PLASMID CONTAINING A SEQUENCE ENCODING AN MRNA WITH A SEGMENTED POLY(A) TAIL

Information

  • Patent Application
  • 20210348172
  • Publication Number
    20210348172
  • Date Filed
    October 10, 2019
    4 years ago
  • Date Published
    November 11, 2021
    2 years ago
Abstract
The present disclosure provides a DNA plasmid comprising a sequence which encodes an mRNA molecule and a modified poly(A) tail, wherein the part of the sequence that encodes the modified poly(A) tail is characterized in that it consists of at least two A elements each defined as a nucleotide sequence consisting of 55 to 65 T nucleotides and at least one S element each S element consisting of one nucleotide that is not a T nucleotide, or 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide, wherein the number of A elements is one more than the number of S elements, and wherein any two A elements are separated by one S element; said DNA plasmid exhibiting a reduced recombination during amplification in a bacterial host cell compared to the same DNA plasmid without said at least one S element.
Description

The present invention relates to a DNA plasmid comprising a DNA sequence which contains a first nucleotide sequence which encodes an mRNA molecule and, located downstream thereof, a second nucleotide sequence which encodes a modified poly(A) tail, wherein said second nucleotide sequence is characterized in that it consists of at least two A elements each defined as a nucleotide sequence consisting of 55 to 65 T nucleotides and at least one S element each S element consisting of i) one nucleotide that is not a T nucleotide, or ii) 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide, wherein the total number of A elements is one more than the total number of S elements, and wherein any two A elements are separated by one S element.


Genetic information is stored as deoxyribonucleic acid (DNA) in the cell and can be transcribed into ribonucleic acid (RNA) when required. Both, DNA and RNA molecules, are built up of nucleotides consisting of a nitrogenous base, a five-carbon sugar, and at least one phosphate group. Different types of RNA molecules exist including mRNA molecules that carry the genetic information for protein synthesis. In eukaryotes, these mRNA molecules are transcribed from the DNA as pre-mature mRNA molecules and subsequently modified by adding for example a 3′ polyadenosine (poly(A)) tail. The poly(A) tail is characteristic for mature, functional mRNA molecules with one of the few known exceptions being histone mRNAs (Marzluff et al., 2008, Nat. Rev. Genet., 9(11):843-854; Yang et al., 2011, Genome Biology, 12:R16). The mature mRNA molecule is then transferred from the cell nucleus into the cytoplasm where it is translated into proteins. Thus, the DNA sequence as well as the amount, stability and translational efficiency of the mature mRNA molecule mainly determine the synthesis of the respective protein in a cell.


For optimizing the synthesis of a desired protein in a cell, for example in therapeutic contexts, mRNA based approaches represent a promising tool. The use of mRNA molecules has the advantage that the molecules have to be introduced only into the cytoplasm of a cell for protein translation (Tavernier et al., 2011, J Control Release, 150(3):238-247; Yamamoto et al., 2009, Eur J Pharm Biopharm, 71(3):484-489). Compared to the use of the respective DNA sequence comprised in an appropriate carrier such as a plasmid, the use of mRNA molecules is furthermore less difficult, more efficient, and avoids the considerable risk of chromosomal DNA being altered if the plasmid or parts thereof become incorporated into the genome. Initial challenges associated with introducing mRNA molecules into cells such as the instability of mRNA molecules and immune responses have been successfully met using for example chemically modified nucleotides (WO 2011/012316; EP patent application number 18 15 6466.7; KarikO et al., 2008, Mol. Ther., 16(11):1833-40). Several determinants of mRNA efficiency have been identified and investigated so far, including the efficiency of 5′ capping and the composition of the 5′ cap, the nature of untranslated regions, codon optimization of protein coding sequences, the presence of miRNA target sequences in the protein coding sequence and untranslated regions, and the length of the 3′ poly(A) tail (e.g. Thess, et al., 2015, Mol. Ther., 23(9), 1456-1464; Trepotec et al., 2018, Tissue Engineering. Part A, ten.TEA.2017.0485; Ziemniak et al., 2013, Future Med. Chem., 5(10), 1141-1172).


For improving the efficiency of mRNA based approaches, optimizing the length of the poly(A) tail is crucial. The length of the poly(A) tail is considered as major determinant of mRNA turnover as the degradation of the poly(A) tail is the first step in mRNA decay before either 5′ cap hydrolysis or further 3′ to 5′ degradation occurs (e.g. reviewed in Lodish et al., Mol. Cell Biol. 4th edition, New York: W. H. Freeman, 2000, Section 11.2; Ramanathan et al., 2016, Nucleic Acids Res., 44(16): 7511-7526). In mammals for example, poly(A) tails have been reported to have an average length of approximately 200 to 300 nucleotides. Furthermore, it has been reported that poly(A) tails can exhibit terminal nucleotides other than A nucleotides and that for example a terminal G nucleotide can retard the degradation of the poly(A) tail (Chang et al., 2014, Mol. Cell 53(6):1044-1052). However, as the poly(A) tail is not encoded in the respective DNA sequence, but enzymatically added to the transcribed pre-mature mRNA molecule, its length remains difficult to optimize.


Although the length of the poly(A) tail is known to be one of the most important physiological factors influencing mRNA stability and translational efficiency, existing technologies struggle with the production of mRNA molecules with defined poly(A) tail length on large scale. mRNA molecules for protein synthesis, e.g. in therapeutic applications, are primarily generated by in vitro transcription. Such approaches are based on DNA templates, in which the poly(A) tail is already encoded and which are amplified by cloning or polymerase chain reaction (PCR). Compared to an enzymatically catalyzed addition of a poly(A) tail to a generated mRNA these approaches theoretically have the advantage of providing poly(A) tails of defined and reproducible length (Holtkamp et al., 2006, Blood, 108(13), 4009-4018). In view of large scale production of mRNA molecules, plasmid based approaches are of special interest. Plasmid production is well established, can easily be scaled up and performed under Good Manufacturing Practice conditions. Compared to PCR based approaches, plasmid production costs and the risk of obtaining undesired mutations in coding sequences of the mRNA molecule are comparatively low. However, one main challenge persists using plasmids for bacterial amplification of template DNA sequences: homopolymeric sequences, such as the sequence encoding the poly(A) tail, can recombine during bacterial amplification of the plasmid DNA and, as a consequence, the sequences encoding the poly(A) tails are shortened over time in an unpredictable manner (Grier et al., 2016, Mol. Ther. Nucleic Acids, 5(4):e306). Grier and colleagues reported for example that sequences encoding a 70 nucleotide long poly(A) tail remained stable in contrast to sequences encoding poly(A) tails that consisted of at least 100 nucleotides, which is still much shorter than the on average 300 nucleotides long poly(A) tail of newly synthesized mature mRNA molecules in eukaryotes (Grier et al., 2016, Mol. Ther. Nucleic Acids, 5(4):e306). In an attempt to increase the poly(A) tail length, research focused on the use of a linear plasmid system, which allows stable cloning of sequences encoding poly(A) tails of up to 500 nucleotides (Grier et al., 2016, Mol. Ther. Nucleic Acids, 5(4):e306). In particular, a sequence encoding the mRNA molecule can be incorporated into a plasmid using a multiple cloning site (MCS). Using a unique site for a site-specific restriction enzyme, sequences encoding a comparatively short poly(A) tail can be added in an iterative process, thus allowing the expansion of the poly(A) tail encoding sequence comprised in the plasmid. However, it remains unclear how this system can efficiently be scaled up for large scale production of templates for in vitro transcribed mRNAs as the used linear plasmid is a low copy plasmid (BigEasy® v2.0 Linear Cloning System (pJAZZ® Vectors), Lucigen). Hence, there is still a need to have at hand alternative solutions for being able to deliver on large scale templates for in vitro transcription of mRNA molecules with defined and stable poly(A) tail length.


The present application addresses the need for in vitro transcription templates that encode mRNA molecules with defined poly(A) tail lengths and that exhibit a reduced recombination of the poly(A) tail encoding sequence during amplification in a bacterial cell by providing the embodiments as recited in the claims.


In particular, the present invention relates to a DNA plasmid comprising a DNA sequence which contains (i) a first nucleotide sequence which encodes an mRNA molecule and, located downstream thereof, (ii) a second nucleotide sequence which encodes a modified poly(A) tail, wherein said second nucleotide sequence is characterized in that it consists of (a) at least two A elements each defined as a nucleotide sequence consisting of 55 to 65 T nucleotides, and (b) at least one S element each S element consisting of (b1) one nucleotide that is not a T nucleotide, or (b2) 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide; wherein the total number of A elements is one more than the total number of S elements, and wherein any two A elements are separated by one S element. In other words, the present invention relates to a DNA plasmid comprising a DNA sequence that can be transcribed, preferably in vitro, into an mRNA molecule with a modified, i.e. segmented poly(A) tail.


It has surprisingly been found that a DNA plasmid comprising such a DNA sequence which encodes an mRNA molecule with a modified poly(A) tail shows reduced recombination during amplification of said DNA plasmid in a bacterial cell compared to the same DNA plasmid comprising a DNA sequence which encodes the same mRNA molecule with the same poly(A) tail, but without said S elements.


In the context of the present invention, the terms “DNA” and “RNA” refer to single- or double-stranded DNA or RNA molecules. If not stated otherwise, the terms “DNA” and “DNA molecule” refer to a double-stranded DNA molecule built up of A, C, G, and/or T nucleotides, and the terms “RNA” and “RNA molecule” refer to a single-stranded RNA molecule built up of A, C, G, and/or U nucleotides. Herein, said A, C, G, T, and U nucleotides refer to nucleotides comprising adenine, guanine, cytosine, thymine, and uracil as the respective nitrogenous base.


In the context of the present invention the term “DNA plasmid” refers to a plasmid consisting of a double-stranded DNA molecule. If not stated otherwise, the term “plasmid” refers to a circular DNA molecule, though the term can also encompass linear DNA molecules. In particular, the term “plasmid” also covers molecules which result from linearizing a circular plasmid by cutting it, e.g. with a restriction enzyme, thereby converting the circular plasmid molecule into a linear molecule. Plasmids can replicate, i.e. amplify in a cell independently from the genetic information stored as chromosomal DNA in the cell and can be used for cloning, i.e. for amplifying genetic information in a bacterial cell. Preferably, the DNA plasmid according to the present invention is a medium- or high-copy plasmid, more preferably a high-copy plasmid. Examples for such high-copy plasmids are vectors based on pUC, pBluescript®, pGEM®, pTZ plasmids or any other plasmids which contain an origin of replication (e.g. pMB1, pColE1) that support high copies of the plasmid.


The plasmid according to the present invention preferably comprises at least an origin of replication (ORI), a marker gene or a fragment thereof and/or a reporter gene or a fragment thereof, and unique restriction sites which allow for the insertion of DNA elements, preferably restriction sites in the form of a multiple cloning site (MCS). The ORI is essential for replication. A marker gene such as an antibiotics resistance gene is advantageous to identify cells containing a plasmid that comprises said marker gene. A reporter gene such as lacZ or luciferase is advantageous to measure the strength and/or regulation of expression of a target gene it is linked to or it can be used as a vicarious target gene for testing the expression and/or establishing a new approach or system. An MCS comprises several restriction sites that are unique within the DNA plasmid and that are each specifically recognized by a restriction enzyme. In particular, an MCS is advantageous for incorporating genetic elements such as the DNA sequence or the second nucleotide sequence according to the present invention into a suitable DNA plasmid, preferably a high-copy DNA plasmid. In case of the latter, the DNA plasmid further comprises a target gene (encoding an mRNA) to which the modified poly(A) encoded by the second nucleotide sequence is linked to.


