009(HlNl)) was prepared and used for subsequent in vitro transcription reactions. The respective mRNA sequence as well as further details on the vaccination regimen are provided below.
G/C-enriched mRNA sequence R2564 coding for the hemagglutinin (HA) protein of influenza A virus (A/Netherlands/602/2009(HlNl)) (SEQ ID NO: 21):
GGGGCGCUGCCUACGGAGGUGGCAGCCAUCUCCUUCUCGGCAUCAAGCUUACCAUGAAGGCCAU CCUGGUGGUCCUCCUGUACACCUUCGCCACCGCGAACGCCGACACGCUGUGCAUCGGCUACCACGCCAA CAACAGCACCGACACCGUGGACACCGUGCUCGAGAAGAACGUCACGGUGACCCACUCCGUGAACCUGCU GGAGGACAAGCACAACGGGAAGCUCUGCAAGCUGCGGGGCGUCGCCCCGCUGCACCUCGGGAAGUGCAA CAUCGCCGGCUGGAUCCUGGGGAACCCGGAGUGCGAGAGCCUGUCCACCGCGAGCUCCUGGAGCUACAU CGUGGAGACCUCCAGCUCCGACAACGGCACGUGCUACCCCGGCGACUUCAUCGACUACGAGGAGCUCCG CGAGCAGCUGAGCUCCGUGAGCUCCUUCGAGCGGUUCGAGAUCUUCCCCAAGACCAGCUCCUGGCCCAA CCACGACAGCAACAAGGGGGUCACCGCCGCCUGCCCGCACGCCGGCGCGAAGUCCUUCUACAAGAACCU GAUCUGGCUCGUGAAGAAGGGGAACAGCUACCCCAAGCUGUCCAAGAGCUACAUCAACGACAAGGGCA AGGAGGUGCUGGUCCUCUGGGGGAUCCACCACCCCAGCACCUCCGCCGACCAGCAGAGCCUGUACCAGA ACGCCGACGCCUACGUGUUCGUGGGCUCCAGCCGCUACUCCAAGAAGUUCAAGCCCGAGAUCGCCAUCC GGCCGAAGGUCCGCGACCAGGAGGGCCGGAUGAACUACUACUGGACGCUGGUGGAGCCCGGGGACAAGA UCACCUUCGAGGCGACCGGCAACCUCGUGGUCCCCCGCUACGCCUUCGCCAUGGAGCGGAACGCCGGGA GCGGCAUCAUCAUCUCCGACACCCCCGUGCACGACUGCAACACGACCUGCCAGACCCCGAAGGGCGCCA UCAACACCAGCCUGCCCUUCCAGAACAUCCACCCCAUCACGAUCGGGAAGUGCCCCAAGUACGUGAAGU CCACCAAGCUGCGCCUCGCGACCGGCCUGCGGAACGUCCCGAGCAUCCAGUCCCGCGGGCUGUUCGGCG CCAUCGCCGGGUUCAUCGAGGGCGGCUGGACCGGGAUGGUGGACGGCUGGUACGGGUACCACCACCAGA ACGAGCAGGGCAGCGGGUACGCCGCCGACCUCAAGUCCACGCAGAACGCGAUCGACGAGAUCACCAACA AGGUGAACAGCGUCAUCGAGAAGAUGAACACCCAGUUCACCGCCGUGGGCAAGGAGUUCAACCACCUG GAGAAGCGGAUCGAGAACCUGAACAAGAAGGUCGACGACGGCUUCCUCGACAUCUGGACGUACAACGC CGAGCUGCUGGUGCUCCUGGAGAACGAGCGCACCCUGGACUACCACGACUCCAACGUGAAGAACCUCUA CGAGAAGGUCCGGAGCCAGCUGAAGAACAACGCCAAGGAGAUCGGGAACGGCUGCUUCGAGUUCUACC ACAAGUGCGACAACACCUGCAUGGAGUCCGUGAAGAACGGGACCUACGACUACCCCAAGUACAGCGAG GAGGCCAAGCUGAACCGCGAGGAGAUCGACGGCGUGAAGCUCGAGUCCACGCGGAUCUACCAGAUCCUG GCGAUCUACAGCACCGUCGCCAGCUCCCUGGUGCUCGUGGUCAGCCUGGGGGCCAUCUCCUUCUGGAUG UGCAGCAACGGCUCCCUGCAGUGCCGCAUCUGCAUCUGACCACUAGUGCAUCACAUUUAAAAGCAUCU CAGCCUACCAUGAGAAUAAGAGAAAGAAAAUGAAGAUCAAUAGCUUAUUCAUCUCUUUUUCUUUUUC GUUGGUGUAAAGCCAACACCCUGUCUAAAAAACAUAAAUUUCUUUAAUCAUUUUGCCUCUUUUCUCU
RECTIFIED SHEET RULE 91 ISA EP GUGCUUCAAUUAAUAAAAAAUGGAAAGAACCUAGAUCUAAAAAAAAAAAAAAAAAAAAAAAAAAAAA
AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAUGCAUCCCCCCCCCCCCCCCCCCCCCCCCCCCCC
CCAAAGGCUCUUUUCAGAGCCACCAGAAUU
According to a first preparation, the DNA sequence coding for the above mentioned mRNA was prepared. The construct R2564 (SEQ ID NO:21) was prepared by introducing a 5'-TOP-UTR derived from the ribosomal protein 32L, modifying the wild type coding sequence by introducing a GC-optimized sequence for stabilization, followed by a stabilizing sequence derived from the albumin-3'-UTR, a stretch of 64 adenosines (poly(A)-sequence), a stretch of 30 cytosines (poly(C)-sequence), and a histone stem loop (A64-N5-C30-histone stem loop-N5).
Preparation of the vaccine
The 'naked' mRNA R2564 was administered in Ringer's Lactate solution (RiLa). The co- formulation of naked mRNA R2564 with 3-C12-OH was generated by mixing all components directly before administration.
Immunization
Balb/c mice (n=8 per group) were vaccinated intramuscularly (left M. tibialis) on day 0 and boosted on day 21, with 10 μg 'naked' HA-mRNA or with 10 μg HA-mRNA co-formulated with 0.4 3-C12-OH (HA-mRNA = R2564, SEQ ID NO:21); see table 6 below. Therein, the indicated amount in μg refers to the mass of the nucleic acid molecule per se.
