WO2025164792A1 - High specific activity/high secretion expression-type factor viii variant, nucleic acid encoding high expression-type factor viii, and use thereof - Google Patents
High specific activity/high secretion expression-type factor viii variant, nucleic acid encoding high expression-type factor viii, and use thereofInfo
- Publication number
- WO2025164792A1 WO2025164792A1 PCT/JP2025/003309 JP2025003309W WO2025164792A1 WO 2025164792 A1 WO2025164792 A1 WO 2025164792A1 JP 2025003309 W JP2025003309 W JP 2025003309W WO 2025164792 A1 WO2025164792 A1 WO 2025164792A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- amino acid
- variant
- factor viii
- human factor
- nucleic acid
- 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
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/36—Blood coagulation or fibrinolysis factors
- A61K38/37—Factors VIII
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/46—Ingredients of undetermined constitution or reaction products thereof, e.g. skin, bone, milk, cotton fibre, eggshell, oxgall or plant extracts
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/04—Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/745—Blood coagulation or fibrinolysis factors
- C07K14/755—Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
- C12N15/864—Parvoviral vectors, e.g. parvovirus, densovirus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12P—FERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
- C12P21/00—Preparation of peptides or proteins
- C12P21/02—Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
Definitions
- the present invention relates to human factor VIII variants with high specific activity and high secretory expression, nucleic acids encoding the same, highly expressible human factor VIII-encoding nucleic acids, and uses thereof. More specifically, the present invention relates to human factor VIII variants with enhanced secretory expression efficiency and/or specific activity compared to the wild-type, nucleic acids encoding the same, nucleic acids encoding human factor VIII (including variants) whose expression efficiency has been enhanced by modifying the base sequence, and the use thereof for the treatment of hemophilia A, etc.
- Hemophilia is a disease caused by a genetic deficiency in blood clotting factors involved in hemostasis. According to the World Federation of Hemophilia (WFH), there are approximately 400,000 hemophilia patients worldwide, with approximately 6,000 patients in Japan. There are two types of hemophilia: hemophilia A, which is caused by a deficiency in factor VIII, and hemophilia B, which is caused by a deficiency in factor IX. Hemophilia A patients predominate, with a ratio of 5:1.
- hemophilia A has been treated with factor VIII replacement therapy.
- factor VIII replacement therapy
- the half-life of this clotting factor is short, and patients need to receive intravenous administration two to three times a week from childhood to prevent bleeding.
- factor VIII is administered continuously to hemophilia A patients can develop antibodies against factor VIII, known as inhibitors (25-30%), which is known to reduce the effectiveness of replacement therapy.
- Emicizumab which acts as a substitute for factor VIII, has been launched as a treatment for hemophilia A.
- Emicizumab can treat hemophilia A with subcutaneous injections less frequently (once a month) than factor VIII preparations.
- it is no different from conventional coagulation factor preparations in that it requires continuous administration, and because it is produced using recombinant animal cells, the manufacturing costs are high, placing a significant burden on the medical economy.
- Hemophilia is a hereditary disease caused by a single gene deficiency. Because therapeutic effects can be easily confirmed by measuring clotting factor levels in the blood and because even a relatively small increase in blood levels can be expected to be effective, it has long been considered a good target for gene therapy. Furthermore, preclinical studies have shown that continuous expression of clotting factors through gene therapy can induce immune tolerance via regulatory T cells, potentially eliminating inhibitors.
- AAV adeno-associated virus
- Non-Patent Document 1 In a clinical trial conducted by BioMarin, high factor VIII activity was maintained even after one year in the high-dose administration group (Non-Patent Document 1), but a tendency for blood levels to decrease was observed after three years.
- factor VIII expression from a high-dose AAV vector in liver cells may induce endoplasmic reticulum stress (Non-Patent Document 3).
- Non-Patent Document 2 On the other hand, in Spark's clinical trials, a serotype with high liver tropism was used and a lower dose of AAV vector than BioMarin's was administered, but hepatotoxicity was observed in the 2 x 1012 vg/kg administration group (Non-Patent Document 2). Attempts have been made to reduce the dose and improve safety by achieving high expression through the use of liver-specific promoters and codon optimization of factor VIII, but these approaches are not sufficient in cases where high protein expression itself impairs the long-term stability of the therapeutic effect.
- Non-Patent Document 5 Furthermore, the same group has reported that substituting 12 amino acid residues in the light chain of human factor VIII with canine residues increases specific activity while maintaining wild-type secretion efficiency. Furthermore, Sabatino et al. have disclosed that the specific activity can be increased by substituting or deleting one or more amino acids in the furin recognition site and a3 domain in the human factor VIII light chain with canine forms (Patent Document 2).
- the first object of the present invention is to provide a novel human factor VIII variant and the nucleic acid encoding it that can stably complement factor VIII over the long term in gene therapy for hemophilia A, thereby establishing a gene therapy for hemophilia A with superior long-term efficacy.
- the second object of the present invention is to provide a recombinant factor VIII preparation containing the variant and an mRNA drug encoding the variant.
- the inventors optimized the nucleotide sequence of the nucleic acid encoding the variants for human use using a codon optimization algorithm.
- the resulting nucleotide sequence contained many CpG sequences.
- the inventors predicted that CpG sequences may induce an immune response in the host and inhibit continuous transgene expression, so they performed base substitutions to remove these CpG sequences without changing the amino acid sequence of the variants, and then performed codon optimization again.
- the resulting variant-encoding nucleic acid was ligated downstream of a promoter sequence from which CpG sequences had also been removed, and then incorporated into an AAV vector.
- factor VIII expression was significantly increased compared to the variant-encoding nucleic acid before CpG sequence removal.
- the expression of factor VIII was similarly compared between a codon-optimized nucleic acid encoding wild-type human factor VIII and a nucleic acid that had been further codon-optimized after CpG sequence removal.
- a significant enhancement of expression was observed after CpG sequence removal.
- [Section 1] A human factor VIII variant characterized in that at least one amino acid residue selected from the group consisting of K213, S367 and F2196 of wild-type human factor VIII is replaced with another amino acid, and that the variant has higher specific activity and/or secretory expression efficiency compared to the wild-type variant.
- [Section 2] Item 2. The variant according to Item 1, wherein the amino acid substitution at K213 is K213N or K213H, and/or the amino acid substitution at S367 is S367P, S367N or S367Q, and/or the amino acid substitution at F2196 is F2196L or F2196M.
- [Section 3] Item 3.
- Item 4 The variant according to any one of Items 1 to 3, wherein at least one amino acid residue selected from the group consisting of R-5, P25, A28, L152, M217, W228, Q410, Y487, R489, F501, M539, I566, L603, I642, S727, A736, S1657, Q1659, E1661, I1668, D1681, R1776, H1859, A1993, H2007, N2019, K2085, K2207, F2275, S2296, V2314, Q2316, and M2321 is further substituted with another amino acid.
- the variant according to Item 4 comprising at least one amino acid substitution selected from the group consisting of P, Q1659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N2019K, K2085M, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
- the variant of item 1 further comprising at least one amino acid substitution in the C2 domain selected from the group consisting of K2207Q, Q2316H and M2321L, and optionally F2275L, S2296A and V2314A.
- the variant of item 3 further comprising amino acid substitutions L152P, Y487H and L603P, and optionally further comprising at least one amino acid substitution selected from the group consisting of S727P, Q1659E, H1859R, A1993V, H2007Q, N2019K and K2085M.
- Item 4 The variant according to Item 3, further comprising the amino acid substitutions M539L, I566M, L603P, I642V, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
- Item 11 Item 11.
- the variant of Item 10 further comprising the amino acid substitutions I1668F and D1681G.
- nucleic acid of claim 16 or a nucleic acid encoding wild-type human Factor VIII that has been codon-optimized for human use, which has been further modified to not contain CpG sequences without changing the amino acid sequence that it encodes.
- [Section 18] Item 18.
- An expression vector comprising the nucleic acid according to any one of Items 15 to 17.
- [Section 19] 19.
- the expression vector of claim 18, wherein the nucleic acid is under the control of a liver-specific promoter.
- Proction 20 Item 20.
- Proction 21 Item 21.
- Item 22 The expression vector according to Item 21, wherein the virus is an adeno-associated virus having liver tropism.
- the virus is an adeno-associated virus having liver tropism.
- [Section 24] 24. A method for producing human factor VIII or a variant thereof, comprising culturing the host cell according to Item 23 and recovering human factor VIII or a variant thereof from the resulting culture.
- [Section 25] A pharmaceutical comprising the variant according to any one of Items 1 to 14, the nucleic acid according to any one of Items 15 to 17, the expression vector according to any one of Items 18 to 22, or the host cell according to Item 23.
- Item 26 The pharmaceutical agent according to Item 25, which is for treating hemophilia A.
- a method for treating hemophilia A comprising administering to a patient with hemophilia A an effective amount of the variant of any one of Items 1 to 14, the nucleic acid of any one of Items 15 to 17, the expression vector of any one of Items 18 to 22, or the host cell of Items 23.
- the present invention provides human factor VIII variants with high specific activity and/or secretory expression efficiency, and nucleic acids encoding the same, which, when used, enable the treatment of hemophilia A with stable factor VIII supplementation over a long period of time. Furthermore, the present invention provides human factor VIII-encoding nucleic acids with enhanced protein expression efficiency compared to other nucleic acids encoding the same amino acid sequence.
- the amino acid substitutions of the variants of the present invention are shown below.
- the in vitro (left) and in vivo coagulation factor activities of Ver. 1 to Ver. 10 are shown.
- the in vitro coagulation factor activity of variants such as S3, S5, S7, S4, S6, S8, and S12 is shown (left: one-stage coagulation assay, right: synthetic substrate assay).
- 1 shows the effect of each amino acid substitution contained in the S8 variant on the coagulation factor activity.
- 1 shows the coagulation factor activity of XNPL variants when L152 is substituted with various other amino acids.
- 1 shows the coagulation factor activity of PXPL variants when K213 is substituted with various other amino acids.
- 1 shows the coagulation factor activity of PNXL variants when S367 is substituted with various other amino acids.
- 1 shows the coagulation factor activity of PNPX variants when F2196 is substituted with various other amino acids.
- 1 shows the in vivo coagulation factor activity and blood FVIII antigen levels of various variants.
- 1 shows the intracellular and extracellular localization of wild-type and various mutant FVIII.
- 1 shows the subcellular localization of wild-type and FG mutant FVIII.
- 1 shows the in vitro coagulation factor activity of variant Ver. 12.
- the amino acid substitutions of the variants of the present invention (F8-Tochigi-3, 5, 8, 10, 11, 13, 15) are shown.
- 1 shows the in vitro coagulation factor activity and antigen amount of the variants of the present invention (F8-Tochigi-3, 5, 8, 10, 11, 13, 15, 28).
- 4 shows the in vitro and in vivo expression-enhancing effect of the Ver. 4 variant due to removal of CpG sequences.
- 1 shows the effect of removing CpG sequences on enhancing the expression of wild-type hFVIII in vitro and in vivo.
- an AAV8 vector carrying the Ver. 4 variant with CpG sequences removed can significantly increase factor VIII activity in a cynomolgus monkey model at the dose used in clinical trials.
- the single-chain full-length human factor VIII protein after its 19-amino acid signal peptide is cleaved in the endoplasmic reticulum, consists of three A domains (A1, A2, A3), two C domains (C1, C2), a B domain, and three peptide regions rich in acidic amino acids (a1, a2, a3).
- the present invention provides a human factor VIII variant (hereinafter also referred to as the "variant of the present invention") that has enhanced specific activity and/or secretory expression efficiency compared to the wild-type form.
- the variant of the present invention is characterized in that at least one amino acid residue selected from the group consisting of K213, S367, and F2196 of wild-type human factor VIII is substituted with another amino acid.
- wild-type human factor VIII refers to a protein comprising the amino acid sequence of naturally occurring normal human factor VIII, specifically:
- proteins include (a) a protein consisting of the amino acid sequence represented by SEQ ID NO: 1 (registered in UniProtKB under accession number P00451), or (b) a protein consisting of the amino acid sequence of an allelic variant (minor allele frequency (MAF) less than 1%) or genetic polymorphism (MAF 1% or more; e.g., SNP) of the protein of (a) (e.g., a variant not associated with disease listed in the "Variant" column of UniProtKB under accession number P00451; excluding the amino acid mutations at positions K213, S367, and F2196), which protein exhibits a specific activity and secretory expression efficiency equivalent to that of the protein of (a).
- “equivalent” in specific activity or secretory expression efficiency means that the specific activity or secretory expression efficiency is 0.8 to 1.2 times, preferably 0.9 to 1.1 times, and more preferably 0.95 to 1.05 times that of the protein of (a).
- wild-type human factor VIII is the protein of (a) above.
- variant human factor VIII refers to a protein having an amino acid sequence in which one or more amino acids have been substituted, deleted, inserted, or added in the amino acid sequence (reference sequence) of wild-type human factor VIII (a) or (b) above, and in which the specific activity and/or secretory expression efficiency has been substantially altered compared to human factor VIII consisting of the reference sequence.
- substantially altered preferably means a statistically significant change, but also encompasses cases in which there is a tendency for an increase or decrease even if there is no significant difference.
- the variants of the present invention are human factor VIII variants that have higher specific activity and/or secretory expression efficiency compared to the wild-type.
- the specific activity of human factor VIII can be evaluated by measuring the coagulation factor activity using the publicly known one-stage coagulation assay (OSA) or chromogenic assay (CSA), measuring the protein amount using a publicly known protein quantification method (e.g., immunoassay such as ELISA), and calculating the coagulation factor activity per unit protein amount.
- OSA publicly known one-stage coagulation assay
- CSA chromogenic assay
- a publicly known protein quantification method e.g., immunoassay such as ELISA
- the secretory expression efficiency of human factor VIII can also be evaluated by measuring the concentration of human factor VIII in the culture supernatant of cells transfected with a nucleic acid encoding it or in the plasma of animals administered the nucleic acid using a publicly known method (e.g., immunoassay such as ELISA).
- a publicly known method e.g., immunoassay such as ELISA.
- the amino acid residues corresponding to K213, S367, and F2196 can be identified as the amino acid residues corresponding to K213, S367, and F2196 in SEQ ID NO: 1, respectively, when the amino acid sequence of (b) and the amino acid sequence of SEQ ID NO: 1 are appropriately aligned using any homology search algorithm.
- the other amino acids are not particularly limited, so long as the human factor VIII variant obtained as a result of the amino acid substitution has enhanced specific activity and/or secretory expression efficiency compared to the wild-type.
- the amino acid substitution at K213 is preferably K213N or K213H, with K213N being more preferred.
- the amino acid substitution at S367 is preferably S367P, S367N or S367Q, with S367P being more preferred.
- the amino acid substitution at F2196 is preferably F2196L or F2196M, with F2196L being more preferred.
- the variant of the present invention comprises the amino acid substitutions K213 N , S367 P , and F2196 L in wild-type human factor VIII (hereinafter, sometimes referred to as the "NPL mutation" after the substituted amino acid residues).
- NPL mutation wild-type human factor VIII
- a variant having the NPL mutation is characterized by a higher specific activity than the wild-type, provided that there are no other mutations (combinations of mutations) that would negate the effect of the NPL mutation.
- a high-specific-activity variant can achieve coagulation activity equivalent to or greater than that achieved by overexpressing wild-type factor VIII at an appropriate protein expression level that does not impair the long-term stability of the therapeutic effect, and is therefore useful for gene therapy for hemophilia A.
- amino acid residues at positions other than K213, S367 and F2196 may be substituted with other amino acids.
- substitution sites include R-5, P25, A28, L152, M217, W228, Q410, Y487, R489, F501, M539, I566, L603, I642, S727, A736, S1657, Q1659, E1661, I1668, D1681, R1776, H1859, A1993, H2007, N2019, K2085, K2207, F2275, S2296, V2314, Q2316, and M2321. Accordingly, in one embodiment, the variant of the present invention is one in which at least one of the amino acid residues is further substituted with another amino acid.
- the other amino acids are not particularly limited as long as the human factor VIII variant obtained as a result of the amino acid substitution still has enhanced specific activity and/or secretory expression efficiency compared to the wild-type, but are preferably R-5P, P25H, A28T, L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q or L152V, M217T, W228Q, Q410L, Y487H, and at least one amino acid substitution selected from the group consisting of R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N2019K, K2085M, K2207Q, F2275L, S2296A, V2314A, Q2316H,
- the other amino acid substitution is: (i) L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q, or L152V, and/or (ii) I1668F and D1681G Examples include:
- a particularly preferred embodiment of the present invention is a variant that contains the amino acid substitution (i) above in addition to the NPL mutation (hereinafter, this may be referred to as an "XNPL mutation," and when L152 is substituted with proline (P), this may be referred to as a "PNPL mutation").
- XNPL mutations including PNPL mutations, not only have a higher specific activity than the wild-type, but can also have the property of being secreted at a higher level than the wild-type. Therefore, in addition to the advantages conferred by the NPL mutation, these variants may have the further advantage of suppressing the induction of endoplasmic reticulum stress by improving secretion efficiency, thereby realizing long-term stability of the therapeutic effects of human factor VIII.
- Another preferred embodiment of the present invention is a variant that includes the amino acid substitution (ii) above (hereinafter sometimes referred to as "FG mutation") in addition to the NPL mutation or XNPL mutation.
- FG mutation amino acid substitution
- the variant with the FG mutation exhibits significantly reduced intracellular retention of the light chain, and therefore further improvements in secretion efficiency and/or heavy and light chain reconstitution can be observed.
- the variant of the present invention is the amino acid substitution at K213 is K213N; M539L, and optionally at least one amino acid substitution in the A2 domain selected from the group consisting of I566M, L603P, and I642V;
- the variant further comprises at least one amino acid substitution in the C2 domain selected from the group consisting of K2207Q, Q2316H, and M2321L, and optionally F2275L, S2296A, and V2314A.
- the variant can be conferred the property of high secretory expression compared to wild-type human factor VIII, even without the NPL mutation.
- the secretory expression efficiency of the variant can be further enhanced by further including the FG mutation.
- the variant of the present invention further comprises, in addition to the NPL mutation, amino acid substitutions L152P, Y487H, and L603P, and optionally at least one amino acid substitution selected from the group consisting of S727P, Q1659E, H1859R, A1993V, H2007Q, N2019K, and K2085M. Because the variant contains the PNPL mutation, it has a higher specific activity and is expressed at a higher level of secretion compared to the wild-type.
- the variant of the present invention further comprises the following amino acid substitutions in addition to the NPL mutation: M539L, I566M, L603P, I642V, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
- the high specific activity characteristic of the NPL mutation may be attenuated by the presence of additional amino acid mutations including the 10 amino acid substitutions
- the secretory expression efficiency of the variant is enhanced regardless of the presence or absence of an amino acid substitution at L152 (e.g., L152P) (i.e., even without the XNPL mutation).
- the secretory expression efficiency of the variant can be further enhanced by further including the FG mutation.
- Preferred embodiments of the present invention include modified human factor VIII having the following amino acid substitutions (a) to (n) in wild-type human factor VIII: (a) K213N, S367P, and F2196L (b) K213N, S367P, F2196L, I1668F, and D1681G (c) K213N, M539L, I566M, I642V, K2207Q, F2275L, Q2316H, and M2321L (d) K213N, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, Q2316H, and M2321L (e) K213N, M539L, I566M, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L (f) K213N, M539L, I566M, I
- the B domain of human factor VIII does not contain regions that interact with various proteins and does not contribute to cofactor activity in circulating blood; therefore, deletion of the B domain retains coagulation factor activity. Therefore, the variants of the present invention may lack the B domain.
- B domain deletion refers not only to the complete deletion of the amino acid sequence of the B domain (positions 741-1648), but also to the retention of an amino acid sequence of approximately 1-15 amino acid residues at the N-terminus and/or C-terminus of the B domain between the a2 and a3 regions.
- B domain-deleted (BDD) human factor VIII variants are advantageous when using AAV vectors in gene therapy for hemophilia A.
- AAV vectors can carry a small nucleic acid size of 4.7 kbp
- full-length human factor VIII consists of 2351 amino acids, so the coding region alone exceeds 7 kbp and cannot be carried by AAV vectors.
- the coding sequence for BDD-human factor VIII is less than 4.4 kbp, and the entire expression cassette, including the promoter sequence, fits within the packaging size of an AAV vector.
- the variant of the present invention lacks positions 744 to 1637 of the B domain (corresponding to positions 763 to 1656 in the amino acid sequence represented by SEQ ID NO: 1) (i.e., S743 is linked to Q1638; this may be referred to as "FVIII SQ" or "SQ").
- BDD-human factor VIII that leads to efficient human factor VIII processing is known, other than FVIII SQ, including deletions of the B domain that leave portions of both termini of the B domain, or substitutions of the B domain with three or four arginine (R) residues (e.g., Lind et al., Eur. J. Biochem. 232, 19-27 (1995)), and these can also be used in the present invention.
- the variants of the present invention may contain all or part of the B domain in order to improve secretion efficiency.
- LV lentivirus
- AdV adenovirus
- the variants of the present invention may be expressed in the animal to be treated by administering a nucleic acid encoding them in vivo or ex vivo to the animal, or they may be recombinant proteins obtained by introducing the nucleic acid into suitable host cells, allowing it to be expressed, and culturing the host cells. In either case, the variants of the present invention are produced based on the nucleic acid that encodes them, and the present invention also provides nucleic acids encoding any of the above variants of the present invention.
- nucleic acid encoding a human factor VIII variant used in the present invention may be DNA or RNA, or may be a DNA/RNA chimera.
- the nucleic acid may be double-stranded or single-stranded. If double-stranded, it may be double-stranded DNA, double-stranded RNA, or a DNA:RNA hybrid. If single-stranded, it may be a sense strand (i.e., coding strand) or an antisense strand (i.e., non-coding strand).
- the type of nucleic acid can be appropriately selected depending on its application (such as the type of vector used), but is preferably DNA, more preferably double-stranded DNA.
- the nucleic acid may also be a physiologically acceptable salt with an acid or base, for example, a physiologically acceptable acid addition salt.
- a physiologically acceptable acid addition salt e.g., salts with inorganic acids (e.g., hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or salts with organic acids (e.g., acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) are used.
- DNA encoding human factor VIII variants includes genomic DNA, cDNA (cRNA) derived from cells or tissues of humans or other mammals, and synthetic DNA (RNA).
- nucleic acids encoding variants of the present invention include any nucleotide sequence encoding a variant amino acid sequence having any of the above-mentioned amino acid substitutions in the amino acid sequence of wild-type human factor VIII represented by SEQ ID NO: 1, or in the amino acid sequence of a natural allelic variant or genetic polymorphism (e.g., SNP, etc.) of a protein consisting of said amino acid sequence.
- the nucleotide sequence is codon-optimized for expression in the host cell to be used, preferably a human cell. During gene expression, converting the nucleotide sequence to codons frequently used in the host organism can be expected to increase the amount of protein expression.
- codon usage in the host to be used can be obtained, for example, from the genetic code usage database published on the website of the Kazusa DNA Research Institute (http://www.kazusa.or.jp/codon/index.html), or references listing codon usage in each host can be referenced.
- codon optimization can be performed using a publicly known codon optimization algorithm (e.g., GeneArt Codon Optimizer).
- GeneArt Codon Optimizer Such algorithms can take into account multiple parameters, such as GC content, removal of destabilizing RNA elements, removal of cryptic splice sites, removal of intragenic polyA sites, removal of repetitive sequences, avoidance of RNA secondary structures, and removal of IRES, in addition to frequency of codon usage in the host.
- DNA encoding the desired variant of the present invention can be constructed by chemically synthesizing a DNA strand with the codon-optimized sequence obtained as described above, or by connecting chemically synthesized, partially overlapping short oligo-DNA strands using PCR or Gibson Assembly.
- the nucleic acid encoding the variant of the present invention is a wild-type human factor VIII consisting of the amino acid sequence represented by SEQ ID NO: 1, wherein (n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P,
- An example of such a nucleic acid is a nucleic acid consisting of the nucleotide sequence represented by SEQ ID NO: 2, which is codon-optimized for human use and encodes a variant having the amino acid substitutions Q1659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N2019K, K2085M, F2196L, K2207Q,
- a nucleotide sequence codon-optimized for a target host may contain one or more CpG sequences within the sequence. However, it is predicted that CpG sequences may induce an immune response in the host and inhibit continuous transgene expression. Therefore, by making base substitutions to remove these CpG sequences without changing the amino acid sequence of the variant, and then re-optimizing the variant, the expression efficiency within host cells can be significantly increased.
- nucleic acid encoding a variant codon-optimized for humans consisting of the nucleotide sequence represented by SEQ ID NO: 2, can be modified so that it does not contain any CpG sequences without changing the amino acid sequence it encodes.
- An example of such a nucleotide sequence from which CpG has been removed and which has been re-codon-optimized is the nucleotide sequence represented by SEQ ID NO: 3.
- the present invention also provides a human factor VIII-encoding nucleic acid comprising a nucleotide sequence obtained by modifying a nucleic acid encoding human factor VIII that has been codon-optimized for a target host, preferably for humans, so that it does not contain CpG sequences without changing the amino acid sequence it encodes, and then further codon-optimizing the nucleotide sequence.
- nucleotide sequence that has been codon-optimized for humans and encodes wild-type human factor VIII consisting of the amino acid sequence represented by SEQ ID NO: 1 is the nucleotide sequence represented by SEQ ID NO: 4.
- nucleotide sequence represented by SEQ ID NO: 5 An example of a nucleotide sequence obtained by modifying the sequence so that it does not contain CpG sequences without changing the amino acid sequence it encodes, and then further codon-optimizing the nucleotide sequence for humans is the nucleotide sequence represented by SEQ ID NO: 5.
- a nucleic acid encoding a variant of the present invention, or a nucleic acid encoding human factor VIII whose expression efficiency in host cells has been enhanced by codon optimization and removal of CpG sequences (hereinafter collectively referred to as the "nucleic acid of the present invention") can be linked downstream of a promoter functional in liver cells and inserted into a vector to construct an expression vector that can be expressed in liver cells.
- liver is composed of hepatic parenchymal cells (hepatocytes), which are responsible for the liver's main functions, such as bile production and metabolism, as well as non-parenchymal liver cells such as hepatic sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, pit cells, bile duct epithelial cells, and mesothelial cells.
- hepatic parenchymal cells hepatic parenchymal cells
- non-parenchymal liver cells such as hepatic sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, pit cells, bile duct epithelial cells, and mesothelial cells.
- liver cells refers to one or more of the above cell groups that make up the liver (including cancer cells and cell lines derived from them).
- a "promoter functional in liver cells” refers to a promoter capable of inducing transcription of a gene (a nucleic acid encoding a protein) linked downstream in liver cells.
- a promoter capable of inducing transcription of a gene (a nucleic acid encoding a protein) linked downstream in liver cells.
- the promoter function specifically in liver cells (inducing transcription of a downstream gene).
- the terms “specifically in liver cells” and “liver-specific” are used to encompass not only cases in which expression is limited to liver cells, but also cases in which expression in liver cells is significantly higher than expression in cells of other organs or tissues.
- substantially liver-specific gene expression e.g., to the extent that it does not cause undesirable side effects
- the promoter may further contain other regulatory sequences (e.g., endogenous proximal or distal enhancer sequences, enhancer sequences derived from other genes, etc.).
- Promoters functional in liver cells are not particularly limited as long as they can induce transcription of downstream genes in liver cells. However, they are preferably promoters of genes highly expressed in the liver, and more preferably liver-specific promoters. Here, “liver-specific" has the same meaning as above. Examples of promoters functional in liver cells that can be used in the expression vectors of the present invention include, but are not limited to, the transthyretin (TTR) promoter, the ⁇ 1-antitrypsin (AAT) promoter, the albumin promoter, the ⁇ -fetoprotein promoter, and the thyroxine-binding globulin promoter.
