CN114717229A - Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA - Google Patents

Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA Download PDF

Info

Publication number
CN114717229A
CN114717229A CN202110008455.8A CN202110008455A CN114717229A CN 114717229 A CN114717229 A CN 114717229A CN 202110008455 A CN202110008455 A CN 202110008455A CN 114717229 A CN114717229 A CN 114717229A
Authority
CN
China
Prior art keywords
nucleic acid
acid molecule
utr
rna
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110008455.8A
Other languages
Chinese (zh)
Inventor
邝纬阳
林庭匡
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Massena Therapy Hong Kong Ltd
Original Assignee
Massena Therapy Hong Kong Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Massena Therapy Hong Kong Ltd filed Critical Massena Therapy Hong Kong Ltd
Priority to CN202110008455.8A priority Critical patent/CN114717229A/en
Priority to PCT/CN2021/130552 priority patent/WO2022148131A1/en
Publication of CN114717229A publication Critical patent/CN114717229A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1808Epidermal growth factor [EGF] urogastrone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43595Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from coelenteratae, e.g. medusae
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/485Epidermal growth factor [EGF] (urogastrone)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • C07K14/503Fibroblast growth factors [FGF] basic FGF [bFGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination

Abstract

The present invention relates to cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA. More specifically, the present invention provides a nucleic acid molecule comprising, from the 5 'end to the 3' end, a 5 '-cap structure, a human β -globin 5' untranslated region (5 '-UTR), at least one coding region, a human α -globin 3' untranslated region (3 '-UTR) and a 3' -poly a tail, said at least one coding region being operably linked to said 5 '-UTR and said 3' -UTR, and a method of in vitro transcription. The in vitro transcription methods and nucleic acid molecules of the invention not only enable maximal production of mRNA up to about 2.3mg/mL or even higher per hour, but also the resulting mRNA shows enhanced stability of gene expression and translation efficiency.

