EP3298143A2 - Composition de poudre sèche comprenant de l'arn à chaîne longu - Google Patents

Composition de poudre sèche comprenant de l'arn à chaîne longu

Info

Publication number
EP3298143A2
EP3298143A2 EP16728216.9A EP16728216A EP3298143A2 EP 3298143 A2 EP3298143 A2 EP 3298143A2 EP 16728216 A EP16728216 A EP 16728216A EP 3298143 A2 EP3298143 A2 EP 3298143A2
Authority
EP
European Patent Office
Prior art keywords
dry powder
powder composition
long
rna molecule
diseases
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP16728216.9A
Other languages
German (de)
English (en)
Inventor
Benyamin YAZDAN PANAH
Fabian Johannes EBER
Stefanie SEWING
Thomas Ketterer
Thorsten Mutzke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Curevac SE
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
Publication of EP3298143A2 publication Critical patent/EP3298143A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0091Purification or manufacturing processes for gene therapy compositions

Definitions

  • Dry powder composition comprising long-chain RNA
  • the present invention is directed to a storage-stable formulation of long-chain RNA.
  • the invention concerns a dry powder composition comprising a long-chain RNA molecule.
  • the present invention is furthermore directed to methods for preparing a dry powder composition comprising a long-chain RNA molecule by spray-drying or spray- freeze-drying.
  • the invention further concerns the use of such a dry powder composition comprising a long-chain RNA molecule in the preparation of pharmaceutical compositions and vaccines, to a method of treating or preventing a disorder or a disease, to first and second medical uses of such a dry powder composition comprising a long-chain RNA molecule and to kits, particularly to kits of parts, comprising such a dry powder composition comprising a long-chain RNA molecule.
  • nucleic acid molecules are therefore regarded as important tools for gene therapy and prophylactic and therapeutic vaccination against infectious and malignant diseases.
  • application of RNA also represents a favored tool in modern molecular medicine. It also exhibits some superior properties over DNA cell transfection. As generally known, transfection of DNA molecules may lead to serious complications. For example, application of DNA molecules bears the risk that the DNA integrates into the host genome.
  • RNA particularly mRNA
  • An advantage of using RNA rather than DNA is that no virus-derived promoter element has to be administered in vivo and no integration into the genome may occur. Furthermore, the RNA, in order to exert its function, does not need to overcome the barrier to the nucleus.
  • RNA Ribonucleic acid
  • RNA in solution
  • RNA is typically stored at -20° C or even -80° C and under RNAse free conditions to prevent degradation of the RNA.
  • Such storage conditions do not sufficiently prevent a loss of function over time. Additionally, applying such conditions is very cost-intensive, especially for shipping and storage, e.g. whenever such low temperatures have to be guaranteed.
  • the only method for stabilization of long-chain RNA comprises lyophilization or freeze-drying of the RNA (see e.g. WO2011/069587 and WO2011/069586).
  • Lyophilization is a method, which is known and recognized in the art to enhance storage stability of temperature sensitive biomolecules, such as nucleic acids.
  • water is typically removed from a frozen sample containing nucleic acids via sublimation.
  • the process of lyophilization is usually characterized by a primary and a secondary drying step. In the primary drying step, free, i.e. unbound, water surrounding the nucleic acid (sequence) and optionally further components, evaporates from the frozen solution.
  • lyophilization is the most common processing method for removing moisture from biopharmaceuticals, and it can increase stability, temperature tolerance, and shelf life of these products.
  • lyphilization does have its limitations, especially if scale-up is needed.
  • One major disadvantage of lyophilization is that every single vial containing a sample has to be lyophilized separately. The lyophilized product cannot be separated into distinct charges or aliquots, as the lyophilized product is not provided e.g. in powder form.
  • lyophilization of samples for a scaled-up production involves cost-intensive equipment, since, for example, a lot of lyophilizers are needed for market production, requiring large production facilities. Together with the time required for lyophilization and the additional requirement of a granulation step that renders lyophilization a technique, which is often not suitable for industrial scale production. Especially in an environment where budgets are tightening, and where time and facility space are at a premium, lyophilization may not be considered, e.g. by the pharmaceutical industry, as a competitive process.
  • Double-stranded short interfering RNAs for inhalation were dried by using a spray-drying technology.
  • PLGA poly(D,L-lactide-co-glycolide)
  • NPs nanoparticles
  • US2011/077284 discloses the provision of dry powders of therapeutic and inhalable short siRNAs against influenza virus. Therein, drying temperatures were determined experimentally. Generally, the inlet temperature was from about 65° C to about 125° C, while the outlet temperature ranged from about 30° C to about 70° C. In the examples presented therein, siRNAs are dried at a T ou tiet of ⁇ 55°C. The powders generally had a moisture content of typically less than 10% by weight, or less than 5% by weight, or less than 3% by weight. Also, the study characterized the chemical stability of the dry powder. Less than 10% by weight of the active siRNA were degraded upon storage of the dry powder composition under ambient conditions for a period of 18 months. However, the biological activity of the siRNA stored as a dry powder was not determined.
  • lyophilization of long-chain RNA bears the problem that it is very cost- and time-intensive, particularly if commercial production in a scaled-up process is envisaged.
  • the underlying object is therefore to provide a nucleic acid molecule, in particular a long- chain RNA, exhibiting no loss of activity when stored prior to its use and being available by cost-avoiding production process.
  • One object of the invention is to provide a dry powder composition comprising a long-chain RNA molecule.
  • RNA molecule retains its chemical integrity and its biological activity. It is another object of the present invention that such methods are applicable under industrial large-scale production conditions, preferably by a continuous process. It is a particular object to provide a method that allows drying a liquid comprising long-chain RNA molecules.
  • the invention relates to a long-chain RNA molecule in a particulate formulation.
  • the invention concerns a dry powder composition comprising a long-chain RNA molecule.
  • long-chain RNA in contrast to shorter double-stranded RNAs
  • the dry powder composition according to the invention provides a storage-stable form of a long-chain RNA molecule.
  • the dry powder composition according to the invention is characterized by superior handling properties.
  • the inventive dry powder composition can be packaged in any quantity or in any container or dosage form, respectively. Handling of the dry powder composition according to the invention is further improved by its free-flowing properties.
  • the inventive dry powder composition does not form agglomerates or aggregates that would inhibit packaging and/or dosage. Due to its flowability, the dry powder composition according to the invention can be readily further processed. For instance, the dry powder composition can be transferred, e.g. from one vessel to another or from a larger vessel into a plurality of smaller vessels, simply by pouring. The inventive dry powder composition can readily be packaged in a variety of packages and final dosage forms according to the actual requirements.
  • the dry powder composition according to the invention provides excellent storage stability.
  • the invention provides a dry powder composition comprising a long-chain RNA molecule, wherein the long-chain RNA molecule preferably comprises at least 30 nucleotides.
  • the long-chain RNA molecule is a molecule as defined herein. More preferably, the long-chain RNA molecule is not an RNA molecule selected from the group consisting of a small interfering RNA (siRNA), a microRNA, a small nuclear RNA (snRNA), a small-hairpin (sh) RNA, a riboswitch, a ribozyme or an aptamer.
  • the long-chain RNA is not an siRNA, most preferably not a double-stranded siRNA.
  • the dry powder composition as described herein does not comprise an RNA molecule comprising less than 30 nucleotides, less than 200 nucleotides or less than 250 nucleotides.
  • the dry powder composition does preferably not comprise an RNA molecule selected from the group consisting of a small interfering RNA (siRNA), preferably a single-stranded or a double-stranded siRNA, a microRNA, a small nuclear RNA (snRNA), a small-hairpin (sh) RNA, a riboswitch, a ribozyme or an aptamer.
  • siRNA small interfering RNA
  • siRNA small interfering RNA
  • snRNA small nuclear RNA
  • sh small-hairpin
  • ribozyme a ribozyme or an aptamer.
  • the dry powder composition does not comprise an siRNA, most preferably not a double-stranded siRNA.
  • dry powder typically refers to a composition that is - amongst other features - characterized by its residual moisture content, which is preferably low enough in order to prevent the formation of aggregates that would reduce or inhibit the flowability of the powder.
  • residual moisture content typically refers to the total amount of solvent present in the dry powder composition. Said total amount of residual solvents in the dry powder composition is determined using any suitable method known in the art.
  • the residual moisture content of the inventive dry powder composition is expected to contribute to its excellent storage stability.
  • the residual moisture content of the dry powder composition according to the invention is 15% (w/w) or less, more preferably 10% (w/w) or less, even more preferably 9% (w/w), 8% (w/w), 7% (w/w), 6% (w/w) or 5% (w/w).
  • the residual moisture content of the dry powder composition is 5% (w/w) or less, preferably 4% (w/w) or less. In a particularly preferred embodiment, the residual moisture is 7% (w/w) or less. In a further preferred embodiment, the residual moisture content of the dry powder composition in the range from 0% to 15% (w/w), from 0% to 10% (w/w), from 0% to 7% (w/w), from 0% to 5% (w/w), from 0% to 4% (w/w), from 3% to 6% (w/w) or from 2% to 5% (w/w).
  • the residual moisture content of the dry powder composition as described herein is 5% (w/w) or less, more preferably 4% (w/w) or less, even more preferably 3% (w/w) or less, 2% (w/w) or less, or 1 % (w/w) or less.
  • the residual moisture content of the dry powder composition as described herein is preferably in the range from from 0% to 5% (w/w), from 0% to 4% (w/w), from 0% to 3% (w/w), from 0% to 2% (w/w) or from 0% to 1% (w/w).
  • the dry powder composition comprising a long-chain RNA molecule comprises a plurality of particles.
  • the term “particle” typically refers to an individual solid particle of the dry powder composition.
  • the individual particles of the dry powder composition according to the invention are preferably physically separated from each other, i.e. the individual particles that constitute the dry powder may be in lose and reversible contact with each other (as opposed to an irreversible link between individual particles).
  • the term “particle” refers to the smallest physical entity of the inventive dry powder composition.
  • the particles of the inventive dry powder composition do preferably not stick to each other.
  • the particulate nature of the formulation contributes to the superior characteristics of the inventive dry powder composition, e.g. its free flowability.
  • the plurality of individual particles of the dry powder composition according to the invention is characterized by a size distribution, wherein the size of individual particles may be the same or different from each other.
  • the size of the particles of a dry powder composition is characterized by a Gaussian distribution or a quasi-Gaussian distribution.
  • the dry powder composition according to the invention is characterized by a volume weighted particle size distribution as determined, for instance, by static light scattering techniques, such as laser diffraction, or by using a cascade impactor. In a volume weighted distribution, the contribution of each particle in the dry powder composition relates to the volume of that particle.
  • the dry powder composition according to the invention is preferably characterized by parameters such as, for example, Dv10, Dv50, Dv90 or the mass median aerodynamic diameter (MMAD).
  • the parameter "Dv50" (or “volume D50” or “volume weighted D50”) relates to the median particle size based on a volume weighted particle size distribution.
  • Dv50 thus typically describes the particle size (based on a volume weighted distribution), preferably the diameter of a particle in micrometers (pm), with 50% of the particles in the distribution having a larger size and 50% of the particles in the distribution having a smaller size than Dv50.
  • the parameter "Dv50" typically relates to the diameter (e.g.
  • the dry powder composition according to the invention is characterized by a Dv50 of at least 0.3 ⁇ .
  • the Dv50 of the dry powder composition according to the invention is at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or 50 pm.
  • the Dv50 of the dry powder composition according to the invention is below 1.500, 1.250, 1.000, 750, 600, 500, 400, 300, 200 or 100 pm.
  • the Dv50 may be in a range from 0.3 pm to 2.000 pm, from 1 pm to 1 ,000 pm, from 2 pm to 500 pm or from 2 pm to 200 pm.
  • the Dv50 of the dry powder composition is at least 1 pm or in the range from 1 to 200 pm.
  • the Dv50 of the dry powder composition is at least 3 pm, at least 5 pm or at least 20 pm and/or preferably lower than 50 pm, 100 pm or 200 pm.
  • the parameter "Dv10" corresponds to the cut-off size (preferably in pm) of the particles in a volume weighted distribution, which represent 0% of the total volume of the sample, and which have a particle size equal to or smaller than the Dv 0 value.
  • the Dv10 of the dry powder composition according to the invention is at least 0.2, 0.3, 0.4, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 pm. More preferably, the Dv10 of the dry powder composition is equal to or lower than 5, equal to or lower than 10, or equal to or lower than 20 pm. Most preferably, the Dv10 is in a range from 1 pm to 10 pm or from about 3 pm to about 5 pm.
  • the parameter "Dv90" corresponds to the cut-off size (preferably in pm) of the particles in a volume weighted distribution, which represent 90% of the total volume of the sample, and which have a particle size equal to or smaller than the Dv90 value.
  • the Dv90 of the dry powder composition according to the invention is at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or 50 pm.
  • the Dv90 of the dry powder composition according to the invention is equal to or lower than 1.500, 1.250, 1.000, 750, 600, 500, 400, 300, 200 or 100 pm.
  • the Dv90 may be in a range from 0.3 pm to 2.000 pm, from 1 pm to 1.000 pm, from 2 pm to 500 pm or from 2 pm to 200 pm.
  • the Dv90 of the dry powder composition is at least 1 pm or in the range from 1 to 200 pm.
  • the Dv90 of the dry powder composition is at least 3 pm, at least 5 pm or at least 20 pm.
  • the mass median aerodynamic diameter (MMAD) describes the particle size based on the aerodynamic properties of the respective particle, in particular its settling behaviour.
  • the MMAD is preferably determined using any suitable instrument known in the art, such as, for instance, an APSTM spectrometer (TSI Inc.).
  • the MMAD relates to the median aerodynamic diameter of the particle distribution and is the diameter of a unit density sphere having the same settling velocity, in air, as the particle.
  • the MMAD thus typically describes the particle size, preferably in micrometers ( ⁇ ), with 50% of the particles in the distribution having a larger size and 50% of the particles in the distribution having a smaller size than the MMAD value.
  • the dry powder composition according to the invention is characterized by a MMAD of at least 0.3 pm.
  • the MMAD of the dry powder composition according to the invention is at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or 50 pm.
  • the MMAD of the dry powder composition according to the invention is equal to or less than 1.500, 1.250, 1.000, 750, 600, 500, 400, 300, 200 or 100 pm. Further preferably, the MMAD may be in a range from 0.3 pm to 2.000 pm, from 1 pm to 1.000 pm, from 2 pm to 500 pm or from 2 pm to 200 pm. In a preferred embodiment, the MMAD of the dry powder composition is at least 1 pm or in the range from 1 to 200 pm. In a particularly preferred embodiment, the MMAD of the dry powder composition is at least 3 pm, at least 5 pm or at least 20 pm.
  • the dry powder composition is preferably characterized by using the span of the particle size distribution as a parameter.
  • the span for a volume weighted distribution
  • the inventive dry powder composition is characterized by a low span value, which indicates a narrow (or more homogeneous) particle size distribution. Typically, a narrow distribution enhances the flowability of the dry powder composition.
  • the span of the dry powder composition according to the present invention is equal to or less than 5, more preferably equal to or less than 4, and even more preferably equal to or less than 3.
  • the particle size distribution of the dry powder composition according to the invention is characterized by a span of equal to or less than about 2 or a span of equal to or less than about 1.5.
  • the dry powder composition comprises a plurality of spherical particles.
  • spherical comprises not only geometrically perfect spheres, but also more irregular shapes, such as spheroidal, elipsoid, oval or rounded particles.
  • the shape of an individual particle can be determined by known methods and by using instruments, which are commercially available, such as LasentecTM (particle chord length FBRM), MalvernTM (Fraunhofer diffraction) or Coulter CounterTM (electric zone sensing). Typically, the volume and the surface area of an individual particle are determined.
  • Waddell's sphericity ⁇ herein also referred to as "sphericity" or "circularity" may be calculated, e.g. by using the following formula
  • the average sphericity of the particles, which are contained in the inventive dry powder composition is at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the average sphericity of the particles, which are contained in the dry powder composition is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the average sphericity of the particles, which are contained in the inventive dry powder composition is in the range from 0.7 to 1.