- TTR transthyretin
- AAT ⁇ 1-antitrypsin
- albumin promoter the albumin promoter
- ⁇ -fetoprotein promoter the ⁇ -fetoprotein promoter
- thyroxine-binding globulin promoter the thyroxine-binding globulin promoter.
- the TTR promoter or the AAT promoter can be used as a promoter functional in liver cells.
- a chimeric promoter composed of the human AAT promoter and the liver-type regulatory region (HCR) of the Apo E/C1 gene can also be used as a promoter functional in liver cells.
- Promoters functional in liver cells can be obtained by known methods based on the sequence information of the gene (genomic DNA) from which each promoter is derived. Such sequence information is registered in publicly available databases such as NCBI, EMBL, FASTA, and DDBJ, and is also described in literature. Those skilled in the art can easily access this sequence information.
- nucleotide sequence of a promoter functional in liver cells contains one or more CpG sequences
- the wild-type mouse TTR promoter consists of the nucleotide sequence represented by SEQ ID NO: 6
- a modified promoter can be used in which the CpG sequences in the wild-type mouse TTR promoter have been removed, and the nucleotide sequence represented by SEQ ID NO: 7.
- the linking of a promoter functional in liver cells to the nucleic acid of the present invention can be carried out by methods known in the art. For example, if the promoter and nucleic acid fragments each have blunt ends, the two fragments can be ligated using DNA ligase. Alternatively, any adapter sequence can be added to the ends of both fragments, treated with an appropriate restriction enzyme to generate sticky ends, and then the two can be ligated.
- the nucleic acid of the present invention linked downstream of a promoter functional in liver cells is preferably inserted into a vector in the form of an expression cassette further linked downstream of a transcription termination signal functional in liver cells, i.e., a polyA addition signal (e.g., an SV40 polyA addition signal).
- a polyA addition signal e.g., an SV40 polyA addition signal
- a mouse TTR promoter (SEQ ID NO: 7) that has been modified so as not to contain a CpG sequence, (ii) in wild-type human factor VIII, (n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1659E, E1661K, I1668F, D1681G, R177
- An expression cassette that encodes a variant having the amino acid substitutions 6K, H1859R, A1993V, H2007Q, N2019K, K2085M, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L, that has been codon-optimized for human use
- An example of such an expression cassette is a nucleic acid consisting of the nucleotide sequence (4814 bp) set forth in SEQ ID NO: 8.
- This nucleic acid contains a 17-nucleotide 5'-upstream sequence (positions 283-299) between the promoter sequence (positions 1-282) of SEQ ID NO: 7 and the coding sequence (positions 300-4673) of SEQ ID NO: 3.
- SEQ ID NO: 9 shows the nucleotide sequence (4803 bp) in which these 11 nucleotides have been deleted from the nucleotide sequence of SEQ ID NO: 8.
- SEQ ID NO: 10 shows the nucleotide sequence (4790 bp) in which 13 nucleotides from the 3' end of the SV40 polyA addition signal sequence (positions 4663-4803) of SEQ ID NO: 9 have been deleted
- SEQ ID NO: 11 shows the nucleotide sequence (4768 bp) in which 35 nucleotides from the 3' end have been deleted.
- the vector into which the nucleic acid of the present invention is inserted is not particularly limited as long as it is one generally used in gene therapy.
- suitable vectors include viral vectors such as AAV vectors, LV vectors, retroviral vectors, AdV vectors, Sindbis virus vectors, rabies virus vectors, Sendai virus vectors, and herpes simplex virus vectors, as well as non-viral vectors such as plasmids for animal cells.
- AAV and LV vectors are preferred from the viewpoints of high gene transfer and expression efficiency, ability to transfer to non-dividing cells, and long-term expression of the introduced gene.
- AAV vectors are more preferred from the viewpoints of low frequency of chromosomal integration, elimination of the risk of insertional mutagenesis, low immunogenicity, and high safety.
- AAV vectors are diluted in dividing cells as the cells proliferate, but adult hepatocytes rarely divide, so expression of the introduced gene can be maintained for a long period of time.
- the gene size that can be carried is small at 4.7 kbp, depending on the size of the nucleic acid to be introduced, it may be preferable to use LV or AdV vectors, which can carry larger inserts.
- an AAV vector when using an AAV vector as a vector, it is desirable to use a vector derived from a serotype with high liver tropism.
- serotypes with liver tropism include AAV1, AAV2, AAV3, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAVrhlO, AAV-DJ, and AAV-DJ/8.
- the desired expression vector can be easily constructed using methods such as those described in "Adenovirus Technique," R. Freshney eds., 2nd ed. (1987), Wiley-Liss; Frank L. Graham, Manipulation of adenovirus vectors, Chapter 11, pp. 109-128; E.J. Murray eds., Methods in Molecular Biology, Vol. 7, Gene Transfer and Expression Protocols (1991); Chen, S-H. et al., Combination gene therapy for liver metastases of colon carcinoma in vivo, Proc. Natl. Acad. Sci. USA (1995) 92, 2477-2581.
- the synthetic promoter of the present invention is first inserted between the 5'- and 3'-ITRs of AAV into a plasmid that can be amplified in a suitable host cell (e.g., Escherichia coli, Bacillus subtilis, yeast, etc.).
- a suitable host cell e.g., Escherichia coli, Bacillus subtilis, yeast, etc.
- suitable host cell e.g., Escherichia coli, Bacillus subtilis, yeast, etc.
- E. coli-derived plasmids e.g., pBR322, pBR325, pUC12, pUC13
- Bacillus subtilis-derived plasmids e.g., pUB110, pTP5, pC194
- yeast-derived plasmids e.g., pSH19, pSH15.
- AAV ITRs derived from the desired serotype may be used, AAV2-derived ITRs are commonly used.
- the synthetic promoter of the present invention alone can be inserted between the two ITR sequences, it is preferable to insert a polyA addition signal (e.g., an SV40 polyA addition signal) downstream thereof, and a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) can also be inserted between the synthetic promoter and the polyA addition signal.
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- a shortened WPRE (0.25 kb; Mol. Brain, 7:17, 2014) can also be used.
- Inserting one or more restriction enzyme recognition sites, preferably a multicloning site (MCS), between the promoter of the present invention and the polyA addition signal (or between the promoter and WPRE if the WPRE is included) can facilitate insertion of the nucleic acid of the present invention (construction of an expression cassette for human factor VIII).
- MCS multicloning site
- an expression cassette for human factor VIII is constructed in the plasmid.
- the expression vector can further contain a 5'-UTR downstream of the promoter that can function in the host, and a 3'-UTR downstream of the DNA encoding human factor VIII that can function in the host. It may also contain an enhancer, a splicing signal, etc. Each of these components can be made using known components.
- An AAV vector can be produced from an AAV vector expression plasmid containing the nucleic acid of the present invention downstream of a promoter functional in liver cells by methods known per se, such as the plasmid transfection method, recombinant baculovirus method, recombinant herpes virus vector method, yeast method, etc.
- the AAV vector expression plasmid, a plasmid containing the AAV Rep and Cap genes, and a pHelper plasmid containing the adenovirus-derived E2A, E4orf6, and VARNA genes are transfected into HEK293 cells or the like to produce AAV virus particles.
- the Rep gene does not need to be derived from the desired serotype; the Rep gene from AAV2 is generally used.
- the Cap gene must be derived from the desired serotype, and the cell tropism of the AAV serotype is determined by the capsid protein encoded by the Cap gene. That is, in the expression vector of the present invention that targets liver cells, Cap genes derived from liver-tropic viruses such as AAV1, AAV2, AAV3, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAVrhlO, AAV-DJ, and AAV-DJ/8 can be used as Cap genes.
- the expression vector when a non-viral vector is used as the expression vector of the present invention, the expression vector can be introduced using a polymeric carrier such as a poly-L-lysine-nucleic acid complex, or by encapsulation in a liposome.
- a polymeric carrier such as a poly-L-lysine-nucleic acid complex
- Liposomes are capsules made of phospholipids with a particle size of several tens to several hundreds of nanometers, and a plasmid vector can be encapsulated inside them.
- An expression vector containing a nucleic acid of the present invention downstream of a promoter functional in liver cells can be administered to a subject suffering from a disease for which human factor VIII exerts a therapeutic effect, particularly hemophilia A, to treat the disease. Therefore, the present invention also provides a gene therapy agent for hemophilia A comprising the expression vector of the present invention.
- the gene therapy agent of the present invention may be the expression vector of the present invention used as is, or, if necessary, may be mixed with a pharmacologically acceptable carrier to form various formulations, such as injections, and then used as a medicine.
- organic or inorganic carrier substances commonly used as pharmaceutical ingredients are used as pharmacologically acceptable carriers, and are incorporated as solvents, solubilizers, suspending agents, isotonicity agents, buffers, soothing agents, etc. in liquid formulations.
- formulation additives such as preservatives, antioxidants, and coloring agents can also be used as needed.
- solvents include water for injection, physiological saline, Ringer's solution, alcohol, propylene glycol, polyethylene glycol, sesame oil, corn oil, olive oil, cottonseed oil, etc.
- solubilizing agents include polyethylene glycol, propylene glycol, D-mannitol, trehalose, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate, sodium salicylate, and sodium acetate.
- suspending agents include surfactants such as stearyl triethanolamine, sodium lauryl sulfate, lauryl aminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glycerin monostearate; hydrophilic polymers such as polyvinyl alcohol, polyvinylpyrrolidone, sodium carboxymethylcellulose, methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, and hydroxypropylcellulose; polysorbates; and polyoxyethylene hydrogenated castor oil.
- surfactants such as stearyl triethanolamine, sodium lauryl sulfate, lauryl aminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glycerin monostearate
- hydrophilic polymers such as polyvinyl alcohol, polyvinylpyrrolidone, sodium carboxymethylcellulose, methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, and hydroxy
- Suitable examples of isotonic agents include sodium chloride, glycerin, D-mannitol, D-sorbitol, glucose, etc.
- buffering agents include buffer solutions such as phosphate, acetate, carbonate, and citrate.
- Suitable examples of soothing agents include benzyl alcohol.
- Suitable examples of preservatives include parahydroxybenzoic acid esters, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, and sorbic acid.
- antioxidants include sulfites and ascorbates.
- coloring agents include water-soluble food tar dyes (e.g., food dyes such as Food Red No. 2 and No. 3, Food Yellow No. 4 and No. 5, and Food Blue No. 1 and No. 2), water-insoluble lake dyes (e.g., aluminum salts of the above-mentioned water-soluble food tar dyes), and natural dyes (e.g., ⁇ -carotene, chlorophyll, red iron oxide, etc.).
- water-soluble food tar dyes e.g., food dyes such as Food Red No. 2 and No. 3, Food Yellow No. 4 and No. 5, and Food Blue No. 1 and No. 2
- water-insoluble lake dyes e.g., aluminum salts of the above-mentioned water-soluble food tar dyes
- natural dyes e.g., ⁇ -carotene, chlorophyll, red iron oxide, etc.
- the dosage form of the pharmaceutical composition may be, for example, an injection (e.g., subcutaneous injection, intravenous injection, intramuscular injection, intraperitoneal injection, etc.), a drip infusion, or other parenteral preparation.
- an injection e.g., subcutaneous injection, intravenous injection, intramuscular injection, intraperitoneal injection, etc.
- a drip infusion e.g., a drip infusion, or other parenteral preparation.
- the gene therapy agent of the present invention can be produced by methods commonly used in the pharmaceutical technology field, such as the methods described in the Japanese Pharmacopoeia.
- the content of the viral vector, which is the active ingredient in the formulation varies depending on the dosage form, the dose of the active ingredient, etc., but is, for example, about 0.1 to 100% by weight.
- the viral titer can be appropriately adjusted to, for example, about 1 x 10 10 to 10 13 vp/mL, but is not limited to this range.
- Preparations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions, which may contain antioxidants, buffers, bacteriostats, isotonicity agents, etc. Also included are aqueous and non-aqueous sterile suspensions, which may contain suspending agents, solubilizers, thickeners, stabilizers, preservatives, etc.
- the most preferred dosage form in the present invention is an injection solution.
- the dosage of the formulation varies depending on the type of vector, promoter activity, administration route, severity of the disease, target animal species, drug tolerance, body weight, age, etc. of the target animal.
- an AAV vector e.g., AAV8 with the HCRhAAT promoter as a basal promoter
- the AAV vector when the AAV vector is administered systemically, particularly via a peripheral vein, it can be administered at a single dose of, for example, about 5 ⁇ 10 12 to about 5 ⁇ 10 13 vp/kg body weight.
- the gene therapy agent of the present invention can be systemically administered at a dose of about 1 ⁇ 10 10 to about 5 ⁇ 10 13 vp/kg body weight, preferably about 1 ⁇ 10 11 to about 1 ⁇ 10 13 vp/kg body weight, more preferably about 5 ⁇ 10 11 to about 5 ⁇ 10 12 vp/kg body weight or about 1 ⁇ 10 12 to about 1 ⁇ 10 13 vp/kg body weight, in terms of the amount of AAV vector.
- the present invention also provides a method for expressing human factor VIII in liver cells, which comprises introducing into liver cells in vitro a gene therapy agent containing the expression vector of the present invention or the nucleic acid of the present invention.
- a gene therapy agent containing the expression vector of the present invention or the nucleic acid of the present invention.
- the nucleic acid of the present invention is single-stranded RNA
- the nucleic acid can be formulated in the form of an mRNA pharmaceutical.
- the expression vector is introduced into a suitable host (e.g., mammalian cells) and cultured, and the mRNA is recovered using a method known per se (e.g., the LiCl method), and the mRNA encoding human factor VIII can be purified to obtain the mRNA.
- the mRNA can be obtained by excising the human factor VIII coding sequence (including the 5'- and 3'-UTRs in addition to the CDS) from the expression vector and using it as a template to convert it into mRNA encoding this human factor VIII using a known in vitro transcription system. More specifically, the mRNA coding region is excised using an appropriate restriction enzyme and a phage (T7, T3, SP6, etc.) promoter is ligated to the 5' end, or a fragment of the mRNA coding region linked to the phage promoter is obtained by using the expression vector as a template and performing PCR with primers containing the phage promoter sequence.
- a phage T7, T3, SP6, etc.
- the resulting DNA fragment can be used as a template to react with phage (T7, T3, SP6, etc.) RNA polymerase to synthesize mRNA encoding human factor VIII in vitro.
- phage T7, T3, SP6, etc.
- NTP RNA monomer
- modified NTPs with base substitutions that have been reported to avoid natural immunity (e.g., inosine triphosphate instead of ATP, 5-methylcytidine triphosphate instead of CTP, etc.) can also be used for other NTPs.
- the 5'-cap structure can be achieved by adding a Cap 0 structure after mRNA synthesis using a capping enzyme, and then converting it to a Cap 1 structure using an mRNA 2'-O-methyltransferase.
- transcription and 5'-capping can be performed simultaneously by adding an RNA cap analog (e.g., 3'-O-Me- m7G (5')ppp( 5 ')G, m7G (5')ppp(5')G, 3'- O-Me-m7G(5')ppp(5')A, m7G(5')ppp(5')A, etc.) to the transcription reaction solution.
- PolyA tailing can also be added to the 3' end of mRNA after synthesis using polyA polymerase, or it can be performed simultaneously with the transcription reaction by adding a polyA sequence to the transcription template in advance.
- the mRNA obtained as described above can be purified by removing the template DNA with DNase I and then by, for example, the LiCl method.
- a nucleic acid transfer reagent may be used to promote the transfer of the nucleic acid of the present invention into target cells.
- nucleic acid transfer reagents include atelocollagen; liposomes; nanoparticles; lipofectin, lipofectamine, DOGS (transfectam), DOPE, DOTAP, DDAB, DHDEAB, HDEAB, polybrene, and cationic lipids such as poly(ethyleneimine) (PEI).
- the nucleic acid of the present invention may be a pharmaceutical composition encapsulated in a liposome.
- Liposomes are small, closed vesicles with an internal phase surrounded by one or more lipid bilayers, and can typically hold a water-soluble substance in the internal phase and a lipid-soluble substance within the lipid bilayer.
- the term "encapsulated” used herein refers to the nucleic acid of the present invention being held in the internal phase of the liposome or within the lipid bilayer.
- the liposomes used in the present invention may be monolayer or multilayer membranes, and the particle size can be appropriately selected, for example, from the range of 10 to 1,000 nm, preferably 50 to 300 nm. Considering delivery to target tissues, the particle size may be, for example, 200 nm or less, preferably 100 nm or less.
- Methods for encapsulating water-soluble compounds such as polynucleotides into liposomes include, but are not limited to, the lipid film method (vortex method), reverse phase evaporation, surfactant removal, freeze-thaw method, and remote loading method, and any known method can be selected as appropriate.
- lipid film method vortex method
- reverse phase evaporation surfactant removal
- freeze-thaw method freeze-thaw method
- remote loading method any known method can be selected as appropriate.
- the nucleic acid of the present invention is carried on a lipid nanoparticle and is encapsulated within the particle.
- lipid nanoparticles (sometimes abbreviated as “LNPs” in the present specification) refer to particles having a membrane structure in which the hydrophilic groups of amphipathic lipids are aligned toward the aqueous phase side of the interface, and having a particle diameter of less than 1 ⁇ m
- amphipathic lipid refers to a lipid having both a hydrophilic group and a hydrophobic group.
- the particle diameter of the lipid nanoparticles used in the present invention is preferably 10 nm to 500 nm, and more preferably 30 nm to 300 nm. Particle diameter can be measured using a particle size distribution analyzer such as a Zetasizer Nano (Malvern). The particle diameter of the lipid nanoparticles can be adjusted appropriately depending on the manufacturing method of the lipid nanoparticles. In this specification, "particle diameter” refers to the average particle diameter (zeta mean) measured by dynamic light scattering.
- Amphipathic lipids include, for example, cationic lipids, ionic lipids, phospholipids, and PEG lipids.
- cationic lipid refers to a lipid having a constitutively positively charged hydrophilic group.
- ionizable lipid refers to a lipid that is neutral at physiological pH but becomes protonated and positively charged at low pH.
- PEG refers to polyethylene glycol
- PEG lipid refers to a lipid modified with PEG, i.e., a lipid to which PEG is bound.
- the lipid nanoparticles in which the nucleic acid of the present invention is encapsulated are: (A) a cationic lipid or an ionic lipid, (B) phospholipids, (C) steroids, and (D) lipid nanoparticles containing PEG lipids.
- Each of the component lipids (A) to (D) includes known lipids that are commonly used in lipid nanoparticles, and a person skilled in the art can easily select an appropriate type of lipid and its composition.
- the present invention also provides host cells into which an expression vector containing the nucleic acid of the present invention has been introduced; a method for producing human factor VIII or a modified factor thereof, which comprises culturing the host cells and recovering human factor VIII or a modified factor thereof from the resulting culture; and a pharmaceutical (recombinant protein preparation), particularly a therapeutic agent for hemophilia A, containing the modified human factor VIII obtained by the method.
- An expression vector for producing recombinant human factor VIII can be produced by linking a nucleic acid of the present invention (i.e., a nucleic acid encoding a variant of the present invention, or a nucleic acid encoding a highly expressed form of human factor VIII) downstream of a promoter in an appropriate expression vector.
- Expression vectors that can be used include plasmids derived from Escherichia coli (e.g., pBR322, pBR325, pUC12, pUC13); plasmids derived from Bacillus subtilis (e.g., pUB110, pTP5, pC194); yeast-derived plasmids (e.g., pSH19, pSH15); insect cell expression plasmids (e.g., pFast-Bac); animal cell expression plasmids (e.g., pA1-11, pXT1, pRc/CMV, pRc/RSV, pcDNAI/Neo); bacteriophages such as ⁇ phage; insect virus vectors such as baculovirus (e.g., BmNPV, AcNPV); and animal virus vectors such as retrovirus, lentivirus, vaccinia virus, adenovirus, adeno-
- any promoter may be used as long as it is appropriate for the host used to express the gene.
- the SR ⁇ promoter when the host is an animal cell, the SR ⁇ promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (Rous sarcoma virus) promoter, MoMuLV (Moloney murine leukemia virus) LTR, HSV-TK (herpes simplex virus thymidine kinase) promoter, etc.
- a publicly known promoter may also be selected as appropriate.
- expression vectors can also contain, as desired, enhancers, splicing signals, polyA addition signals, selection markers, SV40 origin of replication (hereinafter sometimes abbreviated as SV40 ori), etc.
- selection markers include the dihydrofolate reductase gene, ampicillin resistance gene, and neomycin resistance gene.
- Human factor VIII or a modified form thereof can be produced by transforming a host with an expression vector containing the nucleic acid of the present invention and culturing the resulting transformant.
- hosts that can be used include Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, and animal cells.
- mammalian cells examples include monkey COS-7 cells, monkey Vero cells, Chinese hamster ovary cells (hereinafter abbreviated as CHO cells), dhfr gene-deficient CHO cells (hereinafter abbreviated as CHO(dhfr ⁇ ) cells), mouse L cells, mouse AtT-20 cells, mouse myeloma cells, rat GH3 cells, human FL cells, HeLa cells, HepG2 cells, and HEK293 cells.
- CHO cells Chinese hamster ovary cells
- CHO(dhfr ⁇ ) cells examples include monkey L cells, mouse AtT-20 cells, mouse myeloma cells, rat GH3 cells, human FL cells, HeLa cells, HepG2 cells, and HEK293 cells.
- CHO cells Chinese hamster ovary cells
- CHO(dhfr ⁇ ) cells examples include monkey L cells, mouse AtT-20 cells, mouse myeloma cells, rat GH3 cells, human
- Transformation can be carried out according to known methods depending on the type of host.
- Animal cells can be transformed, for example, according to the methods described in Cell Engineering Special Issue 8, New Cell Engineering Experimental Protocols, pp. 263-267 (1995) (published by Shujunsha) and Virology, Vol. 52, p. 456 (1973).
- the transformant can be cultured according to a known method depending on the type of host.
- a medium such as minimum essential medium (MEM) containing about 5 to about 20% fetal bovine serum, Dulbecco's modified Eagle's medium (DMEM), RPMI 1640 medium, 199 medium, or Ham's F-12 medium can be used.
- the pH of the medium is preferably about 6 to about 8.
- the culture is usually carried out at about 30°C to about 40°C for about 15 to about 60 hours. Aeration or stirring may be performed as necessary. In this manner, human factor VIII can be produced intracellularly or extracellularly in the transformant.
- Human factor VIII can be separated and purified from the culture obtained by culturing the transformant using methods known per se. Examples of such methods include methods that utilize solubility, such as salting out and solvent precipitation; methods that primarily utilize differences in molecular weight, such as dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis; methods that utilize differences in charge, such as ion exchange chromatography; methods that utilize specific affinity, such as affinity chromatography; methods that utilize differences in hydrophobicity, such as reversed-phase high-performance liquid chromatography; and methods that utilize differences in isoelectric point, such as isoelectric focusing. These methods can also be combined as appropriate.
- solubility such as salting out and solvent precipitation
- methods that primarily utilize differences in molecular weight such as dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis
- methods that utilize differences in charge such as ion exchange chromatography
- methods that utilize specific affinity such as affinity chromat
- the free form can be converted into a salt by a method known per se or a method similar thereto. If the human factor VIII is obtained as a salt, the salt can be converted into the free form or another salt by a method known per se or a method similar thereto.
- Human factor VIII is low in toxicity and can be administered to humans or other mammals parenterally (e.g., intravascular administration (intravenous administration, intraarterial administration, etc.), subcutaneous administration, intradermal administration, intraperitoneal administration, intramuscular injection, topical administration, etc.) either as a liquid preparation or as a pharmaceutical composition in an appropriate dosage form.
- parenterally e.g., intravascular administration (intravenous administration, intraarterial administration, etc.), subcutaneous administration, intradermal administration, intraperitoneal administration, intramuscular injection, topical administration, etc.
- compositions for parenteral administration include, for example, injections and suppositories, and injections may include dosage forms such as intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, and drip infusion injections.
- injections can be prepared according to known methods. For example, injections can be prepared by dissolving, suspending, or emulsifying one or more active ingredients in a sterile aqueous or oily liquid typically used for injections.
- aqueous solutions for injection examples include saline, isotonic solutions containing glucose and other adjuvants, and may be used in combination with appropriate solubilizers, such as alcohols (e.g., ethanol), polyalcohols (e.g., propylene glycol, polyethylene glycol), and nonionic surfactants (e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)).
- solubilizers such as alcohols (e.g., ethanol), polyalcohols (e.g., propylene glycol, polyethylene glycol), and nonionic surfactants (e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)).
- solubilizers such as alcohols (e.g., ethanol), polyalcohols (e.g., propylene glycol, polyethylene glycol), and nonionic surfact
- parenteral pharmaceutical compositions are conveniently prepared in dosage unit forms that correspond to the dosage of the active ingredient.
- dosage unit forms include tablets, pills, capsules, injections (ampoules), and suppositories, and each dosage unit typically contains 100 to 5,000 units, preferably 250 to 3,000 units.
- the dosage of human factor VIII varies depending on the recipient, symptoms, and route of administration.
- a single dose of approximately 10-100 units/kg body weight, preferably approximately 20-50 units/kg body weight is typically administered intravenously or intraperitoneally, preferably two to three times a week. Similar amounts can also be administered for other parenteral administrations. When symptoms are particularly severe, the dosage may be increased accordingly.
- mice were purchased from Japan SLC.
- Factor VIII-deficient mice were obtained from Jackson Laboratory (USA).
- Example 1 Preparation of nucleic acids encoding B domain-deleted human factor VIII (hFVIIISQ) and its variants, and coagulation factor activity of each variant hFVIISQ
- the DNA sequence of human coagulation factor VIII (hFVIIISQ) with most of the B domain removed was designed based on the NCBI reference sequence (NM_000132.4), and codon-optimized and gene-synthesized using GeneArt (Thermo Scientific) (SEQ ID NO: 4). Ver. 1-Ver. 12 A variant, Ver.
- Ver. 1 was prepared by gene synthesis and mutagenesis PCR based on the sequences of hFVIIISQ, canine FVIIISQ, porcine FVIIISQ, ovine FVIIISQ, and bovine FVIIISQ, altering 72 amino acid sites to those of canine FVIIISQ (codon-optimized for human use). Furthermore, variants Ver. 2 and Ver. 3 were prepared by reducing the number of substitutions to 58 and 50, respectively. Ver. 4, which contains only the amino acid substitutions common to Ver. 2 and Ver. 3, was also prepared. Ver. 5, which contains 56 substitutions, was prepared by removing the overlap with the X5 and X10 mutations previously reported (WO 2014/209942). Ver. 6 and Ver.
- Ver. 7 which contain 35 substitutions, were prepared by removing the overlap with the X5 mutation from Ver. 4.
- Ver. 7 which has 51 substitutions, was created by removing overlaps with the JF12 mutation previously reported (Mol Ther Methods Clin Dev. 2020 Jan 15; 17:328-336) from Ver. 2; Ver. 8, which has 32 substitutions, was created by removing overlaps with the JF12 mutation from Ver. 4; Ver. 9, which has 49 substitutions, was created by removing overlaps with the X5, X10, and JF12 mutations from Ver. 2; Ver. 10, which has 31 substitutions, was created by removing overlaps with the X5 and JF12 mutations from Ver. 4; Ver.
- each hFVIIISQ was introduced into a plasmid (p1.1HCRhAAT) carrying the HCRhAAT promoter, which combines HCR, a liver-specific enhancer of the ApoE gene, with the human ⁇ 1-antitrypsin promoter.