Description

Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA
Technical Field
The present disclosure relates generally to the field of molecular biology, and more particularly to cell-free and vector-free in vitro RNA transcription methods of mRNA and nucleic acid molecules.
Background
Messenger rna (mrna) is a relatively new therapeutic molecule with a wide range of potential clinical applications, including cancer treatment, vaccines and regenerative therapies. Advantages of mRNA-based drugs over recombinant protein-based drugs include: 1. the production is cost-effective; 2. longer therapeutic effect; 3. rapid synthesis and purification; 4. no endotoxin and infectious agents; 5. post-translational modifications are advantageous. mRNA is also a very beneficial alternative to gene therapy because it does not integrate the gene into the genome, does not require entry into the nucleus, and expression is directly controllable. Overall, mRNA-based therapies are safe and cost-effective.
Although mRNA-based therapies are promising, the stability of mRNA and the ability to deliver mRNA to cells is poor. Thus, there remains a need in the art for improved in vitro RNA transcription methods and more stable mrnas.
Disclosure of Invention
In one aspect, the present disclosure provides an RNA nucleic acid molecule comprising or consisting of, from 5 'end to 3' end: a 5 '-cap structure, a human β -globin 5' untranslated region (5 '-UTR), at least one coding region, a human α -globin 3' untranslated region (3 '-UTR), and a 3' -poly a tail, the at least one coding region operably linked to the 5 '-UTR and the 3' -UTR.
In one embodiment, the coding region encodes at least one polypeptide or protein of interest, optionally selected from a growth factor, an antigenic polypeptide or protein, an allergenic polypeptide or protein, a therapeutic polypeptide or protein, or a fragment, variant or derivative of the foregoing.
In one embodiment, the coding region further encodes at least one selected from the group consisting of: a signal peptide, a peptide tag or protein tag, a targeting signal or targeting sequence, and a peptide linker.
In one embodiment, the 5' -UTR comprises or consists of a sequence according to SEQ ID NO: 1, or an RNA sequence according to SEQ ID NO: 1, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity.
In one embodiment, the 3' -UTR comprises or consists of a sequence according to SEQ ID NO: 2, or an RNA sequence according to SEQ ID NO: 2, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity.
In one embodiment, the 5' -cap structure is a 5' -anti-reverse cap analog (5 ' -ARCA), preferably the 5' -ARCA is 7m G (3 ' -O-Me) pppG.
In one embodiment, the 3' -poly a tail comprises 10 to 200, 20 to 100, 40 to 80, or 50 to 70 adenine nucleotides.
In another aspect, the present disclosure provides a DNA nucleic acid molecule comprising or consisting of, from 5 'end to 3' end: a promoter, a human β -globin 5 '-untranslated region (5' -UTR), at least one coding region, a human α -globin 3 '-untranslated region (3' -UTR), and a transcription terminator, said at least one coding region operably linked to said 5 '-UTR and said 3' -UTR.
In one embodiment, the coding region encodes at least one polypeptide or protein of interest, optionally selected from a peptide growth factor, an antigenic polypeptide or protein, an allergenic polypeptide or protein, a therapeutic polypeptide or protein, or a fragment, variant or derivative of the foregoing.
In one embodiment, the coding region further encodes at least one selected from the group consisting of: a signal peptide, a peptide tag or protein tag, a targeting signal or targeting sequence, and a peptide linker.
In one embodiment, the promoter is selected from the group consisting of the T3, T7, Sny5, or SP6 promoters.
In one embodiment, the promoter is selected from the group consisting of the T3 promoter and the transcription terminator is selected from the group consisting of the T7 transcription terminator.
In one embodiment, the 5' -UTR comprises or consists of a sequence according to SEQ ID NO: 3, or a DNA sequence according to SEQ ID NO: 3, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity.
In one embodiment, the 3' -UTR comprises or consists of a sequence according to SEQ ID NO: 4, or a DNA sequence according to SEQ ID NO: 4, having a sequence identity of at least 80%, 90%, 95%, 96%, 97%, 98%, or 99%.
In one embodiment, the DNA nucleic acid molecule further comprises a first restriction enzyme site upstream of the promoter and a second restriction enzyme site downstream of the transcription terminator.
In one embodiment, said DNA nucleic acid molecule further comprises a third restriction enzyme site located between said promoter and said 5 '-UTR and a fourth restriction enzyme site located between said 3' -UTR and said transcription terminator.
In yet another aspect, the present disclosure provides an in vitro transcription method comprising the steps of:
(a) providing a DNA nucleic acid molecule according to the present disclosure as a transcription template;
(b) optionally, amplifying the DNA nucleic acid molecule;
(c) the DNA nucleic acid molecule is added to 5' -anti-reverse cap analogue (5 ' -ARCA), preferably 7m G (3 ' -O-Me) pppG
Performing in vitro transcription in the presence of a promoter to obtain a reaction mixture comprising 5' -ARCA-terminated mRNA;
(d) optionally, removing the transcription template in the reaction mixture comprising 5' -ARCA-terminated mRNA by adding dnase; and
(e) adding a polya polymerase reaction mixture to the reaction mixture comprising 5 '-ARCA-terminated mRNA for 3' -polya tail addition to obtain 5 '-ARCA-terminated mRNA with a 3' -polya tail.
In yet another aspect, the present disclosure provides a composition comprising an RNA nucleic acid molecule according to the present disclosure and/or an RNA nucleic acid molecule obtained according to the in vitro transcription method of the present disclosure, and a pharmaceutically acceptable carrier and/or excipient.
In one embodiment, the composition is a pharmaceutical composition or a vaccine or a kit.
In a further aspect, the present disclosure provides the use of an RNA nucleic acid molecule according to the present disclosure and/or an RNA nucleic acid molecule obtained according to the in vitro transcription method of the present disclosure for the preparation of a medicament for the treatment or prevention of a disease or disorder selected from an immune disease, a genetic disease, a cancer, an infectious disease, an inflammatory disease, an allergy and/or for gene therapy and/or immunomodulation.
Drawings
The drawings are only for purposes of illustrating the invention in more detail and are not to be construed as limiting the scope of the disclosure or the claims in any way.
Fig. 1 shows the structure of a nucleic acid molecule according to an embodiment of the present disclosure: a DNA nucleic acid construct (fig. 1A); an RNA nucleic acid molecule (FIG. 1B) and preferably 5' -ARCA (FIG. 1C).
FIG. 2 shows in vitro transcription of EGF and bFGF mRNA.
FIG. 3 shows the fluorescence signal of 293T cells transfected with GFP mRNA.
Fig. 4 shows the results of quantification of intracellular EGF and bFGF mRNA in 293T cells by qPCR.
FIG. 5 shows the expression of EGF and FGF proteins in 293T cells. Molecular weights of EGF and bFGF mRNA were confirmed by gel electrophoresis (fig. 5A); after transfection of EGF and bFGF mRNA into 293T cells, whole cell lysates and cell culture media were analyzed by western blot at the indicated time points, where the transfected 293T cells showed significant increases in extracellular hbFGF (fig. 5B), intracellular hbFGF (fig. 5C), extracellular hEGF (fig. 5D) and intracellular hEGF (fig. 5E).
Detailed Description
For the purposes of promoting an understanding of the principles of the invention, reference will now be made to the embodiments illustrated in the drawings and specific description will be made to the embodiments. It will nevertheless be understood that no limitation of the scope of the invention is thereby intended.
A detailed description of a cell-free and vector-free in vitro transcription method and constructs for use in these methods is provided below. These methods and constructs fulfill at least one need present in the art.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described in any way.
Unless explicitly defined otherwise, terms used herein should be understood according to their ordinary meaning in the art. Unless the context clearly dictates otherwise, the terms "a", "an", and "the" mean "one or more" are used interchangeably.
Standard techniques and procedures are generally performed according to conventional methods in the art and various general references (see, generally, Sambrook et al, molecular Cloning: Alabortory Manual, 2nd ed. (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.).
As used herein, unless otherwise indicated, the term "about" means +/-10%, more preferably +/-5%, such as +/-4%, +/-3%, +/-2%, or +/-1% of the specified value.
It should be noted that the features in the embodiments and examples of the present disclosure may be combined with each other without conflict.
The present disclosure provides a novel in vitro transcribed RNA platform for the production of mRNA, e.g., mRNA therapeutics. The platform can provide cost-effective mRNA production, has high safety and effectiveness, and is easy to administer into cells. The mRNA provided by the present disclosure comprises a 5' -anti-inversion cap analog (5 ' -ARCA), a human beta globin 5' -untranslated region sequence, a signal sequence and a target gene or target gene coding sequence, a human alpha globin 3' untranslated sequence, and a 3' -poly a tail. The mRNA produced on this platform shows enhanced gene expression stability and translation efficiency. Also, the novel platform showed maximum yields of mRNA that could be as high as about 2.3mg/mL or even higher within 60 minutes of incubation. The present disclosure is based, at least in part, on the surprising discovery that: the combination of elements selected in the present disclosure not only enables maximum production of mRNA up to about 2.3mg/mL or even higher per hour, but the resulting mRNA exhibits enhanced gene expression stability and translation efficiency.
As used herein, the term "nucleic acid" or "nucleic acid molecule" refers to any DNA or RNA molecule, and is used interchangeably with polynucleotide. When reference is made herein to a nucleic acid or nucleic acid sequence encoding a particular protein and/or peptide, said nucleic acid or nucleic acid sequence, respectively, preferably further comprises a regulatory sequence, allowing expression, i.e. transcription and/or translation, of the nucleic acid sequence encoding the particular protein or peptide in a suitable host, such as a human.
RNA nucleic acid molecules
In one aspect, the present disclosure provides an RNA nucleic acid molecule comprising, from the 5 'end to the 3' end, a 5 '-cap structure, a human β -globin 5' untranslated region (5 '-UTR), at least one coding region operably linked to the 5' -UTR and the 3 '-UTR, a human α -globin 3' untranslated region (3 '-UTR), and a 3' -poly a tail.
Untranslated region (UTR)
As used herein, the term "untranslated region (UTR)" refers to "untranslated regions" located upstream (5 ') and/or downstream (3') of the coding region of a nucleic acid molecule described herein, and thus generally flank the coding region. Thus, the term "UTR" generally includes the 5 '-untranslated region ("5' -UTR") and the 3 '-untranslated region ("3' -UTR"). UTRs can generally comprise or consist of nucleic acid sequences that are not translated into proteins. The UTR may comprise one or more regulatory elements.
As used herein, the term "regulatory element" refers to a nucleic acid sequence having gene regulatory activity capable of effecting transcription or translation of an operably (in cis or trans) linked transcribable nucleic acid sequence. The term may include promoters, enhancers, Internal Ribosome Entry Sites (IRES), introns, leader sequences, transcription termination signals such as polyadenylation signals, and other expression control elements.
As used herein, the term "operably linked" refers to an arrangement of elements wherein the elements described in that term are assembled to perform their ordinary function. For example, a given promoter operably linked to a nucleic acid sequence can affect the expression of that sequence in the presence of the appropriate enzyme. The promoter need not be contiguous with the sequence, so long as it directs expression of the sequence. Thus, for example, a intervening untranslated yet transcribed sequence can be present between a promoter sequence and a nucleic acid sequence, and the promoter sequence can still be considered "operably linked" to the coding sequence.
In some embodiments, UTRs are "operably linked", i.e., located upstream and downstream of the coding region in a functional relationship, preferably upstream and downstream of the coding region in a manner that allows them to control (i.e., regulate or modulate, preferably enhance) expression of the coding sequence.
The inventors of the present application have unexpectedly discovered that a combination of 5' -and 3' -untranslated regions (UTRs), preferably in combination with the 5' -cap structure and poly a tail of the present disclosure, act synergistically together to synergistically enhance expression of operably linked nucleic acid sequences. Nucleic acid molecules having the UTR combinations of the present invention advantageously enable rapid and transient expression of large amounts of polypeptides or proteins delivered for gene therapy or immunotherapy purposes. Thus, the nucleic acid molecules provided herein are particularly useful for a variety of therapeutic applications in vivo, including, for example, gene therapy, cancer immunotherapy, or vaccination against infectious agents.
The testing of the synergy of UTR combinations (preferably in combination with the 5' -cap structure and poly a tail of the present disclosure) is routine for those skilled in the art, e.g., synergy testing can be performed by luciferase expression after mRNA transfection to demonstrate that synergy, i.e., not just additive, is present.
As used herein, the term "5 '-UTR" refers to a portion of a nucleic acid molecule that is 5' of the open reading frame (i.e., "upstream") and is not translated into protein. In the context of the present disclosure, the 5' -UTR starts with the transcription start site and terminates one nucleotide before the start codon of the open reading frame.
As used herein, the term "3 '-UTR" refers to a portion of a nucleic acid molecule that is located 3' (i.e., "downstream") of an open reading frame and is not translated into protein. In the context of the present disclosure, a 3 '-UTR corresponds to a sequence located between 3' of a stop codon of a protein coding sequence (preferably immediately adjacent to the stop codon of the protein coding sequence) and a polyadenylation sequence.
In one embodiment, the UTR combination of the present disclosure is a human β -globin 5 'untranslated region (5' -UTR) and a human α -globin 3 'untranslated region (3' -UTR). In one embodiment, the UTR combinations of the present disclosure comprise (a) a human β -globin 5 'untranslated region or homolog, variant, or fragment thereof and (b) a human α -globin 3' untranslated region or homolog, variant, or fragment thereof. In some embodiments, a homologue or variant differs from a UTR only by nucleotides other than the regulatory element of the UTR, e.g., one or more nucleotide differences, such as 1-20, 2-15, 3-10, 4-9, 5-8, 6-7 nucleotide differences. In some embodiments, a fragment of a UTR comprises all regulatory elements of said UTR.
In some embodiments, the human β -globin 5' -UTR comprises or consists of a sequence according to SEQ ID NO: 1, or an RNA sequence according to SEQ ID NO: 1, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity. In some embodiments, the polypeptide is identical to a polypeptide according to SEQ ID NO: 1 with the RNA sequence of SEQ ID NO: 1, except for regulatory elements in the UTR, e.g., one or more nucleotide differences, such as 1-20, 2-15, 3-10, 4-9, 5-8, 6-7 nucleotide differences.
SEQ ID NO: 1 (RNA sequence of human beta-globin 5' -UTR)
5’-ACAUUUGCUUCUGACACAACUGUGUUCACUAGCAACCUCAAACAGACACC-3’
In some embodiments, the human α -globin 3' -UTR comprises or consists of a sequence according to SEQ ID NO: 2, or an RNA sequence according to SEQ ID NO: 2, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity. In some embodiments, the polypeptide is identical to a polypeptide according to SEQ ID NO: 2 with the RNA sequence of SEQ ID NO: 2, e.g., one or more nucleotide differences, such as 1-20, 2-15, 3-10, 4-9, 5-8, 6-7 nucleotide differences, are present only outside of the regulatory elements in the UTR.
The amino acid sequence of SEQ ID NO: 2 (RNA sequence of human alpha-globin 3' -UTR)
5’-GCUGGAGCCUCGGUAGCCGUUCCUCCUGCCCGCUGGGCCUCCCAACGGGCCCUCCUCCCCUCCUUGCACCGGCCCUUCCUGGUCUUUG-3’
5' -cap structure
According to a particularly preferred embodiment of the present invention, the RNA nucleic acid molecules of the present disclosure are modified by the addition of a "5' -cap structure" which may further stabilize the RNA nucleic acid molecule.
The "5' -cap structure" can be formed from modified nucleotides, in particular from derivatives of guanine nucleotides. Preferably, the 5 '-cap is linked to the 5' -terminus by a 5 '-5' -triphosphate linkage. In some embodiments, the 5 'cap can be methylated, for example m7GpppN, where N is the 5' terminal nucleotide of the nucleic acid with the 5 '-cap, typically the 5' -end of an mRNA. m7GpppN is a 5' -cap structure (cap 0 structure) that is naturally present in mRNA transcribed by polymerase II.
Other examples of 5 '-cap structures known in the art include glyceryl moieties, inverted deoxyabasic residues (moieties), 4', 5 'methylene nucleotides, 1- (. beta. -D-erythroribofuranosyl) nucleotides, 4' -thio nucleotides, carbocyclic nucleotides, 1, 5-anhydrohexitol nucleotides, L-nucleotides, alpha-nucleotides, modified base nucleotides, threo-pentofuranosyl nucleotides, acyclic 3', 4' -seco nucleotides, acyclic 3, 4-dihydroxybutyryl nucleotides, acyclic 3, 5-dihydroxypentyl nucleotides, 3 '-3' -inverted nucleotide moieties, 3 '-3' inverted abasic moieties, 3 '-2' inverted nucleotide moieties, 3 '-2' inverted abasic moieties, 1, 4-butanediol phosphate, 3 '-phosphoramidate, hexyl phosphate, aminohexyl phosphate, 3' -phosphorothioate, phosphorodithioate, or bridged or unbridged methylphosphonate moieties.