  • the dry powder composition according to the invention consists of particles, which are characterized by a sphericity of at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the dry powder composition consists of particles with a sphericity in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the sphericity of those particles of the dry powder composition that have a particle size equal to Dv50 as defined herein is at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the sphericity of those particles of the dry powder composition that have a particle size equal to Dv50 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • those particles of the dry powder composition that have a particle size equal to Dv90 as defined herein have a sphericity of at least 0.7, preferably of at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the sphericity of those particles of the dry powder composition that have a particle size equal to Dv90 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the average sphericity of those particles of the dry powder composition that have a particle size equal to or lower than Dv50 as defined herein is at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the average sphericity of those particles of the dry powder composition that have a particle size equal to or lower than Dv50 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the average sphericity of those particles of the dry powder composition that have a particle size equal to or lower than Dv90 as defined herein is at least 0.7, preferably of at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the average sphericity of those particles of the dry powder composition that have a particle size equal to or lower than Dv90 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the term "RNA" is used as abbreviation for ribonucleic acid.
  • RNA is a nucleic acid molecule, i.e. a polymer consisting of nucleotides. These nucleotides are usually adenosine-monophosphate, uridine-monophosphate, guanosine-monophosphate and cytidine-monophosphate monomers, which are connected to each other along a so-called backbone.
  • the backbone is formed by phosphodiester bonds between the sugar, i.e. ribose, of a first and a phosphate moiety of a second, adjacent monomer.
  • the specific succession of the monomers is called the RNA sequence.
  • RNA molecule is not limited to any particular type of RNA.
  • RNA may be obtainable by transcription of a DNA-sequence, e.g., inside a cell.
  • transcription is typically performed inside the nucleus or the mitochondria.
  • transcription of DNA usually results in the so-called premature RNA, which has to be processed into so-called messenger-RNA, usually abbreviated as mRNA.
  • Processing of the premature RNA e.g. in eukaryotic organisms, comprises a variety of different posttranscriptional-modifications such as splicing, 5'-capping, polyadenylation, export from the nucleus or the mitochondria and the like. The sum of these processes is also called maturation of RNA.
  • RNA usually provides the nucleotide sequence that may be translated into an amino acid sequence of a particular peptide or protein.
  • a (mature) mRNA comprises a 5'-cap, optionally a 5'UTR, an open reading frame, optionally a 3'UTR and a poly(A) and/or poly(C) sequence.
  • RNA molecule comprises ribonucleic acids comprising more than one open reading frame, such as bicistronic or multicistronic RNA molecules.
  • a bicistronic or multicistronic RNA molecule is typically an RNA molecule, preferably an mRNA molecule, that may typically have two (bicistronic) or more (multicistronic) open reading frames (ORF).
  • RNA molecules further encompass other coding RNA molecules, such as viral RNA, retroviral RNA, self-replicating RNA (replicon RNA), small interfering RNA (siRNA), microRNA, small nuclear RNA (snRNA), small-hairpin (sh) RNA, riboswitches, ribozymes or an aptamers.
  • long-chain RNA molecule typically refers to an RNA molecule, preferably as described herein, which preferably comprises at least 30 nucleotides.
  • the long-chain RNA molecule according to the invention may comprise at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350, 400, 450 or at least 500 nucleotides.
  • the long-chain RNA molecule comprises at least 100 nucleotides, even more preferably at least 200 nucleotides.
  • the long-chain RNA molecule further preferably comprises from 30 to 50.000 nucleotides, from 30 to 20.000 nucleotides, from 100 to 20.000 nucleotides, from 200 to 20.000 nucleotides, from 250 to 15.000 nucleotides or from 500 to 20.000 nucleotides.
  • the long-chain RNA of the dry powder composition as described herein comprises more than 200 nucleotides, preferably at least 250 nucleotides.
  • the long-chain RNA as described herein may comprise more than 210, more than 220, more than 230, more than 240, more than 250, more than 260, more than 270, more than 280, more than 290, more than 300, more than 350, more than 400, more than 450 or more than 500 nucleotides.
  • the long-chain RNA as described herein may comprise at least about 210, at least about 220, at least about 230, at least about 240, at least about 250, at least about 260, at least about 270, at least about 280, at least about 290, at least about 300, at least about 350, at least about 400, at least about 450 or at least about 500 nucleotides.
  • the inventive dry powder composition comprises a (first) long-chain RNA molecule and may further comprise a second or further RNA molecule, which may also be a long-chain RNA molecule, preferably as defined herein.
  • the second or further RNA molecule comprised in the dry powder composition is distinct from the (first) long-chain RNA molecule.
  • the long-chain RNA molecule comprised in the dry powder composition according to the invention is not an RNA molecule selected from the group consisting of a small interfering RNA (siRNA), a microRNA, a small nuclear RNA (snRNA), a small-hairpin (sh) RNA or riboswitch, a ribozyme, and an aptamer.
  • the long-chain RNA as described herein is not an siRNA, most preferably not a double- stranded siRNA.
  • RNA molecule typically refers to a single-stranded or a double- stranded RNA molecule.
  • the long-chain RNA molecule of the inventive dry powder composition is a single-stranded RNA molecule.
  • the long-chain RNA molecule comprised in the dry powder composition according to the invention is a coding RNA molecule or an immunostimulatory RNA molecule.
  • the long-chain RNA is a coding RNA, which comprises at least one open reading frame encoding a peptide or protein.
  • the long-chain RNA molecule may be a coding RNA molecule encoding 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 adjuvant proteins, from antigens, e.g. tumour antigens, pathogenic antigens (e.g.
  • RNA molecule may be transported into a cell, a tissue or an organism and the protein may be expressed subsequently in this cell, tissue or organism.
  • the long-chain RNA molecule is an mRNA molecule.
  • the long-chain RNA molecule of the dry powder composition may further be an immunostimulatory RNA molecule, such as any RNA molecule known in the art, which is capable of inducing an innate immune response. Particularly preferred in this context are immunostimulatory RNA molecules as described in WO 2009/095226.
  • the dry powder composition may further comprise a modified RNA molecule.
  • the long-chain RNA molecule comprises at least one modification as described herein.
  • the dry powder composition may comprise a second or further RNA molecule (distinct from the (first) long-chain RNA molecule), which comprises at least one modification as described herein.
  • the long-chain RNA molecule of the dry powder composition according to the invention comprises an RNA modification, which preferably increases the stability of the RNA molecule and/or the expression of a protein encoded by the RNA molecule.
  • RNA modifications are known in the art, which can be applied to an RNA molecule in the context of the present invention.
  • RNA modification may refer to chemical modifications comprising backbone modifications as well as sugar modifications or base modifications.
  • a modified RNA molecule as defined herein may contain nucleotide analogues/modifications, e.g. backbone modifications, sugar modifications or base modifications.
  • a backbone modification in connection with the present invention is a modification, in which phosphates of the backbone of the nucleotides contained in an RNA molecule as defined herein are chemically modified.
  • a sugar modification in connection with the present invention is a chemical modification of the sugar of the nucleotides of the RNA molecule as defined herein.
  • a base modification in connection with the present invention is a chemical modification of the base moiety of the nucleotides of the RNA molecule.
  • nucleotide analogues or modifications are preferably selected from nucleotide analogues, which are applicable for transcription and/or translation.
  • modified nucleosides and nucleotides which may be incorporated into a modified RNA molecule as described herein, 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.
  • R H, alkyl, cycloalkyl, aryl,
  • “Deoxy” modifications include hydrogen, amino (e.g. Nhfe; 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 O.
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a modified RNA molecule can include nucleotides containing, for instance, arabinose as the sugar.
  • the phosphate backbone may further be modified in the modified nucleosides and nucleotides, which may be incorporated into a modified RNA molecule as described herein.
  • the phosphate groups of the backbone 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).
  • modified nucleosides and nucleotides which may be incorporated into a modified RNA molecule as described herein can further be modified in the nucleobase moiety.
  • nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine and uracil.
  • 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 nucleotide analogues/modifications are selected from base modifications, which are preferably 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'
  • 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, 1 -taurinomethyl-4-thio- uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-1-methyl-pseudouridine, 2-thio-1- methyl-pseudouridine, 1 -methyl-1 -deaza-pseudouridine, 2-thio
  • 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-1-methyl-pseudoisocytidine, 4-thio-1 -methyl-1 -deaza-pseudoisocytidine, 1 -methyl-1 - deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio- zebularine, 2-thi
  • 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-dimethyl
  • 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, -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
  • a modified nucleoside is 5'-0-(1-Thiophosphate)-Adenosine, 5'- 0-(1 -Thiophosphate)-Cytidine, 5'-0-(1 -Thiophosphate)-Guanosine, 5'-0-(1 - Thiophosphate)-Uridine or 5'-0-(1-Thiophosphate)-Pseudouridine.
  • a modified RNA may comprise nucleoside modifications selected from 6-aza-cytidine, 2-thio-cytidine, a-thio-cytidine, Pseudo-iso-cytidine, 5- aminoallyl-uridine, 5-iodo-uridine, N1-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, N1-methyl-adenosine, 2-amino-6-Chloro-purine, N6- methyl-2-amino-purine, Pseudo-
  • a modified RNA molecule as defined herein can contain a lipid modification.
  • a Iipid-modified RNA molecule typically comprises an RNA as defined herein.
  • Such a Iipid-modified RNA molecule as defined herein typically further comprises at least one linker covalently linked with that RNA molecule, and at least one lipid covalently linked with the respective linker.
  • the Iipid-modified RNA molecule comprises at least one RNA molecule as defined herein and at least one (bifunctional) lipid covalently linked (without a linker) with that RNA molecule.
  • the Iipid-modified RNA molecule comprises an RNA molecule as defined herein, at least one linker covalently linked with that RNA molecule, 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 RNA molecule.
  • the lipid modification is present at the terminal ends of a linear RNA sequence.
  • RNA molecule as defined herein can be modified by the addition of a so-called "5' CAP" structure.
  • a 5'-cap is an entity, typically a modified nucleotide entity, which generally "caps" the 5'- end of a mature mRNA.
  • 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 mRNA transcribed by polymerase II and is therefore not considered as modification comprised in a modified RNA in this context. Accordingly, a modified RNA of the present invention may comprise a m7GpppN as 5'-CAP, but additionally the modified RNA 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 nucle
  • modified 5 -CAP structures are CAP1 (methylation of the ribose of the adjacent nucleotide of m7G), CAP2 (methylation of the ribose of the 2nd nucleotide downstream of the m7G), CAP3 (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.
  • the inventive dry powder composition comprises a modified RNA molecule having at least one open reading frame, which encodes at least one peptide or protein.
  • Said modified RNA molecule having at least one open reading frame may be the (first) long-chain RNA molecule, preferably a long-chain mRNA molecule, or a second or further RNA molecule, which may be comprised in the dry powder composition in addition to the (first) long-chain RNA molecule.
  • the sequence of the open reading frame in such an RNA molecule is modified as described herein.
  • the G/C content of the coding region of a modified RNA comprised in the inventive dry powder composition is modified, particularly increased, compared to the G/C content of its particular wild type coding region, i.e. the unmodified coding region.
  • the encoded amino acid sequence of the coding region is preferably not modified compared to the coded amino acid sequence of the particular wild type coding region.
  • the modification of the G/C-content of the coding region of the modified RNA as defined herein is based on the fact that the sequence of any mRNA region to be translated is important for efficient translation of that mRNA. Thus, the composition and the sequence of various nucleotides are important.
  • mRNA sequences having an increased G (guanosine)/C (cytosine) content are more stable than mRNA sequences having an increased A (adenosine)/U (uracil) content.
  • the codons of the coding region are therefore varied compared to its wild type coding region, 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).
  • RNA sequence e.g. the coding region, compared to its wild type coding region.
  • amino acids which are encoded by codons, which contain exclusively G or C nucleotides
  • no modification of the codon is necessary.
  • the codons for Pro CCC or CCG
  • Arg CGC or CGG
  • Ala GCC or GCG
  • 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. Examples of these are: the codons for Pro can be modified from CCU or CCA to CCC or CCG; the codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG; the codons for Ala can be modified from GCU or GCA to GCC or GCG; the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • the codons for 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 lie 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
  • the G/C content of the coding region of the modified RNA as defined herein is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the wild type coding region.
  • 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 coding region encoding at least one peptide or protein, which comprises a pathogenic antigen or a fragment, variant or derivative thereof, are substituted, thereby increasing the G/C content of said coding region.
  • a further preferred modification of the coding region encoding at least one peptide or protein of a modified RNA as defined herein is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the mRNA is translated to a significantly poorer degree than in the case where codons coding for relatively "frequent" tRNAs are present.
  • the coding region of the modified RNA is preferably modified compared to the corresponding wild type coding region 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 coding region of the modified RNA as defined herein is modified such that codons, for which frequently occurring tRNAs are available, are inserted.
  • codons of the wild type coding region 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.
  • the sequential G/C content which is increased, in particular maximized, in the coding region of the modified RNA as defined herein, with the "frequent" codons without modifying the amino acid sequence of the peptide or protein encoded by the coding region of the RNA sequence.
  • This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) RNA sequence as defined herein.
  • the long-chain RNA molecule may also comprise a 5'- and/or 3' untranslated region (5'-UTR or 3'-UTR, respectively). More preferably, the long-chain RNA molecule comprises a 5'-CAP structure. Preferably, the long-chain RNA molecule further comprises a poly(A) sequence. The length of the poly(A) sequence may vary.
  • the poly(A) sequence may have a length of about 20 adenine nucleotides up to about 300 adenine nucleotides, preferably of about 40 to about 200 adenine nucleotides, more preferably from about 50 to about 100 adenine nucleotides, such as about 60, 70, 80, 90 or 100 adenine nucleotides.
  • the long-chain RNA molecule comprises a poly(A) sequence of about 60 to about 70 nucleotides, most preferably 64 adenine nucleotides.
  • the poly(A) sequence in the long-chain RNA molecule is derived from a DNA template by 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.
  • the long-chain RNA molecule optionally comprises a polyadenylation signal, which is defined herein as a signal, which conveys polyadenylation to a (transcribed) mRNA 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)).
  • CPSF cleavage and polyadenylation specificity factor
  • CstF cleavage stimulation factor
  • CF I and CF II cleavage factors I and II
  • PAP poly(A) polymerase
  • a consensus polyadenylation signal is preferred 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 long-chain RNA molecule may also comprise a poly(C) sequence, preferably in the region 3' of the coding region of the RNA.
  • a poly(C) sequence is typically a stretch of multiple cytosine nucleotides, typically about 10 to about 200 cytidine nucleotides, preferably about 10 to about 100 cytidine nucleotides, more preferably about 10 to about 70 cytidine nucleotides or even more preferably about 20 to about 50 or even about 20 to about 30 cytidine nucleotides.
  • a poly(C) sequence may preferably be located 3' of the coding region comprised by a nucleic acid.
  • the long- chain RNA molecule comprises a poly(A) sequence and a poly(C) sequence, wherein the poly(C) sequence is located 3' of the poly(A) sequence.
  • the long-chain RNA molecule in the context of the present invention comprises in 5'-to-3'-direction, a 5'-UTR, an open reading frame, preferably a modified open reading frame as defined herein, a 3'-UTR element and a poly(A) or a poly(C) sequence.
  • the inventive dry powder composition may comprise the long-chain RNA molecule as described herein in free form ("naked RNA") or in the form of a complex with another compound, such as a transfection or complexation agent.
  • the long-chain RNA molecule may be present in the dry powder composition in a complex with a cationic or polycationic carrier or compound, which may serve as transfection or complexation agent.
  • the dry powder composition comprises both, the long-chain RNA in free form as well in a complex with a cationic or polycationic carrier or compound.
  • Such a complex of long-chain RNA with a cationic or polycationic carrier or compound may be present in the inventive dry powder composition or in an intermediate product as a nanoparticle, preferably as defined herein.
  • the preparation of RNA complexes with polycationic or cationic compounds is known in the art and is preferably carried out as described in WO2010/037539 or WO2011/026641 , the entire disclosure of which is herewith incorporated by reference.