- Huh7 cells a human hepatoma cell line
- Huh7 cell culture medium was RPMI-1640 (Sigma) supplemented with 10% FBS, GlutaMAX (Thermo Scientific), and penicillin-streptomycin.
- Huh7 cells were seeded at 1.5 x 10 cells/well in a 24-well plate, and transfection was performed the following day using Lipoefectamin 3000 (Thermo Scientific). The medium was replaced the day after transfection, and the medium was collected the following day after 24 hours of culture. The collected medium was centrifuged, and the supernatant was used as a sample for measuring the activity and antigen of factor VIII.
- Factor VIII activity was measured using the chromogenic assay with the fully automated blood coagulation analyzer CS-1600 (Sysmex).
- AAV8 vectors carrying each FVIII gene were administered to factor VIII-deficient mice at a vector dose of 1 x 108 vg per gram of mouse body weight.
- Factor VIII activity in mouse plasma was measured by the synthetic substrate method.
- Example 2 Identification of Mutation Sites Important for Increased Activity of Ver. 4
- the number of substitutions was reduced from Ver. 4, and a variant S12 with 12 substitutions was identified using the one-stage clotting assay. Further reductions in the number of substitutions led to the identification of PNPL (L152P, K213N, S367P, F2196L) using the synthetic substrate assay.
- S7, with 7 substitutions was created by removing five substitutions from S12 that overlap with the X5 and JF12 mutations.
- S5 and S3, with 5 and 3 substitutions, respectively, were further reduced, and S8, S6, and S4, with the addition of the L152P substitution to S7, S5, and S3, respectively, were created.
- Example 3 Substitution of PNPL variants with other amino acids
- a series of variants (XNPL, PXPL, PNXL, PNPX) were prepared by substituting any of the substitution sites in PNPL with 19 other amino acids. These variants were transfected with plasmids in the same manner as in Example 1, and the coagulation factor activity in the culture supernatant was measured by the one-stage coagulation assay and the synthetic substrate assay. The results are shown in Figures 5-1 to 5-4. It was revealed that substitutions of L152 with Q, V, M, etc., K213 with N, H, etc., S367 with N, Q, etc., and F2196 with M, etc., also showed high activity similar to that of PNPL.
- Example 4 Administration Experiment to FVIII KO Mice
- the Ver. 4, 10, and 11, S12, S8, and PNPL genes which showed particularly high activity in Examples 1 and 2, were loaded onto AAV8 vectors and administered to FVIII knockout hemophilia A model mice.
- blood samples were collected and coagulation factor activity was measured using a one-stage clotting assay and a synthetic chromogenic assay.
- Antigen levels were also measured using ELISA. The results are shown in Figure 6. S12 and PNPL, which contain the PNPL mutation, almost matched the activity of Ver. 4, but the antigen levels were significantly lower. This suggests that these variants have increased specific activity.
- Example 5 Localization of FVIII in and outside FVIII-expressing cells.
- Huh7 cells were transfected with Ver. 4, S12, and PNPL, and the amount of FVIII present in the culture supernatant and cell extract was analyzed by Western blot. The results are shown in Figure 7-1. Intracellular retention of the light chain was observed with wild-type hFVIIISQ, whereas retention was abolished with Ver. 4. S12 and PNPL did not improve intracellular retention of the light chain. The inventors predicted that the improvement in intracellular retention of the light chain in Ver.
- Example 6 Identification of mutation sites important for high secretion and high activity of Ver. 4 (2) Based on Ver. 11, Ver. 12 (FIG. 1) was identified using the synthetic substrate method, in which the number of substitutions was reduced to 13. The results are shown in FIG. The number of substitutions was further reduced based on Ver. 12 to create highly secreted variants with 8 to 15 substitutions (F8-Tochigi-3 (NPL), 5, 8, 10, 11, 13, and 15) (the amino acid substitutions of these variants are shown in Figure 9). Plasmid transfection was performed in the same manner as in Example 1, and the coagulation factor activity in the culture supernatant was measured by the one-stage clotting assay and the synthetic substrate assay. The antigen amount in the culture supernatant was also measured using ELISA. The results are shown in Figure 10. In the figure, F8-Tochigi-28 represents the same variant as Ver. 11.
- Example 7 Construction of a High-Expression hFVIII Gene
- the nucleotide sequences of the codon-optimized wild-type hFVIII DNA (SEQ ID NO: 4) and the Ver. 4 variant DNA (SEQ ID NO: 2) contain numerous CpG sequences.
- the inventors predicted that these CpG sequences may induce an immune response in the host, impairing FVIII expression. Therefore, they removed all of the CpG sequences without changing the amino acid sequence of the FVIII they encode. Then, they re-optimized the codons, taking into account other parameters such as GC content, and designed the nucleotide sequences using GeneArt Codon Optimizer (Thermo Scientific).
- the nucleotide sequences of the CpG-removed and re-optimized wild-type hFVIII DNA and the Ver. 4 variant DNA are shown in SEQ ID NOs: 5 and 3, respectively. These DNAs were ligated downstream of a mouse transthyretin (mTTR) promoter sequence (SEQ ID NO: 7) from which the CpG sequence had also been removed, and then loaded into an AAV6 vector. The vector was then plasmid transfected and introduced into Huh7 cells in the same manner as in Example 1. The expression cassette was also loaded into an AAV8 vector and administered to mice. The coagulation factor activity in the culture supernatant and blood was measured by the one-stage clotting assay and the synthetic substrate assay. The results are shown in Figures 11-1 and 11-2. For both Ver. 4 and the wild-type, removal of the CpG sequence significantly increased the expression efficiency.
- mTTR mouse transthyretin
- Example 8 Administration to a Cynomolgus Monkey Model
- Immunor VIII activity and antigen levels were measured every one or two weeks after administration. The results are shown in Figure 12-1.
- factor VIII activity significantly increased in all individuals after vector administration.
- the above DNA was inserted into an AAV5 vector and administered to a similarly treated cynomolgus monkey model at vector doses 1/10 (6 x 10 12 vg/kg) and 1/30 (2 x 10 12 vg/kg) of those used with Rectavian (registered trademark) (a hemophilia A treatment drug marketed by BioMarin).
- Rectavian registered trademark
- Factor VIII activity and antigen levels were measured every one or two weeks after administration. The results are shown in Figure 12-2. Factor VIII activity significantly increased after vector administration at both doses.
- Example 9 Optimization of Expression Cassette Sequence
- Huh7 cells were transfected with the AAV6 vector (original) prepared in Example 7, in which DNA was inserted into the CpG-deleted and re-optimized Ver. 4 variant coding sequence ligated downstream of the CpG-deleted mTTR promoter sequence (which further contained an SV40 polyA addition signal downstream of the coding sequence and contained the nucleotide sequence shown in SEQ ID NO: 8 as an expression cassette), the AAV6 vector (v1mTTRp) containing, as an expression cassette, a nucleotide sequence (SEQ ID NO: 9) in which the sequence between the promoter and the coding sequence was shortened, and the AAV6 vector (v1-v1, v1-v2, and v1-v3, respectively) containing, as expression cassettes, three nucleotide sequences (SEQ ID NOs: 10, 11, and 12) in which the 3'-end of the polyA addition signal was shortened by different lengths.
- the medium was replaced after 24 hours, and the culture supernatant and cells were collected after another 24 hours.
- the variants of the present invention, the nucleic acids encoding them, expression vectors containing the nucleic acids, and liver cells transfected with the expression vectors are useful for in vivo or ex vivo gene therapy, mRNA medicines, etc. for hemophilia A.
- recombinant human factor VIII variants obtained by culturing host cells transfected with the expression vectors are useful as biological preparations for the treatment of hemophilia A.
- highly expressed human factor VIII-encoding nucleic acids are useful for gene therapy and mRNA medicines for hemophilia A, and are also useful for producing hosts that highly produce recombinant human factor VIII.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Microbiology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biophysics (AREA)
- Epidemiology (AREA)
- Gastroenterology & Hepatology (AREA)
- Hematology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Plant Pathology (AREA)
- General Chemical & Material Sciences (AREA)
- Toxicology (AREA)
- Physics & Mathematics (AREA)
- Virology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Diabetes (AREA)
- Cell Biology (AREA)
- Botany (AREA)
- Immunology (AREA)
- Mycology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
本発明は、高比活性/高分泌発現型のヒト第VIII因子改変体及びそれをコードする核酸、高発現型のヒト第VIII因子コード核酸、並びにそれらの用途に関する。より詳細には、本発明は、野生型と比較して分泌発現効率及び/又は比活性が増強されたヒト第VIII因子改変体及びそれをコードする核酸、ヒト第VIII因子(改変体を含む)をコードする核酸であって、塩基配列の改変により発現効率が増強された核酸、並びにそれらを用いた血友病Aの治療等に関する。 The present invention relates to human factor VIII variants with high specific activity and high secretory expression, nucleic acids encoding the same, highly expressible human factor VIII-encoding nucleic acids, and uses thereof. More specifically, the present invention relates to human factor VIII variants with enhanced secretory expression efficiency and/or specific activity compared to the wild-type, nucleic acids encoding the same, nucleic acids encoding human factor VIII (including variants) whose expression efficiency has been enhanced by modifying the base sequence, and the use thereof for the treatment of hemophilia A, etc.
血友病は、止血に関わる血液凝固因子の遺伝的欠損によって引き起こされる疾患である。世界血友病連盟(WFH)によれば、世界には約40万人の血友病患者がおり、国内には約6,000名の患者が存在する。血友病には、第VIII因子の欠損に起因する血友病Aと、第IX因子の欠損に起因する血友病Bとがあり、比率5:1で血友病Aの患者が多い。 Hemophilia is a disease caused by a genetic deficiency in blood clotting factors involved in hemostasis. According to the World Federation of Hemophilia (WFH), there are approximately 400,000 hemophilia patients worldwide, with approximately 6,000 patients in Japan. There are two types of hemophilia: hemophilia A, which is caused by a deficiency in factor VIII, and hemophilia B, which is caused by a deficiency in factor IX. Hemophilia A patients predominate, with a ratio of 5:1.
従来、血友病Aは、第VIII因子の補充療法によって治療されていた。しかし、凝固因子の半減期は短く、出血予防のために、小児期から週に2~3回の静脈投与を必要とする。また、血友病A患者に第VIII因子を継続的に投与した場合、患者においてインヒビターと称する第VIII因子に対する抗体が生じることがあり(25~30%)、それにより補充療法の有効性が損なわれることが知られている。 Traditionally, hemophilia A has been treated with factor VIII replacement therapy. However, the half-life of this clotting factor is short, and patients need to receive intravenous administration two to three times a week from childhood to prevent bleeding. Furthermore, when factor VIII is administered continuously to hemophilia A patients can develop antibodies against factor VIII, known as inhibitors (25-30%), which is known to reduce the effectiveness of replacement therapy.
近年、血友病Aの治療薬として、第VIII因子の代替物として作用する抗体医薬エミシズマブ(emicizumab)が上市された。エミシズマブは、第VIII因子製剤よりも少ない頻度(月に1回)の皮下注射で血友病Aを治療することができる。しかし、永続的な投与が必要である点で従来の凝固因子製剤と変わりはなく、また、動物細胞による組換え生産のため製造コストが高く、医療経済への負担が大きい。 In recent years, the antibody drug emicizumab, which acts as a substitute for factor VIII, has been launched as a treatment for hemophilia A. Emicizumab can treat hemophilia A with subcutaneous injections less frequently (once a month) than factor VIII preparations. However, it is no different from conventional coagulation factor preparations in that it requires continuous administration, and because it is produced using recombinant animal cells, the manufacturing costs are high, placing a significant burden on the medical economy.
そこで、単回投与で長期間にわたって凝固因子活性が維持されることが期待されるウイルスベクターを用いた遺伝子治療が、血友病の新たな治療法として注目されている。血友病は、単一遺伝子欠損による遺伝性疾患であり、血中の凝固因子レベルで治療効果が容易に確認できることや、比較的わずかな血中レベルの上昇でも効果が期待できる等の理由から、以前より遺伝子治療のよい標的疾患であると考えられていた。また、遺伝子治療による連続的な凝固因子の発現により、制御性T細胞を介した免疫寛容が誘導され、インヒビターを除去し得ることも前臨床研究で示されている。最近、アデノ随伴ウイルス(AAV)ベクターを用いたヒト遺伝子治療の部分的な成功例が相次いで報告されている(例えば、非特許文献1、2を参照)。凝固因子は肝臓で産生されるが、AAVベクターは、静脈投与で肝臓に高効率で遺伝子を送達することができる。 Therefore, gene therapy using viral vectors, which are expected to maintain clotting factor activity for long periods with a single administration, is attracting attention as a new treatment for hemophilia. Hemophilia is a hereditary disease caused by a single gene deficiency. Because therapeutic effects can be easily confirmed by measuring clotting factor levels in the blood and because even a relatively small increase in blood levels can be expected to be effective, it has long been considered a good target for gene therapy. Furthermore, preclinical studies have shown that continuous expression of clotting factors through gene therapy can induce immune tolerance via regulatory T cells, potentially eliminating inhibitors. Recently, a series of partially successful human gene therapy cases using adeno-associated virus (AAV) vectors have been reported (see, for example, Non-Patent Documents 1 and 2). Clotting factors are produced in the liver, and AAV vectors can deliver genes to the liver with high efficiency via intravenous administration.
BioMarin社の臨床試験では、高用量投与群で1年後にも高い第VIII因子活性が維持されたが(非特許文献1)、3年後には血中レベルの低下傾向が認められた。その原因の1つとして、肝臓細胞における高用量AAVベクターからの第VIII因子発現が小胞体ストレスを惹起する可能性が指摘されている(非特許文献3)。
一方、Spark社の臨床試験では、肝指向性の高い血清型を用いてBioMarin社よりも低用量のAAVベクターが投与されたが、2×1012vg/kg投与群では肝毒性が認められた(非特許文献2)。肝臓特異的プロモーターの使用や第VIII因子のコドン最適化により高発現を実現することで、低用量化して安全性を高める試みがなされているが、タンパク質の高発現自体が治療効果の長期安定性を損なう場合には、そのアプローチでは十分に対応できない。
In a clinical trial conducted by BioMarin, high factor VIII activity was maintained even after one year in the high-dose administration group (Non-Patent Document 1), but a tendency for blood levels to decrease was observed after three years. One possible reason for this is that factor VIII expression from a high-dose AAV vector in liver cells may induce endoplasmic reticulum stress (Non-Patent Document 3).
On the other hand, in Spark's clinical trials, a serotype with high liver tropism was used and a lower dose of AAV vector than BioMarin's was administered, but hepatotoxicity was observed in the 2 x 1012 vg/kg administration group (Non-Patent Document 2). Attempts have been made to reduce the dose and improve safety by achieving high expression through the use of liver-specific promoters and codon optimization of factor VIII, but these approaches are not sufficient in cases where high protein expression itself impairs the long-term stability of the therapeutic effect.
高用量ウイルスベクター投与による治療効果の減衰を回避する手段の1つとして、適切なタンパク質レベルで野生型第VIII因子を高発現させた場合と同等以上の凝固活性を発揮し得る、比活性の高い第VIII因子改変体や、小胞体ストレス等の細胞への悪影響を排除するために、細胞外への分泌効率が向上した第VIII因子改変体の開発が望まれる。Xiaoらは、A1ドメイン中の10カ所のアミノ酸残基をブタ型に置換すると、分泌効率が著明に増大し、中でも5カ所のアミノ酸置換が重要であると報告している(特許文献1及び非特許文献4)。さらに、同グループは、ヒト第VIII因子軽鎖中の12カ所のアミノ酸残基をイヌ型に置換すると、野生型の分泌効率を維持しつつ、比活性を増大させ得ることを報告している(非特許文献5)。また、Sabatinoらは、ヒト第VIII因子軽鎖中のフリン認識部位及びa3ドメインの1以上のアミノ酸をイヌ型に置換もしくは欠失させると、比活性を増大させ得ることを開示している(特許文献2)。 As one way to avoid the attenuation of therapeutic efficacy due to high-dose viral vector administration, it is desirable to develop a variant of factor VIII with high specific activity that can exert coagulation activity equivalent to or greater than that of wild-type factor VIII expressed at appropriate protein levels, as well as a variant of factor VIII with improved extracellular secretion efficiency to eliminate adverse effects on cells such as endoplasmic reticulum stress. Xiao et al. have reported that substituting 10 amino acid residues in the A1 domain with porcine residues significantly increases secretion efficiency, with five amino acid substitutions being particularly important (Patent Document 1 and Non-Patent Document 4). Furthermore, the same group has reported that substituting 12 amino acid residues in the light chain of human factor VIII with canine residues increases specific activity while maintaining wild-type secretion efficiency (Non-Patent Document 5). Furthermore, Sabatino et al. have disclosed that the specific activity can be increased by substituting or deleting one or more amino acids in the furin recognition site and a3 domain in the human factor VIII light chain with canine forms (Patent Document 2).
本発明の第1の課題は、血友病Aの遺伝子治療において、長期間安定して第VIII因子を補完できる新規なヒト第VIII因子改変体及びそれをコードする核酸を提供することであり、以って、より持効性に優れた血友病Aの遺伝子治療を確立することである。本発明の第2の課題は、当該改変体を含む組換え第VIII因子製剤や当該改変体をコードするmRNA医薬を提供することである。 The first object of the present invention is to provide a novel human factor VIII variant and the nucleic acid encoding it that can stably complement factor VIII over the long term in gene therapy for hemophilia A, thereby establishing a gene therapy for hemophilia A with superior long-term efficacy. The second object of the present invention is to provide a recombinant factor VIII preparation containing the variant and an mRNA drug encoding the variant.
本発明者らは、上記の課題を解決すべく鋭意研究を重ねた結果、野生型ヒト第VIII因子の213位のリジン(K213)、367位のセリン(S367)及び2196位のフェニルアラニン(F2196)からなる群より選択される少なくとも1個のアミノ酸残基が他のアミノ酸で置換された種々の改変体が、野生型と比較して高比活性及び/又は高分泌発現を示す(本明細書において、「高比活性/高分泌発現型」と略記する場合がある)ことを見出した(本明細書において、ヒト第VIII因子中のアミノ酸残基の位置は、成熟完全長一本鎖ポリペプチド(2332アミノ酸)におけるアミノ酸番号で表記され、n番目のアミノ酸残基X(Xは当該アミノ酸の一文字表記)は、「Xn」と略記される。nが負の整数の場合、シグナルペプチドのアミノ酸残基の位置を示す。また、n番目のアミノ酸残基Xが他のアミノ酸Y(Yは当該他のアミノ酸の一文字表記)で置換されている場合、「XnY」と略記される)。 As a result of extensive research aimed at solving the above-mentioned problems, the present inventors have found that various variants in which at least one amino acid residue selected from the group consisting of lysine at position 213 (K213), serine at position 367 (S367), and phenylalanine at position 2196 (F2196) of wild-type human factor VIII is substituted with another amino acid exhibit higher specific activity and/or higher secretory expression than the wild-type (sometimes abbreviated herein as "high specific activity/high secretory expression type"). (In this specification, the positions of amino acid residues in human factor VIII are represented by the amino acid numbering in the mature full-length single-chain polypeptide (2332 amino acids), and the nth amino acid residue X (X is the one-letter code for the amino acid) is abbreviated as "Xn." When n is a negative integer, it indicates the position of the amino acid residue in the signal peptide. When the nth amino acid residue X is substituted with another amino acid Y (Y is the one-letter code for the other amino acid), it is abbreviated as "XnY.")
さらに、本発明者らは、上記アミノ酸置換を含む改変体のヒトでの発現効率を高めるべく、該改変体をコードする核酸のヌクレオチド配列を、コドン最適化アルゴリズムを用いてヒト用に最適化したが、得られたヌクレオチド配列には、CpG配列が多数含まれていた。本発明者らは、CpG配列は宿主における免疫応答を引き起こし、持続的な導入遺伝子の発現を阻害する可能性があると予測し、該改変体のアミノ酸配列を変化させることなく、これらのCpG配列を除去する塩基置換を行った上で、再度コドン最適化し直した。得られた改変体コード核酸を、やはりCpG配列を除去したプロモーター配列の下流に連結してAAVベクターに搭載し、ヒト肝臓細胞株及び血友病モデルマウスに投与したところ、第VIII因子の発現がCpG配列除去前の改変体コード核酸に比べて著明に上昇した。CpG配列除去による発現増強効果が当該改変体に限らず、野生型を含めた任意のヒト第VIII因子に共通することを実証すべく、野生型ヒト第VIII因子をコードするコドン最適化された核酸と、さらにCpG配列除去を行った上でコドン最適化し直した核酸との間で、同様に第VIII因子の発現を比較したところ、やはりCpG配列の除去により著明な発現増強が認められた。
本発明者らは、これらの知見に基づいてさらに検討を重ねた結果、本発明を完成するに至った。
Furthermore, in order to improve the expression efficiency of the variants containing the above amino acid substitutions in humans, the inventors optimized the nucleotide sequence of the nucleic acid encoding the variants for human use using a codon optimization algorithm. However, the resulting nucleotide sequence contained many CpG sequences. The inventors predicted that CpG sequences may induce an immune response in the host and inhibit continuous transgene expression, so they performed base substitutions to remove these CpG sequences without changing the amino acid sequence of the variants, and then performed codon optimization again. The resulting variant-encoding nucleic acid was ligated downstream of a promoter sequence from which CpG sequences had also been removed, and then incorporated into an AAV vector. When the resulting nucleic acid encoding the variants was administered to a human liver cell line and a hemophilia model mouse, factor VIII expression was significantly increased compared to the variant-encoding nucleic acid before CpG sequence removal. To demonstrate that the expression-enhancing effect of CpG sequence removal is not limited to the variant but is common to any human factor VIII, including wild-type factor VIII, the expression of factor VIII was similarly compared between a codon-optimized nucleic acid encoding wild-type human factor VIII and a nucleic acid that had been further codon-optimized after CpG sequence removal. As a result, a significant enhancement of expression was observed after CpG sequence removal.
Based on these findings, the present inventors have conducted further studies and have completed the present invention.
即ち、本発明は以下のものを提供する。
[項1]
野生型ヒト第VIII因子のK213、S367及びF2196からなる群より選択される少なくとも1個のアミノ酸残基が他のアミノ酸で置換され、野生型と比較して、比活性及び/又は分泌発現効率が高いことを特徴とする、ヒト第VIII因子改変体。
[項2]
K213におけるアミノ酸置換がK213NもしくはK213Hであり、及び/又は
S367におけるアミノ酸置換がS367P、S367NもしくはS367Qであり、及び/又は
F2196におけるアミノ酸置換がF2196LもしくはF2196Mである、項1に記載の改変体。
[項3]
K213N、S367P及びF2196Lのアミノ酸置換を含む、項2に記載の改変体。
[項4]
R-5、P25、A28、L152、M217、W228、Q410、Y487、R489、F501、M539、I566、L603、I642、S727、A736、S1657、Q1659、E1661、I1668、D1681、R1776、H1859、A1993、H2007、N2019、K2085、K2207、F2275、S2296、V2314、Q2316及びM2321からなる群より選択される少なくとも1個のアミノ酸残基が他のアミノ酸でさらに置換されている、項1~3のいずれか一項に記載の改変体。
[項5]
R-5P、P25H、A28T、L152A、L152D、L152E、L152H、L152I、L152M、L152N、L152P、L152Q又はL152V、M217T、W228Q、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321Lからなる群より選択される少なくとも1個のアミノ酸置換を含む、項4に記載の改変体。
[項6]
L152A、L152D、L152E、L152H、L152I、L152M、L152N、L152P、L152Q又はL152V、並びに/或いは
I1668F及びD1681G
のアミノ酸置換をさらに含む、項3に記載の改変体。
[項7]
K213におけるアミノ酸置換がK213Nであり、
M539L、並びに、任意選択でI566M、L603P及びI642Vからなる群より選択される少なくとも1個のA2ドメイン内のアミノ酸置換をさらに含み、
K2207Q、Q2316H及びM2321L、並びに、任意選択でF2275L、S2296A及びV2314Aからなる群より選択される少なくとも1個のC2ドメイン内のアミノ酸置換をさらに含む、項1に記載の改変体。
[項8]
I1668F及びD1681Gのアミノ酸置換をさらに含む、項7に記載の改変体。
[項9]
L152P、Y487H及びL603Pのアミノ酸置換をさらに含み、任意選択でS727P、Q1659E、H1859R、A1993V、H2007Q、N2019K及びK2085Mからなる群より選択される少なくとも1個のアミノ酸置換をさらに含む、項3に記載の改変体。
[項10]
M539L、I566M、L603P、I642V、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321Lのアミノ酸置換をさらに含む、項3に記載の改変体。
[項11]
I1668F及びD1681Gのアミノ酸置換をさらに含む、項10に記載の改変体。
[項12]
野生型ヒト第VIII因子において、以下の(a)~(n)のアミノ酸置換を有するヒト第VIII因子改変体。
(a)K213N、S367P及びF2196L
(b)K213N、S367P、F2196L、I1668F及びD1681G
(c)K213N、M539L、I566M、I642V、K2207Q、F2275L、Q2316H及びM2321L
(d)K213N、M539L、I566M、L603P、I642V、F2196L、K2207Q、F2275L、Q2316H及びM2321L
(e)K213N、M539L、I566M、I642V、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(f)K213N、M539L、I566M、I642V、I1668F、D1681G、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(g)L152P、K213N、S367P及びF2196L
(h)L152P、K213N、S367P、Y487H、L603P及びF2196L
(i)L152P、K213N、S367P、Y487H、L603P、S727P、Q1659E及びF2196L
(j)L152P、K213N、S367P、Y487H、L603P、S727P、Q1659E、H1859R、A1993V、H2007Q、K2085M及びF2196L
(k)K213N、S367P、M539L、I566M、L603P、I642V、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(l)P25H、A28T、K213N、M217T、S367P、Q410L、Y487H、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、N2019K、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(m)R-5P、P25H、A28T、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、N2019K、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(n)R-5P、P25H、A28T、L152P、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
[項13]
Bドメインを欠失している、項1~12のいずれか一項に記載の改変体。
[項14]
Bドメインの欠失が、配列番号1で表されるアミノ酸配列中、763位~1656位のアミノ酸配列の欠失である、項13に記載の改変体。
[項15]
項1~14のいずれか一項に記載の改変体をコードする核酸。
[項16]
ヒト用にコドン最適化されている、項15に記載の核酸。
[項17]
項16に記載の核酸、又はヒト用にコドン最適化された野生型ヒト第VIII因子をコードする核酸であって、それがコードするアミノ酸配列を変化させることなく、CpG配列を含まないようにさらに改変されている、核酸。
[項18]
項15~17のいずれか一項に記載の核酸を含む発現ベクター。
[項19]
前記核酸が肝臓特異的プロモーターの制御下にある、項18に記載の発現ベクター。
[項20]
プロモーターがCpG配列を含まないように改変されている、項18又は19に記載の発現ベクター。
[項21]
ベクターがウイルスベクターである、項18~20のいずれか一項に記載の発現ベクター。
[項22]
ウイルスが肝臓指向性を有するアデノ随伴ウイルスである、項21に記載の発現ベクター。
[項23]
項18~22のいずれか一項に記載の発現ベクターが導入された宿主細胞。
[項24]
項23に記載の宿主細胞を培養し、得られる培養物からヒト第VIII因子又はその改変体を回収することを含む、ヒト第VIII因子又はその改変体の製造方法。
[項25]
項1~14のいずれか一項に記載の改変体、項15~17のいずれか一項に記載の核酸、項18~22のいずれか一項に記載の発現ベクター、又は項23に記載の宿主細胞を含有する医薬。
[項26]
血友病Aの治療用である、項25に記載の医薬。
[項26a]
血友病A患者に、有効量の、項1~14のいずれか一項に記載の改変体、項15~17のいずれか一項に記載の核酸、項18~22のいずれか一項に記載の発現ベクター、又は項23に記載の宿主細胞を投与することを含む、血友病Aの治療方法。
[項26b]
血友病Aの治療における使用のための、項1~14のいずれか一項に記載の改変体、項15~17のいずれか一項に記載の核酸、項18~22のいずれか一項に記載の発現ベクター、又は項23に記載の宿主細胞。
[項26c]
血友病Aの治療剤の製造のための、項1~14のいずれか一項に記載の改変体、項15~17のいずれか一項に記載の核酸、項18~22のいずれか一項に記載の発現ベクター、又は項23に記載の宿主細胞の使用。
That is, the present invention provides the following.