The 5' -cap structure can be cap 0, cap 1 (ribose of adjacent nucleotides of m7G are methylated), cap 2 (ribose of 2nd nucleotide downstream of m7G is additionally methylated), cap 3 (ribose of 3 rd nucleotide downstream of m7G is additionally methylated), cap 4 (ribose of 4 th nucleotide downstream of m7G is methylated).
In some embodiments, a particularly preferred 5' -cap structure useful in the present disclosure is ARCA (anti-inversion cap analog). In one embodiment, the ARCA is 7m G (3' -O-Me) pppG. The ARCA is prepared by reacting the 3' -OH group of the 7mG residue in 7mGpppG with OCH3("OMe") substituted. Several types of ARCA analogs are known in the art (see, e.g., U.S. patent No. 7,074,596). However, the present application surprisingly found that with 7m G (3 '-O-Me) pppG, a large molar excess of ARCA relative to pppG is not required to ensure that most mRNA transcript molecules have a 5' -cap structure, compared to other ARCA analogs. In some embodiments, the molar ratio of 7m G (3' -O-Me) pppG to pppG (gtp) is no greater than 4:1, e.g., 3:1, 2:1, or 1: 1.
It has also been found herein that the use of ARCAs, particularly 7m G (3' -O-Me) pppG, in combination with the UTRs described in this disclosure synergistically enhance the expression of operably linked nucleic acid sequences. Nucleic acid molecules having the combination of the ARCA of the invention, particularly 7m G (3' -O-Me) pppG, and UTR advantageously enable rapid and transient expression of large amounts of polypeptides or proteins delivered for gene therapy or immunotherapy purposes.
3' -PolyA tails
According to a further preferred embodiment, the RNA nucleic acid molecule of the invention may comprise a poly (a) sequence.
As used herein, the term "polya sequence" also referred to as a "polya tail" or "3' -polya tail" refers to a sequence of adenosine nucleotides, e.g., a sequence of up to about 400 adenosine nucleotides, e.g., about 20 to about 400, preferably about 50 to about 400, more preferably about 50 to about 300, even more preferably about 50 to about 250, most preferably about 60 to about 250 adenosine nucleotides. As used herein, a "polyadenylation sequence" may also comprise about 10 to 200 adenosine nucleotides, preferably about 10 to 100 adenosine nucleotides, more preferably about 40 to 80 adenosine nucleotides or even more preferably about 50 to 70 adenosine nucleotides. The poly A sequence is usually located at the 3' end of the RNA, especially the mRNA.
The polyadenylation sequence in the RNA nucleic acid molecule can preferably be obtained from the DNA template by in vitro transcription of the RNA. Alternatively, the polyadenylation sequence may be obtained in vitro by conventional methods of chemical synthesis, without having to be transcribed from the DNA template.
In addition, a polyadenylation sequence or polyadenylation tail may be generated by enzymatic polyadenylation of the RNA nucleic acid molecule using commercially available polyadenylation kits and corresponding protocols known in the art. Polyadenylation is generally understood to be the addition of a polyadenylation sequence to an RNA nucleic acid molecule, e.g. a pre-mature mRNA. Polyadenylation can be induced by a so-called polyadenylation signal. The signal is preferably located within a nucleotide fragment at the 3' end of the nucleic acid (RNA) sequence to be polyadenylated. The polyadenylation signal generally comprises a hexamer consisting of adenine and uracil/thymine nucleotides, preferably the hexamer sequence AAUAAA.
The inventors of the present application further found that the addition of a poly (a) tail to the 3' end of the RNA nucleic acid molecules of the present disclosure further increases the stability of the RNA nucleic acid molecules. This poly a tail in combination with the ARCAs described in the present disclosure, especially 7m G (3' -O-Me) pppG, and the UTRs described in the present disclosure synergistically enhance the stability of operably linked nucleic acid sequences and their expression yield.
Coding region
The nucleic acid molecule according to the invention comprises at least one coding region or coding sequence which is operably linked (typically flanked) to at least one 3 '-UTR element and at least one 5' -UTR element as defined herein. The terms "coding sequence" or "cds" and "coding region" are used interchangeably herein to refer to a segment or portion of nucleic acid that encodes a product of interest (gene). Gene products are the products of gene expression, including polypeptides and nucleic acids, and in general, at least one coding region of a nucleic acid molecule of the present disclosure may encode at least one polypeptide or protein, referred to as a "polypeptide or protein of interest. The coding region is typically composed of exons bounded at their 5 'end by a start codon (e.g., AUG) and bounded at their 3' end by a stop codon (e.g., UAG, UAA, or UGA). In the nucleic acid molecules of the present disclosure, the coding region is bordered by at least one 5 '-UTR element and at least one 3' -UTR element as defined herein.
The polypeptide or protein of interest generally includes any polypeptide or protein that can be encoded by a nucleic acid sequence having at least one coding region and can be expressed under appropriate conditions to produce a functional polypeptide or protein product. As used herein, the term "functional" refers to "capable of performing a desired biological function" and/or "exhibiting a desired biological property". The polypeptide or protein of interest can have a variety of functions and include, for example, growth factors, antibodies, enzymes, signaling proteins, receptors, receptor ligands, peptide hormones, transporters, structural proteins, neurotransmitters, serum proteins, carriers, drugs, immunomodulators, oncogenes, anticancer agents, toxins, tumor antigens, and the like. In some embodiments, the polypeptide or protein of interest may be a therapeutic, antigenic, and allergenic polypeptide or protein.
Cell growth factor
In some embodiments, at least one coding region of a nucleic acid molecule of the present disclosure may encode at least one growth factor. As used herein, the term "growth factor" refers to a class of polypeptides that modulate the multiple effects of cell growth and other cellular functions by binding to specific, high affinity cell membrane receptors. In some embodiments, the growth factor may be selected from platelet-like growth factors (platelet-derived growth factors, PDGF); osteosarcoma-derived growth factor (ODGF), epidermal growth factors (epidermal growth factor EGF, transforming growth factors TGF alpha and TGF beta), fibroblast growth factors (alpha FGF, beta FGF), insulin-like growth factors (IGF-I, IGF-II), Nerve Growth Factor (NGF), interleukin-like growth factors (IL-1, IL-3, etc.), erythrocyte growth factor (EPO), Colony Stimulating Factor (CSF), etc.
Therapeutic polypeptides or proteins
In some embodiments, at least one coding region of a nucleic acid molecule of the present disclosure may encode at least one "therapeutic polypeptide or protein. The term "therapeutic polypeptide or protein" refers to a polypeptide or protein that is capable of mediating a desired diagnostic, prophylactic or therapeutic effect, preferably resulting in the detection, prevention, amelioration and/or cure of a disease.
Preferably, the nucleic acid molecule according to the present disclosure may comprise at least one coding region encoding a therapeutic protein in place of a deleted, defective or mutated protein; therapeutic proteins useful for the treatment of genetic or acquired diseases, infectious diseases or tumors, such as cancer or neoplastic diseases; adjuvant or immunostimulatory therapeutic proteins; a therapeutic antibody or antibody fragment, variant or derivative; a peptide hormone; a gene editing agent; an immune checkpoint inhibitor; a T cell receptor, or a T cell receptor fragment, variant or derivative; and/or an enzyme.
Antigenic polypeptide or protein
The at least one coding region of a nucleic acid molecule of the present disclosure may encode at least one "antigenic polypeptide or protein". The term "antigenic polypeptide or protein" or simply "antigen" generally refers to any polypeptide or protein "antigenic peptide or protein" which is capable of interacting with/being recognized by a component of the immune system (e.g., an antibody or immune cell through its antigenic receptor, such as a B Cell Receptor (BCR) or T Cell Receptor (TCR)) under appropriate conditions, and preferably capable of eliciting an immune response, preferably by interacting with a component of the immune system through its "epitope" or "antigenic determinant".
The choice of a particular antigenic polypeptide or protein will generally depend on the disease to be treated or prevented. In general, the nucleic acid molecule may encode any antigenic polypeptide or protein associated with a disease (e.g., cancer, infectious disease) that can be treated by inducing an immune response against the antigenic infectious disease.
Preferably, the artificial nucleic acid molecule according to the invention may comprise at least one coding region encoding a tumor antigen, a pathogenic antigen, an autoantigen, an alloantigen or an allergenic antigen. Particularly preferred are the tumor antigens NY-ESO-1, 5T4, MAGE-C1, MAGE-C2, Muc-1, PSA, PSMA, PSCA, STEAP and PAP.
Allergenic polypeptides or proteins
The at least one coding region of the nucleic acid molecules of the present disclosure may encode at least one "allergenic polypeptide or protein". The term "allergenic polypeptide or protein" or "allergen" refers to a polypeptide or protein that is capable of inducing an allergic response, i.e., a pathological immune response characterized by altered physical reactivity (e.g., hypersensitivity) when exposed to a subject. Generally, "allergens" are associated with "atopy", i.e. an adverse immune reaction involving immunoglobin e (ige). Thus, the term "allergen" generally refers to a substance (here, a polypeptide or protein) that is associated with atopy and induces IgE antibodies. In some embodiments, the allergen may include insect-derived allergens, mammalian allergens, mollusk-derived allergens, plant allergens, fungal allergens, and the like.
Other domains, tags, linkers, sequences or elements
Preferably, in addition to encoding at least one polypeptide or protein of interest, at least one coding region of a nucleic acid molecule of the present disclosure may encode other polypeptide domains, tags, linkers, sequences or elements. The nucleic acid sequence encoding the further domain, tag, linker, sequence or element is operably linked in frame to a region encoding the polypeptide or protein of interest such that expression of the coding sequence preferably produces a fusion product of the polypeptide or protein of interest coupled to the further domain, tag, linker, sequence or element.
The at least one coding region of the nucleic acid molecules of the present disclosure may further encode at least one of: a signal peptide, a peptide or protein tag, a localization signal or sequence, a peptide linker, etc.
The term "signal peptide" (also sometimes referred to as a secretory signal peptide or signal sequence) refers to a typical short peptide (which can typically be 16 to 30 amino acids in length) that is typically present at the end of a protein that is to be secreted via the secretory pathway.
Preferably, a signal peptide may be introduced into the polypeptide or protein of interest to facilitate secretion of the polypeptide or protein. In particular, where the nucleic acid encoding the antigenic polypeptide or protein is fused to a signal peptide, appropriate secretion may help trigger an immune response against the antigen. The signal peptide may also be effectively combined with any other polypeptide or protein disclosed herein. When encoded in combination with a polypeptide or protein of interest, such a signal peptide may be located at the N-terminus, C-terminus, and/or inside of the polypeptide or protein of interest, preferably at the N-terminus. At the nucleic acid level, the coding sequence for such a signal peptide is usually placed in frame (i.e., in the same reading frame).
Exemplary signal peptides useful in the present disclosure include, but are not limited to, signal sequences of classical or non-classical MHC molecules (e.g., signal sequences of MHC I and MHC II molecules, e.g., signal sequence of MHC class I molecule HLA-a 0201), signal sequences of cytokines or immunoglobin, signal sequences of immunoglobulin or antibody constant chains, signal sequences of Lamp1, Tapasin, Erp57, Calretikulin, calnexin, PLAT, EPO, or albumin, and signal sequences of other membrane-associated proteins or proteins associated with the Endoplasmic Reticulum (ER) or endosomal-lysosomal compartment.
The term "peptide or protein tag" is a short amino acid sequence that is introduced into a polypeptide or protein of interest to impart a desired biological function or property. In general, a "peptide tag" can be used to detect, purify, isolate, or add some desired biological property or function.
A "peptide linker" or "spacer" is a short amino acid sequence that links domains, portions or parts of a polypeptide or protein of interest disclosed herein, e.g., domains, portions or parts of a multi-domain protein or fusion protein. The polypeptide or protein, or a domain, portion or component thereof, is preferably functional, i.e., performs a specific biological function.
DNA nucleic acid molecule
The RNA nucleic acid molecules of the present disclosure can preferably be obtained from DNA templates by RNA in vitro transcription. Alternatively, the polyadenylation sequence may be obtained in vitro by conventional methods of chemical synthesis, without having to be transcribed from the DNA template.
In one embodiment, the RNA nucleic acid molecules of the present disclosure may preferably be obtained from a DNA template by RNA in vitro transcription.
Accordingly, in one aspect, the present disclosure provides a DNA nucleic acid molecule comprising, from the 5 'end to the 3' end, a promoter, a human β -globin 5 '-untranslated region (5' -UTR), at least one coding region, a human α -globin 3 '-untranslated region (3' -UTR), and a transcription terminator, the at least one coding region operably linked to the 5 '-UTR and the 3' -UTR.
In one embodiment, a DNA nucleic acid molecule of the present disclosure includes a UTR combination of a human β -globin 5 'untranslated region (5' -UTR) and a human α -globin 3 'untranslated region (3' -UTR). In one embodiment, the UTR combinations of the present disclosure comprise (a) a human β -globin 5 'untranslated region or homolog, variant, or fragment thereof and (b) a human α -globin 3' untranslated region or homolog, variant, or fragment thereof. In some embodiments, a homologue or variant differs from a UTR only by nucleotides other than the regulatory element of the UTR, e.g., one or more nucleotide differences, such as 1-20, 2-15, 3-10, 4-9, 5-8, 6-7 nucleotide differences. In some embodiments, a fragment of a UTR comprises all regulatory elements of said UTR.
In some embodiments, the human β -globin 5' -UTR comprises or consists of a sequence according to SEQ ID NO: 3, or to an RNA sequence according to SEQ ID NO: 3, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity. In some embodiments, the polypeptide is identical to a polypeptide according to SEQ ID NO: 3 with the DNA sequence of SEQ ID NO: 3, e.g., one or more nucleotide differences, such as 1-20, 2-15, 3-10, 4-9, 5-8, 6-7 nucleotide differences, are present only outside of the regulatory elements in the UTR.
SEQ ID NO: 3 (DNA sequence of human beta-globin 5' -UTR)
5’-ACATTTGCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACC-3’
In some embodiments, the human α -globin 3' -UTR comprises or consists of a sequence according to SEQ ID NO: 4, or a DNA sequence according to SEQ ID NO: 4 has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity. In some embodiments, the polypeptide is identical to a polypeptide according to SEQ ID NO: 4 and the sequence of SEQ ID NO: 4, e.g., one or more nucleotide differences, such as 1-20, 2-15, 3-10, 4-9, 5-8, 6-7 nucleotide differences, are present only outside of the regulatory elements in the UTR.
SEQ ID NO: 4 (DNA sequence of human alpha-globin 3' -UTR)
5’-GCTGGAGCCTCGGTAGCCGTTCCTCCTGCCCGCTGGGCCTCCCAACGGGCCCTCCTCCCCTCCTTGCACCGGCCCTTCCTGGTCTTTG-3’
In some embodiments, the corresponding promoter and transcription terminator or termination element are selected according to the RNA polymerase employed. RNA polymerases useful in the present disclosure include, but are not limited to, T3, T7, Sny5, or SP6 RNA polymerases. Thus, promoters and transcription terminators useful in the present disclosure may be selected from promoters and transcription terminators or termination elements of T3, T7, Sny5, or SP6 RNA polymerase. Exemplary promoters are selected from the T3, T7, Sny5 or SP6 promoters, preferably the T7 promoter. An exemplary transcription terminator or termination element may be the T7 transcription terminator.
In some embodiments, DNA templates for transcription of RNA molecules of the present disclosure may include linear templates obtained by PCR methods or annealing chemically synthesized oligonucleotides, clonally constructed plasmids, and cDNA templates obtained by first and second strand synthesis (e.g., aRNA amplification) based on RNA precursors.
Plasmid vectors used as transcription templates need to be linearized by restriction enzyme digestion. Since the transcription reaction will continue to the end of the DNA template, linearization ensures that RNA transcripts of defined length and sequence are obtained.
In some embodiments, the DNA nucleic acid molecules of the present disclosure further comprise a first restriction enzyme site upstream of the promoter and a second restriction enzyme site downstream of the transcription terminator.