Table 6: Experimental setup
Figure imgf000163_0001
All animals received boost injections on day 21; also on day 21 orbital bleeding was performed. The Experiment was terminated on day 35 (cardiac bleed and generation of serum samples, excision of spleens and isolation of splenocytes were performed) and the serum hemagglutination inhibition (HI) antibody titer (see table 7 below), which is generally used as a surrogate marker of immune protection against influenza virus infection were determined. A HI titer of 1:40 or greater is typically considered to confer protection. Ringer lactate-buffer (RiLa) treated mice served as negative controls.
Table 7: Vaccination schedule
Figure imgf000164_0001
Assays:
For hemagglutination inhibition (HI) assay mouse sera were heat inactivated (56 °C, 30 min), incubated with kaolin, and pre-adsorbed to chicken red blood cells (CRBC) (both Labor Dr. Merck & Ko liege n, Ochsenhausen, Germany). For the HI assay, 50 μΐ of 2-fold dilutions of pre- treated sera were incubated for 45 minutes with 4 hemagglutination units (HAU) of inactivated A/California/5 7/2009 (NIBSC, Potters Bar, UK) and 50 μΐ 0.5 % CRBC were added.
Detection of an antigen-specific immune response (B-cell immune response) was carried out by detecting influenza specific IgGl and IgG2a antibodies by performing ELISA assays.
Therefore, blood samples were taken from the vaccinated mice 21 days post prime and 14 days post boost and sera were prepared. MaxiSorb plates (Nalgene Nunc International) were coated with the inactivated virus. After blocking with lxPBS containing 0.05% Tween-20 and 1% BSA the plates were incubated with diluted mouse serum (as indicated). Subsequently a biotin- coupled secondary antibody (anti-mouse-IgGl and IgG2a, Pharmingen) was added. After washing, the plate was incubated with horseradish peroxidase-streptavidin and subsequently the conversion of the ABTS substrate (2,2'-azino-bis(3-ethyl-benzthiazoline-6-sulfonic acid) was measured.
Induction of antigen-specific T cells was determined 14 days after boost using intracellular cytokine staining (ICS). Splenocytes from vaccinated and control mice were isolated and stimulated with an HA peptide library (PepMix™ Influenza A (HA /California (H1N1)), JPT) and anti-CD28 antibody (BD Biosciences) for 6 hours at 37°C in the presence of the GolgiPlug containing protein transport inhibitor Brefeldin A (BD Biosciences). After stimulation, cells were washed and stained for intracellular cytokines using the Cytofix/Cytoperm reagent (BD
Biosciences) according to the manufacturer's instructions. The following antibodies were used for staining: CD8-APC H7 (1: 100), CD4-BD Horizon V450 (1:200) (BD Biosciences), Thyl.2-F.TC (1:300), TNFa-PE (1: 100), IFN-y-APC (1: 100) (eBioscience), and incubated with FcyR-block diluted 1 : 100. Aqua Dye was used to distinguish live/dead cells (Invitrogen). Cells were acquired using a Canto II flow cytometer (BD Biosciences) and flow cytometry data were analyzed using Flowjo software (Tree Star).
Results hemagglutination inhibition (HI) assay:
As can be seen in figure 7-A/B, all mice vaccinated with the mRNA 3-C12-OH-formulation developed HI-titers≥1:40 after boost vaccination. Figure 7-A/B further shows that the intramuscular vaccination with a formulation comprising HA-mRNA (R2564) and 3-C12-OH after prime and boost vaccination induces significantly higher antibody titers against the HA protein compared to vaccination with the HA-mRNA (R2564) alone.
