US20220010294A1 - Novel recombinant botulinum neurotoxins with increased duration of effect - Google Patents

Novel recombinant botulinum neurotoxins with increased duration of effect Download PDF

Info

Publication number
US20220010294A1
US20220010294A1 US17/482,953 US202117482953A US2022010294A1 US 20220010294 A1 US20220010294 A1 US 20220010294A1 US 202117482953 A US202117482953 A US 202117482953A US 2022010294 A1 US2022010294 A1 US 2022010294A1
Authority
US
United States
Prior art keywords
neurotoxin
amino acid
exosite
recombinant
botulinum
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/482,953
Inventor
Jürgen Frevert
Fred Hofmann
Marcel Jurk
Manuela López De La Paz
Daniel Scheps
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merz Pharma GmbH and Co KGaA
Original Assignee
Merz Pharma GmbH and Co KGaA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merz Pharma GmbH and Co KGaA filed Critical Merz Pharma GmbH and Co KGaA
Priority to US17/482,953 priority Critical patent/US20220010294A1/en
Assigned to MERZ PHARMA GMBH & CO. KGAA reassignment MERZ PHARMA GMBH & CO. KGAA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LÓPEZ DE LA PAZ, Manuela, SCHEPS, Daniel, Frevert, Jürgen , HOFMANN, FRED, JURK, Marcel
Publication of US20220010294A1 publication Critical patent/US20220010294A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/52Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from bacteria or Archaea
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/33Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Clostridium (G)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/24Metalloendopeptidases (3.4.24)
    • C12Y304/24069Bontoxilysin (3.4.24.69), i.e. botulinum neurotoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to novel recombinant botulinum neurotoxins serotype A exhibiting both (i) an increased duration of effect and (ii) a high specific biological activity.
  • the invention also relates to methods for the manufacture of such recombinant botulinum neurotoxins.
  • These novel recombinant botulinum neurotoxins comprise at least two additional domains and least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • the invention further relates to pharmaceutical compositions comprising said recombinant neurotoxins.
  • the invention further relates to novel recombinant single-chain precursor botulinum neurotoxins and nucleic acid sequences encoding such recombinant single-chain precursor botulinum neurotoxins.
  • Clostridium is a genus of anaerobe gram-positive bacteria, belonging to the Firmicutes. Clostridium consists of around 100 species that include common free-living bacteria as well as important pathogens, such as Clostridium botulinum and Clostridium tetani . Both species produce neurotoxins, botulinum toxin and tetanus toxin, respectively. These neurotoxins are potent inhibitors of calcium-dependent neurotransmitter secretion of neuronal cells and are among the strongest toxins known to man. The lethal dose in humans lies between 0.1 ng and 1 ng per kilogram of body weight.
  • botulism which is characterised by paralysis of various muscles. Paralysis of the breathing muscles can cause death of the affected individual.
  • botulinum neurotoxin BoNT
  • tetanus neurotoxin TxNT
  • the botulinum toxin acts at the neuromuscular junction and other cholinergic synapses in the peripheral nervous system, inhibiting the release of the neurotransmitter acetylcholine and thereby causing flaccid paralysis
  • the tetanus toxin acts mainly in the central nervous system, preventing the release of the inhibitory neurotransmitters GABA (gamma-aminobutyric acid) and glycine by degrading the protein synaptobrevin.
  • GABA gamma-aminobutyric acid
  • glycine gamma-aminobutyric acid
  • the consequent overactivity in the muscles results in generalized contractions of the agonist and antagonist musculature, termed a tetanic spasm (rigid paralysis).
  • BoNT/A the immunogenic type of tetanus neurotoxin
  • BoNT/G the botulinum neurotoxins are known to occur in seven different immunogenic types, termed BoNT/A through BoNT/G. Most Clostridium botulinum strains produce one type of neurotoxin, but strains producing multiple toxins have also been described.
  • Botulinum and tetanus neurotoxins have highly homologous amino acid sequences and show a similar domain structure.
  • Their biologically active form comprises two peptide chains, a light chain of about 50 kDa and a heavy chain of about 100 kDa, linked by a disulfide bond.
  • a linker or loop region whose length varies among different clostridial toxins, is located between the two cysteine residues forming the disulfide bond. This loop region is proteolytically cleaved by an unknown clostridial endoprotease to obtain the biologically active toxin.
  • the light chain can then selectively cleave so called SNARE-proteins, which are essential for different steps of neurotransmitter release into the synaptic cleft, e.g. recognition, docking and fusion of neurotransmitter-containing vesicles with the plasma membrane.
  • TeNT, BoNT/B, BoNT/D, BoNT/F, and BoNT/G cause proteolytic cleavage of synaptobrevin or VAMP (vesicle-associated membrane protein), BoNT/A and BoNT/E cleave the plasma membrane-associated protein SNAP-25, and BoNT/C cleaves the integral plasma membrane protein syntaxin and SNAP-25.
  • Clostridial neurotoxins display variable durations of action that are serotype specific.
  • the clinical therapeutic effect of BoNT/A lasts approximately 3 months for neuromuscular disorders and 6 to 12 months for hyperhidrosis.
  • the effect of BoNT/E on the other hand, lasts less than 4 weeks.
  • the longer lasting therapeutic effect of BoNT/A makes it preferable for certain clinical use compared to the other serotypes, for example serotypes B, C 1 , D, E, F, G.
  • One possible explanation for the divergent durations of action might be the distinct subcellular localizations of BoNT serotypes.
  • the protease domain of BoNT/A light chain localizes in a punctate manner to the plasma membrane of neuronal cells, co-localizing with its substrate SNAP-25.
  • the short-duration BoNT/E serotype is cytoplasmic. Membrane association might protect BoNT/A from cytosolic degradation mechanisms allowing for prolonged persistence of BoNT/A in the neuronal cell.
  • botulinum toxin is formed as a protein complex comprising the neurotoxic component and non-toxic proteins.
  • the accessory proteins embed the neurotoxic component thereby protecting it from degradation by digestive enzymes in the gastrointestinal tract.
  • botulinum neurotoxins of most serotypes are orally toxic.
  • Complexes with, for example, 450 kDa or with 900 kDa are obtainable from cultures of Clostridium botulinum.
  • botulinum neurotoxins have been used as therapeutic agents in the treatment of dystonias and spasms.
  • Preparations comprising botulinum toxin complexes are commercially available, e.g. from Ipsen Ltd (Dysport®) or Allergan Inc. (Botox®).
  • a high purity neurotoxic component, free of any complexing proteins, is for example available from Merz Pharmaceuticals GmbH, Frankfurt (Xeomin®).
  • Clostridial neurotoxins are usually injected into the affected muscle tissue, bringing the agent close to the neuromuscular end plate, i.e. close to the cellular receptor mediating its uptake into the nerve cell controlling said affected muscle.
  • Various degrees of neurotoxin spread have been observed. The neurotoxin spread is thought to depend on the injected amount and the particular neurotoxin preparation. It can result in adverse side effects such as paralysis in nearby muscle tissue, which can largely be avoided by reducing the injected doses to the therapeutically relevant level. Overdosing can also trigger the immune system to generate neutralizing antibodies that inactivate the neurotoxin preventing it from relieving the involuntary muscle activity. Immunologic tolerance to botulinum toxin has been shown to correlate with cumulative doses.
  • clostridial neurotoxins are still predominantly produced by fermentation processes using appropriate Clostridium strains.
  • industrial production of clostridial neurotoxin from anaerobic Clostridium culture is a cumbersome and time-consuming process. Due to the high toxicity of the final product, the procedure must be performed under strict containment.
  • the single-chain precursors are proteolytically cleaved by an unknown clostridial protease to obtain the biologically active di-chain clostridial neurotoxin.
  • the degree of neurotoxin activation by proteolytic cleavage varies between different strains and neurotoxin serotypes, which is a major consideration for the manufacture due to the requirement of neurotoxin preparations with a well-defined biological activity.
  • the clostridial neurotoxins are produced as protein complexes, in which the neurotoxic component is embedded by accessory proteins. These accessory proteins have no beneficial effect on biological activity or duration of effect. They can however trigger an immune reaction in the patient, resulting in immunity against the clostridial neurotoxin. Manufacture of recombinant clostridial neurotoxins, which are not embedded by auxiliary proteins, might therefore be advantageous.
  • clostridial neurotoxins have been expressed in eukaryotic expression systems, such as in Pichia pastoris, Pichia methanolica, Saccharomyces cerevisiae , insect cells and mammalian cells (see WO 2006/017749).
  • botulinum neurotoxins may be expressed as single-chain precursors, which subsequently have to be proteolytically cleaved to obtain the final biologically active botulinum neurotoxin.
  • botulinum neurotoxins may be expressed in high yield in rapidly-growing bacteria as relatively non-toxic single-chain polypeptides.
  • botulinum neurotoxin characteristics regarding biological activity, cell specificity, antigenic potential and duration of effect by genetic engineering to obtain recombinant neurotoxins with new therapeutic properties in specific clinical areas.
  • Genetic modification of botulinum neurotoxins might allow altering the mode of action or expanding the range of therapeutic targets.
  • Botulinum toxin variants exhibiting an increased duration of effect and a high specific biological activity in neuromuscular tissue than naturally occurring botulinum toxins would be very advantageous in order to reduce administration frequency and the incidence of neutralizing antibody generation since immunologic tolerance to botulinum toxin is correlated with cumulative doses.
  • US 2002/0127247 describes clostridial neurotoxins comprising modifications in secondary modification sites and exhibiting altered biological persistence.
  • botulinum neurotoxins serotype A with an increased duration of effect and with improved properties, in order to allow for exploitation of the therapeutic potential of BoNT serotype A, which have so far been considered impractical for certain clinical application.
  • the increased duration of effect of a particular botulinum neurotoxin serotype A could be adjusted in a tailor-made fashion in order to address any particular features and demands of a given indication, such as the amount of neurotoxin being administered, frequency of administration etc.
  • botulinum neurotoxins serotype A which exhibit an increased duration of effect than naturally occurring botulinum toxins serotype A with a high specific biological activity. To date, such aspects have not been solved satisfactorily.
  • Such a method and novel precursor botulinum neurotoxins used in such methods would serve to satisfy the great need for recombinant botulinum neurotoxins exhibiting an increased duration of effect with a high specific biological activity.
  • BoNT/A exhibits the longest persistence and was shown to localize in the vicinity of the plasma membrane of neuronal cells.
  • additional factors such as degradation, spread or diffusion, and/or translocation rates might have a decisive impact on the differences in the duration for the individual botulinum toxin serotypes.
  • these neurotoxins comprise at least two additional domains consisting of proline, alanine and an additional amino acid residue.
  • these neurotoxins according to the invention comprise at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • the present invention relates to a recombinant botulinum neurotoxin serotype A comprising at least two domains wherein each domain comprises an amino acid sequence consisting of at least 50 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, wherein the neurotoxin further comprises at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • the present invention relates to the use of the composition of the present invention for cosmetic treatment.
  • the present invention relates to a recombinant single-chain precursor botulinum neurotoxin serotype A comprising at least two additional domains wherein each domain comprises an amino acid sequence consisting of at least 50 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, and a modified alpha-exosite and/or beta-exosite of the light chain.
  • the present invention relates to a nucleic acid sequence encoding the recombinant single-chain precursor botulinum neurotoxin serotype A of the present invention.
  • the present invention relates to a method for obtaining the nucleic acid sequence of the present invention, comprising the step of adding nucleic acid sequences to the heavy and the light chain and in addition modifying a nucleic acid sequence encoding the alpha-exosite and/or at the beta-exosite of the light chain of the parental botulinum neurotoxin serotype A.
  • the present invention relates to a vector comprising the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention.
  • the present invention relates to a recombinant host cell comprising the nucleic acid sequence of the present invention, the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention.
  • the present invention relates to a method for producing the recombinant single-chain precursor botulinum neurotoxin of the present invention, comprising the step of expressing the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention in a recombinant host cell, or cultivating the recombinant host cell of the present invention under conditions that result in the expression of said nucleic acid sequence.
  • a sequence listing containing SEQ ID NOs: 1-5, created Nov. 8, 2019, 48 KB is provided herewith in a computer-readable nucleotide/amino acid .txt file and is specifically incorporated by reference.
  • FIG. 2 SDS ⁇ PAGE of purified BoNT/A MaJ007 (PAS100-BoNT/A-ISA2-PAS100).
  • Lane 1 Molecular weight marker. Prior to applying the samples to the gel, R-mercaptoethanol was added.
  • Lane “v.A.” (before activation) purified, non-activated single-chain PAS100-BoNT/A-ISA2-PAS100.
  • Lanes “n.A.” (after activation) and “n.R.” (after purification) show light chain (LC) and heavy chain (HC) obtained after activation by thrombin under reducing conditions.
  • LC light chain
  • HC heavy chain
  • both the light chain (LC) and the heavy chain (HC) each comprise an additional amino acid sequence consisting of 100 amino acid residues, wherein said amino acid sequence consists of proline, alanine and serine residues (PAS) and where
  • FIG. 4 SDS ⁇ PAGE of purified BoNT/A MaJ024 (PAS100-BoNT/A-ISA5-PAS(100).
  • Lane 1 Molecular weight marker. Prior to applying the samples to the gel, R-mercaptoethanol was added.
  • Lane “v.A.” (before activation) purified, non-activated single-chain PAS100-BoNT/A-ISA5-PAS(100.
  • Lanes “n.A.” (after activation) and “n.R.” (after purification) show light chain (LC) and heavy chain (HC) obtained after activation by thrombin under reducing conditions.
  • LC light chain
  • HC heavy chain
  • FIG. 5 Mouse running assay with BoNT/A PAS100-BoNT/A-ISA5-PAS100 (MaJ024)
  • the present invention relates to a recombinant botulinum neurotoxin serotype A comprising at least two domains wherein each domain comprises an amino acid sequence consisting of at least 50 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, wherein the neurotoxin further comprises at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • botulinum neurotoxin serotype A refers to a natural neurotoxin serotype A obtainable from bacteria Clostridium botulinum , or to a neurotoxin obtainable from alternative sources, including from recombinant technologies or from genetic or chemical modification.
  • the botulinum neurotoxins have a specific biological activity, i.e. endopeptidase activity.
  • Botulinum neurotoxins are produced as single-chain precursors that are proteolytically cleaved by an unknown clostridial endoprotease within the loop region to obtain the biologically active disulfide-linked di-chain form of the neurotoxin, which comprises two chain elements, a functionally active light chain and a functionally active heavy chain, where one end of the light chain is linked to one end of the heavy chain not via a peptide bond, but via a disulfide bond.
  • botulinum neurotoxin light chain refers to that part of a botulinum neurotoxin serotype A that comprises an endopeptidase activity responsible for cleaving one or more proteins that is/are part of the so-called SNARE-complex involved in the process resulting in the release of neurotransmitter into the synaptic cleft:
  • the light chain has a molecular weight of approx. 50 kDa.
  • alpha-exosite of the light chain refers to four LC alpha-helices (102-113, 310-321, 335-348, and 351-358) that interface a helical motif of SNAP-25 that is approximately 30-50 amino acids away from the cleavage site of SNAP-25 which interacts with the substrate SNAP25 (see Xue S, Javor S, Hixon M S, Janda K D, Probing BoNT/A protease exosites: implications for inhibitor design and light chain longevity. Biochemistry. 2014; 53(43):6820-4).
  • beta-exosite of the light chain refers to a beta-sheet region located in a beta-sheet close to the active site which interacts with the substrate SNAP25 comprising AS 162-254 (see Breidenbach MA1, Brunger A T. Substrate recognition strategy for botulinum neurotoxin serotype A, Nature. 2004, 432(7019):925-9).
  • the term “functionally active botulinum neurotoxin chain” refers to a recombinant botulinum neurotoxin serotype A chain able to perform the biological functions of a naturally occurring botulinum neurotoxin serotype A chain to at least about 50%, particularly to at least about 60%, to at least about 70%, to at least about 80%, and most particularly to at least about 90%, where the biological functions of botulinum neurotoxin chains include, but are not limited to, binding of the heavy chain to the neuronal cell, entry of the neurotoxin into a neuronal cell, release of the light chain from the di-chain neurotoxin, and endopeptidase activity of the light chain.
  • WO 95/32738 describes the reconstitution of separately obtained light and heavy chains of tetanus toxin and botulinum toxin. Also cell-based assay methods as described for example in WO2009/114748, WO 2013/049508 and WO2014/207109.
  • the term “about” or “approximately” means within 20%, alternatively within 10%, including within 5% of a given value or range. Alternatively, especially in biological systems, the term “about” means within about a log (i.e. an order of magnitude), including within a factor of two of a given value.
  • the term “recombinant botulinum neurotoxin” refers to a composition comprising a botulinum neurotoxin serotype A that is obtained by expression of the neurotoxin in a heterologous cell such as E. coli , and including, but not limited to, the raw material obtained from a fermentation process (supernatant, composition after cell lysis), a fraction comprising a botulinum neurotoxin serotype A obtained from separating the ingredients of such a raw material in a purification process, an isolated and essentially pure protein, and a formulation for pharmaceutical and/or aesthetic use comprising a botulinum neurotoxin serotype A and additionally pharmaceutically acceptable solvents and/or excipients.
  • the recombinant botulinum neurotoxin serotype A according to the invention comprises at least one amino acid modification which is located at the alpha-exosite at at least one position selected from D102, T109, K340, I348, N353, K356.
  • the recombinant botulinum neurotoxin serotype A according to the invention comprises at least one amino acid modification which is located at the beta-exosite at at least one position selected from G169, T220, P239, S254.
  • the recombinant botulinum neurotoxin serotype A comprises the following six modifications at the alpha-exosite of the light chain:
  • the recombinant botulinum neurotoxin serotype A comprises the following four modifications at the beta-exosite of the light chain:
  • the recombinant botulinum neurotoxin serotype A comprises two domains wherein each domain comprises an amino acid sequence consisting of between 70 and 260 amino acid residues, particularly 100 amino acid residues, 150 amino acid residues, or 200 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one serine residues.
  • the recombinant botulinum neurotoxin serotype A comprises two domains wherein each domain comprises an amino acid sequence consisting of between 70 and 150 amino acid residues or between 80 and 120 amino acid residues or between 90 and 110 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one serine residues.
  • the term “functional variant of a botulinum neurotoxin” refers to a botulinum neurotoxin serotype A that differs in the amino acid sequence and/or the nucleic acid sequence encoding the amino acid sequence from a botulinum neurotoxin serotype A, but is still functionally active.
  • the term “functionally active” refers to the property of a recombinant botulinum neurotoxin serotype A to exhibit a biological activity of at least about 20%, particularly to at least about 40%, at least about 70%, at least about 80%, and most particularly at least about 90% of the biological activity of a naturally occurring parental botulinum neurotoxin, i.e.
  • a parental botulinum neurotoxin serotype A without modifications at the C-terminus of the light chain where the biological functions include, but are not limited to, binding to the neurotoxin receptor, entry of the neurotoxin into a neuronal cell, release of the light chain from the two-chain neurotoxin, and endopeptidase activity of the light chain, and thus inhibition of neurotransmitter release from the affected nerve cell.
  • In vivo assays for assessing biological activity include the mouse LD50 assay and the ex vivo mouse hemidiaphragm assay as described by Pearce et al. (Pearce 1994, Toxicol. Appl. Pharmacol. 128: 69-77) and Dressler et al.
  • MU Mouse Units
  • 1 MU is the amount of neurotoxic component, which kills 50% of a specified mouse population after intraperitoneal injection, i.e. the mouse i.p. LD50.
  • a functional variant will maintain key features of the corresponding botulinum neurotoxin serotype A, such as key residues for the endopeptidase activity in the light chain, or key residues for the attachment to the neurotoxin receptors or for translocation through the endosomal membrane in the heavy chain, but may contain modifications comprising a substitution of one or more amino acids of the corresponding botulinum neurotoxin.
  • the functional variant of a botulinum neurotoxin serotype A additionally comprises a signal peptide.
  • said signal peptide will be located at the N-terminus of the neurotoxin.
  • Many such signal peptides are known in the art and are comprised by the present invention.
  • the signal peptide results in transport of the neurotoxin across a biological membrane, such as the membrane of the endoplasmic reticulum, the Golgi membrane or the plasma membrane of a eukaryotic or prokaryotic cell. It has been found that signal peptides, when attached to the neurotoxin, will mediate secretion of the neurotoxin into the supernatant of the cells.
  • the signal peptide will be cleaved off in the course of, or subsequent to, secretion, so that the secreted protein lacks the N-terminal signal peptide, is composed of separate light and heavy chains, which are covalently linked by disulfide bridges, and is proteolytically active.
  • identity refers to sequence identity characterized by determining the number of identical amino acids between two nucleic acid sequences or two amino acid sequences wherein the sequences are aligned so that the highest order match is obtained. It can be calculated using published techniques or methods codified in computer programs such as, for example, BLASTP, BLASTN or FASTA (Altschul 1990, J Mol Biol 215, 403). The percent identity values are, in one aspect, calculated over the entire amino acid sequence. A series of programs based on a variety of algorithms is available to the skilled worker for comparing different sequences. In this context, the algorithms of Needleman and Wunsch or Smith and Waterman give particularly reliable results.
  • the program PileUp Higgins 1989, CABIOS 5, 151
  • the programs Gap and BestFit Gap and BestFit (Needleman 1970, J Mol Biol 48; 443; Smith 1981, Adv Appl Math 2, 482), which are part of the GCG software packet (Genetics Computer Group 1991, 575 Science Drive, Madison, Wis., USA 53711)
  • the sequence identity values recited above in percent (%) are to be determined, in another aspect of the invention, using the program GAP over the entire sequence region with the following settings: Gap Weight: 50, Length Weight: 3, Average Match: 10.000 and Average Mismatch: 0.000, which, unless otherwise specified, shall always be used as standard settings for sequence alignments.
  • the nucleic acid sequences encoding the functional homologue and the parental botulinum neurotoxin may differ to a larger extent due to the degeneracy of the genetic code. It is known that the usage of codons is different between prokaryotic and eukaryotic organisms. Thus, when expressing a prokaryotic protein such as a botulinum neurotoxin, in a eukaryotic expression system, it may be necessary, or at least helpful, to adapt the nucleic acid sequence to the codon usage of the expression host cell, meaning that sequence identity or homology may be rather low on the nucleic acid level.