A double-stranded DNA molecule such as a DNA plasmid consists of two complementary DNA strands. The term “complementary” refers to nucleotides, the nitrogenous bases of which can naturally bind to each other by hydrogen bonds, i.e. A and T nucleotides, A and U nucleotides as well as C and G nucleotides. According to the present invention, one of the two strands of a double stranded DNA molecule comprises a “DNA sequence”. Herein, the term “DNA sequence” refers to a part of one strand of a double-stranded DNA molecule such as a DNA plasmid. According to the present invention said DNA sequence is built of A, C, G, and T nucleotides and comprises a first nucleotide sequence and a second nucleotide sequence as defined herein.


In the context of the present invention the “first nucleotide sequence” comprised in said DNA sequence is a “nucleotide sequence which encodes an mRNA molecule”. In other words, said first nucleotide sequence refers to the part of said DNA sequence that can be used as a template during transcription, i.e. the synthesis of an mRNA molecule. Hence, in case an “mRNA molecule” is transcribed from the first nucleotide sequence, said mRNA has a nucleotide sequence that is complementary to said first nucleotide sequence except that instead of T nucleotides U nucleotides are used for synthesis of the mRNA molecule. As the poly(A) tail is naturally not encoded in the DNA sequence, but enzymatically added to the mRNA molecule during and/or after transcription, the term a “nucleotide sequence which encodes an mRNA molecule” refers to the part of said DNA sequence that can be used as a template during transcription of an mRNA molecule without any poly(A) tail.


Herein, “encoding” or “encode” refers to i) genetic information comprised in a DNA sequence that can be transcribed into an mRNA molecule, and/or ii) genetic information comprised in an mRNA molecule that can be translated into an amino acid sequence. Hence, these terms also cover genetic information comprised in the DNA that can be converted via transcription of an mRNA molecule into an amino acid sequence such as a protein.


Herein, the terms “mRNA molecule” and “mRNA” are used interchangeable and refer to a class of RNA molecules, preferably single-stranded sequences built up of A, C, G, and U nucleotides that contain one or more coding sequences. Said one or more coding sequence can be used as a template during synthesis of an amino acid sequence during translation. In other words, the term “mRNA” should be understood to mean any RNA molecule which is suitable for the expression of an amino acid sequence or which is translatable into an amino acid sequence such as a protein.


Further features of the first nucleotide sequence encoding an mRNA molecule and of the encoded mRNA molecule are described in further detail below.


According to the present invention, said first nucleotide sequence comprised in the DNA sequence contained in a DNA plasmid, is followed by a second nucleotide sequence comprised in said DNA sequence, i.e. said second nucleotide sequence is located downstream of said first nucleotide sequence. In other words, said two sequences are located on the same strand of a single- or double-stranded DNA molecule such that in case transcription takes place the first nucleotide sequence is transcribed first and then the second nucleotide sequence.


In particular, said two nucleotide sequences are located in close proximity to or directly linked to each other. In a preferred embodiment, the first nucleotide sequence and the second nucleotide sequence are directly linked to each other, i.e. there are no intervening nucleotides. However, it is also possible that the first and the second nucleotide sequence are separated by intervening nucleotides. Preferably, the first and the second nucleotide sequences are separated by not more than 100 nucleotides, more preferably not more than 50, 40, or 30 nucleotides, and most preferably by not more than 20, 10, or 5 nucleotides.


According to the present invention, the DNA sequence of the DNA plasmid comprises a first nucleotide sequence and, located downstream thereof, a second nucleotide sequence. Said “second nucleotide sequence” is a “nucleotide sequence which encodes a modified poly(A) tail”. Herein, this refers to the part of said DNA sequence that can be used as a template for the synthesis of a modified poly(A) tail during transcription, preferably during in vitro transcription, of said DNA sequence. Naturally occurring poly(A) tails which are enzymatically added to a pre-mature mRNA molecule normally consist of A nucleotides. Such naturally occurring poly(A) tails are also referred to in the following as conventional poly(A) tails.


According to the present invention the term “modified poly(A) tail” refers to a segmented poly(A) tail that comprises nucleotides other than A nucleotides. In particular, said second nucleotide sequence encoding a modified poly(A) tail according to the present invention is characterized in that it consists of (a) at least two A elements each defined as a nucleotide sequence consisting of 35 to 65 T nucleotides, preferably of 55 to 65 T nucleotides, and (b) at least one S element each S element consisting of (b1) one nucleotide that is not a T nucleotide, or (b2) 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide; wherein the total number of A elements is one more than the total number of S elements, and wherein any two A elements are separated by one S element.


According to the present invention said A elements and said S elements are alternately disposed in said second nucleotide sequence. Hence, the term “wherein any two A elements are separated by one S element” is intended to be understood as “wherein any two consecutive A elements are separated by one S element”. For illustration, in case of three A elements and two S elements the second sequence according to the present invention consists of one, i.e. the first, A element; one S element; another, i.e. the second, A element; another S element; and a further, i.e. the third, A element. Hence, in this example the term “any two A elements” refers to the first and the second A element as well as the second and the third A element, but not to the first and the third A element. Thus, this term refers to any of the at least two A elements and the closest A element of the remaining A elements.


In some embodiments, the DNA sequence according to the present invention comprises a first nucleotide sequence followed by a second nucleotide sequence followed by one or more additional nucleotides other than T nucleotides. Said additional nucleotides are preferably 1 to 5 C nucleotides, more preferably 1 C nucleotide. In other words, the DNA sequence can encode an mRNA molecule, a modified poly(A) tail, and additional nucleotides, preferably one G nucleotide.


As demonstrated by the appended Examples, it has been found that using a DNA plasmid comprising a DNA sequence which contains a first and a second nucleotide sequence that encode an mRNA molecule and a modified poly(A) tail according to the present invention results in a reduced recombination during amplification of said DNA plasmid in a bacterial cell compared to the same DNA plasmid but with a second nucleotide sequence which encodes an unmodified poly(A) tail consisting of the same total number of A nucleotides as encoded by the second nucleotide sequence according to the present invention. In other words, using a DNA plasmid according to the present invention results in a reduced recombination during amplification of said DNA plasmid in a bacterial cell compared to the same DNA plasmid but without said at least one S element between said at least two A elements.


Surprisingly, it has also been observed that, at the same time, the stability and the translational efficiency of the polyribonucleotide transcribed from the DNA sequence according to the present invention are generally not negatively affected by the modified poly(A) tail as defined above.


Herein, the term “recombination” refers to homologous recombination, in which nucleotide sequences are exchanged between two highly similar or identical nucleotide sequences comprised for example in DNA molecules such as DNA plasmids and/or chromosomes. The term “recombination” comprises intramolecular homologous recombination events, i.e. recombination of two highly similar or identical nucleotide sequences comprised in the same DNA molecule. In the context of the present invention this refers in particular to the recombination of a nucleotide sequence encoding a modified and a conventional poly(A) tail, respectively, and another highly similar or identical nucleotide sequence comprised in the same DNA plasmid during amplification of said plasmid, and/or to the recombination of the two terminal parts of such a nucleotide sequence. Alternatively or additionally, the term “recombination” comprises intermolecular homologous recombination events between a nucleotide sequence encoding a modified and a conventional poly(A) tail comprised in a DNA plasmid, respectively, and a highly similar or identical nucleotide sequence comprised in another DNA molecule, e.g. another DNA plasmid, comprised in the same cell for example.


According to the present invention, the term “reduced recombination” means that recombination is reduced by at least 5%, preferably by at least 10%, more preferably by at least 25%, even more preferably by at least 50%, and most preferably by 100%.


A reduction in recombination can be determined for example by the following procedure: genetically identical bacterial cells are used for amplifying a DNA plasmid according to the present invention (group I) and for amplification of the same DNA plasmid except that the second nucleotide sequence does not comprise said at least one S element (group II), respectively. Using routine lab procedures, bacterial cells can be lysed upon amplification, DNA plasmids obtained and linearized and the comprised DNA sequences or second nucleotide sequences purified. Obtained DNA sequences, especially obtained second nucleotide sequences can then be investigating qualitatively in view of nucleotide composition, sequence, and/or length for example by sequencing and/or electrophoresis such as gel or capillary electrophoresis. Alternatively or additionally, occurrence and frequency of recombination can be determined quantitatively by measuring the length of the second nucleotide sequences, calculating the ratio of second nucleotide sequences that have the expected length as defined by the originally used second nucleotide sequence and the total number of second nucleotide sequences within each group, and comparing the ratio obtained per group between groups. Alternatively, the extracted DNA sequences can be transcribed in vitro before investigating the length of the transcribed poly(A) tails within and between group I and II as described above in case of the second nucleotide sequences.


Such a reduction of recombination has been observed for DNA plasmids comprising a second nucleotide sequence according to the present invention. In particular, it has, e.g., been observed that S elements consisting of a single nucleotide are advantageous for reducing recombination during bacterial amplification. Thus, in a particularly preferred embodiment of the present invention, the DNA plasmid comprises a first nucleotide sequence encoding an mRNA molecule and a second nucleotide sequence encoding a modified poly(A) tail, wherein any one of said S elements consists of one C nucleotide or one A nucleotide, preferably of one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of two A elements separated by one A nucleotide. More preferably, said two A elements are separated by one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of three A elements, wherein any two A elements are separated by one A nucleotide. Preferably, any two of said three A elements are separated by one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of four A elements, wherein any two A elements are separated by one A nucleotide. Preferably, any two of said four A elements are separated by one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of five A elements, wherein any two A elements are separated by one A nucleotide. Preferably, any two of said five A elements are separated by one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of six A elements, wherein any two A elements are separated by one A nucleotide each. Preferably, any two of said six A elements are separated by one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of seven A elements, wherein any two A elements are separated by one A nucleotide. Preferably, any two of said seven A elements are separated by one C nucleotide.


In some embodiments the second nucleotide sequence according to the present invention consists of eight A elements, wherein any two A elements are separated by one A nucleotide. Preferably, any two of said eight A elements are separated by one C nucleotide.


In case more than one S element is comprised in the second nucleotide sequence said at least two S elements can have the same sequence or can vary in their sequence. Preferably, said at least two S elements each consist of i) the same nucleotide in case of one nucleotide, or ii) the same nucleotide sequence in case of 2 to 10 nucleotides, preferably 6 nucleotides.


Hence, in some embodiments each of the at least one S elements comprised in the second nucleotide sequence according to the present invention consists of an A nucleotide. Preferably, each of the at least one S elements comprised in the second nucleotide sequence according to the present invention consists of a C nucleotide.


In case of at least two S elements, in some embodiments at least one, but not all of the S elements can consist of an A nucleotide and each of the at least one remaining S elements can consist of i) one G or preferably of a C nucleotide, or of ii) 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide.


In case of at least two S elements, in some embodiments at least one, but not all of the S elements preferably consist of a G nucleotide and each of the at least one remaining S elements can consist of i) an A or a C nucleotide, or of ii) 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide.


In some embodiments the number of A elements comprised in the nucleotide sequence encoding a modified poly(A) tail contained in said DNA sequence of a DNA plasmid as defined above is two, three or four. In other words, in particularly preferred embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of i) two A elements flanking one S element, ii) three A elements in alternating order with two S elements, or iii) four A elements in alternating order with three S elements; with A and S elements as defined above.


In some embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of two A elements each consisting of the same number of T nucleotides in alternating order with one S element which consists of i) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide or preferably ii) a C nucleotide.


In some embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of three A elements each consisting of the same number of T nucleotides in alternating order with two S elements, wherein all of said two S elements each consist of i) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide or preferably ii) a C nucleotide.


In some embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of four A elements each consisting of the same number of T nucleotides in alternating order with three S elements, wherein all of said three S elements each consist of i) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide or preferably ii) a C nucleotide.


In some embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of two, three, or four A elements, wherein at least two of said A elements differ in their number of T nucleotides, in alternating order with one, two, or three S elements, respectively, wherein all of said S elements each consist of i) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide.


In some embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of two, three, or four A elements, wherein at least two of said A elements differ in their number of T nucleotides, in alternating order with one, two, or three S elements, respectively, wherein preferably all of said S elements each consist one C nucleotide.