  • the long-chain RNA molecule in the inventive dry powder composition is preferably complexed by a compound selected from the group of polymers or complexing agents, typically comprising, without being limited thereto, any polymer suitable for the preparation of a pharmaceutical composition, such as minor/major groove binders, nucleic acid binding proteins, lipoplexes, nanoplexes, non-cationic or non-polycationic compounds, such as PLGA, Polyacetate, Polyacrylate, PVA, Dextran, hydroxymethylcellulose, starch, MMP, PVP, heparin, pectin, hyaluronic acid, and derivatives thereof, or cationic or polycationic compound, particularly cationic or polycationic polymers or cationic or polycationic lipids, preferably a cationic or polycationic polymers.
  • a compound selected from the group of polymers or complexing agents typically comprising, without being limited thereto, any polymer suitable for the preparation of a pharmaceutical composition, such as minor/major groove binders
  • such a cationic or polycationic compound is typically selected from any cationic or polycationic compound, suitable for complexing and thereby stabilizing a long- chain RNA molecule as defined herein, e.g. by associating the RNA molecule with the cationic or polycationic compound.
  • the dry powder composition according to the invention comprises the long-chain RNA as described herein formulated together with one or more cationic or polycationic compounds, preferably with cationic or polycationic polymers, cationic or polycationic peptides or proteins, e.g. protamine, cationic or polycationic polysaccharides and/or cationic or polycationic lipids.
  • the long-chain RNA as described herein may be complexed with lipids to form one or more liposomes, lipoplexes, or lipid nanoparticles. Therefore, in one embodiment, the dry powder composition comprises liposomes, lipoplexes, and/or lipid nanoparticles comprising the long-chain RNA.
  • Lipid-based formulations have been increasingly recognized as one of the most promising delivery systems for RNA due to their biocompatibility and their ease of large-scale production. Cationic lipids have been widely studied as synthetic materials for delivery of RNA. After mixing together, nucleic acids are condensed by cationic lipids to form lipid/nucleic acid complexes known as lipoplexes. These lipid complexes are able to protect genetic material from the action of nucleases and to deliver it into cells by interacting with the negatively charged cell membrane. Lipoplexes can be prepared by directly mixing positively charged lipids at physiological pH with negatively charged nucleic acids.
  • liposomes consist of a lipid bilayer that can be composed of cationic, anionic, or neutral (phospho)lipids and cholesterol, which encloses an aqueous core. Both the lipid bilayer and the aqueous space can incorporate hydrophobic or hydrophilic compounds, respectively. Liposome characteristics and behaviour in vivo can be modified by addition of a hydrophilic polymer coating, e.g. polyethylene glycol (PEG), to the liposome surface to confer steric stabilization. Furthermore, liposomes can be used for specific targeting by attaching ligands (e.g., antibodies, peptides, and carbohydrates) to its surface or to the terminal end of the attached PEG chains (Front Pharmacol. 2015 Dec 1 ;6:286).
  • ligands e.g., antibodies, peptides, and carbohydrates
  • Liposomes are colloidal lipid-based and surfactant-based delivery systems composed of a phospholipid bilayer surrounding an aqueous compartment. They may present as spherical vesicles and can range in size from 20 nm to a few microns. Cationic lipid-based liposomes are able to complex with negatively charged nucleic acids via electrostatic interactions, resulting in complexes that offer biocompatibility, low toxicity, and the possibility of the large-scale production required for in vivo clinical applications. Liposomes can fuse with the plasma membrane for uptake; once inside the cell, the liposomes are processed via the endocytic pathway and the genetic material is then released from the endosome/carrier into the cytoplasm.
  • Liposomes have long been perceived as drug delivery vehicles because of their superior biocompatibility, given that liposomes are basically analogs of biological membranes, and can be prepared from both natural and synthetic phospholipids (Int J Nanomedicine. 2014; 9: 1833-1843). Cationic liposomes have been traditionally the most commonly used non-viral delivery systems for oligonucleotides, including plasmid DNA, antisense oligos, and siRNA/small hairpin RNA-shRNA).
  • Cationic lipids such as DOTAP, (1 ,2-dioleoyl-3-trimethylammonium- propane) and DOTMA (N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethyl-ammonium methyl sulfate) can form complexes or lipoplexes with negatively charged nucleic acids to form nanoparticles by electrostatic interaction, providing high in vitro transfection efficiency .
  • neutral lipid-based nanoliposomes for RNA delivery as e.g. neutral 1 ,2- dioleoyl-sn-glycero-3- phosphatidylcholine (DOPC)-based nanoliposomes were developed. (Adv Drug Deliv Rev. 2014 Feb; 66: 110- 16.).
  • the long-chain RNA of the dry powder composition as described herein is complexed with a cationic lipid and/or a neutral lipid and thereby forms liposomes, lipid nanoparticles, lipoplexes or neutral lipid-based nanoliposomes.
  • Particularly preferred complexation agents in this context are cationic or polycationic compounds, including protamine, nucleoline, spermine or spermidine, or other cationic peptides or proteins, such as poly-L-lysine (PLL), poly-arginine, oligoarginines as defined above, such as Arg 7 , Arg 8 , Arg 9 , Arg 7 , H 3 R 9 , R.H 3 , H3R9H3, YSSR9SSY, (RKH) 4 , Y(RKH) 2 R, etc., basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs), PpT620, proline-rich peptides, arg
  • the dry powder composition according to the invention comprises protamin, wherein the long-chain RNA molecule is preferably complexed by protamine.
  • the dry powder composition according to the invention preferably comprises a cationic or polycationic compound in solution and/or in complex with the long-chain RNA molecule.
  • the inventive dry powder composition comprises a cationic or polycationic compound, preferably protamine, and the long-chain RNA molecule at a weight ratio (RNA: protamine, w/w) in a range from 1 :10 to 10:1 , more preferably from 5:1 to 1 :1 , even more preferably from 3:1 to 1 :1.
  • the weight ratio of the long-chain RNA molecule to cationic or polycationic compound, preferably protamine, in the composition is 2:1 (w/w).
  • cationic or polycationic compounds or carriers may be cationic or polycationic peptides or proteins, which preferably comprise or are additionally modified to comprise at least one -SH moiety.
  • a cationic or polycationic carrier is selected from cationic peptides having the following sum formula (I):
  • the cationic or polycationic peptide or protein when defined according to formula ⁇ (Arg)i;(Lys) m ;(His)n;(Om) 0 ;(Xaa)x ⁇ (formula (I)) as shown above and which comprise or are additionally modified to comprise at least one -SH moeity, may be, without being restricted thereto, selected from subformula (la):
  • cationic or polycationic compounds which can be used as transfection or complexation agent may include cationic polysaccharides, for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g.
  • cationic polysaccharides for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g.
  • PEI polyethyleneimine
  • DOTMA [1- (2,3-sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride, DMRIE, di-C14-amidine, DOTIM, SAINT, DC-Choi, BGTC, CTAP, DOPC, DODAP, DOPE: Dioleyl phosphatidylethanol-amine, DOSPA, DODAB, DOIC, DMEPC, DOGS: Dioctadecylamidoglicylspermin, DIMRI: Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide, DOTAP: dioleoyloxy-3-(trimethylammonio)propane, DC-6-14: ⁇ , ⁇ - ditetradecanoyl-N-(a-trimethylammonioacetyl)diethanolamine chloride, CLIP1 : rac-[(2,3- dioctadecyloxypropyl)(2-hydroxyethyl)]-di
  • modified polyaminoacids such as ⁇ -aminoacid- polymers or reversed polyamides, etc.
  • modified polyethylenes such as PVP (poly(N-ethyl- 4-vinylpyridinium bromide)), etc.
  • modified acrylates such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), etc.
  • modified Amidoamines such as pAMAM (poly(amidoamine)), etc., modified polybetaaminoester (PBAE), such as diamine end modified 1 ,4 butanediol diacrylate-co-5-amino-1-pentanol polymers, etc.
  • dendrimers such as polypropylamine dendrimers or pAMAM based dendrimers, etc.
  • polyimine(s) such as PEI: poly(ethyleneimine), poly(propyleneimine), etc.
  • polyallylamine sugar backbone based polymers
  • the long-chain RNA molecule of the inventive dry powder composition is complexed at least partially with a cationic or polycationic compound, preferably a cationic protein or peptide.
  • a cationic or polycationic compound preferably a cationic protein or peptide.
  • the ratio of complexed long-chain RNA to free long-chain RNA is selected from a range of about 5:1 (w/w) to about 1 :10 (w/w), more preferably from a range of about 4:1 (w/w) to about 1 :8 (w/w), even more preferably from a range of about 3:1 (w/w) to about 1 :5 (w/w) or 1 :3 (w/w), and most preferably the ratio of complexed long-chain RNA molecule to free long-chain RNA molecule is selected from a ratio of about 1 :1 (w/w).
  • a particle of the dry powder composition as defined herein may thus comprise a long-chain RNA molecule in free form or complexed by a cationic or polycationic compound.
  • a particle of the inventive dry powder composition as described herein comprises or consists of long-chain RNA complexed by a cationic or polycationic compound, wherein the complex is preferably present as a nanoparticle as defined herein.
  • nanoparticle typically refers to a complex of the long-chain RNA molecule with a complexation agent as defined herein, preferably with a cationic or polycationic compound.
  • the complexed long-chain RNA molecule as described herein upon reconstitution of the dry powder in a suitable solvent, is present in the solvent in the form of nanoparticles.
  • the size of the nanoparticle comprising or consisting of complexed long-chain RNA molecule after reconstitution is preferably from 50 to 500 nm, more preferably from 50 to 200 nm.
  • the particle size of the nanoparticle comprising or consisting of complexed long-chain RNA molecule after reconstitution is from 75 to 180 nm, more preferably from 100 to 150 nm.
  • the dry powder composition as described herein comprises a long-chain RNA, which is preferably not complexed with poly(lactide-co- glycolide) PLGA. More preferably, the dry powder composition as described herein does not comprise PLGA.
  • the dry powder composition as described herein comprises a long-chain RNA, which is preferably not complexed with a compound selected from the group consisting of PLGA, poly-lactide (PLA), polyethylene imine (PEI) or poly-L- lysine (PLL). More preferably, the dry powder composition as described herein does not comprise a compound selected from the group consisting of PLGA, PLA, PEI or PLL.
  • the dry powder composition as described herein comprises a long-chain RNA, which is preferably not complexed with DOTAP. More preferably, the dry powder composition as described herein does not comprise DOTAP. Even more preferably, the dry powder composition as described herein may comprise a long-chain RNA, which is preferably not complexed with a cationic lipid. More preferably, the dry powder composition as described herein does not comprise a cationic lipid.
  • the dry powder composition as described herein comprises a long-chain RNA, which is preferably not complexed with mannitol, trehalose or lactose. More preferably, the dry powder composition as described herein does not comprise mannitol, trehalose or lactose. Even more preferably, the dry powder composition as described herein may comprise a long-chain RNA, which is preferably not complexed with a carbohydrate. More preferably, the dry powder composition as described herein does not comprise a carbohydrate.
  • the long-chain RNA of the dry powder composition as described herein is not comprised in nanoparticles or in liposomes, preferably as defined herein.
  • the dry powder composition may preferably not comprise a nanoparticle or a liposome, preferably as defined herein.
  • the inventive dry powder composition comprising a long-chain RNA molecule comprises at least one further component or excipient.
  • the inventive dry powder composition comprises a solvent, preferably in the amounts as defined herein with respect to the residual moisture content of the dry powder composition.
  • the solvent is a residue of a solvent, which was used during preparation of the dry powder composition, a residue of which may be present in the inventive dry powder composition.
  • the solvent contained in the inventive dry powder composition is a residue of a solvent used during preparation of the dry powder composition by using the inventive method as described herein.
  • the solvent comprised in the dry powder composition according to the invention is suitable for use in spray drying and/or spray-freeze drying.
  • a solvent is comprised in the inventive composition, in which the long-chain RNA and any other component comprised in the composition, if present, are soluble.
  • the solvent is volatile with a boiling point of preferably below 150°C.
  • the solvent is preferably non-toxic.
  • the solvent is an aqueous solution.
  • the solvent is preferably miscible with water.
  • the solvent comprised in the dry powder composition according to the invention comprises an aequeous solution or water, preferably pyrogen-free water or water for injection (WFI).
  • WFI water for injection
  • WFI water for injection
  • WFI Water for Injection
  • WFI is water purified by distillation or reverse osmosis.
  • WFI is typically produced by either distillation or 2-stage reverse osmosis.
  • WFI typically does not contain more than 0.25 USP endotoxin units (EU) per ml.
  • Endotoxins are a class of pyrogens that are components of the cell wall of Gram-negative bacteria (the most common type of bacteria in water), preferably in an action limit of 10 cfu/100 ml.
  • the microbial quality may be tested by membrane filtration of a 100 ml sample and plate count agar at an incubation temperature of 30 to 35 degrees Celsius for a 48-hour period.
  • the chemical purity requirements of WFI are typically the same as of PW (purified water).
  • the dry powder composition according to the invention may comprise a buffer, preferably selected from a buffer as defined herein, e.g. a buffer containing 2- hydroxypropanoic acid, preferably including at least one of its optical isomers L-(+)-lactic acid, (S)-lactic acid, D-(-)-lactic acid or (R)-lactic acid, more preferably its biologically active optical isomer L-(+)-lactic acid, or a salt or an anion thereof, preferably selected from sodium-lactate, potassium-lactate, or Al 3 + -lactate, NH 4 + -lactate, Fe-lactate, Li-lactate, Mg- lactate, Ca-lactate, Mn-lactate or Ag-lactate, or a buffer selected from Ringer's lactate (RiLa), lactated Ringer's solution (main content sodium lactate, also termed "Hartmann's Solution” in the UK), acetated Ringer ' s solution, or ortho-lactate
  • a buffer preferably
  • a buffer as defined herein may also be a mannose containing buffer, an isotonic buffer or solution, preferably selected from isotonic saline, a lactate or ortho-lactate-containing isotonic solution, a isotonic buffer or solution selected from phosphate-buffered saline (PBS), TRIS-buffered saline (TBS), Hank's balanced salt solution (HBSS), Earle's balanced salt solution (EBSS), standard saline citrate (SSC), HEPES-buffered saline (HBS), Grey's balanced salt solution (GBSS), or normal saline (NaCI), hypotonic (saline) solutions with addition of glucose or dextrose, or any solution as defined herein, etc.
  • PBS phosphate-buffered saline
  • TRIS-buffered saline TRIS-buffered saline
  • HBSS Hank's balanced salt solution
  • EBSS Earle's balanced salt
  • a buffer or, in particular, a residue thereof may be comprised in the dry powder composition according to the invention, more preferably an aqueous (isotonic solution or aqueous) buffer, containing a sodium salt, preferably at least 50 mM of a sodium salt, a calcium salt, preferably at least 0.01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt.
  • the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g.
  • examples of sodium salts include e.g. NaCI, Nal, NaBr, Na2C0 3 , NaHCC»3, Na 2 S0 4
  • examples of the optional potassium salts include e.g. KCI, Kl, KBr, K2CO3, KHCO3, K 2 S0 4
  • examples of calcium salts include e.g. CaCI 2 , Cab, CaBr 2 , CaC03, CaS0 4 , Ca(OH) 2 .
  • the salts are present in such a buffer in a concentration of at least 50 mM sodium chloride (NaCI), at least 3 mM potassium chloride (KCI) and at least 0.01 mM calcium chloride (CaCI 2 ).
  • organic anions of the aforementioned cations may be contained in the buffer.
  • the buffer may contain salts selected from sodium chloride (NaCI), calcium chloride (CaCI 2 ) and optionally potassium chloride (KCI), wherein further anions may be present in addition to the chlorides.
  • CaCI 2 may also be replaced therein by another salt like KCI.
  • the inventive dry powder composition may be reconstituted in a solvent or a buffer as defined herein, preferably as defined above.
  • the inventive dry powder composition may be reconstituted in water, Ringer Lactate solution, a buffer as defined above, or a buffer containing mannose, to obtain the desired salt concentration or alternatively the desired buffer conditions.
  • the reconstitution of the dry powder composition is carried out in WFI (water for injection), if the dry powder composition was prepared from a long-chain RNA molecule dissolved in Ringer Lactate solution (optionally comprising further components), which represents an isotonic solution for injection.
  • the dry powder composition is reconstituted in an isotonic solution, preferably as defined herein, more preferably in Ringer Lactate, especially if the dry powder composition was prepared from a long-chain RNA molecule dissolved in water, preferably WFI (wherein the water optionally comprises further components).
  • the dry powder composition according to the invention does not comprise a lipid compound.
  • the inventive dry powder composition may further comprise any type of suitable component, which is compatible with the long-chain RNA molecule.
  • the term 'component' preferably comprises any additive or excipient, preferably a pharmaceutically acceptable excipient that does preferably not cause or enhance degradation of the long-chain RNA molecule.
  • Such a component may further be in any state, such as liquid, gel-like, solid or semi-solid.
  • a component is preferably selected from the group consisting of cryoprotectants, lyoprotectants, bulking agents, preservatives, antioxidants, antimicrobial agents, colorants, carriers, fillers, film formers, redispersants and disintegrants.
  • the inventive dry powder composition may also comprise excipients, such as defoamers, surfactants, viscosity enhancing agents, force control agents or the like.
  • the inventive dry powder composition comprises at least one component selected from a cryoprotectant, a lyoprotectant or a bulking agent.
  • cryoprotectants are understood as excipients, which allow influencing the structure of a frozen material and/or the eutectical temperature of the mixture.
  • Lyoprotectants are typically excipients, which partially or totally replace the hydration sphere around a molecule and thus prevent catalytic and hydrolytic processes.
  • a bulking agent e.g. a filler
  • a bulking agent is any excipient compatible with the long-chain RNA molecule, which may be comprised in the inventive composition.
  • a bulking agent may be used for increasing the volume and/or the mass of the inventive composition.
  • a bulking agent may also protect the long-chain RNA molecule from degradation.