  • the term “variant” refers to a botulinum neurotoxin serotype A that is a chemically, enzymatically, or genetically modified derivative of a corresponding neurotoxin of C. botulinum neurotoxin serotype A.
  • a chemically modified derivative may be one that is modified by pyruvation, phosphorylation, sulfatation, lipidation, pegylation, glycosylation and/or the chemical addition of an amino acid or a polypeptide comprising between 2 and about 100 amino acids, including modification occurring in the eukaryotic host cell used for expressing the derivative.
  • An enzymatically modified derivative is one that is modified by the activity of enzymes, such as endo- or exoproteolytic enzymes, including modification by enzymes of the eukaryotic host cell used for expressing the derivative.
  • a genetically modified derivative is one that has been modified by deletion or substitution of one or more amino acids contained in, or by addition of one or more amino acids (including polypeptides comprising between 2 and about 100 amino acids) to, the amino acid sequence of said botulinum neurotoxin.
  • the recombinant botulinum neurotoxin serotype A is used in the treatment of a disease requiring improved chemodenervation, wherein the recombinant neurotoxin causes both (i) an increased duration of effect relative to a wildtype botulinum neurotoxin serotype A and (ii) an increased specific biological activity relative to a botulinum neurotoxin serotype A comprising two domains consisting of proline, alanine and serine residues without said modifications in the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • the term “increased duration of effect” or “increased duration of action” refers to a longer lasting denervation mediated by a botulinum neurotoxin serotype A of the present invention.
  • administration of a disulfide-linked di-chain botulinum neurotoxin serotype A comprising two domains according to the invention results in localized paralysis for a longer period of time relative to administration of an identical disulfide-linked di-chain botulinum neurotoxin serotype A without the at least two domains according to the present invention.
  • the term “increased duration of effect/action” is defined as a more than about 20%, particularly more than about 50%, more particularly more than about 90% increased duration of effect of the recombinant neurotoxin of the present invention relative to the identical neurotoxin without the two domains according to the invention.
  • an “increased duration of effect” can be determined using the “Mouse Running Assay”.
  • the “Mouse Running Assay” is well-known to the person skilled in the art and measures the daily running distance of a mouse in a treadmill after a botulinum neurotoxin was injected into the M. gastrocnemius (see Keller J E. Recovery from botulinum neurotoxin poisoning in vivo. Neuroscience.
  • the distance which a mouse is able to run in the treadmill the day before the botulinum neurotoxin is injected is used as comparison and is set as 100%.
  • a daily running distance of no more than 80% of the initial running distance is regarded as paralysis of the muscle.
  • the duration of effect is determined by the time period between the time point attaining a half-maximal paralysis, i.e. about 40% of the initial running distance and the time point when paralysis reaches recovery, i.e. 40% of the initial running distance.
  • the botulinum neurotoxin is considered to exhibit an “increased duration of effect/action” provided that the mutated BoNT exhibits a similar potency i.e shows a similar maximal paralysis (reduction of the running distance) of about 80-90%.
  • the “specific endoprotease activity” is measured in an assay which is described in Jones et al. (J Immunol Methods. 2008 Jan. 1; 329(1-2):92-101) and Hallis et al. (J Clin Microbiol. 1996 (8):1934-8; 1996) and is described further below in example 2.
  • the term “decreased specific endoprotease activity” is defined as an at least 20% lower amount of cleavage product produced by a BoNT/A mutant in the endoprotease assay compared to the BoNT/A wildtype applying the same amount of protein.
  • the term “specific biological activity” relates to the “specific endoprotease activity” mentioned above and can be derived also from the maximal paralysis shown in the “Mouse Running Assay” described above.
  • denervation refers to denervation resulting from administration of a chemodenervating agent, for example a neurotoxin.
  • localized denervation or “localized paralysis” refers to denervation of a particular anatomical region, usually a muscle or a group of anatomically and/or physiologically related muscles, which results from administration of a chemodenervating agent, for example a neurotoxin, to the particular anatomical region.
  • a chemodenervating agent for example a neurotoxin
  • the recombinant botulinum neurotoxins of the present invention might show increased biological half-life, reduced degradation rates, decreased diffusion rates, increased uptake by neuronal cells, and/or modified intracellular translocation rates, in each case relative to an identical parental clostridial neurotoxin without the at least two domains according to the invention.
  • biological half-life specifies the lifespan of a protein, for example of a botulinum neurotoxin serotype A, in vivo.
  • biological half-life refers to the period of time, by which half of a protein pool is degraded in vivo. For example it refers to the period of time, by which half of the amount of botulinum neurotoxin of one administered dosage is degraded.
  • the present invention relates to a composition, in particular a pharmaceutical or cosmetic composition comprising the recombinant botulinum neurotoxin of the present invention.
  • a composition in particular a pharmaceutical or cosmetic composition
  • the toxin can be formulated by various techniques dependent on the desired application purposes which are known in the art.
  • the (biologically active) botulinum neurotoxin polypeptide can be used in combination with one or more pharmaceutically acceptable carriers as a pharmaceutical composition.
  • the pharmaceutically acceptable carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and being not deleterious to the recipient thereof.
  • the pharmaceutical carrier employed may include a solid, a gel, or a liquid.
  • Exemplary of solid carriers are lactose, terra alba, sucrose, talc, gelatine, agar, pectin, acacia, magnesium stearate, stearic acid and the like.
  • Exemplary of liquid carriers are glycerol, phosphate buffered saline solution, water, emulsions, various types of wetting agents, and the like. Suitable carriers comprise those mentioned above and others well known in the art, see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • the pharmaceutical composition can be dissolved in a diluent, prior to administration.
  • the diluent is also selected so as not to affect the biological activity of the Neurotoxin product.
  • the pharmaceutical composition or formulation may also include other carriers or non-toxic, non-therapeutic, non-immunogenic stabilizers and the like.
  • the formulated Neurotoxin product can be present, in an aspect, in liquid or lyophilized form. In an aspect, it can be present together with glycerol, protein stabilizers (HSA) or non-protein stabilizers such as polyvinyl pyrrolidone (PVP), hyaluronic acid or free amino acids.
  • suitable non-proteinaceous stabilizers are disclosed in WO 2005/007185 or WO 2006/020208.
  • the formulated Neurotoxin product may be used for human or animal therapy of various diseases or disorders in a therapeutically effective dose or for cosmetic purposes.
  • the recombinant botulinum neurotoxin of the present invention or the pharmaceutical composition of the present invention is for use in the treatment of a disease or condition taken from the list of: cervical dystonia (spasmodic torticollis), blepharospasm, severe primary axillary hyperhidrosis, achalasia, lower back pain, benign prostate hypertrophy, chronic focal painful neuropathies, migraine and other headache disorders.
  • Additional indications where treatment with botulinum neurotoxins is currently under investigation and where the pharmaceutical composition of the present invention may be used include pediatric incontinence, incontinence due to overactive bladder, and incontinence due to neurogenic bladder, anal fissure, spastic disorders associated with injury or disease of the central nervous system including trauma, stroke, multiple sclerosis, Parkinson's disease, or cerebral palsy, focal dystonias affecting the limbs, face, jaw or vocal cords, temporomandibular joint (TMJ) pain disorders, diabetic neuropathy, wound healing, excessive salivation, vocal cord dysfunction, reduction of the Masseter muscle for decreasing the size of the lower jaw, treatment and prevention of chronic headache and chronic musculoskeletal pain, treatment of snoring noise, assistance in weight loss by increasing the gastric emptying time.
  • pediatric incontinence incontinence due to overactive bladder
  • incontinence due to neurogenic bladder anal fissure
  • spastic disorders associated with injury or disease of the central nervous system including trauma, stroke,
  • clostridial neurotoxins have been evaluated for the treatment of other new indications, for example painful keloid, diabetic neuropathic pain, refractory knee pain, trigeminal neuralgia trigger-zone application to control pain, scarring after cleft-lip surgery, cancer and depression.
  • the present invention relates to the use of the composition of the present invention for cosmetic treatment.
  • the present invention relates to a method of cosmetically treating a patient, comprising the step of administering a composition comprising a recombinant clostridial neurotoxin according to the present invention to a patient desiring such cosmetic treatment
  • cosmetic treatment relates to uses in cosmetic or aesthetic applications, such as the treatment of wrinkles, crow's feet, glabella frown lines, reduction of the masseter muscle, reduction of the calves, removing of facial asymmetries etc.
  • the present invention relates to a method for the generation of the recombinant botulinum neurotoxin of the present invention, comprising the step of obtaining a recombinant nucleic acid sequence encoding a recombinant single-chain precursor botulinum neurotoxin by the insertion of a nucleic acid sequence encoding at least two domains consisting of proline, alanine and additional amino acid residues, selected from the group consisting of serine, threonine, tyrosine and glutamine residues into a nucleic acid sequence encoding a parental clostridial neurotoxin and by modifying the nucleic acid sequence encoding a parental botulinum neurotoxin at the alpha-exosite and/or at the beta-exosite of the light chain.
  • the term “recombinant nucleic acid sequence” refers to a nucleic acid, which has been generated by joining genetic material from two different sources.
  • single-chain precursor botulinum neurotoxin refers to a single-chain precursor for a disulfide-linked di-chain botulinum neurotoxin, comprising a functionally active botulinum neurotoxin light chain, a functionally active neurotoxin heavy chain, and a loop region linking the C-terminus of the light chain with the N-terminus of the heavy chain.
  • the term “recombinant single-chain precursor botulinum neurotoxin” refers to a single-chain precursor botulinum neurotoxin comprising at least two domains consisting of proline, alanine and additional amino acid residues, selected from the group consisting of serine, threonine, tyrosine and glutamine residues, and a modified alpha-exosite and/or beta-exosite of the light chain of the neurotoxin.
  • the recombinant single-chain precursor botulinum neurotoxin comprises a protease cleavage site in said loop region.
  • Single-chain precursor botulinum neurotoxins have to be proteolytically cleaved to obtain the final biologically active botulinum neurotoxins.
  • Proteolytic cleavage may either occur during heterologous expression by host cell enzymes, or by adding proteolytic enzymes to the raw protein material isolated after heterologous expression.
  • Naturally occurring botulinum neurotoxins usually contain one or more cleavage signals for proteases which post-translationally cleave the single-chain precursor molecule, so that the final di- or multimeric complex can form.
  • botulinum neurotoxins are still predominantly produced by fermentation processes using appropriate Clostridium strains.
  • the single-chain precursors are proteolytically cleaved by an unknown clostridial protease to obtain the biologically active di-chain clostridial neurotoxin.
  • the single-chain precursor molecule is the precursor of a protease
  • autocatalytic cleavage may occur.
  • the protease can be a separate non-clostridial enzyme expressed in the same cell.
  • WO 2006/076902 describes the proteolytic cleavage of a recombinant clostridial neurotoxin single-chain precursor at a heterologous recognition and cleavage site by incubation of the E. coli host cell lysate.
  • proteolytic cleavage is carried out by an unknown E. coli protease.
  • modified protease cleavage sites have been introduced recombinantly into the interchain region between the light and heavy chain of clostridial toxins, e.g. protease cleavage sites for human thrombin or non-human proteases (see WO 01/14570).
  • the protease cleavage site is a site that is cleaved by a protease selected from the list of: thrombin, trypsin, enterokinase, factor Xa, plant papain, insect papain, crustacean papain, enterokinase, human rhinovirus 3C protease, human enterovirus 3C protease, tobacco etch virus protease, Tobacco Vein Mottling Virus, subtilisin and caspase 3.
  • a protease selected from the list of: thrombin, trypsin, enterokinase, factor Xa, plant papain, insect papain, crustacean papain, enterokinase, human rhinovirus 3C protease, human enterovirus 3C protease, tobacco etch virus protease, Tobacco Vein Mottling Virus, subtilisin and caspase 3.
  • the recombinant single-chain precursor botulinum neurotoxin serotype A further comprises a binding tag, particularly selected from the group comprising: glutathione-S-transferase (GST), maltose binding protein (MBP), a His-tag, a StrepTag, or a FLAG-tag.
  • GST glutathione-S-transferase
  • MBP maltose binding protein
  • His-tag a StrepTag
  • FLAG-tag FLAG-tag
  • parental botulinum neurotoxin refers to an initial botulinum neurotoxin without modifications selected from a natural botulinum neurotoxin, a functional variant of a natural botulinum neurotoxin or a chimeric botulinum neurotoxin.
  • the method for the generation of the recombinant botulinum neurotoxin of the present invention further comprises the step of heterologously expressing said recombinant nucleic acid sequence in a host cell, particularly in a bacterial host cell, more particularly in an E. coli host cell.
  • the E. coli cells are selected from E. coli XL1-Blue, Nova Blue, TOP10, XL10-Gold, BL21, and K12.
  • the method for the generation of the recombinant botulinum neurotoxin of the present invention additionally comprises at least one of the steps of (i) generating a disulfide-linked di-chain recombinant botulinum neurotoxin according to the invention by causing or allowing contacting of said recombinant single-chain precursor botulinum neurotoxin with an endoprotease and (ii) purification of said recombinant single-chain precursor botulinum neurotoxin or said disulfide-linked di-chain recombinant botulinum neurotoxin by chromatography.
  • the recombinant single-chain precursor botulinum neurotoxin of the present invention, or the recombinant disulfide-linked di-chain botulinum neurotoxin of the present invention is purified after expression, or in the case of the recombinant disulfide-linked di-chain botulinum neurotoxin, after the cleavage reaction.
  • the protein is purified by chromatography, particularly by immunoaffinity chromatography, or by chromatography on an ion exchange matrix, a hydrophobic interaction matrix, or a multimodal chromatography matrix, particularly a strong ion exchange matrix, more particularly a strong cation exchange matrix.
  • the term “causing . . . contacting of said recombinant single-chain precursor botulinum neurotoxin . . . with an endoprotease” refers to an active and/or direct step of bringing said neurotoxin and said endoprotease in contact
  • the term “allowing contacting of a recombinant single-chain precursor botulinum neurotoxin . . . with an endoprotease” refers to an indirect step of establishing conditions in such a way that said neurotoxin and said endoprotease are getting in contact to each other.
  • endoprotease refers to a protease that breaks peptide bonds of non-terminal amino acids (i.e. within the polypeptide chain). As they do not attack terminal amino acids, endoproteases cannot break down peptides into monomers.
  • cleavage of the recombinant single-chain precursor botulinum neurotoxin is near-complete.
  • the term “near-complete” is defined as more than about 95% cleavage, particularly more than about 97.5%, more particularly more than about 99% as determined by SDS-PAGE and subsequent Western Blot or reversed phase chromatography.
  • cleavage of the recombinant single-chain precursor botulinum neurotoxin of the present invention occurs at a heterologous cleavage signal located in the loop region of the recombinant precursor botulinum neurotoxin.
  • the cleavage reaction is performed with crude host cell lysates containing said single-chain precursor protein.
  • the single-chain precursor protein is purified or partially purified, particularly by a first chromatographic enrichment step, prior to the cleavage reaction.
  • the term “purified” relates to more than about 90% purity. In the context of the present invention, the term “partially purified” relates to purity of less than about 90% and an enrichment of more than about two fold.
  • the present invention relates to a recombinant single-chain botulinum neurotoxin serotype A, which is a precursor for the recombinant botulinum neurotoxin of the present invention.
  • the present invention relates to a recombinant single-chain precursor botulinum neurotoxin serotype A comprising two domains consisting of proline, alanine and additional amino acid residues, selected from the group consisting of serine, threonine, tyrosine and glutamine residues, and a modified alpha-exosite and/or beta-exosite of the light chain of the neurotoxin.
  • the present invention relates to a method for obtaining the nucleic acid sequence of the present invention, comprising the step of modifying a nucleic acid sequence encoding a parental botulinum neurotoxin serotype A.
  • the present invention relates to a vector comprising the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention.
  • the present invention relates to a recombinant host cell comprising the nucleic acid sequence of the present invention, the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention.
  • the present invention relates to a method for producing the recombinant single-chain precursor botulinum neurotoxin of the present invention, comprising the step of expressing the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention in a recombinant host cell, or cultivating the recombinant host cell of the present invention under conditions that result in the expression of said nucleic acid sequence.
  • Example 1 Generation and Purification of a PASylated Botulinum Toxin Type A (PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100)
  • PAS100-BoNT/A D102F T109R K340M I348L N353M K356R
  • the nucleic acid construct encoding a “PAS” module comprising 100 amino acid residues built from the amino acids proline (P), alanine (A) and serine (S) was synthetically produced, wherein the following motive was used (ASPAAPAPASPAAPAPSAPA) 5 .
  • ASPAAPAPASPAAPAPSAPA restriction enzymes NdeI and SwaI
  • the corresponding gene module was first inserted at the N-terminus of recombinant BoNT/A (rBoNT/A).
  • the PAS module was inserted at the C-terminus of the heavy chain by using restriction enzymes BglII and AatII. The correct cloning was verified by sequencing.
  • BoNT/A in E.
  • coli gene constructs were cloned into pET29c.
  • the variants contain fused His6- and Strep-affinity tags which can be cleaved after protein purification via thrombin.
  • a synthetic gene with the corresponding mutations of D102F T109R K340M I348L N353M K356R in the BoNT/A1 wild type sequence and restriction sites SwaI and ScaI was used.
  • rBoNT/A variants were performed in Riesenberg media with 50 ⁇ g/mL Kanamycin ⁇ Riesenberg, 1991 #1 ⁇ . Cells were grown in shake flasks (37° C., 175 r.p.m) until an OD600 of 1.5-2 was reached. For induction of protein expression 1 mM IPTG (Fermentas) was added to the E. coli culture. Protein synthesis was performed for 24 h (15° C., 175 r.p.m.).
  • the resulting crude extracts were centrifuged (20,000 r.p.m., 30 min, 4° C.), and the supernatants with the soluble proteins were recovered.
  • Protein purification was carried out by fast protein liquid chromatography (GE Healthcare) using a three step purification protocols. The first capture step was performed by IMAC using a HisTrap HP 1 mL column (GE Healthcare). The column was washed (1 ml min-1 working flow) using a two-step protocol with His elution buffer (50 mM Tris, 150 mM NaCl, 400 mM Imidazol pH 8.0). The elution of the toxin proteins occurred at 400 mM Imidazol.
  • a Strep-Tactin affinity chromatography was performed as previously described (IBA GmbH).
  • a cation exchange chromatography with a HiTrap SP HP 1 mL column (GE Healthcare, Freiburg, Germany) was used.
  • the corresponding samples were diluted with SP binding buffer (50 mM Tris, pH 8) and eluted with SP elution buffer (50 mM Tris, 1 M NaCl, pH 8).
  • SP binding buffer 50 mM Tris, pH 8
  • SP elution buffer 50 mM Tris, 1 M NaCl, pH 8
  • the SEC running buffer (20 mM Tris, 150 mM NaCl, 2.5 mM CaCl 2 ) pH 7.7) was also used to store the purified protein solutions in aliquots at ⁇ 20° C. Each protein was analyzed by applying 0.5-1 ⁇ g on 4-12% gradient SDS-PAGE (Novex Life Technologies) and stained with Coomassie G-250 based SimplyBlue safe stain (Pierce). Each protein was judged >98% pure before applying in vitro or in vivo experiments.
  • BoNT/A preparations were activated with Thrombin (Merck Millipore; 8 U/1 mg BoNT) for 24 h at 20° C. yielding >99% of di-chain toxin. Afterwards the cleavage protease was eliminated with the previously described Strep-Tactin Kit (IBA GmbH).
  • FIG. 2 summarizes the results of purification and activation.
  • the specific endoproteinase activity of wildtype BoNT/A and modified BoNT/A variants was determined by an assay which is described in Jones R G, Ochiai M, Liu Y, Ekong T, Sesardic D. Development of improved SNAP25 endopeptidase immuno-assays for botulinum type A and E toxins. J Immunol Methods. 2008 Jan. 1; 329(1-2):92-101 and Hallis B, James B A, Shone C C Development of novel assays for botulinum type A and B neurotoxins based on their endopeptidase activities. J Clin Microbiol. 1996 (8):1934-8. The “1-step TMB Ultra” solution from Perbio Science was used for staining and quantification.
  • a C-terminal fragment of 70 amino acids of SNAP25 from IBA GmBH was used in the reaction as a substrate.
  • the primary mouse antibody from R&D Systems specifically recognizes SNAP25 fragments cleaved only by BoNT/A.
  • the secondary antibody was an anti-mouse-IgG-HRP-antibody conjugate from Dianova.
  • the assay was performed on a 96 microtiter plate and the amount of cleaved material was determined after 120 minutes by measuring the absorption at 450 nm (Molecular Devices, FilterMax F5). For the assay equal amounts of proteins were used.
  • Table 1 shows that the specific endoproteinase activity of PASylated botulinum toxin A was increased by introduced mutations.
  • Example 3 Generation and Purification of a PASylated Botulinum Toxin Type A (PAS100-BoNT/A G169I, T220R, P239M, S254-PAS100)
  • the nucleic acid construct encoding a “PAS” module comprising 100 amino acid residues built from the amino acids proline (P), alanine (A) and serine (S) was synthetically produced, wherein the following motive was used (ASPAAPAPASPAAPAPSAPA) 5 .
  • ASPAAPAPASPAAPAPSAPA restriction enzymes NdeI and SwaI
  • the corresponding gene module was first inserted at the N-terminus of recombinant BoNT/A (rBoNT/A).
  • the PAS module was inserted at the C-terminus of the heavy chain by using restriction enzymes BglII and AatII. The correct cloning was verified by sequencing.
  • BoNT/A in E.
  • coli gene constructs were cloned into pET29c.
  • the variants contain fused His6- and Strep-affinity tags which can be cleaved after protein purification via thrombin.
  • a synthetic gene with the corresponding mutations of G169I, T220R, P239M, S254 in the BoNT/A1 wild type sequence and restriction sites SwaI and Scat was used.
  • Protein expression was performed as described above (see Example 1) in expression strain E. coli BL21. Purification was done using a combination of his affinity, ion exchange and size exclusion chromatography, followed by activation using thrombin. FIG. 3 summarizes the results of purification and activation.
  • SEQ ID NO 1 recombinant BoNT A including His.tag
  • SEQ ID NO 2 recombinant PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100 including His.tag
  • SEQ ID NO 3 recombinant PAS100-BoNT/A (G169I, T220R, P239M, S254)-PAS100 including His.tag)
  • SEQ ID NO 4 recombinant PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100 including His.tag (nucleic acid sequence)
  • SEQ ID NO 5 recombinant PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS100 including His.tag) (nucleic acid sequence)