In some embodiments, the first restriction site and the second restriction site allow for insertion of the DNA nucleic acid molecule into a vector, such as a plasmid. As used herein, the term "restriction enzyme" or "restriction endonuclease" refers to a class of enzymes that recognize and attach to a particular deoxyribonucleotide sequence and cleave the phosphodiester bond between two deoxyribonucleotides at a particular site in each strand. The cleavage method is to cleave the bond between the sugar molecule and the phosphate, thereby generating a nick on each of the two DNA strands without destroying the nucleotide and the base. There are two types of cleavage formats, a sticky end with protruding single-stranded DNA and a smooth end with a flat end without protrusions. Since the broken DNA fragments can be ligated by DNA ligase, different restriction fragments on the chromosome or DNA can be joined together via splicing. In some embodiments, the first restriction site and the second restriction site may be the same or different, and both the first restriction site and the second restriction site are EcoRI. Restriction enzymes useful in the present disclosure may include, but are not limited to: EcoRI, PstI, XbaI, BamHI, HindIII, TaqI, NotI, HinfI, Sau3A, PovII, SmaI, HaeIII, AluI, SalI, Dra, etc. The insertion of the target polynucleotide is carried out using standard Molecular Biology methods, for example, as described in Sambrook et al (Sambrook et al. Molecular Cloning: A Laboratory Manual, Cold Spring harbor Laboratory Press,1989) and/or Autosubel et al (Current Protocols in Molecular Biology, Greene pub. Association and Wiley-Interscience (1988.) the method of ligating nucleic acids is obvious to one skilled in the art and is described, for example, in Sambrook et al. Molecular Cloning: A Laboratory Manual, Cold Spring library Laboratory Press,1989 and/or Subel et al (editor), Current Protocols in Molecular Biology, Greene protocol in general, and ligation using, for example, DNA ligase T et al, enzymes ligation.
In some embodiments, the DNA nucleic acid molecules of the present disclosure further comprise a third restriction enzyme site located between the promoter and the 5 '-UTR and a fourth restriction enzyme site located between the 3' -UTR and the transcription terminator. In some embodiments, the third restriction site and the fourth restriction site allow for universal cloning. In some embodiments, the third restriction site and the fourth restriction site are independently selected from the group consisting of EcoRI, PstI, XbaI, BamHI, HindIII, TaqI, NotI, HinfI, Sau3A, poviii, SmaI, HaeIII, AluI, SalI, and Dra. In some embodiments, the third restriction site and the fourth restriction site are different, e.g., SalI and NotI, respectively.
In vitro transcription method
In one aspect, the present disclosure provides an in vitro transcription method comprising the steps of: (a) providing a DNA nucleic acid molecule according to the present disclosure as a transcription template; (b) optionally, amplifying the DNA nucleic acid molecule; (c) subjecting said DNA nucleic acid molecule to in vitro transcription in the presence of a 5 '-anti-reverse cap analogue (5' -ARCA), preferably 7m G (3 '-O-Me) pppG, to obtain a reaction mixture comprising 5' -ARCA-terminated mRNA; (d) optionally, removing the transcription template in the reaction mixture comprising 5' -ARCA-terminated mRNA by adding dnase; and (e) adding a polya polymerase reaction mixture to the reaction mixture comprising 5 '-ARCA-terminated mRNA for 3' -polya tail addition to obtain 5 '-ARCA-terminated mRNA having a 3' -polya tail.
The term "in vitro transcription" of RNA relates to a method of synthesizing RNA from a DNA template in a cell-free system (in vitro). DNA, preferably linear DNA (e.g., linearized plasmid DNA, linearized dbDNA) is used as a template for the generation of RNA transcripts. A DNA template for the in vitro transcription of RNA can be obtained by cloning nucleic acids, in particular cDNA corresponding to the corresponding RNA to be transcribed in vitro, and introducing them into a suitable vector for the in vitro transcription of RNA, for example into plasmid DNA.
Templates for in vitro transcription can also be prepared by PCR amplification using the DNA nucleic acid molecules of the disclosure as templates. After purification of the PCR product, RNA synthesis can be performed by standard in vitro transcription methods. The obtained PCR product is mixed and incubated with an in vitro RNA synthesis premix comprising a 5' -anti-reverse cap analogue (5 ' -ARCA) (e.g., for 30 minutes at 37 ℃) to generate 5' -ARCA-terminated RNA. In some embodiments, an in vitro RNA synthesis premix comprises: buffers suitable for in vitro transcription (e.g., Tris-HCl, pH 7.9), cap analogs (e.g., 5' -anti-reverse cap analogs), ribonucleoside triphosphates (ATP, UTP, CTP, and GTP), ribonuclease inhibitors, and RNA polymerases (e.g., T7 RNA polymerase). In some embodiments, the in vitro RNA synthesis premix may also comprise MgCl2At least one or all of an antioxidant and a polyamine (e.g., spermidine).
To obtain high quality RNA suitable for use in RNA-based therapies, DNA templates can be efficiently and reliably removed from the final RNA product to ensure the efficacy and safety of RNA-based therapeutics. Removal of DNA template from an RNA in vitro transcription reaction can be achieved, for example, by enzymatic (e.g., DNase I) digestion of DNA and purification of RNA.
The 3' -polya tail is performed by additional polya polymerase reaction mixture. In some embodiments, the polya polymerase reaction mixture comprises a buffer (e.g., Tris-HCl, pH 8.1) and polya polymerase and salts (e.g., NaCl and MgCl)2One or two ofSeed) and incubated (e.g., at 37 ℃ for 30 minutes) to obtain 5 '-ARCA-terminated mRNA with a 3' -poly a tail.
Compositions and vaccines
In another aspect, the present disclosure provides a composition comprising an RNA nucleic acid molecule of the present disclosure, and at least one pharmaceutically acceptable carrier and/or excipient. In some preferred embodiments, the composition is provided as a pharmaceutical composition. According to a further preferred embodiment, the composition may be provided as a vaccine. A "vaccine" is generally understood as a prophylactic or therapeutic material providing at least one antigen, preferably an antigenic peptide or protein. By "providing at least one antigen" is meant, for example, that the vaccine comprises an antigen or that the vaccine comprises, for example, a molecule encoding an antigen. Accordingly, a vaccine of the present disclosure may comprise at least one RNA nucleic acid molecule encoding at least one antigenic polypeptide or protein as defined herein, which may for example be derived from a tumor antigen, a bacterial antigen, a viral antigen, a fungal antigen or a protozoan antigen, an autoantigen, an allergen or an alloantigen, and preferably induces an immune response against the respective antigen when expressed and presented to the immune system.
The compositions or vaccines of the present disclosure preferably comprise at least one RNA nucleic acid molecule as described herein. Each RNA nucleic acid molecule in the compositions or vaccines of the present disclosure may encode at least one or at least two (the same or different) multiple polypeptides or proteins of interest. The RNA nucleic acid molecule may be provided in the composition or vaccine in "complexed" or "free" form or mixtures thereof. The composition or vaccine may further comprise at least one additional active agent for the treatment of a disease or disorder treated with the RNA nucleic acid molecule or a composition or vaccine comprising the same.
Preferably, the composition or vaccine according to the invention comprises at least one pharmaceutically acceptable carrier and/or excipient. The term "pharmaceutically acceptable" refers to a compound or agent that is compatible with one or more active agents and does not interfere with and/or significantly reduce its pharmaceutical effect. The pharmaceutically acceptable carriers and excipients preferably have a sufficiently high purity and sufficiently low toxicity to render them suitable for administration to a subject to be treated.
Pharmaceutically acceptable excipients may serve different functions and include, but are not limited to, diluents, fillers, bulking agents, carriers, disintegrants, binders, lubricants, glidants, coatings, solvents and co-solvents, buffers, preservatives, adjuvants, antioxidants, wetting agents, antifoaming agents, thickeners, sweeteners, flavoring agents, and humectants.
For compositions in liquid form, useful pharmaceutically acceptable carriers and excipients include solvents, diluents or carriers such as (pyrogen-free) water, (isotonic) salt solutions such as phosphate or citrate buffered saline, fixed oils, vegetable oils such as peanut oil, cottonseed oil, sesame oil, olive oil, corn oil, ethanol, polyols (e.g., glycerol, propylene glycol, polyethylene glycol, and the like); lecithin; a surfactant; preservatives such as benzyl alcohol, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like; isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol or sodium chloride; aluminum monostearate or gelatin; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetate, citrate or phosphate; and agents for adjusting tonicity such as sodium chloride or dextrose. The pH can be adjusted with an acid or base such as hydrochloric acid or sodium hydroxide. The buffer may be hypertonic, isotonic or hypotonic with respect to the specific reference medium, i.e. the buffer may have a higher, the same or a lower salt content with respect to the specific reference medium, wherein preferably such concentrations of the above mentioned salts may be used which do not cause cell damage due to osmosis or other concentration effects. Reference media are, for example, liquids which are produced in an "in vivo" process, such as blood, lymph, cytoplasmic or other body fluids, or liquids which can be used as reference media, for example in an "in vitro" process, such as, for example, customary buffers or liquids. Such common buffers or liquids are known to the skilled person.
For compositions in (semi) solid form, useful pharmaceutically acceptable carriers and excipients include binders, such as microcrystalline cellulose, gum tragacanth or gelatin; starch or lactose; sugars such as lactose, glucose and sucrose; starches, such as corn starch or potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, cellulose acetate; disintegrating agents, such as alginic acid; lubricants, such as magnesium stearate; glidants, such as stearic acid, magnesium stearate; calcium sulfate, colloidal silica, and the like; sweetening agents, such as sucrose or saccharin; and/or a flavoring agent, such as peppermint, methyl salicylate, or orange flavoring.
Suitable pharmaceutically acceptable carriers and excipients can generally be selected based on the desired formulation of the composition.
Liquid compositions for administration by injection, particularly intravenous injection, should be sterile and stable under the conditions of manufacture and storage. Such compositions are typically formulated as parenterally acceptable aqueous solutions which are pyrogen free, have a suitable pH, are isotonic and maintain the stability of the active ingredient. Particularly useful pharmaceutically acceptable carriers and excipients for liquid compositions according to the invention include water, typically pyrogen-free water; isotonic saline or buffer solutions, e.g., phosphate, citrate, etc. Especially for injection of the composition of the present disclosure, water or preferably a buffer, more preferably an aqueous buffer, comprising a sodium salt, preferably at least 50mM sodium salt; a calcium salt, preferably at least 0.01mM calcium salt; and optionally a potassium salt, preferably at least 3mM potassium salt.
According to a preferred embodiment, the sodium, calcium and optionally potassium salts may be present in the form of their halides, for example chlorides, iodides or bromides, in the form of their hydroxides, carbonates, bicarbonates or sulphates, etc. In addition, the buffer may contain an organic anion of the above cation.
According to a preferred embodiment, the buffer suitable for injection purposes as defined above may comprise a buffer selected from the group consisting of sodium chloride (NaCl), calcium chloride (CaCl)2) And optionally potassium chloride (KCl), wherein in addition to chloride ions, further anions may be presentAnd (4) adding the active ingredients. CaCl2Another salt such as KCl may be substituted. Typically, the salt concentration in the injection buffer is at least 50mM sodium chloride (NaCl), at least 3mM potassium chloride (KCl), and at least 0.01mM calcium chloride (CaCl)2)。
Compositions for topical administration may be formulated as emulsions, ointments, gels, pastes or powders using suitable liquid and/or (semi-) solid excipients or carriers as described herein above and below. Orally administered compositions may be formulated into tablets, capsules, liquids, powders, or sustained release forms using suitable liquid and/or (semi-) solid excipients or carriers as described elsewhere herein.
According to some preferred embodiments, the composition or vaccine of the present disclosure is administered parenterally, in particular by intradermal or intramuscular injection, orally, intranasally, pulmonary, inhalation, topically, rectally, buccally, vaginally or by implanted depot, and is provided in the form of a liquid or lyophilized formulation for parenteral administration as discussed elsewhere herein. Parenteral preparations are usually stored in vials, intravenous bags, ampoules, cartridges or prefilled syringes and can be administered in the form of injections, inhalants or aerosols, preferably in the form of injections.
According to a preferred embodiment, the composition or vaccine of the present disclosure may comprise an RNA nucleic acid molecule of the present disclosure complexed to a lipid, which may be, for example, in the form of a lipid nanoparticle, a liposome, a lipid complex, or an emulsion.
According to other preferred embodiments, the composition or vaccine of the present disclosure is provided in lyophilized form. Preferably, the lyophilized composition or vaccine is reconstituted in a suitable buffer prior to administration, the buffer advantageously being based on an aqueous carrier, such as lactated ringer's solution, phosphate buffer, preferably lactated ringer's solution.
According to a preferred embodiment, the composition or vaccine of the present disclosure may further comprise at least one adjuvant. An "adjuvant" or "auxiliary component" in the broad sense is typically a pharmacological and/or immunological agent that may alter, e.g., enhance, the action of other active agents, e.g., therapeutic agents or vaccines. Herein, "adjuvant" may be understood as any compound suitable to support administration and delivery of the compositions of the present disclosure. In particular, the adjuvant may preferably enhance the immunostimulatory properties of the composition or vaccine to which it is added. In addition, such adjuvants may, but are not limited to, elicit or enhance the immune response of the innate immune system, i.e., a non-specific immune response.
Reagent kit
In another aspect, the present disclosure relates to a kit or kit of parts comprising an RNA nucleic acid molecule and/or composition or vaccine of the present disclosure.
In a kit or kit of parts of the present disclosure, at least one RNA nucleic acid molecule in lyophilized or liquid form, optionally together with one or more pharmaceutically acceptable carriers and/or excipients.
Optionally, the kits or kits of reagents of the present disclosure may further comprise other reagents, such as antimicrobial agents, rnase inhibitors, solubilizing agents, and the like.
The kit of reagents may be a two or more part kit and will typically contain its components in suitable containers. For example, each container may be in the form of a vial, bottle, squeeze bottle, jar, sealed pouch, or the like, or any other suitable form, provided that the container is configured to prevent premature mixing of the components. Each of the different components may be provided separately, or may be provided with some of the different components (i.e., in the same container). The kit may also contain instructions for any administration and dosage information regarding its ingredients.
Medical use and treatment
The RNA nucleic acid molecules or compositions or vaccines or kits of the present disclosure can be used in humans, but also for veterinary purposes, preferably for human medical purposes.
According to another aspect, the invention thus relates to an RNA nucleic acid molecule, composition, vaccine, or kit of the present disclosure for use as a medicament.
The RNA nucleic acid molecules, compositions or vaccines or kits of the present disclosure can be used to treat genetic diseases, cancer, autoimmune diseases, inflammatory diseases, and infectious diseases or other diseases or conditions.
According to another aspect, the invention therefore relates to an RNA nucleic acid molecule, composition or vaccine or kit of the present disclosure for use in the treatment of genetic diseases, cancer, autoimmune diseases, inflammatory and infectious diseases or other diseases or conditions.
"Gene therapy" preferably involves regulating gene expression in a subject to achieve a therapeutic effect. For this purpose, gene therapy generally involves the introduction of nucleic acids into cells. Gene therapy may involve transformation of host cells in vivo or in vitro.
The term "treating" a disease includes preventing the disease (i.e., causing clinical symptoms not to develop); inhibiting disease (i.e., arresting or inhibiting the development of clinical symptoms); and/or relieving the disease (i.e., causing regression of clinical symptoms). It will be appreciated that it is not always possible to distinguish between "preventing" and "inhibiting" a disease or condition, as one or more of the ultimate inducing events may be unknown or potential. Thus, the term "prevention" will be understood to constitute a type of "treatment" that encompasses both "prevention" and "inhibition". Thus, the term "treatment" includes "prevention".
As used herein, the term "subject", "patient" or "individual" generally includes humans and non-human animals, and preferably includes mammals (e.g., non-human primates including marmosets, tamarins, spider monkeys, owl monkeys, long tail chimpanzees, squirrel monkeys, and baboons, macaques, chimpanzees, orangutans, gorillas, cattle, horses, sheep, pigs, chickens, cats, dogs, mice, rats, rabbits, guinea pigs, and the like), including chimeric and transgenic animals and disease models. In this context, the term "subject" preferably refers to a non-human primate or human, most preferably a human.
Accordingly, the present disclosure also provides a method of treating a disease disclosed herein by administering to a subject in need thereof a pharmaceutically effective amount of an RNA nucleic acid molecule, composition or vaccine or kit, comprising administering to a patient/subject in need thereof a pharmaceutically effective amount of the RNA nucleic acid molecule, composition or vaccine or kit.