[Section 1]
A human factor VIII variant characterized in that at least one amino acid residue selected from the group consisting of K213, S367 and F2196 of wild-type human factor VIII is replaced with another amino acid, and that the variant has higher specific activity and/or secretory expression efficiency compared to the wild-type variant.
[Section 2]
Item 2. The variant according to Item 1, wherein the amino acid substitution at K213 is K213N or K213H, and/or the amino acid substitution at S367 is S367P, S367N or S367Q, and/or the amino acid substitution at F2196 is F2196L or F2196M.
[Section 3]
Item 3. The variant according to Item 2, comprising the amino acid substitutions K213N, S367P, and F2196L.
[Section 4]
Item 4. The variant according to any one of Items 1 to 3, wherein at least one amino acid residue selected from the group consisting of R-5, P25, A28, L152, M217, W228, Q410, Y487, R489, F501, M539, I566, L603, I642, S727, A736, S1657, Q1659, E1661, I1668, D1681, R1776, H1859, A1993, H2007, N2019, K2085, K2207, F2275, S2296, V2314, Q2316, and M2321 is further substituted with another amino acid.
[Section 5]
R-5P, P25H, A28T, L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q or L152V, M217T, W228Q, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657 Item 5. The variant according to Item 4, comprising at least one amino acid substitution selected from the group consisting of P, Q1659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N2019K, K2085M, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
[Section 6]
L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q or L152V, and/or I1668F and D1681G
Item 4. The variant according to Item 3, further comprising an amino acid substitution of:
[Section 7]
the amino acid substitution at K213 is K213N;
M539L, and optionally at least one amino acid substitution in the A2 domain selected from the group consisting of I566M, L603P, and I642V;
Item 14. The variant of item 1, further comprising at least one amino acid substitution in the C2 domain selected from the group consisting of K2207Q, Q2316H and M2321L, and optionally F2275L, S2296A and V2314A.
[Section 8]
Item 8. The variant of Item 7, further comprising the amino acid substitutions I1668F and D1681G.
[Section 9]
Item 4. The variant of item 3, further comprising amino acid substitutions L152P, Y487H and L603P, and optionally further comprising at least one amino acid substitution selected from the group consisting of S727P, Q1659E, H1859R, A1993V, H2007Q, N2019K and K2085M.
[Section 10]
Item 4. The variant according to Item 3, further comprising the amino acid substitutions M539L, I566M, L603P, I642V, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
[Section 11]
Item 11. The variant of Item 10, further comprising the amino acid substitutions I1668F and D1681G.
[Section 12]
A human factor VIII variant having the following amino acid substitutions (a) to (n) in wild-type human factor VIII:
(a) K213N, S367P, and F2196L
(b) K213N, S367P, F2196L, I1668F, and D1681G
(c) K213N, M539L, I566M, I642V, K2207Q, F2275L, Q2316H, and M2321L
(d) K213N, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, Q2316H, and M2321L
(e) K213N, M539L, I566M, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L
(f) K213N, M539L, I566M, I642V, I1668F, D1681G, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L
(g) L152P, K213N, S367P and F2196L
(h) L152P, K213N, S367P, Y487H, L603P and F2196L
(i) L152P, K213N, S367P, Y487H, L603P, S727P, Q1659E, and F2196L
(j) L152P, K213N, S367P, Y487H, L603P, S727P, Q1659E, H1859R, A1993V, H2007Q, K2085M and F2196L
(k) K213N, S367P, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(l) P25H, A28T, K213N, M217T, S367P, Q410L, Y487H, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P , Q1659E, E1661K, I1668F, D1681G, R1776K, N2019K, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(m) R-5P, P25H, A28T, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736 V, S1657P, Q1659E, E1661K, I1668F, D1681G, R1776K, N2019K, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487 H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1 659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N201 9K, K2085M, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
[Section 13]
Item 13. The variant according to any one of Items 1 to 12, which lacks the B domain.
[Section 14]
Item 14. The variant according to Item 13, wherein the deletion of the B domain is a deletion of the amino acid sequence from positions 763 to 1656 in the amino acid sequence represented by SEQ ID NO:1.
[Section 15]
A nucleic acid encoding the variant according to any one of Items 1 to 14.
[Section 16]
Item 16. The nucleic acid of Item 15, which is codon-optimized for human use.
[Section 17]
17. The nucleic acid of claim 16, or a nucleic acid encoding wild-type human Factor VIII that has been codon-optimized for human use, which has been further modified to not contain CpG sequences without changing the amino acid sequence that it encodes.
[Section 18]
Item 18. An expression vector comprising the nucleic acid according to any one of Items 15 to 17.
[Section 19]
19. The expression vector of claim 18, wherein the nucleic acid is under the control of a liver-specific promoter.
[Section 20]
Item 20. The expression vector according to Item 18 or 19, wherein the promoter has been modified so as not to contain a CpG sequence.
[Section 21]
Item 21. The expression vector according to any one of Items 18 to 20, wherein the vector is a viral vector.
[Section 22]
Item 22. The expression vector according to Item 21, wherein the virus is an adeno-associated virus having liver tropism.
[Section 23]
A host cell into which the expression vector according to any one of Items 18 to 22 has been introduced.
[Section 24]
24. A method for producing human factor VIII or a variant thereof, comprising culturing the host cell according to Item 23 and recovering human factor VIII or a variant thereof from the resulting culture.
[Section 25]
A pharmaceutical comprising the variant according to any one of Items 1 to 14, the nucleic acid according to any one of Items 15 to 17, the expression vector according to any one of Items 18 to 22, or the host cell according to Item 23.
[Section 26]
Item 26. The pharmaceutical agent according to Item 25, which is for treating hemophilia A.
[Section 26a]
A method for treating hemophilia A, comprising administering to a patient with hemophilia A an effective amount of the variant of any one of Items 1 to 14, the nucleic acid of any one of Items 15 to 17, the expression vector of any one of Items 18 to 22, or the host cell of Items 23.
[Section 26b]
The variant of any one of Items 1 to 14, the nucleic acid of any one of Items 15 to 17, the expression vector of any one of Items 18 to 22, or the host cell of Item 23, for use in treating hemophilia A.
[Section 26c]
Use of the variant according to any one of Items 1 to 14, the nucleic acid according to any one of Items 15 to 17, the expression vector according to any one of Items 18 to 22, or the host cell according to Item 23 for the manufacture of a therapeutic agent for hemophilia A.
本発明によれば、比活性及び/又は分泌発現効率の高いヒト第VIII因子改変体及びそれをコードする核酸が提供され、これらを用いることにより、長期間安定して第VIII因子を補完できる血友病Aの治療が可能となる。また、本発明によれば、同一のアミノ酸配列をコードするものであっても、他に比べてタンパク質発現効率が増強されたヒト第VIII因子コード核酸が提供される。 The present invention provides human factor VIII variants with high specific activity and/or secretory expression efficiency, and nucleic acids encoding the same, which, when used, enable the treatment of hemophilia A with stable factor VIII supplementation over a long period of time. Furthermore, the present invention provides human factor VIII-encoding nucleic acids with enhanced protein expression efficiency compared to other nucleic acids encoding the same amino acid sequence.
小胞体でアミノ酸19個からなるシグナルペプチドが切断された1本鎖完全長ヒト第VIII因子タンパク質は、3つのAドメイン(A1,A2,A3)、2つのCドメイン(C1,C2)、Bドメイン、さらに酸性アミノ酸が豊富な3つのペプチド領域(a1,a2,a3)から構成され、アミノ末端からA1(1~336位)、a1(337~372位)、A2(373~710位)、a2(711~740位)、B(741~1648位)、a3(1649~1689位)、A3(1690~2019位)、C1(2020~2172位)及びC2(2173~2332位)の順に並ぶ。ゴルジ体でBドメイン内のR1313やR1648が切断され、重鎖と軽鎖の2本鎖となる。Bドメインは欠失しても凝固活性をほとんど損なわないので、Bドメイン欠失(BDD)-第VIII因子がよく用いられており、本発明においても好ましく用いられる。 The single-chain full-length human factor VIII protein, after its 19-amino acid signal peptide is cleaved in the endoplasmic reticulum, consists of three A domains (A1, A2, A3), two C domains (C1, C2), a B domain, and three peptide regions rich in acidic amino acids (a1, a2, a3). From the amino terminus, these are arranged in the following order: A1 (positions 1-336), a1 (positions 337-372), A2 (positions 373-710), a2 (positions 711-740), B (positions 741-1648), a3 (positions 1649-1689), A3 (positions 1690-2019), C1 (positions 2020-2172), and C2 (positions 2173-2332). In the Golgi apparatus, R1313 and R1648 in the B domain are cleaved, resulting in a two-chain heavy and light chain. Since the B domain is deleted without significantly impairing coagulation activity, B domain-deleted (BDD) factor VIII is commonly used and is also preferably used in the present invention.
本発明は、野生型と比べて比活性及び/又は分泌発現効率が増強されたヒト第VIII因子改変体(以下、「本発明の改変体」ともいう)を提供する。本発明の改変体は、野生型ヒト第VIII因子のK213、S367及びF2196からなる群より選択される少なくとも1個のアミノ酸残基が他のアミノ酸で置換されていることを特徴とする。 The present invention provides a human factor VIII variant (hereinafter also referred to as the "variant of the present invention") that has enhanced specific activity and/or secretory expression efficiency compared to the wild-type form. The variant of the present invention is characterized in that at least one amino acid residue selected from the group consisting of K213, S367, and F2196 of wild-type human factor VIII is substituted with another amino acid.
本明細書において「野生型ヒト第VIII因子」とは、天然に存在する正常なヒト第VIII因子のアミノ酸配列を含むタンパク質を意味し、具体的には、
(a)配列番号1で表されるアミノ酸配列(UniProtKBにアクセッション番号P00451として登録されている)からなるタンパク質、あるいは
(b)(a)のタンパク質のアレル変異体(マイナーアレル頻度(MAF)1%未満)もしくは遺伝子多型(MAF1%以上;例、SNP等)のアミノ酸配列(例えば、UniProtKBのアクセッション番号P00451の「Variants」欄に、疾患と関連しないバリアントとして掲載されているもの等;但し、K213、S367及びF2196の位置にアミノ酸変異を有しない)からなるタンパク質であって、(a)のタンパク質と同等の比活性及び分泌発現効率を示すタンパク質
を挙げることができる。ここで比活性又は分泌発現効率が「同等」であるとは、(a)のタンパク質に対して比活性又は分泌発現効率が0.8~1.2倍、好ましくは0.9~1.1倍、より好ましくは0.95~1.05倍であることを意味する。
好ましい実施態様において、野生型ヒト第VIII因子は、前記(a)のタンパク質である。
As used herein, the term "wild-type human factor VIII" refers to a protein comprising the amino acid sequence of naturally occurring normal human factor VIII, specifically:
Examples of such proteins include (a) a protein consisting of the amino acid sequence represented by SEQ ID NO: 1 (registered in UniProtKB under accession number P00451), or (b) a protein consisting of the amino acid sequence of an allelic variant (minor allele frequency (MAF) less than 1%) or genetic polymorphism (MAF 1% or more; e.g., SNP) of the protein of (a) (e.g., a variant not associated with disease listed in the "Variant" column of UniProtKB under accession number P00451; excluding the amino acid mutations at positions K213, S367, and F2196), which protein exhibits a specific activity and secretory expression efficiency equivalent to that of the protein of (a). Here, "equivalent" in specific activity or secretory expression efficiency means that the specific activity or secretory expression efficiency is 0.8 to 1.2 times, preferably 0.9 to 1.1 times, and more preferably 0.95 to 1.05 times that of the protein of (a).
In a preferred embodiment, wild-type human factor VIII is the protein of (a) above.
本明細書において「ヒト第VIII因子改変体」とは、前記(a)又は(b)の野生型ヒト第VIII因子のアミノ酸配列(参照配列)において、1以上のアミノ酸が置換、欠失、挿入もしくは付加されたアミノ酸配列を有し、参照配列からなるヒト第VIII因子と比較して、比活性及び/又は分泌発現効率が実質的に変化したタンパク質を意味する。ここで「実質的に変化」とは、統計学的に有意に変化することが好ましいが、有意差はなくとも増大もしくは低下傾向にある場合をも包含する。 As used herein, "variant human factor VIII" refers to a protein having an amino acid sequence in which one or more amino acids have been substituted, deleted, inserted, or added in the amino acid sequence (reference sequence) of wild-type human factor VIII (a) or (b) above, and in which the specific activity and/or secretory expression efficiency has been substantially altered compared to human factor VIII consisting of the reference sequence. Here, "substantially altered" preferably means a statistically significant change, but also encompasses cases in which there is a tendency for an increase or decrease even if there is no significant difference.
本発明の改変体は、野生型と比較して、比活性及び/又は分泌発現効率が高いヒト第VIII因子改変体である。ヒト第VIII因子の比活性は、自体公知の凝固一段法(OSA)又は合成基質法(CSA)により凝固因子活性を測定し、自体公知のタンパク質定量法(例、ELISA等のイムノアッセイ等)によりタンパク質量を測定して、単位タンパク質量あたりの凝固因子活性を算出することにより評価することができる。また、ヒト第VIII因子の分泌発現効率は、それをコードする核酸をトランスフェクトした細胞の培養上清や、該核酸を投与した動物の血漿中のヒト第VIII因子濃度を、自体公知の方法(例、ELISA等のイムノアッセイ等)にて測定することにより評価することができる。 The variants of the present invention are human factor VIII variants that have higher specific activity and/or secretory expression efficiency compared to the wild-type. The specific activity of human factor VIII can be evaluated by measuring the coagulation factor activity using the publicly known one-stage coagulation assay (OSA) or chromogenic assay (CSA), measuring the protein amount using a publicly known protein quantification method (e.g., immunoassay such as ELISA), and calculating the coagulation factor activity per unit protein amount. The secretory expression efficiency of human factor VIII can also be evaluated by measuring the concentration of human factor VIII in the culture supernatant of cells transfected with a nucleic acid encoding it or in the plasma of animals administered the nucleic acid using a publicly known method (e.g., immunoassay such as ELISA).
本発明の改変体は、野生型ヒト第VIII因子のK213、S367及びF2196からなる群より選択される少なくとも1個、好ましくは2個(例、K213及びS367、K213及びF2196、あるいはS367及びF2196)、より好ましくは3個(K213、S367及びF2196)のアミノ酸残基が、他のアミノ酸で置換されている。野生型ヒト第VIII因子が前記(b)のタンパク質であって、そのアミノ酸残基の位置が、1以上のアミノ酸の欠失もしくは挿入/付加により、配列番号1における当該アミノ酸残基の位置とは異なる場合、K213、S367及びF2196に対応するアミノ酸残基は、(b)のアミノ酸配列と配列番号1のアミノ酸配列とを、任意の相同性検索アルゴリズムを用いて適切にアラインさせた際の、配列番号1のK213、S367及びF2196にそれぞれ対応するアミノ酸残基として同定することができる。 In the variants of the present invention, at least one, preferably two (e.g., K213 and S367, K213 and F2196, or S367 and F2196), more preferably three (K213, S367 and F2196) amino acid residues selected from the group consisting of K213, S367, and F2196 of wild-type human factor VIII are substituted with other amino acids. When the wild-type human factor VIII is the protein (b) above and the positions of the amino acid residues differ from those in SEQ ID NO: 1 due to the deletion or insertion/addition of one or more amino acids, the amino acid residues corresponding to K213, S367, and F2196 can be identified as the amino acid residues corresponding to K213, S367, and F2196 in SEQ ID NO: 1, respectively, when the amino acid sequence of (b) and the amino acid sequence of SEQ ID NO: 1 are appropriately aligned using any homology search algorithm.
当該他のアミノ酸としては、アミノ酸置換の結果得られるヒト第VIII因子改変体が、野生型と比べて比活性及び/又は分泌発現効率が増強されたものである限り、特に制限はないが、K213におけるアミノ酸置換としては、K213NもしくはK213Hが好ましく、K213Nがより好ましい。S367におけるアミノ酸置換としては、S367P、S367NもしくはS367Qが好ましく、S367Pがより好ましい。F2196におけるアミノ酸置換としては、F2196LもしくはF2196Mが好ましく、F2196Lがより好ましい。 The other amino acids are not particularly limited, so long as the human factor VIII variant obtained as a result of the amino acid substitution has enhanced specific activity and/or secretory expression efficiency compared to the wild-type. However, the amino acid substitution at K213 is preferably K213N or K213H, with K213N being more preferred. The amino acid substitution at S367 is preferably S367P, S367N or S367Q, with S367P being more preferred. The amino acid substitution at F2196 is preferably F2196L or F2196M, with F2196L being more preferred.
特に好ましい一実施様において、本発明の改変体は、野生型ヒト第VIII因子において、K213N、S367P及びF2196Lのアミノ酸置換(以下、置換後のアミノ酸残基に因んで「NPL変異」という場合がある)を含む。NPL変異を有する改変体は、結果としてその効果を打ち消す他の変異(の組み合わせ)が存在しないことを条件として、野生型と比較して高比活性であるとの特徴を有する。前述のとおり、高比活性な改変体は、治療効果の長期安定性を損なわない程度に適切なタンパク質発現レベルで、野生型第VIII因子を高発現させた場合と同等以上の凝固活性を実現し得るので、血友病Aに対する遺伝子治療に有用である。 In a particularly preferred embodiment, the variant of the present invention comprises the amino acid substitutions K213 N , S367 P , and F2196 L in wild-type human factor VIII (hereinafter, sometimes referred to as the "NPL mutation" after the substituted amino acid residues). A variant having the NPL mutation is characterized by a higher specific activity than the wild-type, provided that there are no other mutations (combinations of mutations) that would negate the effect of the NPL mutation. As described above, a high-specific-activity variant can achieve coagulation activity equivalent to or greater than that achieved by overexpressing wild-type factor VIII at an appropriate protein expression level that does not impair the long-term stability of the therapeutic effect, and is therefore useful for gene therapy for hemophilia A.
本発明の改変体は、野生型と比べて比活性及び/又は分泌発現効率が高いという特性が保持される限り、K213、S367及びF2196からなる群より選択される少なくとも1個のアミノ酸置換に加えて、K213、S367及びF2196以外の位置のアミノ酸残基が他のアミノ酸で置換されてもよい。そのような置換部位として、例えば、R-5、P25、A28、L152、M217、W228、Q410、Y487、R489、F501、M539、I566、L603、I642、S727、A736、S1657、Q1659、E1661、I1668、D1681、R1776、H1859、A1993、H2007、N2019、K2085、K2207、F2275、S2296、V2314、Q2316及びM2321を挙げることができる。従って、一実施態様において、本発明の改変体は、前記アミノ酸残基のうちの少なくとも1個が他のアミノ酸でさらに置換されたものである。 As long as the variant of the present invention maintains the properties of higher specific activity and/or secretory expression efficiency compared to the wild type, in addition to at least one amino acid substitution selected from the group consisting of K213, S367 and F2196, amino acid residues at positions other than K213, S367 and F2196 may be substituted with other amino acids. Examples of such substitution sites include R-5, P25, A28, L152, M217, W228, Q410, Y487, R489, F501, M539, I566, L603, I642, S727, A736, S1657, Q1659, E1661, I1668, D1681, R1776, H1859, A1993, H2007, N2019, K2085, K2207, F2275, S2296, V2314, Q2316, and M2321. Accordingly, in one embodiment, the variant of the present invention is one in which at least one of the amino acid residues is further substituted with another amino acid.
当該他のアミノ酸としては、アミノ酸置換の結果得られるヒト第VIII因子改変体が、依然として、野生型と比べて比活性及び/又は分泌発現効率が増強されたものである限り、特に制限はないが、好ましくは、R-5P、P25H、A28T、L152A、L152D、L152E、L152H、L152I、L152M、L152N、L152P、L152Q又はL152V、M217T、W228Q、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321Lからなる群より選択される少なくとも1個のアミノ酸置換である。
より好ましくは、当該他のアミノ酸置換として、
(i)L152A、L152D、L152E、L152H、L152I、L152M、L152N、L152P、L152Q又はL152V、並びに/或いは
(ii)I1668F及びD1681G
を挙げることができる。
The other amino acids are not particularly limited as long as the human factor VIII variant obtained as a result of the amino acid substitution still has enhanced specific activity and/or secretory expression efficiency compared to the wild-type, but are preferably R-5P, P25H, A28T, L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q or L152V, M217T, W228Q, Q410L, Y487H, and at least one amino acid substitution selected from the group consisting of R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N2019K, K2085M, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
More preferably, the other amino acid substitution is:
(i) L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q, or L152V, and/or (ii) I1668F and D1681G
Examples include:
本発明の特に好ましい一実施態様として、前記NPL変異に加えて上記(i)のアミノ酸置換を含む(以下、「XNPL変異」という場合があり、L152がプロリン(P)で置換されている場合を「PNPL変異」という場合がある)改変体が挙げられる。PNPL変異をはじめとするXNPL変異を有する改変体は、野生型と比較して高比活性であることに加えて、さらに、野生型と比較して高分泌発現するという特性を有し得る。従って、当該改変体は、NPL変異により付与される利点に加えて、分泌効率の向上により小胞体ストレスの誘導を抑制し、ヒト第VIII因子による治療効果の長期安定性を実現し得るという更なる利点を有し得る。 A particularly preferred embodiment of the present invention is a variant that contains the amino acid substitution (i) above in addition to the NPL mutation (hereinafter, this may be referred to as an "XNPL mutation," and when L152 is substituted with proline (P), this may be referred to as a "PNPL mutation"). Variants having XNPL mutations, including PNPL mutations, not only have a higher specific activity than the wild-type, but can also have the property of being secreted at a higher level than the wild-type. Therefore, in addition to the advantages conferred by the NPL mutation, these variants may have the further advantage of suppressing the induction of endoplasmic reticulum stress by improving secretion efficiency, thereby realizing long-term stability of the therapeutic effects of human factor VIII.
本発明の別の好ましい一実施態様として、前記NPL変異又はXNPL変異に加えて、上記(ii)のアミノ酸置換(以下、「FG変異」という場合がある)を含む改変体が挙げられる。FG変異を有する改変体は、I1668F及びD1681Gのいずれかの変異を有しない対応するヒト第VIII因子と比較して、軽鎖の細胞内貯留が著明に抑制されるので、分泌効率及び/又は重鎖及び軽鎖の再構成において更なる改善が認められ得る。 Another preferred embodiment of the present invention is a variant that includes the amino acid substitution (ii) above (hereinafter sometimes referred to as "FG mutation") in addition to the NPL mutation or XNPL mutation. Compared to the corresponding human factor VIII lacking either the I1668F or D1681G mutation, the variant with the FG mutation exhibits significantly reduced intracellular retention of the light chain, and therefore further improvements in secretion efficiency and/or heavy and light chain reconstitution can be observed.
好ましい一実施態様において、本発明の改変体は、
K213におけるアミノ酸置換がK213Nであり、
M539L、並びに、任意選択でI566M、L603P及びI642Vからなる群より選択される少なくとも1個のA2ドメイン内のアミノ酸置換をさらに含み、
K2207Q、Q2316H及びM2321L、並びに、任意選択でF2275L、S2296A及びV2314Aからなる群より選択される少なくとも1個のC2ドメイン内のアミノ酸置換をさらに含む。K213N、M539L、K2207Q、Q2316H及びM2321Lのアミノ酸置換を少なくとも含むことにより、前記NPL変異を含まなくとも、野生型ヒト第VIII因子と比較して高分泌発現であるという特性が付与され得る。当該改変体の分泌発現効率は、前記FG変異をさらに含むことでより増強され得る。
In a preferred embodiment, the variant of the present invention is
the amino acid substitution at K213 is K213N;
M539L, and optionally at least one amino acid substitution in the A2 domain selected from the group consisting of I566M, L603P, and I642V;
The variant further comprises at least one amino acid substitution in the C2 domain selected from the group consisting of K2207Q, Q2316H, and M2321L, and optionally F2275L, S2296A, and V2314A. By including at least the amino acid substitutions K213N, M539L, K2207Q, Q2316H, and M2321L, the variant can be conferred the property of high secretory expression compared to wild-type human factor VIII, even without the NPL mutation. The secretory expression efficiency of the variant can be further enhanced by further including the FG mutation.
別の好ましい一実施態様において、本発明の改変体は、前記NPL変異に加えて、L152P、Y487H及びL603Pのアミノ酸置換をさらに含み、任意選択でS727P、Q1659E、H1859R、A1993V、H2007Q、N2019K及びK2085Mからなる群より選択される少なくとも1個のアミノ酸置換をさらに含む。当該改変体はPNPL変異を含むので、野生型と比較して高比活性でかつ高分泌発現する。 In another preferred embodiment, the variant of the present invention further comprises, in addition to the NPL mutation, amino acid substitutions L152P, Y487H, and L603P, and optionally at least one amino acid substitution selected from the group consisting of S727P, Q1659E, H1859R, A1993V, H2007Q, N2019K, and K2085M. Because the variant contains the PNPL mutation, it has a higher specific activity and is expressed at a higher level of secretion compared to the wild-type.
別の好ましい一実施態様において、本発明の改変体は、前記NPL変異に加えて、M539L、I566M、L603P、I642V、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321Lのアミノ酸置換をさらに含む。当該改変体は、上記10個のアミノ酸置換を含む更なるアミノ酸変異を有することにより、NPL変異による高比活性という特性が減衰する場合があり得る一方で、L152におけるアミノ酸置換(例、L152P)の有無にかかわらず(即ち、XNPL変異を有さなくとも)、分泌発現効率が増強されている。当該改変体の分泌発現効率は、前記FG変異をさらに含むことでより増強され得る。 In another preferred embodiment, the variant of the present invention further comprises the following amino acid substitutions in addition to the NPL mutation: M539L, I566M, L603P, I642V, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L. While the high specific activity characteristic of the NPL mutation may be attenuated by the presence of additional amino acid mutations including the 10 amino acid substitutions, the secretory expression efficiency of the variant is enhanced regardless of the presence or absence of an amino acid substitution at L152 (e.g., L152P) (i.e., even without the XNPL mutation). The secretory expression efficiency of the variant can be further enhanced by further including the FG mutation.
