USE OF IFN-LAMBDA MRNA FOR TREATING VIRAL INFECTIONS

Information

  • Patent Application
  • 20240181011
  • Publication Number
    20240181011
  • Date Filed
    November 04, 2021
    2 years ago
  • Date Published
    June 06, 2024
    a month ago
Abstract
The present invention relates to pharmaceutical compositions comprising an mRNA encoding an IFN-λ polypeptide for use in treating a viral-induced disorder, preferably a viral-induced respiratory disorder, such as COVID-19.
Description

The present invention relates to pharmaceutical compositions comprising an mRNA encoding an IFN-λ polypeptide for use in treating a viral-induced disorder, preferably a viral-induced respiratory disorder, such as influenza or COVID-19.


Interferons are a group of proteins which presently are classified into three different families, type I interferons, type II interferons and type III interferons.


Type I interferons are a family of closely related glycoproteins comprised of thirteen IFN-α subtypes as well as IFN-β, IFN-κ, IFNε and IFN-ω. In humans, the IFNα gene family is composed of 12 different subtypes encoded by 14 genes, including one pseudogene and two genes that encode identical proteins (Díaz et al., Genomics 22 (1994), 540-552), i.e. IFN-α1, IFN-α2, IFN-α8, IFN-α14, IFN-α17, IFN-α4, IFN-α5, IFN-α6, IFN-α7, IFN-α10, IFN-α16, IFN-α21 (da Rocha Matos et al., Emerg Microbes Infect. 8, (2019), 1763-1776). As regards IFN-α subtypes, it has been found that they reveal a spectrum of anti-viral, anti-proliferative and immunomodulatory responses. Type I interferons play an important role in the innate immune response to respiratory virus infection. The mechanism involves initial release of interferon-β which in turn stimulates the further release of interferon-β and of the interferon-αs in a cascade mediated via the type-1 interferon receptor. There is one type II interferon, i.e. IFN-gamma, which binds a different receptor than type I interferons and has largely distinct functions from the type I IFNs. Type III interferons are thee most recently discovered family of interferons. They are also referred to as interferon lambdas (IFNλs). The IFNλs are three closely related proteins which have been discovered in 2003. Interferon λ-1 is also known as IL-29, while Interferon λ-2 and λ-3 are also known as IL-28A and IL-28B, respectively. These interferons bind to a third receptor distinct from those of type I and type II interferons. These interferons have been shown to have anti-viral activity. It has been found that interferon λ is important in a wide variety of viral infections including HCV, HBV, influenza virus, rhinovirus, respiratory syncytial virus (RSV), lymphocytic choriomeningitis virus (LCMV), rotavirus, reovirus, norovirus and West Nile virus (WNV). Experimental in vivo approaches using IFNλ receptor knockout mice have highlighted the importance of IFNλ signaling in control of influenza A virus (IAV), SARS coronavirus, RSV and human metapneumovirus levels in the lung as well as norovirus, reovirus and rotavirus levels in the gastrointestinal tract. In vivo studies indicate that IFNλ shows, in comparison to IFNα/β, a reduced ISG response while it is much less inflammatory in vivo than IFNα/β.


Interestingly, IFNλ retains many antiviral properties despite the less inflammatory response compared with type I IFNs. This has spurred development of IFNλ for clinical use as an alternative treatment to IFNα for HCV infection (Muir et al., J. Hepatol. 61 (2014), 1238-1246). More recent developments also indicate that IFNλ treatment could also be utilized to control respiratory viral infections. IFNλ2 and 3 was shown to control IAV pulmonary titers similarly to IFNα or IFNβ treatment (Davidson et al., EMBO Mol. Med. 8 (2016), 1099-1112; Kim et al, Am. J. Respir. Cell. Mol. Biol. 56 (2017), 202-212). Importantly, IFNλ treatment avoided excessive pulmonary inflammation associated with IFNα treatment (Kim et al., Am. J. Respir. Cell. Mol. Biol. 56 (2017), 202-212). Therapeutic applications of IFNλ so far concentrate on the administration of the recombinantly produced protein. However, the use of recombinantly produced protein requires the repeated administration of relatively high doses of protein since the protein is relatively quickly cleared from the system and can, thus, only act for a brief period of time. For example in Dinnon III et al. (Nature https://doi.org/10.1038/s41586-020-2708-8 (2020)) 2 μg of peg-IFNλ1 is administered subcutaneously to mice. Similarly, Davidson et al. (EMBO Mol. Med. 8 (2016), 1099-1112) describe the administration of 2.6 μg/50 μl of IFNλ3 to B6.A2G-Mx mice for treating or preventing influenza infection. In Galani et al. (Immunity 46 (2017), 875-890), it was reported that mice were treated with 5 or 10 μg of recombinant mouse pegylated IFNλ2.


Therefore, there is a need to provide means and methods for an efficient way to deliver IFNλ in vivo to target tissues, i.e. in a manner which ensures that it is active for an extended period of time and can exert its effect for an extended period of time. This would allow to reduce the amount of IFNλ to be administered to a patient within a given period of time and would also allow to reduce the numbers of administrations to a patient.


The present invention addresses this need by providing the embodiments as recited in the claims.


Thus, the present invention relates to a pharmaceutical composition comprising an mRNA encoding an IFN-λ polypeptide for use in treating and/or preventing a viral-induced disorder.


The present invention is based on the finding that mRNA which codes for IFNλ allows for an extremely efficient and long-lasting activating effect on down-stream targets important for antiviral activity of IFN-λ when transfected into cells which serve as a model for alveolar epithelial cells. In particular, in an in vitro model using A-549 cells (a model for type II-like pneumocytes/alveolar epithelial cells), even at extremely low doses of mRNA the downstream-targets IFIT3, ISG15, IFIT1 and OAS3 (which are indicative of the antiviral activity of IFNλ) are activated to high levels and for an extended period of time (more than 120 hours). These results could be confirmed in in vivo experiments in mice. Moreover, it has surprisingly been found that in A-549 cells which had previously been stably transfected so as to express the ACE2 receptor, the induction of the downstream targets is as high as in the case of A-549 cells which do not express the ACE2 receptor when mRNA encoding an IFN-λ polypeptide is used, while in the case of the administration of recombinant IFNλ protein the induction of downstream targets is much lower in ACE2 expressing A-549 cells when compared to A-549 cells which do not express the ACE2 receptor. This indicates that the administration of the recombinant IFNλ polypeptide does not lead to an efficient activation of downstream targets in cells which express the ACE2 receptor while the administration of mRNA encoding IFNλ allows for an efficient activation. Accordingly, the use of mRNA encoding IFNλ is particularly advantageous in cases where viral infections target ACE2 expressing cells, i.e. where the virus enters the cells via the ACE2 receptor.


Moreover, it could be shown in Air-Liquid Interface (ALI) cultures of different cell-types that unexpectedly mRNA encoding IFNλ is able to show an effect as regards activating downstream targets when applied to the apical side of the cells (facing the air) although the corresponding receptor is expected to be located on the basolateral side of the cells (facing the liquid culture medium). These results could be confirmed by in vivo experiments in mice in which the mRNA encoding IFNλ was directly applied to the lung. It is known that airway epithelia form mechanical barriers that separate the external environment from the internal milieu and that structural polarity of epithelia is critical for barrier integrity and is controlled by intricate cell-cell adhesive complexes that contain adherens and tight junctions (see, e.g., Humlicek et al., J. Immunol. 178 (2007), 6395-6403). Moreover, it is known that there is a polarity as regards the functionality of certain cytokines in the sense that only their basolateral location with regard to the airway epithelium leads to an effect since the corresponding receptors are obviously only present at the basolateral side of the cells. Accordingly, airway epithelial responses to certain cytokines could only be observed after application to the airway lumen if the barrier function of the epithelia was physically or pharmacologically disrupted previously (Humlicek et al., loc. cit.). Thus, it is surprising that the administration of IFNλ mRNA to the apical side of epithelial cells cultured in ALI cultures or applied directly to the lung of mice without disruption of the barrier function of the epithelia leads to an efficient activation of downstream targets of IFNλ (which is indicative for an antiviral effect).


In principle, in the context of the present invention, mRNA encoding an IFNλ polypeptide can be used for treating and/or preventing any possible viral-induced disorder. It has been shown that IFNλ exhibits antiviral activity against many viruses in vitro. In vivo the antiviral activity of IFNλ has in particular been observed for viruses that infect epithelial cells of the respiratory, gastrointestinal and urogenital tracts as well as the liver (see, e.g., Lazear et al., Immunity 43 (2015), 15-28; Table 1). Viral-induced diseases which can be treated or prevented according to the present invention include diseases caused by a virus belonging to the family of Pneumoviridae, Orthomyxoviridae, Adenoviridae, Arenaviridae, Paramyxoviridae, Flaviviridae, Retroviridae, Caliciviridae, Picornaviridae, Coronaviridae, Parvoviridae, Reoviridae, Herpesviridae or Hepadnaviridae.


Viruses belonging to the family of Pneumoviridae include Human Metapneumo virus.


Viruses belonging to the family of Orthomyxoviridae include Influenza virus.


Viruses belonging to the group of Adenoviridae include Adenovirus.


Viruses belonging to the family of Arenaviridae include Lymphocytic choriomeningitis virus.


Viruses belonging to the family of Paramyxoviridae include Respiratory syncytial virus.


Viruses belonging to the family of Flaviviridae include Dengue virus, Hepatitis C virus, Zika virus and West Nile virus.


Viruses belonging to the family of Retroviridae include Human immunodeficiency virus.


Viruses belonging to the family of Caliciviridae include norovirus.


Viruses belonging to the family of Picornaviridae include rhinovirus.


Viruses belonging to the family of Coronaviridae include SARS-COV, SARS-COV2, MERS and HCoV-NL63, -OC43, -229E and HKU1.


Viruses belonging to the family of Parvoviridae include bocavirus.


Viruses belonging to the family of Reoviridae include Reovirus and Rotavirus.


Viruses belonging to the family of Herpesviridae include Cytomegalovirus and Herpes simplex virus, such as Herpes simplex virus 1 and 2.


Viruses belonging to the family of Hepadnaviridae include Hepatitis B virus.


In a preferred embodiment, the viral-induced disorder is a viral-induced respiratory disorder. In this context, it is preferred that the virus which induces the respiratory disorder is selected from the group consisting of rhinovirus, influenza virus, parainfluenza virus, metapneumo virus, respiratory syncytial virus, adenovirus and corona virus.


In a particularly preferred embodiment, the virus is a virus which enters cells via the ACE2 receptor. ACE2 (Angiotensin-Converting-Enzyme 2) is an enzyme attached to the cell membranes of cells in the lungs, arteries, heart, kidney and intestines. It lowers blood pressure by catalyzing the hydrolysis of angiotensin II into angiotensin. ACE2 also serves as the entry point into cells for some coronaviruses, including SARS-COV, SARS-COV-2 and HCoV-NL63.


Thus, in a particularly preferred embodiment, the virus is a coronavirus and most preferably a virus selected from the group consisting of SARS-COV, SARS-COV-2 and HCOV-NL63.


Accordingly, in a particularly preferred embodiment, the viral-induced disease is SARS (caused by SARS-COV), COVID-19 (caused by SARS-COV-2) and mild to moderate upper respiratory tract infections, severe lower respiratory tract infection, croup and bronchiolitis (caused by HCoV-NL63).


The IFNλ protein encoded by the mRNA comprised in the pharmaceutical composition can be any possible IFNλ protein, in particular Interferon λ-1 (is also known as IL-29), Interferon λ-2 (also known as IL-28A) or Interferon λ-3 (IL-28B) or any combination thereof. In a preferred embodiment, the IFNλ protein is a human protein.


In one embodiment the IFNλ protein encoded by the mRNA comprised in the pharmaceutical composition is IFNλ-1, preferably IFNλ-1 comprising the amino acid sequence as shown in SEQ ID NO:2. In a more preferred embodiment the mRNA encoding IFNλ-1 comprises a coding region as shown in SEQ ID NO: 1.


In one embodiment the IFNλ protein encoded by the mRNA comprised in the pharmaceutical composition is IFNλ-2, preferably IFNλ-2 comprising the amino acid sequence as shown in SEQ ID NO:4. In a more preferred embodiment the mRNA encoding IFNλ-2 comprises a coding region as shown in SEQ ID NO:3.


In one embodiment the IFNλ protein encoded by the mRNA comprised in the pharmaceutical composition is IFNλ-3, preferably IFNλ-3 comprising the amino acid sequence as shown in SEQ ID NO:6. In a more preferred embodiment the mRNA encoding IFNλ-3 comprises a coding region as shown in SEQ ID NO:5.


In some embodiments of the present invention the polyribonucleotide employed according to the present invention may contain unmodified and modified nucleotides. The term “unmodified nucleotide” used herein refers to A, C, G and U nucleotides. The term “modified nucleotide” used herein refers to any naturally occurring or non-naturally occurring isomers of A, C, G and U nucleotides as well as to any naturally occurring or naturally occurring analogs, alternative or modified nucleotide or isomer thereof having for example chemical modifications or substituted residues. Modified nucleotides can have a base modification and/or a sugar modification. Modified nucleotides can also have phosphate group modifications, e.g., with respect to the 5′-prime cap of an mRNA molecule. Modified nucleotides also include nucleotides that are synthesized post-transcriptionally by covalent modification of the nucleotides. Further, any suitable mixture of non-modified and modified nucleotides is possible. A non-limiting number of examples of modified nucleotides can be found in the literature (e.g. Cantara et al., Nucleic Acids Res, 2011, 39(Issue suppl_1): D195-D201; Helm and Alfonzo, Chem Biol, 2014, 21(2): 174-185; Carell et al., Angew Chem Int Ed Engl, 2012, 51(29):7110-31) and some preferable modified nucleotides are mentioned exemplarily in the following based on their respective nucleoside residue:


1-methyladenosine, 2-methylthio-N6-hydroxynorvalyl carbamoyladenosine, 2-methyladenosine, 2′-O-ribosylphosphate adenosine, N6-methyl-N6-threonylcarbamoyladenosine, N6-acetyladenosine, N6-glycinylcarbamoyladenosine, N6-isopentenyladenosine, N6-methyladenosine, N6-threonylcarbamoyladenosine, N6,N6-dimethyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, N6-hydroxynorvalylcarbamoyladenosine, 1,2′-O-dimethyladenosine, N6,2′-O -dimethyladenosine, 2′-O-methyladenosine, N6,N6,2′-O-trimethyladenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl)adenosine, 2-methylthio-N6-methyladenosine, 2-methylthio-N6-isopentenyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6-2-methylthio-N6-threonyl carbamoyladenosine, 2-methylthio-N6-(cis -hydroxyisopentenyl)adenosine, 7-methyladenosine, 2-methylthio-adenosine, 2-methoxy -adenosine, 2′-amino-2′-deoxyadenosine, 2′-azido-2′-deoxyadenosine, 2′-fluoro-2′-deoxyadenosine, 2-aminopurine, 2,6-diaminopurine, 7-deaza-adenosine, 7-deaza-8-aza -adenosine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine; 2-thiocytidine, 3-methylcytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-hydroxycytidine, lysidine, N4-acetyl-2′-O-methylcytidine, 5-formyl-2′-O-methylcytidine, 5,2′-O-dimethylcytidine, 2-O-methylcytidine, N4,2′-O -dimethylcytidine, N4,N4,2′-O-trimethylcytidine, isocytidine, pseudocytidine, pseudoisocytidine, 2-thio-cytidine, 2′-methyl-2′-deoxycytidine, 2′-amino-2′-deoxycytidine, 2′-fluoro-2′-deoxycytidine, 5-iodocytidine, 5-bromocytidine, 2′-azido-2′-deoxycytidine, 2′-amino-2′-deoxycytidine, 2′-fluor-2′-deoxycytidine, 5-aza-cytidine, 3-methyl-cytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-5-methyl -cytidine, 4-thio-pseudoisocytidine, 4-thio-l-methyl-pseudoisocytidine, 4-thio-l-methyl-1-deaza-pseudoisocytidine, 1-methyl-l-deaza-pseudoisocytidine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-l-methyl -pseudoisocytidine, zebularine,5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio -zebularine, 2-thio-zebularine; 1-methylguanosine, N2,7-dimethylguanosine, N2-methylguanosine, 2′-O-ribosylphosphate guanosine, 7-methylguanosine, hydroxywybutosine, 7-aminomethyl-7-deazaguanosine, 7-cyano-7-deazaguanosine, N2,N2-dimethylguanosine, N2,7,2′-O-trimethylguanosine, N2,2′-O-dimethylguanosine, 1,2′-O-dimethylguanosine, 2′-O-methylguanosine, N2,N2,2′-O-trimethylguanosine, N2,N2J-trimethylguanosine, Isoguanosine, 4-demethylwyosine, epoxyqueuosine, undermodified hydroxywybutosine, methylated undermodified hydroxywybutosine, isowyosine, peroxywybutosine, galactosyl-queuosine, mannosyl-queuosine, queuosine, archaeosine, wybutosine, methylwyosine, wyosine, 7-aminocarboxypropyldemethylwyosine, 7-aminocarboxypropylwyosine, 7-aminocarboxypropylwyosinemethylester, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl -guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-methyl-6-thio -guanosine, N2-methyl-6-thio-guanosine, N2,N2-dimethyl-6-thio-guanosine, N1-methylguanosine, 2′-amino-3′-deoxyguanosine, 2′-azido-2′-deoxyguanosine, 2′-fluoro-2′-deoxyguanosine, 2-thiouridine, 3-(3-amino-3-carboxypropyl)uridine, 3-methyluridine, 4-thiouridine, 5-methyl-2-thiouridine, 5-methylaminomethyluridine, 5-carboxymethyluridine, 5-carboxymethylaminomethyluridine, 5-hydroxyuridine, 5-methyluridine, 5-taurinomethyluridine, 5-carbamoylmethyluridine, 5-(carboxyhydroxymethyl)uridine methyl ester, dihydrouridine, 5-methyldihydrouridine, 5-methylaminomethyl-2-thiouridine, 5-(carboxyhydroxymethyl)uridine, 5-(carboxyhydroxymethyl)-2′-O-methyluridine methyl ester, 5-(isopentenylaminomethyl)uridine, 5-(isopentenylaminomethyl)-2-thiouridine, 3,2′-O-dimethyluridine, 5-carboxymethylaminomethyl-2′-O-methyluridine, 5-carbamoylhydroxymethyluridine, 5-carbamoylmethyl-2′-O-methyluridine, 5-carbamoylmethyl-2-thiouridine, 5-methoxycarbonylmethyl-2′-O-methyluridine, 5-(isopentenylaminomethyl)-2′-O-methyluridine, 5,2′-O-dimethyluridine, 2′-O -methyluridine, 2′-O-methyl-2-thiorudine, 2-thio-2′-O-methyluridine, uridine 5-oxyacetic acid, 5-methoxycarbonylmethyluridine, uridine 5-oxyacetic acid methyl ester, 5-methoxyuridine, 5-aminomethyl-2-thiouridine, 5-carboxymethylaminomethyl-2-thiouridine, 5-methylaminomethyl-2-selenouridine, 5-methoxycarbonylmethyl-2-thiouridine, 5-taurinomethyl-2-thiouridine, pseudouridine, 1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine, 1-methylpseudouridine, 3-methylpseudouridine, 2′-O -methylpseudouridine, 5-formyluridine, 5-aminomethyl-2-geranyluridine, 5-taurinomethyluridine, 5-iodouridine, 5-bromouridine, 2′-methyl-2′-deoxyuridine, 2′-amino -2′-deoxyuridine, 2′-azido-2′-deoxyuridine, 2′-fluoro-2′-deoxyuridine, inosine, 1-methylinosine, 1,2′-O-dimethylinosine, 2′-O-methylinosine, 5-aza-uridine, 2-thio-5-aza -uridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1-taurinomethyl-4-thio -uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-l-methyl-pseudouridine, 2-thio -l-methyl-pseudouridine, 1-methyl-l-deaza-pseudouridine, 2-thio-1-methyl-l-deaza -pseudouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio -dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy pseudouridine, 4-methoxy-2-thio-pseudouridine, 1,2′-O-dimethyladenosine, 1,2′-O- -dimethylguanosine, 1,2′-O-dimethylinosine, 2,8-dimethyladenosine, 2-methylthiomethylenethio-N6-isopentenyl-adenosine, 2-geranylthiouridine, 2-lysidine, 2-methylthio cyclic N6-threonylcarbamoyladenosine, 2-methylthio-N6-(cis -hydroxyisopentenyl) adenosine, 2-methylthio-N6-hydroxynorvalylcarbamoyladenosine, 2-methylthio-N6-threonylcarbamoyladenosine, 2-selenouridine, 2-thio-2′-O -methyluridine, 2′-O-methyladenosine, 2′-O-methylcytidine, 2′-O-methylguanosine, 2′-O -methylinosine, 2′-O-methylpseudouridine, 2′-O-methyluridine, 2′-O-methyluridine 5-oxyacetic acid methyl ester, 2′-O-ribosyladenosinephosphate, 2′-O -ribosylguanosinephosphate, 3,2′-O-dimethyluridine, 3-(3-amino-3-carboxypropyl)-5,6-dihydrouridine, 3-(3-amino-3-carboxypropyl)pseudouridine, 5,2′-O-dimethylcytidine, 5,2′-O-dimethyluridine, 5-(carboxyhydroxymethyl)-2′-O-methyluridine methyl ester, 55-(isopentenylaminomethyl)-2′-O-methyluridine, 5-aminomethyl-2-geranylthiouridine, 5-aminomethyl-2-selenouridine, 5-aminomethyluridine, 5-carbamoylmethyl-2′-O -methyluridine, 5-carboxyhydroxymethyluridine, 5-carboxymethyl-2-thiouridine, 5-carboxymethylaminomethyl-2-geranylthiouridine, 5-carboxymethylaminomethyl-2-selenouridine, 5-carboxymethylaminomethyl-2′-O-methyluridine, 5-cyanomethyluridine, 5-formyl-2′-O-methylcytidine, 5-methoxycarbonylmethyl-2′-O-methyluridine, 5-methylaminomethyl-2-geranylthiouridine, 7-aminocarboxypropyl-demethylwyosine, 7-methylguanosine, 8-methyladenosine, N2,2′-O-dimethylguanosine, N2,7,2′-O -trimethylguanosine, N2,7-dimethylguanosine, N2,N2,2′-O-trimethylguanosine, N2,N2,7-trimethylguanosine, N2,N2,7-trimethylguanosine, N4,2′-O-dimethylcytidine, N4,N4,2′-O -trimethylcytidine, N4,N4-dimethylcytidine, N4-acetyl-2′-O-methylcytidine, N6,2′-O -dimethyladenosine, N6,N6,2′-O-trimethyladenosine, N6-formyladenosine, N6-hydroxymethyladenosine, agmatidine, 2-methylthio cyclic N6-threonylcarbamoyladenosine, glutamyl-queuosine, guanosine added to any nucleotide, guanylylated 5′ end, hydroxy-N6-threonylcarbamoyladenosine; most preferably pseudo -uridine, N1-methyl-pseudo-uridine, 2′-fluoro-2′-deoxycytidine, 5-iodocytidine, 5-methylcytidine, 2-thiouridine, 5-iodouridine and/or 5-methyl-uridine.


Furthermore, the term “modified nucleotide” comprises nucleotides containing isotopes such as deuterium. The term “isotope” refers to an element having the same number of protons but different number of neutrons resulting in different mass numbers. Thus, isotopes of hydrogen for example are not limited to deuterium, but include also tritium. Furthermore, the polyribonucleotide can also contain isotopes of other elements including for example carbon, oxygen, nitrogen and phosphor. It is also possible that modified nucleotides are deuterated or contain another isotope of hydrogen or of oxygen, carbon, nitrogen or phosphor.


The total number of modified nucleotide types in the polyribonucleotide can be 0, 1, 2, 3, or 4. Hence, in some embodiments, at least one nucleotide of one nucleotide type, e.g. at least one U nucleotide, can be a modified nucleotide. In some embodiments, at least one nucleotide of in total two nucleotide types, e.g. at least one U nucleotide and at least one C nucleotide, can be a modified nucleotide. In some embodiments, at least one nucleotide of in total three nucleotide types, e.g. at least one G nucleotide, at least one U nucleotide and at least one C nucleotide, can be a modified nucleotide. In some embodiments, at least one nucleotide of all four nucleotide types can be a modified nucleotide. In all these embodiments one or more nucleotides per nucleotide type can be modified, the percentage of said modified nucleotides of per nucleotide type being 0%, 2.5%, 5%, 7.5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 100%.


In some embodiments, the total percentage of modified nucleotides comprised in the mRNA molecules to be purified is 0%, 2.5%, 5%, 7.5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 100%.


In a preferred embodiment the mRNA is an mRNA which contains a combination of modified and unmodified nucleotides. Preferably, it is an mRNA containing a combination of modified and unmodified nucleotides as described in WO2011/012316. The mRNA described therein is reported to show an increased stability and diminished immunogenicity. In a preferred embodiment, in such a modified mRNA 5 to 50% of the cytidine nucleotides and 5 to 50% of the uridine nucleotides are modified. The adenosine-and guanosine-containing nucleotides can be unmodified. The adenosine and guanosine nucleotides can be unmodified or partially modified, and they are preferably present in unmodified form. Preferably 10 to 35% of the cytidine and uridine nucleotides are modified and particularly preferably the content of the modified cytidine nucleotides lies in a range from 7.5 to 25% and the content of the modified uridine nucleotides in a range from 7.5 to 25%. It has been found that in fact a relatively low content, e.g. only 10% each, of modified cytidine and uridine nucleotides can achieve the desired properties. It is particularly preferred that the modified cytidine nucleotides are 5-methylcytidin residues and the modified uridine nucleotides are 2-thiouridin residues. Most preferably, the content of modified cytidine nucleotides and the content of the modified uridine nucleotides is 25%, respectively.


In certain embodiments of any of the foregoing, the percentage of analogs of a given nucleotide refers to input percentage (e.g., the percentage of analogs in a starting reaction, such as a starting in vitro transcription reaction). In certain embodiments of any of the foregoing, the percentage of analogs of a given nucleotide refers to output (e.g., the percentage in a synthesized or transcribed compound). Both options are equally contemplated.


The mRNA may be produced recombinantly in in vivo systems by methods known to a person skilled in the art.


Alternatively, the modified RNA, preferably the mRNA molecules of the present invention may be produced in an in vitro system using, for example, an in vitro transcription system which is known to the person skilled in the art. An in vitro transcription system capable of producing RNA, preferably mRNA requires an input mixture of modified and unmodified nucleoside triphosphates to produce modified mRNA molecules.


Furthermore, the modified RNA, preferably mRNA molecules may be chemically synthesized, e.g., by conventional chemical synthesis on an automated nucleotide sequence synthesizer using a solid-phase support and standard techniques or by chemical synthesis of the respective DNA sequences and subsequent in vitro or in vivo transcription of the same.


The coding region comprised in the mRNA and which encodes an IFNλ protein can be a partly or fully codon optimized sequence. Codon optimization refers to a technique which is applied to maximize protein expression by increasing the translational efficiency of the respective polyribonucleotide as in some cases codons exist that are preferentially used by some species for a given amino acid. Examples of codon optimized coding regions are the coding regions depicted in SEQ ID NO: 1, 3 and 5.


Further, said polyribonucleotide might comprise further modifications to adjust and/or extend the duration of action. Said polyribonucleotide might also contain an m7GpppG cap, an internal ribosome entry site (IRES) and/or a polyA tail at the 3′ end and/or additional sequences for promoting translation.


In addition, the polyribonucleotide employed according to the present invention may also comprise further functional regions and/or 3′ or 5′ non-coding regions. The 3′ and/or 5′ non-coding regions can be sequences which naturally flank the encoded protein or artificial sequences which contribute to the stabilization and/or regulation of said polyribonucleotide. Suitable sequences may be identified and investigated by routine experiments. Further, said polyribonucleotide can also have further functional regions and may be combined with regulatory elements and target sequences of micro-RNAs for example for spatial and temporal control the activity of the desired polyribonucleotide comprising a sequence which encodes a protein, i.e. for example with respect to specific cells or cell types and/or developmental stages or specific time frames.


In one embodiment, the mRNA also contains a 5′- and/or 3′-UTR. In a particularly preferred embodiment, the UTR sequence is a 5′-UTR sequence as described in WO 2017/167910. Even more preferably, the mRNA contains a 5′-UTR sequence directly upstream of the start codon of the coding region which shows the following sequence: GGGAGACGCCACC (SEQ ID NO:7)


In a further preferred embodiment, the mRNA also contains a 3′-UTR, preferably a 3′-UTR of the sequence 5′-TTCG-3′.


In another preferred embodiment, the mRNA is transcribed from a DNA molecule as described in WO 2017/167910. More preferably, such a DNA molecule comprises one strand with the following elements:

    • (a) a coding region, including a start codon at its 5′ end, coding for an IFNλ polypeptide; and
    • (b) directly upstream of said coding sequence the sequence GGGAGACGCCACC (SEQ ID NO:7) and upstream of this sequence a promoter which is recognized by a DNA-dependent RNA polymerase, preferably a promoter with the sequence TAATACGACTCACTATA (SEQ ID NO: 8) which is recognized by a T7 DNA-dependent RNA polymerase.


Thus, in such an embodiment the sequence upstream of the start codon of the coding region is TAATACGACTCACTATA GGGAGACGCCACC (SEQ ID NO:9)


In a particularly preferred embodiment, the mRNA is transcribed from a DNA molecule as shown in any one of SEQ ID NOs: 10 to 12. SEQ ID NO: 10 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNλ-1. SEQ ID NO: 11 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNλ-2. SEQ ID NO: 12 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNλ-3.


The mRNA which codes for IFNλ can be combined with an mRNA which encodes another type of interferon, preferably with a type I interferon or a type II interferon. In a preferred embodiment the type I interferon is selected from the group consisting IFN-β, IFN-α1, IFN-α2, IFN-α8, IFN-α14, IFN-α17, IFN-α4, IFN-α5, IFN-α6, IFN-α7, IFN-α10, IFN-α16 and IFN-α21 or any combination thereof. In a particularly preferred embodiment the type I interferon is IFN-α16 or IFN-β. In another preferred embodiment the other type of interferon is a type II interferon, in particular IFNγ. For such an mRNA which encodes another interferon, the same applies in connection with the preferred embodiments (e.g. possible modifications, promoter sequences, UTRs etc.) as has been described above for the mRNA encoding IFNλ.


An exemplary RNA sequence encoding IFN-α16 is shown in SEQ ID NO: 16. The encoded protein is shown in SEQ ID NO: 17.


An exemplary RNA sequence encoding IFN-β is shown in SEQ ID NO: 18. The encoded protein is shown in SEQ ID NO: 19.


An exemplary RNA sequence encoding IFNγ is shown in SEQ ID NO: 20. The encoded protein is shown in SEQ ID NO: 21.


An exemplary DNA sequence which can be transcribed into an mRNA encoding IFN-α16 is shown in SEQ ID NO: 22.


An exemplary DNA sequence which can be transcribed into an mRNA encoding IFN-β is shown in SEQ ID NO: 23.


An exemplary DNA sequence which can be transcribed into an mRNA encoding IFNγ is shown in SEQ ID NO: 24.


An exemplary mRNA sequence encoding IFN-α16 is shown in SEQ ID NO: 25.


An exemplary mRNA sequence encoding IFN-β is shown in SEQ ID NO: 26.


An exemplary mRNA sequence encoding c is shown in SEQ ID NO: 27.


The mRNA which is to be administered in accordance with the present invention is in the form of a pharmaceutical composition. In accordance with this invention, the term “pharmaceutical composition” relates to a composition in for administration to a subject. Exemplary subjects include a mammal such as a dog, cat, pig, cow, sheep, horse, rodent, e.g., rat, mouse, and guinea pig, or a primate, e.g., gorilla, chimpanzee, and human. In a most preferable embodiment, the subject is a human.


Generally, the RNA is included in an effective amount in the pharmaceutical composition. The term “effective amount” refers to an amount sufficient to induce a detectable therapeutic response in the subject to which the pharmaceutical composition is to be administered.


The pharmaceutical composition can comprise a pharmaceutically acceptable carrier, i.e. chemical compounds, materials, ingredients, and/or compositions, which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Thus, a pharmaceutically acceptable carrier is an inactive substance formulated alongside the pharmaceutically active substance for facilitating its handling in view of dosage, adsorption, solubility or pharmacokinetic considerations. Examples of suitable pharmaceutical acceptable carriers are well known in the art and include phosphate buffered saline solutions, buffer, water, emulsions, such as oil/water emulsions, various types of wetting agents, and sterile solutions. In particularly, aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and organic esters such as ethyl oleate. Further examples of pharmaceutically acceptable carriers include but are not limited to saline, Ringer's solution and dextrose solution, citrate, phosphate, and other organic acids; salt-forming counter-ions, e.g. sodium and potassium; low molecular weight (>10 amino acid residues) polypeptides; proteins, e.g. serum albumin, or gelatine; hydrophilic polymers, e.g. polyvinylpyrrolidone; amino acids such as histidine, glutamine, lysine, asparagine, arginine, or glycine; carbohydrates including glucose, mannose, or dextrins; monosaccharides; disaccharides; other sugars, e.g. sucrose, mannitol, trehalose or sorbitol; chelating agents, e.g. EDTA; non-ionic surfactants, e.g., polyoxyethylene sorbitan monolaurate, available on the market with the commercial name Tween, propylene glycol, Pluronics or polyethylene glycol; antioxidants including methionine, ascorbic acid and tocopherol; and/or preservatives, e.g. octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, e.g. methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol). Suitable pharmaceutically acceptable carriers and their formulations are described in greater detail in Remington's Pharmaceutical Sciences, 17th ed., 1985, Mack Publishing Co. Furthermore, preservatives, stabilizers and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases, nanosystems or liposomes, and the like.