  • the inventive dry powder composition may additionally contain at least one suspending agent, preferably mannit.
  • the inventive dry powder composition may additionally contain at least one component selected, e.g., from proteins, amino acids, alcohols, carbohydrates, mannose, mannit, metals or metal ions, surfactants, polymers or complexing agents, buffers, etc., or a combination thereof.
  • one preferred component may also be selected from the group of amino acids.
  • group may comprise, without being limited thereto, any naturally occurring amino acid, including alanine, cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, pyrrolysine, proline, glutamine, arginine, serine, threonine, selenocysteine, valine, tryptophan, and tyrosine, more preferably glycine, arginine, and alanine.
  • Cryoprotectants and/or lyoprotectants selected from the group of amino acids may additionally comprise any modification of a naturally occurring amino acid as defined above.
  • a further component may be selected from the group of alcohols.
  • group may comprise, without being limited thereto, any alcohol suitable for the preparation of a pharmaceutical composition, preferably, without being limited thereto, mannitol, polyethyleneglycol, polypropyleneglycol, sorbitol, etc.
  • a further component may be selected from the group of (free) carbohydrates.
  • a carbohydrate such as a sugar
  • group of (free) carbohydrates may comprise, without being limited thereto, any (free) carbohydrate, suitable for the preparation of a pharmaceutical composition, preferably, without being limited thereto, (free) monosaccharides, such as e.g.
  • free glucose preferably glucose, (free) fructose, (free) galactose, (free) sorbose, (free) mannose
  • free preferably means unbound or unconjugated, e.g. the mannose is not covalently bound to the long-chain RNA molecule, or in other words, the mannose is unconjugated, preferably with respect to the long-chain RNA molecule), etc., and mixtures thereof; disaccharides, such as e.g.
  • sugars that are preferably used in the composition according to the invention include lactose, sucrose or trehalose.
  • a sugar that is preferred in this context has a high water displacement activity and a high glass transition temperature.
  • a sugar suitable for use in the composition is preferably hydrophilic but not hygroscopic.
  • the sugar preferably has a low tendency to crystallize, such as trehalose. Trehalose is particularly preferred.
  • the dry powder composition may comprise a cryoprotectant, which is preferably not selected from lactose or trehalose. More preferably, the cryoprotectant is not a carbohydrate.
  • the weight ratio of the long-chain RNA molecule in the composition to the carbohydrate component, preferably a sugar, more preferably trehalose, in the composition is preferably in the range from about 1 :2.000 to about 1 :10, more preferably from about 1 :1 ,000 to about 1 :100.
  • the weight ratio of the long-chain RNA molecule in the composition to the carbohydrate excipient, preferably a sugar, more preferably trehalose, in the composition is in the range from about 1 :250 to about 1 :10 and more preferably in the range from about 1 : 100 to about 1 :10 and most preferably in the range from about 1 : 100 to about 1 :50.
  • the dry powder composition according to the present invention comprises at least 50 % (w/w), preferably at least 70% (w/w), at least 80% (w/w), at least 90% (w/w), or at least 95% (w/w) of a carbohydrate component, preferably a sugar, more preferably trehalose.
  • the inventive dry powder composition comprises trehalose. More preferably, trehalose is present in the inventive dry powder composition in a relative amout of about 5% to about 99.5% (w/w), preferably in a relative amount of about 20% to about 98% (w/w), more preferably in a relative amount of about 50% to about 95% (w/w), even more preferably in a relative amount of about 70 to about 99% (w/w), and most preferably in a relative amount of about 75 to about 90% (w/w).
  • the relative amount of trehalose in the inventive dry powder composition is at least 30% (w/w), at least 40% (w/w), at least 50% (w/w), at least 60% (w/w), at least 70% (w/w), at least 80% (w/w), at least 90% (w/w) or at least 95% (w/w).
  • a further suitable component may also be selected from the group of proteins.
  • Such group may comprise, without being limited thereto, proteins such as albumin, gelatine, therapeutically active proteins, antibodies, antigens, or any further protein encoded by the long-chain RNA molecule as defined herein.
  • a component, which may be contained in the inventive dry powder composition may be selected from the group of metals or metal ions, typically comprising, without being limited thereto, metals or metal ions or salts selected from
  • alkali metals including members of group 1 of the periodic table: lithium (Li), sodium (Na), potassium (K), rubidium (Rb), caesium (Cs), and francium (Fr), and their (monovalent) metal alkali metal ions and salts; preferably lithium (Li), sodium (Na), potassium (K), and their (monovalent) metal alkali metal ions and salts;
  • alkaline earth metals including members of group 2 of the periodic table: beryllium (Be), magnesium (Mg), calcium (Ca), strontium (Sr), barium (Ba) and radium (Ra), and their (divalent) alkaline earth metal ions and salts; preferably magnesium (Mg), calcium (Ca), strontium (Sr), barium (Ba) and their (divalent) alkaline earth metal ions and salts;
  • transition metals including members of groups 3 to 13 of the periodic table and their metal ions and salts;.
  • the transition metals typically comprise the 40 chemical elements 21 to 30, 39 to 48, 71 to 80, and 103 to 112.
  • the name transition originates from their position in the periodic table of elements. In each of the four periods in which they occur, these elements represent the successive addition of electrons to the d atomic orbitals of the atoms. In this way, the transition metals represent the transition between subgroup 2 elements and subgroup 12 (or 13) elements.
  • Transition metals in the context of the present invention particularly comprise members of subgroup 3 of the periodic table: including Scandium (Sc), Yttrium (Y), and Lutetium (Lu), members of subgroup 4 of the periodic table: including Titan (Ti), Zirconium (Zr), and Hafnium (Hf), members of subgroup 5 of the periodic table: including Vanadium (V), Niobium (Nb), and Tantalum (Ta), members of subgroup 6 of the periodic table: including Chrome (Cr), Molybdenum (Mo), and Tungsten (W), members of subgroup 7 of the periodic table: including Manganese (Mn), Technetium (Tc), and Rhenium (Re), members of subgroup 8 of the periodic table: including Iron (Fe), Ruthenium (Ru), and Osmium (Os), members of subgroup 9 of the periodic table: including Cobalt (Co), Rhodium (Rh), and Iridium (Ir), members of subgroup 10 of the periodic table: including Nickel (Ni
  • earth metals or members of the boron group including members of group 3 of the periodic table: including Boron (B), Aluminium (Al), Gallium (Ga), Indium (In) and Thallium (TI) and their metal ions and salts; preferably Boron (B) and Aluminium (Al) and their metal ions and salts;
  • metalloids or semi metals including Boron (B), Silicon (Si), Germanium (Ge), Arsenic (As), Antimony (Sb), Tellurium (Te).and Polonium (Po), and their semi metal ions and salts; preferably Boron (B) and Silicon (Si) and their semi metal ions and salts;
  • a further component may be selected from the group of surfactants may comprise, without being limited thereto, any surfactant, preferably any pharmaceutically acceptable surfactant, which is preferably suitable for spray drying or spray-freeze drying. More preferably, without being limited thereto, the surfactant is selected from the group consisting of Tween, e.g. Tween 80 (0.2%), Pluronics, e.g. Pluronic L121 (1.25%), Triton-X, SDS, PEG, LTAB, Saponin, Cholate, etc.
  • the inventive dry powder composition may additionally contain one or more compatible solid or liquid fillers or diluents or encapsulating compounds, which are preferably suitable for administration to a patient to be treated.
  • compatible means that these constituents are capable of being mixed with the long-chain RNA molecule (free or in a complex with a cationic or polycationic compound), as defined according to the present invention, in such a manner that no interaction occurs, which would substantially reduce the integrity or biological activity of the long-chain RNA molecule, 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 and sucrose; starches, such as, for example, corn starch or potato starch; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium 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.
  • sugars such as, for example, lactose, glucose and sucrose
  • starches such as, for example, corn
  • the dry powder composition according to the invention may optionally contain further excipients or agents, such as stabilizers, for example EDTA, Tween, benzoic acid derivatives or RNAse inhibitors.
  • the dry powder composition may further comprise any type of component or additive, which is compatible with the long-chain RNA molecule.
  • Such an excipient is preferably selected from the group consisting of preservatives, antioxidants, antimicrobial agents, colorants, carriers, fillers, film formers, redispersants and disintegrants.
  • the dry powder composition may also comprise a component or additive, preferably in very small amounts, that were added during the manufacturing process, such as defoamers, surfactants, viscosity enhancing agents, force control agents or the like.
  • the dry powder composition of the invention is obtained by the method as described herein.
  • the dry powder composition according to the invention is particularly suitable as storage-stable form of a long-chain RNA molecule.
  • the inventors have surprisingly found that the storage stability of the long-chain RNA molecule in the dry powder composition is excellent and the long-chain RNA molecule remains functional after extended storage periods.
  • the storage stability of the long-chain RNA molecule is typically determined through determination of the relative (structural) integrity and the biological activity after a given storage period, e.g. via time-course in vitro expression studies.
  • the relative integrity is preferably determined as the percentage of full-length RNA (i.e. non-degraded long-chain RNA) with respect to the total amount of RNA (i.e. long-chain RNA and degraded RNA fragments (which appear as smears in gel electrophoresis)), preferably after deduction of the LOD (3 x background noise), for example, by using the software QuantityOne from BioRad.
  • the dry powder composition according to the invention thus provides the advantageous characteristics of a powder and the potential of such a composition for, e.g. packaging and dosage, while it also allows significantly longer storage at temperatures from -80°C to 60°C than the corresponding RNAs in WFI or other injectable solutions.
  • the dry powder composition can be stored at room temperature, which simplifies shipping and storage.
  • the dry powder composition is stored with or without shielding gas.
  • single doses of the dry powder composition are packaged and sealed.
  • multiple doses can be packaged in one packaging unit.
  • Single dose packaging in blisters or capsules is preferably used in order to prevent cross-contamination.
  • the relative integrity is at least 70%, more preferably at least 75%, at least 80%, at least 85%, at least 90% or at least 95% after storage at room temperature for preferably at least one week, more preferably for at least one month, even more preferably for at least 6 months and most preferably for at least one year.
  • the biological activity of the long-chain RNA molecule of the dry powder composition after storage at room temperature, preferably as defined above with respect to the relative integrity of the long-chain RNA molecule is preferably at least 70%, more preferably at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the biological activity of the freshly prepared long-chain RNA molecule.
  • the biological activity is preferably determined by analysis of the amounts of protein expressed from reconstituted RNA and from freshly prepared RNA, respectively, e.g. after transfection into a mammalian cell line. Alternatively, the biological activity may be determined by measuring the induction of an (adaptive or innate) immune response in a subject.
  • a method that allows preparing a storage form of a long-chain RNA molecule, preferably a long-chain RNA in particulate form as described herein, wherein a liquid comprising the RNA molecule is provided and wherein the liquid is dried by spray-drying or spray-freeze drying.
  • the invention concerns a method for drying a liquid comprising a long-chain RNA molecule.
  • inventive dry powder composition may likewise apply to the inventive method.
  • description of the long-chain RNA molecule and further components of the dry powder composition apply to the inventive method as well.
  • further definitions may apply to the inventive method as specifically indicated in the following.
  • the invention concerns a method for preparing a dry powder comprising a long-chain RNA molecule, wherein the method comprises the following steps: a) providing a liquid comprising the long-chain RNA molecule, b) drying the liquid provided in step a) by spray-drying or spray-freeze drying.
  • a storage form of a long-chain RNA molecule may be obtained by the method as described herein.
  • the inventors found that - by using the method according to the invention - a dry powder comprising a long-chain RNA molecule can be obtained.
  • the method according to the invention is suitable for application at an industrial scale.
  • the invention provides a method that can be carried out by the skilled person using standard equipment, thus providing a cost- and time- effective solution.
  • the method can be carried out in bulk as well as continuously.
  • the storage form, preferably the long-chain RNA in particulate form, obtained by using the method according to the invention therefore represents an effective means for extending the stability of long-chain RNA as an API (active pharmaceutical ingredient), especially during storage at a variety of different temperatures and in different packaging formats.
  • a liquid is provided that comprises a long-chain RNA molecule.
  • the long-chain RNA molecule comprised in the liquid provided in step a) of the inventive method is characterized by any feature or any combination of features described herein with respect to the long-chain RNA molecule that is comprised in the inventive dry powder composition.
  • the liquid in step a) of the method according to the invention is provided by diluting or dissolving the long-chain RNA molecule in a suitable solvent.
  • the solvent is preferably a solvent suitable for use in spray drying and/or spray-freeze drying.
  • a solvent is used, in which the long-chain RNA and any other component, if present, are soluble. Suitable solvents are described above with respect to the inventive dry powder composition.
  • the liquid in step a) of the inventive method preferably comprises the long- chain RNA molecule and a solvent or buffer as described above with respect to the inventive dry powder composition.
  • step a) of the inventive method comprises dissolving or diluting the long-chain RNA molecule as defined herein in a solvent or buffer as defined herein, preferably in an aqueous solution, such as Ringer Lactate, or water, more preferably pyrogen-free water or WFI.
  • a solvent or buffer as defined herein, preferably in an aqueous solution, such as Ringer Lactate, or water, more preferably pyrogen-free water or WFI.
  • the liquid comprising the long-chain RNA molecule comprises at least one further component, preferably as described herein with respect to the dry powder composition disclosed herein.
  • the liquid provided in step a) of the inventive method preferably comprises a further component selected from the group consisting of buffers, cryoprotectants, lyoprotectants, bulking agents, suspending agents, proteins, amino acids, alcohols, carbohydrates, metals, metal ions, salts, surfactants, fillers, diluents, carriers, glidants, vegetable oils, polyols, encapsulating compounds, stabilizers, preservatives, antioxidants, antimicrobial agents, colorants, film formers, redispersants, disintegrants, defoamers, viscosity enhancing agents and force control agents, wherein the respective component is preferably as defined above with respect to the inventive dry powder composition.
  • the long-chain RNA molecule as defined herein may be present in the liquid provided in step a) of the inventive method in free form (as "naked RNA") and/or as a complex with a polycationic or cationic compound, preferably as described herein.
  • the long-chain RNA molecule as defined herein and a cationic or polycationic compound may be comprised in the liquid provided in step a), either as a complex, preferably in the form of a nanoparticle as defined herein, or both in free form, i.e. in solution without being in a complex with each other.
  • RNA complexes with complexation agents preferably with polycationic or cationic compounds
  • RNA complexation agents preferably with polycationic or cationic compounds
  • the liquid provided in step a) of the inventive method comprises a complexation agent, preferably as defined herein, more preferably a cationic or polycationic compound as defined herein, such as protamine, nucleoline, spermin, spermidine, oligoarginines as defined above, such as Arg 7 , Args, Argg, Arg 7 , H3R9, R9H3, H3R9H3, YSSRgSSY, (RKH) 4 , Y(RKH) 2 R, etc.
  • a complexation agent preferably as defined herein, more preferably a cationic or polycationic compound as defined herein, such as protamine, nucleoline, spermin, spermidine, oligoarginines as defined above, such as Arg 7 , Args, Argg, Arg 7 , H3R9, R9H3, H3R9H3, YSSRgSSY, (RKH) 4 , Y(RKH
  • the complexation agent preferably a cationic or polycationic compound as defined herein, is preferably present in the liquid provided in step a) in free form (in solution) or in a complex with the long-chain RNA molecule.
  • Protamine is particularly preferred and is preferably comprised in the liquid provided in step a) of the method at a concentration in a range from 0.01 g/l to 10 g/l, from 0.05 g/l to 5 g/l, or from 0.05 g/l to 2 g/l. More preferably, the concentration of protamine in the liquid provided in step a) of the method is in a range from 0.05 g/l to 3 g/l or from 0.1 to 1 g/l.
  • the liquid provided in step a) further comprises lactate, wherein the lactate concentration is preferably in the range of about 3 mM to about 300 mM, preferably in the range of about 5 mM to about 200 mM, more preferably in the range of about 10 mM to about 150 mM, even more preferably about 15 mM to about 35 mM, and most preferably 20 mM to about 31 mM.
  • the liquid provided in step a) of the method typically comprises a Ringer's lactate concentration (or a concentration of any of the afore mentioned lactate containing solutions) e.g. in the range of about 10% (w/w) to about 100% (w/w), e.g.
  • Ringer's lactate (100 % (w/w)) is typically defined as a solution comprising 131 mM Na + , 5,36 mM K + , 1 ,84 mM Ca 2+ , and 28,3 mM Lactate).
  • the liquid provided in step a) of the inventive method does not comprise a lipid compound.
  • the liquid provided in step a) of the method may additionally contain at least one suspending agent, preferably mannit, preferably in a concentration of about 1 to 15% (w/w), more preferably in a concentration of about 3 to 10% (w/w), and even more preferably in a concentration of about 4 to 6% (w/w).