Abstract

This invention relates to novel recombinant botulinum neurotoxins serotype A exhibiting both (i) an increased duration of effect and (ii) a high specific biological activity. These novel recombinant botulinum neurotoxins comprise at least two additional domains consisting of proline, alanine and an additional amino acid residue and at least one amino acid modification which is located at the alpha-exosite or at the beta-exosite of the light chain of the neurotoxin. The invention further relates to novel recombinant single-chain precursor botulinum neurotoxins and compositions comprising the recombinant botulinum neurotoxin with an increased duration of effect and a high specific biological activity.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. application Ser. No. 16/498,257, filed Sep. 26, 2019, which is a U.S. National Stage Application filed under 35 U.S.C. § 371 of International Application No. PCT/EP2017/066891, filed Jul. 6, 2017, both of which are hereby incorporated by reference in their entirety.
  • FIELD OF THE INVENTION
  • This invention relates to novel recombinant botulinum neurotoxins serotype A exhibiting both (i) an increased duration of effect and (ii) a high specific biological activity. The invention also relates to methods for the manufacture of such recombinant botulinum neurotoxins. These novel recombinant botulinum neurotoxins comprise at least two additional domains and least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin. The invention further relates to pharmaceutical compositions comprising said recombinant neurotoxins. The invention further relates to novel recombinant single-chain precursor botulinum neurotoxins and nucleic acid sequences encoding such recombinant single-chain precursor botulinum neurotoxins.
  • BACKGROUND OF THE INVENTION
  • Clostridium is a genus of anaerobe gram-positive bacteria, belonging to the Firmicutes. Clostridium consists of around 100 species that include common free-living bacteria as well as important pathogens, such as Clostridium botulinum and Clostridium tetani. Both species produce neurotoxins, botulinum toxin and tetanus toxin, respectively. These neurotoxins are potent inhibitors of calcium-dependent neurotransmitter secretion of neuronal cells and are among the strongest toxins known to man. The lethal dose in humans lies between 0.1 ng and 1 ng per kilogram of body weight.
  • Oral ingestion of botulinum toxin via contaminated food or generation of botulinum toxin in wounds can cause botulism, which is characterised by paralysis of various muscles. Paralysis of the breathing muscles can cause death of the affected individual.
  • Although both botulinum neurotoxin (BoNT) and tetanus neurotoxin (TxNT) function via a similar initial physiological mechanism of action, inhibiting neurotransmitter release from the axon of the affected neuron into the synapse, they differ in their clinical response. While the botulinum toxin acts at the neuromuscular junction and other cholinergic synapses in the peripheral nervous system, inhibiting the release of the neurotransmitter acetylcholine and thereby causing flaccid paralysis, the tetanus toxin acts mainly in the central nervous system, preventing the release of the inhibitory neurotransmitters GABA (gamma-aminobutyric acid) and glycine by degrading the protein synaptobrevin. The consequent overactivity in the muscles results in generalized contractions of the agonist and antagonist musculature, termed a tetanic spasm (rigid paralysis).
  • While the tetanus neurotoxin exists in one immunologically distinct type, the botulinum neurotoxins are known to occur in seven different immunogenic types, termed BoNT/A through BoNT/G. Most Clostridium botulinum strains produce one type of neurotoxin, but strains producing multiple toxins have also been described.
  • Botulinum and tetanus neurotoxins have highly homologous amino acid sequences and show a similar domain structure. Their biologically active form comprises two peptide chains, a light chain of about 50 kDa and a heavy chain of about 100 kDa, linked by a disulfide bond. A linker or loop region, whose length varies among different clostridial toxins, is located between the two cysteine residues forming the disulfide bond. This loop region is proteolytically cleaved by an unknown clostridial endoprotease to obtain the biologically active toxin.
  • The molecular mechanism of intoxication by TeNT and BoNT appears to be similar as well: entry into the target neuron is mediated by binding of the C-terminal part of the heavy chain to a specific cell surface receptor; the toxin is then taken up by receptor-mediated endocytosis. The low pH in the so formed endosome then triggers a conformational change in the clostridial toxin which allows it to embed itself in the endosomal membrane and to translocate through the endosomal membrane into the cytoplasm, where the disulfide bond joining the heavy and the light chain is reduced. The light chain can then selectively cleave so called SNARE-proteins, which are essential for different steps of neurotransmitter release into the synaptic cleft, e.g. recognition, docking and fusion of neurotransmitter-containing vesicles with the plasma membrane. TeNT, BoNT/B, BoNT/D, BoNT/F, and BoNT/G cause proteolytic cleavage of synaptobrevin or VAMP (vesicle-associated membrane protein), BoNT/A and BoNT/E cleave the plasma membrane-associated protein SNAP-25, and BoNT/C cleaves the integral plasma membrane protein syntaxin and SNAP-25.
  • Clostridial neurotoxins display variable durations of action that are serotype specific. The clinical therapeutic effect of BoNT/A lasts approximately 3 months for neuromuscular disorders and 6 to 12 months for hyperhidrosis. The effect of BoNT/E, on the other hand, lasts less than 4 weeks. The longer lasting therapeutic effect of BoNT/A makes it preferable for certain clinical use compared to the other serotypes, for example serotypes B, C1, D, E, F, G. One possible explanation for the divergent durations of action might be the distinct subcellular localizations of BoNT serotypes. The protease domain of BoNT/A light chain localizes in a punctate manner to the plasma membrane of neuronal cells, co-localizing with its substrate SNAP-25. In contrast, the short-duration BoNT/E serotype is cytoplasmic. Membrane association might protect BoNT/A from cytosolic degradation mechanisms allowing for prolonged persistence of BoNT/A in the neuronal cell.
  • In Clostridium botulinum, the botulinum toxin is formed as a protein complex comprising the neurotoxic component and non-toxic proteins. The accessory proteins embed the neurotoxic component thereby protecting it from degradation by digestive enzymes in the gastrointestinal tract. Thus, botulinum neurotoxins of most serotypes are orally toxic. Complexes with, for example, 450 kDa or with 900 kDa are obtainable from cultures of Clostridium botulinum.
  • In recent years, botulinum neurotoxins have been used as therapeutic agents in the treatment of dystonias and spasms. Preparations comprising botulinum toxin complexes are commercially available, e.g. from Ipsen Ltd (Dysport®) or Allergan Inc. (Botox®). A high purity neurotoxic component, free of any complexing proteins, is for example available from Merz Pharmaceuticals GmbH, Frankfurt (Xeomin®).
  • Clostridial neurotoxins are usually injected into the affected muscle tissue, bringing the agent close to the neuromuscular end plate, i.e. close to the cellular receptor mediating its uptake into the nerve cell controlling said affected muscle. Various degrees of neurotoxin spread have been observed. The neurotoxin spread is thought to depend on the injected amount and the particular neurotoxin preparation. It can result in adverse side effects such as paralysis in nearby muscle tissue, which can largely be avoided by reducing the injected doses to the therapeutically relevant level. Overdosing can also trigger the immune system to generate neutralizing antibodies that inactivate the neurotoxin preventing it from relieving the involuntary muscle activity. Immunologic tolerance to botulinum toxin has been shown to correlate with cumulative doses.
  • At present, clostridial neurotoxins are still predominantly produced by fermentation processes using appropriate Clostridium strains. However, industrial production of clostridial neurotoxin from anaerobic Clostridium culture is a cumbersome and time-consuming process. Due to the high toxicity of the final product, the procedure must be performed under strict containment. During the fermentation process, the single-chain precursors are proteolytically cleaved by an unknown clostridial protease to obtain the biologically active di-chain clostridial neurotoxin. The degree of neurotoxin activation by proteolytic cleavage varies between different strains and neurotoxin serotypes, which is a major consideration for the manufacture due to the requirement of neurotoxin preparations with a well-defined biological activity. Furthermore, during fermentation processes using Clostridium strains the clostridial neurotoxins are produced as protein complexes, in which the neurotoxic component is embedded by accessory proteins. These accessory proteins have no beneficial effect on biological activity or duration of effect. They can however trigger an immune reaction in the patient, resulting in immunity against the clostridial neurotoxin. Manufacture of recombinant clostridial neurotoxins, which are not embedded by auxiliary proteins, might therefore be advantageous.
  • Methods for the recombinant expression of clostridial neurotoxins in E. coli are well known in the art (see, for example, WO 00/12728, WO 01/14570, or WO 2006/076902). Furthermore, clostridial neurotoxins have been expressed in eukaryotic expression systems, such as in Pichia pastoris, Pichia methanolica, Saccharomyces cerevisiae, insect cells and mammalian cells (see WO 2006/017749).
  • Recombinant botulinum neurotoxins may be expressed as single-chain precursors, which subsequently have to be proteolytically cleaved to obtain the final biologically active botulinum neurotoxin. Thus, botulinum neurotoxins may be expressed in high yield in rapidly-growing bacteria as relatively non-toxic single-chain polypeptides.
  • Furthermore, it might be advantageous to modify botulinum neurotoxin characteristics regarding biological activity, cell specificity, antigenic potential and duration of effect by genetic engineering to obtain recombinant neurotoxins with new therapeutic properties in specific clinical areas. Genetic modification of botulinum neurotoxins might allow altering the mode of action or expanding the range of therapeutic targets.
  • Botulinum toxin variants exhibiting an increased duration of effect and a high specific biological activity in neuromuscular tissue than naturally occurring botulinum toxins would be very advantageous in order to reduce administration frequency and the incidence of neutralizing antibody generation since immunologic tolerance to botulinum toxin is correlated with cumulative doses.
  • US 2002/0127247 describes clostridial neurotoxins comprising modifications in secondary modification sites and exhibiting altered biological persistence.
  • There is a strong demand to produce new botulinum neurotoxins serotype A with an increased duration of effect and with improved properties, in order to allow for exploitation of the therapeutic potential of BoNT serotype A, which have so far been considered impractical for certain clinical application. Ideally, the increased duration of effect of a particular botulinum neurotoxin serotype A could be adjusted in a tailor-made fashion in order to address any particular features and demands of a given indication, such as the amount of neurotoxin being administered, frequency of administration etc. In addition, it would be desirable to produce botulinum neurotoxins serotype A which exhibit an increased duration of effect than naturally occurring botulinum toxins serotype A with a high specific biological activity. To date, such aspects have not been solved satisfactorily.
  • OBJECTS OF THE INVENTION
  • It was an object of the invention to overcome the above illustrated drawbacks. In particular, it was an object of the invention to provide recombinant botulinum neurotoxins serotype A exhibiting an increased duration of effect than naturally occurring botulinum toxins with a high specific biological activity in neuromuscular tissue and to establish a reliable and accurate method for manufacturing and obtaining such recombinant botulinum neurotoxins. Such a method and novel precursor botulinum neurotoxins used in such methods would serve to satisfy the great need for recombinant botulinum neurotoxins exhibiting an increased duration of effect with a high specific biological activity.
  • SUMMARY OF THE INVENTION
  • The naturally occurring botulinum toxin serotypes display highly divergent durations of effect, probably due to their distinct subcellular localization. BoNT/A exhibits the longest persistence and was shown to localize in the vicinity of the plasma membrane of neuronal cells. However, additional factors such as degradation, spread or diffusion, and/or translocation rates might have a decisive impact on the differences in the duration for the individual botulinum toxin serotypes.
  • So far, except for the approach described and claimed in WO 2015/132004, no generally applicable method for modifying clostridial neurotoxins to increase their duration of effect is available. According to WO 2015/132004, a recombinant botulinum neurotoxin comprising a domain consisting of proline (P), alanine (A) and serine (S) residues (hereafter referred to “PASylated” botulinum neurotoxins) exhibits an increased duration of effect compared to a corresponding wildtype botulinum neurotoxin, but it was shown that such a “PASylated” botulinum neurotoxin also exhibits a decreased specific potency, i.e. a decreased specific biological activity compared to a corresponding wildtype botulinum neurotoxin. This means for the clinical setting, that a higher amount of such a modified botulinum neurotoxin has to be injected into a subject/patient to reach the same paralytic effect in comparison to a wildtype botulinum neurotoxin which in turn increases the risk of generating neutralizing antibodies that inactivate the neurotoxin.
  • Surprisingly, it has been found that recombinant botulinum neurotoxins serotype A having an increased duration of effect compared to a corresponding wildtype botulinum neurotoxin and a high specific potency, i.e. a high specific biological activity can be obtained by a two-fold modification. On the one hand these neurotoxins comprise at least two additional domains consisting of proline, alanine and an additional amino acid residue. Secondly, these neurotoxins according to the invention comprise at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • Thus, in one aspect, the present invention relates to a recombinant botulinum neurotoxin serotype A comprising at least two domains wherein each domain comprises an amino acid sequence consisting of at least 50 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, wherein the neurotoxin further comprises at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • In another aspect, the present invention relates to a composition, in particular to a pharmaceutical composition comprising the recombinant botulinum neurotoxin of the present invention.
  • In yet another aspect, the present invention relates to the use of the composition of the present invention for cosmetic treatment.
  • In another aspect, the present invention relates to a method for the generation of the recombinant botulinum neurotoxin of the present invention, comprising the step of obtaining a recombinant nucleic acid sequence encoding a recombinant single-chain precursor botulinum neurotoxin serotype A by modifying the nucleic acid sequence of the botulinum neurotoxin serotype A accordingly.
  • In another aspect, the present invention relates to a recombinant single-chain precursor botulinum neurotoxin serotype A comprising at least two additional domains wherein each domain comprises an amino acid sequence consisting of at least 50 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, and a modified alpha-exosite and/or beta-exosite of the light chain.
  • In another aspect, the present invention relates to a nucleic acid sequence encoding the recombinant single-chain precursor botulinum neurotoxin serotype A of the present invention.
  • In another aspect, the present invention relates to a method for obtaining the nucleic acid sequence of the present invention, comprising the step of adding nucleic acid sequences to the heavy and the light chain and in addition modifying a nucleic acid sequence encoding the alpha-exosite and/or at the beta-exosite of the light chain of the parental botulinum neurotoxin serotype A.
  • In another aspect, the present invention relates to a vector comprising the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention.
  • In another aspect, the present invention relates to a recombinant host cell comprising the nucleic acid sequence of the present invention, the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention.
  • In another aspect, the present invention relates to a method for producing the recombinant single-chain precursor botulinum neurotoxin of the present invention, comprising the step of expressing the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention in a recombinant host cell, or cultivating the recombinant host cell of the present invention under conditions that result in the expression of said nucleic acid sequence.
  • REFERENCE TO A SEQUENCE LISTING
  • A sequence listing containing SEQ ID NOs: 1-5, created Nov. 8, 2019, 48 KB is provided herewith in a computer-readable nucleotide/amino acid .txt file and is specifically incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1: Schematic Presentation of a modified botulinum toxin A (MaJ007=PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100=PAS100-BoNT/A-ISA2-PAS100), wherein both the light chain (LC) and the heavy chain (HC) each comprise an additional amino acid sequence consisting of 100 amino acid residues, wherein said amino acid sequence consists of proline (P), alanine (A) and serine (S) residues (PAS) and wherein the light chain (LC) comprises six amino acid modifications which are located at the alpha-exosite at positions D102, T109, K340, I348, N353, K356, wherein these amino acids are substituted as follows D102F, T109R, K340M, I348L, N353M, K356R.
  • FIG. 2: SDS⋅PAGE of purified BoNT/A MaJ007 (PAS100-BoNT/A-ISA2-PAS100). Lane 1: Molecular weight marker. Prior to applying the samples to the gel, R-mercaptoethanol was added. Lane “v.A.” (before activation): purified, non-activated single-chain PAS100-BoNT/A-ISA2-PAS100. Lanes “n.A.” (after activation) and “n.R.” (after purification) show light chain (LC) and heavy chain (HC) obtained after activation by thrombin under reducing conditions.
  • FIG. 3: Schematic Presentation of a modified botulinum toxin PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS(100)=PAS100-BoNT/A-ISA5-PAS100), wherein both the light chain (LC) and the heavy chain (HC) each comprise an additional amino acid sequence consisting of 100 amino acid residues, wherein said amino acid sequence consists of proline, alanine and serine residues (PAS) and wherein the light chain (LC) comprises four amino acid modifications are located at the beta-exosite at positions G169, T220, P239, S254, wherein these amino acids are substituted as follows G169I, T220R, P239M, S254T.
  • FIG. 4: SDS⋅PAGE of purified BoNT/A MaJ024 (PAS100-BoNT/A-ISA5-PAS(100). Lane 1: Molecular weight marker. Prior to applying the samples to the gel, R-mercaptoethanol was added. Lane “v.A.” (before activation): purified, non-activated single-chain PAS100-BoNT/A-ISA5-PAS(100. Lanes “n.A.” (after activation) and “n.R.” (after purification) show light chain (LC) and heavy chain (HC) obtained after activation by thrombin under reducing conditions.
  • FIG. 5: Mouse running assay with BoNT/A PAS100-BoNT/A-ISA5-PAS100 (MaJ024)
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention may be understood more readily by reference to the following detailed description of the invention and the examples included therein.
  • In one aspect, the present invention relates to a recombinant botulinum neurotoxin serotype A comprising at least two domains wherein each domain comprises an amino acid sequence consisting of at least 50 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, wherein the neurotoxin further comprises at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • In the context of the present invention, the term “botulinum neurotoxin serotype A” refers to a natural neurotoxin serotype A obtainable from bacteria Clostridium botulinum, or to a neurotoxin obtainable from alternative sources, including from recombinant technologies or from genetic or chemical modification. Particularly, the botulinum neurotoxins have a specific biological activity, i.e. endopeptidase activity.
  • Botulinum neurotoxins are produced as single-chain precursors that are proteolytically cleaved by an unknown clostridial endoprotease within the loop region to obtain the biologically active disulfide-linked di-chain form of the neurotoxin, which comprises two chain elements, a functionally active light chain and a functionally active heavy chain, where one end of the light chain is linked to one end of the heavy chain not via a peptide bond, but via a disulfide bond.