The administration of the mRNA encoding an IFNλ protein for the treatment and/or prevention of a viral-induced disorder can be achieved by means and methods known to the person skilled in the art, in particular for ensuring that the mRNA reaches the intended target tissue/target cells. Possible routes are, e.g., intravenous, intramuscular, intradermal, subcutaneous and delivery into the respiratory system.


If the viral-induced disorder is a respiratory disorder, the administration into the respiratory system is preferred. Possibilities for delivery into the respiratory system include instillation and inhalation. Alternatively, for delivery of the mRNA into the lung, also an i.v. administration with formulations that have a tropism for the lung is possible.


In a preferred embodiment, the pharmaceutical composition comprising the mRNA encoding an IFN-λ polypeptide is administered to a patient via inhalation.


The mRNA can be inhaled in any form which is suitable for inhalation. In a preferred embodiment, the mRNA is present in the pharmaceutical composition in a form suitable for inhalation in the form of an aerosol. A particularly suitable way of administering the mRNA to the respiratory system of a patient is by nebulization.


In a preferred embodiment, the inhalation is bolus inhalation. This means that during inhalation only for a short amount of time the aerosol containing the active agent is mixed to the inhaled air. If the aerosol containing the active agent is mixed with the air at the beginning of the inhalation, the active agent reaches with the first part of the inhaled air the deeper parts of the lung. If at the end of inhalation the addition of the active agent is stopped, no active agent is deposited in the central region of the lung, i.e. the airways, at the end of the inhalation. By using bolus inhalation it is possible to better target the different regions of the lung, e.g. the periphery of the lung or the central region, as regards the deposition of the active agent, depending on the requirements of the underlying disease state.


The mRNA can advantageously be combined in the pharmaceutical composition with compounds which ease delivery of the mRNA to the target cells or the target tissue and/or which increase its stability. One possibility in this regard is the formation of the RNA into nanoparticles with suitable substances such as those described, e.g. in PCT/EP2020/053774.


Examples for preferred agents or reagents for delivering and/or introducing the RNA into a target cell or a target tissue are in this context liposomal transfection reagents (LTR'S). One particular mode for delivering and/or introducing the mRNA into target cells or target tissue is transfection. Hence, the mRNA to be employed can be envisaged to be transfected (into (target) cells or tissue), to be delivered/administered via transfection and/or to be prepared for transfection. Means and methods for transfecting mRNA are well known in the art and are, for example, described in Tavernier (loc. cit.), Yamamoto (Eur J Pharm Biopharm. 71(3) (2009), 484-9) and Kormann (Nat Biotechnol. 29(2) (2011), 154-7). Particular modes of transfection are lipofection, magnetofection, magnetolipofection or complexation with polymers. Hence, the mRNA to be employed may be prepared for lipofection, prepared to be transfected by lipofection, delivered/introduced via lipofection and/or administered via lipofection.


Thus, the pharmaceutical composition may (further) comprise at least one lipid or liposomal transfection reagent or enhancer (LTR; liposomal transfection reagent). The mRNA to be employed may be comprised in, complexed with and/or delivered by the LTR. In particular, the mRNA to be employed may be comprised in and/or delivered by (respective) lipofection complexes comprising the mRNA and the LTR. The pharmaceutical composition may (further) comprise the lipofection complexes.


LTRs are known in the art and are, for example, distributed by OzBiosciences, Marseille, France. For example, such LTRs may be lipids or lipidoids, preferably cationic lipids or cationic lipidoids, like the lipidoids as disclosed in PCT/EP2014/063756 (e.g. C12-(2-3-2), the lipids as disclosed in EP2285772 (e.g. Dogtor) and the lipopolyamines as disclosed in EP1003711 (e. g. DreamFect™ and DreamFect Gold™). A particular LTR may be selected from the group consisting of

    • (i) C12-(2-3-2);
    • (ii) Dream Fect™, preferably DreamFect Gold™ (DF™M/DF-Gold™; OzBiosciences, Marseille, France);
    • (iii) Dogtor (OzBiosciences, Marseille, France); and
    • (iv) Lipofectamine like, for example, Lipofectamine 2000 (Invitrogene, CA, USA).


In principle, Dogtor is a preferred, DreamFect™ is a more preferred and DF-Gold™ and C12-(2-3-2) are even more preferred LTR(s).


LTRs like Dogtor are, for example, described in EP2285772. LTRs like DF™ or DF-Gold™ are, for example, described in EP1003711. In principle, the oligomers, polymers or lipidoids as disclosed in PCT/EP2014/063756, the particular cationic lipids as disclosed in EP2285772 and the particular lipopolyamines as disclosed in EP1003711 are preferred LTRs. LTRs like C12-(2-3-2) and DF-Gold™ are most preferred.


Non-limiting examples of lipofection complexes are DF-Gold™/RNA lipoplexes and C12-(2-3-2)/RNA lipoplexes.


C12-(2-3-2) is a particularly preferred LTR having the structure shown in formula (V) (cf. Jarzębińska et al., Angew Chem Int Ed Engl., 2016; 55(33):9591-5):




embedded image


C12-(2-3-2) is preferably prepared as described e.g. in WO 2016/075154 A1, EP 3013964, and Jarzębińska et al. (Angew Chem Int Ed Engl., 2016; 55(33):9591-5). The cationic lipidoid can be prepared by mixing N1-(2-aminoethyl)-N3-(2-((3,4-dimethoxybenzyl)amino)ethyl)propane-1,3-diamine (8.9 g, 1 eq., 28.67 mmol) with 1,2-Epoxydodecane (42.27, 8 eq., 229.4 mmol) and mixed for 24 h at 80° C. under constant shaking followed by purification and removal of the 3,4-dimethoxybenzyl protection group.


Different isomers, of C12-(2-3-2) can be used such as a racemate, an S-isomer, and/or an R-isomer. Preferably, C12-(2-3-2) is used as pure R-isomer and has the structure shown in formula (VI). For obtaining pure R-isomers of C12-(2-3-2) it can be prepared as described above for C12-(2-3-2) using the R-isomer of 1,2-Epxoydodecane for synthesis.




embedded image


Hence, in preferred embodiments the pharmaceutical composition comprises an mRNA encoding IFNλ and further comprises a lipidoid having the structure shown in formula (V), preferably as shown in formula (VI).


A further particularly preferred LTR is a cationic lipidoid having formula (VII), herein also referred to as “dL_P” which can be synthesized via reaction of N, N′-Bis(2-aminoethyl) -1,3-propanediamine with N-Dodecylacrylamide using boric acid as catalyst. For the reaction, the mixture can be stirred at 100° C. under microwave irradiations.


Hence, in further preferred embodiments the pharmaceutical composition comprises an mRNA encoding IFNλ and further comprises a lipidoid having the structure shown in formula (VII).




embedded image


Furthermore, the pharmaceutical composition comprises a cationic lipidoid having formula (V), (VI) and/or (VII), preferably dL_P and/or C12-(2-3-2), more preferably dL_P and/or the R-isomer of C12-(2-3-2), comprised in a formulation as described in the following. In particular, the herein described agents and reagents for delivering and/or introducing the RNA into a target cell or a target tissue and the herein described LTRs may be combined with one or more (e.g. two, three or four) further lipid(s) (like, for example, cholesterol, DPPC, DOPE and/or PEG-lipids (e.g. DMPE-PEG, DMG-PEG2000)). These further lipids may support the desired function of the agents/reagents and LTRs (support and/or increase the delivering and/or introducing of RNA into the cell or tissue and improve transfection efficiency, respectively) and function as respective “helper lipids”. Particular examples of such “helper lipids” are cholesterol, DPPC, DOPE and/or PEG-lipids (e.g. DMPE-PEG, DMG-PEG (e.g. DMG-PEG2000). The further lipids (e.g. “helper lipids”) may also be part(s) of the herein disclosed complexes/particles. The skilled person is readily in the position to prepare complexes/particles in accordance with the invention. Examples of further lipids (e.g. “helper lipids”) are also known in the art. The skilled person is readily in the position to choose suitable further lipids (e.g. “helper lipids”) and ratios of the agents/reagents/LTRs and the further lipids (e.g. “helper lipids”). Such ratios may be molar ratios of 1-4:1-5, 3-4:4-6, about 4:about 5, about 4:about 5.3 of agents/reagents/LTRs: further lipid(s) (the more narrow ranges are preferred). For example, the agents/reagents/LTRs may be combined with three further lipids, like DPPC, cholesterol, and DMG-PEG2000, at a molar ratio of 8:5.3:4.4:0.9, respectively, or, more particular, 8:5.29:4.41:0.88, respectively.


Preferably, dL_P and/or C12-(2-3-2), more preferably dL_P and/or the R-isomer of C12-(2-3-2), is generated as described above and used with helper lipids DPPC and cholesterol and PEG-lipid DMG-PEG2000 at the molar ratios 8:5.29:4.41:0.88 for formulating lipoid particles.


A composition in which the R-isomer of C12-(2-3-2) (formula VI) is formulated with the lipids DPPC and cholesterol and PEG-lipid DMG-PEG2000 at the molar ratios 8:5.29:4.41:0.88 is also referred herein as “LF92”. A composition in which the dL_P (formula VII) is formulated with the lipids DPPC and cholesterol and PEG-lipid DMG-PEG2000 at the molar ratios 8:5.29:4.41:0.88 is also referred herein as “LF111”. As also exemplarily described e.g. in WO 2016/075154 A1, EP 3013964, and Zhang et al. (TERMIS, 2019, Tissue Engineering: Part A, Vol. 25, Numbers 1 and 2), dL_P and/or C12-(2-3-2) can be used as a non-viral vector, to make a stable lipoplex with mRNA molecules, based on electrostatic interaction between the positive amino groups of lipidoid and negative phosphate groups of mRNA molecules (Anderson, Human Gene Therapy 14, 2003, 191-202). To stabilize the lipoplex structure and reduce the leakage, dL_P and/or C12-(2-3-2), more preferably dL_P and/or the R-isomer of C12-(2-3-2), can be supplied with two helper lipids entitled 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and cholesterol (Anderson, Drug Delivery 11, 2004, 33-39; Liang, Journal of Colliod and Interface Science 278, 2004, 53-62). At the end, 1,2-Dimyristoyl-sn-glycerol, methoxypolyethylene Glycol (DMG-PEG) 2kD (DMG-PEG2000) is to be added to the lipid mix to provide a PEGylated liposome. It is already well known that PEGylation improves the physico-chemical characteristic of liposome formulation by increasing water solubility, protecting from enzymatic degradation, and limiting immunogenic and antigenic reactions (Milla, Current Drug Metabolism 13, 2012, 105-119). Final N/P ratios for entire ethanoic lipid mixture are to be 8:5.29:4.41:0.88 standing for molar ratios of amino group of dL_P and/or C12-(2-3-2)/DPPC/cholesterol/DMG-PEG2000, respectively, to one phosphate group of mRNA molecule.


Hence, in preferred embodiments the pharmaceutical composition comprises an mRNA encoding IFNλ and further comprises dL_P and/or C12-(2-3-2), preferably dL_P and/or the R-isomer of C12-(2-3-2), formulated with DPPC, cholesterol, and DMG-PEG2000. Moreover, in particularly preferred embodiments the pharmaceutical composition comprises an mRNA encoding IFNλ and further comprises dL_P and/or C12-(2-3-2), preferably dL_P and/or the R-isomer of C12-(2-3-2), formulated with DPPC, cholesterol, and DMG-PEG2000 with final N/P ratios for entire ethanoic lipid mixture of 8:5.29:4.41:0.88 for molar ratios of amino group of dL_P and/or C12-(2-3-2)/DPPC/cholesterol/DMG-PEG2000, respectively, to one phosphate group of mRNA molecule.


R-isomers of C12-(2-3-2) formulated with DPPC, cholesterol, and DMG-PEG2000 as stated above are also referred to as LF92 formulation.


Hence, in particularly preferred embodiments of the pharmaceutical composition comprising an mRNA encoding IFNλ, said pharmaceutical composition further comprises an LF92 formulation.


dL_P formulated with DPPC, cholesterol, and DMG-PEG2000 as stated above are also referred to as LF111 formulation.


Hence, in another preferred embodiment of the pharmaceutical composition comprising an mRNA encoding IFNλ, said pharmaceutical composition further comprises an LF111 formulation.


When the pharmaceutical composition is administered via inhalation to a human patient, one dose to be inhaled by a patient contains preferably between 200 μg and 15 mg of mRNA encoding an IFN-λ polypeptide. As shown in the appended Examples, experiments in mice show that an induction of downstream targets of IFNλ (which is indicative of antiviral activity) can already be achieved with extremely low levels of IFNλ mRNA when administered via instillation into the lung. When extrapolated to human patients based on the weight of the lung, it can be expected that effective amounts of IFNλ mRNA will lie in the range of 200 μg to 15 mg per dose, preferably between 250 μg and 5 mg per dose, even more preferably 250 μg and 1 mg per dose, even more preferably between 250 μg and 750 μg per dose.


The number of doses to be administered to a subject depend on the actual purpose. In the case of an acute viral infection, the administration is preferably effected:

    • One dose once (e.g. in cases of a mild infection); or
    • One dose every two days over a period of 2, 4, 6, 8, 10 or 14 days; or
    • One dose daily over a period of time of 2, 3, 4, 5, 6, 7, 8, 9, or 10 days; or
    • Two doses daily over a period of time of 2, 3, 4, 5, 6, 7, 8, 9, or 10 days.


In the case of a prevention of a virus infection or of a virus-induced exacerbation of lung diseases such as asthma or COPD the administration is preferably effected:

    • One dose per week, preferably for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 weeks, preferably for 1, 2, 3, 4, 5, 6 or 7 months.


The present invention also relates to an mRNA molecule comprising a sequence as shown in any one of SEQ ID Nos: 13 to 15, 25, 26 or 27.


SEQ ID Nos: 13 to 15, 25, 26 and 27 contain a codon optimized coding region for IFNλ-1, -2 and -3, of IFN α16, IFNβ and IFNγ, respectively. Moreover, each of these mRNAs contains directly upstream of the start codon the sequence GGGAGACGCCACC (SEQ ID NO:7) and as a 3′-UTR the sequence 5′-TTCG-3′.


Preferably, such an mRNA further comprises a poly-A tail, e.g. of about 200 nucleotides.


The present invention also relates to a DNA molecule comprising a sequence as shown in any one of SEQ ID NOs: 10 to 12 or 22 to 24. SEQ ID NO: 10 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNλ-1. SEQ ID NO: 11 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNλ-2. SEQ ID NO: 12 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNλ-3. SEQ ID NO: 22 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFN α16. SEQ ID NO: 23 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNβ. SEQ ID NO: 24 shows a DNA molecule for the transcription of an mRNA molecule which encodes IFNγ.






FIG. 1 shows IFNλ1 concentration in supernatants of cells treated with rec. hIL-29. Supernatants of A-549 cells treated with doses of rec. hIL-29 as indicated in x-axis were collected and stored at −80° C. At day of analysis, hIL-29 ELISA was performed according to kit protocol. TMB was incubated for 15 min. Delta O.D of 650 and 450 nm was used for analysis using GraphPad Prism V.8.



FIG. 2 shows IFNλ1 concentrations in supernatants of cells transfected with IFNλ mRNA. Supernatants of A-549 cells transfected with doses of mRNA coding for hIL-29, as indicated in x-axis, were collected and stored at −80° C. At day of analysis, hIL-29 ELISA was performed according to kit protocol. TMB was incubated for 15 min. Delta O.D of 450 and 650 nm is used for analysis using GraphPad Prism V.8.



FIG. 3 shows target activation after treatment or transfection with high dose. A549 cells were transfected with 15 ng/well IFN-Lambda mRNA and stimulated with 2.9 ng/well human rec. hIL-29. As a control, A549 were transfected with 15 ng/well BMP2-Stop encoding mRNA. At different time points (6, 10, 24, 48, 72 and 120 h) cells for Real-Time PCR analysis are collected and lysed.