In some embodiments of the DNA plasmid as defined above, the number of A elements is four and the nucleotide sequences of the four A elements together have an overall length of 240 nucleotides.


Preferably, each A element has a length of 60 nucleotides. In other words, in particularly preferred embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of four A elements in alternating order with three S elements, wherein the four A elements together consist of 240 nucleotides and preferably have the same length each.


More preferably, said second nucleotide sequence which encodes a modified poly(A) tail consists of four A elements each consisting of 60 T nucleotides in alternating order with three S elements, wherein all three S elements each consist of i) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide or preferably ii) one C nucleotide.


In some embodiments of the DNA plasmid as defined above, the number of A elements is two and the nucleotide sequences of the two A elements together have an overall length of 120 nucleotides.


Preferably, each A element has a length of 60 nucleotides. In other words, in particularly preferred embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of two A elements having together a total length of 120 nucleotides. In other words, in particularly preferred embodiments said second nucleotide sequence which encodes a modified poly(A) tail consists of two A elements in alternating order with one S element, wherein the two A elements together consist of 120 nucleotides and preferably have the same length each. More preferably, said second nucleotide sequence which encodes a modified poly(A) tail consists of two A elements each consisting of 60 T nucleotides flanking one S element consisting of i) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide or preferably ii) one C nucleotide.


In some embodiments of the DNA plasmid as defined above, said DNA plasmid contains in the nucleotide sequence encoding a modified poly(A) tail three A elements having together a total length of 120 nucleotides. In other words, in this embodiment of the DNA plasmid as defined above, the number of A elements is three and said nucleotide sequences of the three A elements together have an overall length of 120 nucleotides, preferably each A element having a length of 40 nucleotides. Hence, the second nucleotide sequence encoding said modified poly(A) tail can consist of three A elements alternating ordered with two S elements. Preferably, each of the three A elements consists of 40 T nucleotides and each of the two S elements of i) preferably one C nucleotide or ii) 6 nucleotides wherein each of the two terminal nucleotides is not a T nucleotide.


According to the present invention, the DNA plasmid comprises a DNA sequence comprising a first nucleotide sequence followed by a second nucleotide sequence as defined above. Preferably, the first nucleotide sequence is preceded by a promoter comprised in the DNA sequence or the DNA plasmid. The promoter is involved in controlling and initiating the transcription of the following nucleotide sequence such as the DNA sequence comprising the first nucleotide sequence or the first nucleotide sequence. Hence, a promoter comprised in the DNA plasmid, preferably a strong promoter, can be advantageous for comparing effects of other genetic elements on the transcription duration and/or intensity of the following nucleotide sequence encoding an mRNA molecule. On the other hand, a promoter comprised in the DNA sequence, preferably a strong promoter, can be advantageous for optimizing transcription duration and/or intensity of the following nucleotide sequence encoding an mRNA molecule.


The promoter comprises at least a recognition site followed by a binding site. Recognition and binding site can interact with amino acid sequences that mediate or regulate transcription. The binding site is located closer to the first nucleotide sequence comprised in the DNA sequence according to the invention compared to the recognition site. For example, the recognition site can be a −35 box and the binding site a −10 box located in a distance of approximately 35 nucleotides and 10 nucleotides before the transcription start site, respectively. The binding site can be for example a Pribnow box in prokaryotes and a TATA box in eukaryotes. Preferably the binding site is a Pribnow box comprised in the DNA plasmid or the DNA sequence close to the first nucleotide sequence.


Optionally, the promoter comprises at least one additional regulatory element such as an AT-rich upstream element located approximately 40 and/or 60 nucleotides before the transcription start site, and/or additional regulatory elements located between the recognition and the binding site that enhance the activity of the promoter.


In a particularly preferred embodiment, the promoter is a strong promoter. In other words, the promoter comprises sequences that promote transcription of the following DNA sequence according to the present invention. Strong promoters are known to the person skilled in the art, for example the RecA promoter derived OXB18, OXB19, and OXB20 promoters for Escherichia coli, or can be identified or synthesized by routine lab procedures.


Optionally, the promoter is preceded by additional regulatory elements such as enhancers comprised in the DNA plasmid that promote transcription of the DNA sequence according to the present invention.


The DNA sequence comprises a first nucleotide sequence which encodes an mRNA molecule, i.e. can be converted into an mRNA molecule during transcription, wherein synthesis of the mRNA molecule starts with its 5′ end and ends with its 3′ end. Features of the first nucleotide sequence encoding an mRNA molecule are described in more detail below in connection with the encoded mRNA molecule.


The first nucleotide sequence comprises at least one region that encodes an untranslated region (UTR) and at least one coding region. Preferably, the first nucleotide sequence encodes at least the 5′ UTR and the coding region of the encoded mRNA molecule. The coding region can comprise non-coding and coding parts. Preferably, the coding region consists of at least one coding part that encodes the part of the mRNA molecule that can be translated into an amino acid sequence, or parts thereof.


In a particularly preferred embodiment, the first nucleotide sequence comprises a region encoding a 5′ UTR followed by a coding region followed by a region encoding a 3′ UTR of the encoded mRNA molecule.


According to the present invention, the first nucleotide sequence encodes an mRNA molecule. Preferably, the first nucleotide sequence encodes an mRNA comprising a 5′ UTR, a coding sequence and a 3′ UTR.


The 5′ end of the 5′ UTR is defined by the transcriptional start site and its 3′ end is followed by the coding sequence. The coding sequence is terminated by the start and the stop codon, i.e. the first and the last three nucleotides of the mRNA molecule that can be translated, respectively. The 3′ UTR starts after the stop codon of the coding sequence and is followed by the modified poly(A) tail encoded by the second nucleotide sequence according to the present invention.


The 5′ UTR generally comprises at least one ribosomal binding site (RBS) such as the Shine-Dalgarno sequence in prokaryotes and the Kozak sequence or translation initiation site in eukaryotes. RBS promote efficient and accurate translation of an mRNA molecule by recruiting ribosomes during initiation of translation. The activity of a given RBS can be optimized by varying its length and sequence as well as its distance to the start codon. Alternatively or optionally, the 5′ UTR comprises internal ribosome entry sites or IRES.


In some embodiments, the mRNA molecule comprises a 5′ UTR that comprises additionally one or more additional regulatory sequences such as a binding site for an amino acid sequence that enhances the stability of the mRNA molecule, a binding site for an amino acid sequence that enhances the translation of the mRNA molecule, a regulatory element such as a riboswitch, a binding site for a regulatory RNA molecule such as an miRNA molecule, and/or a nucleotide sequence that positively affects initiation of translation. Furthermore, within the 5′ UTR there are preferably no functional upstream open reading frames, out-of-frame upstream translation initiation sites, out-of-frame upstream start codons and/or nucleotide sequences giving rise to a secondary structure that reduces or prevents translation. The presence of such nucleotide sequences in the 5′ UTR can have a negative effect on translation.


The coding sequence is encoded by the at least one coding part of the first nucleotide sequence and comprises codons that can be translated into an amino acid sequence. Further features of the encoded amino acid sequence are described in detail below.


The coding sequence can contain the codons of a naturally occurring coding sequence or it can be a partially or completely synthetic coding sequence. Alternatively, the coding sequence can be a partly or fully codon optimized sequence derived from the natural sequence to be used. Most of the amino acids are encoded by more than one codon, i.e. three consecutive nucleotides of an mRNA molecule that can be translated into an amino acid. Codons exist that are used preferentially in some species for a given amino acid. The presence of more often occurring codons can enhance the amount of amino acid sequences translated based on a given mRNA molecule compared to the same mRNA molecule but comprising comparably rare codons. Hence, it is advantageous to species specific adapt the codons in a given coding sequence by avoiding rare codons and enhancing the occurrence of abundant codons for a given amino acid to improve the translation of said mRNA.


The mRNA may optionally also comprise a 3′ UTR. The 3′ UTR may comprise one or more regulatory sequences such as a binding site for an amino acid sequence that enhances the stability of the mRNA molecule, a binding site for a regulatory RNA molecule such as a miRNA molecule, and/or a signal sequence involved in intracellular transport of the mRNA molecule.


In a particularly preferred embodiment, the mRNA molecule encoded by the first nucleotide sequence comprises a 5′ UTR with at least one RBS and/or IRES, a coding sequence with optimized codons and a 3′ UTR with at least one regulatory sequence as defined above.


The mRNA molecule according to the present invention comprises a coding sequence, i.e. a sequence encoding an amino acid sequence. As regards the function of the encoded amino acid sequence, there is no limitation and possible amino acid sequences to be encoded by said polyribonucleotide are described further below. Herein, the term “amino acid sequence” encompasses any kind of amino acid sequence, i.e. chains of two or more amino acids which are each linked via peptide bonds and refers to any amino acid sequence of interest. Preferably, the encoded amino acid sequence is at least 5 amino acids long, more preferably at least 10 amino acids, even more preferably at least 50, 100, 200 or 500 amino acids. In other words, the term “amino acid sequence” covers short peptides, oligopeptides, polypeptides, fusion proteins, proteins as well as fragments thereof, such as parts of known proteins, preferably functional parts. These can, for example be biologically active parts of a protein or antigenic parts such as epitopes which can be effective in raising antibodies.


Preferably, the function of the encoded amino acid sequence in the cell or in the vicinity of the cell is needed or beneficial, e.g. an amino acid sequence the lack or defective form of which is a trigger for a disease or an illness, the provision of which can moderate or prevent a disease or an illness, or an amino acid sequence which can promote a process which is beneficial for the body, in a cell or its vicinity. The encoded amino acid sequence can be the complete amino acid sequence or a functional variant thereof. Further, the encoded amino acid sequence can act as a factor, inducer, regulator, stimulator or enzyme, or a functional fragment thereof, where this amino acid sequence is one whose function is necessary in order to remedy a disorder, in particular a metabolic disorder or in order to initiate processes in vivo such as the formation of new blood vessels, tissues, etc. Here, functional variant is understood to mean a fragment which in the cell can undertake the function of the amino acid sequence whose function in the cell is needed or the lack or defective form whereof is pathogenic.


Preferably, such an amino acid sequence is advantageous with respect to applications in supplemental or medical purposes to generate or regenerate physiological functions caused by suboptimal amino acid sequence biosynthesis and thus also to favorably influence directly or indirectly the course of diseases. Disorders with known genetic base are for example cystic fibrosis, haemophilia, hypertension, elevated cholesterol level, cancer, neurodegenerative disorders, mental illness and others. An online catalogue with presently 22,993 entries of Human Genes and Genetic Disorders together with their respective genes and a description of their phenotypes are available at the ONIM (Online Mendelian Inheritance in Man) webpage (http://onim.org); sequences of each are available from the Uniprot database (http://www.uniprot.org). As non-limiting examples, the following Table 1 lists some congenital diseases, and the corresponding gene(s). Due to the high degree of interaction of cellular signalling pathways, the mutation of a certain gene causes a multiply of pathogenic symptoms, of which only a characteristic one is listed in Table 1.