Results ELISA assays: Formulated HA-mRNA with lipid 3-C12-OH induced significantly higher functional antibody titers, both IgGl and IgG2a subtypes, when compared to mRNA without lipid already after a single i.m. injection as apparent from figures 8-A-D 21 days after prime vaccination and 14 days after boost vaccination.
Results T cell assays: As apparent from figure 9, it was observed that 10 μg HA-mRNA, formulated with lipid 3-
C12-OH, led to significant increased IFNy production compared to non-lipid-formulated mRNA. The medium control as expected never generated a response
Example 8: Effect of different polymer-lipid formulations on transfection efficiency on Hep G2 cells
This example describes the evaluation of the effect of different lipid formulations on transfection efficiency on Hep G2 cells. As a read-out for transfection efficiency, Gaussia princeps luciferase GpLuc mRNA was used as a cargo. Successful transfection with the cargo leads to the translation of the luciferase protein and to a secretion of luciferase protein into the cell culture supernatant.
Accordingly, Hep G2 cells were seeded in 24-well-plates at a density of 75.000 cells per well in cell culture medium (RPMI 1640 w/ 25mM Hepes 500ml, 10% FCS, 1% L-Glutamine, 1%
Penicillin/Streptomycin; Lonza Group AG BE12-115F / 6MB205; Basel/Switzerland). Hep G2 cells were transfected in duplicates as described below with different carrier-lipid formulations and with mRNA encoding GpLuc (SEQ ID NO:14; R2851). As a negative control, naked mRNA encoding GpLuc without lipid was used. Luciferase expression was quantified after 24 h.
Results: Figure 10 shows that GpLuc protein was expressed in A549 cells transfected with the mRNA construct R2851 and that the tested formulations with added lipids were highly efficient when compared to the control w/o added lipids. This shows that the combination of mRNA with very small amounts of lipid was able to increase the transfection efficiency.
Example 9: This example describes the evaluation of the effect of different polymer - lipid formulations on transfection efficiency on A549 cells (human lung carcinoma cell line). As a read-out for transfection efficiency, Gaussia princeps luciferase GpLuc mRNA was used as a cargo. Successful transfection with the cargo leads to the translation of the luciferase protein and to a secretion of luciferase protein into the cell culture supernatant. Accordingly, A549 cells were seeded in 24-well-plates at a density of 75.000 cells per well in cell culture medium (Gibco (Thermo Fisher) Ham's F-12K (Kaighn's) Medium, 10% Fetal Bovine Serum (FBS), 1% L-Glutamine, 1 % Penicillin/Streptomycin). A549 cells were transfected in duplicates as described below with different carrier-lipid formulations and with mRNA encoding GpLuc (SEQ ID NO:14; R2851). As a negative control, mRNA encoding GpLuc without CVCM/PB83 carrier was used. Luciferase expression was quantified after 24 h.
In this working example, the cationic lipid DDAB (dimethyldioctadecylammonium ; CAS Number 3700-67-2; Avanti Polar Lipids, Alabaster, USA) was used:
Figure imgf000166_0001
Table 8: Transfection conditions
Figure imgf000167_0001
Results:
Figure 11 shows that GpLuc protein was expressed in A549 cells transfected with the mRNA construct R2851 and that the tested formulations with added lipids were highly efficient when compared to the control w/o added lipids. This shows that the combination of mRNA with very small amounts of lipid was able to increase the transfection efficiency.