  • at least one suspending agent preferably mannit, preferably in a concentration of about 1 to 15% (w/w), more preferably in a concentration of about 3 to 10% (w/w), and even more preferably in a concentration of about 4 to 6% (w/w).
  • the liquid provided in step a) of the method comprises a carbohydrate component, preferably a sugar, more preferably trehalose.
  • a carbohydrate component, preferably a sugar, more preferably trehalose is present in the liquid provided in step a) of the method at a concentration of about 0.01 to about 20% (w/w), preferably in a concentration of about 0.01 to about 15% (w/w), more preferably in a concentration of about 0.1 to about 10% (w/w), even more preferably in a concentration of about 0.5 to about 10% (w/w), and most preferably in a concentration of about 2.5 to about 7.5% (w/w), e.g. at a concentration of about 4 to about 7% (w/w), such as about 5 % (w/w).
  • the pH of the liquid provided in step a) of the method may be in the range of about 4 to 8, preferably in the range of about 6 to about 8, more preferably from about 7 to about 8.
  • the liquid provided in step a) of the method contains the herein defined contents, optional components, additives, etc. in such a concentration so as to lead to an osmolarity comparable to that of blood plasma.
  • osmolarity is typically to be understood as a measure of all contents, optional components, additives, etc. of the liquid as defined herein. More precisely, osmolarity is typically the measure of solute concentration, defined as the number of osmoles (Osm) of all solubilized contents, optional components, additives, etc. per liter (I) of solution (osmol/l or osm/l).
  • the liquid provided in step a) of the method may comprise an osmolarity preferably in the range of about 200 mosmol/l to about 400 mosmol/l, more preferably in the range of about 250 mosmol/l to about 350 mosmol/l, even more preferably in the range of about 270 mosmol/l to about 330 mosmol/l or in the range of about 280 mosmol/l to about 320 mosmol/l, or in the range of about e.g. about 290 mosmol/l to about 310 mosmol/l, e.g.
  • the method according to the present invention further comprises a step b), wherein the liquid provided in step a) of the method is dried by spray-drying or by spray-freeze drying.
  • the term 'spray-drying' typically relates to a process that involves breaking up a liquid into small droplets (atomization) and rapidly removing solvent from the droplets in a spray-drying apparatus, where there is a strong driving force for evaporation of solvent from the droplets, which provide a favourable surface to mass ratio.
  • the strong driving force for solvent evaporation is generally provided by a high surface to mass ratio of the droplets and by maintaining the partial pressure of solvent in the spray-drying appartus well below the vapor pressure of the solvent at the temperature of the drying droplets. This may be achieved, for example, by maintaining the pressure in the spray-drying apparatus at a partial vacuum or by mixing the droplets with a warm drying gas or a combination of both.
  • particles preferably dry particles, more preferably in the form of a dry powder composition, are typically obtained.
  • the term 'spray-freeze drying' relates to a spray-drying process, preferably as defined herein, wherein the droplets are typically frozen after droplet formation by contacting the droplets with a coolant, such as a cooling gas or a cooling liquid. The solvent subsequently sublimes from the frozen droplets. Accordingly, particles, preferably dry particles, are obtained as a result of the process.
  • the spray-freeze drying process is preferably characterized by any of the features of the spray-drying process as described herein. For instance, the features concerning the atomization or the apparatus in general may likewise apply to the spray-drying process. Spray-freeze drying processes were described in the prior art, such as US 7,007,406.
  • the method according to the invention may be carried out in bulk or as a continuous process.
  • the method is carried out as a continuous process.
  • the spray-drying process or the spray-freeze drying process may be carried out in bulk or as a continuous process in the context of the inventive method.
  • the spray-drying process or the spray-freeze drying process is carried out in a continuous process.
  • the liquid provided in step a) of the method is used as liquid feed in a spray- drying process or in a spray-freeze drying process.
  • the liquid feed is supplied at a rate of preferably at least 200 g/h, more preferably at least 300 g/h or even more preferably at least 500 g/h.
  • the feed rate is in a range from 200 g/h to 1 ,000 g/h, from 300 g/h to 800 g/h or from 300 g/h to 600 g/h.
  • the liquid comprising the long-chain RNA molecule which is provided in step a) is first broken up into a plurality of small droplets that are preferably suspended in a gas or a gas mixture, such as air.
  • a gas or a gas mixture such as air.
  • the obtained mixture of droplets and gas is typically referred to as 'spray' or 'fog'.
  • the process of breaking up the liquid feed into droplets is known as 'atomization' and may be carried out using any suitable device known in the art (atomizer).
  • atomizers are known in the art, which are suitable for being used in the inventive method, such as rotary atomizers, pressure nozzles, two-fluid nozzles, fountain nozzles, ultrasonic nebulizers and vibrating orifice aerosol generators.
  • a rotary atomizer is used as atomizer in the spray-drying or spray- freeze drying process.
  • Rotary atomizers exploit the energy of high-speed rotation to produce fine droplets.
  • the liquid feed is introduced into a reservoir, typically in the center of the rotary wheel.
  • a two-fluid nozzle is used in the spray-drying process or the spray-freeze drying process.
  • Two-fluid nozzles combine two fluids, where one fluid is typically the liquid feed to be dried and the second fluid is typically a compressed gas (e.g. air, nitrogen or C0 2 at, for example, 0.1 to 7 bar).
  • the energy of the compressed gas is used to atomize the liquid feed.
  • the produced spray droplets are usually mixed with a drying gas stream, allowing the liquid to quickly evaporate. The rapid evaporation typically results in a cooling effect, so that the dried particles do not reach the drying air temperature, which is particularly advantageous if heat sensitive material is dried.
  • a pressure nozzle is used as atomizer.
  • a pressure nozzle is used, which comprises a swirl chamber, causing the liquid passing through them to rotate.
  • a pressure nozzle is used as atomizer, wherein the nozzle pressure is preferably not higher than about 1 bar, more preferably not higher than about 0.7 bar, not higher than about 0.5 bar or not higher than about 0.3 bar.
  • the nozzle pressure is in a range from about 1 to about 0.1 bar, more preferably in a range from about 0.7 to about 0.3 bar. In a particularly preferred embodiment, the nozzle pressure is not higher than about about 0.3 bar.
  • an atomizer is preferably used that produces droplets, which are preferably characterized by a mass median aerodynamic diameter (MMAD), preferably as defined herein, of at least 0.3 ⁇ .
  • MMAD mass median aerodynamic diameter
  • the MMAD of the droplets according to the invention is at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or 50 m.
  • the MMAD of the droplets is equal to or less than 1 ,500, 1 ,250, 1 ,000, 750, 600, 500, 400, 300, 200 or 100 Mm.
  • the MMAD of the droplets may be in a range from 0.5 ⁇ to 2,000 ⁇ , from 1 m to 1 ,000 ⁇ , from 2 Mm to 500 ⁇ or from 2 ⁇ to 200 ⁇ . In a preferred embodiment, the MMAD of the droplets is at least 1 ⁇ or in the range from 1 to 200 ⁇ . In a particularly preferred embodiment, the MMAD of the droplets is at least 3 ⁇ , at least 5 ⁇ or at least 20 Mm.
  • the droplet size distribution is narrow, i.e. the size of the individual droplets that are formed by the atomizer is relatively uniform. More preferably, the droplets formed by the atomizer are characterized by using the span of the droplet size distribution as a parameter. Therein, the span (for a volume weighted distribution) is defined as outlined above with respect to the particle size of the inventive dry powder composition. In a preferred embodiment, the droplet size distribution is characterized by a low span value, which preferably results in a narrow particle size distribution in the dry powder composition. Typically, a narrow droplet size distribution after atomization results in increased flowability of the resulting dry powder.
  • the span of the droplets formed by the atomizer is equal to or less than 5, more preferably equal to or less than 4, and even more preferably equal to or less than 3.
  • the particle size distribution of the dry powder composition according to the invention is characterized by a span of less than about 2 or less than about 1.5.
  • atomization of the liquid feed results in spherical droplets.
  • spherical comprises not only geometrically perfect spheres, but also more irregular shapes, such as spheroidal, elipsoid, oval or rounded droplet.
  • Waddell's sphericity ⁇ (herein also referred to as “sphericity” or “circularity”) may be calculated, e.g. by using the following equation
  • a droplet formed by atomization of the liquid feed is characterized by a sphericity of at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the atomizer generates a plurality of droplets comprising at least one droplet with a sphericity in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the average sphericity of the droplets, which are formed by the atomizer is at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the average sphericity of the droplets, which are formed by the atomizer is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the sphericity of those droplets that have a particle size (i.e. droplet size) equal to Dv50 in the drople size distribution as defined herein is at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the sphericity of those droplets that have a particle size equal to Dv50 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • those droplets that have a particle size equal to Dv90 as defined herein have a sphericity of at least 0.7, preferably of at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the sphericity of those droplets that have a particle size equal to Dv90 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the average sphericity of those droplets that have a particle size equal to or lower than Dv50 as defined herein is at least 0.7, preferably at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the average sphericity of those droplets that have a particle size equal to or lower than Dv50 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the average sphericity of those droplets that have a particle size equal to or lower than Dv90 as defined herein is at least 0.7, preferably of at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.95 or 1.
  • the average sphericity of those droplets that have a particle size equal to or lower than Dv90 as defined herein is in the range from 0.7 to 1 , more preferably in the range from 0.8 to 1 , from 0.85 to 1 , or from 0.9 to 1.
  • the droplets formed by the atomizer are dried by evaporation of the solvent from the droplets.
  • a drying chamber which may be of any shape and which may consist of one or more chambers.
  • the droplets are contacted with a gas stream that is preferably capable of absorbing, at least partially, the solvent that evaporates from the droplets.
  • the gas stream is preferably introduced into the drying chamber via an inlet, such as a disperser, which is preferably located in the upper half of the drying chamber, more preferably in the vicinity of the atomizer, thus allowing rapid mixing of the drying gas and the droplets.
  • the gas stream leaves the drying chamber through an outlet, which is preferably located at the bottom of the drying chamber.
  • the drying chamber comprises a cone-shaped part, wherein the tip of the cone comprises the outlet, preferably for the drying gas as well as for dried particles.
  • the characteristics of the drying chamber are matched with, amongst others, the atomizer that is used.
  • the droplets preferably contact a surface only when they are sufficiently dry.
  • Spray-drying devices that use centrifugal atomizer typically require relatively larger diameter vessels, but less cylinder height for optimal drying.
  • Spray-drying devices that use pressure nozzles usually require relatively small diameters with larger cylinder height for sufficient drying.
  • the dry powder composition is preferably collected at the bottom of the drying chamber that is preferably designed as a cone. In the center of the cone area, the outlet of the gas-stream is preferably positioned, where cool and moist air is removed from the drying chamber.
  • Such a design of the cone and outlet is acting as a cyclone separator and leads to an accumulation of the dry powder composition at the bottom of the drying chamber.
  • Cyclonic separation is preferably used to separate dry particles or fine droplets from the drying gas, preferably without the use of filters, through vortex separation.
  • a high speed rotating flow is preferably established within a cylindrical or conical container, the cyclone.
  • drying gas flows in a helical pattern, from the top (wide end) of the cyclone to the bottom (narrow) end before exiting the cyclone in a straight stream through the center of the cyclone and out the top.
  • a filter e.g. a bag filter or a combination of a cyclone separator and a filter may be used for separation.
  • a filter e.g. a bag filter or a combination of a cyclone separator and a filter may be used for separation.
  • the spray-drying or spray-freeze drying device is set up as a co-current flow device (spray and drying gas move into the same directions), as a counter-current flow device (spray and drying gas move into opposite directions) or as a mixed flow device (co-current and counter-current flow combined).
  • the spray-drying or spray-freeze drying device is a co-current flow device.
  • common spray-drying or spray-freeze drying devices may be categorized depending on the type of air cycle that is used.
  • a spray-drying or spray-freeze drying device as used herein is preferably an open cycle device (drying air that enters the system through the inlet is exhausted through the outlet into the atmosphere) or a closed cycle device (drying air that enters the system through the inlet, exits the system via the outlet and is reused).
  • a closed cycle device is preferably employed, if a drying gas is used that should not accumulate in the facility, where the device is located (such as nitrogen or C0 2 ), and/or if toxic substances are released from the drying material.
  • the spray-drying or spray-freeze drying device preferably reduces the residual moisture content of the composition to the desired level, preferably as defined herein, in one pass through the system. If the moisture content of the product after one cycle is higher than desired, the moisture content of the powder may be further reduced by a second drying stage (or several of those) until the desired residual moisture content of the product is achieved.
  • FIG. 3 An example of a spray-drying apparatus is shown in Figure 3, which further illustrates the principle of spray-drying.
  • Liquid input stream (A) is sprayed through a nozzle (3) into a hot vapor stream (1 , 2) and is vaporized (4).
  • the droplets Upon introduction into the hot air stream the droplets are cooled down due to the evaporation of water or a chemical solvent from the concentrate. Solid particles form, while moisture quickly leaves the droplets.
  • a nozzle is used in order to achieve a sufficiently small droplet size (atomizer) and in order to maximize heat transfer and the rate of water/solvent evaporation.
  • a co-current flow of hot drying gas is used in the exemplary device shown in Figure 3. Particles are further dried and separated in a cyclone device (6). The dry particles are cooled and collected, ready for packaging in different formats (8).
  • the final product is collected as described above and is preferably in the form of a dry powder comprising the particles as defined herein with respect to the inventive dry powder composition.
  • the method comprises spray-freeze drying of the liquid comprising a long-chain RNA molecule.
  • the spray-freeze drying process may occur in bulk or as a continuous process, for example through a barrel lyophilizer.
  • the droplets formed by atomization are frozen immediately upon atomization.
  • the liquid feed is atomized, wherein the outlet of the atomizer is exposed to a coolant.
  • the atomizer is a rotary atomizer, a pressure nozzle, a vibrating orifice aerosol generator (VOAG), an ultrasonic nebulizer or a fountain nozzle.
  • the atomizer preferably sprays the droplets into the coolant.
  • the coolant is selected from a cryogenic gas or a cryogenic liquid, preferably an inert gas or inert liquid at low temperature.
  • coolants include, but are not limited to, air, carbon dioxide, nitrogen or argon.
  • the atomizer sprays the droplets into liquid nitrogen, which leads to immediate freezing of the droplets.
  • the temperature of the coolant is below -50°C, more preferably below -60°C, below -70°C, below -80°C, below -90°C, below -100°C or below -196°C.
  • the droplets contact the coolant, preferably a cryogenic gas or cryogenic liquid as defined herein, and are rapidly frozen.
  • the medium is changed to a cooling gas, which is characterized by a somewhat higher temperature, such as preferably a temperature in a range from -10°C to -30°C.
  • the cooling gas is selected from air, an inert gas, such as nitrogen, carbon dioxide or argon, and a mixture of gases.
  • the frozen particles are lyophilized.
  • the solvent comprised in the frozen particles typically sublimes into the cooling gas.
  • the cooling gas is continuously desiccated and/or heated in order to enhance the sublimation process.
  • the frozen particles may be dried in a vibrating fluid bed, preferably of the plug flow type like the VIBRO-FLUIDIZERTM from GEA Niro. It is preferably operated as a separate drying or cooling unit for powders or agglomerates or as a part of a spray drying plant for final drying or cooling.
  • the moist powder layer is typically vibrated on a cooling gas distributor plate. The effect of the vibration combined with the flow of drying gas creates suitable conditions for powder drying.
  • the vibration preferably allows to work with fluidized powder layers of less than 200 mm thickness. Attrition is thus preferably reduced by a narrow and controlled residence time.
  • the powder is collected from the device.
  • the coolant in the spray-freeze drying process is directly injected into the drying chamber, preferalby by using a nozzle, while spray droplets from atomizer are frozen immediately after contacting the surrounding curtain of e.g. liquid nitrogen and then conveyed to the separator or collector.
  • the coolant enters through a porous wall that encloses the atomizer.
  • a coolant is supplied in the space between the housing of the drying chamber and the drying chamber itself, so that the coolant forms a cooling layer, whose temperature is adjusted accordingly.
  • an apparatus for spray-freeze drying, which comprises: a chamber having an atomizer, preferably at one end of the chamber, the atomizer being connected to a liquid feed to produce a flow of liquid droplets; a nozzle for providing a flow of coolant that entrains atomized fluid sprayed by the atomizer; a coolant feed for the nozzle system; and a collector spaced from the atomizer sufficiently so that liquid droplets atomized by the atomizer are frozen by the flow of coolant before contacting the collector.