  • In the context of the present invention, the term “botulinum neurotoxin light chain” refers to that part of a botulinum neurotoxin serotype A that comprises an endopeptidase activity responsible for cleaving one or more proteins that is/are part of the so-called SNARE-complex involved in the process resulting in the release of neurotransmitter into the synaptic cleft: In naturally occurring botulinum neurotoxins, the light chain has a molecular weight of approx. 50 kDa.
  • In the context of the present invention, the term “alpha-exosite of the light chain” refers to four LC alpha-helices (102-113, 310-321, 335-348, and 351-358) that interface a helical motif of SNAP-25 that is approximately 30-50 amino acids away from the cleavage site of SNAP-25 which interacts with the substrate SNAP25 (see Xue S, Javor S, Hixon M S, Janda K D, Probing BoNT/A protease exosites: implications for inhibitor design and light chain longevity. Biochemistry. 2014; 53(43):6820-4).
  • In the context of the present invention, the term “beta-exosite of the light chain” refers to a beta-sheet region located in a beta-sheet close to the active site which interacts with the substrate SNAP25 comprising AS 162-254 (see Breidenbach MA1, Brunger A T. Substrate recognition strategy for botulinum neurotoxin serotype A, Nature. 2004, 432(7019):925-9).
  • In the context of the present invention, the term “botulinum neurotoxin heavy chain” refers to that part of a botulinum neurotoxin serotype A that is responsible for entry of the neurotoxin into the neuronal cell: In naturally occurring botulinum neurotoxins, the heavy chain has a molecular weight of approx. 100 kDa.
  • In the context of the present invention, the term “functionally active botulinum neurotoxin chain” refers to a recombinant botulinum neurotoxin serotype A chain able to perform the biological functions of a naturally occurring botulinum neurotoxin serotype A chain to at least about 50%, particularly to at least about 60%, to at least about 70%, to at least about 80%, and most particularly to at least about 90%, where the biological functions of botulinum neurotoxin chains include, but are not limited to, binding of the heavy chain to the neuronal cell, entry of the neurotoxin into a neuronal cell, release of the light chain from the di-chain neurotoxin, and endopeptidase activity of the light chain. Methods for determining a neurotoxic activity can be found, for example, in WO 95/32738, which describes the reconstitution of separately obtained light and heavy chains of tetanus toxin and botulinum toxin. Also cell-based assay methods as described for example in WO2009/114748, WO 2013/049508 and WO2014/207109.
  • In the context of the present invention, the term “about” or “approximately” means within 20%, alternatively within 10%, including within 5% of a given value or range. Alternatively, especially in biological systems, the term “about” means within about a log (i.e. an order of magnitude), including within a factor of two of a given value.
  • In the context of the present invention, the term “recombinant botulinum neurotoxin” refers to a composition comprising a botulinum neurotoxin serotype A that is obtained by expression of the neurotoxin in a heterologous cell such as E. coli, and including, but not limited to, the raw material obtained from a fermentation process (supernatant, composition after cell lysis), a fraction comprising a botulinum neurotoxin serotype A obtained from separating the ingredients of such a raw material in a purification process, an isolated and essentially pure protein, and a formulation for pharmaceutical and/or aesthetic use comprising a botulinum neurotoxin serotype A and additionally pharmaceutically acceptable solvents and/or excipients.
  • In the context of the present invention, the term “comprises” or “comprising” means “including, but not limited to”. The term is intended to be open-ended, to specify the presence of any stated features, elements, integers, steps or components, but not to preclude the presence or addition of one or more other features, elements, integers, steps, components, or groups thereof. The term “comprising” thus includes the more restrictive terms “consisting of” and “consisting essentially of”.
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention comprises at least one amino acid modification which is located at the alpha-exosite at at least one position selected from D102, T109, K340, I348, N353, K356.
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention comprises at least one amino acid modification which is located at the beta-exosite at at least one position selected from G169, T220, P239, S254.
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention comprises the following six modifications at the alpha-exosite of the light chain:
      • (i) D102F modification (aspartic acid at position 102 of the light chain is replaced by a phenylalanine),
      • (ii) T109R modification (threonine at position 109 is replaced by an arginine),
      • (iii) K340M modification (lysine at position 340 is replaced by a methionine),
      • (iv) I348L modification (isoleucine at position 348 of the light chain is replaced by a leucine),
      • (v) N353M modification (asparagine at position 353 of the light chain is replaced by a methionine),
      • (vi) K356R modification (lysine at position 356 of the light chain is replaced by an arginine).
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention comprises the following four modifications at the beta-exosite of the light chain:
      • (i) G169I modification (glycine at position 169 of the light chain is replaced by a isoleucine),
      • (ii) T220R modification (threonine at position 220 is replaced by an arginine),
      • (iii) P239M modification (proline at position 239 is replaced by a methionine),
      • (iv) S254T modification (serine at position 254 of the light chain is replaced by a threonine).
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention comprises two domains wherein each domain comprises an amino acid sequence consisting of between 70 and 260 amino acid residues, particularly 100 amino acid residues, 150 amino acid residues, or 200 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one serine residues.
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention comprises two domains wherein each domain comprises an amino acid sequence consisting of between 70 and 150 amino acid residues or between 80 and 120 amino acid residues or between 90 and 110 amino acid residues, wherein said amino acid sequence consists of at least one proline, at least one alanine and at least one serine residues.
  • In the context of the present invention, the term “functional variant of a botulinum neurotoxin” refers to a botulinum neurotoxin serotype A that differs in the amino acid sequence and/or the nucleic acid sequence encoding the amino acid sequence from a botulinum neurotoxin serotype A, but is still functionally active. In the context of the present invention, the term “functionally active” refers to the property of a recombinant botulinum neurotoxin serotype A to exhibit a biological activity of at least about 20%, particularly to at least about 40%, at least about 70%, at least about 80%, and most particularly at least about 90% of the biological activity of a naturally occurring parental botulinum neurotoxin, i.e. a parental botulinum neurotoxin serotype A without modifications at the C-terminus of the light chain, where the biological functions include, but are not limited to, binding to the neurotoxin receptor, entry of the neurotoxin into a neuronal cell, release of the light chain from the two-chain neurotoxin, and endopeptidase activity of the light chain, and thus inhibition of neurotransmitter release from the affected nerve cell. In vivo assays for assessing biological activity include the mouse LD50 assay and the ex vivo mouse hemidiaphragm assay as described by Pearce et al. (Pearce 1994, Toxicol. Appl. Pharmacol. 128: 69-77) and Dressler et al. (Dressler 2005, Mov. Disord. 20:1617-1619, Keller 2006, Neuroscience 139: 629-637) or a cell-based assay as described in WO2009/114748, WO2014/207109 or WO 2013/049508. The biological activity is commonly expressed in Mouse Units (MU). As used herein, 1 MU is the amount of neurotoxic component, which kills 50% of a specified mouse population after intraperitoneal injection, i.e. the mouse i.p. LD50.
  • On the protein level, a functional variant will maintain key features of the corresponding botulinum neurotoxin serotype A, such as key residues for the endopeptidase activity in the light chain, or key residues for the attachment to the neurotoxin receptors or for translocation through the endosomal membrane in the heavy chain, but may contain modifications comprising a substitution of one or more amino acids of the corresponding botulinum neurotoxin.
  • In another embodiment, the functional variant of a botulinum neurotoxin serotype A additionally comprises a signal peptide. Usually, said signal peptide will be located at the N-terminus of the neurotoxin. Many such signal peptides are known in the art and are comprised by the present invention. In particular, the signal peptide results in transport of the neurotoxin across a biological membrane, such as the membrane of the endoplasmic reticulum, the Golgi membrane or the plasma membrane of a eukaryotic or prokaryotic cell. It has been found that signal peptides, when attached to the neurotoxin, will mediate secretion of the neurotoxin into the supernatant of the cells. In certain embodiments, the signal peptide will be cleaved off in the course of, or subsequent to, secretion, so that the secreted protein lacks the N-terminal signal peptide, is composed of separate light and heavy chains, which are covalently linked by disulfide bridges, and is proteolytically active.
  • In particular embodiments, the functional variant has a sequence identity of at least about 40%, at least about 50%, at least about 60%, at least about 70% or most particularly at least about 80%, and a sequence homology of at least about 60%, at least about 70%, at least about 80%, at least about 90%, or most particularly at least about 95% to the corresponding part in the parental botulinum neurotoxin serotype A. Methods and algorithms for determining sequence identity and/or homology, including the comparison of variants having deletions, additions, and/or substitutions relative to a parental sequence, are well known to the practitioner of ordinary skill in the art. The term “identity” as used herein refers to sequence identity characterized by determining the number of identical amino acids between two nucleic acid sequences or two amino acid sequences wherein the sequences are aligned so that the highest order match is obtained. It can be calculated using published techniques or methods codified in computer programs such as, for example, BLASTP, BLASTN or FASTA (Altschul 1990, J Mol Biol 215, 403). The percent identity values are, in one aspect, calculated over the entire amino acid sequence. A series of programs based on a variety of algorithms is available to the skilled worker for comparing different sequences. In this context, the algorithms of Needleman and Wunsch or Smith and Waterman give particularly reliable results. To carry out the sequence alignments, the program PileUp (Higgins 1989, CABIOS 5, 151) or the programs Gap and BestFit (Needleman 1970, J Mol Biol 48; 443; Smith 1981, Adv Appl Math 2, 482), which are part of the GCG software packet (Genetics Computer Group 1991, 575 Science Drive, Madison, Wis., USA 53711), may be used. The sequence identity values recited above in percent (%) are to be determined, in another aspect of the invention, using the program GAP over the entire sequence region with the following settings: Gap Weight: 50, Length Weight: 3, Average Match: 10.000 and Average Mismatch: 0.000, which, unless otherwise specified, shall always be used as standard settings for sequence alignments. On the DNA level, the nucleic acid sequences encoding the functional homologue and the parental botulinum neurotoxin may differ to a larger extent due to the degeneracy of the genetic code. It is known that the usage of codons is different between prokaryotic and eukaryotic organisms. Thus, when expressing a prokaryotic protein such as a botulinum neurotoxin, in a eukaryotic expression system, it may be necessary, or at least helpful, to adapt the nucleic acid sequence to the codon usage of the expression host cell, meaning that sequence identity or homology may be rather low on the nucleic acid level.
  • In the context of the present invention, the term “variant” refers to a botulinum neurotoxin serotype A that is a chemically, enzymatically, or genetically modified derivative of a corresponding neurotoxin of C. botulinum neurotoxin serotype A. A chemically modified derivative may be one that is modified by pyruvation, phosphorylation, sulfatation, lipidation, pegylation, glycosylation and/or the chemical addition of an amino acid or a polypeptide comprising between 2 and about 100 amino acids, including modification occurring in the eukaryotic host cell used for expressing the derivative. An enzymatically modified derivative is one that is modified by the activity of enzymes, such as endo- or exoproteolytic enzymes, including modification by enzymes of the eukaryotic host cell used for expressing the derivative. As pointed out above, a genetically modified derivative is one that has been modified by deletion or substitution of one or more amino acids contained in, or by addition of one or more amino acids (including polypeptides comprising between 2 and about 100 amino acids) to, the amino acid sequence of said botulinum neurotoxin. Methods for designing and constructing such chemically or genetically modified derivatives and for testing of such variants for functionality are well known to anyone of ordinary skill in the art.
  • In particular embodiments, the recombinant botulinum neurotoxin serotype A according to the invention is used in the treatment of a disease requiring improved chemodenervation, wherein the recombinant neurotoxin causes both (i) an increased duration of effect relative to a wildtype botulinum neurotoxin serotype A and (ii) an increased specific biological activity relative to a botulinum neurotoxin serotype A comprising two domains consisting of proline, alanine and serine residues without said modifications in the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
  • In the context of the present invention, the term “increased duration of effect” or “increased duration of action” refers to a longer lasting denervation mediated by a botulinum neurotoxin serotype A of the present invention. For example, as disclosed herein, administration of a disulfide-linked di-chain botulinum neurotoxin serotype A comprising two domains according to the invention results in localized paralysis for a longer period of time relative to administration of an identical disulfide-linked di-chain botulinum neurotoxin serotype A without the at least two domains according to the present invention.
  • In the context of the present invention, the term “increased duration of effect/action” is defined as a more than about 20%, particularly more than about 50%, more particularly more than about 90% increased duration of effect of the recombinant neurotoxin of the present invention relative to the identical neurotoxin without the two domains according to the invention. For example, an “increased duration of effect” can be determined using the “Mouse Running Assay”. The “Mouse Running Assay” is well-known to the person skilled in the art and measures the daily running distance of a mouse in a treadmill after a botulinum neurotoxin was injected into the M. gastrocnemius (see Keller J E. Recovery from botulinum neurotoxin poisoning in vivo. Neuroscience. 2006 May 12; 139(2):629-37). The distance which a mouse is able to run in the treadmill the day before the botulinum neurotoxin is injected is used as comparison and is set as 100%. A daily running distance of no more than 80% of the initial running distance is regarded as paralysis of the muscle. The duration of effect is determined by the time period between the time point attaining a half-maximal paralysis, i.e. about 40% of the initial running distance and the time point when paralysis reaches recovery, i.e. 40% of the initial running distance. If this time period is longer than 2 days compared with the standard (wildtype BoNT), the botulinum neurotoxin is considered to exhibit an “increased duration of effect/action” provided that the mutated BoNT exhibits a similar potency i.e shows a similar maximal paralysis (reduction of the running distance) of about 80-90%.
  • In the context of the present invention, the “specific endoprotease activity” is measured in an assay which is described in Jones et al. (J Immunol Methods. 2008 Jan. 1; 329(1-2):92-101) and Hallis et al. (J Clin Microbiol. 1996 (8):1934-8; 1996) and is described further below in example 2.
  • In the context of the present invention, the term “decreased specific endoprotease activity” is defined as an at least 20% lower amount of cleavage product produced by a BoNT/A mutant in the endoprotease assay compared to the BoNT/A wildtype applying the same amount of protein.
  • In the context of the present invention, the term “specific biological activity” relates to the “specific endoprotease activity” mentioned above and can be derived also from the maximal paralysis shown in the “Mouse Running Assay” described above.
  • In the context of the present invention the term “denervation” refers to denervation resulting from administration of a chemodenervating agent, for example a neurotoxin.
  • In the context of the present invention, the term “localized denervation” or “localized paralysis” refers to denervation of a particular anatomical region, usually a muscle or a group of anatomically and/or physiologically related muscles, which results from administration of a chemodenervating agent, for example a neurotoxin, to the particular anatomical region.
  • Without wishing to be bound by theory, the recombinant botulinum neurotoxins of the present invention might show increased biological half-life, reduced degradation rates, decreased diffusion rates, increased uptake by neuronal cells, and/or modified intracellular translocation rates, in each case relative to an identical parental clostridial neurotoxin without the at least two domains according to the invention.
  • In the context of the present invention, the term “biological half-life” specifies the lifespan of a protein, for example of a botulinum neurotoxin serotype A, in vivo. In the context of the present invention, the term “biological half-life” refers to the period of time, by which half of a protein pool is degraded in vivo. For example it refers to the period of time, by which half of the amount of botulinum neurotoxin of one administered dosage is degraded.
  • In another aspect, the present invention relates to a composition, in particular a pharmaceutical or cosmetic composition comprising the recombinant botulinum neurotoxin of the present invention. For preparing a preparation comprising a botulinum neurotoxin serotype A the toxin can be formulated by various techniques dependent on the desired application purposes which are known in the art. For example, the (biologically active) botulinum neurotoxin polypeptide can be used in combination with one or more pharmaceutically acceptable carriers as a pharmaceutical composition. The pharmaceutically acceptable carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and being not deleterious to the recipient thereof. The pharmaceutical carrier employed may include a solid, a gel, or a liquid. Exemplary of solid carriers are lactose, terra alba, sucrose, talc, gelatine, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemplary of liquid carriers are glycerol, phosphate buffered saline solution, water, emulsions, various types of wetting agents, and the like. Suitable carriers comprise those mentioned above and others well known in the art, see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. In an aspect, the pharmaceutical composition can be dissolved in a diluent, prior to administration. The diluent is also selected so as not to affect the biological activity of the Neurotoxin product. Examples of such diluents are distilled water or physiological saline. In addition, the pharmaceutical composition or formulation may also include other carriers or non-toxic, non-therapeutic, non-immunogenic stabilizers and the like. Thus, the formulated Neurotoxin product can be present, in an aspect, in liquid or lyophilized form. In an aspect, it can be present together with glycerol, protein stabilizers (HSA) or non-protein stabilizers such as polyvinyl pyrrolidone (PVP), hyaluronic acid or free amino acids. In an aspect, suitable non-proteinaceous stabilizers are disclosed in WO 2005/007185 or WO 2006/020208. The formulated Neurotoxin product may be used for human or animal therapy of various diseases or disorders in a therapeutically effective dose or for cosmetic purposes.
  • In particular embodiments, the recombinant botulinum neurotoxin of the present invention or the pharmaceutical composition of the present invention is for use in the treatment of a disease or condition taken from the list of: cervical dystonia (spasmodic torticollis), blepharospasm, severe primary axillary hyperhidrosis, achalasia, lower back pain, benign prostate hypertrophy, chronic focal painful neuropathies, migraine and other headache disorders.