FIG. 4 shows target induction after treatment or transfection with mid dose. A549 cells were transfected with 4 ng/well IFN-Lambda mRNA and stimulated with 0.6 ng/well human rec. hIL-29. As a control, A549 were transfected with 4 ng/well BMP2-Stop encoding mRNA. At different time points (6, 10, 24, 48, 72 and 120 h) cells for Real-Time PCR analysis are collected and lysed.



FIG. 5 shows target activation after transfection or treatment with low dose. A549 cells were transfected with 2 ng/well IFN-Lambda mRNA and stimulated with 0.3 ng/well human rec. hIL-29. As a control, A549 were transfected with 2 ng/well BMP2-Stop encoding mRNA. At different time points (6, 10, 24, 48, 72 and 120 h) cells for Real-Time PCR analysis were collected and lysed.



FIG. 6 shows target activation after treatment or transfection with very low dose. A549 cells were transfected with 1 ng/well IFN-Lambda mRNA and stimulated with 0.1 ng/well human rec. hIL-29. As a control, A549 were transfected with 1 ng/well BMP2-Stop encoding mRNA. At different time points (6, 10, 24, 48, 72 and 120 h) cells for Real-Time PCR analysis were collected and lysed



FIG. 7 shows the downstream target activation 6 hours post treatment with recombinant IFNλ1 or mRNA coding for IFNλ1 as assessed via qPCR for IFIT1, IFIT3, OAS3 and ISG15. Downstream target activation is plotted against measured IFNλ1 levels.



FIG. 8 shows:

    • (a) IFNλ1 downstream signaling in A-549 and A-549-ACE2 cells 48 hours after mRNA transfection with different doses. Similar induction patterns are observed for both cell lines.
    • (b) IFNλ1 downstream signaling in A-549 and A-549-ACE2 cells using recombinant protein after 48 hours. In this case higher induction levels are observed in A-549 cells.
    • (c) IFNλ1 expression in A-549-ACE2 cells: Transfection of A549-ACE2 cells with mRNA coding for IFNλ1 resulted in dose-dependent production of IFNλ1 after 48 hours.



FIG. 9 shows the IFNλ1 expression in supernatants of transfected cells after transfection of submersed cells. Comparable expression levels are observed between HEK293, A-549 and FreeStyle 293-F cells. Lower levels were observed in 16HBE14o-cells; untreated wells showed background in ELISA. Peak expression was observed at 24 h post transfection and levels remained constant at least up to 48 h post transfection. In all cell lines except 16HBE14o-cells, transfection doses below 4 ng/well were sufficient to reach an effective antiviral IFNλ concentration of 0.3 ng/ml at 6 h post transfection.



FIG. 10 shows:

    • (a) IFNλ1 downstream signaling in submersed A-549 cells after transfection with IFN-Lambda mRNA. The indicated values are normalized to UT (untransfected). An upregulation of all target genes was observed and peak expression was at 24 h after transfection. A very strong target inducement was observed, even with low mRNA doses.
    • (b) IFNλ1 downstream signaling in submersed 16HBE14o-cells after transfection with IFN-Lambda mRNA. The indicated values are normalized to UT (untransfected). An upregulation of all target genes (lower for OAS3) was observed and peak expression was at 24 h after transfection. Overall, a weak target inducement was observed compared to A-549 cells.



FIG. 11 shows:

    • (a) ELISA results for the detection of IFNλ1 in ALI cultures after transfection with IFNλ1 mRNA. Values were measured 24 h post transfection. The total amount of IFNλ1 was measured in the in apical and basolateral compartment. The detection of IFNλ1 was as follows: A-549>16HBE14o->Epithelix human primary wt ALIs. Maximum basal yield in Epithelix ALIs is 500 fold lower compared to that of A-549 ALIs.
    • (b) Measurement of the downstream target activation in ALI cultures 24 hours after transfection with IFNλ1 mRNA. The highest downstream target activation was observed in A-549 ALI culture. The indicated values are normalized to UT (untransfected). The same activation pattern was observed for all cell lines: ISG15>22 IFIT1≥IFIT3>OAS3.
    • (c) The endogenous levels of downstream targets in untreated 16HBE and Epithelix primary human ALIs compared to A-549 ALIs. 16HBE14o- and Epithelix human primary ALIs have up to 40-fold higher basal endogenous levels of downstream targets than A-549 ALIs.



FIG. 12 shows:

    • (a) The quantification of IFNλ1 mRNA in lung homogenate of treated mice 5 and 24 hours after treatment. IFNλ1 mRNA is detected dose-dependently at 5 h post application with very low levels at 24 h post application.
    • (b) The results of an ELISA assay detecting IFNλ1 in lung homogenate and in BALF at 5 h and 24 h post application. IFNλ1 was measurable in lung homogenate and in BALF at 5 h post application. 24 h post application only low levels of IFNλ1 were detectable.
    • (c) IFNλ1 downstream activation in lung homogenate. The functionality of human IFNλ1 mRNA in mouse is demonstrated. The indicated values are normalized to UT (untreated) mouse. Target genes are highly induced 5 h post application and still upregulated at 24 h post application although less strongly. OAS3 is induced less strongly compared to other target genes but induction is higher at 24 h than 5 h.



FIG. 13 shows comparison of hIFNλ1 mRNA transfection with recombinant protein (see also Example 8).



FIG. 14 shows hIFNλ ELISA from supernatants of transfected A-549 cells: LF92 vs. commercially available transfection reagent (see also Example 9).



FIG. 15 shows Target gene induction by LF92 formulated mRNA (see also Example 10).



FIG. 16 shows in vitro tolerability of LF92 formulated mRNA encoding hIFNλ1 (see also Example 11).



FIG. 17 shows correlation of mRNA translation and target gene induction of (a) IFIT1 (b) OAS3 (c) ISG15 and (d) MX1 (see also Example 12).



FIG. 18 shows (a) SARS-COV-2 and (b) IAV inhibition in vitro assessed by qPCR (see also Example 13).



FIG. 19 shows detection of hIFNλ1 mRNA in lung homogenate (see also Example 14).



FIG. 20 shows detection of hIFNλ1 protein in lung homogenate (see also Example 15).



FIG. 21 shows target gene activation of (a) IFIT1 (b) IFIT3 (c) ISG15 and (d) OAS3 in lung tissue after hIFNλ1 mRNA application (see also Example 16).



FIG. 22 shows detection of chemokines in plasma after single nasal application of hIFNλ1 mRNA (see also Example 17).



FIG. 23 shows target gene activation of (a) IFIT1 (b) IFIT3 (c) ISG15 and (d) OAS3 in lung tissue upon treatment with hIFNλ1 mRNA (see also Example 18).



FIG. 24 shows detection of hIFNλ1 mRNA deposited in the lung after nasal application in mice (see also Example 19).



FIG. 25 shows body weight during repeated administration of hIFNλ1 mRNA (see also Example 20).



FIG. 26 shows quantification of hIFNλ1 mRNA in ferret lung homogenate (see also Example 21).



FIG. 27 shows target gene activation in ferret lung homogenate (see also Example 22).



FIG. 28 shows quantification of IFN mRNA, target gene activation, and cytokine induction in ferret lung homogenate (see also Example 23).



FIG. 29 shows reduction of IAV replication by mRNAs encoding IFNs (see also Example 24).



FIG. 30 shows Reduction of SARS-COV2 replication by mRNA encoding hIFNλ1 (see also Example 25).





Other aspects and advantages of the invention will be described in the following examples, which are given for purposes of illustration and not by way of limitation. Each publication, patent, patent application or other document cited in this application is hereby incorporated by reference in its entirety.


EXAMPLES

Examples 1 and 2 described in the following aimed at assessing downstream target activation by IFNλ1 treatment with recombinant protein or mRNA coding for IFNλ1 in A-549 cells. For this purpose, A549 cells were incubated for 6 h with recombinant IFNλ1 or transfected with IFNλ1 coding mRNA and downstream activation was analyzed 6, 10, 24, 48, 72, 120 and 168 h post treatment via qPCR for IFIT1, IFIT3, OAS3 and ISG15. In addition, IFNλ1 levels were measured by hIL-29 ELISA in cell supernatant at all sampling time points.


Upon treatment of A-549 cells with recombinant IFNλ1, downstream target activation was observed at 6 h post treatment but declined rapidly at later time points after medium exchange. IFNλ1-ELISA revealed that about 30% of added protein was recovered in the supernatant at 6 h post treatment. Upon transfection of A-549 cells with mRNA coding for IFNλ1 and medium exchange 6 h post transfection, IFNλ1 was detectable in supernatants of these cells up to 168 h with a peak at 48 h. Downstream targets were highly induced also with low doses of transfected mRNA and remained upregulated up to 120 h post mRNA transfection


The following abbreviations and definitions are used in the appended Examples:
















Abbreviation
Description









DMEM
Dulbecco's Modified Eagle Medium



ELISA
Enzyme linked immunosorbent assay



FBS
Fetal bovine serum



IFNλ1
Interferon lambda 1



PBS
Phosphate buffered saline



qPCR
Quantitative Polymerase Chain Reaction



IFIT1/3
Interferon induced protein with




tetratricopeptide repeat 1/3



OAS3
2′-5′-oligoadenylate synthetase 3



ISG15
Interferon stimulated gene 15



MX1
MX dynamin like GTPase 1



IL-6/8
Interleukin 6/8



MCP-1
C-C motif chemokine ligand 2



CXCL9/10/11
C-X-C motif chemokine ligand 9/10/11



CXCL10
C-X-C motif chemokine ligand 10










In the appended Examples the following Materials and Methods were used.


1. Materials














Material
Supplier
Cat no.







MEM with GlutaMAX ™
Gibco
41090-028


FBS, heat-inactivated
Thermo Fisher Scientific
10500064


Penicillin-Streptomycin 100×
Thermo Fisher Scientific
15140122


Lipofectamine MessengerMax
Thermo Fisher Scientific
LMRNA008


DPBS 1× without Ca and Mg
Thermo Fisher Scientific
Gibco




14190-250


Human IL-29 ELISA Kit
Abcam
ab100568


Recombinant hIL-29
Abcam
ab155625
























TaqMan Probes
Supplier
Cat no.









IFIT3
Thermo Fisher Scientific
Hs01922752_s1



IFIT1
Thermo Fisher Scientific
Hs03027069_s1



OAS3
Thermo Fisher Scientific
Hs00196324_m1



ISG15
Thermo Fisher Scientific
Hs01921425_s1






















Device
Supplier
Cat no. or type







Countess Cell Counter
Thermo Fisher Scientific
C10281


Countess Cell Counting
Thermo Fisher Scientific
C10283


chamber slides


96-well cell culture plate
Corning
Costar 3599


96-well storage plate
Corning
Costar 3363


Microplate shaker
Heidolph
Titramax


Microplate reader
Tecan
Tecan Infinite




M200 Pro


Light Cycler ® 96
Roche
Light




Cycler ® 96









2. Methods
2.1 Cell Culture

A-549 cells were cultivated in MEM supplemented with 10% heat inactivated FBS and 1% P/S at 37° C., in a humidified atmosphere with 5% CO2. 24 h prior transfection 20.000 cells per well were seeded in 96-well plates in a total volume of 100 μL.


2.2 In Vitro Transcription

To generate templates for in vitro transcription, circular plasmids were linearized by restriction digestion with Bsp1191 and further purified by chloroform ethanol precipitation.


mRNA was produced using a standard in vitro transcription mix (including indicated modified triphosphate nucleotides) containing T7 RNA polymerase, inorganic pyrophosphatase, and RNase inhibitor. Co-transcriptional capping was achieved by addition of an ARCA cap analogue. For in vitro transcription of chemically modified RNA, 25% of Cytidine-5′-Triphosphate were replaced by 5-Methylcytidine-5′-Triphosphate and 25% Uridine-5′-Triphosphate were replaced by 2-Thiouridine-5′-Triphosphate (Jena Biosciences), respectively (cf. e.g. SEQ ID NO: 1). Residual template DNA was digested using DNasel. Subsequently mRNA was purified by a proprietary tangential flow filtration process (Verweis auf unser Patent).


Dephosphorylation of residual uncapped mRNA was carried out using a phosphatase (Quick cip) followed by purification via a proprietary tangential flow filtration process (Verweis auf unser Patent).


mRNA was further polyadenylated by using a poly(A) polymerase and purified again by a proprietary tangential flow filtration process (Verweis auf unser Patent). Finally, mRNA was filtrated using a 0.22 μm membrane. Critical quality attributes including poly(A) tail length, integrity, proportion of cap, incorporation of modified nucleotides were measured in a subsequent quality control of the mRNA.


2.3 Transfection

mRNA coding for IFNλ1 (SEQ ID NO:13) (which was produced as described in section 2.2., above) was transfected using Lipofectamine® MessengerMAX™ in a RNA to Lipofectamine ratio of 1:1.5 (w/v). For lipoplex formation mRNA was diluted in dH2O. Lipofectamine® MessengerMAX™ was diluted in serum free medium and was mixed by pipetting. After incubation of 10 min at RT, the RNA solution was added to the Lipofectamine® MessengerMAX™ solution, mixed and incubated for another 5 min at RT. A dose titration ranging from 500 to 0.98 ng mRNA/well was performed. Afterwards, 25 μL of the Lipoplex solution for 15.6, 3.9, 1.95 and 0.98 ng mRNA/well was added to the respective wells.


2.4 Harvest

6, 10, 24, 48, 72, 120 and 168 h post transfection, supernatants were collected and stored in a fresh 96-well storage plate at −80° C. until ELISA. Prior ELISA the supernatants were centrifuged at 500×g for 2 min to pellet cell debris.


For qPCR, after collecting the supernatant, cells were washed with 150 μL D-PBS per well and frozen at −80 C.


2.5 IFNλ1 ELISA

Protocol for IFNλ ELISA was performed as established according to Kit protocol with the exception, that TMB incubation is shortened to 15 minutes.


Example 1: IFNλ1 Expression in Supernatants

Presence of IFNλ1 in supernatants of cells transfected with mRNA or treated with rec. hIL-29 were measured via hIL-29 ELISA. Treatment with recombinant hIL-29 was done in doses of 2.9, 0.6, 0.3 and 0.1 ng/well. The doses corresponded to 23, 5, 2.5 and 1 ng/ml in the supernatant. 6 h post treatment, concentrations between 0.3 and 8 ng/ml were quantified in supernatants by ELISA. 70% of the added protein was consumed by the cells or degraded (see FIG. 1).


In supernatants of cells transfected with mRNA coding for IFNλ1, increasing concentrations of IFNλ1 were measurable for each dose up to 48 h. From 48 h to 168 h, IFNλ1 was still detectable with only a small decrease (see FIG. 2).


Example 2: Downstream Target Activation

Downstream target activation post treatment with recombinant IFNλ1 or mRNA coding for IFNλ1 was assessed via qPCR for IFIT1, IFIT3, OAS3 and ISG15.


For all doses of mRNA transfection, high induction for IFIT1, IFIT3 and ISG15 was observed. Peak induction was reached between 24 and 48 h depending on target and dose. Upon mRNA transfection, target genes remained upregulated up to 120 h. (see FIGS. 3 to 6). For OAS3 induction peak was reached at 120 h upon all transfection doses.


Treatment with recombinant protein showed comparable target induction to mRNA transfection at 6 h post treatment. At all later time points after medium was exchanged and the protein was no longer available for the cells, downstream activation declined rapidly (see FIGS. 3 to 6).


Downstream target activation 6 hours post treatment with recombinant IFNλ1 or mRNA coding for IFNλ1 was assessed via qPCR for IFIT1, IFIT3, OAS3 and ISG15. Downstream target activation is plotted against measured IFNλ1 levels. IFIT1, IFIT3 and ISG15 activation was higher at low IFNλ1 levels when provided by transfection of IFNλ1 mRNA compared to incubation with the recombinant IFNλ1 protein (see FIG. 7).


Summary of Examples 1 and 2

Upon transfection of A-549 cells with mRNA coding for IFNλ1 and medium exchange 6 h post transfection, IFNλ1 was detectable in supernatants of these cells up to 168 h with a peak at 48 h. Downstream targets were highly induced also with low doses of mRNA transfection and remained upregulated up to 120 h post mRNA transfection. Regarding treatment with recombinant hIL-29, downstream target activation was observed at 6 h post treatment and declined rapidly after medium exchange.