The RNA nucleic acid molecules or compositions or vaccines or kits of the present disclosure can be administered, e.g., systemically or locally. Systemic routes of administration typically include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal, and intraperitoneal injection and/or intranasal routes of administration. Topical routes of administration generally include, for example, topical routes, but also include intradermal, transdermal, subcutaneous or intramuscular injections or intralesional, intratumoral, intracranial, intrapulmonary, intracardial, and sublingual injections.
According to a preferred embodiment, the RNA nucleic acid molecule, composition or vaccine or kit is administered by parenteral route, preferably by intradermal, subcutaneous or intramuscular route. Preferably, the RNA nucleic acid molecule composition or vaccine or kit can be administered by injection, e.g. subcutaneous, intramuscular, or intradermal injection, which can be needle-free and/or needle injection. Thus, in a preferred embodiment, the medical use and/or the method of treatment according to the present invention comprises administering the RNA nucleic acid molecule, composition or vaccine or kit by subcutaneous, intramuscular or intradermal injection, preferably by intramuscular or intradermal injection, more preferably by intradermal injection. Such injection may be performed by injection using a conventional needle or (needle-free) jet, preferably by injection using a (needle-free) jet.
The RNA nucleic acid molecule, (pharmaceutical) composition or vaccine or kit of the present disclosure may be administered to a subject in need thereof several times per day, daily, every other day, weekly or monthly; and may be administered sequentially or simultaneously.
If different RNA nucleic acid molecules, or compositions or vaccines or kits comprising several components, e.g.different RNA nucleic acid molecules and optionally separately other active agents as described herein, are administered, each component may be administered simultaneously (simultaneously by the same or different routes of administration) or separately (administered at different times by the same or different routes of administration).
The RNA nucleic acid molecules, compositions or vaccines or kits of the present disclosure can preferably be administered in a therapeutically effective amount. As used herein, "therapeutically effective amount" refers to an amount of an active agent sufficient to elicit a desired biological or pharmaceutical response in a tissue, system, animal or human that is being sought. The therapeutically effective amount is preferably sufficient to induce a positive change in the disease to be treated, i.e. to alleviate symptoms of the disease to be treated, to reduce disease progression or to prevent symptoms of the disease to be prevented. At the same time, however, the "therapeutically effective amount" is preferably small enough to avoid serious side effects, that is to say to allow a reasonable relationship between advantage and risk, i.e. a safe and therapeutically effective amount.
The "therapeutically effective amount" will also vary with the particular condition being treated and the age, physical condition, weight, sex, and diet of the patient being treated, the severity of the condition, the duration of the treatment, the nature of the concomitant therapy, the particular pharmaceutically acceptable carrier or excipient used, the treatment regimen, and like factors.
A "therapeutically effective amount" of an RNA nucleic acid molecule may also be selected depending on the type of RNA nucleic acid molecule, e.g. monocistronic, bicistronic or even polycistronic RNA, since a bicistronic or even polycistronic RNA may result in significantly higher expression of the encoded polypeptide or protein of interest in case the amount of monocistronic RNA is equal.
Therapeutic efficacy and toxicity of the RNA nucleic acid molecules, compositions or vaccines or kits of the present disclosure can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining LD50 (the dose lethal to 50% of the population) and ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED 50. RNA nucleic acid molecules, compositions or kits that exhibit a large therapeutic index are generally preferred. Data obtained from cell culture assays and animal studies can be used to formulate a range of dosages for use in humans. The dosage of such compounds is preferably within a range that includes circulating concentrations of ED50 with little or no toxicity.
For example, a therapeutically effective dose of an RNA nucleic acid molecule, composition or vaccine or kit described herein may be about 0.001mg to 10mg per dosage unit, preferably about 0.01mg to 5mg per dosage unit, more preferably about 0.1mg to 2mg per dosage unit or about 0.01nmol to 1mmol per dosage unit, in particular 1nmol to 1mmol per dosage unit, preferably 1 μmol to 1mmol per dosage unit. In some embodiments, a therapeutically effective dose of an RNA nucleic acid molecule, composition or vaccine or kit of the present disclosure can be about 0.01g/kg to 10g/kg, preferably about 0.05mg/kg to 5g/kg, more preferably about 0.1mg/kg to 2.5g/kg (per kg body weight).
Genetic diseases
In a preferred embodiment, the RNA nucleic acid molecule, the (pharmaceutical) composition or the vaccine or the kit is for use in the treatment or prevention of a genetic disease.
As used herein, the term "genetic disorder" includes any disease, disorder or condition caused by, characterized by, or associated with genomic abnormalities (i.e., deviations from wild-type, healthy and asymptomatic states). Such abnormalities may include changes in chromosome copy number (e.g., aneuploidy) or partial changes thereof (e.g., deletions, duplications, amplifications); or a change in chromosome structure (e.g., translocation, point mutation). Genomic abnormalities may be genetic (recessive or dominant) or non-genetic. Genomic abnormalities can be present in certain cells of an organism or in all cells of the organism, and include autosomal abnormalities, X-linked abnormalities, Y-linked abnormalities, and mitochondrial abnormalities.
Cancer treatment
In a preferred embodiment, the RNA nucleic acid molecule, composition or vaccine or kit is for use in the treatment or prevention of cancer.
As used herein, the term "cancer" refers to a neoplasm characterized by uncontrolled and often rapid proliferation of cells that have a tendency to invade surrounding tissues and metastasize to distant body sites. The term includes benign tumors and malignant tumors. Malignant tumors in cancer are often characterized by anaplasia, invasion, and metastasis; while benign tumors do not usually possess these properties. The term includes neoplasms that grow as tumors, as well as cancers of the blood and lymphatic system.
In some embodiments, the RNA nucleic acid molecule, composition or vaccine or kit according to the present disclosure may be used as a medicament, in particular for the treatment of tumors or cancer diseases. In this case, the treatment preferably involves intratumoral administration, in particular by intratumoral injection. Thus, the RNA nucleic acid molecule, composition or vaccine or kit according to the present disclosure may be used for the preparation of a medicament for the treatment of a tumor or cancer disease, which medicament is particularly suitable for intratumoral application (administration) for the treatment of a tumor or cancer disease.
Preferably, the tumors and cancer diseases mentioned herein are selected from the group of tumors or cancer diseases preferably comprising: such as acute lymphocytic leukemia, acute myelocytic leukemia, adrenocortical carcinoma, AIDS-related cancer, AIDS-related lymphoma, anal cancer, appendiceal cancer, astrocytoma, basal cell carcinoma, cholangiocarcinoma, bladder cancer, bone cancer, osteosarcoma/malignant fibrous histiocytoma, brain stem glioma, brain tumor, cerebellar astrocytoma, brain astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumor, visual pathway and hypothalamic glioma, breast cancer, bronchial adenoma/carcinoid, Burkitt's lymphoma, childhood carcinoid tumor, gastrointestinal carcinoid tumor, unknown primary, primary central nervous system lymphoma, childhood cerebellar astrocytoma, childhood astrocytoma/malignant glioma, cervical cancer, childhood cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, and acute myelogenous leukemia, Chronic myeloid leukemia, chronic myeloproliferative disease, colon cancer, cutaneous T cell lymphoma, desmoplastic small round cell tumor, endometrial cancer, ependymoma, esophageal cancer, Ewing's sarcoma in Ewing's family of tumors, childhood extracranial germ cell tumor, extragonadal germ cell tumor, extrahepatic bile duct cancer, intraocular melanoma, retinoblastoma, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, extracranial, extragonadal or ovarian germ cell tumor, gestational trophoblastoma, brain stem glioma, childhood brain astrocytoma, childhood visual pathway and hypothalamic glioma, gastric carcinoid, hairy cell leukemia, head and neck cancer, heart cancer, hepatocellular carcinoma, Hodgkin's lymphoma, hypopharyngeal cancer, childhood hypothalamic and visual pathway glioma, intraocular melanoma, islet cell carcinoma, Kaposi's sarcoma, cancer, neuroblastoma, melanoma, and tumor associated with renal cell carcinoma of the origin of the, Renal cancer, laryngeal cancer, leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, hairy cell leukemia, lip and oral cancer, liposarcoma, liver cancer, non-small cell lung cancer, lymphoma, AIDS-related lymphoma, Burkitt's lymphoma, cutaneous T cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, primary central nervous system lymphoma, megaglobulinemia, malignant fibrous histiocytoma of bone/osteosarcoma, childhood medulloblastoma, melanoma, intraocular (eye) melanoma, Merck's cell carcinoma, adult malignant mesothelioma, childhood mesothelioma, metastatic squamous neck cancer with occult primary, oral cancer, childhood multiple endocrine tumor syndrome, multiple myeloma/plasmacytoma, mycosis fungoides, sarcoidosis, multiple myeloma/plasmacytoma, multiple myeloma, multiple sclerosis, Myelodysplastic syndrome, myelodysplastic/myeloproliferative disorders, chronic myelogenous leukemia, adult acute myelogenous leukemia, childhood acute myelogenous leukemia, multiple myeloma, chronic myelodysplasia, cancers of the nasal and paranasal sinuses, nasopharyngeal carcinoma, neuroblastoma, oral cancer, oropharyngeal cancer, osteosarcoma/malignant fibrous histiocytoma of the bone, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, pancreatic islet cell pancreatic cancer, paranasal sinuses and nasal cavities, parathyroid cancer, penile cancer, throat cancer, pheochromocytoma, pineal astrocytoma, pineal germ cell tumor, childhood pineal and supratentorial primitive neuroectodermal tumors, pituitary adenoma, plasmacytoma/multiple myeloma, pleuropneumoblastoma, primary central nervous system lymphoma, cervical myelogenous leukemia, neuroblastoma, ovarian cancer, ovarian germ cell tumor, ovarian cancer, pancreatic carcinoma, paranasal sinuses and nasal cavity cancer, parathyroid carcinoma, penile carcinoma, and primary neuroblastoma, Prostate cancer, rectal cancer, renal cell carcinoma, carcinoma of the renal pelvis and ureter, retinoblastoma, rhabdomyosarcoma of children, salivary gland carcinoma, Ewing ' S family sarcoma, Kaposi ' S sarcoma, soft tissue sarcoma, uterine sarcoma, Szary ' S syndrome, skin cancer (non-melanoma), skin cancer (melanoma), Mercury cell skin cancer, small intestine cancer, squamous cell carcinoma, metastatic squamous neck cancer with occult primary, childhood primitive neuroectodermal tumors, testicular cancer, laryngeal cancer, childhood thymoma, thymoma and thymus cancer, thyroid cancer, childhood thyroid cancer, transitional cell carcinoma of the renal pelvis and ureter, gestational trophoblastic tumors, cancer of the urethra, endometrial uterine carcinoma, uterine sarcoma, vaginal cancer, childhood visual pathway and hypothalamic gliomas, vulvar cancer, megaglobulinemia, and childhood Wilms' tumor (renal carcinoma).
Infectious diseases
In a preferred embodiment, the RNA nucleic acid molecule, composition or vaccine or kit is for use in the treatment or prevention of an infectious disease.
The term "infection" or "infectious disease" relates to the invasion and reproduction of microorganisms, such as bacteria, viruses and parasites, which are not normally present in the body. Infection may not cause any symptoms and is subclinical, or may cause symptoms and is clinically significant. The infection may remain localized or may spread through the blood or lymphatic system to become a systemic infection. In this case, the infectious disease preferably includes a viral, bacterial, fungal or protozoan infectious disease.
Autoimmune diseases
In a preferred embodiment, the RNA nucleic acid molecule, composition or vaccine or kit is for use in the treatment or prevention of an autoimmune disease.
The term "autoimmune disease" refers to any disease, disorder or condition in a subject characterized by cellular, tissue and/or organ damage caused by the subject's immune response to its own cells, tissues and/or organs. In general, "autoimmune diseases" are caused or exacerbated by antibodies reactive with self-antigens (i.e., antigens expressed by healthy human cells).
Autoimmune diseases can be classified as systemic symptoms, including but not limited to Systemic Lupus Erythematosus (SLE), sjogren's syndrome, scleroderma, rheumatoid arthritis, and polymyositis; or local syndromes, which may be endocrine (type I diabetes, hashimoto's thyroiditis, addison's disease, etc.), dermatological (pemphigus vulgaris), haematological (autoimmune hemolytic anemia), neurological (multiple sclerosis) or may involve almost any defined body tissue. In this context, the autoimmune disease may be selected from the group consisting of type I autoimmune diseases or type II autoimmune diseases or type III autoimmune diseases or type IV autoimmune diseases, such as Multiple Sclerosis (MS), rheumatoid arthritis, diabetes, type I diabetes (type 1 diabetes), chronic multiple arthritis, goiter, autoimmune forms of chronic hepatitis, ulcerative colitis, type I allergic diseases, type II allergic diseases, type III allergic diseases, type IV allergic diseases, fibromyalgia, alopecia, beheigh's disease, crohn's disease, myasthenia gravis, neurodermatitis, polymyalgia rheumatica, Progressive Systemic Sclerosis (PSS), reiter's syndrome, rheumatoid arthritis, psoriasis, vasculitis and type II diabetes.
Inflammatory diseases
In a preferred embodiment, the RNA nucleic acid molecule, composition or vaccine or kit is for use in the treatment or prevention of an inflammatory disease.
The term "inflammatory disease" refers to any disease, disorder or condition in a subject characterized by, caused by, or associated with inflammation, preferably chronic inflammation. Autoimmune diseases may or may not be associated with inflammation. Moreover, the inflammation may or may not be caused by an autoimmune disease. Thus, certain diseases can be characterized as both autoimmune and inflammatory diseases.
Herein, exemplary inflammatory diseases include, but are not limited to, rheumatoid arthritis, crohn's disease, diabetic retinopathy, psoriasis, endometriosis, alzheimer's disease, ankylosing spondylitis, arthritis (e.g., osteoarthritis, Rheumatoid Arthritis (RA), psoriatic arthritis), asthma, atherosclerosis, colitis, dermatitis, diverticulitis, fibromyalgia, hepatitis, Irritable Bowel Syndrome (IBS), Systemic Lupus Erythematosus (SLE), nephritis, parkinson's disease, and ulcerative colitis.
Allergic reactions
In a preferred embodiment, the RNA nucleic acid molecule, composition or vaccine or kit is for use in the treatment or prevention of allergy.
The term "allergy" or "allergic hypersensitivity" refers to any disease, disorder or condition that is caused by or characterized by hypersensitivity induced by immune mechanisms to allergens, typically in genetically susceptible individuals (atopy). The allergy may be antibody-mediated or cell-mediated. In most patients, the antibodies that normally cause allergic reactions are of the IgE isotype (IgE-mediated allergy, type I allergy). In non-IgE-mediated allergy, the antibody may belong to the IgG isotype. Allergies may be classified according to the source of the antigen causing the allergy. In this context, the allergy may be selected from (a) food allergy, (b) drug allergy, (c) house dust allergy, (d) insect venom or bite allergy, and (e) pollen allergy. Alternatively, allergy may be classified according to its chief symptoms. Herein, the allergy may be selected from (a) asthma, (b) rhinitis, (c) conjunctivitis, (d) rhinoconjunctivitis (rhinocon juctives), (e) dermatitis, (f) urticaria and (g) anaphylaxis.
Combination therapy
The RNA nucleic acid molecules, compositions or vaccines or kits of the present disclosure may also be used in combination therapy. Any other therapy useful for treating or preventing the diseases and disorders defined herein may be combined with the uses and methods disclosed herein.
For example, a subject receiving an RNA nucleic acid molecule, composition or vaccine or kit of the present disclosure can be a patient with cancer or a related disorder receiving chemotherapy (e.g., first-or second-line chemotherapy), radiation therapy, chemoradiation therapy (a combination of chemotherapy and radiation therapy), a tyrosine kinase inhibitor (e.g., an EGFR tyrosine kinase inhibitor), an antibody therapy and/or an inhibitory and/or stimulatory checkpoint molecule (e.g., a CTLA4 inhibitor), or a patient who has achieved partial remission or disease stabilization after receiving one or more of the above treatments. Alternatively, the subject receiving the RNA nucleic acid molecule, composition or vaccine or kit of the present disclosure may be a patient suffering from an infectious disease, preferably as defined herein, receiving an antibiotic, antifungal or antiviral treatment.
Administration of the RNA nucleic acid molecules, compositions, or vaccines or kits of the present disclosure can be performed prior to, concurrently with, and/or subsequent to administration of another therapeutic agent or to subject a patient to another therapy useful for treating a particular disease or condition.
The present disclosure, taking as an example mrnas encoding human basic fibroblast growth factor (hbFGF), human epidermal growth factor (hEGF), and Green Fluorescent Protein (GFP), introduces a novel cell-free in vitro transcription RNA platform or system for producing therapeutic mrnas. In which system one or more of the following advantages may be present: the mRNA produced has high stability; can be easily administered into cells; the translation efficiency is better; has a fast production time (no longer than 1 hour); the yield can reach 2mg/ml/h, even about 2.3 mg/ml/h; the management is easy; and low production cost.