Claims

Claims
1. A composition comprising
(a) a cationisable or permanently cationic lipid or lipidoid, and
(c) a nucleic acid compound;
wherein the lipid or lipidoid and the nucleic acid compound are non-covalently associated, and wherein the ratio of the lipid or lipidoid to the nucleic acid compound is not higher than about 2 nmol lipid per μg nucleic acid compound.
2. The composition of claim 1, wherein the ratio of the cationisable or permanently cationic lipid or lipidoid to the nucleic acid compound is not higher than about 1 nmol/ μg, or is in the range from about 0.05 to about 2 nmol/ μg, or from about 0.1 to about 1.5 nmol/ μg, or from about 0.25 to about 1.0 nmol/ μg, or from about 0.3 to about 0.8 nmol/ μg, such as about 0.4 nmol^g, respectively; and/or wherein the N/P ratio as defined herein is not higher than about 1.
3. The composition of claim 1 or 2, wherein the cationisable or permanently cationic lipid is a compound according to formula
X-Y-Z (formula la) or
X-Y(Zi)-Z2 (formula lb) or
X-Y(Zi) (Z2) -Z3 (formula ic)
Ζΐΐ-Χ-Υ2-Ζ2 (formula Id)
wherein
X is a hydrophilic head group comprising a cationisable or permanently cationic nitrogen;
Y, Y1 and Y2 are linking groups, each comprising an ether, ester, amide, urethane, thioether, disulphide, orthoester, or phosphoramide bond; and
Z, Z1, Z2, and Z3 are independently selected and represent hydrophobic groups each comprising a linear or branched hydrocarbon chain or a cyclic hydrocarbon group, such as a steroid residue, wherein the number of carbon atoms in the linear or branched hydrocarbon chain is
6 or higher for Z; and
4 or higher for Z1 or Z2 or Z3, provided that, for a compound of formula lb, Z1 and Z2 together have at least 12 carbon atoms in their hydrocarbon chains, and for a compound of formula Ic, Z1, Z2 and Z3 together have at least 12 carbon atoms in their hydrocarbon chains.
4. The composition of claim 3, wherein
X is selected from a quaternary ammonium group, in particular a trimethylammonium group, and/or
Y, Y1 and/or Y2 are selected from linking groups comprising an ester or amide bond or a dioxolane ring; and/or
Z is a steroid residue; and/or
Z1, Z2, and/or Z3 are selected from saturated or unsaturated hydrocarbon chains with 14 to 22 carbon atoms.
5. The composition of any one of claims 1 to 4, wherein the cationisable or permanently
cationic lipid is a compound according to formula la, lb, Ic or Id which is not zwitterionic under substantially neutral or physiological conditions, and is optionally selected from the group consisting of
N,N-di[(0-hexadecanoyl)hydroxyethyl]-N-hydroxyethyl-N-methyl ammonium bromide ("DOHEMAB");
N-[l-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTMA"; also known as l,2-dioleyloxy-3-trimethylaminopropane chloride);
N-[l-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTAP" or "DOTAP.C1", also known as l,2-dioleoyloxy-3-trimethylaminopropane chloride);
l,2-dioleoyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide („DORI"); l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide („DORIE"); l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxypropyl ammonium bromide („DORIE-
HP");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxybutyl ammonium bromide („DORIE-
HB");
l,2-dioleyloxypropyl-N,N-dimethyl-N-hydroxypentyl ammonium bromide („DORIE- HPe");
l,2-dimyristyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE" or "DIMRI")
l,2-dimpalmityloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DPRIE");
l,2-distearyloxypropyl-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DSRIE"); l,2-dilinoleyloxy-3-trimethylaminopropane chloride ("DLin-TMA.Cl");
l,2-dilinoleoyl-3-trimethylaminopropane chloride ("DLin-TAP.Cl");
rac-[(2,3-dioctadecyloxypropyl) (2-hydroxyethyl)]-dimethylammonium chloride ("CLIPl");
rac-[2(2,3-dihexadecyloxypropyl-oxymethyloxy)ethyl]trimethylammonium ("CLIP6");
rac-[2(2,3-dihexadecyloxypropyl-oxysuccinyloxy)ethyl]-trimethylammonium ("CLIP9");
N-[l-(2,3-dioleyloxy)propyl]-N-2-(sperminecarboxamido)ethyl)-N,N-dimethyl- ammonium trifluoracetate ("DOSPA"; also referred to as 2,3-dioleyloxy- [2(sperminecarboxamido)ethyl]-N,N-dimethyl-l-propanaminiumtrifluoroacetate);
0,0-ditetradecanoyl-N-(a-trimethylammonioacetyl)diethanolamine chloride ("DC-6-
14");
(6Z,9Z,28Z,31Z) ieptatriaconta-6,9,28,31-tetraen-19-yl-4- (trimethylamino)butanoate and its salts ("DLin-MC3-TMA", also referred to as "MC3- cationised");
2,2-dilinoleyl-4-(2-trimethylaminoethyl)-[l,3]-dioxolane ("DLin-KC2-TMA", also referred to as "KC2 cationised");
3-beta-[N-(N',N',N'-trimethylaminoethane)carbamoyl]cholesterol iodide ("TC-Chol") l-(2-octylcyclopropyl)heptadec-8-yl-4-(trimethylammonium)butanoate ("C9-C17-C3 cat");
l-(2-octylcyclopropyl)heptadec-8-yl-l,l-dimethyl-3-pyrrolidiniumcarboxylate ("C9-C17-P cat").
6. The composition of claim 1 or 2, wherein the cationisable or permanently cationic lipidoid is a compound comprising at least one moiety of formula III:
N+(Ri) (R2)-CH2-CH(R3)-R4
(formula III)
wherein independently for each individual moiety of formula III
- Ri and R2 are independently selected from Ci-C4-alkyl,
- R3 is hydrogen or hydroxyl; and
- R4 is selected from linear or branched, saturated or unsaturated C6-C16 hydrocarbyl chain.
7. The composition of claim 6, wherein lipidoid compound is a compound comprising three identical moieties of formula III, and wherein
- Ri and R2 are methyl;
- R3 is hydroxyl, and
- R4 is a linear or branched C6-C16 alkyl chain.
8. The composition of claim 7, wherein lipidoid comprises the cation depicted in the formula IV
Figure imgf000171_0001
Figure imgf000171_0002
9. The composition of any one of the preceding claims, wherein the nucleic acid compound forms a complex with the permanently cationic lipid or lipidoid.
10. The composition of any one of the preceding claims, being substantially free of non-cationic lipids or lipidoids.
11. The composition of any one of the preceding claims, wherein the nucleic acid compound and the cationisable or permanently cationic lipid or lipidoid are comprised in a nanoparticle.
12. A nanoparticle comprising a composition as defined in claims 1 to 11.
13. The nanoparticle of claim 12, wherein the nucleic acid compound is selected from