  • the spray-drying process or the spray-freeze drying process in the context of the present invention, may be carried out using any suitable spray-drying device known in the art.
  • Mini Spray Dryer B-290 (Buchi); Nano Spray Dryer B-90 (Buchi); Anhydro MicraSpray Dryer GMP (SPX.com); Anhydro MicraSpray Dryer Aseptic series (SPX.com); MDL-50 series; B,C,S,M sub-types, (fujisaki electric); MDL-015 (C) MGC lab-scale, (fujisaki electric); MDL-050 (C) MGC lab-scale, (fujisaki electric); LSD-1500 Mini spray dryer (cndryer.com); MSD-8 Multi-functional laboratory spray dryer (cndryer.com); PSD-12 Precision pharmacy spray dryer (cndryer.com); PSD-12 Precision pharmacy spray dryer (cndryer.com); TALL FORM DRYERTM - TFD (GEA Process Engineering); COMPACT DRYERTM - CD (GEA Process Engineering); Multi-Stage Dryer - MSDTM (GEA
  • the drying gas may be any suitable gas or mixture of gases, such as air.
  • an inert gas is used as drying gas, for example nitrogen, nitrogen-enriched air, helium or argon.
  • the spray-drying process is influenced to a considerable degree by the temperature of the drying gas. That temperature is typically characterized by two parameters, i.e. the inlet temperature (T in iet) and the outlet temperature (T ou tiet).
  • T in iet the inlet temperature
  • T ou tiet the outlet temperature
  • the term 'inlet temperature' refers to the drying gas temperature as measured at the drying gas inlet of the drying chamber as described herein.
  • Outlet temperature' refers to the drying gas temperature as measured at the drying gas outlet of the drying chamber as described herein.
  • the inlet temperature is preferably chosen sufficiently high in order to allow rapid and efficient drying, while at the same time avoiding degradation of the material. If heat sensitive material is dried, the residence time of the material in the drying chamber is preferably increased, e.g. by using larger drying chambers, which allows operating at lower temperatures.
  • the drying gas inlet temperature is in a range from about 30°C to about 250°C, more preferably from about 70°C to about 200°C or from about 90°C to about 150°C. In a preferred embodiment, the inlet temperature is at least about 30°C, at least about 50°C, at least about 70°C, at least about 85°C, at least about 95°C, at least about 100°C or at least about 110°C.
  • the inlet temperature is at least about 95°C or at least about 110°C.
  • the inlet temperature is at least about 95, at least about 96, at least about 97, at least about 98 or at least about 99°C. More preferably, the inlet temperature is at least about 100°C.
  • the drying gas outlet temperature is preferably in a range from about 30°C to about 200°C, more preferably from about 50°C to about 180°C, even more preferably from about 70°C to about 180°C, most preferably from about 70°C to 90°C or from 71 °C to 85°C.
  • the outlet temperature is at least about 50°C, more preferably at least about 60°C, at least about 65°C, at least about 70°C, at least about 71 °C, at least about 72°C, at least about 73°C, at least about 74°C, at least about 75°C, at least about 76°C, at least about 77°C, at least about 78°C, at least about 79°C, at least about 80°C, at least about 85°C, at least about 86°C, at least about 87°C, at least about 88°C, at least about 89°C, at least about 90°C, at least about 91 °C, at least about 92°C, at least about 93°C, at least about 94°C, at least about 95°C, at least about 96°C, at least about 97°C, at least about 98°C, at least about 99°C, at least about 100°C, at least about 101 °C, at least about 98°C, at
  • the inlet temperature by be at least about 95°C or at least about 100°C and/or the outlet temperature may be at least about 55, at least about 60, at least about 65, at least about 70 or at least about 71 °C. Most preferably, the inlet temperature is at least about 100°C and/or the outlet temperature is at least about 65, at least about 70 or at least about 71 °C.
  • higher temperatures increase the efficiency of the spray-drying process. The disadvantage of higher temperatures may be, however, an increased risk of degradation depending on the active ingredient or the excipients. Another caveat with spray-drying in general are shear forces that are acting on the liquid to be dried, in particular during atomization.
  • the shear stress is typically expected to be particularly detrimental to larger molecules.
  • a long-chain RNA molecule may be dried by spray-drying, while the long-chain RNA molecule's integrity and biological activity is retained notwithstanding the mechanical stress involved in the process.
  • the long-chain RNA molecule is not degraded, even when relatively high temperatures, in particular outlet temperatures, are used.
  • high outlet temperatures (T ou tiet) used in the spray-drying process for example, Toutiet > 60°C
  • One particular advantage of the inventive dry powder composition and the inventive method is that a dry powder composition is provided, which can be divided into packages useful for shipping, storage and use as medicament. Furthermore dry powder formation of long-chain RNA represents a cost- and time effective process, which can readily be scaled- up for commercial production. In this context, it is particularly advantageous that spray- freeze drying and spray drying can be carried out as a continuous process.
  • One advantage of a continuous process is that the product produced in one run has the same properties, therefore reducing the amount of required quality controls.
  • the residual moisture content of the dry powder composition obtained by the method according to the invention is as defined above with regard to the inventive dry powder composition.
  • the relative integrity and the biological activity of the long-chain RNA molecule in the dry powder composition obtained by using the inventive method is preferably as defined above for the inventive dry powder composition comprising a long- chain RNA molecule.
  • the inventive method thus provides long-chain RNA as defined herein in a particulate formulation.
  • the particles comprised in the dry powder composition obtained by the inventive method are characterized by a size distribution, which is preferably as defined herein for the particles of the inventive dry powder composition.
  • the product, which is obtained from the method according to the invention is the inventive dry powder composition as described herein.
  • the invention concerns a particle, or a plurality of particles, comprising a long-chain RNA molecule, which is preferably obtainable by the inventive method.
  • the invention is directed to a dry powder composition comprising a long-chain RNA molecule, which is obtainable by the inventive method as defined herein.
  • the inventive dry powder composition, the particle obtainable by the inventive method or the dry powder composition obtainable by the inventive method are packaged in single dosages after the drying process is completed.
  • the method may further comprise purification or selection steps, for example in order to separate particles of a certain size or shape.
  • the inventive dry powder composition, the particle obtainable by the inventive method or the dry powder composition obtainable by the inventive method may also be extended at any stage after the production process per se is finished.
  • an excipient preferably as described herein, may be added to the inventive dry powder composition or to the product of the inventive method, respectively.
  • the product of the inventive method provides considerable flexibility and allows extension of weight/volume (e.g. for better handling) as well as combination with other active ingredients and excipients.
  • a suitable excipient, preferably as defined herein, such as a carbohydrate may be added, for example inulin, starch or trehalose.
  • the excipient, which is added to inventive dry powder composition or to the particles or dry powder composition obtained by the inventive method is characterized by a low osmolarity.
  • the powder or the particles can easily be further processed to other dosage forms, such as tablets, capsules, granules or the like.
  • the present invention provides a pharmaceutical composition, comprising or consisting of the inventive dry powder composition, the particles as obtainable by the inventive method or the dry powder composition obtainable by the inventive method.
  • the inventive dry powder composition, the particles as obtainable by the inventive method or the dry powder composition obtainable by the inventive method are pharmaceutical compositions.
  • the inventive pharmaceutical composition comprises the inventive dry powder composition, the particles as obtainable by the inventive method or the dry powder composition obtainable by the inventive method and optionally a pharmaceutically acceptable carrier and/or vehicle.
  • the inventive pharmaceutical composition may optionally be supplemented with further components as defined above for the inventive dry powder composition or for the inventive method.
  • the inventive pharmaceutical composition may be prepared as a whole by the inventive method.
  • the inventive pharmaceutical composition comprises the long-chain RNA in particulate form as defined herein.
  • the first ingredient of the inventive pharmaceutical composition is the inventive dry powder composition, the particles as obtainable by the inventive method or the dry powder composition obtainable by the inventive method, as defined above.
  • the long-chain RNA molecule as defined herein represents a pharmaceutically active ingredient of the pharmaceutical composition.
  • the inventive pharmaceutical composition may comprise another class of compounds, which may be added to the inventive pharmaceutical composition in this context, may be selected from at least one pharmaceutically active component.
  • a pharmaceutically active component in this context is a compound that has a therapeutic effect against a particular medical indication, preferably cancer diseases, autoimmune disease, allergies, infectious diseases or a further disease as defined herein.
  • Such compounds include, without implying any limitation, preferably compounds including, without implying any limitation, peptides or proteins (e.g. as defined herein), nucleic acid molecules, (therapeutically active) low molecular weight organic or inorganic compounds (molecular weight less than 5,000, preferably less than 1 ,000), sugars, antigens or antibodies (e.g. as defined herein), therapeutic agents already known in the prior art, antigenic cells, antigenic cellular fragments, cellular fractions; modified, attenuated or deactivated (e.g. chemically or by irridation) pathogens (virus, bacteria etc.), etc.
  • inventive pharmaceutical composition may comprise a pharmaceutically acceptable carrier and/or vehicle.
  • a pharmaceutically acceptable carrier typically includes the liquid or non-liquid basis of the inventive pharmaceutical composition. If the inventive pharmaceutical composition is provided in liquid form, the carrier will typically be pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g phosphate, citrate etc. buffered solutions.
  • water or preferably a buffer preferably an aqueous buffer
  • 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.
  • examples of the optional potassium salts include e.g. KCI, Kl, KBr, K2CO3, KHCO3, K2SO4, and examples of calcium salts include e.g. CaCI 2 , Cal 2 , CaBr 2 , CaC0 3 , CaS0 4 , Ca(OH) 2 .
  • organic anions of the aforementioned cations may be contained in the buffer.
  • the buffer suitable for injection purposes as defined above may contain salts selected from sodium chloride (NaCI), calcium chloride (CaCI 2 ) and optionally potassium chloride (KCI), wherein further anions may be present additional to the chlorides.
  • CaCI 2 can also be replaced by another salt like KCI.
  • the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCI), at least 3 mM potassium chloride (KCI) and at least 0,01 mM calcium chloride (CaCI 2 ).
  • the injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e.
  • the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects.
  • Reference media are e.g. liquids occurring in "in vivo” methods, 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 and may be as defined above.
  • one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well for the inventive pharmaceutical composition, which are suitable for administration to a patient to be treated.
  • the term "compatible” as used here means that these constituents of the inventive pharmaceutical composition are capable of being mixed with the dry powder composition or the particles as defined herein in such a manner that no interaction occurs, which would substantially reduce the pharmaceutical effectiveness of the inventive pharmaceutical composition 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 and sucrose; starches, such as, for example, corn starch or potato starch; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium 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.
  • sugars such as, for example, lactose, glucose and sucrose
  • starches such as, for example, corn
  • inventive pharmaceutical composition may be administered orally, parenterally, by inhalation, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques.
  • the inventive pharmaceutical composition may be administered by parenteral injection, more preferably by subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or via infusion techniques.
  • Sterile injectable forms of the inventive pharmaceutical compositions may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol.
  • a non-toxic parenterally-acceptable diluent or solvent for example as a solution in 1 ,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation of the inventive pharmaceutical composition.
  • inventive pharmaceutical composition as defined above may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • the active ingredient i.e. the dry powder composition or the particles, as defined above, is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • the inventive pharmaceutical composition may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including diseases of the skin or of any other accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs.
  • the inventive pharmaceutical composition may be formulated in a suitable ointment, containing the components as defined above suspended or dissolved in one or more carriers. Carriers for topical administration include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the inventive pharmaceutical composition can be formulated in a suitable lotion or cream.
  • suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the inventive pharmaceutical composition is for mucosal, intranasal, inhalation or pulmonary delivery.
  • the pharmaceutical composition comprises the powder or the particles, preferably as defined herein, which are respirable, i.e. which can readily be dispersed in air or in a gas (e.g. by using an inhalation device) and inhaled by a subject.
  • the particles of the pharmaceutical composition are as defined herein with respect to the inventive dry powder composition so that at least a portion of the aerosolized particles reaches the lungs.
  • the pharmaceutical composition according to the invention comprises the inventive dry powder composition, wherein the dry powder composition comprises particles that have a MMAD of 10 m or less.
  • the inventive pharmaceutical composition typically comprises a "safe and effective amount" of the components of the inventive pharmaceutical composition as defined above, particularly of long-chain RNA molecule as comprised in the inventive dry powder composition or in the particles obtainable by the inventive method.
  • a "safe and effective amount” means an amount of the long-chain RNA molecule that is sufficient to significantly induce a positive modification of a disease or disorder as defined herein.
  • a "safe and effective amount” is small enough to avoid serious side-effects, that is to say, to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • a "safe and effective amount" of the components of the inventive pharmaceutical composition, particularly of the long-chain RNA molecule will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the activity of the specific (lyophilized) nucleic acid (sequence) employed, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying doctor.
  • the inventive pharmaceutical composition may be used for human and also for veterinary medical purposes, preferably for human medical purposes, as a pharmaceutical composition in general or as a vaccine.
  • the inventive dry powder composition, the particles or the dry powder composition obtainable by the inventive method or the inventive pharmaceutical composition may be provided as a vaccine.
  • Such an inventive vaccine is typically composed like the inventive pharmaceutical composition, i.e. it contains a long-chain RNA molecule formulated as defined above and optionally a pharmaceutically acceptable carrier and/or vehicle. Further components may be as defined above for the inventive pharmaceutical composition.
  • the inventive vaccine preferably supports at least an innate immune response of the immune system of a patient to be treated.
  • inventive vaccine furthermore may also elicit an adaptive immune response, preferably, if the long-chain RNA molecule of the inventive vaccine encodes any of the antigens (or antibodies) mentioned herein, which elicit an adaptive immune response or any antigen as defined herein is added to the inventive vaccine, which can effectively induce an adaptive immune response.
  • the inventive vaccine may also comprise a pharmaceutically acceptable carrier, adjuvant, and/or vehicle as defined above for the inventive pharmaceutical composition.
  • a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered.
  • the inventive vaccine can be administered, for example, systemically or locally.
  • 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/intrapulmonal 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 herein may be administered by an intradermal, subcutaneous, or intramuscular route. Inventive vaccines are therefore preferably formulated in liquid (or sometimes in solid, e.g. as an aerosol) form. The suitable amount of the inventive vaccine to be administered can be determined by routine experiments with animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non- human primate models. Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof.
  • 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 vaccine is to be administered orally, 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.
  • 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 may comprise an adjuvant.
  • an adjuvant may be understood as any compound, which is suitable to initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response.
  • the inventive vaccine when administered, the inventive vaccine preferably elicits an innate immune response due to the adjuvant, optionally contained therein.
  • an adjuvant may be selected from an adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal.
  • the adjuvant is preferably selected from compounds, which are known to be immune-stimulating due to their binding affinity (as ligands) to human Toll-like 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, TLR1 1 , TLR12 or TLR13.
  • the present invention furthermore provides several applications and uses of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method.
  • the invention concerns the use of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method for the preparation of a medicament for the prophylaxis, treatment and/or amelioration of a disorder or a disease, preferably as defined herein.