Examples
The disclosure is described herein by way of the following examples, which are intended to be illustrative only and not limiting as to the scope of the disclosure.
Materials and methods:
design of DNA oligonucleotides for RNA in vitro transcription
As shown in fig. 1A, DNA oligonucleotide constructs were designed starting from EcoRI before the T7 promoter. The signal and coding sequences of the gene of interest (hEGF, hbFGF, GFP) were added between the human beta globin 5 'untranslated sequence and the human alpha globin 3' untranslated sequence, respectively. To terminate transcription, a T7 terminator was added to the 5' end of the construct. For universal cloning, SalI and NotI were added to the spacer sequence. DNA oligonucleotides were synthesized by ThermoFisher Scientific. The sequences of the DNA oligonucleotide constructs are shown as SEQ ID NO 5-7 respectively.
SEQ ID NO:5(hEGF)
5’-GGGAGAGTCGACAAATAAGAGAGAAAAGAAGAGTACATTTGCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGCTGCTCACTCTTATCATTCTGTTGCCAGTAGTTTCAAAAAATAGTGACTCTGAATGTCCCCTGTCCCACGATGGGTACTGCCTCCATGATGGTGTGTGCATGTATATTGAAGCATTGGACAAGTATGCATGCAACTGTGTTGTTGGCTACATCGGGGAGCGATGTCAGTACCGAGACCTGAAGTGGTGGGAACTGCGCTGAGCTGGAGCCTCGGTAGCCGTTCCTCCTGCCCGCTGGGCCTCCCAACGGGCCCTCCTCCCCTCCTTGCACCGGCCCTTCCTGGTCTTTGGCGGCCGCCTGCTAACAAAGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGGAACTATATCCGGATGAATTCGC-3’
SEQ ID NO:6(hbFGF)
5’-GGGAGAGTCGACAAATAAGAGAGAAAAGAAGAGTACATTTGCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGGCAGCCGGGAGCATCACCACGCTGCCCGCCTTGCCCGAGGATGGCGGCAGCGGCGCCTTCCCGCCCGGCCACTTCAAGGACCCCAAGCGGCTGTACTGCAAAAACGGGGGCTTCTTCCTGCGCATCCACCCCGACGGCCGAGTTGACGGGGTCCGGGAGAAGAGCGACCCTCACATCAAGCTACAACTTCAAGCAGAAGAGAGAGGAGTTGTGTCTATCAAAGGAGTGTGTGCTAACCGTTACCTGGCTATGAAGGAAGATGGAAGATTACTGGCTTCTAAATGTGTTACGGATGAGTGTTTCTTTTTTGAACGATTGGAATCTAATAACTACAATACTTACCGGTCAAGGAAATACACCAGTTGGTATGTGGCACTGAAACGAACTGGGCAGTATAAACTTGGATCCAAAACAGGACCTGGGCAGAAAGCTATACTTTTTCTTCCAATGTCTGCTAAGAGCTGAGCTGGAGCCTCGGTAGCCGTTCCTCCTGCCCGCTGGGCCTCCCAACGGGCCCTCCTCCCCTCCTTGCACCGGCCCTTCCTGGTCTTTGGCGGCCGCCTGCTAACAAAGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGGAACTATATCCGGATGAATTCGC-3’
SEQ ID NO:7(GFP)
5’-GGGAGAGTCGACAAATAAGAGAGAAAAGAAGAGTACATTTGCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCCTGACCTACGGCGTGCAGTGCTTCAGCCGCTACCCCGACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAAGCTGGAGCCTCGGTAGCCGTTCCTCCTGCCCGCTGGGCCTCCCAACGGGCCCTCCTCCCCTCCTTGCACCGGCCCTTCCTGGTCTTTGGCGGCCGCCTGCTAACAAAGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGGAACTATATCCGGATGAATTCGC-3’
In vitro transcription of RNA
PCR templates for in vitro transcription were prepared by PCR extension using oligonucleotides P1 (5'-AATTCATCCGGATATAGTTC-3' (SEQ ID NO:8)) and P2 (5'-TGTACATAATACGACTCACTAT-3' (SEQ ID NO:9)) as primers and the DNA oligonucleotides designed above as templates. PCR products through New England Biolabs (Epstein, Mass.)
Figure BDA0002884026540000211
PCR&The DNA Cleanup kit was purified and then subjected to standard mRNA synthesis, which was performed as described by New England Biolabs. Mu.g of the purified PCR product was mixed with a premix for in vitro mRNA synthesis [1.5mM ATP, 1.25mM UTP, 1.25mM CTP, 1mM GTP, 4mM 7m G (3' -O-Me) pppG (ARCA), ] 1U/. mu.l RNase inhibitor, 0.4U/. mu. l T7 RNA polymerase, 40mM Tris-HCl (pH 7.9), 6mM MgCl22mM spermidine]And incubated at 37 ℃ for 30 minutes to generate 5' -ARCA-terminated mRNA. Then 0.2U/. mu.l DNase I was added to the reaction mixture and incubated at 37 ℃ for 15 min to remove the PCR template. By additional Poly (A) polymerase reaction mixture [50mM Tris-HCl (pH 8.1), 250mM NaCl, 10mM MgCl20.05U/. mu.l Poly (A) polymerase]Poly (A) tailing was performed and incubated at 37 ℃ for 30 minutes. The final mRNA product was obtained by New England Biolabs (Epstein, Mass.)
Figure BDA0002884026540000221
The clearup kit was purified and stored for analysis. The structure of the obtained mRNA is shown in fig. 1B, and the sequences of the transcribed mRNA are shown in SEQ ID NO: 10-12.
SEQ ID NO:10(hEGF)
5’-GGGAGAGUCGACAAAUAAGAGAGAAAAGAAGAGUACAUUUGCUUCUGACACAACUGUGUUCACUAGCAACCUCAAACAGACACCAUGCUGCUCACUCUUAUCAUUCUGUUGCCAGUAGUUUCAAAAAAUAGUGACUCUGAAUGUCCCCUGUCCCACGAUGGGUACUGCCUCCAUGAUGGUGUGUGCAUGUAUAUUGAAGCAUUGGACAAGUAUGCAUGCAACUGUGUUGUUGGCUACAUCGGGGAGCGAUGUCAGUACCGAGACCUGAAGUGGUGGGAACUGCGCUGAGCUGGAGCCUCGGUAGCCGUUCCUCCUGCCCGCUGGGCCUCCCAACGGGCCCUCCUCCCCUCCUUGCACCGGCCCUUCCUGGUCUUUGGCGGCCGC-3’
SEQ ID NO:11(hbFGF)
5’-GGGAGAGUCGACAAAUAAGAGAGAAAAGAAGAGUACAUUUGCUUCUGACACAACUGUGUUCACUAGCAACCUCAAACAGACACCAUGGCAGCCGGGAGCAUCACCACGCUGCCCGCCUUGCCCGAGGAUGGCGGCAGCGGCGCCUUCCCGCCCGGCCACUUCAAGGACCCCAAGCGGCUGUACUGCAAAAACGGGGGCUUCUUCCUGCGCAUCCACCCCGACGGCCGAGUUGACGGGGUCCGGGAGAAGAGCGACCCUCACAUCAAGCUACAACUUCAAGCAGAAGAGAGAGGAGUUGUGUCUAUCAAAGGAGUGUGUGCUAACCGUUACCUGGCUAUGAAGGAAGAUGGAAGAUUACUGGCUUCUAAAUGUGUUACGGAUGAGUGUUUCUUUUUUGAACGAUUGGAAUCUAAUAACUACAAUACUUACCGGUCAAGGAAAUACACCAGUUGGUAUGUGGCACUGAAACGAACUGGGCAGUAUAAACUUGGAUCCAAAACAGGACCUGGGCAGAAAGCUAUACUUUUUCUUCCAAUGUCUGCUAAGAGCUGAGCUGGAGCCUCGGUAGCCGUUCCUCCUGCCCGCUGGGCCUCCCAACGGGCCCUCCUCCCCUCCUUGCACCGGCCCUUCCUGGUCUUUGGCGGCCGC-3’
SEQ ID NO:12(GFP)
5’-GGGAGAGUCGACAAAUAAGAGAGAAAAGAAGAGUACAUUUGCUUCUGACACAACUGUGUUCACUAGCAACCUCAAACAGACACCAUGGUGAGCAAGGGCGAGGAGCUGUUCACCGGGGUGGUGCCCAUCCUGGUCGAGCUGGACGGCGACGUAAACGGCCACAAGUUCAGCGUGUCCGGCGAGGGCGAGGGCGAUGCCACCUACGGCAAGCUGACCCUGAAGUUCAUCUGCACCACCGGCAAGCUGCCCGUGCCCUGGCCCACCCUCGUGACCACCCUGACCUACGGCGUGCAGUGCUUCAGCCGCUACCCCGACCACAUGAAGCAGCACGACUUCUUCAAGUCCGCCAUGCCCGAAGGCUACGUCCAGGAGCGCACCAUCUUCUUCAAGGACGACGGCAACUACAAGACCCGCGCCGAGGUGAAGUUCGAGGGCGACACCCUGGUGAACCGCAUCGAGCUGAAGGGCAUCGACUUCAAGGAGGACGGCAACAUCCUGGGGCACAAGCUGGAGUACAACUACAACAGCCACAACGUCUAUAUCAUGGCCGACAAGCAGAAGAACGGCAUCAAGGUGAACUUCAAGAUCCGCCACAACAUCGAGGACGGCAGCGUGCAGCUCGCCGACCACUACCAGCAGAACACCCCCAUCGGCGACGGCCCCGUGCUGCUGCCCGACAACCACUACCUGAGCACCCAGUCCGCCCUGAGCAAAGACCCCAACGAGAAGCGCGAUCACAUGGUCCUGCUGGAGUUCGUGACCGCCGCCGGGAUCACUCUCGGCAUGGACGAGCUGUACAAGUAAGCUGGAGCCUCGGUAGCCGUUCCUCCUGCCCGCUGGGCCUCCCAACGGGCCCUCCUCCCCUCCUUGCACCGGCCCUUCCUGGUCUUUGGCGGCCGC-3’
mRNA transfection
Transfection of mRNA was accomplished with lipid nanoparticles. Briefly, DC-cholesterol (3 β - [ N- (N ', N' -dimethylaminoethane) -carbamoyl ] cholesterol hydrochloride) and DOPE (dioleoylphosphatidylethanolamine) were dissolved in chloroform at 25 mg/mL. mu.L of DC-cholesterol was mixed with 80. mu.L of DOPE and dried in a vacuum concentrator for 15 minutes to evaporate chloroform. When the lipids were resuspended in 1mL nuclease-free water and emulsified in a 35kHz ultrasonic bath for 1 hour, then extruded in a mini-extruder.
mu.L of lipid nanoparticles were diluted in 100. mu.L of serum-free DMEM medium, then mixed with 1. mu.g of the obtained mRNA and incubated for 15 minutes at room temperature. The mRNA-lipid nanoparticle complexes were then added to 293T cells to incubate for the indicated time.
Protein detection
At 24 hours post-transfection, 293T cells transfected with GFP mRNA were observed under a Plan Apo 20X/0.75DIC lens under a Nikon Eclipse Ti inverted microscope equipped with an Andor EM-CCD camera. Brightfield and GFP images were taken at the respective filter settings as shown in figure 3. 293T cells transfected with hEGF and hbFGF mRNA were incubated for 24 hours. Cells and media were collected at 0, 2, 4, 6, 8, 24 hours post transfection. Cells were lysed in lysis buffer (PBS + 1% Triton and complete protease inhibitor). Cell lysate samples were analyzed by Western blot for hbFGF and hEGF expression. The results are shown in FIG. 5.
Real-time PCR
Transfected 293T cells were lysed and their total RNA extracted using RNAzol reagent (molecular research center). RNA yield was quantified using a Qubit. 100ug of RNA was reverse transcribed using Oligodt primer with GoScript reverse transcriptase (Promega). cDNA samples were analyzed by pre-designed real-time PCR primers/probes to detect hEGF and hbFGF using a premix of QuantStaudio 3 and SYBR Green I. All samples were run in triplicate. Mean gene expression was calculated in 3 independent experiments. As shown in fig. 2, the in vitro transcription yields of EGF and bFGF mRNA collected at different time points were quantified with Qubit. When in vitro transcription was performed for one hour, the maximum yield reached 2250. mu.g/mL.
As a result:
engineering and in vitro transcription of mRNA
The DNA oligonucleotide consists of the T7 promoter, the human β globin 5 '-untranslated sequence, the signal and coding sequences of the gene of interest (hEGF, hbFGF and GFP), the human α globin 3' -untranslated sequence and the T7 terminator. This oligonucleotide serves as a stable in vitro transcriptional backbone for hEGF, hbFGF and GFP. mRNA was transcribed by T7 polymerase, and anti-reverse cap analogs and polyA tails were added at the 5 'and 3' ends, respectively, to enhance mRNA stability and translation efficiency. Various time points during in vitro transcription were used for analysis. The maximum yield per ml of mRNA was about 2.3mg when the reaction time was 1 hour.
Figure 4 shows the quantification of intracellular EGF and bFGF mRNA in 293T cells by qPCR. 0.5 μ g of EGF and bFGF mRNA encapsulated in lipid nanoparticles was transfected into 293T cells. EGF and bFGF mRNA collected at different time points after transfection was quantified by qPCR. As can be seen from FIG. 4, under standard 293T cell culture conditions, the concentration of EGF and bFGF mRNA remained stable for 8 hours and eventually declined by 24 hours after transfection.
293T cells transfected with GFP-expressing mRNA
293T cells were transfected with lipid nanoparticles encapsulating 0.5ug of GFP expressing mRNA and incubated for 24 hours. Under a fluorescence microscope, more than 85% of the transfected cells showed a fluorescence signal under the GFP filter, as shown in figure 3. mRNA encoding GFP encapsulated by lipid nanoparticles has been successfully transfected into 293T cells. The fluorescence signal of 293T cells transfected with GFP mRNA increased with time.
qPCR detection of hEGF and hbFGF mRNA
293T cells were transfected with lipid nanoparticles encapsulating 0.5ug hEGF mRNA and 0.5ug hbFGF mRNA, respectively. RNA probes with high specificity for hEGF and hbFGF were used to quantify mRNA levels. hEGF and hbFGF mRNA levels remained stable throughout the first 8 hours of culture and only slightly decreased at 24 hours, demonstrating that the mRNA was very stable, as shown in FIG. 4.
Western blotting method for detecting hEGF and hbFGF
293T cells transfected with hEGF and hbFGF mRNA were harvested at 0, 2, 4, 6, 8, 24 hours post-transfection. Cell lysates and cell culture media were prepared and analyzed by western blotting for hEGF and hbFGF antibodies. As shown in fig. 5, expression of hEGF and hbFGF in cell lysates were both significantly increased after 4 hours post-transfection; on the other hand, hEGF and hbFGF levels in cell culture medium were observed and significantly increased after 6 hours post-transfection.
Although various embodiments of RNA in vitro transcription methods and constructs have been described herein in considerable detail, these embodiments are provided merely as non-limiting examples of the disclosure described herein. Accordingly, it will be understood by those skilled in the art that various changes and modifications may be made to the arrangements described in the present disclosure without departing from the spirit of the invention. Indeed, this disclosure is not intended to be exhaustive or to limit the scope of the invention.
Further, in describing representative embodiments, the present disclosure has presented the methods and/or processes of the present invention as a particular sequence of steps. However, the method or process should not be limited to the particular sequence of steps described. Other sequences of steps are possible. Therefore, the particular order of the steps disclosed herein is not to be construed as limitations on the invention. Further, the disclosure of methods and/or processes should not be limited to the performance of their steps in the order described. Such order may vary and still be within the scope of the present invention.
Sequence listing
<110> Mengqian scientific and technological intellectual Property Limited
<120> cell-free and vector-free in vitro RNA transcription method for therapeutic mRNA and nucleic acid molecule
<130> GWHWW204169DI
<141> 2021-01-05
<160> 12
<170> SIPOSequenceListing 1.0
<210> 1
<211> 50
<212> RNA
<213> human (Homo sapiens)
<400> 1
acauuugcuu cugacacaac uguguucacu agcaaccuca aacagacacc 50
<210> 2
<211> 88
<212> RNA
<213> human (Homo sapiens)
<400> 2
gcuggagccu cgguagccgu uccuccugcc cgcugggccu cccaacgggc ccuccucccc 60
uccuugcacc ggcccuuccu ggucuuug 88
<210> 3
<211> 50
<212> DNA
<213> human (Homo sapiens)
<400> 3
acatttgctt ctgacacaac tgtgttcact agcaacctca aacagacacc 50
<210> 4
<211> 88
<212> DNA
<213> human (Homo sapiens)
<400> 4
gctggagcct cggtagccgt tcctcctgcc cgctgggcct cccaacgggc cctcctcccc 60
tccttgcacc ggcccttcct ggtctttg 88
<210> 5
<211> 521
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 5
gggagagtcg acaaataaga gagaaaagaa gagtacattt gcttctgaca caactgtgtt 60
cactagcaac ctcaaacaga caccatgctg ctcactctta tcattctgtt gccagtagtt 120
tcaaaaaata gtgactctga atgtcccctg tcccacgatg ggtactgcct ccatgatggt 180
gtgtgcatgt atattgaagc attggacaag tatgcatgca actgtgttgt tggctacatc 240
ggggagcgat gtcagtaccg agacctgaag tggtgggaac tgcgctgagc tggagcctcg 300
gtagccgttc ctcctgcccg ctgggcctcc caacgggccc tcctcccctc cttgcaccgg 360
cccttcctgg tctttggcgg ccgcctgcta acaaagcccg aaaggaagct gagttggctg 420
ctgccaccgc tgagcaataa ctagcataac cccttggggc ctctaaacgg gtcttgaggg 480
gttttttgct gaaaggagga actatatccg gatgaattcg c 521
<210> 6
<211> 785
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 6
gggagagtcg acaaataaga gagaaaagaa gagtacattt gcttctgaca caactgtgtt 60
cactagcaac ctcaaacaga caccatggca gccgggagca tcaccacgct gcccgccttg 120
cccgaggatg gcggcagcgg cgccttcccg cccggccact tcaaggaccc caagcggctg 180
tactgcaaaa acgggggctt cttcctgcgc atccaccccg acggccgagt tgacggggtc 240
cgggagaaga gcgaccctca catcaagcta caacttcaag cagaagagag aggagttgtg 300
tctatcaaag gagtgtgtgc taaccgttac ctggctatga aggaagatgg aagattactg 360
gcttctaaat gtgttacgga tgagtgtttc ttttttgaac gattggaatc taataactac 420
aatacttacc ggtcaaggaa atacaccagt tggtatgtgg cactgaaacg aactgggcag 480
tataaacttg gatccaaaac aggacctggg cagaaagcta tactttttct tccaatgtct 540
gctaagagct gagctggagc ctcggtagcc gttcctcctg cccgctgggc ctcccaacgg 600
gccctcctcc cctccttgca ccggcccttc ctggtctttg gcggccgcct gctaacaaag 660
cccgaaagga agctgagttg gctgctgcca ccgctgagca ataactagca taaccccttg 720
gggcctctaa acgggtcttg aggggttttt tgctgaaagg aggaactata tccggatgaa 780
ttcgc 785
<210> 7
<211> 1037
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 7
gggagagtcg acaaataaga gagaaaagaa gagtacattt gcttctgaca caactgtgtt 60
cactagcaac ctcaaacaga caccatggtg agcaagggcg aggagctgtt caccggggtg 120
gtgcccatcc tggtcgagct ggacggcgac gtaaacggcc acaagttcag cgtgtccggc 180
gagggcgagg gcgatgccac ctacggcaag ctgaccctga agttcatctg caccaccggc 240
aagctgcccg tgccctggcc caccctcgtg accaccctga cctacggcgt gcagtgcttc 300
agccgctacc ccgaccacat gaagcagcac gacttcttca agtccgccat gcccgaaggc 360
tacgtccagg agcgcaccat cttcttcaag gacgacggca actacaagac ccgcgccgag 420
gtgaagttcg agggcgacac cctggtgaac cgcatcgagc tgaagggcat cgacttcaag 480
gaggacggca acatcctggg gcacaagctg gagtacaact acaacagcca caacgtctat 540
atcatggccg acaagcagaa gaacggcatc aaggtgaact tcaagatccg ccacaacatc 600
gaggacggca gcgtgcagct cgccgaccac taccagcaga acacccccat cggcgacggc 660
cccgtgctgc tgcccgacaa ccactacctg agcacccagt ccgccctgag caaagacccc 720
aacgagaagc gcgatcacat ggtcctgctg gagttcgtga ccgccgccgg gatcactctc 780
ggcatggacg agctgtacaa gtaagctgga gcctcggtag ccgttcctcc tgcccgctgg 840
gcctcccaac gggccctcct cccctccttg caccggccct tcctggtctt tggcggccgc 900
ctgctaacaa agcccgaaag gaagctgagt tggctgctgc caccgctgag caataactag 960
cataacccct tggggcctct aaacgggtct tgaggggttt tttgctgaaa ggaggaacta 1020
tatccggatg aattcgc 1037
<210> 8
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 8
aattcatccg gatatagttc 20
<210> 9
<211> 22
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 9
tgtacataat acgactcact at 22
<210> 10
<211> 384
<212> RNA
<213> Artificial sequence (Artificial sequence)
<400> 10
gggagagucg acaaauaaga gagaaaagaa gaguacauuu gcuucugaca caacuguguu 60
cacuagcaac cucaaacaga caccaugcug cucacucuua ucauucuguu gccaguaguu 120
ucaaaaaaua gugacucuga auguccccug ucccacgaug gguacugccu ccaugauggu 180
gugugcaugu auauugaagc auuggacaag uaugcaugca acuguguugu uggcuacauc 240
ggggagcgau gucaguaccg agaccugaag uggugggaac ugcgcugagc uggagccucg 300
guagccguuc cuccugcccg cugggccucc caacgggccc uccuccccuc cuugcaccgg 360
cccuuccugg ucuuuggcgg ccgc 384
<210> 11
<211> 648
<212> RNA
<213> Artificial sequence (Artificial sequence)
<400> 11
gggagagucg acaaauaaga gagaaaagaa gaguacauuu gcuucugaca caacuguguu 60
cacuagcaac cucaaacaga caccauggca gccgggagca ucaccacgcu gcccgccuug 120
cccgaggaug gcggcagcgg cgccuucccg cccggccacu ucaaggaccc caagcggcug 180
uacugcaaaa acgggggcuu cuuccugcgc auccaccccg acggccgagu ugacgggguc 240
cgggagaaga gcgacccuca caucaagcua caacuucaag cagaagagag aggaguugug 300
ucuaucaaag gagugugugc uaaccguuac cuggcuauga aggaagaugg aagauuacug 360
gcuucuaaau guguuacgga ugaguguuuc uuuuuugaac gauuggaauc uaauaacuac 420
aauacuuacc ggucaaggaa auacaccagu ugguaugugg cacugaaacg aacugggcag 480
uauaaacuug gauccaaaac aggaccuggg cagaaagcua uacuuuuucu uccaaugucu 540
gcuaagagcu gagcuggagc cucgguagcc guuccuccug cccgcugggc cucccaacgg 600
gcccuccucc ccuccuugca ccggcccuuc cuggucuuug gcggccgc 648
<210> 12
<211> 900
<212> RNA
<213> Artificial sequence (Artificial sequence)
<400> 12
gggagagucg acaaauaaga gagaaaagaa gaguacauuu gcuucugaca caacuguguu 60
cacuagcaac cucaaacaga caccauggug agcaagggcg aggagcuguu caccggggug 120
gugcccaucc uggucgagcu ggacggcgac guaaacggcc acaaguucag cguguccggc 180
gagggcgagg gcgaugccac cuacggcaag cugacccuga aguucaucug caccaccggc 240
aagcugcccg ugcccuggcc cacccucgug accacccuga ccuacggcgu gcagugcuuc 300
agccgcuacc ccgaccacau gaagcagcac gacuucuuca aguccgccau gcccgaaggc 360
uacguccagg agcgcaccau cuucuucaag gacgacggca acuacaagac ccgcgccgag 420
gugaaguucg agggcgacac ccuggugaac cgcaucgagc ugaagggcau cgacuucaag 480
gaggacggca acauccuggg gcacaagcug gaguacaacu acaacagcca caacgucuau 540
aucauggccg acaagcagaa gaacggcauc aaggugaacu ucaagauccg ccacaacauc 600
gaggacggca gcgugcagcu cgccgaccac uaccagcaga acacccccau cggcgacggc 660
cccgugcugc ugcccgacaa ccacuaccug agcacccagu ccgcccugag caaagacccc 720
aacgagaagc gcgaucacau gguccugcug gaguucguga ccgccgccgg gaucacucuc 780
ggcauggacg agcuguacaa guaagcugga gccucgguag ccguuccucc ugcccgcugg 840
gccucccaac gggcccuccu ccccuccuug caccggcccu uccuggucuu uggcggccgc 900