  • Additional indications where treatment with botulinum neurotoxins is currently under investigation and where the pharmaceutical composition of the present invention may be used, include pediatric incontinence, incontinence due to overactive bladder, and incontinence due to neurogenic bladder, anal fissure, spastic disorders associated with injury or disease of the central nervous system including trauma, stroke, multiple sclerosis, Parkinson's disease, or cerebral palsy, focal dystonias affecting the limbs, face, jaw or vocal cords, temporomandibular joint (TMJ) pain disorders, diabetic neuropathy, wound healing, excessive salivation, vocal cord dysfunction, reduction of the Masseter muscle for decreasing the size of the lower jaw, treatment and prevention of chronic headache and chronic musculoskeletal pain, treatment of snoring noise, assistance in weight loss by increasing the gastric emptying time.
  • Most recently, clostridial neurotoxins have been evaluated for the treatment of other new indications, for example painful keloid, diabetic neuropathic pain, refractory knee pain, trigeminal neuralgia trigger-zone application to control pain, scarring after cleft-lip surgery, cancer and depression.
  • In yet another aspect, the present invention relates to the use of the composition of the present invention for cosmetic treatment.
  • Thus, in another aspect, the present invention relates to a method of cosmetically treating a patient, comprising the step of administering a composition comprising a recombinant clostridial neurotoxin according to the present invention to a patient desiring such cosmetic treatment
  • In the context of the present invention, the term “cosmetic treatment” relates to uses in cosmetic or aesthetic applications, such as the treatment of wrinkles, crow's feet, glabella frown lines, reduction of the masseter muscle, reduction of the calves, removing of facial asymmetries etc.
  • In another aspect, the present invention relates to a method for the generation of the recombinant botulinum neurotoxin of the present invention, comprising the step of obtaining a recombinant nucleic acid sequence encoding a recombinant single-chain precursor botulinum neurotoxin by the insertion of a nucleic acid sequence encoding at least two domains consisting of proline, alanine and additional amino acid residues, selected from the group consisting of serine, threonine, tyrosine and glutamine residues into a nucleic acid sequence encoding a parental clostridial neurotoxin and by modifying the nucleic acid sequence encoding a parental botulinum neurotoxin at the alpha-exosite and/or at the beta-exosite of the light chain.
  • In the context of the present invention, the term “recombinant nucleic acid sequence” refers to a nucleic acid, which has been generated by joining genetic material from two different sources.
  • In the context of the present invention, the term “single-chain precursor botulinum neurotoxin” refers to a single-chain precursor for a disulfide-linked di-chain botulinum neurotoxin, comprising a functionally active botulinum neurotoxin light chain, a functionally active neurotoxin heavy chain, and a loop region linking the C-terminus of the light chain with the N-terminus of the heavy chain.
  • In the context of the present invention, the term “recombinant single-chain precursor botulinum neurotoxin” refers to a single-chain precursor botulinum neurotoxin comprising at least two domains consisting of proline, alanine and additional amino acid residues, selected from the group consisting of serine, threonine, tyrosine and glutamine residues, and a modified alpha-exosite and/or beta-exosite of the light chain of the neurotoxin.
  • In particular embodiments, the recombinant single-chain precursor botulinum neurotoxin comprises a protease cleavage site in said loop region.
  • Single-chain precursor botulinum neurotoxins have to be proteolytically cleaved to obtain the final biologically active botulinum neurotoxins. Proteolytic cleavage may either occur during heterologous expression by host cell enzymes, or by adding proteolytic enzymes to the raw protein material isolated after heterologous expression. Naturally occurring botulinum neurotoxins usually contain one or more cleavage signals for proteases which post-translationally cleave the single-chain precursor molecule, so that the final di- or multimeric complex can form. At present, botulinum neurotoxins are still predominantly produced by fermentation processes using appropriate Clostridium strains. During the fermentation process, the single-chain precursors are proteolytically cleaved by an unknown clostridial protease to obtain the biologically active di-chain clostridial neurotoxin. In cases, where the single-chain precursor molecule is the precursor of a protease, autocatalytic cleavage may occur. Alternatively, the protease can be a separate non-clostridial enzyme expressed in the same cell. WO 2006/076902 describes the proteolytic cleavage of a recombinant clostridial neurotoxin single-chain precursor at a heterologous recognition and cleavage site by incubation of the E. coli host cell lysate. The proteolytic cleavage is carried out by an unknown E. coli protease. In certain applications of recombinant expression, modified protease cleavage sites have been introduced recombinantly into the interchain region between the light and heavy chain of clostridial toxins, e.g. protease cleavage sites for human thrombin or non-human proteases (see WO 01/14570).
  • In particular embodiments, the protease cleavage site is a site that is cleaved by a protease selected from the list of: thrombin, trypsin, enterokinase, factor Xa, plant papain, insect papain, crustacean papain, enterokinase, human rhinovirus 3C protease, human enterovirus 3C protease, tobacco etch virus protease, Tobacco Vein Mottling Virus, subtilisin and caspase 3.
  • In a particular embodiment, the recombinant single-chain precursor botulinum neurotoxin serotype A further comprises a binding tag, particularly selected from the group comprising: glutathione-S-transferase (GST), maltose binding protein (MBP), a His-tag, a StrepTag, or a FLAG-tag.
  • In the context of the present invention, the term “parental botulinum neurotoxin” refers to an initial botulinum neurotoxin without modifications selected from a natural botulinum neurotoxin, a functional variant of a natural botulinum neurotoxin or a chimeric botulinum neurotoxin.
  • In particular embodiments, the method for the generation of the recombinant botulinum neurotoxin of the present invention further comprises the step of heterologously expressing said recombinant nucleic acid sequence in a host cell, particularly in a bacterial host cell, more particularly in an E. coli host cell.
  • In certain embodiments, the E. coli cells are selected from E. coli XL1-Blue, Nova Blue, TOP10, XL10-Gold, BL21, and K12.
  • In particular embodiments, the method for the generation of the recombinant botulinum neurotoxin of the present invention additionally comprises at least one of the steps of (i) generating a disulfide-linked di-chain recombinant botulinum neurotoxin according to the invention by causing or allowing contacting of said recombinant single-chain precursor botulinum neurotoxin with an endoprotease and (ii) purification of said recombinant single-chain precursor botulinum neurotoxin or said disulfide-linked di-chain recombinant botulinum neurotoxin by chromatography.
  • In particular embodiments, the recombinant single-chain precursor botulinum neurotoxin of the present invention, or the recombinant disulfide-linked di-chain botulinum neurotoxin of the present invention, is purified after expression, or in the case of the recombinant disulfide-linked di-chain botulinum neurotoxin, after the cleavage reaction. In particular such embodiments, the protein is purified by chromatography, particularly by immunoaffinity chromatography, or by chromatography on an ion exchange matrix, a hydrophobic interaction matrix, or a multimodal chromatography matrix, particularly a strong ion exchange matrix, more particularly a strong cation exchange matrix.
  • In the context of the present invention, the term “causing . . . contacting of said recombinant single-chain precursor botulinum neurotoxin . . . with an endoprotease” refers to an active and/or direct step of bringing said neurotoxin and said endoprotease in contact, whereas the term “allowing contacting of a recombinant single-chain precursor botulinum neurotoxin . . . with an endoprotease” refers to an indirect step of establishing conditions in such a way that said neurotoxin and said endoprotease are getting in contact to each other.
  • In the context of the present invention, the term “endoprotease” refers to a protease that breaks peptide bonds of non-terminal amino acids (i.e. within the polypeptide chain). As they do not attack terminal amino acids, endoproteases cannot break down peptides into monomers.
  • In particular embodiments, cleavage of the recombinant single-chain precursor botulinum neurotoxin is near-complete.
  • In the context of the present invention, the term “near-complete” is defined as more than about 95% cleavage, particularly more than about 97.5%, more particularly more than about 99% as determined by SDS-PAGE and subsequent Western Blot or reversed phase chromatography.
  • In particular embodiments, cleavage of the recombinant single-chain precursor botulinum neurotoxin of the present invention occurs at a heterologous cleavage signal located in the loop region of the recombinant precursor botulinum neurotoxin.
  • In particular embodiments, the cleavage reaction is performed with crude host cell lysates containing said single-chain precursor protein.
  • In other particular embodiments, the single-chain precursor protein is purified or partially purified, particularly by a first chromatographic enrichment step, prior to the cleavage reaction.
  • In the context of the present invention, the term “purified” relates to more than about 90% purity. In the context of the present invention, the term “partially purified” relates to purity of less than about 90% and an enrichment of more than about two fold.
  • In another aspect, the present invention relates to a recombinant single-chain botulinum neurotoxin serotype A, which is a precursor for the recombinant botulinum neurotoxin of the present invention. Thus, in such aspect, the present invention relates to a recombinant single-chain precursor botulinum neurotoxin serotype A comprising two domains consisting of proline, alanine and additional amino acid residues, selected from the group consisting of serine, threonine, tyrosine and glutamine residues, and a modified alpha-exosite and/or beta-exosite of the light chain of the neurotoxin.
  • In another aspect, the present invention relates to a method for obtaining the nucleic acid sequence of the present invention, comprising the step of modifying a nucleic acid sequence encoding a parental botulinum neurotoxin serotype A.
  • In another aspect, the present invention relates to a vector comprising the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention.
  • In another aspect, the present invention relates to a recombinant host cell comprising the nucleic acid sequence of the present invention, the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention.
  • In another aspect, the present invention relates to a method for producing the recombinant single-chain precursor botulinum neurotoxin of the present invention, comprising the step of expressing the nucleic acid sequence of the present invention, or the nucleic acid sequence obtainable by the method of the present invention, or the vector of the present invention in a recombinant host cell, or cultivating the recombinant host cell of the present invention under conditions that result in the expression of said nucleic acid sequence.
  • EXAMPLES Example 1: Generation and Purification of a PASylated Botulinum Toxin Type A (PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100)
  • The nucleic acid construct encoding a “PAS” module comprising 100 amino acid residues built from the amino acids proline (P), alanine (A) and serine (S) was synthetically produced, wherein the following motive was used (ASPAAPAPASPAAPAPSAPA)5. By using restriction enzymes NdeI and SwaI, the corresponding gene module was first inserted at the N-terminus of recombinant BoNT/A (rBoNT/A). In a second step, the PAS module was inserted at the C-terminus of the heavy chain by using restriction enzymes BglII and AatII. The correct cloning was verified by sequencing. For expression of BoNT/A in E. coli gene constructs were cloned into pET29c. The variants contain fused His6- and Strep-affinity tags which can be cleaved after protein purification via thrombin. In order to construct a plasmid for the expression of BoNT/A a synthetic gene with the corresponding mutations of D102F T109R K340M I348L N353M K356R in the BoNT/A1 wild type sequence and restriction sites SwaI and ScaI was used.
  • Protein Expression and Purification.
  • Expression of rBoNT/A variants was performed in Riesenberg media with 50 μg/mL Kanamycin{Riesenberg, 1991 #1}. Cells were grown in shake flasks (37° C., 175 r.p.m) until an OD600 of 1.5-2 was reached. For induction of protein expression 1 mM IPTG (Fermentas) was added to the E. coli culture. Protein synthesis was performed for 24 h (15° C., 175 r.p.m.). Cells were collected by centrifugation (5,000 r.p.m., 20 min, 4° C.) and resuspended in His binding buffer pH 8.0 (50 mM Tris, 150 mM NaCl, 5 mM Imidazol) containing EDTA-free protease inhibitor complete (Roche Diagnostics). For the determination of endopeptidase activity and in vivo characterization, the different toxin variants were extracted and purified. Resuspended pellets were disrupted in 2-3 cycles by a French Press Cell Disrupter (Thermo Electron Corporation) at 4° C. The resulting crude extracts were centrifuged (20,000 r.p.m., 30 min, 4° C.), and the supernatants with the soluble proteins were recovered. Protein purification was carried out by fast protein liquid chromatography (GE Healthcare) using a three step purification protocols. The first capture step was performed by IMAC using a HisTrap HP 1 mL column (GE Healthcare). The column was washed (1 ml min-1 working flow) using a two-step protocol with His elution buffer (50 mM Tris, 150 mM NaCl, 400 mM Imidazol pH 8.0). The elution of the toxin proteins occurred at 400 mM Imidazol. In a further step a Strep-Tactin affinity chromatography was performed as previously described (IBA GmbH). As an alternative instead of a second affinity chromatography a cation exchange chromatography with a HiTrap SP HP 1 mL column (GE Healthcare, Freiburg, Germany) was used. The corresponding samples were diluted with SP binding buffer (50 mM Tris, pH 8) and eluted with SP elution buffer (50 mM Tris, 1 M NaCl, pH 8). This procedure was followed by a SEC using a Superdex 200 10/300 column (GE Healthcare). The SEC running buffer (20 mM Tris, 150 mM NaCl, 2.5 mM CaCl2) pH 7.7) was also used to store the purified protein solutions in aliquots at −20° C. Each protein was analyzed by applying 0.5-1 μg on 4-12% gradient SDS-PAGE (Novex Life Technologies) and stained with Coomassie G-250 based SimplyBlue safe stain (Pierce). Each protein was judged >98% pure before applying in vitro or in vivo experiments.
  • Thrombin Cleavage
  • BoNT/A preparations were activated with Thrombin (Merck Millipore; 8 U/1 mg BoNT) for 24 h at 20° C. yielding >99% of di-chain toxin. Afterwards the cleavage protease was eliminated with the previously described Strep-Tactin Kit (IBA GmbH).
  • Expression was performed in expression strain E. coli B121. Purification was done using a combination of his affinity, ion exchange and size exclusion chromatography, followed by activation using thrombin. FIG. 2 summarizes the results of purification and activation.
  • Example 2: Determination of Endoproteinase Activity of Wild Type BoNT/A and PASylated BoNT/A
  • The specific endoproteinase activity of wildtype BoNT/A and modified BoNT/A variants was determined by an assay which is described in Jones R G, Ochiai M, Liu Y, Ekong T, Sesardic D. Development of improved SNAP25 endopeptidase immuno-assays for botulinum type A and E toxins. J Immunol Methods. 2008 Jan. 1; 329(1-2):92-101 and Hallis B, James B A, Shone C C Development of novel assays for botulinum type A and B neurotoxins based on their endopeptidase activities. J Clin Microbiol. 1996 (8):1934-8. The “1-step TMB Ultra” solution from Perbio Science was used for staining and quantification. A C-terminal fragment of 70 amino acids of SNAP25 from IBA GmBH was used in the reaction as a substrate. The primary mouse antibody from R&D Systems specifically recognizes SNAP25 fragments cleaved only by BoNT/A. The secondary antibody was an anti-mouse-IgG-HRP-antibody conjugate from Dianova. The assay was performed on a 96 microtiter plate and the amount of cleaved material was determined after 120 minutes by measuring the absorption at 450 nm (Molecular Devices, FilterMax F5). For the assay equal amounts of proteins were used.
  • The results of the measured endopeptidase activities are shown in the following table 1:
  • TABLE 1
    Endoproteinase activities of mutants
    Batch Construct Endopeptidase assay
    Xeomin Wildtype BoNT/A # 100%
    DaSch021 PAS100-BoNT/A-PAS100 30-40%
    MaJ007 PAS(100)-BoNT/A-ISA2-PAS(100) 85-95%
    ISA2 = (D102F T109R K340M I348L N353M K356R);
    # Standard
  • Table 1 shows that the specific endoproteinase activity of PASylated botulinum toxin A was increased by introduced mutations.
  • Example 3: Generation and Purification of a PASylated Botulinum Toxin Type A (PAS100-BoNT/A G169I, T220R, P239M, S254-PAS100)
  • The nucleic acid construct encoding a “PAS” module comprising 100 amino acid residues built from the amino acids proline (P), alanine (A) and serine (S) was synthetically produced, wherein the following motive was used (ASPAAPAPASPAAPAPSAPA)5. By using restriction enzymes NdeI and SwaI the corresponding gene module was first inserted at the N-terminus of recombinant BoNT/A (rBoNT/A). In a second step, the PAS module was inserted at the C-terminus of the heavy chain by using restriction enzymes BglII and AatII. The correct cloning was verified by sequencing. For expression of BoNT/A in E. coli gene constructs were cloned into pET29c. The variants contain fused His6- and Strep-affinity tags which can be cleaved after protein purification via thrombin. In order to construct a plasmid for the expression of BoNT/A a synthetic gene with the corresponding mutations of G169I, T220R, P239M, S254 in the BoNT/A1 wild type sequence and restriction sites SwaI and Scat was used.
  • Protein expression was performed as described above (see Example 1) in expression strain E. coli BL21. Purification was done using a combination of his affinity, ion exchange and size exclusion chromatography, followed by activation using thrombin. FIG. 3 summarizes the results of purification and activation.
  • Example 4: Duration of Effect of PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS100 in the “Mouse Running Assay”
  • Two different dosages of PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS100 (=MaJ024), i.e. 6.0 pg and 9.0 pg were injected into the M. gastrocnemius of each mice in comparison to standard Xeomin® (3 pg; 0.6 U) and PASylated Botulinum Toxin Type A without the introduced mutations (=Dasch021). The mice had been trained in a treadmill. The daily running distance in the treadmill was measured over 21 days. The paralysis caused by the toxins was plotted as percentage of the running distance on the day before the injection, which was set as 100%, against the time (see FIG. 5).
  • As shown in FIG. 5, only 6 pg of PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS(100) (=MaJ024) was required to achieve the same paralysis in comparison to 9 pg of Dasch021 (PASylated BoNT/A without introduced mutations) which indicates that the specific potency of MaJ024 was clearly increased in comparison to Dasch021.
  • Table 1: Sequences
  • SEQ ID NO 1: recombinant BoNT A including His.tag
    SEQ ID NO 2: recombinant PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100 including His.tag
    SEQ ID NO 3: recombinant PAS100-BoNT/A (G169I, T220R, P239M, S254)-PAS100 including His.tag)
    SEQ ID NO 4: recombinant PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100 including His.tag (nucleic acid sequence)
    SEQ ID NO 5: recombinant PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS100 including His.tag) (nucleic acid sequence)
  • SEQ ID NO 1:
    MPFVNKQFNYKDPVNGVDIAYIKIPNAGQMQPVKAFKIHNKIWVIPERDTFTNPEEGDLN
    PPPEAKQVPVSYYDSTYLSTDNEKDNYLKGVTKLFERIYSTDLGRMLLTSIVRGIPFWGG
    STIDTELKVIDTNCINVIQPDGSYRSEELNLVIIGPSADIIQFECKSFGHEVLNLTRNGY
    GSTQYIRFSPDFTFGFEESLEVDTNPLLGAGKFATDPAVTLAHELIHAGHRLYGIAINPN
    RVFKVNTNAYYEMSGLEVSFEELRTFGGHDAKFIDSLQENEFRLYYYNKFKDIASTLNKA
    KSIVGTTASLQYMKNVFKEKYLLSEDTSGKFSVDKLKFDKLYKMLTEIYTEDNFVKFFKV
    LNRKTYLNFDKAVFKINIVPKVNYTIYDGFNLRNTNLAANFNGQNTEINNMNFTKLKNFT
    GLFEFYKLLCVRGIITSKAGAGKSLVPRGSAGAGALNDLCIKVNNWDLFFSPSEDNFTND
    LNKGEEITSDTNIEAAEENISLDLIQQYYLTFNFDNEPENISIENLSSDIIGQLELMPNI
    ERFPNGKKYELDKYTMFHYLRAQEFEHGKSRIALTNSVNEALLNPSRVYTFFSSDYVKKV
    NKATEAAMFLGWVEQLVYDFTDETSEVSTTDKIADITIIIPYIGPALNIGNMLYKDDFVG
    ALIFSGAVILLEFIPEIAIPVLGTFALVSYIANKVLTVQTIDNALSKRNEKWDEVYKYIV
    TNWLAKVNTQIDLIRKKMKEALENQAEATKAIINYQYNQYTEEEKNNINFNIDDLSSKLN
    ESINKAMININKFLNQCSVSYLMNSMIPYGVKRLEDFDASLKDALLKYIYDNRGTLIGQV
    DRLKDKVNNTLSTDIPFQLSKYVDNQRLLSTFTEYIKNIINTSILNLRYESNHLIDLSRY
    ASKINIGSKVNFDPIDKNQIQLFNLESSKIEVILKNAIVYNSMYENFSTSFWIRIPKYFN
    SISLNNEYTIINCMENNSGWKVSLNYGEIIWTLQDTQEIKQRVVFKYSQMINISDYINRW
    IFVTITNNRLNNSKIYINGRLIDQKPISNLGNIHASNNIMFKLDGCRDTHRYIWIKYFNL
    FDKELNEKEIKDLYDNQSNSGILKDFWGDYLQYDKPYYMLNLYDPNKYVDVNNVGIRGYM
    YLKGPRGSVMTTNIYLNSSLYRGTKFIIKKYASGNKDNIVRNNDRVYINVVVKNKEYRLA
    TNASQAGVEKILSALEIPDVGNLSQVVVMKSKNDQGITNKCKMNLQDNNGNDIGFIGFHQ
    FNNIAKLVASNWYNRQIERSSRTLGCSWEFIPVDDGWGERPLGDLVPRGSANSSSVDKLW
    SHPQFEKLEHHHHHH