For the Examples described in the Examples, the following Materials and Methods were used:


1 Methods
1.1 In Vitro
1.1.1 Cell Culture
1.1.1.1 Submersed Cells

HEK293, A-549 and 16HBE14o-cells were cultivated in MEM supplemented with 10% heat inactivated FBS and 1% P/S at 37° C., in a humidified atmosphere with 5% CO2. For 16HBE14o-cells, coated flasks were used. FreeStyle 293-F cells were cultivated in suspension in a 125 mL vented Erlenmeyer flask using FreeStyle F17 Expression Medium. 24 h prior transfection cells were seeded in 96-well plates in a total volume of 100 μL with following densities:

    • HEK293: 25.000 c/well
    • FreeStyle 293-F: 25.000 c/well
    • 16HBE14o-: 25.000 c/well
    • A-549: 20.000 c/well


1.1.1.2 ALI (Air-Liquid-Interface) Cultures

For 16HBE14o- and A-549 ALI cultures, 6×104 submersed cells (at 80-90% confluency) were seeded in coated 24-well inserts. For 16HBE14o-ALIs, inserts were coated with 50 μg/mL collagen type I diluted in 20 mM acetic acid. Cells were seeded in 250 μL 16HBE14o- or A-549 medium apical. For 16HBE14o-500 μL medium and for A-549 700 μL medium was added to the basolateral side. Cells were incubated for 72 h to attach to the membrane with medium on the apical and basolateral side. 24 h prior transfection, basal medium was replaced by fresh medium and the seeding medium on the apical side was carefully aspirated (air-lift). The cells were maintained as ALI cultures without liquid on the apical side.


MucilAir™ inserts were purchased at Epithelix and cultivated in 700 μL MucilAir™ culture medium at 37° C., in a humidified atmosphere with 5% CO2 as ALI (Air-Liquid-Interface) cultures. Prior transfection the cells were let rest for 2-3 days upon arrival. To maintain culture, medium was exchanged every 2-3 days.


1.1.2 Transfection
1.1.2.1 Submersed Cultures

mRNA was transfected using Lipofectamine® MessengerMAX™ in a RNA to Lipofectamine ratio of 1:1.5 (w/v). A dose titration ranging from 500 to 0.98 ng mRNA/well was performed. For lipoplex formation mRNA was diluted in dH2O and Lipofectamine® MessengerMAX™ was diluted in serum free medium and mixed by pipetting. After Lipofectamine/MMax incubation of 10 min at RT, the RNA solution was added to the Lipofectamine® MessengerMAX™ solution, mixed and incubated for another 5 min at RT. Afterwards, 25 μL of the Lipoplex solution for desired concentrations is added to the respective wells.


1.1.2.2 ALI Cultures

Prior transfection of Epithelix primary human ALIs a mucus wash was performed. 200 μL of PBS (w/o Mg/Ca) was added to the apical side and incubated for 20 min at 27° C. Total time of apical washing should not exceed 30 min. To detach the mucus from the apical surface three back and forth movements were done with 100 μL from the apical liquid with a P200 pipet. PBS from the apical surface of the ALI culture was removed by gentle aspiration without damaging the epithelium. To remove traces of PBS, WFI wash using 200 μL WFI was performed. 16HBE14o- and A-549 ALIs were washed solely using WFI. Subsequently, the cells were transfected with 0.1, 0.3, 1, 3 and 6 μg mRNA in LNP/insert. Formulations were thawed at RT and kept on ice until further use. Detailed calculations are shown in Table 1.









TABLE 1







Calculation for ALI transfection













0.1
0.3
1
3
6



μg
μg
μg
μg
μg

















Dose
μg/cm2
0.3
0.9
3
9
18



μg/insert
0.1
0.3
1
3
6


transfection. Vol
[μl]
25
25
25
25
25


final LNP conc. in well
[ng/μL]
4
12
40
120
240


LNP conc. of stock wells
[μg/mL]
500
500
500
500
500


dilution (incl. excess)
μL LNP
1.0
2.1
7
21
36



Vehicle
124.0
85.4
80.5
66.5
39.0









6 h post transfection, LNP was aspirated.


1.1.3 Harvest
1.1.3.1 Submersed Cultures

At respective time points post transfection, supernatant of cells were collected and stored in a fresh 96-well storage plate at −80° C. until ELISA. Prior ELISA the supernatants were centrifuged at 500×g for 2 min to pellet cell debris.


For qPCR, after collecting the supernatant, submersed cells were washed with 150 μL D-PBS per well and cell pellets were frozen at −80° C.


1.1.3.2 ALI Cultures

24 h post transfection, apical side of the inserts was washed using 200 μL PBS, which was pipetted up and down three times. Wash solution was collected in a 96-well storage plate. Also 200 μL of basal medium was collected in a storage plate. Samples were stored at −80° C. until ELISA was performed.


For qPCR, medium was removed completely from the inserts without PBS washing. 175 μL RLT buffer from RNease Mini Kit (Qiagen, 74104), complemented with DTT (40 μL/mL) was added on the insert. With a mini cell scraper the cells were loosened from the insert. The RLT buffer was transferred into a QIAshredder column. To ensure all cells were transferred, the insert was washed with another 175 μL of RLT buffer. To finish the cell lysis, the columns were centrifuged for 2.5 min at max speed. Cell lysates can be frozen at −80° C. or used immediately to isolate RNA.


1.1.4 IFNλ1 ELISA

IFNλ ELISA was performed according to Kit instructions (IL-29 Human ELISA Kit, Abcam, ab100568) except that TMB was incubated for 10 instead of 30 min.


1.1.5 RNA Isolation
1.1.5.1 Submersed Cells

For submersed cells, RNA isolation was performed using SingleShot™ Cell Lysis Kit (BioRad, Art. Nr.: 1725080).


Preparation of the SingleShot™ cell lysis buffer was done according to Table 2. Preparation was done always fresh and on ice. The lysis buffer was mixed thoroughly, centrifuged and was used within 2 h.









TABLE 2







Lysis Buffer










Volume per
Volume for 96-well plate


Component
well [μL]
[μL] (with excess)












SingleShot ™ Cell
48
4800


Lysis Buffer


Proteinase K Solution
1
100


DNase Solution
1
100









50 μL lysis buffer for RNA isolation was added per well to frozen samples and incubated without agitation for 10 min at RT. Samples were processed within 20 min. Subsequently the cell lysate was transferred to a PCR plate. Protein and DNA was digested using the program “BioDNA” with the following conditions (see Table 3): Thermal cycler protocol for cell lysis









TABLE 3







Thermal cycler protocol for cell lysis










Time
Temperature







5 min
37° C.



5 min
75° C.










The cell lysate can be stored for up to 4 h on ice, for up to 2 months at −20° C., or for up to 12 months at −80° C.


1.1.5.2 ALI Cultures

RNA was isolated using the RNeasy Mini Kit (Qiagen, 74104) according to the manufacturer's protocol. Elution was done using 30 μL RNAse free water.


1.1.6 cDNA Synthesis


1.1.6.1 For Submersed Cells

iScript™ Select cDNA Synthesis Kit (BioRad, Art. Nr.: 1708897) was used. The plates with cell lysates were thawed on ice. All kit components, except iScript™ reverse transcriptase, were thawed on ice and mixed thoroughly and centrifuged briefly. cDNA synthesis was done using OligodT primers. Following components were added to a 2 mL tube, where the iScript reverse transcriptase was added after the other components (see Table 4).









TABLE 4







Master Mix iScript ™ Select cDNA Synthesis










Volume per
Volume for 96-well


Component
well [μL]
plate [μL]












Nuclease free water
9
900


5× iScript ™ reaction mix
4
400


OligodT
2
200


iScript reverse
1
400


transcriptase




Total
16
1600









4 μL of the cell lysate were pipetted in a new PCR plate using a multi-channel pipet, before adding 16 μL of the master mix on top. The plate was sealed using a cover foil and mixed gently at 400 rpm before the plate was spun down briefly. cDNA synthesis was performed using a thermal cycler with following protocol (“ISCRIPT2”).









TABLE 5







Thermal cycler protocol for cDNA synthesis











Time
Temperature
Step














60
min
42° C.
cDNA synthesis


5
min
85° C.
Heat inactivation









cDNA can be stored at −20° C. until qPCR is performed.


1.1.6.2 ALI Cultures

For cDNA synthesis of ALI cultures Transcriptor First Strand cDNA Synthesis Kit (Roche, 4896866001) was used. In accordance to manufacturers protocol RNA was transcribed to cDNA. For a two-step cDNA synthesis 1 μg of total RNA and 1 μL Oligo(dT) Primers in a total volume of 13 μL were necessary to prepare the first step. One example for the preparation of template-primer mixture is given in Table 6:









TABLE 6







Template-Primer-Mix













Volume

Water



Nucleic Acid
RNA
Oligo(dT)
(RNAse


Name
Conc. [ng/μL]
[μL]
Primer [μL]
free) [μL]














Sample No. 1
147.7
6.8
1.0
5.2





Total volume
13









The optional denaturation step at 65° C. for 10 min was done in a thermal block cycler. Afterwards, all remaining reagents except enzymes, Reverse Transcriptase and RNAse Inhibitor, were thawed on ice and briefly centrifuged. To prepare a master-mix following components listed in Table 7 were added into a fresh tube:









TABLE 7







Master-Mix











Name
1× [μL]
n = 4















5× reaction buffer
4
16



RNAse Inhibitor
0.5
2



dNTPS
2
8



Reverse transcriptase
0.5
2



Total volume [μL]
7
28










At last, enzymes were added to the reagents listed in Table 7. Volume of master-mix was adapted according to number of samples. 7 μL of master-mix were added to each tube, which contained template-primer mixture to get a final volume of 20 μL per tube, see Table 6. The tubes were mixed carefully and briefly centrifuged. Tubes were put back to a thermal block cycler and following program for reverse transcription (here step two of cDNA synthesis) was used:









TABLE 8







Program for reverse transcription using Oligo(dT) primer









Step in thermal
Duration
Temperature


block cycler
[min]
[° C.]












1
30
55


2
5
85









cDNA can be stored at −20° C. until qPCR is performed.


1.1.7 qPCR Using TaqMan Probes


The following components shown in Table 9 were combined and vortexed briefly. To bring the reaction mix to the bottom of the tube and to eliminate air bubbles the mix was briefly centrifuged. 18 μL of TaqMan master mix were transferred to an optical 96-well qPCR reaction plate. 2 μL cDNA template (cDNA and nuclease free water) were added as shown in


Table 10 to the optical 96-well qPCR reaction plate to obtain a final volume of 20 μL. The optical 96-well qPCR reaction plate was sealed with optical adhesive film and briefly centrifuged.









TABLE 9







Master mix qPCR TaqMan












Volume per
Volume for 96-well



Component
well [μL]
plate [μL]















Nuclease free water
7
700



TaqMan Probe
1
100



TaqMan Fast Advanced
10
1000



Master Mix





Total
18
1800

















TABLE 10







cDNA template preparation












Volume per
Volume for 96-well



Component
well [μL]
plate [μL]















cDNA
1
96



Nuclease free water
1
96



Total
2
192










TaqMan assay was performed using following parameters:

















Mode
Cycle
Step









2 Step
40
95° C. for 5 s



Amplification

60° C. for 30 s

























TaqMan Probes
Supplier
Cat no.









IFIT3
Thermo Fisher Scientific
Hs01922752_s1



IFIT1
Thermo Fisher Scientific
Hs03027069_s1



OAS3
Thermo Fisher Scientific
Hs00196324_m1



ISG15
Thermo Fisher Scientific
Hs01921425_s1










1.2 In Vivo
1.2.1 Animal Housing

Mice were housed under specific pathogen free conditions (facility tested negative for any FELASA listed pathogens according to the annual health and hygiene survey 2017) in individually ventilated cages under a circadian light cycle (lights on from 7 a.m. to 7 p.m.). Food and drinking water were provided ad libitum. After arrival, animals were given at least 7 days for acclimatization until they enter the study.


1.2.2 Intratracheal Application

Animals were anesthetized by the inhalation of pure oxygen containing 4% Isoflurane (Isothesia, Henry Shine, Germany). Unconscious animals were intubated using a 20 gauge catheter shortened to 37 mm. Test item in a final volume of 50 μL was applied as one drop at the proximal tip of the tubus and thereby aspirated during the physiological inspiratory movement of the animal. Finally, 150 μL of air was applied, to assure that no liquid remains within the catheter.


1.2.3 Clinical Examination

Animals were examined clinically before application of the test item and 24 hours later using a clinical mouse scoring system. Clinical examination consisted of 4 different categories which were scored separately. The scorings of each of the 4 categories were summarized to a total clinical score. A summarized score of more than 4 points or a score of more than 1 point in a single category are considered as moderate suffering and thereby as a humane endpoint.


1.2.4 Necropsy

Animals were set under full anesthesia by intraperitoneal injection of Fentanyl/Midazolam/Medetomidin (0.05/5.0/0.5 mg/kg bw). Subsequently, mice were killed by cervical dislocation.


1.2.5 BALF and Lung Withdrawal

The thoracal and abdominal cavity were opened and a 18G catheter was placed into the trachea. 0.5 mL PBS was injected and retrieved from the lungs and centrifuged at 300×g for 10 min at 4° C. Supernatant was taken and stored at −80° C. until further processing. The cell pellet was biobanked at −80° C. Next, the lungs were explanted and as well stored at −80° C. until further processing.


1.2.6 Lung Homogenisation

Samples were homogenized using liquid nitrogen. Therefore, the organ was put in a mortar which is placed in an ice box. Liquid nitrogen was added in a volume, which covers the lung completely. The lung was pulverized in the mortar using a pistle. A stapula was immersed in nitrogen and used to devide the organ powder in two halfs. Both halfs were weighed in an empty tared tube each. Mortar, pistle and stapula were cleaned using Ethanol after every sample.


For ELISA, lysis was done in 250 μL Triton X-100 lysis buffer (0.25 M Triethanolamine, 0.1% Triton X-100, pH7.7), complemented with protease inhibitor. For qPCR lysis was done in 350 μL lysis buffer per 30 mg organ. Lysis buffer RLT was provided by the kit and complemented with 40 μL/mL DTT. For qPCR, additional homogenizing in lysing tubes was performed. Therefore, volumes were applied as described above and procedure was performed for 3×20 sec in a Tissue homogenizer (MP FastPrep-24 Tissue and Cell Homogenizer).


Samples were incubated for 10 min on ice and centrifuged for 10 min at 4° C. in a Mikro 22R centrifuge (Hettich Zentrifugen) with maximum speed. Supernatant was transferred in fresh tubes and stored at −80° C. until further analysis.


1.2.7 Cell Lysis and cDNA Synthesis for qPCR


RNA isolation was done using the NucleoSpin® RNA Kit (Macherey & Nagel, 740984.50) according to manufacturer's protocol.


CDNA synthesis was done using Transcriptor First Strand cDNA Synthesis Kit (Roche, 4896866001). In accordance to manufacturer's protocol RNA was transcribed to cDNA. For a two-step cDNA synthesis 1 μg of total RNA and 1 μL OligoDT Primers in a total volume of 13 μL were necessary to prepare the first step. One example for the preparation of template-primer mixture is given in Table 11:









TABLE 11







Template primer mixture













Volume

Water



Nucleic Acid
mRNA
OligoDT
(RNase


Name
Conc. [ng/μL]
[μL]
Primer [μL]
free) [μL]














Sample No. 1
147.7
6.8
1
5.2





Total volume
13









The denaturation step at 65° C. for 10 min was done in a thermo block cycler. Afterwards, all remaining reagents except enzymes, Reverse Transcriptase and RNAse Inhibitor were thawed on ice and briefly centrifuged. To prepare a master-mix following components listed in Table 12 are added into a fresh tube:









TABLE 12







Master mix










Name
1× [μL]














5× reaction buffer
4



RNAse Inhibitor
0.5



dNTPs
2



Reverse transcriptase
0.5



Total volume [μL]
7










At last, enzymes were added to the reagents listed in Table 12. Volume of master mix was adapted according to number of samples. 7 μL of master mix were added to each tube, which contains template-primer mixture to get a final volume of 20 μL per tube. The tubes were mixed carefully and briefly centrifuged. Tubes were put back to a thermo block cycler and following program for reverse transcription was started:

















Step in thermo
Duration
Temperature



block cycler
[min]
[° C.]




















Reverse transcription
30
55



Enzyme denaturation
5
85










cDNA can be stored at −20° C. until qPCR is performed.


1.2.8 Real-Time PCR Using TaqMan Probes for Downstream Targets See 1.1.7.

















TaqMan Probes
Supplier
Cat no.