- chemically modified or unmodified DNA, single stranded or double stranded DNA, coding or non-coding DNA, optionally selected from plasmid, oligodesoxynucleotide, genomic DNA, DNA primers, DNA probes, immunostimulatory DNA, aptamer, or any combination thereof, and/or
- chemically modified or unmodified RNA, single-stranded or double-stranded RNA, coding or non-coding RNA, optionally selected from messenger RNA (mRNA),
oligoribonucleotide, viral RNA (vRNA), replicon RNA, transfer RNA (tRNA), ribosomal RNA (rRNA), immunostimulatory RNA (isRNA), microRNA, small interfering RNA (siRNA), small nuclear RNA (snRNA), small-hairpin RNA (shRNA) or a riboswitch, an RNA aptamer, an RNA decoy, an antisense RNA, a ribozyme, or any combination thereof.
14. The nanoparticle of claim 12 or 13, wherein the nanoparticle has a hydrodynamic diameter as determined by dynamic laser scattering from about 30 nm to about 800 nm, and preferably from about 50 nm to about 300 nm, or from about 60 nm to about 250 nm, or from about 60 nm to about 150 nm, or from about 60 nm to about 120 nm, respectively.
15. The nanoparticle of any one of claims 12 to 14, wherein the nanoparticle has a zeta
potential in the range from about 0 mV to about -50 mV, or from about 0 mV to about - 10 mV.
16. The nanoparticle of any one of claims 12 to 15, wherein the nanoparticle further comprises one or more compounds independently selected from targeting agents, cell penetrating agents, and stealth agents.
17. The nanoparticle of claim 16, wherein the nanoparticle essentially consists of
(a) one or more cationisable or permanently cationic lipids or lipidoids;
(b) one or more nucleic acid compounds; and optionally
(c) one or more compounds independently selected from targeting agents, cell penetrating agent, and stealth agents.
18. A pharmaceutical composition comprising a plurality of the nanoparticles of any one of claims 11 to 17.
19. The pharmaceutical composition of claim 18, wherein the nucleic acid compound is a
coding nucleic acid which encodes at least one peptide or protein.
20. The pharmaceutical composition of claim 19 wherein the coding nucleic acid encodes a therapeutically active protein or an antigen.
21. A vaccine comprising the pharmaceutical composition of claim 31, wherein the coding nucleic acid encodes at least one antigen.
22. The composition of any one of claims 1 to 11 or claims 18 to 20, wherein the composition is formulated as a sterile liquid.
23. The pharmaceutical composition of any one of claims 18 to 20 or 22, wherein the
nanoparticles have a hydrodynamic mean diameter as determined by dynamic laser scattering from about 30 nm to about 800 nm, and preferably from about 50 nm to about 300 nm, or from about 60 nm to about 250 nm, or from about 60 nm to about 150 nm, or from about 60 nm to about 120 nm, respectively.
24. The pharmaceutical composition of any one of claims 1 to 11 or 18 to 20, wherein the
composition is formulated as a steril solid composition, such as a powder or lyophilised form for reconstitution with an aqueous liquid carrier.
25. A kit for preparing the composition of any one of claims 1 to 11 or 18 to 20 or 22 to 24, comprising
(a) a first kit component comprising the cationisable or permanently cationic lipid or lipidoid; and
(b) a second kit component comprising the nucleic acid compound.
26. A permanently cationic lipidoid comprising the cation depicted in the formula IV:
Figure imgf000173_0001
further optionally comprising a pharmaceutically acceptable anion. A cationisable li idoid comprising the compound depicted in formula IVa:
Figure imgf000173_0002
28. The nanoparticle of any one of claims 12 to 17 or the composition of any one of claims 1 to 11 or 18 to 20 or 22 to 24 or the vaccine of claim 21 or the kit of claim 25 for use as a medicament.
29. The nanoparticle or composition or vaccine or kit for use according to claim 28, wherein the use comprises the prophylaxis, treatment and/or amelioration of diseases selected from cancer or tumour diseases, infectious diseases, preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases, monogenetic diseases, i.e. (hereditary) diseases, or genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a defined gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, diseases of the respiratory system, diseases of the digestive system, diseases of the skin, musculoskeletal disorders, disorders of the connective tissue, neoplasms, immune deficiencies, endocrine, nutritional and metabolic diseases, eye diseases, ear diseases and diseases associated with a peptide or protein deficiency.
30. The nanoparticle or composition or vaccine or kit for use according to claim 28 or 29,
wherein the use comprises the extravascular administration of the composition and/or nanoparticle to a subject by
(a) local or locoregional injection, infusion or implantation, in particular intradermal, subcutaneous, intramuscular, interstitial, locoregional, intravitreal, periocular,
intratumoural, intralymphatic, intranodal, intra-articular, intrasynovial, periarticular, intraperitoneal, intra-abdominal, intracardial, intrapericardial, intraventricular, intrapleural, perineural, intrathoracic, epidural, intradural, peridural, intrathecal, intramedullary intracerebral, intracavernous, intracorporus cavernosum, intraprostatic, intratesticular, intracartilaginous, intraosseous, intradiscal, intraspinal, intracaudal, intrabursal, intragingival, intraovarian, intrauterine, periodontal, retrobulbar,
subarachnoid, subconjunctival or intralesional injection, infusion or implantation;
(b) topical administration to the skin or a mucosa, in particular dermal or cutaneous, nasal, buccal, sublingual, otic or auricular, ophthalmic, conjunctival, vaginal, rectal, intracervical, endosinusial, laryngeal, oropharyngeal, ureteral, or urethral administration;
(c) administration to the respiratory system by inhalation, in particular by aerosol administration to the lungs, bronchi, bronchioli, alveoli, or paranasal sinuses; or
(d) transdermal or percutaneous administration.
PCT/EP2017/064057 2016-06-09 2017-06-09 Cationic carriers for nucleic acid delivery WO2017212007A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP17730740.2A EP3468609A1 (en) 2016-06-09 2017-06-09 Cationic carriers for nucleic acid delivery
US16/308,595 US20190336608A1 (en) 2016-06-09 2017-06-09 Cationic carriers for nucleic acid delivery