    SEQ ID NO 2: recombinant PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100 including His.tag
  • MGSSHHHHHHGSLVPRSSSASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAA
    SPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAA
    PFVNKQFNYKDPVNGVDIAYIKIPNAGQMQPVKAFKIHNKIWVIPERDTFTNPEEGDLNP
    PPEAKQVPVSYYDSTYLSTDNEKDNYLKGVTKLFERIYSTFLGRMLLRSIVRGIPFWGGS
    TIDTELKVIDTNCINVIQPDGSYRSEELNLVIIGPSADIIQFECKSFGHEVLNLTRNGYG
    STQYIRFSPDFTFGFEESLEVDTNPLLGAGKFATDPAVTLAHELIHAGHRLYGIAINPNR
    VFKVNTNAYYEMSGLEVSFEELRTFGGHDAKFIDSLQENEFRLYYYNKFKDIASTLNKAK
    SIVGTTASLQYMKNVFKEKYLLSEDTSGKFSVDKLKFDMLYKMLTELYTEDMFVRFFKVL
    NRKTYLNFDKAVFKINIVPKVNYTIYDGFNLRNTNLAANFNGQNTEINNMNFTKLKNFTG
    LFEFYKLLCVRGIITSKAGAGKSLVPRGSAGAGALNDLCIKVNNWDLFFSPSEDNFTNDL
    NKGEEITSDTNIEAAEENISLDLIQQYYLTFNFDNEPENISIENLSSDIIGQLELMPNIE
    RFPNGKKYELDKYTMFHYLRAQEFEHGKSRIALTNSVNEALLNPSRVYTFFSSDYVKKVN
    KATEAAMFLGWVEQLVYDFTDETSEVSTTDKIADITIIIPYIGPALNIGNMLYKDDFVGA
    LIFSGAVILLEFIPEIAIPVLGTFALVSYIANKVLTVQTIDNALSKRNEKWDEVYKYIVT
    NWLAKVNTQIDLIRKKMKEALENQAEATKAIINYQYNQYTEEEKNNINFNIDDLSSKLNE
    SINKAMININKFLNQCSVSYLMNSMIPYGVKRLEDFDASLKDALLKYIYDNRGTLIGQVD
    RLKDKVNNTLSTDIPFQLSKYVDNQRLLSTFTEYIKNIINTSILNLRYESNHLIDLSRYA
    SKINIGSKVNFDPIDKNQIQLFNLESSKIEVILKNAIVYNSMYENFSTSFWIRIPKYFNS
    ISLNNEYTIINCMENNSGWKVSLNYGEIIWTLQDTQEIKQRVVFKYSQMINISDYINRWI
    FVTITNNRLNNSKIYINGRLIDQKPISNLGNIHASNNIMFKLDGCRDTHRYIWIKYFNLF
    DKELNEKEIKDLYDNQSNSGILKDFWGDYLQYDKPYYMLNLYDPNKYVDVNNVGIRGYMY
    LKGPRGSVMTTNIYLNSSLYRGTKFIIKKYASGNKDNIVRNNDRVYINVVVKNKEYRLAT
    NASQAGVEKILSALEIPDVGNLSQVVVMKSKNDQGITNKCKMNLQDNNGNDIGFIGFHQF
    NNIAKLVASNWYNRQIERSSRTLGCSWEFIPVDDGWGERPLASPAAPAPASPAAPAPSAP
    AASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAP
    AASPAAPAPASPAAPAPSAPAGDLVPRGSANSSSVDKLWSHPQFEK

    SEQ ID NO 3: recombinant PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS100 including His.tag)
  • MGSSHHHHHHGSLVPRSSSASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAA
    SPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAA
    PFVNKQFNYKDPVNGVDIAYIKIPNAGQMQPVKAFKIHNKIWVIPERDTFTNPEEGDLNP
    PPEAKQVPVSYYDSTYLSTDNEKDNYLKGVTKLFERIYSTDLGRMLLTSIVRGIPFWGGS
    TIDTELKVIDTNCINVIQPDGSYRSEELNLVIIGPSADIIQFECKSFIHEVLNLTRNGYG
    STQYIRFSPDFTFGFEESLEVDTNPLLGAGKFATDPAVRLAHELIHAGHRLYGIAINMNR
    VFKVNTNAYYEMTGLEVSFEELRTFGGHDAKFIDSLQENEFRLYYYNKFKDIASTLNKAK
    SIVGTTASLQYMKNVFKEKYLLSEDTSGKFSVDKLKFDKLYKMLTEIYTEDNFVKFFKVL
    NRKTYLNFDKAVFKINIVPKVNYTIYDGFNLSNTNLAANFNGQNTEINNMNFTKLKNFTG
    LFEFYKLLCVRGIITSKAGAGKSLVPRGSAGAGALNDLCIKVNNWDLFFSPSEDNFTNDL
    NKGEEITSDTNIEAAEENISLDLIQQYYLTFNFDNEPENISIENLSSDIIGQLELMPNIE
    RFPNGKKYELDKYTMFHYLRAQEFEHGKSRIALTNSVNEALLNPSRVYTFFSSDYVKKVN
    KATEAAMFLGWVEQLVYDFTDETSEVSTTDKIADITIIIPYIGPALNIGNMLYKDDFVGA
    LIFSGAVILLEFIPEIAIPVLGTFALVSYIANKVLTVQTIDNALSKRNEKWDEVYKYIVT
    NWLAKVNTQIDLIRKKMKEALENQAEATKAIINYQYNQYTEEEKNNINFNIDDLSSKLNE
    SINKAMININKFLNQCSVSYLMNSMIPYGVKRLEDFDASLKDALLKYIYDNRGTLIGQVD
    RLKDKVNNTLSTDIPFQLSKYVDNQRLLSTFTEYIKNIINTSILNLRYESNHLIDLSRYA
    SKINIGSKVNFDPIDKNQIQLFNLESSKIEVILKNAIVYNSMYENFSTSFWIRIPKYFNS
    ISLNNEYTIINCMENNSGWKVSLNYGEIIWTLQDTQEIKQRVVFKYSQMINISDYINRWI
    FVTITNNRLNNSKIYINGRLIDQKPISNLGNIHASNNIMFKLDGCRDTHRYIWIKYFNLF
    DKELNEKEIKDLYDNQSNSGILKDFWGDYLQYDKPYYMLNLYDPNKYVDVNNVGIRGYMY
    LKGPRGSVMTTNIYLNSSLYRGTKFIIKKYASGNKDNIVRNNDRVYINVVVKNKEYRLAT
    NASQAGVEKILSALEIPDVGNLSQVVVMKSKNDQGITNKCKMNLQDNNGNDIGFIGFHQF
    NNIAKLVASNWYNRQIERSSRTLGCSWEFIPVDDGWGERPLASPAAPAPASPAAPAPSAP
    AASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAPAASPAAPAPASPAAPAPSAP
    AASPAAPAPASPAAPAPSAPAGDLVPRGSANSSSVDKLWSHPQFEK