TABLE 1





Disease
Pathology
Gene, heredity















Blood diseases









Fanconi Anemia
Anemia and neutropenia,
FANCA, autosomal



evidence that a DNA repair
recessive



mechanism is affected



Hemophilia-A
Abnormal bleeding
Coagulation Factor VIII,




X-chromosomal




recessive


Hemophilia-B
Abnormal bleeding
Coagulation Factor IX, X-




chromosomal recessive


Hereditary
spherical-shaped
Ankyrin (ANK1)


Spherocytosis
erythrocytes



(various types)
(spherocytes)



Paroxysmal
Anemia and presence
PIG-A, X-chromosomal


nocturnal
of blood in the urine



hemoglobinuria




Porphyria cutanea
Overproduction of heme,
Uroporphyrinogen


tarda
iron overload
decarboxylase (UROD),




autosomal recessive


Severe combined
Due to impaired DNA
Adenosine deaminase,


immune deficiency
synthesis severe immune
autosomal recessive,


(SCID)
deficiency in humoral and
IL-2R-γ, JAK3, (IL-7R-α,



cellular immunity
RAG1/2, Artemis, CD3δ,




CD3ε


Sickle-cell anemia
Abnormal hemoglobin (HbS)
β-Hemoglobin (HB),




autosomal recessive


Thalassemia (α- and
Lack of α- or β hemoglobin
Deletion of HBA1 and/or


β form)
resulting in anemia
HBA2,


Von Willebrand
Abnormal bleeding,
Autosomal dominant


disease
hemorrhage similar to
and recessive forms


(three types known,
hemophilia A and B



Type-III is most




severe)









Cancer









Malignant
P16 mutation leads to
Cyclie dependant kinase


melanoma
uncontrolled proliferation
inhibitor 2 (CDKN2)



of fibroblasts



Neurofibromatosis
Benign tumors on auditory
NF1, NF2, autosomal


(2 types)
nerves leads to deafness
dominant







Deafness (Ear)









Deafness
Hearing loss
Deafness-1A (DFNB1),




autosomal recessive


Pendred syndrome
Hearing loss
Pendrin (PDS),




autosomal recessive







Heart









Ataxia
DNA damage repair
ATM,


telangiectasia
disturbed,



Atherosclerosis
Increase of blood cholesterol
apoE,


LQT Syndrome
Potassium channel defect
LQT1 and other genes


(Long QT)




Von-Hippel Lindau
Abnormal growth of blood
VHL, autosomal


Syndrome
vessels, can lead to cancer
dominant


William's Beuren
Deletion of elastin results
Deletion of elastin and


Syndrome
in vascular defects,
LIM kinase genes



supravalvular aortic stenosis








Metabolic disorders and glycogen storage diseases









Adrenoleukodystrophy
Disturbed fatty acid transport
ABCD1, X-chromosomal



and metabolism



Alkaptonuria
Nitrogen metabolism defect,
Homogentisic Oxidase,



Urine turns dark when
autosomal recessive



exposed to oxygen



Diabetes type I
Disturbed insulin production
IDDM1, IDDM2, GCK, . . .


Galactosemia
disorder of galactose
Galactose-1-phosphate



metabolism
uridyltransferase gene




(GALT), autosomal




recessive


Gauche disease
Disturbance of fat metabolism
Glucocerebrosidase


Glucose
Disturbed glucose and
SGLT1, autosomal


Galactosidase
galactose transport out of the
recessive


Malabsorption
intestinal lumen resulting in




diarrhea



Glycogen storage
Accumulation of glucose
Glucose-6-Phosphatase,


diseaseType I,
in liver and kidney
autosomal recessive


Von-Gierke's




disease




Glycogen storage
Accumulation of glycogen in
α-1-Glucosidase,


disease Type II,
liver, heart, skeletal muscle,
autosomal recessive


Pompe's disease
cardiomegaly



Glycogen storage
Accumulation of glycogen in
Debranching enzyme,


disease Type III,
liver, heart, skeletal muscle,
autosomal recessive


Cori's disease
hepatoomegaly



Glycogen storage
Cannot untilize glycogen
Muscle phosphorylase,


disease Type V,
in muscle cells
autosomal recessive


McArdle's disease




Glucose-6-
Inability to maintain,
G6PD X-chromosomal


Phosphate
glutathione leads to
recessive


Dehydrogenase
hemolytic anemia



Hereditary
Excess of iron in the body
Hemochromatosis (HFE)


Hemochromatosis
(esp. liver) due to excessive



(4 types)
iron absorption in the gut



Homocystinuria
Nitrogen metabolism defect
Cystathione synthetase




defect, autosomal




recessive


Lesh Nyhan
Accumulation of uric acid
HPRT1,


Syndrome
leading to gout, ureate
X-chromosomal



stones and muscle loss



Maple Syrup Urine
Amino acid metabolism
Branched-chain-alpha-


Disease
defect leads to the
dehydrogenase (BCKDH)



accumulation of




α-Ketoacides and death in




the first months if untreated



Menkes’ Syndrome
Reduced ability to absorb
ATP7A, X-chromosomal



copper, leads to death in
recessive



infancy if untreated



Obesity
Elevated body weight
Polygenic, elevated leptin




levels may play a role


Phenylketonuria
Inability to break down
Phenylalanine



Phenylalanine into tyrosine
hydroxylase (PAH),



leads to mental retardation
autosomal recessive


Tangier disease
reduced levels of plasma high
ATP-binding cassette-1



density lipoproteins
gene (ABCA1)


Zellweger
High levels of iron and copper
PXR1 (receptor on the


Syndrome (leads to
in the blood
surface of peroxisomes)


death in infants)




Wilsons Disease
Copper accumulation
ATP7B (P-type ATPase),



in brain and liver
autosomal recessive







Musculoskeletal system









Achondroplasis
Short stature with a large
Fibroblast growth factor



head due to slow proliferation
receptor 3 (FGF3R),



of chondrocytes



Charcot-Marie-
Degeneration of the
Different forms caused


Tooth Syndrome
muscles in limbs
by different gene


and its more severe

mutations, autosomal


form Dejerine-

recessive and X-


Sottas Syndrome

chromosomal


Cockayne
Premature aging and short
group 8 excision repair


syndrome (2 types)
stature, loss of “on the fly”
cross-complementing



DNA repair
protein (ERCC8)


Chondroectodermal
Malformation of bones and
EVC, autosomal


dysplasia
polydactyly
recessive


Diastrophic
Malformed hands, sulfate
DTDST gene


dysplasia (DTD)
transporter defect



Duchenne muscular
Enlargement of muscle tissue
DMD, X-chromosomal


dystrophy
with subsequent loss
of recessive



function



Fibrodysplasia
Heterotopic bone formation
NOG, BMP,


Ossificans

Autosomal


Progressiva

dominant


Friedreich's ataxia
Heart enlargement and
Frataxin, autosomal



progressive loss of muscular
recessive



coordination



Hypophosphatasia
Production of an abnormal
ALPL, autosomal



version of alkaline
recessive



phosphatase affecting the




mineralization process



Marfan Syndrome
Connective tissue disorder
Fibrillin 1 (FBN),



due fibrillin deficiency
autosomal dominant


Myotonic dystrophy
Protein kinase defect in
Dystrophia myotonica


(onset during young
skeletal muscle cells
protein kinase (DMPK),


adulthood)

autosomal dominant


Osteogenesis
Defect in type-I collagen
COL1A1, COL1A2


imperfect (various
formation leads to multiple



types)
fractures after birth



Prader-Willi
Decreased muscle tone and
SNRPN (small


Syndrome
mental retardation
ribonucleoprotein N)




deleted due to a deletion




on chromosome 15







Neurons and Brain









Alzheimer disease
Increased amyloid
Polygenic, PS1, PS2, . . .



production, progressive




inability to remember facts



Amyotrophic lateral
Progressive degeneration of
Superoxide dismutase 1


sclerosis (ALS)
motor neuron cells (defect
(SOD1), various genes


(various forms)
in elimination superoxide
involved



radicals)



Angelman
Mental retardation with
Genomic imprinting on


syndrome
inadequate laughing
chromosome 15


Pyruvat
Neurological defects
Pyruvat dehydrogenase,


dehydrogenase
if untreated
autosomal recessive


Refsum disease
Accumulation of phytanic acid
Phytanoyl-CoA



leads to peripheral
hydroxylase (PHYH),



neuropathy
autosomal recessive


Rett's syndrome
Mental retardation with
Methyl-CpG-binding



arrested development
protein-2 (MECP2), X-



between 6 and 18 months of
chromosomal dominant



age



Tay-Sachs disease
Disturbed break down of GM2
HEXA(β-


(various forms of
ganglioside leads to
hexosaminidase A),


severity)
neurological damage
autosomal recessive


LaFora Disease
Aggressive form of epilepsy
EPM2A, autosomal




recessive


Essential tremor
Uncontrollable shaking
ETM1, ETM2, autosomal


(variable forms)

dominant


Fragile X syndrome
Lack of FMR1 RNA binding
FMR1 gene is not



protein, mental retardation
expressed due to an




CGG amplification in




the 5'UTR region


Huntington's
Progressive dementia with
HTT (huntington),


disease
onset in adulthood
autosomal dominant







Intestine









Bartter's syndrome
Renal disease
Kidney chloride channel


(3 types)

B gene (CLCNKB),




autosomal recessive


Polycystic kidney
renal disease
PDK1, PDK2, autosomal


disease (2 types)

dominant, there is also




an autosomal recessive




form known (ARPKD)







Lung









Alpha-1-antitrypsin
Defect alveoli due to
SERPINA1, autosomal



uncontrolled release of
codominant



elastase



Asthma
Chronic inflammatory
Polygenic



disorder of the airways



Cystic fibrosis
Excessively viscous mucous
CFTR (cystic



due to defective Cl ion
fibrosis conductance



transport
transmembrane




regulator), autosomal




recessive


Surfactant
Newborns are of normal body
ATP-binding cassette


metabolism
weight, but all fail to inflate
transporter (ABCA3)


dysfunction




(various types)




Primary cliliary
Excessively viscous mucous
DNAI1, CCNO, CCDC40


dyskinesia
due to defective/missing cilia
among others



function








Lysosomal storage diseases









Fabry's disease
Beyond others, skin lesions
α-Galactosidase A, X-



due to the accumulation of
chromosomal recessive



ceramide trihexoside



Gaucher's Disease
Accumulation of
Glucocerebrosidase,


Type-I: adult form
glucocerebrosides
autosomal recessive,


(normal lifespan
(gangliosides,



under treatment)
sphingolipids)



Type-II: infantile




form (death




before age 1)




Type-Ill: juvenile




form (onset in early




childhood, less




severe than Type-II)




Hunter's Syndrome
Accumulation of
L-iduronosulfat sulfatase,



mucopolysaccharides
X-chromosomal




recessive


Hurler's Syndrome
Accumulation of
α-L-iduronidase,


(death by age of 10)
mucopolysaccharides
autosomal recessive


Niemann-Pick
Defect in releasing
Sphingomyelinase,


Disease (three
Cholesterol from lysosomes,
autosomal recessive


distinct forms A, B,
accumulation of



C)
Sphingomyelin



Tay-Sachs disease
Accumulation of GM2
Hexosaminidase A,


(death by age of 4)
ganglioside in neuronal cells
autosomal recessive







Skin









Albinism
Nitrogen metabolism
Tyrosinase deficiency,



defect
autosomal recessive


Albinism,
Reduced biosynthesis
OCA2, autosomal


oculocutaneous,
of melanin pigment
recessive


type II




Ehlers-Danlos
Diaphragmatic hernia.
Various defects in


Syndrome (various
common, retinal detachment
collagen synthesis


types)




Epidermolysis
Defects in maintenance of
Epidermolysis bullosa


bullosa
keratinocyte structural
macular type (EBM),


(various types
stability or adhesion of the
Epidermolysis bullosa 3


including EB
keratinocyte to the
progressiva (EBR3),


simplex, Junctional
underlying dermis
Epidermolysis bullosa 4


EB, Dystrophic EB

pseudojunctual (EBR4),


and Kindler

Desmoplakin (DSP),


syndrome)