  • the present invention is directed to the use of the long-chain RNA molecule in particulate form as defined herein in the treatment or prevention of a disease.
  • the invention concerns the use of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method, in the treatment or the prevention of a disease, preferably as defined herein.
  • the present invention concerns the first medical use of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method as a medicament.
  • the medicament may be in the form of a pharmaceutical composition or in the form of a vaccine as a specific form of pharmaceutical compositions.
  • a pharmaceutical composition in the context of the present invention typically comprises or consists of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method as defined above, optionally further ingredients, preferably as defined above, and optionally a pharmaceutically acceptable carrier and/or vehicle, preferably as defined above.
  • the present invention concerns a method of treating or preventing a disorder or a disease by administering to a subject in need thereof a pharmaceutically effective amount, preferably as defined herein, of the inventive dry powder composition, the inventive pharmaceutical composition, or the inventive vaccine.
  • the method is for treating or preventing a disorder or a disease selected from cancer or tumor 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 single gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, or any further disease mentioned herein.
  • infectious diseases preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases
  • monogenetic diseases i.e. (hereditary) diseases
  • genetic diseases in general, diseases which have a genetic inherited background and which are typically caused by a single gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, or any further disease mentioned herein.
  • the present invention is directed to the use of the long- chain RNA molecule in particulate form, preferably in the form of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method, for the prophylaxis, treatment and/or amelioration of a disease or disorder as defined herein, wherein the disease or disorder is preferably selected from cancer or tumor 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 single gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, or any further disease mentioned herein.
  • infectious diseases preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases
  • monogenetic diseases i.e. (hereditary) diseases
  • genetic diseases in general, diseases which have a
  • the present invention is directed to the second medical use of the long-chain RNA in particulate form as defined herein, preferably in the form of the inventive dry powder composition, the particles obtainable by the inventive method or the dry powder composition obtainable by the inventive method for the treatment of diseases as defined herein, preferably to the use therof for the preparation of a medicament for the prophylaxis, treatment and/or amelioration of various diseases as defined herein, preferably selected from cancer or tumor 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 single gene defect and are inherited according to Mendel's laws, cardiovascular diseases, neuronal diseases, or any further disease mentioned herein.
  • infectious diseases preferably (viral, bacterial or protozoological) infectious diseases, autoimmune diseases, allergies or allergic diseases
  • monogenetic diseases i.e.
  • the present invention also allows treatment of diseases, which have not been inherited, or which may not be summarized under the above categories.
  • diseases may include e.g. the treatment of patients, which are in need of a specific protein factor, e.g. a specific therapeutically active protein as mentioned above.
  • This may e.g. include dialysis patients, e.g. patients, which undergo a (regular) a kidney or renal dialysis, and which may be in need of specific therapeutically active proteins as defined above, e.g. erythropoietin (EPO), etc.
  • diseases in the context of the present invention may include cardiovascular diseases chosen from, without being limited thereto, coronary heart disease, arteriosclerosis, apoplexy and hypertension, etc.
  • diseases in the context of the present invention may be chosen from neuronal diseases including e.g. Alzheimer's disease, amyotrophic lateral sclerosis, dystonia, epilepsy, multiple sclerosis and Parkinson's disease etc.
  • the present invention also provides a kit, particularly as a kit of parts.
  • a kit of parts may contain e.g. the inventive dry powder composition, the inventive pharmaceutical composition or the inventive vaccine as defined above, preferably divided into different parts of the kit.
  • the inventive pharmaceutical composition or the inventive vaccine may be prepared as a kit of parts, e.g.
  • the inventive pharmaceutical composition or the inventive vaccine as described herein (whereby at least the long-chain RNA in particulate form is included), or the inventive dry powder composition as such, as a dry formulation, i.e. devoid of any liquid component, and in at least one further separate part of the kit a solvent and/or a buffer as described herein with respect to the liquid provided in step a) of the inventive method, the inventive pharmaceutical composition or the inventive vaccine or any further solvent and/or buffer as described herein for lyophilization, transfection and/or injection.
  • the inventive pharmaceutical composition or the inventive vaccine may be prepared as a kit of parts, e.g.
  • kits by incorporating into one or more parts of the kit only the inventive dry powder composition, the particles obtainable by the inventive method, or the dry powder composition obtainable by the inventive method, as described herein, and in at least one further separate part of the kit a solvent and/or a buffer as described herein for the liquid provided in step a) of the inventive method, for the inventive pharmaceutical composition or the inventive vaccine or any further liquid and/or buffer as described herein for lyophilization, transfection and/or injection.
  • further ingredients of the kit may include components as defined above, e.g. (solutions comprising) proteins, amino acids, alcohols, carbohydrates, metals or metal ions, surfactants, polymers or complexing agents, and/or buffers, preferably all as defined above.
  • kit or kit of parts
  • kit or kit of parts as described above may contain optionally technical instructions with information on the administration and dosage of the inventive composition.
  • Such a kit, preferably kit of parts may be applied, e.g., for any of the above mentioned applications or uses.
  • FIG. 1 Schematic diagram of a spray-freeze drying apparatus
  • A Spray-freeze drying process, where liquid feed is atomized and frozen in a cooling zone.
  • B Frozen particles are subsequently freeze-dried in a lyophilizer.
  • Figure 2 Scheme of a continuous spray-freeze drying process line showing the three process compartments (A: droplet formation through nozzle; cooling and fast freezing of droplets to particles; B: continuous freeze-drying drum; C: flexible multi or single-dose packaging of bulk powder).
  • A solution or suspension to be dried in
  • B atomization gas (e.g. nitrogen) in
  • 1 drying gas (e.g. nitrogen) in
  • 2 heating of drying gas
  • 3 spraying of solution or suspension
  • 4 drying chamber
  • 5 part between drying chamber and cyclone
  • 6 cyclone
  • 7 drying gas out
  • 8 product collection vessel.
  • RNA was formulated at 0.8 g/L + 0.2 g/L protamine + 50 g/L trehalose in WFI.
  • the samples from test runs 6 and 7 was spray-dried using a SDMicro spray dryer from GEA Niro. Dried powders were reconstited in WFI to yield the initial concentration of the RNA formulation of 51 g/L total mass corresponding to 0.8 g/L RNA, UV absorbance was measured at 260nm with Nanodrop.
  • Figure 5 Size determination of reconstituted RNA complexes using dynamic light scattering analysis by Zetasizer
  • Zeta potential was measured using tenfold diluted reconstituted spray dried samples and compared to lyophilized controls
  • Particle size distribution was measured through polydispersity index analysis with Zetasizer and non-diluted reconstituted spray dried samples compared to lyophilized controls.
  • Figure 8 Clarity of reconstituted RNA complex dispersions. As a measure of clarity, the absorbance at 350 nm was measured in non-diluted samples. Reconstituted spray-dried samples were compared to lyophilized controls.
  • a vector for in vitro transcription was constructed containing a T7 promoter followed by a GC-enriched sequence encoding the hemagglutinin (HA) protein of influenza A virus (A/Netherlands/602/2009(H1 N1 )) and used for subsequent in vitro transcription .reactions.
  • HA hemagglutinin
  • the constructs R2564 (SEQ ID NO: 1 ) was prepared by introducing a 5'- TOP-UTR derived from the ribosomal protein 32L4, 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.
  • SEQ ID NO: 1 see Figure 12
  • the sequence of the corresponding mRNA is shown.
  • RNA was diluted (0.87 g/L RNA final concentration) and a protamine/trehalose mixture was prepared (0.43 g/L protamine; 10.87 % trehalose in water for injection). Both solutions were mixed in a RNA:protamine ratio of 2: 1 (w/w).
  • RNA/protamine complexes were supplemented with R2564 to yield final concentrations of 0.4 g/L RNA complexed with 0.2 g/L protamine, 0.4 g/L free RNA and 5% trehalose (w/w).
  • trehalose- containing solution 5% trehalose (w/w) in water
  • test composition 5% trehalose (w/w) in water
  • the objective was to spray-dry a solution comprising 50 g of RNA formulated with protamine according to Example 3 with a yield as high as possible (> 10% yield) resulting in a free-flowing non-sticky powder. Residual moisture content should be below 5%, preferably below 4%.
  • the formulation was spray dried using a SDMicro spray dryer from GEA Niro. Following parameters were used: Heating gas (nitrogen) rate at 30 kg/h, nozzle gas rate at 6 kg/h.
  • the drying chamber has a diameter of 200 mm, a cylindrical height of 350 mm and a 60° conical bottom. Nitrogen is used as drying gas and heated electrically. The drying gas enters the top of the chamber through a ceiling gas distributor.
  • the feed is transferred to the two-fluid nozzle at the top of the drying chamber through silicone hoses by a peristaltic pump.
  • the particle-laden nitrogen flows in co-current with the heating gas towards a cyclone for primary particle separation.
  • Primary particles were collected in a sterile 100 ml glass bottle.
  • Very fine particles not successfully collected by the cyclone passed to a bag filter for secondary particle collection.
  • particle-laden nitrogen is passed through HEPA filters and an active carbon filter before being exhausted.
  • the plant was cleaned in place with a 0.5 M sodium hydroxide solution followed by extensive washing with WFI (water for injection).
  • the two main parameters which were adjusted during the test runs were atomization gas pressure and outlet temperature.
  • Atomization gas pressure was adjusted in order to investigate yields of different particle sizes created by the two-fluid nozzle.
  • the outlet temperature was changed in order to evaluate residual moisture content of different powders and the biological activity of the RNA subjected to different temperatures.
  • 5 tests were carried out with each 100 g of a test solution containing 5 % excipient (trehalose) in WFI (according to Example 4) to setup the process parameters.
  • the formulation containing the long-chain single-stranded RNA (2% of the solid matter, of which 80% is RNA and 20% protamine) and 5% trehalose (w/w) as excipient was divided in two samples and subjected to two different tests with parameters initially confirmed as successful (Test No. 6 and 7). Parameters chosen and results are summarized in Table 1.
  • Example drying test no. 1
  • Outlet and inlet temperatures were further decreased to 50°C and 87°C respectively in test no. 3 with atomization gas pressure of 0.3 bars and the feed rate at 500 g/h.
  • Test resulted in a yield of 52 % corresponding to 1.3 g of dry powder and residual humidity at 4.8 % as tested in a Karl Fischer assay.
  • Example drying test no. 6 For final drying tests, the plant was sanitized in place and all parameters were kept as in test no. 1 , 4 and 5 (outlet temperature was at 71 °C, atomization gas pressure at 0.3 bars and feed rate and inlet temperature at 500 g/h and 1 13°C respectively) except that 25 g of the RNA/protamine formulation was fed into the plant. Result: Test resulted in a yield of 42 % corresponding to 530 mg of dry powder and residual humidity expected at near 4 %.
  • Example drying test no. 7 Outlet temperature was stabilized at 55°C with an inlet temperature at 92°C, all other parameters were kept as in test no. 1 , 4 and 5.
  • an outlet temperature of at least 50°C results in a residual moisture below 7%.
  • An outlet temperature of 71 °C results in a residual moisture below 5%, more preferably below 4%.
  • Spray dried powder was filled into 2R vials manually (about 20-40 mg per vial) using a sterile rubber spatula. For storage, the powder was then overlaid with argon and sealed.
  • Dried powder was reconstituted in WFI to yield 51 g/L total mass corresponding to 0.8 g/L RNA.
  • spray-dried powder had to be portioned manually UV absorbance was controlled at 260nm with Nanodrop (Thermo scientific ND-2000c, pathlength: 1 cm) and compared to the absorbance of reconstituted lyophilized RNA.
  • Fig. 4 shows that the whole amount spray-dried RNA could be reconstituted. This corresponds to the reconstitution of lyophilized RNA used as control.
  • 8.2 Measurement of nanoparticle size, zeta potential and polydispersity of protamine- formulated RNA
  • the hydrodynamic diameter of nanoparticles was measured by dynamic light scattering using a Zetasizer Nano (Malvern Instruments, Malvern, UK) according to the instructions provided by the manufacturer. The measurements were performed at 25°C and a scattering angle of 173° in the specified buffer analyzed by a cumulant method to obtain the hydrodynamic diameters and polydispersity indices of the particles.
  • the Zeta potential of the nanoparticles was evaluated by the laser Doppler electrophoresis method using a Zetasizer Nano (Malvern Instruments, Malvern, UK). The measurement was performed after 10-times dilution of the reconstituted material in water for injection at 25°C and a scattering angle of 173°.
  • Nanoparticle size was measured using Zetasizer nano (Malvern instruments) with 70 ⁇ of complexation solution in a UV transmittable cuvette (UVette, Eppendorf) and the following settings: Refractive index of material: 1.450; absorption of material: 0.001 ; dispersant temperature: 25 °C; dispersant viscosity: 0.8753; dispersant refractive index: 1.331 ; Mark- Houwink parameters: A parameter 0.428; K-parameter 7.67-05 cm2/s; use dispersant viscosity as sample viscosity; sample temperature: 25 °C; sample cuvette: Zen 0040 disposable cuvette; equilibration time: 0s; measurement angle: 173° backscatter; automatic measurement duration; number of measurements: 1 ; automatic attenuation setting; positioning method: seek for optimal position; no extension duration for large particles; analysis model: normal resolution.
  • the average nanoparticle size (hydrodynamic diameter of a spherical particle, in nm) is given as the Z-average and the polydispersity is given as the polydispersity index (PDI), both of which were calculated by the instrument's software (Zetasizer software version 6.34, Malvern Instruments). Results:
  • Fig. 5 shows that the nanoparticles comprised in the reconstituted spray-dried samples form test runs 6 and 7 are comparable in size to nanoparticles comprised in the sample, which was lyophilized before reconstitution.
  • Fig. 6 shows that the nanoparticles comprised in the reconstituted spray-dried samples from test runs 6 and 7 have the same zeta potential as nanoparticles in the sample, which was lyophilized before reconstitution.
  • Fig. 7 shows that the nanoparticles comprised in the reconstituted spray-dried samples form test runs 6 and 7 have the same polydispersity index as nano in the sample, which was lyophilized before reconstitution.
  • the absorption at 350 nm was determined. 200 ⁇ of each complexation reaction were applied to a microwell plate (Costar, UV Plate, 96 well, no lid, UV transmittable flat bottom). A350 was measured with a Synergy HT plate reader (BioTek systems). Path length correction was performed by Gen5 software (BioTek, Installation version: 1.1 1.5) with a test wavelength of 977 nm and a reference wavelength of 900 nm. Correction was performed with a constant K-factor of 0.18 to yield the A350 value corrected to 1 cm path length.
  • Fig. 8 shows that the nanoparticles comprised in the reconstituted spray-dried samples form test runs 6 and 7 have the same turbidity (A350 nm) as nanoparticles in the sample, which was lyophilized before reconstitution.
  • RNA and protamine sodium chloride was added to 2.5 M final concentration of sodium chloride and 0.4 g/L RNA. After incubation at 80 °C for 3 min, the RNA was precipitated with isopropanole at -20 °C for 2 h, centrifuged, washed with 75 % (v/v) ethanol, dried and dissolved in water for injection.
  • RNA was separated in formaldehyde-containing agarose gels (0.7 % w/w formaldehyde, 1.2 % wA/ agarose) in 3-Morpholinopropane sulfonic acid buffer (for details on method see Sambrook, Russel: Molecular Cloning: A Laboratory Manual, vol. 3, Cold Spring Harbor Laboratory, 2000.).
  • RNA samples were denatured in RNA sample buffer (Thermo scientific) at 80 °C for 5 min before loading to the gel. 1 of RNA were loaded per lane. A voltage of about 5 V/cm was applied until the marker of 6 RNAs of different size (CureVac) was well-separated. Integrity of the RNA was calculated using Quantity One (BioRad, Version 4.6.9).
  • RNA/protamine formulations after reconstitution did not differ significantly if the formulation was spray- or freeze-dried ( Figure 7, Figure 8).
  • the RNA concentrations recovered from RNA/protamine formulations dried by spray- or freeze-drying comparable. With the two drying methods, spray- and freeze-drying, the high integrity (>90 %) of RNA preparations remained unchanged ( Figure 10).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biotechnology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Manufacturing & Machinery (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne une formulation d'ARN à chaîne longue stable au stockage. En particulier, l'invention concerne une composition de poudre sèche comprenant une molécule d'ARN à chaîne longue. La présente invention concerne en outre des procédés de préparation d'une composition de poudre sèche comprenant une molécule d'ARN à chaîne longue par séchage par atomisation ou pulvérisation-cryodessiccation. L'invention concerne en outre l'utilisation de ladite composition de poudre sèche comprenant une molécule d'ARN à chaîne longue dans la préparation de compositions pharmaceutiques et de vaccins, un procédé de traitement ou de prévention d'un trouble ou d'une maladie, une première et une seconde utilisations médicales de ladite composition de poudre sèche comprenant une molécule d'ARN à chaîne longue et des kits, en particulier des kits de parties, comprenant ladite composition de poudre sèche comprenant une molécule d'ARN à chaîne longue.
EP16728216.9A 2015-05-20 2016-05-20 Composition de poudre sèche comprenant de l'arn à chaîne longu Withdrawn EP3298143A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15001517 2015-05-20
PCT/EP2016/000846 WO2016184577A2 (fr) 2015-05-20 2016-05-20 Composition de poudre sèche comprenant de l'arn à chaîne longu