Claims (12)

1. An RNA nucleic acid molecule comprising, from the 5 'end to the 3' end, a 5 '-cap structure, a human β -globin 5' untranslated region (5 '-UTR), at least one coding region, a human α -globin 3' untranslated region (3 '-UTR), and a 3' -poly a tail, the at least one coding region operably linked to the 5 '-UTR and the 3' -UTR.
2. The RNA nucleic acid molecule of claim 1, wherein the coding region encodes at least one polypeptide or protein of interest, optionally selected from a growth factor, an antigenic polypeptide or protein, an allergenic polypeptide or protein, a therapeutic polypeptide or protein, or a fragment, variant or derivative of the foregoing; wherein preferably the coding region further encodes at least one selected from the group consisting of: a signal peptide, a peptide tag or protein tag, a targeting signal or targeting sequence, and a peptide linker.
3. The RNA nucleic acid molecule according to claim 1 or 2, wherein the 5' -UTR comprises or consists of a sequence according to SEQ ID NO: 1, or an RNA sequence according to SEQ ID NO: 1, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity; and/or wherein the 3' -UTR comprises or consists of a sequence according to SEQ ID NO: 2, or an RNA sequence according to SEQ ID NO: 2, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity.
4. The RNA nucleic acid molecule according to any of claims 1 to 3, wherein the 5' -cap structure is a 5' -anti-reverse cap analogue (5 ' -ARCA), preferably the 5' -ARCA is 7m G (3 ' -O-Me) pppG.
5. The RNA nucleic acid molecule of any of claims 1-4, wherein the 3' -poly A tail comprises 10 to 200, 20 to 100, 40 to 80, or 50 to 70 adenine nucleotides.
6. A DNA nucleic acid molecule comprising, from the 5 'end to the 3' end, a promoter, a human β -globin 5 '-untranslated region (5' -UTR), at least one coding region, a human α -globin 3 '-untranslated region (3' -UTR), and a transcription terminator, the at least one coding region operably linked to the 5 '-UTR and the 3' -UTR.
7. The DNA nucleic acid molecule according to claim 6, wherein the coding region encodes at least one polypeptide or protein of interest, optionally selected from a peptide growth factor, an antigenic polypeptide or protein, an allergenic polypeptide or protein, a therapeutic polypeptide or protein, or a fragment, variant or derivative of the foregoing; wherein preferably the coding region further encodes at least one selected from the group consisting of: a signal peptide, a peptide tag or protein tag, a targeting signal or targeting sequence, and a peptide linker.
8. The DNA nucleic acid molecule according to claim 6 or 7, wherein the promoter is selected from the group consisting of the T3, T7, Sny5 or SP6 promoter, and/or the promoter is selected from the group consisting of the T3 promoter, and the transcription terminator is selected from the group consisting of the T7 transcription terminator.
9. The DNA nucleic acid molecule according to any one of claims 6-8, wherein the 5' -UTR comprises or consists of a sequence according to SEQ ID NO: 3, or a DNA sequence according to SEQ ID NO: 3, has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity; and/or wherein the 3' -UTR comprises or consists of a sequence according to SEQ ID NO: 4, or a DNA sequence according to SEQ ID NO: 4 has at least 80%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity.
10. An in vitro transcription method comprising the steps of:
(a) providing a DNA nucleic acid molecule according to any one of claims 6-9 as a transcription template;
(b) optionally, amplifying the DNA nucleic acid molecule;
(c) subjecting said DNA nucleic acid molecule to in vitro transcription in the presence of a 5 '-anti-reverse cap analogue (5' -ARCA), preferably 7m G (3 '-O-Me) pppG, to obtain a reaction mixture comprising 5' -ARCA-terminated mRNA;
(d) optionally, removing the transcription template in the reaction mixture comprising 5' -ARCA-terminated mRNA by adding dnase; and
(e) adding a polya polymerase reaction mixture to the reaction mixture comprising 5 '-ARCA-terminated mRNA for 3' -polya tail addition to obtain 5 '-ARCA-terminated mRNA with a 3' -polya tail.
11. A composition comprising an RNA nucleic acid molecule according to any one of claims 1-5 and/or obtained according to the method of claim 10, and a pharmaceutically acceptable carrier and/or excipient; wherein preferably the composition is a pharmaceutical composition or a vaccine or a kit.
12. Use of an RNA nucleic acid molecule according to any one of claims 1 to 5 and/or an RNA nucleic acid molecule obtained according to the method of claim 10 for the manufacture of a medicament for the treatment or prevention of a disease or disorder selected from an immune disease, a genetic disease, a cancer, an infectious disease, an inflammatory disease, an allergy and/or for gene therapy and/or immunomodulation.
CN202110008455.8A 2021-01-05 2021-01-05 Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA Pending CN114717229A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202110008455.8A CN114717229A (en) 2021-01-05 2021-01-05 Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA
PCT/CN2021/130552 WO2022148131A1 (en) 2021-01-05 2021-11-15 Cell-free and vector-free in vitro rna transcription method and nucleic acid molecule for therapeutic mrna