本発明の好ましい実施態様として、野生型ヒト第VIII因子において、以下の(a)~(n)のアミノ酸置換を有するヒト第VIII因子改変体を挙げることができる。
(a)K213N、S367P及びF2196L
(b)K213N、S367P、F2196L、I1668F及びD1681G
(c)K213N、M539L、I566M、I642V、K2207Q、F2275L、Q2316H及びM2321L
(d)K213N、M539L、I566M、L603P、I642V、F2196L、K2207Q、F2275L、Q2316H及びM2321L
(e)K213N、M539L、I566M、I642V、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(f)K213N、M539L、I566M、I642V、I1668F、D1681G、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(g)L152P、K213N、S367P及びF2196L
(h)L152P、K213N、S367P、Y487H、L603P及びF2196L
(i)L152P、K213N、S367P、Y487H、L603P、S727P、Q1659E及びF2196L
(j)L152P、K213N、S367P、Y487H、L603P、S727P、Q1659E、H1859R、A1993V、H2007Q、K2085M及びF2196L
(k)K213N、S367P、M539L、I566M、L603P、I642V、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(l)P25H、A28T、K213N、M217T、S367P、Q410L、Y487H、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、N2019K、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(m)R-5P、P25H、A28T、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、N2019K、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(n)R-5P、P25H、A28T、L152P、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
Preferred embodiments of the present invention include modified human factor VIII having the following amino acid substitutions (a) to (n) in wild-type human factor VIII:
(a) K213N, S367P, and F2196L
(b) K213N, S367P, F2196L, I1668F, and D1681G
(c) K213N, M539L, I566M, I642V, K2207Q, F2275L, Q2316H, and M2321L
(d) K213N, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, Q2316H, and M2321L
(e) K213N, M539L, I566M, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L
(f) K213N, M539L, I566M, I642V, I1668F, D1681G, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L
(g) L152P, K213N, S367P and F2196L
(h) L152P, K213N, S367P, Y487H, L603P and F2196L
(i) L152P, K213N, S367P, Y487H, L603P, S727P, Q1659E, and F2196L
(j) L152P, K213N, S367P, Y487H, L603P, S727P, Q1659E, H1859R, A1993V, H2007Q, K2085M and F2196L
(k) K213N, S367P, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(l) P25H, A28T, K213N, M217T, S367P, Q410L, Y487H, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P , Q1659E, E1661K, I1668F, D1681G, R1776K, N2019K, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(m) R-5P, P25H, A28T, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736 V, S1657P, Q1659E, E1661K, I1668F, D1681G, R1776K, N2019K, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487 H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1 659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N201 9K, K2085M, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
ヒト第VIII因子のBドメイン中には、各種タンパク質との相互作用領域がなく、Bドメインは循環血液中でのコファクター活性に寄与しないので、Bドメインを欠失しても凝固因子活性は保持される。従って、本発明の改変体は、Bドメインを欠失していてもよい。「Bドメインを欠失している」とは、Bドメイン(741~1648位)のアミノ酸配列を完全に欠失している場合だけでなく、BドメインのN末端及び/又はC末端の1~15アミノ酸残基程度のアミノ酸配列をa2領域とa3領域との間に保持している場合も包含する。Bドメイン欠失(BDD)ヒト第VIII因子改変体は、血友病Aに対する遺伝子治療においてAAVベクターを利用する場合に利点を有する。AAVベクターは搭載可能な核酸のサイズが4.7kbpと小さいが、全長ヒト第VIII因子は2351アミノ酸からなるので、コード領域だけで7kbpを超え、AAVベクターに搭載することができない。一方、BDD-ヒト第VIII因子のコード配列は4.4kbp未満であり、プロモーター配列等を含めた発現カセット全体でもAAVベクターのパッケージングサイズ内に収まる。 The B domain of human factor VIII does not contain regions that interact with various proteins and does not contribute to cofactor activity in circulating blood; therefore, deletion of the B domain retains coagulation factor activity. Therefore, the variants of the present invention may lack the B domain. "B domain deletion" refers not only to the complete deletion of the amino acid sequence of the B domain (positions 741-1648), but also to the retention of an amino acid sequence of approximately 1-15 amino acid residues at the N-terminus and/or C-terminus of the B domain between the a2 and a3 regions. B domain-deleted (BDD) human factor VIII variants are advantageous when using AAV vectors in gene therapy for hemophilia A. Although AAV vectors can carry a small nucleic acid size of 4.7 kbp, full-length human factor VIII consists of 2351 amino acids, so the coding region alone exceeds 7 kbp and cannot be carried by AAV vectors. On the other hand, the coding sequence for BDD-human factor VIII is less than 4.4 kbp, and the entire expression cassette, including the promoter sequence, fits within the packaging size of an AAV vector.
好ましい一実施態様において、本発明の改変体は、Bドメインのうち、744~1637位(配列番号1で表されるアミノ酸配列中、763位~1656位に相当する)を欠失する(即ち、S743とQ1638とが連結している;「FVIII SQ」又は「SQ」という場合がある)。FVIII SQ以外の、Bドメインの両末端の一部を残すBドメインの欠失、あるいはBドメインの3~4個のアルギニン(R)残基による置換など、効率的なヒト第VIII因子のプロセッシングを導くBDD-ヒト第VIII因子は知られており(例えば、Lind et al.,Eur.J.Biochem.232,19-27(1995))、本発明においても利用することができる。 In a preferred embodiment, the variant of the present invention lacks positions 744 to 1637 of the B domain (corresponding to positions 763 to 1656 in the amino acid sequence represented by SEQ ID NO: 1) (i.e., S743 is linked to Q1638; this may be referred to as "FVIII SQ" or "SQ"). BDD-human factor VIII that leads to efficient human factor VIII processing is known, other than FVIII SQ, including deletions of the B domain that leave portions of both termini of the B domain, or substitutions of the B domain with three or four arginine (R) residues (e.g., Lind et al., Eur. J. Biochem. 232, 19-27 (1995)), and these can also be used in the present invention.
一方で、Bドメインは第VIII因子の細胞内輸送に関与することが知られているので、分泌効率の向上を企図して、本発明の改変体はBドメインの全部又は一部を含むものであってもよい。例えば、遺伝子治療において、よりパッケージングサイズの大きいレンチウイルス(LV)ベクターやアデノウイルス(AdV)ベクターを用いる場合や、本発明の改変体をコードするmRNAをmRNA医薬として用いる場合、あるいは、本発明の改変体を組換え生産し、タンパク質製剤として使用する場合などには、Bドメインを含む改変体を用いることもまた好ましい。 On the other hand, since the B domain is known to be involved in the intracellular transport of factor VIII, the variants of the present invention may contain all or part of the B domain in order to improve secretion efficiency. For example, when using lentivirus (LV) vectors or adenovirus (AdV) vectors with larger packaging sizes in gene therapy, when using mRNA encoding the variants of the present invention as mRNA pharmaceuticals, or when recombinantly producing the variants of the present invention and using them as protein preparations, it is also preferable to use variants that contain the B domain.
本発明の改変体は、それをコードする核酸を、治療対象である動物にインビボ又はエクスビボ投与することにより、該動物体内で発現するものであってもよいし、該核酸を適切な宿主細胞に導入して発現させ、該宿主細胞を培養することにより得られる組換えタンパク質であってもよい。いずれにしても、本発明の改変体は、それをコードする核酸をもとに製造されるので、本発明はまた、上記いずれかの本発明の改変体をコードする核酸を提供する。 The variants of the present invention may be expressed in the animal to be treated by administering a nucleic acid encoding them in vivo or ex vivo to the animal, or they may be recombinant proteins obtained by introducing the nucleic acid into suitable host cells, allowing it to be expressed, and culturing the host cells. In either case, the variants of the present invention are produced based on the nucleic acid that encodes them, and the present invention also provides nucleic acids encoding any of the above variants of the present invention.
本発明に使用される「ヒト第VIII因子改変体をコードする核酸」は、DNAであってもRNAであってもよく、あるいはDNA/RNAキメラであってもよい。また、該核酸は二本鎖であっても、一本鎖であってもよい。二本鎖の場合は、二本鎖DNA、二本鎖RNAまたはDNA:RNAのハイブリッドでもよい。一本鎖の場合は、センス鎖(即ち、コード鎖)であっても、アンチセンス鎖(即ち、非コード鎖)であってもよい。核酸の種類はその用途(使用するベクターの種類等)に応じて適宜選択することができるが、好ましくはDNA、より好ましくは二本鎖DNAである。また、該核酸は酸または塩基との生理学的に許容される塩であってもよく、例えば、生理学的に許容される酸付加塩が好ましい。この様な塩としては、中でも、無機酸(例えば、塩酸、リン酸、臭化水素酸、硫酸)との塩、あるいは有機酸(例えば、酢酸、ギ酸、プロピオン酸、フマル酸、マレイン酸、コハク酸、酒石酸、クエン酸、リンゴ酸、蓚酸、安息香酸、メタンスルホン酸、ベンゼンスルホン酸)との塩などが用いられる。
ヒト第VIII因子改変体をコードするDNAとしては、ゲノムDNA、ヒトもしくは他の哺乳動物の細胞・組織由来のcDNA(cRNA)、合成DNA(RNA)などが挙げられる。
The "nucleic acid encoding a human factor VIII variant" used in the present invention may be DNA or RNA, or may be a DNA/RNA chimera. The nucleic acid may be double-stranded or single-stranded. If double-stranded, it may be double-stranded DNA, double-stranded RNA, or a DNA:RNA hybrid. If single-stranded, it may be a sense strand (i.e., coding strand) or an antisense strand (i.e., non-coding strand). The type of nucleic acid can be appropriately selected depending on its application (such as the type of vector used), but is preferably DNA, more preferably double-stranded DNA. The nucleic acid may also be a physiologically acceptable salt with an acid or base, for example, a physiologically acceptable acid addition salt. Among such salts, salts with inorganic acids (e.g., hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or salts with organic acids (e.g., acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) are used.
DNA encoding human factor VIII variants includes genomic DNA, cDNA (cRNA) derived from cells or tissues of humans or other mammals, and synthetic DNA (RNA).
より具体的には、「本発明の改変体をコードする核酸」として、例えば、配列番号1で表される野生型ヒト第VIII因子のアミノ酸配列、あるいは、該アミノ酸配列からなるタンパク質の天然のアレル変異体もしくは遺伝子多型(例、SNP等)のアミノ酸配列において、上記したいずれかのアミノ酸置換を有する、改変アミノ酸配列をコードする任意のヌクレオチド配列を挙げることができる。好ましくは、当該ヌクレオチド配列は、使用する宿主細胞、好ましくはヒト細胞での発現のためにコドン最適化された配列である。遺伝子発現に際し、そのヌクレオチド配列を宿主生物において使用頻度の高いコドンに変換することで、タンパク質発現量の増大が期待できる。使用する宿主におけるコドン使用頻度のデータは、例えば(公財)かずさDNA研究所のホームページに公開されている遺伝暗号使用頻度データベース(http://www.kazusa.or.jp/codon/index.html)を用いることができ、または各宿主におけるコドン使用頻度を記した文献を参照してもよい。あるいは、後述の実施例において用いられるように、自体公知のコドン最適化アルゴリズム(例、GeneArt Codon Optimizer)を用いて、コドン最適化を行うこともできる。そのようなアルゴリズムにおいては、宿主での使用頻度に加えて、GC含量、不安定化RNAエレメントの除去、隠れたスプライス部位の除去、遺伝子内polyAサイトの除去、繰り返し配列の除去、RNA2次構造の回避、IRESの除去などの複数のパラメータが考慮され得る。上記のようにして得られるコドン最適化された配列のDNA鎖を化学的に合成するか、あるいは、化学合成した一部オーバーラップするオリゴDNA短鎖を、PCR法やGibson Assembly法を利用して接続することにより、所望の本発明の改変体をコードするDNAを構築することが可能である。 More specifically, examples of "nucleic acids encoding variants of the present invention" include any nucleotide sequence encoding a variant amino acid sequence having any of the above-mentioned amino acid substitutions in the amino acid sequence of wild-type human factor VIII represented by SEQ ID NO: 1, or in the amino acid sequence of a natural allelic variant or genetic polymorphism (e.g., SNP, etc.) of a protein consisting of said amino acid sequence. Preferably, the nucleotide sequence is codon-optimized for expression in the host cell to be used, preferably a human cell. During gene expression, converting the nucleotide sequence to codons frequently used in the host organism can be expected to increase the amount of protein expression. Data on codon usage in the host to be used can be obtained, for example, from the genetic code usage database published on the website of the Kazusa DNA Research Institute (http://www.kazusa.or.jp/codon/index.html), or references listing codon usage in each host can be referenced. Alternatively, as used in the examples described below, codon optimization can be performed using a publicly known codon optimization algorithm (e.g., GeneArt Codon Optimizer). Such algorithms can take into account multiple parameters, such as GC content, removal of destabilizing RNA elements, removal of cryptic splice sites, removal of intragenic polyA sites, removal of repetitive sequences, avoidance of RNA secondary structures, and removal of IRES, in addition to frequency of codon usage in the host. DNA encoding the desired variant of the present invention can be constructed by chemically synthesizing a DNA strand with the codon-optimized sequence obtained as described above, or by connecting chemically synthesized, partially overlapping short oligo-DNA strands using PCR or Gibson Assembly.
好ましい一実施態様において、本発明の改変体をコードする核酸として、配列番号1で表されるアミノ酸配列からなる野生型ヒト第VIII因子において、前記(n)R-5P、P25H、A28T、L152P、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321Lのアミノ酸置換を有する改変体をコードし、ヒト用にコドン最適化された、配列番号2で表されるヌクレオチド配列からなる核酸を挙げることができる。 In a preferred embodiment, the nucleic acid encoding the variant of the present invention is a wild-type human factor VIII consisting of the amino acid sequence represented by SEQ ID NO: 1, wherein (n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, An example of such a nucleic acid is a nucleic acid consisting of the nucleotide sequence represented by SEQ ID NO: 2, which is codon-optimized for human use and encodes a variant having the amino acid substitutions Q1659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N2019K, K2085M, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L.
目的の宿主(血友病Aの遺伝子治療又はmRNA医薬における使用においてはヒト)用にコドン最適化されたヌクレオチド配列は、該配列内に1以上のCpG配列を有し得るが、CpG配列は宿主における免疫応答を引き起こし、持続的な導入遺伝子の発現を阻害する可能性があると予測し、該改変体のアミノ酸配列を変化させることなく、これらのCpG配列を除去する塩基置換を行った上で、再度コドン最適化し直すことにより、宿主細胞内での発現効率を著明に増大させることができる。例えば、配列番号2で表されるヌクレオチド配列からなる、ヒト用にコドン最適化された改変体コード核酸において、それがコードするアミノ酸配列を変化させることなくCpG配列を含まないように改変することができる。そのようなCpG除去され、かつコドン最適化し直されたヌクレオチド配列として、配列番号3で表されるヌクレオチド配列を挙げることができる。 A nucleotide sequence codon-optimized for a target host (humans in the case of use in gene therapy for hemophilia A or mRNA pharmaceuticals) may contain one or more CpG sequences within the sequence. However, it is predicted that CpG sequences may induce an immune response in the host and inhibit continuous transgene expression. Therefore, by making base substitutions to remove these CpG sequences without changing the amino acid sequence of the variant, and then re-optimizing the variant, the expression efficiency within host cells can be significantly increased. For example, a nucleic acid encoding a variant codon-optimized for humans, consisting of the nucleotide sequence represented by SEQ ID NO: 2, can be modified so that it does not contain any CpG sequences without changing the amino acid sequence it encodes. An example of such a nucleotide sequence from which CpG has been removed and which has been re-codon-optimized is the nucleotide sequence represented by SEQ ID NO: 3.
CpG配列の除去による発現効率の向上は、上記の特定のヒト第VIII因子改変体のみならず、任意の改変体、さらには野生型ヒト第VIII因子をコードする核酸においても当該効果を奏する。従って、本発明はまた、目的の宿主、好ましくはヒト用にコドン最適化されたヒト第VIII因子をコードする核酸において、それがコードするアミノ酸配列を変化させることなくCpG配列を含まないように改変した上で、さらにコドン最適化し直したヌクレオチド配列からなるヒト第VIII因子コード核酸を提供する。例えば、配列番号1で表されるアミノ酸配列からなる野生型ヒト第VIII因子をコードする、ヒト用にコドン最適化されたヌクレオチド配列として、配列番号4で表されるヌクレオチド配列が挙げられるが、当該配列においてコードするアミノ酸配列を変化させずにCpG配列を含まないように改変した上で、さらにヒト用にコドン最適化し直したヌクレオチド配列として、配列番号5で表されるヌクレオチド配列を挙げることができる。 The improvement in expression efficiency achieved by removing CpG sequences is effective not only for the specific human factor VIII variants described above, but also for any variants, and even for nucleic acids encoding wild-type human factor VIII. Therefore, the present invention also provides a human factor VIII-encoding nucleic acid comprising a nucleotide sequence obtained by modifying a nucleic acid encoding human factor VIII that has been codon-optimized for a target host, preferably for humans, so that it does not contain CpG sequences without changing the amino acid sequence it encodes, and then further codon-optimizing the nucleotide sequence. For example, an example of a nucleotide sequence that has been codon-optimized for humans and encodes wild-type human factor VIII consisting of the amino acid sequence represented by SEQ ID NO: 1 is the nucleotide sequence represented by SEQ ID NO: 4. An example of a nucleotide sequence obtained by modifying the sequence so that it does not contain CpG sequences without changing the amino acid sequence it encodes, and then further codon-optimizing the nucleotide sequence for humans is the nucleotide sequence represented by SEQ ID NO: 5.
本発明の改変体をコードする核酸、又はコドン最適化とCpG配列の除去とにより宿主細胞内での発現効率が増強されたヒト第VIII因子コード核酸(それらを包括して、以下「本発明の核酸」という場合がある)は、肝臓細胞で機能的なプロモーターの下流に連結してベクター中に挿入することにより、肝臓細胞内で発現可能な発現ベクターを構築することができる。 A nucleic acid encoding a variant of the present invention, or a nucleic acid encoding human factor VIII whose expression efficiency in host cells has been enhanced by codon optimization and removal of CpG sequences (hereinafter collectively referred to as the "nucleic acid of the present invention") can be linked downstream of a promoter functional in liver cells and inserted into a vector to construct an expression vector that can be expressed in liver cells.
肝臓は、胆汁の産生や物質代謝などの肝臓としての主要な機能を担う肝実質細胞(肝細胞)の他に、肝類洞内皮細胞、クッパー細胞、肝星細胞、ピット細胞、胆管上皮細胞、中皮細胞などの肝非実質細胞とから構成されている。本明細書において「肝臓細胞」とは、肝臓を構成する前記細胞群(それら由来のがん細胞や細胞株を含む)の1種以上を意味する。 The liver is composed of hepatic parenchymal cells (hepatocytes), which are responsible for the liver's main functions, such as bile production and metabolism, as well as non-parenchymal liver cells such as hepatic sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, pit cells, bile duct epithelial cells, and mesothelial cells. As used herein, "liver cells" refers to one or more of the above cell groups that make up the liver (including cancer cells and cell lines derived from them).
本明細書において「肝臓細胞で機能的なプロモーター」とは、肝臓細胞内で、その下流に連結された遺伝子(タンパク質をコードする核酸)の転写を誘導し得るプロモーターを意味するが、本発明の好ましい用途がインビボ遺伝子治療である点を考慮すると、該プロモーターは、肝臓細胞で特異的に機能する(下流遺伝子の転写を誘導する)ものであることが望ましい。ここで「肝臓細胞で特異的に」、「肝臓特異的」とは、肝臓細胞のみに発現が限局される場合の他、他の臓器・組織の細胞での発現と比較して、肝臓細胞での発現が有意に高い場合も包含する意図で用いられる。肝臓細胞以外での遺伝子発現を誘導する場合でも、例えば、臓器・組織・細胞指向性の高いベクター(例、ウイルスベクターの各種血清型)と組み合わせることで、実質的に(例えば、望ましくない副作用を引き起こさない程度に)肝臓特異的な遺伝子発現を達成し得る。
該プロモーターはbasalな転写活性を発揮する最小限のヌクレオチド配列を含む限り、他の調節配列(例、内因性の近位又は遠位のエンハンサー配列、他の遺伝子由来のエンハンサー配列等)をさらに含んでいてもよい。
As used herein, a "promoter functional in liver cells" refers to a promoter capable of inducing transcription of a gene (a nucleic acid encoding a protein) linked downstream in liver cells. However, considering that the preferred use of the present invention is in vivo gene therapy, it is desirable that the promoter function specifically in liver cells (inducing transcription of a downstream gene). Here, the terms "specifically in liver cells" and "liver-specific" are used to encompass not only cases in which expression is limited to liver cells, but also cases in which expression in liver cells is significantly higher than expression in cells of other organs or tissues. Even when inducing gene expression in cells other than liver cells, for example, by combining it with a vector with high organ/tissue/cell tropism (e.g., various serotypes of viral vectors), substantially liver-specific gene expression (e.g., to the extent that it does not cause undesirable side effects) can be achieved.
As long as the promoter contains a minimal nucleotide sequence that exerts basal transcriptional activity, it may further contain other regulatory sequences (e.g., endogenous proximal or distal enhancer sequences, enhancer sequences derived from other genes, etc.).
肝臓細胞で機能的なプロモーターとしては、肝臓細胞内で下流遺伝子の転写を誘導し得る限り特に限定されないが、好ましくは、肝臓で高発現する遺伝子のプロモーターであり、より好ましくは、肝臓特異的なプロモーターである。ここで「肝臓特異的」とは前記と同義である。本発明の発現ベクターに用いることのできる肝臓細胞で機能的なプロモーターとしては、例えば、トランスサイレチン(TTR)プロモーター、α1-アンチトリプシン(AAT)プロモーター、アルブミンプロモーター、α-フェトプロテインプロモーター、チロキシン結合グロブリンプロモーター等が挙げられるが、それらに限定されない。好ましい一実施態様において、例えば、AAVベクターを用いた血友病Aの遺伝子治療への使用を目的とする場合、肝臓細胞で機能的なプロモーターとして、TTRプロモーターやAATプロモーターを用いることができる。別の好ましい実施態様においては、肝臓細胞で機能的なプロモーターとして、ヒトAATプロモーターとApo E/C1遺伝子の肝臓型制御領域(HCR)とのキメラプロモーター(HCRhAAT)を用いることもできる。 Promoters functional in liver cells are not particularly limited as long as they can induce transcription of downstream genes in liver cells. However, they are preferably promoters of genes highly expressed in the liver, and more preferably liver-specific promoters. Here, "liver-specific" has the same meaning as above. Examples of promoters functional in liver cells that can be used in the expression vectors of the present invention include, but are not limited to, the transthyretin (TTR) promoter, the α1-antitrypsin (AAT) promoter, the albumin promoter, the α-fetoprotein promoter, and the thyroxine-binding globulin promoter. In a preferred embodiment, for example, when using an AAV vector for gene therapy of hemophilia A, the TTR promoter or the AAT promoter can be used as a promoter functional in liver cells. In another preferred embodiment, a chimeric promoter (HCRhAAT) composed of the human AAT promoter and the liver-type regulatory region (HCR) of the Apo E/C1 gene can also be used as a promoter functional in liver cells.
肝臓細胞で機能的なプロモーターは、各プロモーターの由来する遺伝子(ゲノムDNA)の配列情報に基づいて、自体公知の方法により、取得することができる。それらの配列情報は、NCBI、EMBL、FASTA、DDBJ等の公共に利用可能なデータベースに登録されており、また、文献に記載されている。当業者は容易に該配列情報にアクセスすることができる。 Promoters functional in liver cells can be obtained by known methods based on the sequence information of the gene (genomic DNA) from which each promoter is derived. Such sequence information is registered in publicly available databases such as NCBI, EMBL, FASTA, and DDBJ, and is also described in literature. Those skilled in the art can easily access this sequence information.
肝臓細胞で機能的なプロモーターのヌクレオチド配列が1以上のCpG配列を含む場合、下流に連結される本発明の核酸の宿主細胞内での発現効率を増強させるために、本発明の核酸の場合と同様に、当該CpG配列を除去するように改変することが望ましい。例えば、野生型マウスTTRプロモーターは配列番号6で表されるヌクレオチド配列からなるが、該配列中のCpG配列を除去した、配列番号7で表されるヌクレオチド配列からなる改変プロモーターを使用することができる。 If the nucleotide sequence of a promoter functional in liver cells contains one or more CpG sequences, it is desirable to modify it to remove the CpG sequences, as in the case of the nucleic acid of the present invention, in order to enhance the expression efficiency in host cells of the nucleic acid of the present invention linked downstream. For example, while the wild-type mouse TTR promoter consists of the nucleotide sequence represented by SEQ ID NO: 6, a modified promoter can be used in which the CpG sequences in the wild-type mouse TTR promoter have been removed, and the nucleotide sequence represented by SEQ ID NO: 7.
肝臓細胞で機能的なプロモーターと本発明の核酸との結合は、当該技術分野において自体公知の方法で行うことができる。例えば、該プロモーター及び該核酸の各フラグメントが平滑末端を有する場合、両フラグメントをDNAリガーゼを用いてライゲーションすることができる。あるいは、両フラグメントの末端に任意のアダプター配列を付加し、適当な制限酵素で処理して粘着末端を生じさせた後、両者をライゲーションしてもよい。 The linking of a promoter functional in liver cells to the nucleic acid of the present invention can be carried out by methods known in the art. For example, if the promoter and nucleic acid fragments each have blunt ends, the two fragments can be ligated using DNA ligase. Alternatively, any adapter sequence can be added to the ends of both fragments, treated with an appropriate restriction enzyme to generate sticky ends, and then the two can be ligated.
好ましい実施態様において、肝臓細胞で機能的なプロモーターの下流に連結された本発明の核酸は、その下流に肝臓細胞で機能的な転写終結シグナル、即ちpolyA付加シグナル(例、SV40 polyA付加シグナル)をさらに連結した発現カセットの形態で、ベクター中に挿入されることが好ましい。 In a preferred embodiment, the nucleic acid of the present invention linked downstream of a promoter functional in liver cells is preferably inserted into a vector in the form of an expression cassette further linked downstream of a transcription termination signal functional in liver cells, i.e., a polyA addition signal (e.g., an SV40 polyA addition signal).
例えば、(i)CpG配列を含まないように改変されたマウスTTRプロモーター(配列番号7)の下流に、(ii)野生型ヒト第VIII因子において、前記(n)R-5P、P25H、A28T、L152P、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321Lのアミノ酸置換を有する改変体をコードし、ヒト用にコドン最適化され、さらにCpG配列が除去され、かつコドン最適化し直されたヒト第VIII因子コード配列(配列番号3)、並びに(iii)SV40 polyA付加シグナル配列が連結された発現カセットが、好ましく用いられ得る。そのような発現カセットとして、配列番号8で表されるヌクレオチド配列(4814bp)からなる核酸が挙げられる。当該核酸は、配列番号7のプロモーター配列(1-282位)と配列番号3のコード配列(300-4673位)との間に、17ヌクレオチドからなる5’-上流配列(283-299位)を含むが、そのうち283-293位の11ヌクレオチドを欠失させ短縮化することで、ヒト第VIII因子の分泌発現効率を向上させることができる。配列番号8のヌクレオチド配列から当該11ヌクレオチドが欠失したヌクレオチド配列(4803bp)を配列番号9に示す。また、配列番号9において、SV40 polyA付加シグナル配列(4663-4803位)のうち、3’末端側の13ヌクレオチドを欠失させたヌクレオチド配列(4790bp)を配列番号10に、3’末端側の35ヌクレオチドを欠失させたヌクレオチド配列(4768bp)を配列番号11に示す。これらのpolyA付加シグナルの改変(短縮化)により、ヒト第VIII因子の分泌発現効率はさらに向上し得る。 For example, (i) downstream of a mouse TTR promoter (SEQ ID NO: 7) that has been modified so as not to contain a CpG sequence, (ii) in wild-type human factor VIII, (n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1659E, E1661K, I1668F, D1681G, R177 An expression cassette that encodes a variant having the amino acid substitutions 6K, H1859R, A1993V, H2007Q, N2019K, K2085M, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L, that has been codon-optimized for human use, further CpG sequence-removed, and re-codon-optimized (SEQ ID NO: 3), and (iii) is linked to an SV40 polyA additional signal sequence, can be preferably used. An example of such an expression cassette is a nucleic acid consisting of the nucleotide sequence (4814 bp) set forth in SEQ ID NO: 8. This nucleic acid contains a 17-nucleotide 5'-upstream sequence (positions 283-299) between the promoter sequence (positions 1-282) of SEQ ID NO: 7 and the coding sequence (positions 300-4673) of SEQ ID NO: 3. By deleting and shortening the 11 nucleotides at positions 283-293, the efficiency of secretory expression of human factor VIII can be improved. SEQ ID NO: 9 shows the nucleotide sequence (4803 bp) in which these 11 nucleotides have been deleted from the nucleotide sequence of SEQ ID NO: 8. Furthermore, SEQ ID NO: 10 shows the nucleotide sequence (4790 bp) in which 13 nucleotides from the 3' end of the SV40 polyA addition signal sequence (positions 4663-4803) of SEQ ID NO: 9 have been deleted, and SEQ ID NO: 11 shows the nucleotide sequence (4768 bp) in which 35 nucleotides from the 3' end have been deleted. By modifying (shortening) these polyA addition signals, the efficiency of secretory expression of human factor VIII can be further improved.