IFIT3
Thermo Fisher
Mm01704846_s1





(ifit3,



IFIT1
Thermo Fisher
mM07295796_m1



OAS3
Thermo Fisher
Mm00460944_m1



ISG15
Thermo Fisher
Mm01705338_s1



IFNλ
Thermo Fisher
Mm00558035_m1











1.2.9 qPCR Using UPL for Quantification of SNIM® RNA


For quantification of ETH061T02 SNIM RNA in mouse lungs qPCR was done. Therefore, 1 μg of IFNλ1 SNIM RNA was used for cDNA Synthesis and synthesis was done as described in section 1.1.6. cDNA of IFNλ1 mRNA was diluted in RNAse-free water in the following dilutions: 1:101, 1:102, 1:103, 1:104, 1:105, 1:106 resulting in a standard curve. It is important that the cDNA dilution series is prepared in a larger volume (e.g. 20 μL), to avoid inaccuracies during pipetting. 0.25 μL cDNA of mouse lung samples or the respective standard curve samples were mixed with 3.75 μL RNAse-free water and pipetted in a LightCycler® 480 Multiwell Plate 96. Preparation of qPCR Master Mix is done according to Table 13.









TABLE 13







UPL Master-Mix












Volume per
Volume for 96-well



Component
well [μL]
plate [μL]















Nuclease free water
0.625
60



Primer 1 [20 μM]
0.150
14.4



Primer 2 [20 μM]
0.150
14.4



UPL [stock]
0.075
7.2



Fast Start Essential
5
480



DNA Probes Master





Total
6
576










The master mix was added to 4 μL of diluted cDNA sample or standard in each well to LightCycler® 480 Multiwell Plate 96 using a multi-pipette. The plate was covered with LightCycler® 480 Sealing Foil and spun down briefly. qPCR was done using program for UniversalProbes on LightCyler® 96 system.


Example 3: Effectiveness of IFNλ1 Encoding mRNA in A-549 Cells Expressing ACE2 in Comparison to Recombinant IFNλ1

Downstream target activation 48 hours post treatment of A549 or A549-ACE2 cells with recombinant IFNλ1 (FIG. 8a) or mRNA coding for IFNλ1 (FIG. 8b) was assessed via qPCR for IFIT1, IFIT3, OAS3 and ISG15. Whereas transfection with mRNA coding for IFNλ1 resulted in similar induction of IFIT1, IFIT3, OAS3 and ISG15, induction of the same downstream targets was reduced on A549-ACE2 cells compared with A459 cells after treatment with recombinant IFNλ1. Transfection of A549-ACE2 cells with mRNA coding for IFNλ1 resulted in dose-dependent production of IFNλ1 (FIG. 8c).


Example 4: IFNλ1 Encoding mRNA is Translated in Various Cells Types

Transfection of mRNA coding for IFNλ1 leads to a time- and dose-dependent production of IFNλ1 in HEK293, A549, 16HBE14o- and FreeStyle 293-F cells as measured by IFNλ1 ELISA (FIG. 9).


Example 5: Time- and Dose-Dependent Induction of Downstream Targets in A549 and 16HBE14o-Cells in Submerse Culture After Transfection With IFNλ1 Encoding mRNA

Transfection of mRNA coding for IFNλ1 leads to a time- and dose-dependent induction of IFIT1, IFIT3, OAS3 and ISG15 as assessed via qPCR (FIG. 10a and b).


Example 6: Dose-Dependent Production of IFNλ1 and Induction of Downstream Targets in Airway Liquid Interface (ALI) Cultures Derived From A549, 16HBE14o- or Primary Human Lung Cells (Epithelix)

Transfection of mRNA coding for IFNλ1 formulated in LF92 leads to dose-dependent production of IFNλ1 (FIG. 11a) and induction of IFIT1, IFIT3, OAS3 and ISG15 as assessed via qPCR 24 hours after treatment (FIG. 11b). Endogenous levels of IFIT1, IFIT3, OAS3 and ISG15 are elevated in ALI cultures derived from 16HBE14o- or primary human lung cells compared with A459-derived ALI cultures (FIG. 11c).


Example 7: Dose-Dependent Production of IFNλ1 and Induction of Downstream Targets in the Lungs of Mice Instilled With mRNA Coding for IFNλ1

Delivery of mRNA coding for IFNλ1 formulated in LF92 leads to dose- and time-dependent IFNλ1 mRNA lung deposition (FIG. 12a), production of IFNλ1 in lung tissue and broncheoalveolar lavage fluid (BALF) (FIG. 12b) and induction of IFIT1, IFIT3, OAS3 and ISG15 as assessed via qPCR 5 and 24 hours after treatment (FIG. 12c).


Example 8: Comparison of hIFNλ1 mRNA Transfection With Recombinant Protein

Enhanced target gene activation following single apical transfection of primary bronchial epithelial airway liquid interface (ALI) cultures with hIFNλ1 encoding mRNA compared to apical treatment with recombinant protein. Due to basolateral receptor expression, inhaled (apical) administration of hIFNλ1 encoding mRNA and subsequently greater basal than apical secretion of type III IFN is expected to be more efficacious in humans than inhaled administration of recombinant protein.


Transfection with hIFNλ1 mRNA led to 30-40 pg total protein at apical compartment 24 h after transfection when transfection mixture was aspirated at 6 h post transfection (FIG. 13a). This corresponds to a concentration of 90 ng/ml in the airway surface lining fluid (ASL), assuming that the volume of the ASL in ALI cultures is 0.33 μL (0.33 cm2 surface area of the insert×0.001 cm height of the cilia layer). For this reason, target gene activation post transfection was compared to treatment with 100 ng/ml of recombinant protein.


Following apical mRNA transfection, target gene expression was 4-fold higher compared to Stop mRNA control (FIG. 13b). In contrast, following apical administration of recombinant hIFNλ1 protein at a concentration of 100 ng/mL, target gene induction was lower compared to mRNA transfection and did not exceed a 2-fold induction relative to Stop mRNA control (FIG. 13b). This effect might be due to longer exposure to hIFNλ following mRNA transfection (mRNA translation continues after exchange of medium at 6 hours) and hence potential accumulation and therefore higher protein level on apical and basal compartments compared to treatment with recombinant protein. These observations highlight the potential for added PK/PD benefit of mRNA treatment over treatment with recombinant protein (FIG. 13).


Example 9: hIFNλ ELISA From Supernatants of Transfected A-549 Cells: LF92 Vs. Commercially Available Transfection Reagent

At 6 and 24 h post transfection, medium was exchanged at each plate that was not harvested at these time points. Hence, 24 h time point reflects translation between 6 and 24 h, and 48 and 72 h time points reflect translation after 24 h. Translation following mRNA transfection occurred in a dose- and time dependent manner and to a similar extent with LF92 formulated mRNA as when using a commercial transfection reagent (FIG. 14).


Example 10: Target Gene Induction by LF92 Formulated mRNA

At 6 and 24 h post transfection, medium was exchanged at each plate that was not harvested at these time points. Hence, 24 h time point reflects translation between 6 and 24 h, and 48 and 72 h time points reflect translation after 24 h. Dose-dependent and sustained hIFNλ1 target gene activation following single transfection of A549 lung cells with LF92 formulated mRNA encoding hIFNλ1 was observed (FIG. 15).


Example 11: In Vitro Tolerability of LF92 Formulated mRNA Encoding hIFNλ1

At 6 and 24 h post transfection, medium was exchanged at each plate that was not harvested at these time points. Hence, 24 h time point reflects translation between 6 and 24 h, and 48 and 72 h time points reflect translation after 24 h. Induction of cytokine mRNA expression following single transfection of A549 lung cells with LF92 formulated mRNA encoding hIFNλ1 was observed only at high doses associated with plateau of target gene induction (FIG. 16).


Example 12: Correlation of mRNA Translation and Target Gene Induction

At 6 and 24 h post transfection, medium was exchanged at each plate that was not harvested at these time points. Hence, 24 h time point reflects translation between 6 and 24 h, and 48 and 72 h time points reflect translation after 24 h. Amount of hIFNλ1 measured in supernatant correlates with extent of target gene induction (FIG. 17).


Example 13: Virus Inhibition In Vitro Assessed by qPCR

Prophylactic treatment with hIFNλ1 mRNA reduced SARS-COV-2 and IAV virus load while Stop mRNA did not (FIG. 18). Dose dependent reduction of virus load by recombinant hIFNλ1 was observed, but was lower with recombinant protein compared to mRNA treatment (FIG. 18). Treatment was done 24 h prior infection. Samples were taken 48 h post infection.


Example 14: Detection of hIFNλ1 mRNA in Lung Homogenate

Dose-dependent deposition of mRNA encoding human IFNλ1 was observed at 5 h following single nasal administration in mice (FIG. 19). Three animals per group were assessed.


Example 15: Detection of hIFNλ1 Protein in Lung Homogenate

Human IFNλ1 quantification in lung homogenate revealed dose and time dependent protein translation upon single intranasal treatment with mRNA encoding human IFNλ1 protein (FIG. 20). Three animals per group were assessed.


Example 16: Target Gene Activation in Lung Tissue After hIFNλ1 mRNA Application

hIFNλ1 mRNA application was well tolerated and potently induced pulmonary target gene expression at low doses following single nasal administration in mice (FIG. 21). The analyzed target genes OAS3, ISG15, IFIT1 and IFIT3 displayed a dose dependent activation for all dose levels at the 5 hours' time point (FIG. 21). Only OAS3 showed a dose dependent activation up to 24 h after application (FIG. 21d). Three animals per group were assessed.


Example 17: Detection of Chemokines in Plasma After Single Nasal application of hIFNλ1 mRNA

There was no change in plasma concentrations of the measured chemokines following single nasal administration of hIFNλ1-encoding mRNA or STOP mRNA compared to vehicle at all dose levels in mice (FIG. 22). Three animals per group were assessed.


Example 18: Target Gene Activation in Lung Tissue Upon Treatment With hIFNλ1 mRNA

LF92 formulated hIFNλ mRNA was well tolerated and potently induced pulmonary target gene expression at low dose following multiple nasal administrations in mice (FIG. 23). Three doses in 48 h intervals were administered and samples collected 24 h after the last application. Expression of hIFNλ target genes in lung homogenates increased in a dose-dependent manner (FIG. 23). An up to 15-fold activation of all target genes analysed was seen at a dose level 1 μg hlFNA mRNA (FIG. 23). Lack of target gene activation in STOP and vehicle groups suggest that the observed effect is target-induced (FIG. 23). Six animals per group were assessed.


Example 19: Detection of hIFNλ1 mRNA Deposited in the Lung After Nasal application in Mice

Three doses in 48 h intervals were administered and samples collected 24 h after the last application. Dose-dependent deposition of mRNA encoding human IFNλ1 was observed (FIG. 24). Six animals per group were assessed.


Example 20: Body Weight During Repeated Administration of hIFNλ1 mRNA

Three doses in 48 h intervals were administered by nasal application in mice and body weight was measured daily. No changes in body weight were recorded (FIG. 25). Six animals per group were assessed.


Example 21: Quantification of hIFNλ1 mRNA in Ferret Lung Homogenate

hIFNλ1 mRNA was deposited in all lung lobes after single nasal administration in ferret (FIG. 26). Three animals per group were assessed.


Example 22: Target Gene Activation in Ferret Lung Homogenate

hIFNλ1 treatment lead to induction of ISG15, MX1 and OAS3 after single nasal administration in ferret (FIG. 27). Three animals per group were assessed.


Example 23: Quantification of IFN mRNA, Target Gene Activation, and Cytokine Induction in Ferret Lung Homogenate

Single nasal administration of mRNAs encoding hIFNλ1, hIFNβ or both resulted in mRNA deposition in all lung lobes (FIG. 28). Mx-1 was induced by all three treatments while none of the analyzed cytokines was induced (FIG. 28). Three animals per group were assessed.


Example 24: Reduction of IAV Replication by mRNAs Encoding IFNs

Intra-nasal administration of mRNA encoding hIFNλ1 (FIG. 29a), hIFNβ (FIG. 29b) or both (FIG. 29c) was well tolerated and reduced early virus replication and clinical symptoms in a ferret influenza model (FIG. 29). Treatment was performed on study days −1, 1 and 3 while infection was done on day 0. Ten animals per group were assessed.


Example 25: Reduction of SARS-COV2 Replication by mRNA Encoding hIFNλ1

Intra-nasal administration of mRNA encoding hIFNλ1 was well tolerated and reduced virus replication and body weight reduction in a hACE2-TG mouse model challenged with SARS-COV-2 alpha compared to vehicle treated animals (FIG. 30). Viral replication was assessed by measurement of genomic viral RNA via RT-qPCR (60% reduction compared to vehicle) (FIG. 30a) and by determination of infectious virus particles using the TCID50 method (97% reduction compared to vehicle) (FIG. 30b). Body weight is shown as percentual weight change of day 3 after virus inoculation (FIG. 30c). Two to five animals per group were assessed. Following controls were employed: Mock treated, hIFNλ1 mRNA but not virus treated and vehicle but not virus treated mice.


In Examples 8 to 25 the following Materials were used.


2.1 Materials














Material
Supplier
Cat no.

















QIAshredder
Qiagen
79656


RNeasy Mini Kit
Qiagen
74104


DL-Dithiothreitol
Sigma-Aldrich
646563-10X.5ML


solution (DTT)


Transcriptor First Strand
Roche
4896866001


cDNA Synthesis Kit


IL-29 Human ELISA
Abcam
ab100568


Kit (IFNλ1)


FBS, heat-inactivated
Thermo Fisher
10500064



Scientific


MEM with GlutaMAX ™
Gibco
41090-028


Lipofectamine MMax
Thermo Fisher
LMRNA008



Scientific


SingleShot ™ cell
BioRad
1725080


lysis Kit


iScript ™ Select cDNA
BioRad
1708897


Synthesis Kit


DMEM
Sigma
D6429


DPBS 1× without
Thermo Fisher
Gibco


Ca and Mg
Scientific
14190-250


FBS
Sigma
F7524


Penicillin-Streptomycin
Sigma
P4333


Recombinant hIFNλ1
Abcam
ab155625


Carboxymethylcellulose
Sigma
SAFS21904-250G


Crystal violet
Carl Roth
T123.1



GmbH


Fast Start Essential
Roche
06402682001


DNA Probes Master


Isoflurane
Isothesia,
1311758



Henry Shine


LightCycler ® 480
Roche
4729692001


Multiwell Plate 96, white;


VE: 50 plates with foils


Lysing Matrix D Tubes
MP Biomedicals
6913500


TaqMan ™ Fast
Thermo Fischer
4444557


Advanced Master Mix


Twin.tec PCR Plate96,
Omnilab
5425895


unskirted, low profile,


farblos VE: 20


Casein
Thermo Fisher
37528



Scientific


Mouse CXCL9/MIG
R&D System
DY572


DuoSet ELISA, 15 Plate


Mouse
R&D System
DY466


CXCL10/IP-10/CRG-2


DuoSet ELISA, 15 Plate


Mouse CXCL11/I-TAC
R&D System
DY492


DuoSet ELISA, 15 Plate


10× PBS
Roche
11666789001


Custom Premix Mouse
Merck
MCYTOMAG-70K-09C


Cyto/Chemokine MAG09


Tween-20
Carl Roth
9127.1





















Device
Supplier
Cat no.







96-well cell culture plate
Corning
Costar 3599


96-well storage plate
Corning
Costar 3363


Countess Cell Counter
Thermo Fisher
C10281



Scientific


Countess Cell Counting
Thermo Fisher
C10283


chamber slides
Scientific


IncuCyte
Sartorius
Incucyte S3


Light Cycler ®96
Roche
Light Cycler ®96


Microplate reader
Tecan
Infinite M200 Pro


Microplate shaker
Heidolph
Titramax


Tissue and Cell
MP
MP FastPrep-24


Homogenizer
Biomedicals


Magpix
Luminex
MAGPX14184703









2.2 Methods
2.2.1 Cell Cultures
2.2.1.1 ALI Cultures

MucilAir™ inserts were purchased at Epithelix and cultivated in 700 μL MucilAir™ culture medium at 37° C., in a humidified atmosphere with 5% CO2 as Air-Liquid-Interface (ALI) cultures. Prior transfection the cells were let rest for 2-3 days upon arrival. To maintain culture, medium was exchanged every 2-3 days.


2.2.1.2 A-549 Cell Culture

A-549 cells were cultured as described in 2.1, supra.


2.2.1.3 A549-ACE2 Cell Culture

A549-ACE2 cells were cultivated in DMEM with 10% FBS and 100 μg/mL Streptomycin and 100 IU/mL Penicillin. 10,000 cells were seeded in 96-well plates in a total volume of 100 μL 24 h before treatment.