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110008455.8A CN114717229A (en) 2021-01-05 2021-01-05 Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA

Publications (1)

Publication Number Publication Date
CN114717229A true CN114717229A (en) 2022-07-08

Family

ID=82233909

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110008455.8A Pending CN114717229A (en) 2021-01-05 2021-01-05 Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA

Country Status (2)

Country Link
CN (1) CN114717229A (en)
WO (1) WO2022148131A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104220599A (en) * 2012-03-27 2014-12-17 库瑞瓦格有限责任公司 Artificial nucleic acid molecules
CN105940110A (en) * 2014-01-31 2016-09-14 菲克特生物科学股份有限公司 Methods and products for nucleic acid production and delivery
CN109477102A (en) * 2015-02-13 2019-03-15 菲克特生物科学股份有限公司 Nucleic acid product and its method of administration
CN109803977A (en) * 2016-08-17 2019-05-24 菲克特生物科学股份有限公司 Nucleic acid product and its method of administration
US20190351040A1 (en) * 2017-02-01 2019-11-21 Modernatx, Inc. Rna cancer vaccines
US20200149026A1 (en) * 2016-12-08 2020-05-14 Curevac Ag Rnas for wound healing

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2964234A4 (en) * 2013-03-09 2016-12-07 Moderna Therapeutics Inc Heterologous untranslated regions for mrna
EP3687581A1 (en) * 2017-09-29 2020-08-05 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing
WO2019067999A1 (en) * 2017-09-29 2019-04-04 Intellia Therapeutics, Inc. In vitro method of mrna delivery using lipid nanoparticles
CN110714015B (en) * 2019-10-29 2021-03-16 珠海丽凡达生物技术有限公司 mRNA rabies vaccine
CN110974954B (en) * 2019-12-24 2021-03-16 珠海丽凡达生物技术有限公司 Lipid nanoparticle for enhancing immune effect of nucleic acid vaccine and preparation method thereof
CN111218458B (en) * 2020-02-27 2020-11-20 珠海丽凡达生物技术有限公司 mRNAs encoding SARS-CoV-2 virus antigen and vaccine and preparation method of vaccine

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104220599A (en) * 2012-03-27 2014-12-17 库瑞瓦格有限责任公司 Artificial nucleic acid molecules
CN105940110A (en) * 2014-01-31 2016-09-14 菲克特生物科学股份有限公司 Methods and products for nucleic acid production and delivery
CN109477102A (en) * 2015-02-13 2019-03-15 菲克特生物科学股份有限公司 Nucleic acid product and its method of administration
CN109803977A (en) * 2016-08-17 2019-05-24 菲克特生物科学股份有限公司 Nucleic acid product and its method of administration
US20200149026A1 (en) * 2016-12-08 2020-05-14 Curevac Ag Rnas for wound healing
US20190351040A1 (en) * 2017-02-01 2019-11-21 Modernatx, Inc. Rna cancer vaccines

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NANCY D RODGERS等: "Regulated alpha-globin mRNA decay is a cytoplasmic event proceeding through 3\'-to-5\' exosome-dependent decapping", RNA, vol. 8, no. 12, pages 1526 - 1537 *

Also Published As

Publication number Publication date
WO2022148131A1 (en) 2022-07-14

Similar Documents

Publication Publication Date Title
CA2801218C (en) Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
AU2023201630A1 (en) Circular RNA for translation in eukaryotic cells
JP2020182450A (en) Genome editing without nucleases
US11046975B2 (en) Bicistronic expression vector for antibody expression and method for producing antibody using same
Zarghampoor et al. Improved translation efficiency of therapeutic mRNA
CA3128626A1 (en) Circular polyribonucleotides and pharmaceutical compositions thereof
CA3097857A1 (en) Fusion proteins and fusion ribonucleic acids for tracking and manipulating cellular rna
CN114787357A (en) Antisense guide RNA to which functional region is added for editing target RNA
CN103370415A (en) HBV treatment
CN114717238A (en) Methods and nucleic acid molecules for cell-free and vector-free in vitro RNA transcription of epidermal growth factor mRNA
US20220064642A1 (en) Oligomeric nucleic acid molecule and application thereof
US20200215114A1 (en) Compositions and methods for producing exosome loaded therapeutics for the treatment of multiple oncological disorders
Chen et al. A flexible, efficient, and scalable platform to produce circular RNAs as new therapeutics
JP2024503623A (en) Encapsulated RNA polynucleotides and methods of use
CN114717229A (en) Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA
KR20240035691A (en) A composition comprising UTR sequences that enhance intracellular stability and translation of mRNA, and use thereof
CN114717230A (en) Methods and nucleic acid molecules for cell-free and vector-free in vitro RNA transcription of fibroblast growth factor mRNA
WO2020106771A1 (en) Compositions and methods for producing exosome loaded therapeutics for the treatment of multiple oncological disorders
US20240131095A1 (en) Artificial oncolytic viruses and related methods
WO2023182948A1 (en) Internal ribosome entry site (ires), plasmid vector and circular mrna for enhancing protein expression
CN117904198A (en) Method for preparing circular RNA in vitro by using chimeric PIE system and application
CA3180332A1 (en) Artificial oncolytic viruses and related methods
JP2023518734A (en) In vitro transcript mRNA and pharmaceutical composition containing same
WO2021137742A1 (en) Gene-therapy dna vector
CN117683143A (en) mRNA molecule for encoding vascular endothelial growth factor A fusion protein and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40073357

Country of ref document: HK