本発明の核酸が挿入されるベクターは、一般に遺伝子治療に用いられるものであれば特に制限はなく、例えば、AAVベクター、LVベクター、レトロウイルスベクター、AdVベクター、シンドビスウイルスベクター、狂犬病ウイルスベクター、センダイウイルスベクター、単純ヘルペスウイルスベクター等のウイルスベクター、動物細胞用プラスミド等の非ウイルスベクターを使用することができる。遺伝子導入・発現効率が高い、非分裂細胞にも導入可能である、導入遺伝子の長期的な発現が可能である等の観点から、AAVベクター、LVベクターが好ましく、染色体組込みが低頻度で挿入変異のリスクがなく、免疫原性も低く安全性が高いい等の観点から、AAVベクターを使用することがより好ましい。AAVベクターは分裂細胞では細胞増殖に伴い希釈されるが、成体の肝細胞はほとんど分裂しないので、導入遺伝子の発現は長期間保持され得る。但し、搭載可能な遺伝子サイズが4.7kbpと小さいため、導入する核酸のサイズによっては、より大きなインサートを搭載できるLVベクターやAdVベクターの使用もまた好ましい場合がある。 The vector into which the nucleic acid of the present invention is inserted is not particularly limited as long as it is one generally used in gene therapy. Examples of suitable vectors include viral vectors such as AAV vectors, LV vectors, retroviral vectors, AdV vectors, Sindbis virus vectors, rabies virus vectors, Sendai virus vectors, and herpes simplex virus vectors, as well as non-viral vectors such as plasmids for animal cells. AAV and LV vectors are preferred from the viewpoints of high gene transfer and expression efficiency, ability to transfer to non-dividing cells, and long-term expression of the introduced gene. AAV vectors are more preferred from the viewpoints of low frequency of chromosomal integration, elimination of the risk of insertional mutagenesis, low immunogenicity, and high safety. AAV vectors are diluted in dividing cells as the cells proliferate, but adult hepatocytes rarely divide, so expression of the introduced gene can be maintained for a long period of time. However, because the gene size that can be carried is small at 4.7 kbp, depending on the size of the nucleic acid to be introduced, it may be preferable to use LV or AdV vectors, which can carry larger inserts.
例えば、ベクターとしてAAVベクターを用いる場合、肝臓指向性の高い血清型由来のベクターを用いるのが望ましい。肝臓指向性を有する血清型としては、例えば、AAV1、AAV2、AAV3、AAV5、AAV6、AAV6.2、AAV7、AAV8、AAV9、AAVrh10、AAV-DJ、AAV-DJ/8等が挙げられる。以下に、AAVベクターを例にとって、ベクター構築の手順を説明するが、当業者であれば、他のウイルスベクターや非ウイルスベクター(例、プラスミドベクター)についても、自体公知の方法[例えば、Current Protocols in Molecular Biology, F. Ausubel et al. eds. (1994) John Wiley & Sons,Inc.; Molecular Cloning (A Laboratory Manual),3rd ed. Volumes 1-3, Josseph Sambrook & David W.Russeleds., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, New York)(2001); Culture of Animal Cells; A Manual of Basic Technique, R. Freshney eds., 2nd ed. (1987), Wiley-Liss; Frank L. Graham, Manipulation of adenovirus vector, Chapter 11. p109-p128; E.J. Murray eds., Methods in Molecular Biology, Vol.7, Gene Transfer and Expression Protocols (1991); Chen, S-H.et al., Combination gene therapy for liver metastases of colon carcinoma in vivo., Proc. Natl. Acad. Sci. USA (1995) 92, 2477-2581等]を用いて、容易に所望の発現ベクターを構築することができる。 For example, when using an AAV vector as a vector, it is desirable to use a vector derived from a serotype with high liver tropism. Examples of serotypes with liver tropism include AAV1, AAV2, AAV3, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAVrhlO, AAV-DJ, and AAV-DJ/8. Below, the procedure for constructing a vector will be explained using an AAV vector as an example. However, those skilled in the art will be able to construct other viral vectors and non-viral vectors (e.g., plasmid vectors) using methods known per se [e.g., Current Protocols in Molecular Biology, F. Ausubel et al. eds. (1994) John Wiley & Sons, Inc.; Molecular Cloning (A Laboratory Manual), 3rd ed. Volumes 1-3, Joseph Sambrook & David W. Russell, Cold Spring Harbor Laboratory Press (Cold Spring Harbor, New York) (2001); Culture of Animal Cells; A Manual of Basic The desired expression vector can be easily constructed using methods such as those described in "Adenovirus Technique," R. Freshney eds., 2nd ed. (1987), Wiley-Liss; Frank L. Graham, Manipulation of adenovirus vectors, Chapter 11, pp. 109-128; E.J. Murray eds., Methods in Molecular Biology, Vol. 7, Gene Transfer and Expression Protocols (1991); Chen, S-H. et al., Combination gene therapy for liver metastases of colon carcinoma in vivo, Proc. Natl. Acad. Sci. USA (1995) 92, 2477-2581.
AAV発現ベクターの作製において、本発明の合成プロモーターはまず、適切な宿主細胞(例、大腸菌、枯草菌、酵母等)内で増幅可能なプラスミド中に、AAVの5’-及び3’-ITRとともに、両ITRの間に挿入される。該プラスミドとしては、例えば、大腸菌由来のプラスミド(例、pBR322,pBR325,pUC12,pUC13)、枯草菌由来のプラスミド(例、pUB110,pTP5,pC194)、酵母由来のプラスミド(例、pSH19,pSH15)等が挙げられる。AAV ITRとしては、目的の血清型由来のITRを用いてもよいが、通常AAV2由来のITRを共通して使用することができる。両ITR配列の間には、本発明の合成プロモーターのみを挿入することもできるが、polyA付加シグナル(例、SV40 polyA付加シグナル)をその下流に挿入することが望ましく、該合成プロモーターとpolyA付加シグナルとの間に、さらにウッドチャック肝炎ウイルス転写後調節エレメント(WPRE)を挿入することもできる。WPREとして、短縮化されたもの(0.25kb;Mol.Brain,7:17,2014)を用いることもできる。本発明のプロモーターとpolyA付加シグナルとの間(WPREを含む場合は該プロモーターとWPREとの間)に1以上の制限酵素認識部位、好ましくはマルチクローニング部位(MCS)を挿入することにより、本発明の核酸の挿入(ヒト第VIII因子の発現カセットの構築)を容易にすることができる。 In constructing an AAV expression vector, the synthetic promoter of the present invention is first inserted between the 5'- and 3'-ITRs of AAV into a plasmid that can be amplified in a suitable host cell (e.g., Escherichia coli, Bacillus subtilis, yeast, etc.). Examples of such plasmids include E. coli-derived plasmids (e.g., pBR322, pBR325, pUC12, pUC13), Bacillus subtilis-derived plasmids (e.g., pUB110, pTP5, pC194), and yeast-derived plasmids (e.g., pSH19, pSH15). While AAV ITRs derived from the desired serotype may be used, AAV2-derived ITRs are commonly used. While the synthetic promoter of the present invention alone can be inserted between the two ITR sequences, it is preferable to insert a polyA addition signal (e.g., an SV40 polyA addition signal) downstream thereof, and a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) can also be inserted between the synthetic promoter and the polyA addition signal. A shortened WPRE (0.25 kb; Mol. Brain, 7:17, 2014) can also be used. Inserting one or more restriction enzyme recognition sites, preferably a multicloning site (MCS), between the promoter of the present invention and the polyA addition signal (or between the promoter and WPRE if the WPRE is included) can facilitate insertion of the nucleic acid of the present invention (construction of an expression cassette for human factor VIII).
得られたAAVベクター用発現プラスミドのプロモーターとpolyA付加シグナルとの間(WPREを含む場合は該プロモーターとWPREとの間)に、肝臓細胞で発現させたい本発明の核酸を挿入することにより、該プラスミド中にヒト第VIII因子の発現カセットが構築される。 By inserting the nucleic acid of the present invention to be expressed in liver cells between the promoter and polyA addition signal of the resulting AAV vector expression plasmid (between the promoter and WPRE if WPRE is included), an expression cassette for human factor VIII is constructed in the plasmid.
発現ベクターは、所望により、プロモーターの下流に宿主において機能し得る5’-UTR、ヒト第VIII因子をコードするDNAの下流に宿主において機能し得る3’-UTRをさらに含有することもできる。また、エンハンサー、スプライシングシグナル等を含んでいてもよい。これらの各構成要素としては、自体公知のものを用いることができる。 If desired, the expression vector can further contain a 5'-UTR downstream of the promoter that can function in the host, and a 3'-UTR downstream of the DNA encoding human factor VIII that can function in the host. It may also contain an enhancer, a splicing signal, etc. Each of these components can be made using known components.
肝臓細胞で機能的なプロモーターの下流に本発明の核酸を含むAAVベクター発現用プラスミドから、自体公知の方法、例えば、プラスミドトランスフェクション法、組換えバキュロウイルス法、組換えヘルペスウイルスベクター法、酵母法等により、AAVベクターを製造することができる。例えば、プラスミドトランスフェクション法では、前記AAVベクター発現用プラスミド、AAVのRep及びCap遺伝子を含むプラスミド、アデノウイルス由来のE2A、E4orf6、VARNA遺伝子を含むpHelperプラスミドをHEK293細胞等にトランスフェクトし、AAVウイルス粒子を産生させることができる。Rep遺伝子は目的の血清型由来である必要はなく、一般的にAAV2由来のRep遺伝子が用いられる。一方、Cap遺伝子は目的の血清型由来のものを用いる必要があり、Cap遺伝子にコードされるカプシドタンパク質によりAAV血清型の細胞指向性が決定される。即ち、肝臓細胞を標的とする本発明の発現ベクターにおいては、Cap遺伝子として、肝臓指向性を有するAAV1、AAV2、AAV3、AAV5、AAV6、AAV6.2、AAV7、AAV8、AAV9、AAVrh10、AAV-DJ、AAV-DJ/8等由来のCap遺伝子を用いることができる。 An AAV vector can be produced from an AAV vector expression plasmid containing the nucleic acid of the present invention downstream of a promoter functional in liver cells by methods known per se, such as the plasmid transfection method, recombinant baculovirus method, recombinant herpes virus vector method, yeast method, etc. For example, in the plasmid transfection method, the AAV vector expression plasmid, a plasmid containing the AAV Rep and Cap genes, and a pHelper plasmid containing the adenovirus-derived E2A, E4orf6, and VARNA genes are transfected into HEK293 cells or the like to produce AAV virus particles. The Rep gene does not need to be derived from the desired serotype; the Rep gene from AAV2 is generally used. On the other hand, the Cap gene must be derived from the desired serotype, and the cell tropism of the AAV serotype is determined by the capsid protein encoded by the Cap gene. That is, in the expression vector of the present invention that targets liver cells, Cap genes derived from liver-tropic viruses such as AAV1, AAV2, AAV3, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAVrhlO, AAV-DJ, and AAV-DJ/8 can be used as Cap genes.
一方、本発明の発現ベクターとして非ウイルスベクターを使用する場合、該発現ベクターの導入は、ポリL-リジン-核酸複合体などの高分子キャリアーを用いるか、リポソームに被包して行うことができる。リポソームはリン脂質からなる数10~数100nmの粒径のカプセルで、その内部にプラスミドベクターを封入できる。 On the other hand, when a non-viral vector is used as the expression vector of the present invention, the expression vector can be introduced using a polymeric carrier such as a poly-L-lysine-nucleic acid complex, or by encapsulation in a liposome. Liposomes are capsules made of phospholipids with a particle size of several tens to several hundreds of nanometers, and a plasmid vector can be encapsulated inside them.
肝臓細胞で機能的なプロモーターの下流に本発明の核酸を含む発現ベクターは、ヒト第VIII因子が治療効果を発揮する疾患、特に血友病Aに罹患した対象に投与することにより、該疾患を治療することができる。従って、本発明はまた、本発明の発現ベクターを含んでなる血友病Aの遺伝子治療剤を提供する。 An expression vector containing a nucleic acid of the present invention downstream of a promoter functional in liver cells can be administered to a subject suffering from a disease for which human factor VIII exerts a therapeutic effect, particularly hemophilia A, to treat the disease. Therefore, the present invention also provides a gene therapy agent for hemophilia A comprising the expression vector of the present invention.
本発明の遺伝子治療剤は、本発明の発現ベクターを原体のまま用いてもよいが、必要に応じて薬理学的に許容し得る担体とともに混合して注射剤などの種々の製剤形態とした後に医薬として用いることもできる。 The gene therapy agent of the present invention may be the expression vector of the present invention used as is, or, if necessary, may be mixed with a pharmacologically acceptable carrier to form various formulations, such as injections, and then used as a medicine.
ここで、薬理学的に許容される担体としては、製剤素材として慣用の各種有機あるいは無機担体物質が用いられ、液状製剤における溶剤、溶解補助剤、懸濁化剤、等張化剤、緩衝剤、無痛化剤などとして配合される。また必要に応じて、防腐剤、抗酸化剤、着色剤などの製剤添加物を用いることもできる。 Here, various organic or inorganic carrier substances commonly used as pharmaceutical ingredients are used as pharmacologically acceptable carriers, and are incorporated as solvents, solubilizers, suspending agents, isotonicity agents, buffers, soothing agents, etc. in liquid formulations. Furthermore, formulation additives such as preservatives, antioxidants, and coloring agents can also be used as needed.
溶剤の好適な例としては、注射用水、生理的食塩水、リンゲル液、アルコール、プロピレングリコール、ポリエチレングリコール、ゴマ油、トウモロコシ油、オリーブ油、綿実油などが挙げられる。 Suitable examples of solvents include water for injection, physiological saline, Ringer's solution, alcohol, propylene glycol, polyethylene glycol, sesame oil, corn oil, olive oil, cottonseed oil, etc.
溶解補助剤の好適な例としては、ポリエチレングリコール、プロピレングリコール、D-マンニトール、トレハロース、安息香酸ベンジル、エタノール、トリスアミノメタン、コレステロール、トリエタノールアミン、炭酸ナトリウム、クエン酸ナトリウム、サリチル酸ナトリウム、酢酸ナトリウムなどが挙げられる。 Suitable examples of solubilizing agents include polyethylene glycol, propylene glycol, D-mannitol, trehalose, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate, sodium salicylate, and sodium acetate.
懸濁化剤の好適な例としては、ステアリルトリエタノールアミン、ラウリル硫酸ナトリウム、ラウリルアミノプロピオン酸、レシチン、塩化ベンザルコニウム、塩化ベンゼトニウム、モノステアリン酸グリセリンなどの界面活性剤、例えばポリビニルアルコール、ポリビニルピロリドン、カルボキシメチルセルロースナトリウム、メチルセルロース、ヒドロキシメチルセルロース、ヒドロキシエチルセルロース、ヒドロキシプロピルセルロースなどの親水性高分子、ポリソルベート類、ポリオキシエチレン硬化ヒマシ油などが挙げられる。 Suitable examples of suspending agents include surfactants such as stearyl triethanolamine, sodium lauryl sulfate, lauryl aminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glycerin monostearate; hydrophilic polymers such as polyvinyl alcohol, polyvinylpyrrolidone, sodium carboxymethylcellulose, methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, and hydroxypropylcellulose; polysorbates; and polyoxyethylene hydrogenated castor oil.
等張化剤の好適な例としては、塩化ナトリウム、グリセリン、D-マンニトール、D-ソルビトール、ブドウ糖などが挙げられる。 Suitable examples of isotonic agents include sodium chloride, glycerin, D-mannitol, D-sorbitol, glucose, etc.
緩衝剤の好適な例としては、リン酸塩、酢酸塩、炭酸塩、クエン酸塩などの緩衝液などが挙げられる。 Suitable examples of buffering agents include buffer solutions such as phosphate, acetate, carbonate, and citrate.
無痛化剤の好適な例としては、ベンジルアルコールなどが挙げられる。 Suitable examples of soothing agents include benzyl alcohol.
防腐剤の好適な例としては、パラオキシ安息香酸エステル類、クロロブタノール、ベンジルアルコール、フェネチルアルコール、デヒドロ酢酸、ソルビン酸などが挙げられる。 Suitable examples of preservatives include parahydroxybenzoic acid esters, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, and sorbic acid.
抗酸化剤の好適な例としては、亜硫酸塩、アスコルビン酸塩などが挙げられる。 Suitable examples of antioxidants include sulfites and ascorbates.
着色剤の好適な例としては、水溶性食用タール色素(例:食用赤色2号及び3号、食用黄色4号及び5号、食用青色1号及び2号などの食用色素)、水不溶性レーキ色素(例:前記水溶性食用タール色素のアルミニウム塩など)、天然色素(例:β-カロチン、クロロフィル、ベンガラなど)などが挙げられる。 Suitable examples of coloring agents include water-soluble food tar dyes (e.g., food dyes such as Food Red No. 2 and No. 3, Food Yellow No. 4 and No. 5, and Food Blue No. 1 and No. 2), water-insoluble lake dyes (e.g., aluminum salts of the above-mentioned water-soluble food tar dyes), and natural dyes (e.g., β-carotene, chlorophyll, red iron oxide, etc.).
前記医薬組成物の剤形としては、例えば注射剤(例:皮下注射剤、静脈内注射剤、筋肉内注射剤、腹腔内注射剤など)、点滴剤等の非経口剤が挙げられる。 The dosage form of the pharmaceutical composition may be, for example, an injection (e.g., subcutaneous injection, intravenous injection, intramuscular injection, intraperitoneal injection, etc.), a drip infusion, or other parenteral preparation.
本発明の遺伝子治療剤は、製剤技術分野において慣用の方法、例えば日本薬局方に記載の方法等により製造することができる。該製剤中の有効成分であるウイルスベクターの含量は、剤形、有効成分の投与量などにより異なるが、例えば約0.1ないし100重量%である。ウイルス力価としては、例えば1×1010~1013vp/mL程度となるように適宜調整することができるが、この範囲に限定されない。 The gene therapy agent of the present invention can be produced by methods commonly used in the pharmaceutical technology field, such as the methods described in the Japanese Pharmacopoeia. The content of the viral vector, which is the active ingredient in the formulation, varies depending on the dosage form, the dose of the active ingredient, etc., but is, for example, about 0.1 to 100% by weight. The viral titer can be appropriately adjusted to, for example, about 1 x 10 10 to 10 13 vp/mL, but is not limited to this range.
非経口的な投与(例えば、静脈内注射、皮下注射、筋肉注射、腹腔内投与など)に好適な製剤としては、水性及び非水性の等張な無菌の注射液剤があり、これには抗酸化剤、緩衝液、制菌剤、等張化剤等が含まれていてもよい。また、水性及び非水性の無菌の懸濁液剤が挙げられ、これには懸濁剤、可溶化剤、増粘剤、安定化剤、防腐剤等が含まれていてもよい。本発明において最も好適な剤形は注射液剤である。 Preparations suitable for parenteral administration (e.g., intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal administration, etc.) include aqueous and non-aqueous isotonic sterile injection solutions, which may contain antioxidants, buffers, bacteriostats, isotonicity agents, etc. Also included are aqueous and non-aqueous sterile suspensions, which may contain suspending agents, solubilizers, thickeners, stabilizers, preservatives, etc. The most preferred dosage form in the present invention is an injection solution.
該製剤の投与量は、ベクターの種類、プロモーター活性、投与経路、病気の重篤度、投与対象となる動物種、投与対象の薬物受容性、体重、年齢等によって異なるが、例えば、血友病Aに対するヒト第VIII因子発現ベクターとして、HCRhAATプロモーターをbasalプロモーターとするAAVベクター(例、AAV8)を用いる場合、全身投与、特に末梢静脈からAAVベクターを投与する場合には、例えば、1回用量として約5×1012~約5×1013vp/kg体重で投与することができる。AAVベクターの最近の臨床試験において、高用量(3×1014vp/kg体重)投与群で重篤な肝障害による死亡例が報告されたことから(Audentes Therapeutics. Letter to the MTM disease community. https://myotubulartrust.org/audentes-therapeutics-letter-23-june-2020/;Hum Gene Ther 2020; 31: 695-696)、1014vp/kg体重以下の用量で投与することが望ましい。例えば、好ましい一実施態様において、本発明の遺伝子治療剤を、AAVベクター量として、約1×1010~約5×1013vp/kg体重、好ましくは約1×1011~約1×1013vp/kg体重、より好ましくは約5×1011~約5×1012vp/kg体重又は約1×1012~約1×1013vp/kg体重の用量で全身投与することができる。 The dosage of the formulation varies depending on the type of vector, promoter activity, administration route, severity of the disease, target animal species, drug tolerance, body weight, age, etc. of the target animal. For example, when an AAV vector (e.g., AAV8) with the HCRhAAT promoter as a basal promoter is used as a human factor VIII expression vector for hemophilia A, when the AAV vector is administered systemically, particularly via a peripheral vein, it can be administered at a single dose of, for example, about 5 × 10 12 to about 5 × 10 13 vp/kg body weight. In recent clinical trials of AAV vectors, deaths due to severe liver damage were reported in the high-dose (3 × 10 14 vp/kg body weight) administration group (Audentes Therapeutics. Letter to the MTM disease community. https://myotubulartrust.org/audentes-therapeutics-letter-23-june-2020/; Hum Gene Ther 2020; 31: 695-696), so it is desirable to administer at a dose of 10 14 vp/kg body weight or less. For example, in one preferred embodiment, the gene therapy agent of the present invention can be systemically administered at a dose of about 1×10 10 to about 5×10 13 vp/kg body weight, preferably about 1×10 11 to about 1×10 13 vp/kg body weight, more preferably about 5×10 11 to about 5×10 12 vp/kg body weight or about 1×10 12 to about 1×10 13 vp/kg body weight, in terms of the amount of AAV vector.
本発明はまた、本発明の発現ベクター又は本発明の核酸を含む遺伝子治療剤を、インビトロで肝臓細胞に導入することを含む、肝臓細胞においてヒト第VIII因子を発現させる方法を提供する。本発明の遺伝子治療剤でトランスフェクトされた肝臓細胞を、血友病Aを有する対象に移植することにより、該疾患のエクスビボ(ex vivo)治療法を提供することができる。従って、本発明はまた、本発明の遺伝子治療剤でトランスフェクトされた肝臓細胞を有効成分とする医薬(細胞製剤)、特に血友病の治療剤を提供する。 The present invention also provides a method for expressing human factor VIII in liver cells, which comprises introducing into liver cells in vitro a gene therapy agent containing the expression vector of the present invention or the nucleic acid of the present invention. By transplanting liver cells transfected with the gene therapy agent of the present invention into a subject with hemophilia A, an ex vivo treatment for the disease can be provided. Therefore, the present invention also provides a pharmaceutical (cell preparation), particularly a therapeutic agent for hemophilia, containing liver cells transfected with the gene therapy agent of the present invention as an active ingredient.
本発明の核酸が一本鎖RNAである場合、当該核酸をmRNA医薬の形態で製剤化することができる。例えば、前記発現ベクターを適当な宿主(例えば、哺乳動物細胞)に導入して培養し、自体公知の方法(例、LiCl法)を用いてmRNAを回収し、ヒト第VIII因子をコードするmRNAを精製することにより、該mRNAを取得することができる。 When the nucleic acid of the present invention is single-stranded RNA, the nucleic acid can be formulated in the form of an mRNA pharmaceutical. For example, the expression vector is introduced into a suitable host (e.g., mammalian cells) and cultured, and the mRNA is recovered using a method known per se (e.g., the LiCl method), and the mRNA encoding human factor VIII can be purified to obtain the mRNA.
あるいは、前記発現ベクターからヒト第VIII因子コーディング配列(CDS)に加えて5’-及び3’-UTRを含む)を切り出し、それを鋳型として、自体公知のインビトロ転写系を用いて本ヒト第VIII因子をコードするmRNAに変換することによっても、該mRNAを取得することができる。より具体的には、適当な制限酵素を用いて該mRNAコード領域を切り出し、その5’末端にファージ(T7、T3、SP6等)プロモーターをライゲーションするか、前記発現ベクターを鋳型とし、該ファージプロモーター配列を含むプライマーを用いてPCRを行うことにより、該ファージプロモーターが連結された該mRNAコード領域の断片を得る。得られたDNA断片を鋳型として、ファージ(T7、T3、SP6等)RNAポリメラーゼを反応させ、ヒト第VIII因子をコードするmRNAをインビトロ合成することができる。この時、RNAモノマー(NTPs)として、UTPの代わりにシュードウリジン(Ψ)やN1-メチルシュードウリジン(N1mΨ)の三リン酸を反応液に添加することにより、得られるmRNAはUの代わりにΨ又はN1mΨを含むことになる。それにより該mRNAに対する自然免疫の攻撃を回避することができ、mRNAの安定性及び翻訳効率が向上する。同様に、他のNTPsについても自然免疫を回避することが報告されている塩基置換がされた修飾NTPs(例、ATPに代えてイノシン三リン酸、CTPに代えて5-メチルシチジン三リン酸等)を用いることもできる。 Alternatively, the mRNA can be obtained by excising the human factor VIII coding sequence (including the 5'- and 3'-UTRs in addition to the CDS) from the expression vector and using it as a template to convert it into mRNA encoding this human factor VIII using a known in vitro transcription system. More specifically, the mRNA coding region is excised using an appropriate restriction enzyme and a phage (T7, T3, SP6, etc.) promoter is ligated to the 5' end, or a fragment of the mRNA coding region linked to the phage promoter is obtained by using the expression vector as a template and performing PCR with primers containing the phage promoter sequence. The resulting DNA fragment can be used as a template to react with phage (T7, T3, SP6, etc.) RNA polymerase to synthesize mRNA encoding human factor VIII in vitro. In this case, by adding pseudouridine (Ψ) or N1-methylpseudouridine (N1mΨ) triphosphate instead of UTP as an RNA monomer (NTP) to the reaction solution, the resulting mRNA will contain Ψ or N1mΨ instead of U. This allows the mRNA to avoid attack by natural immunity, improving mRNA stability and translation efficiency. Similarly, modified NTPs with base substitutions that have been reported to avoid natural immunity (e.g., inosine triphosphate instead of ATP, 5-methylcytidine triphosphate instead of CTP, etc.) can also be used for other NTPs.