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP2016063229 2016-06-09
EPPCT/EP2016/063229 2016-06-09

Publications (1)

Publication Number Publication Date
WO2017212007A1 true WO2017212007A1 (en) 2017-12-14

Family

ID=56134339

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/064057 WO2017212007A1 (en) 2016-06-09 2017-06-09 Cationic carriers for nucleic acid delivery

Country Status (3)

Country Link
US (1) US20190336608A1 (en)
EP (1) EP3468609A1 (en)
WO (1) WO2017212007A1 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019038332A1 (en) 2017-08-22 2019-02-28 Curevac Ag Bunyavirales vaccine
CN110283223A (en) * 2018-03-19 2019-09-27 广州市锐博生物科技有限公司 A kind of cation lipid molecule and its application in delivery of nucleic acids
WO2019193183A2 (en) 2018-04-05 2019-10-10 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
WO2020002525A1 (en) 2018-06-27 2020-01-02 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
WO2020261227A1 (en) * 2019-06-26 2020-12-30 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
US10988754B2 (en) 2017-07-04 2021-04-27 Cure Vac AG Nucleic acid molecules
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
JP2021519595A (en) * 2018-04-17 2021-08-12 キュアバック アーゲー New RSV RNA molecule and vaccination composition
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
US11141474B2 (en) 2016-05-04 2021-10-12 Curevac Ag Artificial nucleic acid molecules encoding a norovirus antigen and uses thereof
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
US11225682B2 (en) 2015-10-12 2022-01-18 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
US11464836B2 (en) 2016-12-08 2022-10-11 Curevac Ag RNA for treatment or prophylaxis of a liver disease
US11464847B2 (en) 2016-12-23 2022-10-11 Curevac Ag Lassa virus vaccine
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
US11542490B2 (en) 2016-12-08 2023-01-03 CureVac SE RNAs for wound healing
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
US11723967B2 (en) 2016-02-17 2023-08-15 CureVac SE Zika virus vaccine
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
US11975064B2 (en) 2011-03-02 2024-05-07 CureVac SE Vaccination with mRNA-coded antigens

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2015010880A (en) 2013-02-22 2015-12-03 Curevac Gmbh Combination of vaccination and inhibition of the pd-1 pathway.
AU2014310934B2 (en) 2013-08-21 2019-09-12 CureVac SE Respiratory syncytial virus (RSV) vaccine
WO2016091391A1 (en) 2014-12-12 2016-06-16 Curevac Ag Artificial nucleic acid molecules for improved protein expression
EP3294885B1 (en) 2015-05-08 2020-07-01 CureVac Real Estate GmbH Method for producing rna
CN107873055B (en) 2015-05-29 2021-09-17 库瑞瓦格房地产有限公司 Method for producing and purifying RNA comprising at least one tangential flow filtration step
WO2017109134A1 (en) 2015-12-22 2017-06-29 Curevac Ag Method for producing rna molecule compositions
WO2018096179A1 (en) 2016-11-28 2018-05-31 Curevac Ag Method for purifying rna
US11524066B2 (en) 2016-12-23 2022-12-13 CureVac SE Henipavirus vaccine
SG11201906297QA (en) 2017-03-24 2019-10-30 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
CN111511924A (en) 2017-11-08 2020-08-07 库瑞瓦格股份公司 RNA sequence modulation
US11931406B2 (en) 2017-12-13 2024-03-19 CureVac SE Flavivirus vaccine
SG11202005760PA (en) 2017-12-21 2020-07-29 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
US11541105B2 (en) 2018-06-01 2023-01-03 The Research Foundation For The State University Of New York Compositions and methods for disrupting biofilm formation and maintenance
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
AU2021252164A1 (en) 2020-04-09 2022-12-15 Finncure Oy Mimetic nanoparticles for preventing the spreading and lowering the infection rate of novel coronaviruses
MX2023007574A (en) 2020-12-22 2023-09-29 CureVac SE Rna vaccine against sars-cov-2 variants.
TW202404614A (en) * 2022-04-04 2024-02-01 美商星火治療公司 Immune enhancement and infectious disease treatment
WO2023235818A2 (en) 2022-06-02 2023-12-07 Scribe Therapeutics Inc. Engineered class 2 type v crispr systems
WO2023240074A1 (en) 2022-06-07 2023-12-14 Scribe Therapeutics Inc. Compositions and methods for the targeting of pcsk9
TW202405175A (en) 2022-06-07 2024-02-01 美商斯奎柏治療公司 Compositions and methods for the targeting of pcsk9