Plakophilin-1 (PKP1),




kreatin (KRT5, KRT14),




plectin (PLEC), ITGA6,




integrin subunit (ITGB4),




laminin subunits




(LAMA3, LAMP3,




LAMB3, LAMC2),




collagen (COL17A1 ,




COL7A1 (autosomal




dominant), FERMT1,




autosomal recessive


Hartnup's disease
Defect in tryptophan uptake
SLC6A19, autosomal



in the gastrointestinal tract,
recessive



light-sensitive skin



Hereditary
Telangiectasia of the skin
Endoglin (ENG),


Hemorrhagic
and mucous membranes
autosomal dominant


Telangiectasia,




Osler-Weber-Rendu




Syndrome




Hypercholesterolemia,
elevation of serum
Low-density lipoprotein


familial
cholesterol bound to low
receptor (LDLR),



density lipoprotein,
apolipoprotein B (APOB),



accumulation in skin and
autosomal dominant



arteriosclerosis



Xeroderma
skin defect and melanoma
DNA repair defect,


pigmentosa
due to UV exposure
autosomal recessive


Male pattern
Disturbed conversion
5-α-reductase


baldness
of testosterone into




dihydrotestosterone




in the skin








Genetic liver diseases









Amino acid
Disruptions in the multistep
FAH, TAT, HPD,


metabolism
process that breaks down the
autosomal recessive


disorders
amino acid tyrosine and




phenylalanine



Beta-thalassemia
Shortage of mature red blood
HBB, autosomal


intermedia
cells
recessive


Crigler-Najjar
Deficiency in glucuronidation
UGT1A1, autosomal


syndrome
in which bilirubin gets
recessive



dissolvable in water



Fatty acid
Deficiency in processing of
HADHA, ACADVL


oxidation
long-chain fatty acids and
autosomal recessive


disorders
very long-chain fatty acids




resulting in lethargy and




hypoglycemia



Fructose
Impaired gluconeogenesis
FBP1, ALDOB,


metabolism
causing hypoglycemia
autosomal recessive


disorders




Galactosemia
Deficiency in processing
GALT, GALK1, GALE,



galactose
autosomal recessive


Glycogen
Disturbed breackdown
G6PC, SLC37A4,


storage diseases
of glucose 6-phosphate
AGL, GBE1,



and glycogen leads to
autosomal recessive



accumulation of glycogen as




well as abnormal glycogen




molecules causing cell damage



Heme
Decrease of uroporphyrinogen
UROD autosomal


biosynthesis
decarboxylase resulting in
dominant, ALAS2


disorder
accumulation of compounds
X-linked dominant,



called porphyrins
ALAD autosomal



causing toxic
recessive



levels in liver



Lipid metabolism
Shortage of functional protein,
NPC1, NPC2


(transport)
which prevents movement
of autosomal recessive,


disorders
cholesterol and other lipids,
LDLR, autosomal



leading to their accumulation
dominant



in cells



Metal
Disorders in the storage and
ATP7B, HAMP, HFE,


metabolism
transport of iron and copper
HFE2, autosomal


disorders
resulting in accumulation in
recessive



tissues and organs



Organic acid
Disrupted break down of several
BCKDHA, BCKDHB,


disorders
protein building blocks (amino
and DBT, PCCA and


(Acidurias/
acids), certain lipids, and
PCCB, MUT, MMAA,


Acidemias)
cholesterol
MMAB, MMADHC,




MCEE, IVD, MCCC1




or MCCC2, autosomal




recessive


Primary
Disrupted breakdown of
AGXT, GRHPR,


hyperoxaluria
glyoxylate leading to renal
autosomal recessive


type 1
damage



Progressive
Buildup of bile acids in
ATP8B1, autosomal


familial
liver cells causing
recessive


intrahepatic
liver damage



cholestasis




Thrombocyte
Lack of enzyme activity
ADAMTS13,


activity disorder
disrupts the usual balance
autosomal recessive



between bleeding




and clotting



Urea cycle
Disorder of the urea cycle
OTC (X-linked


disorders
which causes a form of
disorder), CPS1, ASS1



hyperammonemia
and SLC25A13, ASL,




autosomal recessive









The protein encoded by the polyribonucleotide according to the present invention can also have the potential to induce an immunogenic reaction acting, e.g., as an antigen. Thus, polyribonucleotides according to the invention lend themselves to applications in supplemental or medical purposes including vaccination.


Suitable nucleotide sequences of the genetic elements described above such as UTRs, regulatory sequences and binding sites are nucleotide sequences that enhance the action and/or duration of action of the mRNA molecule. Such nucleotide sequences as well as coding regions and/or coding sequences as described above can be naturally occurring, mutagenized, or synthesized nucleotide sequences or a combination thereof and can be identified and adapted by routine experiments known to the person skilled in the art.


The present invention further relates to a bacterial host cell comprising the DNA plasmid as described above. The DNA plasmid according to the present invention can be stored and/or amplified in a bacterial host cell that is competent for transformation. Competent host cells are cells that have the ability to take up free, extracellular genetic material such as a DNA plasmid independent of its sequence. Naturally competent bacteria have the ability to bind extracellular genetic material using for example receptor proteins or protein complexes. Moreover, said cells have the ability to transfer genetic material from the extracellular space into the cell using for example peptides, proteins or protein complexes such as a DNA translocase. A wide range of bacterial cells known to the person skilled in the art is naturally capable of taking up exogenous DNA from the environment and can thus act as a bacterial host cell according to the present invention. Furthermore, competent bacterial host cells can be obtained from naturally non-competent bacterial cells using for example electroporation or chemicals such as a treatment with calcium ions accompanied with the exposure to elevated temperature. Upon uptake, the DNA plasmid is preferably neither degraded nor integrated in the bacterial host cell's genomic information.


Bacterial host cells include Escherichia coli (E. coli) cells which are well-known to the person skilled in the art. In E. coli cells, integration of extracellular genetic material into the host cell's genetic material can take place in a RecA-dependent process, i.e. a process that is dependent on the RecA protein. This protein is essential for repair and maintenance of DNA and involved in homologous recombination. Hence, in some embodiments the bacterial host cell according to the present invention is an E. coli cell, preferably an E. coli recA cell. This might be advantageous to avoid integration of the genetic information comprised in the DNA plasmid according to the present invention into the E. coli cell's original genetic material.


The present invention further relates to the use of a nucleotide sequence which encodes a modified poly(A) tail as defined above for preparing a DNA plasmid showing reduced recombination during amplification in a bacterial host cell, wherein said nucleotide sequence is located downstream of a nucleotide sequence which encodes an mRNA molecule.


The DNA plasmid according of the present invention can be prepared by methods known to the person skilled in the art. Preferably, an MCS comprised in the DNA plasmid is cut, using a site specific restriction enzyme and the generated DNA plasmid ends are ligated to the ends of the DNA sequence. Thus, the DNA sequence according to the present invention is preferably terminated at both sites by said MCS.


As regards the nucleotide sequence which encodes a modified poly(A) tail, the nucleotide sequence which encodes an mRNA molecule, the DNA plasmid, and the reduction of recombination during amplification of said DNA plasmid in a bacterial host cell the same applies as it has been described above in connection with the DNA plasmid and the DNA sequence according to the invention. Moreover, also the other features of such a DNA plasmid and DNA sequence can be as described above.


The present invention further relates to the use of a nucleotide sequence which encodes a modified poly(A) tail as defined above for reducing recombination during amplification of a DNA plasmid in a bacterial host cell. In other words, a nucleotide sequence encoding a modified poly(A) tail as defined above can be used to reduce recombination during amplification of a DNA plasmid containing this nucleotide sequence in a bacterial cell compared to the same DNA plasmid but with a sequence encoding a conventional poly(A) tail, in particular in comparison to the same DNA plasmid in which the nucleotide sequence encoding a poly(A) tail does not contain any S elements. The DNA plasmid is preferably a plasmid which contains a nucleotide sequence encoding an mRNA molecule, wherein the nucleotide sequence encoding the modified poly(A) tail is placed downstream of the nucleotide sequence encoding the mRNA molecule. It is particularly advantageous to reduce recombination of the plasmid by replacing a nucleotide sequence encoding a conventional poly(A) tail by a nucleotide sequence encoding a modified poly(A) tail as defined above.


As regards the nucleotide sequence which encodes a modified poly(A) tail, the nucleotide sequence which encodes an mRNA molecule, the DNA plasmid, and the reduction of recombination during amplification of said DNA plasmid in a bacterial host cell the same applies as it has been described above in connection with the DNA plasmid according to the invention. Moreover, also the other features of such a DNA plasmid can be as described above.


The present invention further relates to a method for reducing recombination of a DNA plasmid comprising a DNA sequence which encodes an mRNA molecule and, located downstream thereof, a poly(A) tail, during amplification in a bacterial host cell, wherein said reduction is achieved by replacing the part of the DNA sequence which encodes the poly(A) tail by a nucleotide sequence which encodes a modified poly(A) tail as defined above.


Replacing a part of the DNA sequence can be performed for example using an MCS flanking said part of the DNA sequence and the nucleotide sequence that is used for replacement. Hence, the DNA plasmid can be cut using restriction enzymes at said MCS and the obtained fragments investigated using for example capillary or gel electrophoresis. Using said MCS, the part of the DNA plasmid not containing the part of the DNA sequence which encodes the poly(A) tail and the nucleotide sequence which encodes a modified poly(A) tail can then be ligated.


As regards the nucleotide sequence which encodes a modified poly(A) tail, the nucleotide sequence which encodes an mRNA molecule, the DNA plasmid, the bacterial host cell, and the reduction of recombination during amplification of said DNA plasmid in a bacterial host cell the same applies as it has been described above in connection with the DNA plasmid according to the invention. Moreover, also the other features of such a DNA plasmid can be as described above.


The present invention further relates to a method of producing a polyribonucleotide comprising a sequence encoding an amino acid sequence and a modified poly(A) tail as encoded by the nucleotide sequence as defined above, said method comprising the step of producing said polyribonucleotide by in vitro transcription from a DNA plasmid according to the invention.


Herein, the term “polyribonucleotide” refers to a single-stranded RNA molecule. The polyribonucleotide according to the present invention comprises the mRNA molecule encoded by the first nucleotide sequence and the modified poly(A) tail encoded by the second nucleotide sequence. In other words, the polyribonucleotide comprises a 5′ UTR, a coding sequence, optionally a 3′ UTR, and a modified poly(A) tail. In a particularly preferred embodiment, the polyribonucleotide comprises a 5′ UTR with at least one RBS and/or IRES, a coding sequence with optimized codons, optionally a 3′ UTR with at least one regulatory sequence, and a modified poly(A) tail. Optionally, the polyribonucleotide comprises additional nucleotides other than A nucleotides following the modified poly(A) tail, preferably one G nucleotide. This can prolong the lifetime and thus the duration of action of the produced polyribonucleotide when it is brought into a cell for example.


As regards the 5′ UTR, the coding sequence, the 3′ UTR, and the modified poly(A) tail the same applies as it has been described above. Moreover, also the other features of such genetic elements can be as described above.


The polyribonucleotide according to the present invention can be produced by any method known in the art. Preferably, the polyribonucleotide is transcribed in vitro using the DNA sequence according to the present invention as a template. In vitro transcription requires a purified linear DNA template containing a promoter, ribonucleotide triphosphates, a buffer system, and an appropriate RNA polymerase such as a T7 RNA polymerase. Hence, the DNA plasmid according to the invention is generally obtained by cell lysis and purified. The purified DNA plasmid is then cut, for example using a site-specific restriction enzyme, i) before the promoter preceding the first nucleotide sequence and ii) directly after the second nucleotide sequence encoding the modified poly(A) tail or, in case of additional nucleotides not being T nucleotides, directly after said additional nucleotides, preferably after one C nucleotide. The obtained linear template sequence is then used for the in vitro transcription of the polyribonucleotide according to the present invention in the presence of A, C, G, and U nucleotides using standard lab protocols. As the template is cut at the positions i) and ii), the template for transcription ends exactly with the desired modified poly(A) tail or the desired additional nucleotide that is not a T nucleotide. Hence, RNA polymerases can be used that do have or that do not have the ability to terminate transcription at a given position as for example, thus allowing run-off transcription. Thus, cutting the DNA plasmid at position i) and ii) and using the generated nucleotide sequence as a template for transcription, preferably for in vitro transcription, ensures the synthesis of polyribonucleotides according to the present invention comprising an mRNA molecule and a modified poly(A) tail of defined length. Quantity and quality of the in vitro transcribed polyribonucleotides can be measured for example by spectrophotometry, capillary electrophoresis and/or sequencing.