また、mRNAの安定化と翻訳の効率化に必要な5’-キャップ構造とポリAテールを付加することが望ましい。5’-キャップ構造は、mRNAの合成後にキャッピング酵素を用いてCap 0構造を付加し、さらにmRNA 2’-O-メチルトランスフェラーゼによりCap 1構造に変換することができる。あるいは、RNAキャップアナログ(例、3’-O-Me-m7G(5’)ppp(5’)G、m7G(5’)ppp(5’)G、3’-O-Me-m7G(5’)ppp(5’)A、m7G(5’)ppp(5’)A等)を転写反応液に添加することにより、転写反応と5’-キャッピングを同時に行うことができる。ポリAテーリングも、mRNA合成後にポリAポリメラーゼを用いて3’末端に付加することもできるし、予め転写反応の鋳型にポリA配列を付加しておくことで、転写反応と同時に行うこともできる。以上のようにして得られたmRNAは、DNase Iにより鋳型DNAを除去し、例えばLiCl法などによって精製することができる。 It is also desirable to add a 5'-cap structure and a polyA tail, which are necessary for stabilizing mRNA and improving translation efficiency. The 5'-cap structure can be achieved by adding a Cap 0 structure after mRNA synthesis using a capping enzyme, and then converting it to a Cap 1 structure using an mRNA 2'-O-methyltransferase. Alternatively, transcription and 5'-capping can be performed simultaneously by adding an RNA cap analog (e.g., 3'-O-Me- m7G (5')ppp( 5 ')G, m7G (5')ppp(5')G, 3'- O-Me-m7G(5')ppp(5')A, m7G(5')ppp(5')A, etc.) to the transcription reaction solution. PolyA tailing can also be added to the 3' end of mRNA after synthesis using polyA polymerase, or it can be performed simultaneously with the transcription reaction by adding a polyA sequence to the transcription template in advance. The mRNA obtained as described above can be purified by removing the template DNA with DNase I and then by, for example, the LiCl method.
本発明の核酸の標的細胞内への導入を促進するために、核酸導入用試薬を用いてもよい。該核酸導入用試薬としては、アテロコラーゲン;リポソーム;ナノパーティクル;リポフェクチン、リポフェクタミン(lipofectamine)、DOGS(トランスフェクタム)、DOPE、DOTAP、DDAB、DHDEAB、HDEAB、ポリブレン、あるいはポリ(エチレンイミン)(PEI)等の陽イオン性脂質等を用いることができる。 A nucleic acid transfer reagent may be used to promote the transfer of the nucleic acid of the present invention into target cells. Examples of such nucleic acid transfer reagents include atelocollagen; liposomes; nanoparticles; lipofectin, lipofectamine, DOGS (transfectam), DOPE, DOTAP, DDAB, DHDEAB, HDEAB, polybrene, and cationic lipids such as poly(ethyleneimine) (PEI).
一実施態様において、本発明の核酸はリポソームに封入されてなる医薬組成物であってもよい。リポソームは、1以上の脂質二重層により包囲された内相を有する微細閉鎖小胞であり、通常は水溶性物質を内相に、脂溶性物質を脂質二重層内に保持することができる。ここで「封入」という場合に、本発明の核酸はリポソーム内相に保持されてもよいし、脂質二重層内に保持されてもよい。本発明に用いられるリポソームは単層膜であっても多層膜であってもよく、また、粒子径は、例えば10~1000nm、好ましくは50~300nmの範囲で適宜選択できる。標的組織への送達性を考慮すると、粒子径は、例えば200nm以下、好ましくは100nm以下であり得る。 In one embodiment, the nucleic acid of the present invention may be a pharmaceutical composition encapsulated in a liposome. Liposomes are small, closed vesicles with an internal phase surrounded by one or more lipid bilayers, and can typically hold a water-soluble substance in the internal phase and a lipid-soluble substance within the lipid bilayer. The term "encapsulated" used herein refers to the nucleic acid of the present invention being held in the internal phase of the liposome or within the lipid bilayer. The liposomes used in the present invention may be monolayer or multilayer membranes, and the particle size can be appropriately selected, for example, from the range of 10 to 1,000 nm, preferably 50 to 300 nm. Considering delivery to target tissues, the particle size may be, for example, 200 nm or less, preferably 100 nm or less.
ポリヌクレオチドのような水溶性化合物のリポソームへの封入法としては、リピドフィルム法(ボルテックス法)、逆相蒸発法、界面活性剤除去法、凍結融解法、リモートローディング法等が挙げられるが、これらに限定されず、任意の公知の方法を適宜選択することができる。 Methods for encapsulating water-soluble compounds such as polynucleotides into liposomes include, but are not limited to, the lipid film method (vortex method), reverse phase evaporation, surfactant removal, freeze-thaw method, and remote loading method, and any known method can be selected as appropriate.
好ましい実施態様においては、本発明の核酸は脂質ナノ粒子を担体とし、該粒子中に内封されている。
本明細書中、「脂質ナノ粒子」(Lipid Nano Particle、本明細書中「LNP」と略称することがある。)とは、両親媒性脂質の親水性基が界面の水相側に向かって配列した膜構造を有し、粒子径が1μm未満である粒子を意味し、「両親媒性脂質」とは、親水性基及び疎水性基の両方を有する脂質を意味する。
In a preferred embodiment, the nucleic acid of the present invention is carried on a lipid nanoparticle and is encapsulated within the particle.
As used herein, "lipid nanoparticles" (sometimes abbreviated as "LNPs" in the present specification) refer to particles having a membrane structure in which the hydrophilic groups of amphipathic lipids are aligned toward the aqueous phase side of the interface, and having a particle diameter of less than 1 μm, and "amphipathic lipid" refers to a lipid having both a hydrophilic group and a hydrophobic group.
本発明に用いられる脂質ナノ粒子の粒子径は、好ましくは10nm~500nmであり、より好ましくは30nm~300nmである。粒子径の測定は、例えばZetasizer Nano(Malvern社)などの粒度分布測定装置を用いて行うことができる。脂質ナノ粒子の粒子径は、脂質ナノ粒子の製造方法により、適宜調整することができる。本明細書中、「粒子径」とは、動的光散乱法により測定した平均粒子径(ゼータ平均)を意味する。 The particle diameter of the lipid nanoparticles used in the present invention is preferably 10 nm to 500 nm, and more preferably 30 nm to 300 nm. Particle diameter can be measured using a particle size distribution analyzer such as a Zetasizer Nano (Malvern). The particle diameter of the lipid nanoparticles can be adjusted appropriately depending on the manufacturing method of the lipid nanoparticles. In this specification, "particle diameter" refers to the average particle diameter (zeta mean) measured by dynamic light scattering.
両親媒性脂質としては、例えば、陽イオン性脂質、イオン性脂質、リン脂質、PEG脂質等を挙げることができる。本明細書中、「陽イオン性脂質(cationic lipid)」とは、恒常的に正に荷電した親水性基を有する脂質を意味する。本明細書中、「イオン性脂質(ionizable lipid)」とは、生理学的pHでは中性であるが、低pHではプロトン付加して正に荷電する脂質を意味する。本明細書中、「PEG」とは、ポリエチレングリコールを意味し、「PEG脂質」とは、PEGで修飾した脂質、即ち、PEGが結合した脂質を意味する。 Amphipathic lipids include, for example, cationic lipids, ionic lipids, phospholipids, and PEG lipids. As used herein, "cationic lipid" refers to a lipid having a constitutively positively charged hydrophilic group. As used herein, "ionizable lipid" refers to a lipid that is neutral at physiological pH but becomes protonated and positively charged at low pH. As used herein, "PEG" refers to polyethylene glycol, and "PEG lipid" refers to a lipid modified with PEG, i.e., a lipid to which PEG is bound.
好ましい実施態様において、本発明の核酸が内封される脂質ナノ粒子として、
(A)陽イオン性脂質又はイオン性脂質、
(B)リン脂質、
(C)ステロイド類、及び
(D)PEG脂質
を含む脂質ナノ粒子を挙げることができる。(A)~(D)の各成分脂質としては、脂質ナノ粒子に通常使用されている既知の脂質が挙げられ、当業者であれば、容易に適切な脂質の種類及びそれらの組成を選択することができる。
In a preferred embodiment, the lipid nanoparticles in which the nucleic acid of the present invention is encapsulated are:
(A) a cationic lipid or an ionic lipid,
(B) phospholipids,
(C) steroids, and (D) lipid nanoparticles containing PEG lipids. Each of the component lipids (A) to (D) includes known lipids that are commonly used in lipid nanoparticles, and a person skilled in the art can easily select an appropriate type of lipid and its composition.
本発明はまた、本発明の核酸を含む発現ベクターが導入された宿主細胞、該宿主細胞を培養し、得られる培養物からヒト第VIII因子又はその改変体を回収することを含む、ヒト第VIII因子又はその改変体の製造方法、当該方法により得られるヒト第VIII因子改変体を含む医薬(組換えタンパク質製剤)、特に血友病Aの治療剤を提供する。 The present invention also provides host cells into which an expression vector containing the nucleic acid of the present invention has been introduced; a method for producing human factor VIII or a modified factor thereof, which comprises culturing the host cells and recovering human factor VIII or a modified factor thereof from the resulting culture; and a pharmaceutical (recombinant protein preparation), particularly a therapeutic agent for hemophilia A, containing the modified human factor VIII obtained by the method.
組換えヒト第VIII因子生産用の発現ベクターは、本発明の核酸(即ち、本発明の改変体をコードする核酸、又は高発現型のヒト第VIII因子コード核酸)を、適当な発現ベクター中のプロモーターの下流に連結することにより製造することができる。発現ベクターとしては、大腸菌由来のプラスミド(例、pBR322,pBR325,pUC12,pUC13);枯草菌由来のプラスミド(例、pUB110,pTP5,pC194);酵母由来プラスミド(例、pSH19,pSH15);昆虫細胞発現プラスミド(例、pFast-Bac);動物細胞発現プラスミド(例、pA1-11,pXT1,pRc/CMV,pRc/RSV,pcDNAI/Neo);λファージなどのバクテリオファージ;バキュロウイルスなどの昆虫ウイルスベクター(例、BmNPV,AcNPV);レトロウイルス、レンチウイルス、ワクシニアウイルス、アデノウイルス、アデノ随伴ウイルス、ヘルペスウイルスなどの動物ウイルスベクターなどが用いられる。 An expression vector for producing recombinant human factor VIII can be produced by linking a nucleic acid of the present invention (i.e., a nucleic acid encoding a variant of the present invention, or a nucleic acid encoding a highly expressed form of human factor VIII) downstream of a promoter in an appropriate expression vector. Expression vectors that can be used include plasmids derived from Escherichia coli (e.g., pBR322, pBR325, pUC12, pUC13); plasmids derived from Bacillus subtilis (e.g., pUB110, pTP5, pC194); yeast-derived plasmids (e.g., pSH19, pSH15); insect cell expression plasmids (e.g., pFast-Bac); animal cell expression plasmids (e.g., pA1-11, pXT1, pRc/CMV, pRc/RSV, pcDNAI/Neo); bacteriophages such as λ phage; insect virus vectors such as baculovirus (e.g., BmNPV, AcNPV); and animal virus vectors such as retrovirus, lentivirus, vaccinia virus, adenovirus, adeno-associated virus, and herpes virus.
プロモーターとしては、遺伝子の発現に用いる宿主に対応して適切なプロモーターであればいかなるものでもよい。例えば、宿主が動物細胞である場合、SRαプロモーター、SV40プロモーター、LTRプロモーター、CMV(サイトメガロウイルス)プロモーター、RSV(ラウス肉腫ウイルス)プロモーター、MoMuLV(モロニーマウス白血病ウイルス)LTR、HSV-TK(単純ヘルペスウイルスチミジンキナーゼ)プロモーターなどが用いられる。他の宿主においても自体公知のプロモーターを適宜選択することができる。 Any promoter may be used as long as it is appropriate for the host used to express the gene. For example, when the host is an animal cell, the SRα promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (Rous sarcoma virus) promoter, MoMuLV (Moloney murine leukemia virus) LTR, HSV-TK (herpes simplex virus thymidine kinase) promoter, etc. may be used. For other hosts, a publicly known promoter may also be selected as appropriate.
発現ベクターとしては、上記の他に、所望によりエンハンサー、スプライシングシグナル、ポリA付加シグナル、選択マーカー、SV40複製起点(以下、SV40 oriと略称する場合がある)などを含有しているものを用いることができる。選択マーカーとしては、例えば、ジヒドロ葉酸還元酵素遺伝子、アンピシリン耐性遺伝子、ネオマイシン耐性遺伝子等が挙げられる。 In addition to the above, expression vectors can also contain, as desired, enhancers, splicing signals, polyA addition signals, selection markers, SV40 origin of replication (hereinafter sometimes abbreviated as SV40 ori), etc. Examples of selection markers include the dihydrofolate reductase gene, ampicillin resistance gene, and neomycin resistance gene.
本発明の核酸を含む発現ベクターで宿主を形質転換し、得られる形質転換体を培養することによって、ヒト第VIII因子又はその改変体を製造することができる。宿主としては、例えば、エシェリヒア属菌、バチルス属菌、酵母、昆虫細胞、昆虫、動物細胞などが用いられる。哺乳動物細胞としては、例えば、サルCOS-7細胞、サルVero細胞、チャイニーズハムスター卵巣細胞(以下、CHO細胞と略記)、dhfr遺伝子欠損CHO細胞(以下、CHO(dhfr-)細胞と略記)、マウスL細胞,マウスAtT-20細胞、マウスミエローマ細胞,ラットGH3細胞、ヒトFL細胞、HeLa細胞、HepG2細胞、HEK293細胞などが用いられる。他の宿主についても自体公知の細胞をそれぞれ適宜選択することができる。 Human factor VIII or a modified form thereof can be produced by transforming a host with an expression vector containing the nucleic acid of the present invention and culturing the resulting transformant. Examples of hosts that can be used include Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, and animal cells. Examples of mammalian cells that can be used include monkey COS-7 cells, monkey Vero cells, Chinese hamster ovary cells (hereinafter abbreviated as CHO cells), dhfr gene-deficient CHO cells (hereinafter abbreviated as CHO(dhfr − ) cells), mouse L cells, mouse AtT-20 cells, mouse myeloma cells, rat GH3 cells, human FL cells, HeLa cells, HepG2 cells, and HEK293 cells. For other hosts, cells known per se can be appropriately selected.
形質転換は、宿主の種類に応じ、公知の方法に従って実施することができる。動物細胞は、例えば、細胞工学別冊8 新細胞工学実験プロトコール,263-267 (1995)(秀潤社発行)、ヴィロロジー(Virology),52巻,456 (1973)に記載の方法に従って形質転換することができる。 Transformation can be carried out according to known methods depending on the type of host. Animal cells can be transformed, for example, according to the methods described in Cell Engineering Special Issue 8, New Cell Engineering Experimental Protocols, pp. 263-267 (1995) (published by Shujunsha) and Virology, Vol. 52, p. 456 (1973).
形質転換体の培養は、宿主の種類に応じ、公知の方法に従って実施することができる。例えば、宿主が動物細胞である形質転換体を培養する場合の培地としては、例えば、約5~約20%の胎児ウシ血清を含む最小必須培地(MEM)、ダルベッコ改変イーグル培地(DMEM)、RPMI 1640培地、199培地、Ham’s F-12培地などが用いられる。培地のpHは、好ましくは約6~約8である。培養は、通常約30℃~約40℃で、約15~約60時間行なわれる。必要に応じて通気や撹拌を行ってもよい。
以上のようにして、形質転換体の細胞内又は細胞外にヒト第VIII因子を製造せしめることができる。
The transformant can be cultured according to a known method depending on the type of host. For example, when the host is an animal cell, a medium such as minimum essential medium (MEM) containing about 5 to about 20% fetal bovine serum, Dulbecco's modified Eagle's medium (DMEM), RPMI 1640 medium, 199 medium, or Ham's F-12 medium can be used. The pH of the medium is preferably about 6 to about 8. The culture is usually carried out at about 30°C to about 40°C for about 15 to about 60 hours. Aeration or stirring may be performed as necessary.
In this manner, human factor VIII can be produced intracellularly or extracellularly in the transformant.
前記形質転換体を培養して得られる培養物からヒト第VIII因子を自体公知の方法に従って分離精製することができる。このような方法としては、塩析や溶媒沈澱法などの溶解度を利用する方法;透析法、限外ろ過法、ゲルろ過法、およびSDS-ポリアクリルアミドゲル電気泳動法などの主として分子量の差を利用する方法;イオン交換クロマトグラフィーなどの荷電の差を利用する方法;アフィニティークロマトグラフィーなどの特異的親和性を利用する方法;逆相高速液体クロマトグラフィーなどの疎水性の差を利用する方法;等電点電気泳動法などの等電点の差を利用する方法;などが用いられる。これらの方法は、適宜組み合わせることもできる。 Human factor VIII can be separated and purified from the culture obtained by culturing the transformant using methods known per se. Examples of such methods include methods that utilize solubility, such as salting out and solvent precipitation; methods that primarily utilize differences in molecular weight, such as dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis; methods that utilize differences in charge, such as ion exchange chromatography; methods that utilize specific affinity, such as affinity chromatography; methods that utilize differences in hydrophobicity, such as reversed-phase high-performance liquid chromatography; and methods that utilize differences in isoelectric point, such as isoelectric focusing. These methods can also be combined as appropriate.
かくして得られるヒト第VIII因子が遊離体である場合には、自体公知の方法あるいはそれに準じる方法によって、該遊離体を塩に変換することができ、ヒト第VIII因子が塩として得られた場合には、自体公知の方法あるいはそれに準じる方法により、該塩を遊離体または他の塩に変換することができる。 If the human factor VIII thus obtained is in the free form, the free form can be converted into a salt by a method known per se or a method similar thereto. If the human factor VIII is obtained as a salt, the salt can be converted into the free form or another salt by a method known per se or a method similar thereto.
ヒト第VIII因子は低毒性であり、そのまま液剤として、または適当な剤型の医薬組成物として、ヒト又は他の哺乳動物に対して非経口的(例、血管内投与(静脈内投与、動脈内投与等)、皮下投与、皮内投与、腹腔内投与、筋肉注射、局所投与など)に投与することができる。 Human factor VIII is low in toxicity and can be administered to humans or other mammals parenterally (e.g., intravascular administration (intravenous administration, intraarterial administration, etc.), subcutaneous administration, intradermal administration, intraperitoneal administration, intramuscular injection, topical administration, etc.) either as a liquid preparation or as a pharmaceutical composition in an appropriate dosage form.
非経口投与のための組成物としては、例えば、注射剤、坐剤等が用いられ、注射剤は静脈注射剤、皮下注射剤、皮内注射剤、筋肉注射剤、点滴注射剤等の剤形を包含しても良い。このような注射剤は、公知の方法に従って調製できる。注射剤の調製方法としては、例えば、1以上の有効成分を、通常注射剤に用いられる無菌の水性液、または油性液に溶解、懸濁または乳化することによって調製できる。注射用の水性液としては、例えば、生理食塩水、ブドウ糖やその他の補助薬を含む等張液等が用いられ、適当な溶解補助剤、例えば、アルコール(例、エタノール)、ポリアルコール(例、プロピレングリコール、ポリエチレングリコール)、非イオン界面活性剤〔例、ポリソルベート80、HCO-50(polyoxyethylene(50mol)adduct of hydrogenated castor oil)〕等と併用してもよい。油性液としては、例えば、ゴマ油、大豆油等が用いられ、溶解補助剤として安息香酸ベンジル、ベンジルアルコール等を併用してもよい。調製された注射液は、適当なアンプルに充填されることが好ましい。直腸投与に用いられる坐剤は、有効成分を通常の坐薬用基剤に混合することによって調製されてもよい。 Compositions for parenteral administration include, for example, injections and suppositories, and injections may include dosage forms such as intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, and drip infusion injections. Such injections can be prepared according to known methods. For example, injections can be prepared by dissolving, suspending, or emulsifying one or more active ingredients in a sterile aqueous or oily liquid typically used for injections. Examples of aqueous solutions for injection include saline, isotonic solutions containing glucose and other adjuvants, and may be used in combination with appropriate solubilizers, such as alcohols (e.g., ethanol), polyalcohols (e.g., propylene glycol, polyethylene glycol), and nonionic surfactants (e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)). As the oily liquid, for example, sesame oil, soybean oil, etc. are used, and solubilizing agents such as benzyl benzoate and benzyl alcohol may be used in combination. The prepared injection solution is preferably filled into appropriate ampoules. Suppositories for rectal administration may be prepared by mixing the active ingredient with a standard suppository base.
上記の非経口用医薬組成物は、有効成分の投与量に適合するような投薬単位の剤形に調製されることが好都合である。このような投薬単位の剤形としては、例えば、錠剤、丸剤、カプセル剤、注射剤(アンプル)、坐剤が挙げられ、投薬単位剤形当たり通常100~5000単位、好ましくは250~3000単位含有されている。 The above-mentioned parenteral pharmaceutical compositions are conveniently prepared in dosage unit forms that correspond to the dosage of the active ingredient. Examples of such dosage unit forms include tablets, pills, capsules, injections (ampoules), and suppositories, and each dosage unit typically contains 100 to 5,000 units, preferably 250 to 3,000 units.
ヒト第VIII因子の投与量は、投与対象、症状、投与ルートなどによっても異なるが、例えば、血友病Aの治療のために使用する場合には、1回量として、通常10~100単位/kg体重程度、好ましくは20~50単位/kg体重程度を、好ましくは週2~3回程度、静脈内又は腹腔内投与により投与するのが好都合である。他の非経口投与の場合もこれに準ずる量を投与することができる。症状が特に重い場合には、その症状に応じて増量してもよい。 The dosage of human factor VIII varies depending on the recipient, symptoms, and route of administration. For example, when used to treat hemophilia A, a single dose of approximately 10-100 units/kg body weight, preferably approximately 20-50 units/kg body weight, is typically administered intravenously or intraperitoneally, preferably two to three times a week. Similar amounts can also be administered for other parenteral administrations. When symptoms are particularly severe, the dosage may be increased accordingly.
以下に実施例を示して、本発明をより詳細に説明するが、これらは単なる例示であって、本発明の範囲を何ら限定するものではない。 The present invention will be explained in more detail below using examples, but these are merely illustrative and do not in any way limit the scope of the present invention.
以下の実施例において、すべての動物実験のプロトコールは、自治医科大学における動物のケア及び関連の制度委員会により承認され、動物のケアは当委員会のガイドラインに準じた。マウスはジャパンSLCから購入した。第VIII因子欠損マウスは、ジャクソンラボラトリー(米国)より入手した。 In the following examples, all animal experimental protocols were approved by the Jichi Medical University Animal Care and Related Institutional Committee, and animal care followed the committee's guidelines. Mice were purchased from Japan SLC. Factor VIII-deficient mice were obtained from Jackson Laboratory (USA).
実施例1 Bドメイン欠失ヒト第VIII因子(hFVIIISQ)及びその改変体をコードする核酸の作製と各改変体の凝固因子活性
hFVIIISQ
Bドメインをほぼ除いたヒト凝固第VIII因子(hFVIIISQ)のDNA配列は、NCBIの参照配列(NM_000132.4)をもとに設計し、GeneArt(Thermo Scientific)でコドン最適化および遺伝子合成した(配列番号4)。
Ver.1―Ver.12
hFVIIISQとイヌFVIIISQ、ブタFVIIISQ、ヒツジFVIIISQ、ウシFVIIISQの配列をもとにした遺伝子合成およびMutagenesis PCRにより72ヶ所のアミノ酸部位をイヌ型に置換するように改変(但し、ヒト用にコドン最適化)した改変体Ver.1を作製した。さらに、置換数をそれぞれ58、50ヶ所に削減した改変体Ver.2及びVer.3を作製した。Ver.2とVer.3に共通のアミノ酸置換のみを有する置換数36のVer.4も作製した。また、Ver.2から既報(WO 2014/209942)のX5、X10変異との重複を除いた置換数56のVer.5、Ver.4からX5変異との重複を除いた置換数35のVer.6、Ver.2から既報(Mol Ther Methods Clin Dev. 2020 Jan 15; 17:328-336)のJF12変異との重複を除いた置換数51のVer.7、Ver.4からJF12変異との重複を除いた置換数32のVer.8、Ver.2からX5、X10及びJF12変異との重複を除いた置換数49のVer.9、Ver.4からX5及びJF12変異との重複を除いた置換数31のVer.10、Ver.10から置換数を3ヶ所削減した置換数28のVer.11、Ver.11から置換数を15ヶ所削減した置換数13のVer.12を作製した(Ver.1~Ver.12のアミノ酸置換を図1にまとめた)。比較のために、既報のX5変異を有する改変体(但し、ヒト用にコドン最適化)も作製した。
Example 1: Preparation of nucleic acids encoding B domain-deleted human factor VIII (hFVIIISQ) and its variants, and coagulation factor activity of each variant
hFVIISQ
The DNA sequence of human coagulation factor VIII (hFVIIISQ) with most of the B domain removed was designed based on the NCBI reference sequence (NM_000132.4), and codon-optimized and gene-synthesized using GeneArt (Thermo Scientific) (SEQ ID NO: 4).
Ver. 1-Ver. 12
A variant, Ver. 1, was prepared by gene synthesis and mutagenesis PCR based on the sequences of hFVIIISQ, canine FVIIISQ, porcine FVIIISQ, ovine FVIIISQ, and bovine FVIIISQ, altering 72 amino acid sites to those of canine FVIIISQ (codon-optimized for human use). Furthermore, variants Ver. 2 and Ver. 3 were prepared by reducing the number of substitutions to 58 and 50, respectively. Ver. 4, which contains only the amino acid substitutions common to Ver. 2 and Ver. 3, was also prepared. Ver. 5, which contains 56 substitutions, was prepared by removing the overlap with the X5 and X10 mutations previously reported (WO 2014/209942). Ver. 6 and Ver. 7, which contain 35 substitutions, were prepared by removing the overlap with the X5 mutation from Ver. 4. Ver. 7, which has 51 substitutions, was created by removing overlaps with the JF12 mutation previously reported (Mol Ther Methods Clin Dev. 2020 Jan 15; 17:328-336) from Ver. 2; Ver. 8, which has 32 substitutions, was created by removing overlaps with the JF12 mutation from Ver. 4; Ver. 9, which has 49 substitutions, was created by removing overlaps with the X5, X10, and JF12 mutations from Ver. 2; Ver. 10, which has 31 substitutions, was created by removing overlaps with the X5 and JF12 mutations from Ver. 4; Ver. 11, which has 28 substitutions, was created by reducing the number of substitutions by three from Ver. 10; and Ver. 12, which has 13 substitutions, was created by reducing the number of substitutions by 15 from Ver. 11 (the amino acid substitutions of Ver. 1 to Ver. 12 are summarized in Figure 1). For comparison, a variant containing the previously reported X5 mutation (but codon-optimized for humans) was also prepared.
各hFVIIISQはクローニング後に、ApoE遺伝子の肝臓特異的エンハンサーであるHCRとヒトα1アンチトリプシンプロモーターを結合させたHCRhAATプロモーターを搭載したプラスミド(p1.1HCRhAAT)に導入した。 After cloning, each hFVIIISQ was introduced into a plasmid (p1.1HCRhAAT) carrying the HCRhAAT promoter, which combines HCR, a liver-specific enhancer of the ApoE gene, with the human α1-antitrypsin promoter.