2.2.2 Transfection/Treatment

Prior transfection a mucus wash was performed. Therefore, 200 μL of PBS (w/o Mg/Ca) were added to the apical side and incubated for 20 min at 37° C. Total time of apical washing should not exceed 30 min. To detach the mucus from the apical surface three back and forth movements were done with 100 μL from the apical liquid with a P200 pipet. PBS from the apical surface of the ALI culture was removed by gentle aspiration without damaging the epithelium. To remove traces of PBS, WFI wash using 200 μL WFI was performed.


Subsequently, the cells were transfected/treated with desired dose. Formulations were thawed at RT and kept on ice until further use. Recombinant protein was diluted in medium.


mRNA and recombinant protein was removed from the apical side 6 h post transfection. In case of recombinant protein, also the basal medium was renewed. At 24 h post transfection, apical side of the inserts was washed using 200 μL PBS, which was pipetted up and down three times. Wash solution was collected in a 96-well storage plate for ELISA analysis. Also 200 μL of basal medium was collected in a storage plate. Samples were stored at −80° C. until ELISA.


For qPCR, 175 μL RLT buffer from RNeasy Mini Kit, complemented with DTT (40 L/mL), were added on the insert. With a mini cell scraper the cells were loosened from the insert. The RLT buffer was transferred into a QIAshredder column. To ensure all cells were transferred, the insert was washed with another 175 μL of RLT buffer. To finish the cell lysis, the columns were centrifuged for 2.5 min at max. speed. Column was removed and cell lysates in tube can be frozen at −80° C. or used immediately to isolate RNA.


2.2.2.1 Transfection Using Lipofectamine MMax in Examples 9 to 12

Transfection using Lipofectamine MMax was performed as described in 2.3, supra, with the exception that a dose titration ranging from 0.005 to 100 ng mRNA/well was performed and afterwards, 25 μL of the Lipoplex solution was added to the respective wells, in which medium was exchanged by 100 μL fresh complete medium.


2.2.2.2 Transfection Using Lipofectamine MMax in Example 13

Transfection using Lipofectamine MMax was performed as described in 2.3, supra, with the exception that a dose titration ranging from 15 ng/25 μL down to desired concentration was performed. Medium of the cells was exchanged by 100 μL fresh medium. Afterwards, 25 L of the Lipoplex solution was added to the respective wells. For treatment with recombinant protein, proteins were pre-diluted in medium and added to the cells in 25 μL with desired concentrations.


2.2.2.3 Transfection Using LF92 Formulated mRNA


LF92 formulated mRNA was diluted in Vehicle (10% (w/v) sucrose, 50 mM NaCl and a proprietary excipient) to desired concentrations. 25 μL of each dilution was added to the respective wells, in which medium was exchanged by 100 μL fresh complete medium.


2.2.3 RNA Isolation

RNA isolation was performed as described in 1.1.5.2, supra.


2.2.4 cDNA Synthesis cDNA synthesis was performed as described in 1.1.6, supra.


2.2.5 qPCR Using TaqMan Probes


qPCR using TaqMan probes was performed as described in 1.1.7, supra. Analysis of qPCR results was done with the ΔΔCt method. To do so, first the ΔCt value of each sample was calculated. This was done by subtracting the average Ct value of the housekeeper (RPLPO) from the target Ct value. Then the ΔΔCt was calculated by subtracting the average ΔCt of the reference samples (vehicle control) from the target ΔCt. Finally, the fold change was calculated with the formula: 2−ΔΔCt.


2.2.6 hIFNλ1 ELISA (IL-29 ELISA)


hIFNλ1 ELISA was performed as described in 1.1.4, supra. Interpolation was done using a 4PL standard curve. Analysis was done with GraphPad Prism Software and Excel.


2.2.7 Harvest

Harvest was done at 3, 6, 24, 48 and 72 h post transfection. For each plate a medium exchange was performed at 6 and 24 h post transfection.


For ELISA, supernatant of the cells was collected in a 96-well storage plate and frozen at −80° C. As two different ELISAs were performed, the supernatants were splitted on two plates to enable separate thawing. For qPCR, cells are washed using 150 μL PBS−/− and cells are frozen without liquid at −80° C.


2.2.8 RNA Isolation Using the SingleShot™ Cell Lysis Kit

RNA Isolation using the SingleShot™ Cell Lysis Kit was performed as described in 2.2.5.


2.2.9 Preparation of Virus Stocks

SARS-COV-2-MUC-IMB-1, SARS-COV-2-GFP strains, wild type Influenza A virus (IAV-WT) (SC35M) and IAV (SC35M) NS1-GFP were produced by infecting Vero E6 cells cultured in DMEM medium (10% FCS, 100 μg/ml Streptomycin, 100 IU/ml Penicillin) for 2 days (MOI 0.01). Viral stock was harvested and spun twice (1000 g/10 min) before storage at −80° C.


Titer of viral stock was determined by plaque assay. For this, confluent monolayers of Vero E6 cells were infected with serial five-fold dilutions of virus supernatants for 1 hour at 37° C. The inoculum was removed and replaced with serum-free MEM containing 0.5% carboxymethylcellulose. Two days post-infection, cells were fixed for 20 minutes at room temperature with formaldehyde directly added to the medium to a final concentration of 5%.


Fixed cells were washed extensively with PBS before staining with H2O containing 1% crystal violet and 10% ethanol for 20 minutes. After rinsing with PBS, the number of plaques was counted, and the virus titer was calculated.


2.2.10 Virus Infection

Cells were infected 24 h after transfection with SARS-COV-2 (MOI 3) or IAV (MOI 0.5). Virus was added in 25 μL medium directly to the cell culture. For the analysis of infection kinetics under live imaging system, the SARS-COV-2-GFP and IAVGFP were used.


2.2.11 Cell Monitoring in the IncuCyte

After the infection, plates were placed in the IncuCyte S3 Live-Cell Analysis System where whole-well real-time images of mock (Phase channel) and infected (GFP and Phase channel) cells were captured every 4 hours for 48 hours. Cell viability (mock) and virus growth (mock and infected) were assessed as the cell confluency per well (Phase area) and integrated GFP intensity normalized on cell confluency per well (Integrated GFP intensity/Phase area) respectively using IncuCyte S3 Software (Essen Bioscience; version 2019B Rev2).


2.2.12 qPCR for SARS-COV-2 and IAV mRNA


For relative transcript quantification PowerUp SYBR Green was used. All steps were performed according to manufacturer's instructions. RPLP0 was employed as housekeeper.


2.2.13 Animal Housing

All animals were housed as described in 1.2.1, supra. All procedures were approved by the local animal welfare authorities (Regierung von Oberbayern) under the file number Az. 2532.Vet_03-17-114 and were conducted according to the German animal protection law (Tierschutzgesetz).


2.2.14 Nasal Application in Examples 14 to 17

Animals were anesthetized by inhalation of pure oxygen at a flow rate of 2 L/min supplemented with approximately 4% of the anesthetic gas Isoflurane in an inhalation chamber. hIFNλ1 mRNA was applied in two 25 μL boluses, by using a laboratory pipette, on both nose holes so that the liquid was actively inhaled through the nose by the animal during a physiological inspiratory movement. The animal was held in perpendicular position during this procedure and was subsequently placed in supine position until recovery from anesthesia.


2.2.15 Clinical Examination in Examples 14 to 17

Clinical examination was performed as described in 1.2.3, supra, with the exception that Animals were examined clinically before and 5 hours after treatment as well as daily until the day of necropsy. Furthermore, body weight was measured before application and daily until the final day of the experiment.


2.2.16 Necropsy

Scheduled necropsy time points were chosen 5, 24 or 48 h after application, respectively. In A01, an additional 72h time point for necropsy was included. In A02, group 2, additional necropsies at 72 and 96 h after application of hIFNλ1 mRNA were performed. Animals were set under full anesthesia by intraperitoneal injection of Fentanyl/Midazolam/Medetomidin (0.05/5.0/0.5 mg/kg bw). Capillary blood was taken from retrobulbar venous plexus using nonheparinized 0.8 mm capillaries and collected in EDTA tubes. Blood samples were centrifuged at 2.000×g for 5 min at 4° C. Subsequently, mice were killed by cervical dislocation.


The thoracical and abdominal cavity were opened, and the lung was explanted unflushed into, and snap frozen on dry ice and stored at −80° C. until further processing.


2.2.17 Lung Homogenization

2.2.17.1


Samples were homogenized using liquid nitrogen. Therefore, the organ was put in a mortar which was placed in an ice box. Liquid nitrogen was added in a volume, which covered the lung completely. The lung was pulverized in the mortar using a pistle. It was always ensured that the organ was covered by liquid nitrogen. A stapula was immersed in nitrogen and used to devide the organ powder in three parts. The organ powder was put in an empty and tared Eppendorf Tube. About 5 mg of the powder was taken for bDNA assay and the remaining powder was divided in two halfs, one for ELISA, one for qPCR. Each organ weight was noted.


Mortar, pistle and stapula were cleaned using Ethanol after every sample.


2.2.17.2


For ELISA, lysis was done in Triton X-100 lysis buffer (0.25 M Triethanolamine, 0.1% Triton X-100, pH7.7). Lysis was done on ice for at least 10 min. After every 6th sample, lysates were centrifuged at 14000 rpm for 10 min. Supernatant was collected in a fresh tube and stored at −80° C. until further processing.


2.2.17.3


For qPCR, 600 μL lysis buffer was added to the tube containing the organ powder. Lysis buffer RLT was provided by the kit and complemented with 40 μL/mL DTT. The solution was then transferred to a Lysing Matrix D tube and homogenization was done for 3×20 sec in a Tissue homogenizer. The lysate was transferred to a fresh Eppendorf tube. The volume of homogenate which refers to 10 mg lung was calculated, transferred into a fresh


Eppendorf tube and mixed using lysis buffer to a final volume of 350 μL. These samples were used directly for isolation of RNA without freezing. The left-over lung homogenates were stored at −80° C.


2.2.18 RNA Isolation and cDNA Synthesis for qPCR in Examples 14 to 17 and 18 to 20


RNA isolation from the lung lysates was done using 10 mg of lysed lung homogenate and the RNeasy mini kit from Qiagen according to manufacturer's protocol.


cDNA synthesis was performed as described in 1.1.6.1, supra. For a two-step cDNA synthesis 1 μg of total RNA and 1 μL OligoDT Primers in a total volume of 13 μL were used to prepare the first step, here called the template primer mixture.


2.2.19 Preparation of Standard Curve for Quantification of hIFNλ1 mRNA


For the exact calculation of amount of mRNA encoding human IFNλ1 in the samples a standard curve is needed. Therefore, 1 μg of hIFNλ1 mRNA was reverse. Then a serial dilution of cDNA in nuclease free water was made in following dilutions: 1:101, 1:102., 1:103, 1:104, 1:105, 1:106 1:107. The dilutions were prepared independently. This means that e.g. the 1:102 dilution was not made from the 1:101 dilution used in the assay, but from a separate 1:101 dilution. From each dilution 2 μL were added to 8 μL master mix to perform qPCR.


2.2.20 qPCR Using UPL Probe


For quantification of mRNA encoding human IFNλ1, including standard curve, an universal probe library (UPL) probe was used.


The following components shown in Table 14 were combined and vortexed briefly. To bring the reaction mix to the bottom of the tube and to eliminate air bubbles the mix was briefly centrifuged at 3200 g for 2 min.


8 μL of TaqMan master mix were transferred to an optical 96-well qPCR reaction plate. 2 μL cDNA template were added to the optical 96-well qPCR reaction plate to obtain a final volume of 10 μL. The optical 96-well qPCR reaction plate was sealed with optical adhesive film and briefly centrifuged at 3200 g for 2 min.


Run mode “new experiment based on Roche Template” was selected and “HydrolysisProbes” was chosen on LightCycler96.









TABLE 14







Master mix qPCR UPL











Volume per



Component
well [μL]














Nuclease free water
2.625



Primer 1 [20 μM]
0.150



Primer 2 [20 μM]
0.150



UPL [stock]
0.075



Fast Start Essential DNA Probes Master
5



Total
8











2.2.21 Calculation of hIFNλ1 mRNA Content


The amount of mRNA encoding human IFNλ1 in the sample can be determined with the standard curve. The standard curve was made by placing the log (amount of hIFNλ1 mRNA) on the x-axis and the ct values on the y-axis from the standard curve.


Exemplary Equation From the Standard Curve





Y=−3,2449X+9,1445


From this the amount of IFNλ1 mRNA in the sample (X: log of concentration) was determined (Y: ct value of sample).









TABLE 15







Example calculation: Calculation of the


total amount of IFNλ1 mRNA per lung


















IFNλ1
Total





Log

mRNA in
amount of





(measured
Measured
30 mg
IFNλ1



Lung

IFNλ1
IFNλ1
lung
mRNA in



weight
Ct
mRNA
mRNA
tissue
lung


Sample
[mg]
Value
[ng]) = X
[pg]
[pg]
[pg]
















Sam-
360.8
18.25
−2.81
1.56
78.14
939.77


ple 1


Sam-
270.9
16.75
−2.34
4.53
226.54
2045.62


ple 2









2.2.22 CXCL9 and CXCL10 ELISA in Plasma

Plasma samples of this study were measured according to the manufacturer's instructions. Changes to the protocol can be found in Table 16.









TABLE 16







Parameters of the CXCL9 + 10 ELISA protocols in plasma


not included in the manufacturer's instructions.
















Diluent for





Sample

Blocking
antibodies
Time for
Sample


Type
Cytokine
solution
and samples
TMB
dilution
LLOQ





Plasma
CXCL9
Casein
0.05% Tween-
16 min
1:2 
40





20 in 1x PBS


pg/mL



CXCL10
1% BSA in
1% BSA in
25 min
1:20
100




1x PBS
1x PBS


pg/mL









2.2.23 CXCL9 and CXCL11 ELISA in Lung Homogenate

Lung homogenates of this study were measured according to the manufacturer's instructions. Changes to the protocol can be found in Table 17.









TABLE 17







Parameters of the CXCL9-11 ELISA protocols in plasma


not included in the manufacturer's instructions.
















Diluent for





Sample

Blocking
antibodies
Time for
Sample


Type
Cytokine
solution
and samples
TMB
dilution
LLOQ





Lung
CXCL9
Casein
0.05% Tween-
16 min
1:50
100


homogenate


20 in 1x PBS


pg/mL



CXCL11
Casein
0.05% Tween-
16 min
1:30
120





20 in 1x PBS


pg/mL










2.2.24 Intranasal application in Examples 18 to 20


Animals were anesthetized by inhalation of pure oxygen at a flow rate of 2 L/min supplemented with approximately 3% of the anesthetic gas Isoflurane in an inhalation chamber. hIFNλ1 mRNA was applied at a volume of 50 μL on the nosetip as one droplet and was subsequently actively inhaled through both nostrils during the next following physiological inspiratory movement. The animal was held in the perpendicular position during this procedure and was subsequently placed in the supine position until recovery from anesthesia. Treatment was performed every other day for a total of 3 drug administrations. Necropsy was performed 24 hours after the last application.


2.2.25 Clinical Examination in Examples 18 to 20

Clinical examination was performed as described in 1.2.3, supra, with the exception that animals were examined clinically before and 3 hours after the first treatment and subsequently daily until the day of necropsy. Furthermore, body weight was measured before application and daily until the final day of the experiment.


2.2.26 Necropsy in Examples 18 to 20

Animals were set under full anesthesia by intraperitoneal injection of Fentanyl/Midazolam/Medetomidin (0.05/5.0/0.5 mg/kg bw). Capillary blood was taken from retrobulbar venous plexus using non-heparinized 0.8 mm capillaries and was collected in EDTA tubes. Subsequently, mice were killed by cervical dislocation. The thoracical and abdominal cavity were opened and the lung was explanted unflushed into.


The right main bronchus was ligated, the right lung explanted and snap frozen on dry ice and stored at −80° C. until further processing. The left lung was insufflated with 0.4 mL fixative (4% paraformaldehyde-solution) via the trachea. Subsequently, the left bronchus was ligated and the insufflated left lung including the trachea transferred to 4% paraformaldehyde solution for diffusion fixation for about 24 h. After completion of fixation time, the lung samples were transferred to 70% ethanol.


2.2.27 Preparation of Plasma

EDTA-blood samples were centrifuged at 2.000×g for 5 min at 4° C. Supernatant was frozen on dry ice and subsequently stored at −80° C. until further processing.


2.2.28 Lung Homogenization in Examples 18 to 20

Lung homogenization was performed as described in 2.2.17.3, supra.


2.2.29 Test System in Examples 21 to 24





    • Species: Mustela putorius (ferret)

    • Strain: Sable

    • Source of supply: Triple F Farms (Gillett, PA)





Ferrets were between 20 and 24 weeks old at the time of LF92 formulated hIFN mRNA dosing. In Example 24, ferrets were five months old at the time of LF92 formulated hIFN mRNA dosing. Animals ranged from 0.7 kg to 1.1 kg in weight on the day before dosing (Day −1). Each animal was permanently identified by unique ear tag at NLS following animal receipt.