In a preferred embodiment, the produced polyribonucleotide is further modified by enzymatically adding a 5′ cap such as a C1-m7G cap or an m7GpppG cap. This might be advantageous for adjusting and/or extending the duration of action of the polyribonucleotide in a cell.


In some embodiments of the present invention the polyribonucleotide is produced by in vitro transcription in the presence of unmodified and/or modified nucleotides. In other words, a polyribonucleotide as described above can be synthesized by in vitro transcription using at least a DNA sequence comprising a nucleotide sequence encoding an mRNA molecule and a nucleotide sequence encoding a modified poly(A) tail according to the present invention as a template using unmodified and/or modified nucleotides.


The term “unmodified nucleotide” used herein refers to A, C, G, T, and U nucleotides as described above. Particularly, in case of the in vitro transcription of a polyribonucleotide said term refers to A, C, G, and U nucleotides. The latter four are referred to herein as the four nucleotide types comprised in a polyribonucleotide.


The term “modified nucleotide” used herein refers to any naturally occurring or chemically synthesized isomers of A, C, G, T, and U nucleotides as well as to any naturally occurring or chemically synthesized 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 five prime cap of polyribonucleotides comprising a sequence which encodes a protein. 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-d imethyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, N6-hydroxynorvalylcarbamoyladenosine, 1,2-O-dimethyladenosine, N6,2-O-dimethyladenosine, 2-O-methyladenosine, N6,N6,0-2-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-2-0-methylcytidine, 5-formyl-2-0-methylcytidine, 5,2-O-dimethylcytidine, 2-O-methylcytidine, N4,2-0-dimethylcytidine, N4,N4,2-0-trimethylcytidine, isocytidine, pseudocytidine, pseudoisocytidine, 2-thio-cytidine, 2″-methyl-2″-deoxycytidine, 2″-amino-2″-deoxycytidine, 2″-fluoro-2″-deoxycytidine, 5-iodocytidine, 5-bromocytidine and 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-I-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-1-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, N2,7,2-0-trimethylguanosine, N2,2-0-dimethylguanosine, 1,2-0-dimethylguanosine, 2-0-methylguanosine, N2,N2,2-0-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-am inocarboxypropyldemethylwyosine, 7-aminocarboxypropylwyosine, 7-am inocarboxypropylwyosine methylester, 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-d methyluridine, 5-carboxymethylaminomethyl-2-O-methyluridine, 5-carbamoylhydroxymethyluridine, 5-carbamoylmethyl-2-0-methyluridine, 5-carbamoylmethyl-2-thiouridine, 5-methoxycarbonylmethyl-2-O-methyluridine, 5-(isopentenylaminomethyl)-2-O-methyluridine, 5,2-0-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, 2-0-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, and 2″-fluoro-2″-deoxyuridine; inosine, 1-methylinosine, 1,2-0-dimethylinosine and 2-0-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-1-methyl-pseudouridine, 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine; 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. 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.


In case a polyribonucleotide according to the present invention is produced by in vitro transcription in the presence of four nucleotide types, i.e. A, C, G, and U nucleotides, the total number of modified nucleotide types can be 0, 1, 2, 3, or 4. In other words, 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 polyribonucleotide according to the present invention 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%.


Preferably, the polyribonucleotide according the invention is characterized in that 5 to 30% of the U nucleotides and 5 to 30% of the C nucleotides are modified. Said modified U nucleotides are preferably 5-ioduridine and said modified C nucleotides are preferably 5-iodcytidine.


More preferably, the polyribonucleotide according the invention is characterized in that 7.5 to 25% of the U nucleotides and 7.5 to 25% of the C nucleotides are modified. Said modified U nucleotides are preferably 5-ioduridine and said modified C nucleotides are preferably 5-iodcytidine.


The present invention further relates to a polyribonucleotide obtainable by any of the methods described above.


As described above, the polyribonucleotide according to the present invention comprises a 5′ UTR, a coding sequence, optionally a 3′ UTR, and a modified poly(A) tail as encoded by the first and the second nucleotide sequence. Preferably, the polyribonucleotide comprises a 5′ UTR with at least one RBS and/or IRES, a coding sequence with optimized codons, optionally a 3′ UTR with at least one regulatory sequence, and a modified poly(A) tail. Furthermore, the polyribonucleotide can optionally comprise an additional nucleotide other than A nucleotides following the modified poly(A) tail, preferably one G nucleotide.


As regards the polyribonucleotide and the methods for obtaining said polyribonucleotide the same applies as described above in connection with the nucleotide sequence which encodes an mRNA molecule and the nucleotide sequence which encodes a modified poly(A) tail, the mRNA molecule and the modified poly(A) tail as well as methods of producing a polyribonucleotide comprising a sequence encoding an amino acid sequence and a modified poly(A) tail according to the present invention. Moreover, also the other features of such a polyribonucleotide can be as described above.


In some embodiments the polyribonucleotide as described above comprises one or more types of modified nucleotides. This might be advantageous for reducing immunogenicity in case of transfecting a cell with said polyribonucleotide. Furthermore, it might be advantageous for enhancing the amount of amino acid sequence that can be obtained by said polyribonucleotide in a cell.


As regards the polyribonucleotide and the methods for obtaining said polyribonucleotide the same applies as described above in connection with the nucleotide sequence which encodes an mRNA molecule and the nucleotide sequence which encodes a modified poly(A) tail, the mRNA molecule and the modified poly(A) tail as well as methods of producing a polyribonucleotide comprising a sequence encoding an amino acid sequence and a modified poly(A) tail using modified nucleotides according to the present invention. Moreover, also the other features of such a polyribonucleotide can be as described above.


The present invention further relates to a pharmaceutical composition containing a polyribonucleotide as described above together with a pharmaceutically acceptable carrier. The polyribonucleotide is preferably included in an effective amount, i.e. an amount sufficient to induce a detectable therapeutic response in the subject to which the pharmaceutical composition is to be administered. The polyribonucleotide or the pharmaceutical composition of the invention can be in sterile aqueous or non-aqueous solutions, suspensions, and emulsions as well as creams and suppositories, but can also have the form of powders, tablets or aerosols.


The term “pharmaceutically acceptable carrier” used herein refers to 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 can also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases, nanosystems or liposomes, and the like.


The pharmaceutical composition of the present invention can be administered via a large range of classes of forms of administration known to the skilled person, such as needle injection, the use of inhalators, creams, foams, gels, lotions and ointments. Dose and duration of action depend on the function which said polyribonucleotide is to fulfill and have to be deliberately adjusted in each case. The duration of action will be as long as possible for example, if said polyribonucleotide is used for the chronic therapy of a disease due to a deficient gene, while with other indications it can be adjusted to a specific time window. Furthermore, systemic administration of said one or more polyribonucleotides as described above is possible.


As regards the polyribonucleotide the same applies as described above in connection with the nucleotide sequence which encodes an mRNA molecule and the nucleotide sequence which encodes a modified poly(A) tail as well as methods of producing a polyribonucleotide comprising a sequence encoding an amino acid sequence and a modified poly(A) tail and polyribonucleotides according to the present invention. Moreover, also the other features of such a polyribonucleotide can be as described above.






FIG. 1: Quantification of poly(A) tail recombination based on the percentage of recombinant clones for poly(A)120, poly(A)3×40_6 and poly(A)2×60_6 constructs.



FIG. 2: Quantification of poly(A) tail recombination based on the percentage of recombinant clones using different one nucleotide spacer.



FIG. 3: Determination of luciferase protein activity and mRNA decay kinetics of different poly(A) constructs 24 h post-transfection (250 ng/well) in A549 cells. (a) Luciferase activity in protein lysates from A549 cells transfected with different poly(A) constructs. (b) Luciferase mRNA quantification in A549 cells. (c) mRNA productivity was calculated by dividing the luciferase luminescence values (RLU; a) by the mRNA amounts (real time qPCR data; b) and normalizing these ratios to those observed with poly(A)120 construct. Statistical significance was assessed by two-way ANOVA test with p-values: *p<0.5, **p<0.01, ***p<0.001, ****p<0.0001, n=6.



FIG. 4: Quantification of secreted human erythropoietin (hEPO) protein levels as measured via ELISA in supernatants from HEK293 cells transfected either with poly(A)120 or poly(A)2×60_6 constructs 24 h post-transfection (250 ng/well). Values represent mean±standard deviation of three replicates. Statistical significance was assessed by two-way ANOVA test with p-values: **p<0.01, ***p<0.001, n=3.



FIG. 5: Determination of luciferase protein activity and mRNA quantification of different poly(A) tail constructs 24 h post-transfection (250 ng/well) in A549 cells. (a) Luciferase activity in protein lysates from A549 cells transfected with different poly(A) tail constructs. (b) Luciferase mRNA quantification in A549 cells transfected with different poly(A) tail constructs. (c) Luciferase mRNA productivity was calculated by dividing the luciferase luminescence values (RLU; a) by the mRNA amounts (real time qPCR data; b) and normalizing these ratios to those observed with the poly(A)120 construct. Statistical significance was assessed by two-way ANOVA test with p-values: *p<0.5, ****p<0.0001, n=6.





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

Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting.


Abbreviations used herein and their respective descriptions are listed in Table 2.










TABLE 2





Abbreviation
Description







A
Nucleotide with an adenine residue


C
Nucleotide with an cytosine residue


° C.
Degree Celsius


ELISA
Enzyme-linked Immunosorbent Assay


FA
Fragment Analyzer


G
Nucleotide with an guanine residue


h
Hour(s)


hIL-6
Human interleukin 6


min
Minutes


mRNA
Messenger ribonucleic acid


n
Total number of clones with a particular poly(A)



tail sequence


n/a
Not applicable


ND
NanoDrop


nm
Nanometer


nt
Nucleotide(s)


poly(A)120
Nucleotide sequence encoding a poly(A) tail without



S element consisting of 120 A nucleotides


poly(A)3×40_6
Nucleotide sequence encoding a poly(A) tail



consisting of three A elements each consisting of 40 A



nucleotides with any two A elements separated by



a 6 nt long S element as defined in Table 6


poly(A)2×60_6
Nucleotide sequenceen coding a poly(A) tail consisting



of two A elements consisting of 60 A nucleotides



each and separated by a 6 nt long S element



as defined in Table 6


poly(A)2×60_C
Nucleotide sequence encoding a poly(A) tail



consisting of two A elements consisting of 60



A nucleotides each and separated by a C



nucleotide


poly(A)2×60_G
Nucleotide sequence encoding a poly(A) tail



consisting of two A elements, consisting of 60 A



nucleotides each and separated by a G nucleotide


poly(A)2×60_T
Nucleotide sequence encoding a poly(A) tail



consisting of two A elements consisting of 60



A nucleotides each and separated by a T nucleotide


qPCR
Quantitative real-time polymerase chain reaction


RLU
Relative light unit


T
Nucleotide with an thymine residue


U
Nucleotide with an uracile residue


%
Percent









Material and Methods


Materials, Devices, Software, and Test System Used


Materials are listed in Table 3.