各FVIII(Ver.1~Ver.10)遺伝子導入によるFVIII発現の評価のため、Huh7細胞(ヒト肝がん細胞株)へのプラスミドトランフェクションにより、培養上清中に一過性に分泌するFVIIIを測定した。Huh7細胞の培地は、10% FBS、GlutaMAX(Thermo Scientific)及びペニシリン-ストレプトマイシンを添加したRPMI-1640(Sigma)を用いた。1.5×105個/wellのHuh7細胞を24wellプレートに播種し、翌日にLipoefectamin 3000(Thermo Scientific)によるトランスフェクションを行った。トランスフェクション翌日に培地交換を行い、その翌日に培養24時間後の培地を回収した。回収した培地を遠心分離し、その上清を検体として第VIII因子の活性と抗原測定に用いた。 To evaluate FVIII expression following transfection of each FVIII gene (Ver. 1 to Ver. 10), Huh7 cells (a human hepatoma cell line) were transfected with the plasmid, and FVIII transiently secreted into the culture supernatant was measured. Huh7 cell culture medium was RPMI-1640 (Sigma) supplemented with 10% FBS, GlutaMAX (Thermo Scientific), and penicillin-streptomycin. Huh7 cells were seeded at 1.5 x 10 cells/well in a 24-well plate, and transfection was performed the following day using Lipoefectamin 3000 (Thermo Scientific). The medium was replaced the day after transfection, and the medium was collected the following day after 24 hours of culture. The collected medium was centrifuged, and the supernatant was used as a sample for measuring the activity and antigen of factor VIII.
第VIII因子の活性測定は、全自動血液凝固測定装置CS-1600(Sysmex)を用いた合成基質法により測定した。 Factor VIII activity was measured using the chromogenic assay with the fully automated blood coagulation analyzer CS-1600 (Sysmex).
各FVIII遺伝子(Ver.1、2、4、10)を搭載したAAV8ベクターを第VIII因子欠損マウスにマウスの体重1gあたり1×108 vgのベクター量で投与した。マウス血漿中の第VIII因子活性を合成基質法により測定した。 AAV8 vectors carrying each FVIII gene (Ver. 1, 2, 4, 10) were administered to factor VIII-deficient mice at a vector dose of 1 x 108 vg per gram of mouse body weight. Factor VIII activity in mouse plasma was measured by the synthetic substrate method.
結果を図2に示す。すべての改変体で野生型hFVIIISQよりも凝固因子活性は増大した。特に、Ver.4は、インビトロ及びインビボ実験の両方で高活性を示した。 The results are shown in Figure 2. All variants had increased coagulation factor activity compared to wild-type hFVIIISQ. In particular, Ver. 4 showed high activity in both in vitro and in vivo experiments.
実施例2 Ver.4の活性上昇に重要な変異部位の同定
Ver.4から置換数を削減していき、凝固一段法をもとに置換数12の改変体S12を同定した。さらに、置換数を削減して、合成基質法をもとにPNPL(L152P、K213N、S367P、F2196L)を同定した。S12からX5及びJF12変異と重複する5ヶ所を除いた置換数7のS7、さらに置換数をそれぞれ5及び3に削減したS5及びS3、S7、S5及びS3にL152Pの置換を加えたS8、S6及びS4を作製し、実施例1と同様にプラスミドトランスフェクションして、培養上清中の凝固因子活性を、凝固一段法及び合成基質法で測定した。これらの結果を図3に示す。PNPL変異を含むS12、S8及びS6が高活性を示した。尚、Ver.10から置換数を28にまで削減したVer.11でも、Ver.4と同等の凝固因子活性が認められた。
Example 2: Identification of Mutation Sites Important for Increased Activity of Ver. 4 The number of substitutions was reduced from Ver. 4, and a variant S12 with 12 substitutions was identified using the one-stage clotting assay. Further reductions in the number of substitutions led to the identification of PNPL (L152P, K213N, S367P, F2196L) using the synthetic substrate assay. S7, with 7 substitutions, was created by removing five substitutions from S12 that overlap with the X5 and JF12 mutations. S5 and S3, with 5 and 3 substitutions, respectively, were further reduced, and S8, S6, and S4, with the addition of the L152P substitution to S7, S5, and S3, respectively, were created. These were transfected with plasmids as in Example 1, and the coagulation factor activity in the culture supernatant was measured using the one-stage clotting assay and the synthetic substrate assay. These results are shown in Figure 3 . S12, S8, and S6, which contain the PNPL mutation, exhibited high activity. Furthermore, Ver. 10, with the number of substitutions reduced to 28, was also developed. Ver. 11 also showed coagulation factor activity equivalent to that of Ver. 4.
次に、S8に含まれる各アミノ酸置換単独の効果について、同様に凝固一段法及び合成基質法で評価した。結果を図4に示す。L152P、K213N、S367P、S727P又はF2196Lが野生型と比較して凝固因子活性を上昇させた。 Next, the effect of each amino acid substitution in S8 was evaluated individually using the one-stage clotting assay and the synthetic substrate assay. The results are shown in Figure 4. L152P, K213N, S367P, S727P, and F2196L increased clotting factor activity compared to the wild type.
実施例3 PNPL改変体の他のアミノ酸による置換
PNPLのいずれかの置換部位を他の19のアミノ酸に置換した改変体シリーズ(XNPL、PXPL、PNXL、PNPX)を作製し、実施例1と同様にプラスミドトランスフェクションして、培養上清中の凝固因子活性を、凝固一段法及び合成基質法で測定した。これらの結果を図5-1~5-4に示す。L152をQ、V、M等、K213をN、H等、S367をN、Q等、F2196をM等に置換しても、PNPLと同様の高活性を示すことが明らかとなった。
Example 3: Substitution of PNPL variants with other amino acids A series of variants (XNPL, PXPL, PNXL, PNPX) were prepared by substituting any of the substitution sites in PNPL with 19 other amino acids. These variants were transfected with plasmids in the same manner as in Example 1, and the coagulation factor activity in the culture supernatant was measured by the one-stage coagulation assay and the synthetic substrate assay. The results are shown in Figures 5-1 to 5-4. It was revealed that substitutions of L152 with Q, V, M, etc., K213 with N, H, etc., S367 with N, Q, etc., and F2196 with M, etc., also showed high activity similar to that of PNPL.
実施例4 FVIII KOマウスへの投与実験
実施例1及び2において特に高活性を示したVer.4、10及び11、S12、S8及びPNPL遺伝子をAAV8ベクターに搭載させ、FVIIIノックアウト血友病Aモデルマウスに投与し、4週間後に採血して、凝固因子活性を凝固一段法及び合成基質法で測定した。また、ELISAを用いて抗原量を測定した。結果を図6に示す。PNPL変異を有するS12及びPNPLは、Ver.4の活性値をほぼ担保するが、抗原量は顕著に少なかった。従って、これらの改変体は比活性が増大していることが示唆された。
Example 4: Administration Experiment to FVIII KO Mice The Ver. 4, 10, and 11, S12, S8, and PNPL genes, which showed particularly high activity in Examples 1 and 2, were loaded onto AAV8 vectors and administered to FVIII knockout hemophilia A model mice. Four weeks later, blood samples were collected and coagulation factor activity was measured using a one-stage clotting assay and a synthetic chromogenic assay. Antigen levels were also measured using ELISA. The results are shown in Figure 6. S12 and PNPL, which contain the PNPL mutation, almost matched the activity of Ver. 4, but the antigen levels were significantly lower. This suggests that these variants have increased specific activity.
実施例5 FVIII発現細胞内外でのFVIIIの局在
Ver.4、S12及びPNPLをHuh7細胞にトランスフェクトし、培養上清及び細胞抽出液中のFVIIIの存在量をウェスタンブロットで解析した。結果を図7-1に示す。野生型hFVIIISQでは、軽鎖の細胞内貯留が認められたのに対し、Ver.4では貯留がなくなっていた。S12及びPNPLでは軽鎖の細胞内貯留は改善されなかった。本発明者らは、Ver.4における軽鎖の細胞内貯留の改善がI1668F及びD1681Gの2ヶ所の変異によるのではないかと予測し、これら2ヶ所の置換のみを含むFVIIISQ改変体を作製し、同様に細胞抽出液中のFVIIIの存在量を調べた。その結果、図7-2に示すように、FG変異は軽鎖の細胞内貯留を改善することが実証された。
Example 5: Localization of FVIII in and outside FVIII-expressing cells. Huh7 cells were transfected with Ver. 4, S12, and PNPL, and the amount of FVIII present in the culture supernatant and cell extract was analyzed by Western blot. The results are shown in Figure 7-1. Intracellular retention of the light chain was observed with wild-type hFVIIISQ, whereas retention was abolished with Ver. 4. S12 and PNPL did not improve intracellular retention of the light chain. The inventors predicted that the improvement in intracellular retention of the light chain in Ver. 4 was due to the two mutations, I1668F and D1681G, and prepared FVIIISQ variants containing only these two substitutions, and similarly examined the amount of FVIII present in cell extracts. As shown in Figure 7-2, the FG mutation demonstrated improved intracellular retention of the light chain.
実施例6 Ver.4の高分泌・高活性に重要な変異部位の同定(2)
Ver.11をベースにして、合成基質法をもとに置換数を13にまで削減したVer.12(図1)を同定した。結果を図8に示す。
Ver.12をもとに置換数をさらに削減し、高分泌する置換数8~15の改変体(F8-Tochigi-3(NPL)、5、8、10、11、13、15)を作製した(これらの改変体のアミノ酸置換を図9に示す)。実施例1と同様にプラスミドトランスフェクションして、培養上清中の凝固因子活性を、凝固一段法及び合成基質法で測定した。また、培養上清中の抗原量をELISAを用いて測定した。結果を図10に示す。図中、F8-Tochigi-28はVer.11と同じ改変体を示す。
Example 6 Identification of mutation sites important for high secretion and high activity of Ver. 4 (2)
Based on Ver. 11, Ver. 12 (FIG. 1) was identified using the synthetic substrate method, in which the number of substitutions was reduced to 13. The results are shown in FIG.
The number of substitutions was further reduced based on Ver. 12 to create highly secreted variants with 8 to 15 substitutions (F8-Tochigi-3 (NPL), 5, 8, 10, 11, 13, and 15) (the amino acid substitutions of these variants are shown in Figure 9). Plasmid transfection was performed in the same manner as in Example 1, and the coagulation factor activity in the culture supernatant was measured by the one-stage clotting assay and the synthetic substrate assay. The antigen amount in the culture supernatant was also measured using ELISA. The results are shown in Figure 10. In the figure, F8-Tochigi-28 represents the same variant as Ver. 11.
実施例7 高発現型のhFVIII遺伝子の構築
コドン最適化された野生型hFVIII DNA(配列番号4)及びVer.4改変体DNA(配列番号2)のヌクレオチド配列は、配列内に多数のCpG配列を含む。本発明者らは、これらのCpG配列が宿主内で免疫応答を誘発してFVIIIの発現が損われる可能性があると予測し、それらがコードするFVIIIのアミノ酸配列を変化させることなく、該CpG配列をすべて除去した上で、GC含量などの他のパラメータも考慮してコドン最適化し直したヌクレオチド配列を、GeneArt Codon Optimizer(Thermo Scientific)を用いて設計し、遺伝子合成した。CpG除去及び再最適化された野生型hFVIII DNA及びVer.4改変体DNAのヌクレオチド配列を、それぞれ配列番号5及び3に示す。これらのDNAを、やはりCpG配列を除去したマウストランスサイレチン(mTTR)プロモーター配列(配列番号7)の下流に連結してAAV6ベクターに搭載し、実施例1と同様にプラスミドトランスフェクションし、Huh7細胞に導入した。また、該発現カセットをAAV8ベクターに搭載させて、マウスに投与した。培養上清及び血中の凝固因子活性を、凝固一段法及び合成基質法で測定した。結果を図11-1及び11-2に示す。Ver.4、野生型のいずれについても、CpG配列を除去することにより、発現効率が著明に増大した。
Example 7 Construction of a High-Expression hFVIII Gene The nucleotide sequences of the codon-optimized wild-type hFVIII DNA (SEQ ID NO: 4) and the Ver. 4 variant DNA (SEQ ID NO: 2) contain numerous CpG sequences. The inventors predicted that these CpG sequences may induce an immune response in the host, impairing FVIII expression. Therefore, they removed all of the CpG sequences without changing the amino acid sequence of the FVIII they encode. Then, they re-optimized the codons, taking into account other parameters such as GC content, and designed the nucleotide sequences using GeneArt Codon Optimizer (Thermo Scientific). The nucleotide sequences of the CpG-removed and re-optimized wild-type hFVIII DNA and the Ver. 4 variant DNA are shown in SEQ ID NOs: 5 and 3, respectively. These DNAs were ligated downstream of a mouse transthyretin (mTTR) promoter sequence (SEQ ID NO: 7) from which the CpG sequence had also been removed, and then loaded into an AAV6 vector. The vector was then plasmid transfected and introduced into Huh7 cells in the same manner as in Example 1. The expression cassette was also loaded into an AAV8 vector and administered to mice. The coagulation factor activity in the culture supernatant and blood was measured by the one-stage clotting assay and the synthetic substrate assay. The results are shown in Figures 11-1 and 11-2. For both Ver. 4 and the wild-type, removal of the CpG sequence significantly increased the expression efficiency.
実施例8 カニクイザルモデルへの投与
実施例7で作製した、CpG配列を除去したmTTRプロモーター配列の下流にCpG除去及び再最適化されたVer.4改変体コード配列が連結されたDNAを、AAV8ベクターに挿入し、University College of Londonによる臨床試験(NCT0300183)で使用されたベクター投与量(2×1012vg/kg)で、免疫抑制剤(プレドニゾロン1mg/kg/日及びタクロリムス0.05mg/kg/日)を投与したカニクイザル血友病モデルに投与し、投与後1又は2週間おきに第VIII因子活性及び抗原量を測定した。結果を図12-1に示す。個体ごとに推移に差はあるものの、いずれの個体でもベクター投与後、第VIII因子活性は著明に増大した。
また、上記DNAをAAV5ベクターに挿入し、Roctavian(登録商標)(BioMarin社から上市されている血友病A治療薬)の1/10(6×1012vg/kg)及び1/30(2×1012vg/kg)のベクター投与量で、同様に処理したカニクイザルモデルに投与し、投与後1又は2週間おきに第VIII因子活性及び抗原量を測定した。結果を図12-2に示す。いずれの投与量でもベクター投与後、第VIII因子活性は著明に増大した。
Example 8: Administration to a Cynomolgus Monkey Model The DNA prepared in Example 7, in which the CpG-deleted and re-optimized Ver. 4 variant coding sequence was ligated downstream of the CpG-deleted mTTR promoter sequence, was inserted into an AAV8 vector and administered to a cynomolgus monkey hemophilia model treated with immunosuppressants (prednisolone 1 mg/kg/day and tacrolimus 0.05 mg/kg/day) at the vector dose (2 x 10 12 vg/kg) used in a clinical trial conducted by the University of London (NCT0300183). Factor VIII activity and antigen levels were measured every one or two weeks after administration. The results are shown in Figure 12-1. Although the time course differed between individuals, factor VIII activity significantly increased in all individuals after vector administration.
Furthermore, the above DNA was inserted into an AAV5 vector and administered to a similarly treated cynomolgus monkey model at vector doses 1/10 (6 x 10 12 vg/kg) and 1/30 (2 x 10 12 vg/kg) of those used with Rectavian (registered trademark) (a hemophilia A treatment drug marketed by BioMarin). Factor VIII activity and antigen levels were measured every one or two weeks after administration. The results are shown in Figure 12-2. Factor VIII activity significantly increased after vector administration at both doses.
実施例9 発現カセットの配列最適化
実施例7で作製した、CpG配列を除去したmTTRプロモーター配列の下流にCpG除去及び再最適化されたVer.4改変体コード配列が連結されたDNAを挿入したAAV6ベクター(コード配列の下流にSV40 polyA付加シグナルをさらに含み、発現カセットとして配列番号8で示されるヌクレオチド配列を含む)(original)、プロモーターとコード配列との間の配列が短縮化されたヌクレオチド配列(配列番号9)を発現カセットとして含むAAV6ベクター(v1mTTRp)、さらにpolyA付加シグナルの3’末端側が異なる長さで短縮化された3種のヌクレオチド配列(配列番号10、11及び12)を発現カセットとして含むAAV6ベクター(それぞれv1-v1、v1-v2及びv1-v3)を、Huh7細胞に遺伝子導入した。24時間後に培地交換し、さらに24時間後に培養上清と細胞とをそれぞれ回収して、上清中の第VIII因子活性及び抗原量と、細胞あたりのAAVゲノム量を測定した。結果を図13に示す。プロモーターとコード配列との間の配列を短縮化することで活性は増大し、polyA付加シグナルの短縮化を組み合わせることでさらに活性は向上した。v1-v2において最も活性が高かった。しかし、v1-v3までpolyA付加シグナルの3’末端を欠失させると、第VIII因子活性は大きく低下した。
Example 9 Optimization of Expression Cassette Sequence Huh7 cells were transfected with the AAV6 vector (original) prepared in Example 7, in which DNA was inserted into the CpG-deleted and re-optimized Ver. 4 variant coding sequence ligated downstream of the CpG-deleted mTTR promoter sequence (which further contained an SV40 polyA addition signal downstream of the coding sequence and contained the nucleotide sequence shown in SEQ ID NO: 8 as an expression cassette), the AAV6 vector (v1mTTRp) containing, as an expression cassette, a nucleotide sequence (SEQ ID NO: 9) in which the sequence between the promoter and the coding sequence was shortened, and the AAV6 vector (v1-v1, v1-v2, and v1-v3, respectively) containing, as expression cassettes, three nucleotide sequences (SEQ ID NOs: 10, 11, and 12) in which the 3'-end of the polyA addition signal was shortened by different lengths. The medium was replaced after 24 hours, and the culture supernatant and cells were collected after another 24 hours. The Factor VIII activity and antigen content in the supernatant, as well as the AAV genome content per cell, were measured. The results are shown in Figure 13. Activity increased when the sequence between the promoter and coding sequence was shortened, and activity was further improved by shortening the polyA addition signal in combination. v1-v2 showed the highest activity. However, when the 3' end of the polyA addition signal was deleted up to v1-v3, Factor VIII activity was significantly reduced.
本発明の改変体、それをコードする核酸、該核酸を含む発現ベクター及び該発現ベクターを導入された肝臓細胞は、血友病Aに対するインビボもしくはエクスビボ遺伝子治療、mRNA医薬等に有用である。また、前記発現ベクターを導入された宿主細胞を培養して得られる組換えヒト第VIII因子改変体は、血友病Aの治療用生物製剤として有用である。さらに、高発現型のヒト第VIII因子コード核酸は、血友病Aの遺伝子治療、mRNA医薬に有用であり、かつ組換えヒト第VIII因子の高産生宿主の製造用としても有用である。 The variants of the present invention, the nucleic acids encoding them, expression vectors containing the nucleic acids, and liver cells transfected with the expression vectors are useful for in vivo or ex vivo gene therapy, mRNA medicines, etc. for hemophilia A. Furthermore, recombinant human factor VIII variants obtained by culturing host cells transfected with the expression vectors are useful as biological preparations for the treatment of hemophilia A. Furthermore, highly expressed human factor VIII-encoding nucleic acids are useful for gene therapy and mRNA medicines for hemophilia A, and are also useful for producing hosts that highly produce recombinant human factor VIII.
Claims (26)
S367におけるアミノ酸置換がS367P、S367NもしくはS367Qであり、及び/又は
F2196におけるアミノ酸置換がF2196LもしくはF2196Mである、請求項1に記載の改変体。 The variant according to claim 1, wherein the amino acid substitution at K213 is K213N or K213H, and/or the amino acid substitution at S367 is S367P, S367N or S367Q, and/or the amino acid substitution at F2196 is F2196L or F2196M.
I1668F及びD1681G
のアミノ酸置換をさらに含む、請求項3に記載の改変体。 L152A, L152D, L152E, L152H, L152I, L152M, L152N, L152P, L152Q or L152V, and/or I1668F and D1681G
The variant of claim 3 further comprising an amino acid substitution of:
M539L、並びに、任意選択でI566M、L603P及びI642Vからなる群より選択される少なくとも1個のA2ドメイン内のアミノ酸置換をさらに含み、
K2207Q、Q2316H及びM2321L、並びに、任意選択でF2275L、S2296A及びV2314Aからなる群より選択される少なくとも1個のC2ドメイン内のアミノ酸置換をさらに含む、請求項1に記載の改変体。 the amino acid substitution at K213 is K213N;
M539L, and optionally at least one amino acid substitution in the A2 domain selected from the group consisting of I566M, L603P, and I642V;
2. The variant of claim 1, further comprising at least one amino acid substitution in the C2 domain selected from the group consisting of K2207Q, Q2316H and M2321L, and optionally F2275L, S2296A and V2314A.
(a)K213N、S367P及びF2196L
(b)K213N、S367P、F2196L、I1668F及びD1681G
(c)K213N、M539L、I566M、I642V、K2207Q、F2275L、Q2316H及びM2321L
(d)K213N、M539L、I566M、L603P、I642V、F2196L、K2207Q、F2275L、Q2316H及びM2321L
(e)K213N、M539L、I566M、I642V、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(f)K213N、M539L、I566M、I642V、I1668F、D1681G、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(g)L152P、K213N、S367P及びF2196L
(h)L152P、K213N、S367P、Y487H、L603P及びF2196L
(i)L152P、K213N、S367P、Y487H、L603P、S727P、Q1659E及びF2196L
(j)L152P、K213N、S367P、Y487H、L603P、S727P、Q1659E、H1859R、A1993V、H2007Q、K2085M及びF2196L
(k)K213N、S367P、M539L、I566M、L603P、I642V、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(l)P25H、A28T、K213N、M217T、S367P、Q410L、Y487H、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、N2019K、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(m)R-5P、P25H、A28T、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、N2019K、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L
(n)R-5P、P25H、A28T、L152P、K213N、M217T、W228Q、S367P、Q410L、Y487H、R489G、F501M、M539L、I566M、L603P、I642V、S727P、A736V、S1657P、Q1659E、E1661K、I1668F、D1681G、R1776K、H1859R、A1993V、H2007Q、N2019K、K2085M、F2196L、K2207Q、F2275L、S2296A、V2314A、Q2316H及びM2321L A human factor VIII variant having the following amino acid substitutions (a) to (n) in wild-type human factor VIII:
(a) K213N, S367P, and F2196L
(b) K213N, S367P, F2196L, I1668F, and D1681G
(c) K213N, M539L, I566M, I642V, K2207Q, F2275L, Q2316H, and M2321L
(d) K213N, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, Q2316H, and M2321L
(e) K213N, M539L, I566M, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L
(f) K213N, M539L, I566M, I642V, I1668F, D1681G, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H, and M2321L
(g) L152P, K213N, S367P and F2196L
(h) L152P, K213N, S367P, Y487H, L603P and F2196L
(i) L152P, K213N, S367P, Y487H, L603P, S727P, Q1659E, and F2196L
(j) L152P, K213N, S367P, Y487H, L603P, S727P, Q1659E, H1859R, A1993V, H2007Q, K2085M and F2196L
(k) K213N, S367P, M539L, I566M, L603P, I642V, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(l) P25H, A28T, K213N, M217T, S367P, Q410L, Y487H, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P , Q1659E, E1661K, I1668F, D1681G, R1776K, N2019K, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(m) R-5P, P25H, A28T, K213N, M217T, W228Q, S367P, Q410L, Y487H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736 V, S1657P, Q1659E, E1661K, I1668F, D1681G, R1776K, N2019K, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
(n) R-5P, P25H, A28T, L152P, K213N, M217T, W228Q, S367P, Q410L, Y487 H, R489G, F501M, M539L, I566M, L603P, I642V, S727P, A736V, S1657P, Q1 659E, E1661K, I1668F, D1681G, R1776K, H1859R, A1993V, H2007Q, N201 9K, K2085M, F2196L, K2207Q, F2275L, S2296A, V2314A, Q2316H and M2321L
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2024015293 | 2024-02-02 | ||
| JP2024-015293 | 2024-02-02 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025164792A1 true WO2025164792A1 (en) | 2025-08-07 |
Family
ID=96590345
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2025/003309 Pending WO2025164792A1 (en) | 2024-02-02 | 2025-01-31 | High specific activity/high secretion expression-type factor viii variant, nucleic acid encoding high expression-type factor viii, and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025164792A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2005511038A (en) * | 2001-11-30 | 2005-04-28 | エモリー ユニバーシテイ | Variant of factor VIII C2 domain |
| JP2010518830A (en) * | 2007-02-23 | 2010-06-03 | ビオメトード | Novel factor VIII for the treatment of hemophilia A |
| JP2013541521A (en) * | 2010-09-15 | 2013-11-14 | ノヴォ ノルディスク アー/エス | Factor VIII mutant with reduced cellular uptake |
| JP2019503649A (en) * | 2015-10-30 | 2019-02-14 | スパーク セラピューティクス インコーポレイテッドSpark Therapeutics, Inc. | CpG reduced factor VIII variants, compositions and methods and uses for the treatment of hemostatic disorders |
-
2025
- 2025-01-31 WO PCT/JP2025/003309 patent/WO2025164792A1/en active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2005511038A (en) * | 2001-11-30 | 2005-04-28 | エモリー ユニバーシテイ | Variant of factor VIII C2 domain |
| JP2010518830A (en) * | 2007-02-23 | 2010-06-03 | ビオメトード | Novel factor VIII for the treatment of hemophilia A |
| JP2013541521A (en) * | 2010-09-15 | 2013-11-14 | ノヴォ ノルディスク アー/エス | Factor VIII mutant with reduced cellular uptake |
| JP2019503649A (en) * | 2015-10-30 | 2019-02-14 | スパーク セラピューティクス インコーポレイテッドSpark Therapeutics, Inc. | CpG reduced factor VIII variants, compositions and methods and uses for the treatment of hemostatic disorders |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20250108093A1 (en) | Factor VIII Sequences | |
| TWI791433B (en) | Gene therapy for treating hemophilia a | |
| JP2025148444A (en) | Methods and compositions for treating premature stop codon-mediated disorders | |
| JP6730193B2 (en) | Treatment of hyperbilirubinemia | |
| KR20210112339A (en) | Gene therapy constructs to treat Wilson's disease | |
| CN110325199A (en) | For treating the gene therapy of phenylketonuria | |
| WO2019210267A2 (en) | Engineered aav capsids with increased tropism and aav vectors comprising the engineered capsids and methods of making and using same | |
| KR20150005521A (en) | Aav vector compositions and methods for gene transfer to cells, organs and tissues | |
| KR20170084012A (en) | An improved expression cassette for packaging and expression of variant factor viii for the treatment of hemostasis disorders | |
| KR102814622B1 (en) | Methods and compositions for increasing protein expression and/or treating hepatotoxicity disorders | |
| KR20240025507A (en) | Methods and compositions for treating premature stop codon-mediated disorders | |
| WO2017123961A1 (en) | Factor viii variants, nucleic acid sequences, and methods and uses for treatment of hemostasis disorders | |
| WO2025164792A1 (en) | High specific activity/high secretion expression-type factor viii variant, nucleic acid encoding high expression-type factor viii, and use thereof | |
| US20240076691A1 (en) | Codon-optimized nucleic acid encoding the fix protein | |
| CN114591921B (en) | Method for purifying adeno-associated virus and eluent | |
| RU2807158C2 (en) | Genotherapy constructions for treatment of wilson's disease | |
| WO2024254232A2 (en) | Two-plasmid system for aav vector particle production | |
| WO2025006829A1 (en) | Improved adeno-associated virus vectors | |
| CN116669774A (en) | Compositions and methods for treating Fabry disease | |
| TW202521698A (en) | Expression cassette for target genes and applications thereof can achieve disease treatment effects with low side effects and lifelong benefits from a single administration | |
| Mcintosh et al. | Factor VIII sequences |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25749157 Country of ref document: EP Kind code of ref document: A1 |