    SEQ ID NO 4: recombinant PAS100-BoNT/A (D102F T109R K340M I348L N353M K356R)-PAS100 including His.tag (nucleic acid sequence)
  • ATGGGTAGCAGCCATCATCATCACCATCATGGTAGCCTGGTTCCGCGTAGCTCTTCTGCA
    AGTCCGGCAGCACCGGCACCGGCTTCACCAGCTGCACCAGCACCTAGCGCACCGGCAGCA
    TCTCCAGCAGCCCCTGCACCGGCAAGCCCTGCAGCTCCAGCACCGTCAGCACCAGCAGCA
    AGCCCAGCTGCTCCTGCTCCAGCGAGCCCAGCAGCGCCAGCTCCTAGTGCCCCTGCTGCC
    TCTCCTGCTGCTCCGGCACCAGCAAGTCCTGCTGCGCCTGCACCGAGTGCTCCGGCTGCT
    AGTCCTGCCGCACCAGCTCCGGCTAGTCCAGCTGCTCCAGCCCCTTCAGCCCCTGCAGCA
    CCATTTGTGAACAAGCAGTTTAACTATAAGGACCCGGTGAACGGTGTGGATATCGCGTAT
    ATCAAAATCCCGAATGCGGGCCAGATGCAACCAGTCAAGGCGTTCAAGATTCATAACAAG
    ATTTGGGTTATTCCGGAACGTGATACCTTCACCAATCCGGAAGAAGGCGATTTAAATCCG
    CCGCCAGAAGCCAAACAAGTGCCGGTGAGCTACTATGATAGCACGTATCTTAGCACCGAT
    AATGAAAAAGACAATTACCTGAAGGGCGTGACCAAGTTGTTCGAGCGCATCTACAGTACC
    TTCTTAGGCCGCATGTTGTTGCGTAGCATCGTTCGCGGTATCCCGTTCTGGGGCGGCTCG
    ACCATTGATACCGAGTTGAAAGTCATTGACACGAACTGTATCAATGTTATCCAACCGGAC
    GGCAGTTATCGCAGCGAGGAGTTAAATTTGGTCATCATCGGTCCAAGCGCAGATATTATT
    CAGTTCGAATGCAAGAGCTTCGGCCATGAGGTCTTGAATTTGACGCGCAACGGTTACGGC
    AGCACCCAATACATCCGCTTTAGCCCGGATTTCACCTTTGGCTTCGAGGAGAGCTTGGAG
    GTGGACACCAACCCGCTGTTAGGTGCCGGCAAATTCGCAACCGACCCGGCAGTGACGTTG
    GCGCACGAATTGATTCATGCGGGTCACCGCTTATACGGTATCGCGATCAATCCGAATCGC
    GTCTTTAAAGTCAATACCAACGCGTACTACGAAATGAGCGGCTTAGAGGTTAGCTTTGAA
    GAATTACGCACCTTCGGTGGCCACGACGCCAAGTTCATCGACAGCCTGCAGGAAAATGAG
    TTCCGCTTGTACTATTACAATAAATTCAAGGACATCGCGAGCACCTTAAATAAAGCAAAG
    AGCATTGTGGGCACCACCGCAAGCTTGCAGTACATGAAGAACGTATTTAAGGAAAAATAT
    TTGTTGTCGGAGGATACCAGCGGGAAATTCAGCGTCGATAAGCTGAAATTCGACATGTTG
    TATAAAATGCTGACCGAGCTGTACACCGAGGATATGTTCGTCCGTTTTTTTAAGGTGTTA
    AATCGTAAGACCTATTTAAACTTTGATAAAGCGGTGTTTAAAATTAATATCGTGCCGAAG
    GTGAATTACACCATCTACGATGGTTTCAATTTACGCAACACGAATCTGGCGGCGAATTTT
    AATGGCCAAAACACCGAAATTAACAACATGAACTTTACGAAGTTAAAGAATTTCACGGGC
    TTATTCGAATTCTACAAGTTATTATGCGTGCGCGGCATCATTACCAGCAAGGCAGGTGCG
    GGCAAGTCCTTGGTTCCGCGTGGCAGCGCCGGCGCCGGCGCGCTCAATGATCTGTGTATT
    AAAGTCAATAACTGGGACCTGTTCTTCAGCCCGAGCGAGGATAACTTTACCAACGACTTA
    AACAAAGGCGAGGAGATCACGAGCGATACGAACATCGAGGCGGCGGAGGAAAATATTAGC
    CTGGACCTCATTCAGCAGTACTATCTGACGTTCAATTTTGACAATGAGCCGGAGAACATC
    AGCATTGAAAATCTCAGCAGCGACATCATCGGTCAGTTGGAACTGATGCCGAACATTGAA
    CGCTTTCCGAACGGCAAAAAATATGAACTGGACAAGTATACCATGTTCCATTACTTACGC
    GCACAGGAATTTGAGCACGGCAAGAGCCGCATTGCGCTGACCAATAGCGTTAACGAGGCC
    TTGTTAAATCCGAGCCGTGTCTACACGTTCTTCAGCAGCGATTATGTCAAAAAAGTGAAC
    AAGGCGACCGAAGCCGCGATGTTTTTGGGCTGGGTCGAGCAATTGGTTTACGATTTTACC
    GACGAAACCAGCGAGGTGAGCACGACCGACAAAATTGCAGATATCACCATCATCATTCCG
    TACATCGGTCCGGCGCTCAATATCGGCAATATGTTATACAAGGACGACTTTGTGGGCGCG
    CTGATCTTTAGCGGCGCGGTTATCTTATTAGAATTCATCCCGGAGATCGCAATCCCGGTC
    TTGGGCACCTTTGCGTTGGTGAGCTATATCGCGAATAAAGTGCTCACGGTCCAAACCATC
    GATAACGCGCTCAGCAAGCGTAATGAGAAATGGGACGAGGTTTATAAGTATATCGTGACC
    AACTGGTTAGCAAAAGTCAATACGCAGATCGATCTCATCCGCAAAAAAATGAAAGAAGCC
    TTGGAAAATCAAGCGGAGGCAACCAAAGCCATCATTAATTACCAGTATAACCAATATACC
    GAAGAAGAAAAAAACAATATCAACTTCAATATCGATGATTTGAGCAGCAAACTGAACGAG
    AGCATTAACAAAGCGATGATTAACATCAACAAGTTCTTGAATCAATGCAGCGTGAGCTAT
    CTCATGAACAGCATGATCCCGTATGGCGTCAAACGCTTGGAAGATTTTGACGCCAGCCTG
    AAAGATGCGCTCCTCAAGTATATTTATGACAACCGCGGCACCCTCATTGGCCAGGTGGAC
    CGCTTGAAGGATAAAGTGAACAATACGCTCAGCACGGATATCCCGTTCCAGCTGAGCAAG
    TACGTCGACAACCAGCGCTTACTGAGCACCTTTACCGAGTATATCAAGAACATCATTAAT
    ACCAGCATCCTCAACTTGCGCTATGAGAGCAATCACCTGATCGACCTCAGCCGCTACGCC
    AGCAAGATCAACATCGGCAGCAAGGTCAATTTCGACCCGATCGATAAGAATCAGATCCAA
    TTGTTTAACCTGGAAAGCAGCAAGATCGAGGTTATCTTGAAGAACGCGATTGTGTACAAC
    AGCATGTACGAGAACTTTAGCACGAGCTTCTGGATTCGTATCCCGAAGTATTTCAATAGC
    ATTAGCCTGAATAACGAATATACCATTATCAACTGCATGGAAAATAATAGCGGCTGGAAG
    GTGAGCTTAAATTACGGCGAGATCATTTGGACCTTACAGGATACCCAAGAAATCAAACAG
    CGCGTCGTCTTTAAGTATAGCCAGATGATCAACATCAGCGATTACATCAACCGCTGGATC
    TTCGTGACCATCACCAATAATCGCTTGAATAATAGCAAGATTTACATCAATGGTCGCTTG
    ATTGATCAAAAACCGATCAGCAATCTCGGTAATATCCATGCCAGCAATAACATCATGTTT
    AAGTTAGACGGTTGCCGCGATACCCACCGCTATATCTGGATCAAGTATTTTAACTTATTT
    GATAAGGAACTCAACGAAAAGGAAATTAAAGACTTATATGACAATCAGAGCAATAGCGGC
    ATCCTGAAGGATTTCTGGGGCGACTACCTGCAGTACGATAAGCCGTACTATATGTTGAAC
    TTGTATGACCCGAACAAATATGTCGATGTGAACAATGTGGGTATTCGTGGCTATATGTAC
    TTAAAGGGCCCGCGTGGTAGCGTGATGACCACGAATATTTACTTAAACAGCAGCTTATAC
    CGCGGCACGAAGTTTATTATCAAGAAGTATGCCAGCGGCAACAAGGACAATATCGTCCGC
    AACAACGACCGTGTGTATATTAACGTGGTGGTGAAGAATAAAGAGTACCGCTTGGCCACG
    AATGCGAGCCAGGCGGGCGTGGAAAAAATCTTGAGCGCGTTGGAGATCCCGGACGTCGGC
    AACCTCAGCCAGGTTGTGGTGATGAAGTCTAAAAACGACCAGGGCATCACGAACAAGTGC
    AAAATGAATTTGCAAGATAACAACGGCAACGACATCGGCTTTATTGGTTTTCACCAGTTC
    AATAACATCGCCAAACTCGTGGCCAGCAATTGGTATAACCGCCAAATTGAACGCAGCAGC
    CGCACGCTCGGCTGTAGCTGGGAGTTCATCCCGGTGGACGATGGCTGGGGCGAGCGCCCG
    CTCGCAAGTCCGGCAGCACCGGCACCGGCTTCACCAGCTGCACCAGCACCTAGCGCACCG
    GCAGCATCTCCAGCAGCCCCTGCACCGGCAAGCCCTGCAGCTCCAGCACCGTCAGCACCA
    GCAGCAAGCCCAGCTGCTCCTGCTCCAGCGAGCCCAGCAGCGCCAGCTCCTAGTGCCCCT
    GCTGCCTCTCCTGCTGCTCCGGCACCAGCAAGTCCTGCTGCGCCTGCACCGAGTGCTCCG
    GCTGCTAGTCCTGCCGCACCAGCTCCGGCTAGTCCAGCTGCTCCAGCCCCTTCAGCCCCT
    GCAGGAGATCTGGTGCCACGCGGTTCCGCGAATTCGAGCTCCGTCGACAAGCTTTGGAGC
    CACCCGCAGTTCGAAAAATAA

    SEQ ID NO 5: recombinant PAS100-BoNT/A (G169I, T220R, P239M, S254T)-PAS100 including His.tag) (nucleic acid sequence)
  • ATGGGTAGCAGCCATCATCATCACCATCATGGTAGCCTGGTTCCGCGTAGCTCTTCTGCA
    AGTCCGGCAGCACCGGCACCGGCTTCACCAGCTGCACCAGCACCTAGCGCACCGGCAGCA
    TCTCCAGCAGCCCCTGCACCGGCAAGCCCTGCAGCTCCAGCACCGTCAGCACCAGCAGCA
    AGCCCAGCTGCTCCTGCTCCAGCGAGCCCAGCAGCGCCAGCTCCTAGTGCCCCTGCTGCC
    TCTCCTGCTGCTCCGGCACCAGCAAGTCCTGCTGCGCCTGCACCGAGTGCTCCGGCTGCT
    AGTCCTGCCGCACCAGCTCCGGCTAGTCCAGCTGCTCCAGCCCCTTCAGCCCCTGCAGCA
    CCATTTGTGAACAAGCAGTTTAACTATAAGGACCCGGTGAACGGTGTGGATATCGCGTAT
    ATCAAAATCCCGAATGCGGGCCAGATGCAACCAGTCAAGGCGTTCAAGATTCATAACAAG
    ATTTGGGTTATTCCGGAACGTGATACCTTCACCAATCCGGAAGAAGGCGATTTAAATCCG
    CCGCCAGAAGCCAAACAAGTGCCGGTGAGCTACTATGATAGCACGTATCTTAGCACCGAT
    AATGAAAAAGACAATTACCTGAAGGGCGTGACCAAGTTGTTCGAGCGCATCTACAGTACC
    GACTTAGGCCGCATGTTGTTGACGAGCATCGTTCGCGGTATCCCGTTCTGGGGCGGCTCG
    ACCATTGATACCGAGTTGAAAGTCATTGACACGAACTGTATCAATGTTATCCAACCGGAC
    GGCAGTTATCGCAGCGAGGAGTTAAATTTGGTCATCATCGGTCCAAGCGCAGATATTATT
    CAGTTCGAATGCAAGAGCTTCATCCATGAGGTCTTGAATTTGACGCGCAACGGTTACGGC
    AGCACCCAATACATCCGCTTTAGCCCGGATTTCACCTTTGGCTTCGAGGAGAGCTTGGAG
    GTGGACACCAACCCGCTGTTAGGTGCCGGCAAATTCGCAACCGACCCGGCAGTGCGCTTG
    GCGCACGAATTGATTCATGCGGGTCACCGCTTATACGGTATCGCGATCAATATGAATCGC
    GTCTTTAAAGTCAATACCAACGCGTACTACGAAATGACCGGCTTAGAGGTTAGCTTTGAA
    GAATTACGCACCTTCGGTGGCCACGACGCCAAGTTCATCGACAGCCTGCAGGAAAATGAG
    TTCCGCTTGTACTATTACAATAAATTCAAGGACATCGCGAGCACCTTAAATAAAGCAAAG
    AGCATTGTGGGCACCACCGCAAGCTTGCAGTACATGAAGAACGTATTTAAGGAAAAATAT
    TTGTTGTCGGAGGATACCAGCGGGAAATTCAGCGTCGATAAGCTGAAATTCGACAAATTG
    TATAAAATGCTGACCGAGATTTACACCGAGGATAACTTCGTCAAGTTTTTTAAGGTGTTA
    AATCGTAAGACCTATTTAAACTTTGATAAAGCGGTGTTTAAAATTAATATCGTGCCGAAG
    GTGAATTACACCATCTACGATGGTTTCAACTTAAGCAACACGAATCTGGCGGCGAATTTT
    AATGGCCAAAACACCGAAATTAACAACATGAACTTTACGAAGTTAAAGAATTTCACGGGC
    TTATTCGAATTCTACAAGTTATTATGCGTGCGCGGCATCATTACCAGCAAGGCAGGTGCG
    GGCAAGTCCTTGGTTCCGCGTGGCAGCGCCGGCGCCGGCGCGCTCAATGATCTGTGTATT
    AAAGTCAATAACTGGGACCTGTTCTTCAGCCCGAGCGAGGATAACTTTACCAACGACTTA
    AACAAAGGCGAGGAGATCACGAGCGATACGAACATCGAGGCGGCGGAGGAAAATATTAGC
    CTGGACCTCATTCAGCAGTACTATCTGACGTTCAATTTTGACAATGAGCCGGAGAACATC
    AGCATTGAAAATCTCAGCAGCGACATCATCGGTCAGTTGGAACTGATGCCGAACATTGAA
    CGCTTTCCGAACGGCAAAAAATATGAACTGGACAAGTATACCATGTTCCATTACTTACGC
    GCACAGGAATTTGAGCACGGCAAGAGCCGCATTGCGCTGACCAATAGCGTTAACGAGGCC
    TTGTTAAATCCGAGCCGTGTCTACACGTTCTTCAGCAGCGATTATGTCAAAAAAGTGAAC
    AAGGCGACCGAAGCCGCGATGTTTTTGGGCTGGGTCGAGCAATTGGTTTACGATTTTACC
    GACGAAACCAGCGAGGTGAGCACGACCGACAAAATTGCAGATATCACCATCATCATTCCG
    TACATCGGTCCGGCGCTCAATATCGGCAATATGTTATACAAGGACGACTTTGTGGGCGCG
    CTGATCTTTAGCGGCGCGGTTATCTTATTAGAATTCATCCCGGAGATCGCAATCCCGGTC
    TTGGGCACCTTTGCGTTGGTGAGCTATATCGCGAATAAAGTGCTCACGGTCCAAACCATC
    GATAACGCGCTCAGCAAGCGTAATGAGAAATGGGACGAGGTTTATAAGTATATCGTGACC
    AACTGGTTAGCAAAAGTCAATACGCAGATCGATCTCATCCGCAAAAAAATGAAAGAAGCC
    TTGGAAAATCAAGCGGAGGCAACCAAAGCCATCATTAATTACCAGTATAACCAATATACC
    GAAGAAGAAAAAAACAATATCAACTTCAATATCGATGATTTGAGCAGCAAACTGAACGAG
    AGCATTAACAAAGCGATGATTAACATCAACAAGTTCTTGAATCAATGCAGCGTGAGCTAT
    CTCATGAACAGCATGATCCCGTATGGCGTCAAACGCTTGGAAGATTTTGACGCCAGCCTG
    AAAGATGCGCTCCTCAAGTATATTTATGACAACCGCGGCACCCTCATTGGCCAGGTGGAC
    CGCTTGAAGGATAAAGTGAACAATACGCTCAGCACGGATATCCCGTTCCAGCTGAGCAAG
    TACGTCGACAACCAGCGCTTACTGAGCACCTTTACCGAGTATATCAAGAACATCATTAAT
    ACCAGCATCCTCAACTTGCGCTATGAGAGCAATCACCTGATCGACCTCAGCCGCTACGCC
    AGCAAGATCAACATCGGCAGCAAGGTCAATTTCGACCCGATCGATAAGAATCAGATCCAA
    TTGTTTAACCTGGAAAGCAGCAAGATCGAGGTTATCTTGAAGAACGCGATTGTGTACAAC
    AGCATGTACGAGAACTTTAGCACGAGCTTCTGGATTCGTATCCCGAAGTATTTCAATAGC
    ATTAGCCTGAATAACGAATATACCATTATCAACTGCATGGAAAATAATAGCGGCTGGAAG
    GTGAGCTTAAATTACGGCGAGATCATTTGGACCTTACAGGATACCCAAGAAATCAAACAG
    CGCGTCGTCTTTAAGTATAGCCAGATGATCAACATCAGCGATTACATCAACCGCTGGATC
    TTCGTGACCATCACCAATAATCGCTTGAATAATAGCAAGATTTACATCAATGGTCGCTTG
    ATTGATCAAAAACCGATCAGCAATCTCGGTAATATCCATGCCAGCAATAACATCATGTTT
    AAGTTAGACGGTTGCCGCGATACCCACCGCTATATCTGGATCAAGTATTTTAACTTATTT
    GATAAGGAACTCAACGAAAAGGAAATTAAAGACTTATATGACAATCAGAGCAATAGCGGC
    ATCCTGAAGGATTTCTGGGGCGACTACCTGCAGTACGATAAGCCGTACTATATGTTGAAC
    TTGTATGACCCGAACAAATATGTCGATGTGAACAATGTGGGTATTCGTGGCTATATGTAC
    TTAAAGGGCCCGCGTGGTAGCGTGATGACCACGAATATTTACTTAAACAGCAGCTTATAC
    CGCGGCACGAAGTTTATTATCAAGAAGTATGCCAGCGGCAACAAGGACAATATCGTCCGC
    AACAACGACCGTGTGTATATTAACGTGGTGGTGAAGAATAAAGAGTACCGCTTGGCCACG
    AATGCGAGCCAGGCGGGCGTGGAAAAAATCTTGAGCGCGTTGGAGATCCCGGACGTCGGC
    AACCTCAGCCAGGTTGTGGTGATGAAGTCTAAAAACGACCAGGGCATCACGAACAAGTGC
    AAAATGAATTTGCAAGATAACAACGGCAACGACATCGGCTTTATTGGTTTTCACCAGTTC
    AATAACATCGCCAAACTCGTGGCCAGCAATTGGTATAACCGCCAAATTGAACGCAGCAGC
    CGCACGCTCGGCTGTAGCTGGGAGTTCATCCCGGTGGACGATGGCTGGGGCGAGCGCCCG
    CTCGCAAGTCCGGCAGCACCGGCACCGGCTTCACCAGCTGCACCAGCACCTAGCGCACCG
    GCAGCATCTCCAGCAGCCCCTGCACCGGCAAGCCCTGCAGCTCCAGCACCGTCAGCACCA
    GCAGCAAGCCCAGCTGCTCCTGCTCCAGCGAGCCCAGCAGCGCCAGCTCCTAGTGCCCCT
    GCTGCCTCTCCTGCTGCTCCGGCACCAGCAAGTCCTGCTGCGCCTGCACCGAGTGCTCCG
    GCTGCTAGTCCTGCCGCACCAGCTCCGGCTAGTCCAGCTGCTCCAGCCCCTTCAGCCCCT
    GCAGGAGATCTGGTGCCACGCGGTTCCGCGAATTCGAGCTCCGTCGACAAGCTTTGGAGC
    CACCCGCAGTTCGAAAAATAA