TABLE 3





Material
Supplier
Cat#







pUC57-Kanamycin vector
GenScript
n/a


Oligonucleotides
IDT
n/a


Annealing buffer
Ethris GmbH
n/a


Tris HCl
Roth
9090.1


NaCl
Roth
9265.1


EDTA
Roth
8040.1


Phusion High-fidelity PCR master mix
Thermo Fisher Scientific
F531S


MgCl2
Roth
KK36.2


DMSO
Sigma Aldrich
67-68-5


NucleoSpin Gel and PCR Clean-Up
Macherey-Nagel
740609.250


Mini prep kit
Macherey-Nagel
740588.250


Maxi Prep kit
Macherey-Nagel
740414.10


Agarose
Sigma Aldrich
A9539


BgIII
Thermo Fisher Scientific
FD0083


Nhel
Thermo Fisher Scientific
FD0973


BstBl
Thermo Fisher Scientific
FD0124


Chloroform
Sigma Aldrich
288306


Firefly luciferase coding region
Promega
n/a


d2EGFP coding region
Clontech
n/a


hEPO coding region
Pubmed
n/a


Ethanol
Roth
5054.1


T7 RNA polymerase
Thermo Fisher Scientific
EP0111


Transcription buffer II
Ethris GmbH
n/a


RiboLock Rnase inhibitor
Thermo Fisher Scientific
EO0381


Inorganic pyrophosphatase 1
Thermo Fisher Scientific
EF0221


Ribonucleotides
Jena Biosciences
NU-1010-NU-1013


DNase I
Thermo Fisher Scientific
EN0525


Ammonium acetate
Applichem
131114.1210


aqua ad injectabilia
B. Braun
3703444


Vaccinia Virus Capping Enzyme
NEB
M2080S


1x capping buffer
NEB
M2080


GTP
Jena Biosciences
NU-1012


S-Methyladenosine
NEB
B9003S


mRNA Cap 2'-o-Methyltransferase
NEB
M0366S


Minimum Essential Media
GlutaMAX Gibco Life
11095-080



Technologies



Glutamax
Gibco/Life Technologies
35050061


Fetal bovine serum
Gibco/Life Technologies
10500064


Penicillin/streptomycin
Gibco/Life Technologies
15140122


Lipofectamine ® 2000
Thermo Fisher Scientific
11668027


PBS
Gibco/Life Technologies
10010023


TritonX-100
Sigma Aldrich
9002-93-1


BioRad protein assay dye
Bio-Rad
5000006


reagent concentrate




Bovine serum albumin
Sigma Aldrich
A2058


Propidium iodide
Sigma Aldrich
11348639001


TrypLE
Gibco/Life Technologies
12604-013


Single Shot Cell Lysis kit
Bio-Rad
1725080


iScript Select cDNA Synthesis kit
Bio-Rad
1708896


Universal Probe Library #29
Roche
4687612001









Devices are listed in Table 4.












TABLE 4







Device
Supplier









Roche Light Cycler 96
Roche Diagnostics



NanoDrop2000C
Thermo Fisher Scientific



Fragment Analyzer
Advanced Analytical



Humidified 5% CO2 incubator
Sanyo



InfiniteR 200 PRO
Tecan



Attune Acoustic Focusing Cytometer
Life Techologies



Gene Pulser II
Biorad










Software is listed in Table 5.












TABLE 5







Software
Provider









GraphPad Prism software (version 6)
GraphPad Software Inc.



Attune Cytometric Software (version 2.1)
Life Technologies



FlowJo (version 10)
FlowJo



LightCycler ® 96 (version 1.1)
Roche



PROSize 3.0
Advanced analytical










The test system is listed in Table 6.











TABLE 6







Test System
Species
Strain





Cell line

E. coli


E. coli





DH10B strain


Cell line
Human
A549




(ACC-107)


Cell line
Human
HEK293




(ACC-305)





Thawed
Used passage



passage no.
no.
Supplier










4-7
Up to 15
DSMZ


4-7
Up to 15
DSMZ









Plasmid Preparation


Synthetic poly(A) tail sequences were introduced to the pUC57-Kanamycin vector backbone either as annealed complementary oligonucleotides or fragments created by PCR-based strategy. A specific set of complementary oligonucleotides was designed for sequences comprising poly(A)2×60_6 constructs and poly(A)3×40_6 constructs and annealed. The synthetic poly(A) constructs of poly(A)120, Poly(A)2×60_C, poly(A)2×60_G, and poly(A)2X60_T were created by PCR.


The two sets of complementary oligonucleotides were annealed in the following way: 100 μM of each oligonucleotide were mixed with 40 μl annealing buffer and incubated for 5 min at 95° C. (10 mM Tris HCl, 50 mM NaCl, 1 mM EDTA, pH 7.5). After the reaction, the mixture was let to cool down to room temperature before proceeding with restriction digestion (BgIII-BstBI).


For the high performance of PCR reaction, Phusion High-fidelity PCR master mix was used. In addition to the master mix which contains 2× Phusion DNA Polymerase, nucleotides and optimized reaction buffer including MgCl2, 0.5 μM of forward and reverse primer, 3% DMSO and 100 ng of template DNA were added to the reaction. The total volume of 25 μl per reaction was initially denatured at 98° C. for 30 sec, following by 30 cycles at 98° C. for 10 sec, annealing at 72° C. for 30 sec and extension at 72° C. for 30 sec/kb. The final extension was performed at 72° C. for 10 min. The size of the PCR product was confirmed on 1% agarose gel and the desired band was purified using NecleoSpin Gel and PCR clean up kit. Purified PCR product was digested with Nhel-BstBI and stored at −20° C. till further use. After restriction enzyme digestion of annealed oligonucleotides (BgIII-BstBI) and PCR fragments (Nhel-BstBI), the poly(A) tail constructs were cloned into accordingly digested pUC57-Kanamycin vectors comprising the coding region of choice (firefly luciferase, hEPO, d2EGFP).


A list of segmented poly(A) sequences and corresponding cloning strategy with PCR primer sets or oligonucleotides is shown in Table 7.












TABLE 7





Construct
Strategy
PCR primer forward/Oligo I
PCR primer reverse/Oligo II







A120
PCR
GTGACTGCTAGCTAATACGACTCACTAT
AGTCACTTCGAATTTTTTTTTTTTTTTTT




AGGGAG (SEQ ID No: 1)
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTAGATCTTTACACGGC





GATCTTGCCGCCCTTC (SEQ ID No: 2)





2x60_6
Oligonucleotides
GTGACTAGATCTAAAAAAAAAAAAAAAAA
AGTCACTTCGAATTTTTTTTTTTTTTTTT




AAAAAAAAAAAAAAAAAAAAAAAAAAAAA
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT




AAAAAAAAAAAAAAATGCATAAAAAAAAA
TTTTTTTTTTTTTATGCATTTTTTTTTTTT




AAAAAAAAAAAAAAAAAAAAAAAAAAAAA
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT




AAAAAAAAAAAAAAAAAAAAAATTCGAAG
TTTTTTTTTTTTTTTTTTTAGATCTAGTC




TGACT (SEQ ID No: 3)
AC (SEQ ID No: 4)





3x40_6
Oligonucleotides
GTGACTAGATCTAAAAAAAAAAAAAAAAA
AGTCACTTCGAATTTTTTTTTTTTTTTTT




AAAAAAAAAAAAAAAAAAAAAAAATGCAT
TTTTTTTTTTTTTTTTTTTTTTTGATATCT




AAAAAAAAAAAAAAAAAAAAAAAAAAAAA
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT




AAAAAAAAAAAGATATCAAAAAAAAAAAA
TTTTTTTTTATGCATTTTTTTTTTTTTTTT




AAAAAAAAAAAAAAAAAAAAAAAAAAAAT
TTTTTTTTTTTTTTTTTTTTTTTTTAGATC




TCGAAGTGACT (SEQ ID No: 5)
TAGTCAC (SEQ ID No: 6)





2x60_C
PCR
GTGACTGCTAGCTAATACGACTCACTAT
AGTCACTTCGAATTTTTTTTTTTTTTTTT




AGGGAG (SEQ ID No: 1)
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTGTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTAGATCTTTACACGG





CGATCTTGCCGCCCTTC (SEQ ID No: 7)





2x60_G
PCR
GTGACTGCTAGCTAATACGACTCACTAT
AGTCACTTCGAATTTTTTTTTTTTTTTTT




AGGGAG (SEQ ID No: 1)
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTCTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTAGATCTTTACACGG





CGATCTTGCCGCCCTTC (SEQ ID No: 8)





2x60_T
PCR
GTGACTGCTAGCTAATACGACTCACTAT
AGTCACTTCGAATTTTTTTTTTTTTTTTT




AGGGAG (SEQ ID No: 1)
TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTATTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTTTTTTTTTTTTTTTTT





TTTTTTTTTTTTTTAGATCTTTACACGG





CGATCTTGCCGCCCTTC (SEQ ID No: 9)









Cloning into E. coli


The ligations were purified using chloroform-ethanol precipitation and electroporated into DH10B strain of E. coli. For electroporation, Gene Pulser II from Biorad was used. Electroporation conditions followed were: 25 μF, 200 ohms, 1.8 kV. Post electroporation, the bacteria were grown in 2 mL of LB-Medium at 30° C. for 1.5 hours. Subsequently, the culture was centrifuged at 5000 rpm for 10 min at room temperature. Supernatant was discarded and the pellet was resuspended in 200 μL of fresh LB-Medium. From this, 100 μL were plated in LB-Agar plates containing the appropriate antibiotic (Kanamycin at a final concentration of 50 μg/mL or Ampicillin at a final concentration of 100 μg/mL). The plates were incubated overnight at 30° C.


Plasmid Preparation Using Mini-Prep Kit


Clones were inoculated in LB-Medium containing the appropriate antibiotic (Kanamycin at a final concentration of 50 μg/mL or ampicillin at a final concentration of 100 μg/mL) and grown at 30° C. overnight in a bacterial shaker (250 rpm). Subsequently plasmids were isolated from the overnight cultures using Mini-Prep kit. Plasmids were tested for insert using restriction digestion and confirmed via sequencing. For the correct clones, glycerol stocks were prepared by adding 200 μL autoclaved glycerol to 800 μL of overnight bacterial culture. Glycerol stocks were stored at −80° C.


Plasmid Preparation Using Maxi-Prep Kit


Plasmid for RNA production was prepared using the Maxi-Prep kit. Glycerol stock from the desired clone(s) was inoculated in 5 mL of LB-Medium containing appropriate antibiotics (Kanamycin at final concentration of 50 μg/mL or Ampicillin at a final concentration of 100 μg/mL) and the culture was grown overnight at 30° C. in a bacterial shaker (250 rpm). 3 mL from this starter culture were used to inoculate 300 mL of LB-Medium containing appropriate antibiotic (Kanamycin at a final concentration of 50 μg/mL or Ampicillin at a final concentration of 100 μg/mL) which was subsequently incubated overnight at 30° C. in a bacterial shaker (250 rpm).Overnight culture was centrifuged at 5000 rpm, 4° C. for 30 min. Supernatant was discarded and the bacterial pellet was used to isolate the plasmid.


Generation of mRNA


To generate in vitro transcribed mRNA, plasmids were linearized by BstBI digestion and purified by chloroform extraction and ethanol precipitation. Purified linear plasmids were used as a template for in vitro transcription. Plasmid templates (0.5 μg/μl) were subjected to in vitro transcription using 3 U/μl T7 RNA polymerase, transcription buffer II, 1 U/μl RiboLock Rnase inhibitor, 0.015 U/μl inorganic pyrophosphatase 1 with a defined choice of ribonucleotides. The complete IVT-mix was incubated at 37° C. for 2 h. Afterwards, 0.01 U/μl DNase I was added for additional 45 min at 37° C. to remove the plasmid template. RNA was precipitated with ammonium acetate at a final concentration of 2.5 mM, followed by two washing steps with 70% ethanol. The pellet was re-suspended in aqua ad injectabilia. A C1-m7G cap structure was added enzymatically by 0.5 mM Vaccinia Virus Capping Enzyme to the 5′ end of the previously denaturated transcript (1 mg/ml) at 80° C. for 5 min. The capping reaction mix also contained 1× capping buffer, 0.5 mM GTP, 0.2 mM S-Methyladenosine, 2.5 U/μl Mrna Cap 2′-o-Methyltransferase and 1 U/μl RiboLock Rnase Inhibitor. The capping mixture was incubated for 60 min at 37° C., followed by RNA precipitation with ammonium acetate at a final concentration of 2.5 mM and two washing steps with 70% ethanol. The pellet was re-suspended in aqua ad injectabilia.


RNA quality and concentration were measured spectrophotometrically on a NanoDrop2000C. Its correct size and purity were determined via automated capillary electrophoresis.