Publications (1)

Publication Number Publication Date
EP3298143A2 true EP3298143A2 (fr) 2018-03-28

Family

ID=53275951

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16728216.9A Withdrawn EP3298143A2 (fr) 2015-05-20 2016-05-20 Composition de poudre sèche comprenant de l'arn à chaîne longu

Country Status (2)

Country Link
EP (1) EP3298143A2 (fr)
WO (1) WO2016184577A2 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2718269T3 (en) 2011-06-08 2018-04-09 Translate Bio Inc SPLITLY LIPIDS
WO2018115527A2 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre le coronavirus du syndrome respiratoire du moyen-orient

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070172430A1 (en) * 2006-01-20 2007-07-26 Nastech Pharmaceutical Company Inc. Dry powder compositions for rna influenza therapeutics
WO2011069529A1 (fr) * 2009-12-09 2011-06-16 Curevac Gmbh Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques
WO2011069528A1 (fr) * 2009-12-09 2011-06-16 Curevac Gmbh Lyophilisation d'acides nucléiques dans des solutions contenant du lactate

Also Published As

Publication number Publication date
WO2016184577A3 (fr) 2017-01-12
WO2016184577A2 (fr) 2016-11-24

Similar Documents

Publication Publication Date Title
US11534405B2 (en) Dry powder composition comprising long-chain RNA
US11433027B2 (en) Dry powder composition comprising long-chain RNA
US11491112B2 (en) Lyophilization of RNA
JP2022095702A (ja) 脂質ナノ粒子の安定化製剤
EP3852728A1 (fr) Préparation de nanoparticules lipidiques et leurs méthodes d'administration
CN111315359A (zh) 制备脂质纳米颗粒的方法
TW202139976A (zh) 製備脂質奈米顆粒之方法
WO2017105138A1 (fr) Procédé de préparation de micelle polymère contenant un médicament anionique
EP3298143A2 (fr) Composition de poudre sèche comprenant de l'arn à chaîne longu
Yihunie et al. Recent advances in messenger ribonucleic acid (mRNA) vaccines and their delivery systems: a review
EP3984527A1 (fr) Poudre sèche d'arnsi nano-in-micro-encapsulée, son procédé de fabrication et utilisation d'une formulation de poudre sous forme de dosage pharmaceutique, en particulier pour l'administration pulmonaire
CN118302155A (en) Method for freezing and freeze-drying Lipid Nanoparticles (LNPs) and LNPs obtained thereby

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20171004

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: EBER, FABIAN, JOHANNES

Inventor name: KETTERER, THOMAS

Inventor name: SEWING, STEFANIE

Inventor name: YAZDAN PANAH, BENYAMIN

Inventor name: MUTZKE, THORSTEN

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20180925

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

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20200307