2.2.30 Test System Housing in Examples 21 to 24

Animals were acclimated for 10 to 12 days prior to study initiation. During the acclimation period, the health status of animals was evaluated daily by technical staff for clinical presentation and behavioral signs indicating normality or illness. Animals were housed in groups of three in stainless steel cages with wire bottoms during acclimation and throughout the in-life phase of the study.


2.2.31 Test Dosing in Examples 21 to 23

Animals were administered 1 mL of LF92 formulated hIFN mRNA by nasal instillation with a pipet (0.5 mL/nostril). LF92 formulated human IFNλ1 or Stop mRNAs were applied at a mRNA concentration of 0.07 mg/mL. In Example 23 a mRNA concentration of 1.25 mg/mL was applied leading to a total mRNA concentration of 2.5 mg/mL in case of the combination of human IFNλ1 and human IFNβ.


2.2.32 Test Dosing in Example 24

On study Day −1, 1 and 3, animals were administered LF92 formulated hIFN mRNA by nasal instillation with a pipet (0.5 mL/nostril of 0.125 or 0.25 mg/mL solution). LF92 formulated mRNA encoding human IFNλ1, human IFNβ or a combination of both was applied at a mRNA concentration of 1.25 mg/mL, leading to a total mRNA concentration of 2.5 mg/mL in case of the combination. Vehicle was applied as control.


On study day 0 animals of all groups were challenged by nasal instillation with 1 ml of freshly diluted H1N1 A/California/04/09 virus until 2×102 TCID 50/ml titer. The viral titer was confirmed the same day additionally (back titration).


2.2.33 Necropsy in Examples 21 to 23

All animals were humanely euthanized while under a heavy plane of sedation at 6 hours post dosing. Lung tissue specimens were collected from the upper, middle, and lower regions of the upper left (left cranial), upper right (right cranial), lower left (left caudal), and lower right (right caudal) lobes of the lungs. Tissue specimens were weighed, cut into 3 pieces and placed into cryovial tubes containing 1 mL RNAlater and stored at 4° C. overnight to allow RNAlater to penetrate tissue. RNAlater solution was removed from the tubes and the tissues were snap frozen and stored at −80° C. until shipped to ethris.


2.2.34 Lung Homogenization in Examples 21 to 23

600 μL lysis buffer was added to Lysing D tubes and stored at ice. Lysis buffer RLT was provided by the kit and complemented with 40 μL/mL DTT. The frozen organ was weighed and depending on weight transferred to one lysing tube or cut into pieces and transferred to several lysing tubes. 300-350 mg in 600 μL lysis buffer was determined to be the maximum amount of tissue and volume of lysis buffer for one lysing tube. While processing further samples, organs in lysis buffer were stored on ice. Homogenization was done for 3×20 sec in a tissue homogenizer. Lysates were then centrifuged for 3 min at maximum speed (14000 rpm). The supernatant was transferred to a fresh Eppendorf tube. The volume of homogenate which referred to 10 mg lung was transferred into an additional fresh Eppendorf tube. To this tube, lysis buffer was added to a final volume of 600 μL. These samples as well as residual lysates were frozen at −80° C. before isolation of RNA.


2.2.35 Clinical Observations in Example 24

Ferrets were observed twice daily and scored for clinical signs of illness, including sneezing, nasal discharge, and activity. A score of 0 or 1 was assigned for the absence or presence of sneezing or nasal discharge. Activity was scored as 0=normal activity, 1=reduced activity, and 2=inactive. Daily scores were reported for each individual ferret displaying the specific clinical sign on a given day. Scores were also reported as the number of ferrets within the groups displaying one or more signs of infection on a given day.


2.2.36 Body Weights and Body Temperatures in Example 24

Body weights were measured at the time of receipt, prior to first administration of the LF92 formulated hIFN mRNA and on each following day of the study.


Body temperatures were measured daily using transponders implanted subdermally into the ferrets on Day −1.


2.2.37 Viral Titers in Nasal Washes

Nasal washes were collected on Days 1, 2, 3, 4, 5, and 6 post-viral challenge by flushing the nasal cavities with 2 mL of sterile PBS containing 0.5% bovine serum albumin (BSA), penicillin, streptomycin, and amphotericin B. Washes on Days 1 and 3 were collected prior to LF92 formulated hIFN mRNA administration. Nasal washes were collected and aliquoted into sterile 1.5-ml Eppendorf tubes and immediately frozen on dry ice then stored at −80° C. until used for determination of viral titer by TCID50.


Serial 1/2-log10 dilutions of nasal washes from 10−1 to 10−10 were prepared in viral growth medium (DMEM supplemented with 0.3% BSA) without TPCK-trypsin. Twenty-five microliters (25 μl) of ½-log10 dilutions of each nasal wash were added (four (4) replicates for each dilution) to MDCK cells in a 96-well microplate and incubated at 37° C., 5% CO2 for 60 min. One hundred seventy-five microliter (175 μl) of viral growth medium (DMEM supplemented with 0.3% BSA with 1 μg/ml TPCK-trypsin) was added to each well following the 60-minute incubation. The cells were then incubated 48 h at 37° C., 5% CO2. The content of each well was tested for hemagglutination by incubating 50 μl of tissue culture supernatant with 50 μl of 0.5% turkey red blood cells in 1×PBS for 30 min at room temperature. The TCID50 was calculated by the Reed and Muench method. Briefly, the presence of influenza virus will agglutinate red blood cells. The number of positives (wells with hemagglutination) and negatives wells at each dilution was determined. The ‘cumulative positive’, ‘cumulative negative”, ratio and % of positives were calculated. Then the ‘proportional distance’ between dilution showing >50% positives and the dilutions to the dilution showing <50% positives was calculated using next equation: Proportional distance=((% positive value above 50%) 50)/(%positive value above 50%-% positive value below 50%)×0.5 (correction factor for ½ log dilution). The viral TCID50 for each specimen was calculated by adding the proportional distance to the dilution showing >50% positive.


2.2.38 Necropsy in Example 24

Animals were humanely euthanized on Day 6 and lung tissue was harvested from all four lung lobes. Lungs were photographed, and individually weighed. Three portions of each lobe were collected. Two of the portions were placed into pre-weighed tubes containing RNAlater (or equivalent) and weighed, the third lung portion was placed into 10% buffered formalin.


2.2.39 Quantification of Viral RNA in Lung

Quantification of influenza virus in ferret lung tissues was determined by one-step RT-qPCR in an Applied Biosystems QuantStudio 6-flex thermal cycler (Applied Biosystems, Foster City, CA, USA). Viral RNA was extracted from lung tissue specimens harvested from the upper left lobe using Zymo Research Quick-RNA Miniprep Plus Kit (Santa Ana, CA) obtaining 60 μL of eluted viral RNA which was aliquoted and stored at −80° C. until used for quantification. Five microliters (5 μL) of extracted RNA was used to program 20 μL one-step RT-qPCR reactions containing qScript XLT One-Step RT-qPCR ToughMix plus Rox (QuantBio) and 0.2 μM of primers M+24 F (5′-AGA TGA GTC TTC TAA CCG AGG TCG-3′) and Rev-mod (5′-TGC AAA GAC ACT TTC CAG TCT CTG-3′) and TaqMan probe M+64 (5′6-FAM/TC AGG CCC C/ZEN/C TCA AAG CCG A-3′BkFQ) for amplification of sequences on viral gene segment 7 encoding the viral Matrix protein. Amplification was performed in a 96-well plate with a single cycle of 50° C. for 10 minutes for reverse transcription followed by 3 minutes at 95° C. and 45 cycles of 95° C. for 10 seconds, 60° C. for 30 seconds for DNA amplification. The quantity of viral RNA in each sample was interpolated from a standard curve generated by amplification of serial dilutions of viral RNA extracted from cell culture stock (8×106 pfu/mL) of Influenza A/California/04/2009 pdmH1N1. The total viral RNA in each sample was normalized to the relative quantity of ferret GAPDH RNA in one milligram of total RNA. Three microliters (3 μL) of extracted RNA was used to program 20 μL one-step RT-qPCR reactions containing qScript XLT One-Step RT-qPCR ToughMix plus Rox (QuantBio) and 0.2 μM of primers Fer_GAPDH FWD (5′-CAA CGG ATT TGG CCG TAT TG-3′) and Fer_GAPDH REV (5′-CTG GAA CAT GTA GAC CAT GTA GT-3′) and TaqMan probe Fer_GAPDH PRB (5Cy5/AGGGTCATT/TAO/GATGGCGACAATATCCAC/3IAbRQSp/) for amplification of GAPDH sequences. Amplification was performed with a single cycle of 50° C. for 10 minutes for reverse transcription followed by 3 minutes at 95° C. and 45 cycles of 95° C. for 10 seconds, 60° C. for 30 seconds for DNA amplification. The quantity of GAPDH in the samples was interpolated from a standard curve generated by amplification of serial dilutions of pooled extracted ferret lung RNA (250 ng/μL). Results were processed by 21 CFR Part 11 Software Module for QuantStudio™ 6 and 7 Systems and data from RT-qPCR analysis were imported into GraphPad Prism for analysis.


2.2.40 Quantification of Viral Reproduction in Example 25
2.2.40.1 Study Approval

All experiments involving animals were approved in advance by the Animal Ethics Committee at the Danish Veterinary and Food Administration (Stationsparken 31-33, 2600 Glostrup, Denmark) and were carried out in accordance with the Danish Animal Welfare Act for the Care and Use of Animals for Scientific Purposes.


2.2.40.2 Biosafety

All aspects of this study were approved by the office of Danish Working Environment Authority, Landskronagade 33, 2100 Copenhagen Ø, before initiation of this study. Work with SARS-COV-2 was performed in a biosafety level 2+ laboratory by personnel equipped with powered air-purifying respirators.


2.2.40.3 SARS-COV-2 Propagation

B.1.1.7 SARS-COV2 (Kent, UK, isolate) was provided under MTA by Professor Arvind Patel, University of Glasgow. The viruses used are clinical isolates. The B.1.1.7 variant is in the database as MZ314997. The viruses were propagated in VeroE6 cells expressing human TMPRSS2 (VeroE6-hTMPRSS2) (kindly provided by Professor Stefan Pohlmann, University of Göttingen) (Hoffmann et al., 2020). Briefly, VeroE6-hTMPRSS2 cells were infected with a multiplicity of infection (MOI) of 0.05, in DMEM (Gibco) +2% FCS (Sigma-aldrich)+1% Pen/Strep (Gibco)+L-Glutamine (Sigma-Aldrich) (From here, complete medium). 72h post infection supernatant, containing new virus progeny, was harvested, and concentrated on 100 kDa Amicon ultrafiltration columns (Merck) by centrifugation at 4000×g for 30 minutes. Virus titer was determined by TCID50% assay and calculated by Reed-Muench method. To convert to the mean number of plaque forming units (pfu)/mL, the TCID50/mL was multiplied by factor 0.7 (ATCC—Converting TCID[50] to plaque forming units (PFU)).


2.2.40.4 Animal Housing

K18-hACE c57BL/6J mice (strain: 2B6.Cg-Tg(K18-ACE2)2Prlmn/J) were obtained from The Jackson Laboratory. Hemizygote offspring were used for experiment. Age matched male and female mice and fed standard chow diets groups and fed standard chow diets and were housed in in pathogen free facility. Mice were weighed every day at the same time of the day until day 3 of post infection (post infection, and the animals were killed at 20% weight loss or when they reached humane endpoint.


2.2.40.5 Treatment of Mice

Mice were treated intranasally under Isoflurane anesthesia with an 15 μL inoculum of formulated human IFNλ1 mRNA (7.5 μg mRNA, corresponds to 3 μg deposited in the lung) one day before and one day following infection.


2.2.40.6 Infection of Mice

Inoculation of SARS-COV-2 was performed in the same way as the application of mRNA (intranasal application of a 15 μL aliquot under Isoflurane inhalation anaesthesia).


2.2.40.7 RNA Isolation, Real-Time qPCR


Lunges were homogenized with steel beads (Qiagen) in a Tissuelyser (II) (Qiagen) in PBS and immediately used for RNA isolation. RNA was isolated using the High Pure RNA Isolation Kit (Roche) and equal amount of RNA was used for standard One-Step RT-PCR (Applied Biosystems TaqMan RNA to CT One Step Kit). For the SARS-COV-2 N gene qPCR primers AAATTTTGGGGACCAGGAAC and TGGCACCTGTGTAGGTCAAC and probe FAM-ATGTCGCGCATTGGCATGGA-BHQ were used. For the IFN-β, Mx1 and β-Actin the Taqman Gene Expression Assays were used (Applied Biosystems). RNA levels of SARS-COV-2 N gene, IFNβ, or Mx1 were normalized to the mouse housekeeping gene β-Actin using the formula 2{circumflex over ( )}(Ct(18S-rRNA)-Ct(Sars-COV-2 RNA)).


2.2.40.8 TCID50 Assay

To determine the amount of infectious virus in cell culture supernatant or generated virus stocks, a limiting dilution assay was performed. 2×104 VeroE6-TMPRRS2 cells were seeded in 90 μL DMEM5 in a 96 well plate. The next day, samples were thawed and 10× diluted, followed by 10-fold serial dilution using DMEM, and 10 μL of each dilution was added to the cells in eight replicates. The cells were incubated 72 h in a humidified CO2 incubator at 37° C., 5% CO2. Cytopathic effect (CPE) was scored after fixing with 5% formalin (Sigma-Aldrich) and crystal violet stain (Sigma-Aldrich), using a light microscope (Leica DMi1), and the tissue culture infectious dose 50 (TCID50/mL) was calculated using the Reed and Muench method.

Claims
  • 1. A method of treating or preventing a viral-induced disorder comprising administering to a subject a pharmaceutical composition comprising an mRNA encoding an IFN-λ polypeptide.
  • 2. The method of claim 1, wherein said viral-induced disorder is a viral-induced respiratory disorder.
  • 3. The method of claim 2, wherein the virus which causes said viral-induced respiratory disorder is selected from the group consisting of rhinovirus, influenza virus, parainfluenza virus, metapneumo virus, respiratory syncytial virus, adenovirus and corona virus.
  • 4. The method of claim 2, wherein the virus which causes said viral-induced respiratory disorder is a virus which enters cells via the ACE2 receptor.
  • 5. The method of claim 4, wherein said virus is SARS- COV, SARS-COV-2 or HCoV-NL63.
  • 6. The method of claim 2, wherein the mRNA is to be administered by delivery into the respiratory system.
  • 7. The method of claim 6, wherein said delivery into the respiratory system is inhalation.
  • 8. The method of claim 7, wherein said inhalation is inhalation of an aerosol comprising said mRNA.
  • 9. The method of claim 1, wherein the mRNA contains a combination of unmodified and modified nucleotides, wherein 5 to 50% of the uridine nucleotides and 5 to 50% of the cytidine nucleotides are modified uridine nucleotides and modified cytidine nucleotides.
  • 10. The method of claim 1, wherein said IFN-λ polypeptide is selected from the group consisting of IFNλ1, IFN80 2 and IFNλ3 or a combination thereof.
  • 11. The method of claim 10, wherein the coding region of the mRNA coding for IFNλ1 is as shown in SEQ ID NO: 1, wherein the coding region of the mRNA coding for IFNλ2 is as shown in SEQ ID NO: 3 or wherein the coding region of the mRNA coding for IFNλ3 is as shown in SEQ ID NO: 5.
  • 12. The method of claim 10, wherein the mRNA coding for IFNλ1 has a sequence as shown in SEQ ID NO: 13, wherein the mRNA coding for IFNλ2 has a sequence as shown in SEQ ID NO: 14 or wherein the mRNA coding for IFNλ3 has a sequence as shown in SEQ ID NO: 15.
  • 13. The method of claim 1, further comprising an mRNA encoding a type I interferon and/or an mRNA encoding a type II interferon.
  • 14. The method of claim 13, wherein the type I interferon is selected from the group consisting of IFN-α, and IFN-β, and wherein the type II interferon is IFNγ.
  • 15. A DNA molecule having a sequence as shown in any one of SEQ ID NOs: 10 to 12.
  • 16. The method of claim 9, wherein the modified uridine nucleotides are 2-thiouridine and the modified cytidine nucleotides are 5-methylcytidine.
  • 17. The method of claim 14, wherein IFN-α is IFN-α16.
Priority Claims (1)
Number Date Country Kind
20205674.3 Nov 2020 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2021/080658 11/4/2021 WO