Cell Culture


A549 (ACC-107) and HEK293 (ACC-305) cells were purchased from DSMZ. All cells were cultivated in Minimum Essential Media (MEM) with Glutamax. Media were supplemented with 10% heat-inactivated fetal bovine serum (FBS) and 1 penicillin/streptomycin. Cells were cultured in a humidified 5% CO2 incubator at 37° C.


In Vitro Transfection Both cell lines, A549 and HEK293, were transfected with 250 ng mRNA per well. A549 and HEK293 cells were seeded at the density of 2×104 cells/well and 4×104 cells/well, respectively, in a 96 well plate, for the purpose of firefly luciferase and hEPO ELISA assay. 24 hours post-seeding, cells were transfected using the commercial transfection reagent Lipofectamine® 2000. Complexes were prepared at a ratio of 2 μl Lipofectamine® 2000 per 1 μg mRNA.


The mRNA was diluted 1:20 in water, and Lipofectamine® 2000 1:10 separately in a serum-free MEM. mRNA was added to the Lipofectamine® 2000 solution followed by 20 min incubation time at RT. The concentration of the final mRNA/Lipofectamine® 2000 solution was 25 ng/μl, and a serial dilution 1:2 was performed. 10 μl of the complex solution was added to the cells and cells were incubated for 24 and 48 h, respectively. For every mRNA construct, replicates of three or six were prepared.


Flow Cytometry Analysis for d2EGFP


Cells were washed with PBS, detached with TrypLE, and re-suspended in flow cytometry buffer (PBS supplemented with 10% FBS). Shortly before measurement, cells were stained with propidium iodide for discrimination between live and dead cells (1 μg/mL). Live cells (>97%) were further gated to discriminate between d2EGFP-expressing cells and those that did not express. Analysis was performed on an Attune Acoustic Focusing Cytometer with Attune Cytometric Software (version 2.1) and FlowJo (version 10).


Firefly Luciferase Assay


For detection of firefly luciferase activity, the assay was performed 24 h post-transfection. At the appropriate time point, cells were washed with PBS, followed by addition of 100 μl of lysis buffer (25 mM Tris-HCl, 0.1% TritonX-100, pH 7.4). Cells were shaken for 20 min at room temperature. After lysation, 50 μl of the cell lysate was used to measure luciferase activity via photon luminescence emission for 5 s using InfiniteR 200 PRO. The protein amount in each sample was quantified in 5 μl of the cell lysate with BioRad protein assay, using bovine serum albumin as a standard. Luciferase values were normalized to the protein concentration.


RNA Isolation and Reverse Transcription


In order to determine the actual mRNA amount 24 h post-transfection, the cultured cells (A549, HEK293) were lysed and RNA was isolated according to the manufacturer's protocol using Single Shot Cell Lysis kit. From the lysates (1 μg of RNA), cDNA was synthesized using iScript Select cDNA Synthesis kit with oligo(dT) primers following the manufacturer's instructions. The synthesized cDNA was stored at −20° C.


Quantitative Real-Time Polymerase Chain Reaction (qPCR)


Real-time qPCR was performed with short hydrolysis probes for d2EGFP and luciferase targets (Universal Probe Library #37 and #29) on a Roche Light Cycler 96. The following primers for d2EGFP were used: 5′-cctgaagttcatctgcacca-3′ and 5′-ctcgtgaccaccctgacc-3′; and for the luciferase target: 5′-acgccgagtacttcgagatg-3′ and 5′-attcagcccatagcgcttc-3′. Absolute mRNA values were calculated by interpolation from the standard curve.


Statistical Analysis


Each experiment was performed with at least three technical replicates per sample. Results are shown as means±SD unless otherwise stated. Statistical analysis was performed using GraphPad Prism software (version 6). Data was tested for normal distribution using D'Agostino-Pearson omnibus normality test. Multiple comparisons were conducted by two-way ANOVA, followed by Sidak's test (pairwise comparison) or Dunett's test (many-to-one comparison). A p-value ≤0.05 was considered statistically significant.


Segmented Poly(A) Tails Drastically Reduce Bacterial Recombination


It was examined whether the use of segmented poly(A) tails affected the recombination of plasmids post transformation into E. coli. To test this, open reading frame sequences of different genes (luciferase, d2EGFP, hEPO) were combined with either of the three poly(A) constructs poly(A)120, poly(A)2×60_6, and poly(A)3×40_6 and cloned into a pUC57-Kanamycin vector. Post transformation into E. coli, clones were screened for insert and positive clones containing the desired insert were additionally screened for the length of the respective poly(A) tail. For each of the three poly(A) tail constructs, the poly(A) tail was digested with restriction enzymes and the digestions were resolved on Fragment Analyzer (capillary gel electrophoresis) to measure the size of the respective poly(A) tail. Recombination in the poly(A)-tail was observed for more than 50% of the clones containing the homologous poly(A) tail poly(A)120. By splitting the poly(A) tail into either poly(A)3×40_6 or poly(A)2×60_6, recombination in E. coli could be significantly reduced with most stable clones (<20% recombination) obtained with plasmids containing poly(A)2×60_6 (FIG. 1; Table 8). This trend was observed for all three tested open reading frame sequences indicating that this reduction in recombination is sequence independent. The most favorable effect was observed for poly(A)2×60_6 constructs with recombination 20% or less.












TABLE 8










Target genes












Poly(A)
Luc2
d2EGFP
EPO







Poly(A)2×60_6
56
10
15



Poly(A)3×40_6
10
10




Poly(A)120
11
10
16










Effect of a One Nucleotide Long Spacer within a Poly(A) Tail on Recombination


The effect of a one nucleotide long spacer in a poly(A)2×60 construct (C, T or G) on recombination in E. coli was examined by investigating clones comprising the open reading frame sequence of the firefly luciferase and the respective poly(A)2×60_C, poly(A)2×60_G, or poly(A)2×60_T construct. Interestingly, the constructs comprising G as a spacer in the poly(A) tail, did not recombine at all. A spacer with a single T recombined in 10% of cases, and the one with a C as a spacer nucleotide recombined in 50% of cases (FIG. 2), which, at first sight seems to be comparable to recombination efficiencies observed with A120 (FIG. 1) but is still significantly lower. This is evident from the following Table 9 which summarizes the recombination efficiencies observed for the constructs tested in FIGS. 1 and 2.














TABLE 9










Recombination



Poly(A)
Figure
n
rate









Poly(A)2×60_6
1
81
19%



Poly(A)3×40_6
1
20
35%



Poly(A)120
1
37
54%



Poly(A)2×60_G
2
16
 0%



Poly(A)2×60_T
2
10
10%



Poly(A)2×60_C
2
10
50%










Effect of a Six Nucleotide Long Spacer on mRNA and Protein Levels


Luciferase protein and mRNA decay was investigated in A549 cells at 24 h post-transfection with luciferase mRNA, containing either a poly(A)2×60_6, poly(A)3×40_6 or poly(A)120 construct. Use of the segmented poly(A)2×60_6 construct significantly increased protein levels post-transfection when compared to the poly(A)120 benchmark (FIG. 3). No significant differences were observed between the mRNA amounts for the different poly(A) format containing luciferase mRNAs across modifications.


Further, the effects of poly(A) segmentation on transcription and translation of a physiological target was tested using human erythropoietin (hEPO) as a prototype of secretory proteins and short mRNAs (0.9 kb). The codon optimized sequence encoding hEPO was cloned into a pUC57-Kanamycin vector upstream of either a poly(A)120 or a poly(A)2×60_6 construct. hEPO protein concentrations were determined via ELISA 24 h post-transfection (FIG. 4). A significant difference was observed between the two compared poly(A) tail constructs with the poly(A)2×60_6 construct resulting in significantly more protein compared to the standard poly(A)120 construct.


Effect of a One Nucleotide Long Spacer on mRNA and Protein Levels


The effect of a single spacer nucleotide within a poly(A)2×60-tail on protein expression and mRNA productivity was tested. Luciferase mRNA expression and protein activity was determined by transfecting A549 cells with mRNA constructs containing a single C, T, or G spacer nucleotide within the respective poly(A)2×60-tail. As a benchmark, the standard poly(A)120 construct was used. Between all three single nucleotide spacer constructs, there was no significant difference in protein expression, but all of them resulted in significantly more protein compared to the poly(A)120 construct (FIG. 5). Calculating mRNA productivity, the construct with a C as a spacer appears to have the highest productivity overall. mRNA productivity of poly(A)120 was significantly lower compared to that of any of the three tested segmented constructs poly(A)2×60_C, poly(A)2X60_G, poly(A)2×60_T.

Claims
  • 1. A DNA plasmid comprising a DNA sequence which contains (i) a first nucleotide sequence which encodes an mRNA molecule and, located downstream thereof,(ii) a second nucleotide sequence which encodes a modified poly(A) tail, wherein said second nucleotide sequence is characterized in that it consists of (a) at least two A elements each defined as a nucleotide sequence consisting of 55 to 65 T nucleotides, and(b) at least one S element each S element consisting of (b1) one nucleotide that is not a T nucleotide, or(b2) 2 to 10 nucleotides, preferably 6 nucleotides, wherein each of the two terminal nucleotides is not a T nucleotide;wherein the total number of A elements is one more than the total number of S elements, andwherein any two A elements are separated by one S element.
  • 2. The DNA plasmid of claim 1, wherein any one of said S elements consists of one C nucleotide or one A nucleotide, preferably of one C nucleotide.
  • 3. The DNA plasmid of claim 1 or 2, wherein the number of A elements is two, three or four.
  • 4. The DNA plasmid of any one of claims 1 to 3, wherein the number of A elements is four and wherein said nucleotide sequences of the four A elements together have an overall length of 240 nucleotides, preferably each A element having a length of 60 nucleotides.
  • 5. The DNA plasmid of any one of claims 1 to 3, wherein the number of A elements is two and wherein said nucleotide sequences of the two A elements together have an overall length of 120 nucleotides, preferably each A element having a length of 60 nucleotides.
  • 6. A bacterial host cell comprising the DNA plasmid of any one of claims 1 to 5.
  • 7. The bacterial host cell of claim 6, which is an E. coli cell, preferably an E. coli recA− cell.
  • 8. Use of a nucleotide sequence which encodes a modified poly(A) tail as defined in claim 1 (ii) and in claims 2 to 5 for preparing a DNA plasmid showing reduced recombination during amplification in a bacterial host cell, wherein said nucleotide sequence is located downstream of a nucleotide sequence which encodes an mRNA molecule.
  • 9. Use of a nucleotide sequence which encodes a modified poly(A) tail as defined in claim 1 (ii) and in claims 2 to 5 for reducing recombination during amplification of a DNA plasmid in a bacterial host cell.
  • 10. A method for reducing recombination of a DNA plasmid comprising a DNA sequence which encodes an mRNA molecule and, located downstream thereof, a poly(A) tail, during amplification in a bacterial host cell, wherein said reduction is achieved by replacing the part of the DNA sequence which encodes the poly(A) tail by a nucleotide sequence which encodes a modified poly(A) tail as defined in claim 1 (ii) and in claims 2 to 5.
  • 11. A method of producing a polyribonucleotide comprising a sequence encoding an amino acid sequence and a modified poly(A) tail as encoded by the nucleotide sequence as defined in claim 1 (ii) and in claims 2 to 5, said method comprising the step of producing said polyribonucleotide by in vitro transcription from a DNA plasmid of any one of claims 1 to 5.
  • 12. The method of claim 11, wherein said polyribonucleotide is produced by in vitro transcription in the presence of unmodified and/or modified nucleotides.
  • 13. A polyribonucleotide obtainable by the method of claim 11 or 12.
  • 14. The polyribonucleotide of claim 13, wherein the polyribonucleotide comprises one or more types of modified nucleotides.
  • 15. A pharmaceutical composition containing a polyribonucleotide of any one of claims 13 to 14 together with a pharmaceutically acceptable carrier.
Priority Claims (1)
Number Date Country Kind
18 19 9857.6 Oct 2018 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2019/077477 10/10/2019 WO 00