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999015206A1 (en) * 1997-09-23 1999-04-01 Megabios Corporation Methods for preparing lipids/polynucleotide transfection complexes
WO2001092542A2 (en) * 2000-05-30 2001-12-06 Ich Productions Limited Integrin-targeting vectors having enhanced transfection activity
WO2002098443A2 (en) 2001-06-05 2002-12-12 Curevac Gmbh Stabilised mrna with an increased g/c content and optimised codon for use in gene therapy
WO2003086280A2 (en) 2002-04-04 2003-10-23 Coley Pharmaceutical Gmbh Immunostimulatory g,u-containing oligoribonucleotides
US6770740B1 (en) 1999-07-13 2004-08-03 The Regents Of The University Of Michigan Crosslinked DNA condensate compositions and gene delivery methods
US20080153166A1 (en) * 1995-01-23 2008-06-26 Leaf Huang Stable lipid-comprising drug delivery complexes and methods for their production
WO2008077592A1 (en) 2006-12-22 2008-07-03 Curevac Gmbh Method for purifying rna on a preparative scale by means of hplc
US20120009222A1 (en) * 2008-10-27 2012-01-12 Massachusetts Institute Of Technology Modulation of the immune response
WO2012019780A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
WO2013143700A2 (en) 2012-03-27 2013-10-03 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr
US8969353B2 (en) 2008-11-07 2015-03-03 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080153166A1 (en) * 1995-01-23 2008-06-26 Leaf Huang Stable lipid-comprising drug delivery complexes and methods for their production
WO1999015206A1 (en) * 1997-09-23 1999-04-01 Megabios Corporation Methods for preparing lipids/polynucleotide transfection complexes
US6770740B1 (en) 1999-07-13 2004-08-03 The Regents Of The University Of Michigan Crosslinked DNA condensate compositions and gene delivery methods
WO2001092542A2 (en) * 2000-05-30 2001-12-06 Ich Productions Limited Integrin-targeting vectors having enhanced transfection activity
WO2002098443A2 (en) 2001-06-05 2002-12-12 Curevac Gmbh Stabilised mrna with an increased g/c content and optimised codon for use in gene therapy
WO2003086280A2 (en) 2002-04-04 2003-10-23 Coley Pharmaceutical Gmbh Immunostimulatory g,u-containing oligoribonucleotides
WO2008077592A1 (en) 2006-12-22 2008-07-03 Curevac Gmbh Method for purifying rna on a preparative scale by means of hplc
US20120009222A1 (en) * 2008-10-27 2012-01-12 Massachusetts Institute Of Technology Modulation of the immune response
US8969353B2 (en) 2008-11-07 2015-03-03 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
WO2012019780A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
WO2013143700A2 (en) 2012-03-27 2013-10-03 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr

Non-Patent Citations (27)

* Cited by examiner, † Cited by third party
Title
AKASHI, CURR. OPIN. GENET. DEV., vol. 11, no. 6, 2001, pages 660 - 666
BAUER S ET AL., PROC NATLACADSCI USA, vol. 98, 2001, pages 9237 - 42
BINDER ET AL., EMBO J., vol. 13, 1994, pages 1969 - 1980
CAPUT ET AL., PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 1670 - 1674
ELBASHIR ET AL., NATURE, vol. 411, 2001, pages 494 - 498
FOERG; MERKLE, JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 97, no. 1, 2008, pages 144 - 62, Retrieved from the Internet <URL:www.interscience.wiley.com>
FRANCES M. P. WONG ET AL: "Cationic Lipid Binding to DNA: Characterization of Complex Formation +", BIOCHEMISTRY, vol. 35, no. 18, 1 January 1996 (1996-01-01), US, pages 5756 - 5763, XP055348342, ISSN: 0006-2960, DOI: 10.1021/bi952847r *
GAO ET AL., THE AAPS JOURNAL, vol. 9, no. 1, 2007
GAO, X.; K. S. KIM, AAPS J, vol. 9, no. 1, 2007, pages E92 - 104
HANNON, NATURE, vol. 41, 2002, pages 244 - 251
HE; KATZE, VIRAL IMMUNOL., vol. 15, 2002, pages 95 - 119
HEMMI H, NATURE, vol. 408, 2000, pages 740 - 5
KWOK, K. Y.; D. L. MCKENZIE ET AL.: "Formulation of highly soluble poly(ethylene glycol)-peptide DNA condensates.", J PHARM SCI, vol. 88, no. 10, 1999, pages 996 - 1003, XP002165042, DOI: doi:10.1021/js990072s
LOVE ET AL., PNAS, vol. 107, no. 5, 2010, pages 1864 - 1869
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
MARTIN, M. E.; K. G. RICE, AAPS J, vol. 9, no. 1, 2007, pages E18 - 29
MARTIN; RICE, THE AAPS JOURNAL, vol. 9, no. 1, 2007
MCKENZIE, D. L.; E. SMILEY ET AL., BIOCONJUG CHEM, vol. 11, no. 6, 2000, pages 901 - 9
MCKENZIE, D. L.; K. Y. KWOK ET AL., J BIOL CHEM, vol. 275, no. 14, 2000, pages 9970 - 7
MEYLAN, E.; TSCHOPP, J.: "Toll-like receptors and RNA helicases: Two parallel ways to trigger antiviral responses.", MOL. CELL, vol. 22, 2006, pages 561 - 569
READ, M. L.; K. H. BREMNER ET AL.: "Vectors based on reducible polycations facilitate intracellular release of nucleic acids", J GENE MED, vol. 5, no. 3, 2003, pages 232 - 45, XP002481542, DOI: doi:10.1002/jgm.331
READ, M. L.; S. SINGH: "A versatile reducible polycation-based system for efficient delivery of a broad range of nucleic acids", NUCLEIC ACIDS RES, vol. 33, no. 9, 2005, pages e86
SHARP, GENES DEV, vol. 5, 2001, pages 485 - 490
STAHL ET AL.: "Handbook of Pharmaceutical Salts", 2002, WILEY-VCH
STARK ET AL., ANNU. REV. BIOCHEM., vol. 67, 1998, pages 227 - 264
VIVES, E.; P. BRODIN ET AL.: "A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus", J BIOL CHEM, vol. 272, no. 25, 1997, pages 16010 - 7, XP002940007, DOI: doi:10.1074/jbc.272.25.16010
ZAMORE, NAT. STRUCT. BIOL., vol. 9, 2001, pages 746 - 750

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11975064B2 (en) 2011-03-02 2024-05-07 CureVac SE Vaccination with mRNA-coded antigens
US11225682B2 (en) 2015-10-12 2022-01-18 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
US11786590B2 (en) 2015-11-09 2023-10-17 CureVac SE Rotavirus vaccines
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US11723967B2 (en) 2016-02-17 2023-08-15 CureVac SE Zika virus vaccine
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
US11141474B2 (en) 2016-05-04 2021-10-12 Curevac Ag Artificial nucleic acid molecules encoding a norovirus antigen and uses thereof
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
US11542490B2 (en) 2016-12-08 2023-01-03 CureVac SE RNAs for wound healing
US11464836B2 (en) 2016-12-08 2022-10-11 Curevac Ag RNA for treatment or prophylaxis of a liver disease
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
US11464847B2 (en) 2016-12-23 2022-10-11 Curevac Ag Lassa virus vaccine
US10988754B2 (en) 2017-07-04 2021-04-27 Cure Vac AG Nucleic acid molecules
WO2019038332A1 (en) 2017-08-22 2019-02-28 Curevac Ag Bunyavirales vaccine
CN110283223B (en) * 2018-03-19 2022-01-04 广州市锐博生物科技有限公司 Cationic lipid molecule and application thereof in nucleic acid delivery
CN110283223A (en) * 2018-03-19 2019-09-27 广州市锐博生物科技有限公司 A kind of cation lipid molecule and its application in delivery of nucleic acids
WO2019193183A2 (en) 2018-04-05 2019-10-10 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
EP4227319A1 (en) 2018-04-17 2023-08-16 CureVac SE Novel rsv rna molecules and compositions for vaccination
JP2021519595A (en) * 2018-04-17 2021-08-12 キュアバック アーゲー New RSV RNA molecule and vaccination composition
WO2020002525A1 (en) 2018-06-27 2020-01-02 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
WO2020128031A2 (en) 2018-12-21 2020-06-25 Curevac Ag Rna for malaria vaccines
WO2020161342A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophtalmic diseases
WO2020254535A1 (en) 2019-06-18 2020-12-24 Curevac Ag Rotavirus mrna vaccine
KR20220068979A (en) * 2019-06-26 2022-05-26 주식회사 바이오오케스트라 Micellar nanoparticles and uses thereof
WO2020261227A1 (en) * 2019-06-26 2020-12-30 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
KR102658962B1 (en) 2019-06-26 2024-04-23 주식회사 바이오오케스트라 Micellar nanoparticles and their uses
US11839624B2 (en) 2019-06-26 2023-12-12 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
WO2021028439A1 (en) 2019-08-14 2021-02-18 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
DE202021004130U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
DE202021004123U1 (en) 2020-02-04 2022-10-26 Curevac Ag Coronavirus Vaccine
EP4147717A1 (en) 2020-02-04 2023-03-15 CureVac SE Coronavirus vaccine
WO2021156267A1 (en) 2020-02-04 2021-08-12 Curevac Ag Coronavirus vaccine
DE202021003575U1 (en) 2020-02-04 2022-01-17 Curevac Ag Coronavirus Vaccine
DE112021000012T5 (en) 2020-02-04 2021-11-18 Curevac Ag Coronavirus vaccine
WO2021239880A1 (en) 2020-05-29 2021-12-02 Curevac Ag Nucleic acid based combination vaccines
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
WO2022043551A2 (en) 2020-08-31 2022-03-03 Curevac Ag Multivalent nucleic acid based coronavirus vaccines
WO2022162027A2 (en) 2021-01-27 2022-08-04 Curevac Ag Method of reducing the immunostimulatory properties of in vitro transcribed rna
WO2022233880A1 (en) 2021-05-03 2022-11-10 Curevac Ag Improved nucleic acid sequence for cell type specific expression
WO2023073228A1 (en) 2021-10-29 2023-05-04 CureVac SE Improved circular rna for expressing therapeutic proteins
WO2023144330A1 (en) 2022-01-28 2023-08-03 CureVac SE Nucleic acid encoded transcription factor inhibitors
WO2023227608A1 (en) 2022-05-25 2023-11-30 Glaxosmithkline Biologicals Sa Nucleic acid based vaccine encoding an escherichia coli fimh antigenic polypeptide

Also Published As

Publication number Publication date
EP3468609A1 (en) 2019-04-17
US20190336608A1 (en) 2019-11-07

Similar Documents

Publication Publication Date Title
US20230302146A1 (en) Hybrid carriers for nucleic acid cargo
US20190336608A1 (en) Cationic carriers for nucleic acid delivery
US20190336611A1 (en) Hybrid carriers for nucleic acid cargo
US20190381180A1 (en) Hybrid carriers for nucleic acid cargo
US10912826B2 (en) 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
JP7289265B2 (en) Lipid nanoparticle mRNA vaccine
EP3319622B1 (en) Method for producing rna molecule compositions
EP3310384A1 (en) Vaccine composition
JP2023546908A (en) Ionizable lipids and methods of production and use thereof
EP3348645A1 (en) 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
CN116917266A (en) Ionizable lipids and methods of making and using the same

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: 17730740

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017730740

Country of ref document: EP

Effective date: 20190109