Claims (20)

1.-15. (canceled)
16. A method of treating a disease requiring improved chemodenervation, said method comprising the step of administering a recombinant neurotoxin to a patient in need thereof, said recombinant neurotoxin comprising:
at least two domains wherein each domain comprises an amino acid sequence comprising at least 50 amino acid residues, wherein said amino acid sequence comprises at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, wherein the neurotoxin further comprises at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin,
wherein the said at least one amino acid modification is located at at least one position selected from D102, T109, K340, I348, N353, and K356 of the alpha-exosite, or at at least one position selected from G169, T220, P239, and S254 of the beta-exosite, and
wherein the recombinant neurotoxin causes both (i) an increased duration of effect relative to a wildtype botulinum neurotoxin serotype A and (ii) an increased specific biological activity relative to a botulinum neurotoxin serotype A comprising two domains consisting of proline, alanine and serine residues without said modifications in the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
17. The method of claim 16, wherein six amino acid modifications are located at the alpha-exosite at positions D102, T109, K340, I348, N353, K356.
18. The method of claim 17, wherein the six amino acids are substituted as follows: D102F, T109R, K340M, I348L, N353M, K356R.
19. The method of claim 16, wherein four amino acid modifications are located at the beta-exosite at positions G169, T220, P239, S254
20. The method of claim 19, wherein the four amino acids are substituted as follows: G169I, T220R, P239M, S254T.
21. The method of claim 16, wherein the recombinant neurotoxin comprises two domains wherein each domain comprises an amino acid sequence consisting of between 70 and 260 amino acid residues.
22. The method of claim 16, wherein the recombinant neurotoxin is administered with a solvent or excipient.
23. The method of claim 16, wherein the recombinant neurotoxin is administered with one or more pharmaceutically acceptable carriers.
24. The method of claim 16, wherein the recombinant neurotoxin is administered through an injection.
25. The method of claim 16, wherein the disease is selected from the group consisting of: cervical dystonia (spasmodic torticollis), blepharospasm, severe primary axillary hyperhidrosis, achalasia, lower back pain, benign prostate hypertrophy, chronic focal painful neuropathies, migraines and other headache disorders.
26. A method of using a recombinant neurotoxin for cosmetic treatment, said method comprising the step of administering the recombinant neurotoxin to a subject, said recombinant neurotoxin comprising:
at least two domains wherein each domain comprises an amino acid sequence comprising at least 50 amino acid residues, wherein said amino acid sequence comprises at least one proline, at least one alanine and at least one additional amino acid residue, selected from the group consisting of serine, threonine, tyrosine and glutamine, wherein the neurotoxin further comprises at least one amino acid modification which is located at the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin,
wherein the said at least one amino acid modification is located at at least one position selected from D102, T109, K340, I348, N353, and K356 of the alpha-exosite, or at at least one position selected from G169, T220, P239, and S254 of the beta-exosite, and
wherein the recombinant neurotoxin causes both (i) an increased duration of effect relative to a wildtype botulinum neurotoxin serotype A and (ii) an increased specific biological activity relative to a botulinum neurotoxin serotype A comprising two domains consisting of proline, alanine and serine residues without said modifications in the alpha-exosite and/or at the beta-exosite of the light chain of the neurotoxin.
27. The method of claim 26, wherein six amino acid modifications are located at the alpha-exosite at positions D102, T109, K340, I348, N353, K356.
28. The method of claim 27, wherein the six amino acids are substituted as follows: D102F, T109R, K340M, I348L, N353M, K356R.
29. The method of claim 26, wherein four amino acid modifications are located at the beta-exosite at positions G169, T220, P239, S254
30. The method of claim 29, wherein the four amino acids are substituted as follows: G169I, T220R, P239M, S254T.
31. The method of claim 26, wherein the recombinant neurotoxin comprises two domains wherein each domain comprises an amino acid sequence consisting of between 70 and 260 amino acid residues.
32. The method of claim 26, wherein the recombinant neurotoxin is administered with a solvent or excipient.
33. The method of claim 26, wherein the recombinant neurotoxin is administered with one or more pharmaceutically acceptable carriers.
34. The method of claim 26, wherein cosmetic treatment is selected from the group consisting of: treatment of wrinkles, treatment of crow's feet, treatment of glabella frown lines, reduction of masseter muscles, reduction of calves, and removing of facial asymmetries.
US17/482,953 2017-07-06 2021-09-23 Novel recombinant botulinum neurotoxins with increased duration of effect Pending US20220010294A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/482,953 US20220010294A1 (en) 2017-07-06 2021-09-23 Novel recombinant botulinum neurotoxins with increased duration of effect

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
PCT/EP2017/066891 WO2019007509A1 (en) 2017-07-06 2017-07-06 Novel recombinant botulinum neurotoxins with increased duration of effect
US201916498257A 2019-09-26 2019-09-26
US17/482,953 US20220010294A1 (en) 2017-07-06 2021-09-23 Novel recombinant botulinum neurotoxins with increased duration of effect

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US16/498,257 Division US11155802B2 (en) 2017-07-06 2017-07-06 Recombinant botulinum neurotoxins with increased duration of effect
PCT/EP2017/066891 Division WO2019007509A1 (en) 2017-07-06 2017-07-06 Novel recombinant botulinum neurotoxins with increased duration of effect

Publications (1)

Publication Number Publication Date
US20220010294A1 true US20220010294A1 (en) 2022-01-13

Family

ID=59384133

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/498,257 Active US11155802B2 (en) 2017-07-06 2017-07-06 Recombinant botulinum neurotoxins with increased duration of effect
US17/482,953 Pending US20220010294A1 (en) 2017-07-06 2021-09-23 Novel recombinant botulinum neurotoxins with increased duration of effect

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/498,257 Active US11155802B2 (en) 2017-07-06 2017-07-06 Recombinant botulinum neurotoxins with increased duration of effect

Country Status (4)

Country Link
US (2) US11155802B2 (en)
EP (1) EP3649143B1 (en)
ES (1) ES2930237T3 (en)
WO (1) WO2019007509A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10441640B2 (en) * 2014-08-12 2019-10-15 Biomadison, Inc. Botulinum neurotoxins with modified light chain specificity and methods for producing same
TW201718627A (en) * 2015-06-11 2017-06-01 梅茲製藥有限兩合公司 Recombinant clostridial neurotoxin, a use thereof, and a method for generating the same, a pharmaceutical composition comprising the same and a precursor corresponding to the same, a nucleic acid sequence encoding the precursor and a method for obtaining
WO2018233813A1 (en) 2017-06-20 2018-12-27 Merz Pharma Gmbh & Co. Kgaa Novel recombinant botulinum toxin with increased duration of effect
US11155802B2 (en) * 2017-07-06 2021-10-26 Merz Pharma Gmbh & Co. Kgaa Recombinant botulinum neurotoxins with increased duration of effect

Family Cites Families (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6203794B1 (en) 1994-05-31 2001-03-20 Allergan Sales, Inc. Modification of clostridial toxins for use as transport proteins
AU4746096A (en) 1995-06-06 1996-12-24 Wisconsin Alumni Research Foundation Analogs of botulinum toxin and pharmaceutical compositions o f botulinum toxin
US6214602B1 (en) 1998-08-28 2001-04-10 Promega Corporation Host cells for expression of clostridial toxins and proteins
KR100876060B1 (en) 1999-08-25 2008-12-26 알러간, 인코포레이티드 Activatable Recombinant Neurotoxins
US6903187B1 (en) 2000-07-21 2005-06-07 Allergan, Inc. Leucine-based motif and clostridial neurotoxins
US7491799B2 (en) 2000-07-21 2009-02-17 Allergan, Inc. Modified botulinum neurotoxins
US20020127247A1 (en) 2000-11-17 2002-09-12 Allergen Sales, Inc. Modified clostridial neurotoxins with altered biological persistence
DE10333317A1 (en) 2003-07-22 2005-02-17 Biotecon Therapeutics Gmbh Formulation for protein medicines without the addition of human serum albumin (HSA)
DK1755661T3 (en) 2004-05-12 2014-06-16 Brigham & Womens Hospital GELSOLIN FOR USE FOR TREATMENT OF INFECTIONS
KR100852822B1 (en) 2004-07-26 2008-08-18 메르츠 파마 게엠베하 운트 코. 카가아 Therapeutic composition with a botulinum neurotoxin
AU2005271372B2 (en) 2004-08-04 2012-05-03 Allergan, Inc. Optimizing expression of active botulinum toxin type A
DE102005002978B4 (en) 2005-01-21 2013-04-25 Merz Pharma Gmbh & Co. Kgaa Recombinant expression of proteins in a disulfide-bonded, two-chain form
EP2131857B1 (en) 2007-03-22 2015-07-29 The Regents of the University of Colorado, a body corporate Method of preparing an immunologically-active adjuvant-bound dried vaccine composition
ATE502114T1 (en) 2007-06-21 2011-04-15 Univ Muenchen Tech BIOLOGICALLY ACTIVE PROTEINS WITH INCREASED IN-VIVO AND/OR IN-VITRO STABILITY
US9044477B2 (en) 2007-12-12 2015-06-02 Allergan, Inc. Botulinum toxin formulation
US9161970B2 (en) 2007-12-12 2015-10-20 Allergan, Inc. Dermal filler
BR122012027456A2 (en) 2008-03-14 2015-07-14 Allergan Inc Pharmaceutical composition comprising botulinum neurotoxin serotype a
BRPI0916964A2 (en) 2008-08-29 2015-11-24 Merz Pharma Gmbh & Co Kgaa polypeptide, antibody, nucleic acid, vector, host cell, method for making a polypeptide, composition and use of the polypeptide
AU2009288118B2 (en) 2008-09-02 2014-12-11 Allergan, Inc. Threads of hyaluronic acid and/or derivatives thereof, methods of making thereof and uses thereof
AU2010204445B2 (en) 2009-01-07 2015-09-03 2405871 Ontario Inc. Treatment of soft tissue injury using hyaluronic acid and botulinum toxin
CN116925238A (en) 2009-02-03 2023-10-24 阿穆尼克斯制药公司 Extended recombinant polypeptides and compositions comprising the same
WO2010136585A2 (en) 2009-05-29 2010-12-02 Galderma Research & Development Combination of adrenergic receptor agonist -1 or -2, preferably brimonidine with fillers, preferablyhyaluronic acid
IN2012DN00733A (en) 2009-07-02 2015-06-19 Merz Pharma Gmbh & Co Kgaa
WO2011109130A1 (en) 2010-03-01 2011-09-09 Tautona Group Lp Threads of hyaluronic acid and methods of use thereof
US8557961B2 (en) 2010-04-02 2013-10-15 Amunix Operating Inc. Alpha 1-antitrypsin compositions and methods of making and using same
WO2011123813A2 (en) 2010-04-02 2011-10-06 Amunix Operating Inc. Binding fusion proteins, binding fusion protein-drug conjugates, xten-drug conjugates and methods of making and using same
EA024755B1 (en) 2010-05-21 2016-10-31 ИксЭль-ПРОТЕИН ГМБХ Biosynthetic proline/alanine random coil polypeptides and their uses
FR2966348B1 (en) 2010-10-22 2012-11-09 Galderma Res & Dev COMPOSITIONS COMPRISING A WRINKLE FILLING MEDIUM AND A CHEMICALLY MODIFIED TETRACYCLINE
KR101640694B1 (en) 2011-09-29 2016-07-18 셀스냅, 엘엘씨 Compositions and methods for toxigenicity testing
RU2646110C2 (en) * 2011-11-09 2018-03-01 Мерц Фарма Гмбх Унд Ко. Кгаа Neurotoxins showing shortened biological activity
AU2012334076A1 (en) 2011-11-09 2014-05-15 Merz Pharma Gmbh & Co. Kgaa Modified neurotoxins with poly-Glycine and uses thereof
JP2014534265A (en) 2011-11-28 2014-12-18 フェーズバイオ ファーマシューティカルズ,インコーポレイテッド Therapeutic drugs containing insulin amino acid sequence
AR089797A1 (en) 2012-01-27 2014-09-17 Merck Sharp & Dohme VACCINES AGAINST CLOSTRIDUM DIFFICILE THAT INCLUDE RECOMBINANT TOXINS
GB201219602D0 (en) 2012-10-31 2012-12-12 Syntaxin Ltd Recombinant clostridium botulinum neurotoxins
US20150322118A1 (en) 2012-12-05 2015-11-12 Merz Pharma Gmbh & Co. Kgaa Recombinant clostridial neurotoxins with enhanced membrane localization
CA2899961C (en) 2013-02-14 2022-08-09 Cornell University Methods to protect against and treat multiple sclerosis
WO2014207109A1 (en) 2013-06-28 2014-12-31 Merz Pharma Gmbh & Co. Kgaa Means and methods for the determination of the biological activity of neurotoxin polypeptides in cells
EP3113792B1 (en) 2014-03-05 2018-12-05 Merz Pharma GmbH & Co. KGaA Novel recombinant clostridial neurotoxins with increased duration of effect
EP3156412B1 (en) 2014-05-29 2020-01-08 Procell Therapeutics Inc. Novel cell penetrating peptide, conjugate thereof with botulinum toxin, and use thereof
US10441640B2 (en) 2014-08-12 2019-10-15 Biomadison, Inc. Botulinum neurotoxins with modified light chain specificity and methods for producing same
WO2016073562A1 (en) 2014-11-04 2016-05-12 Synthetic Biologics, Inc. Methods and compositions for inhibiting clostridium difficile
PL3242884T3 (en) 2015-01-09 2021-08-16 Ipsen Bioinnovation Limited Cationic neurotoxins
WO2016180533A1 (en) * 2015-05-12 2016-11-17 Merz Pharma Gmbh & Co. Kgaa Novel recombinant clostridial neurotoxins with increased duration of effect
TW201718627A (en) 2015-06-11 2017-06-01 梅茲製藥有限兩合公司 Recombinant clostridial neurotoxin, a use thereof, and a method for generating the same, a pharmaceutical composition comprising the same and a precursor corresponding to the same, a nucleic acid sequence encoding the precursor and a method for obtaining
ES2929347T3 (en) * 2016-01-20 2022-11-28 Merz Pharma Gmbh & Co Kgaa Novel recombinant clostridial neurotoxins with increased duration of effect
EP3335719A1 (en) * 2016-12-14 2018-06-20 Merz Pharma GmbH & Co. KGaA Novel recombinant botulinum neurotoxins with a stabilized light chain
WO2018233813A1 (en) 2017-06-20 2018-12-27 Merz Pharma Gmbh & Co. Kgaa Novel recombinant botulinum toxin with increased duration of effect
US11155802B2 (en) * 2017-07-06 2021-10-26 Merz Pharma Gmbh & Co. Kgaa Recombinant botulinum neurotoxins with increased duration of effect
EP3700919A1 (en) 2017-10-26 2020-09-02 Merz Pharma GmbH & Co. KGaA Novel recombinant botulinum neurotoxins with increased duration of effect
WO2019101308A1 (en) * 2017-11-22 2019-05-31 Merz Pharma Gmbh & Co. Kgaa Novel recombinant botulinum toxin with increased duration of effect
CA3118412A1 (en) * 2018-11-02 2020-05-07 Institute Of Basic Medical Sciences Chinese Academy Of Medical Sciences Cell-penetrating peptide based on influenza virus m2 protein

Also Published As

Publication number Publication date
EP3649143A1 (en) 2020-05-13
US11155802B2 (en) 2021-10-26
ES2930237T3 (en) 2022-12-09
US20200131494A1 (en) 2020-04-30
WO2019007509A1 (en) 2019-01-10
EP3649143B1 (en) 2022-08-31

Similar Documents

Publication Publication Date Title
US20220010294A1 (en) Novel recombinant botulinum neurotoxins with increased duration of effect
EP3113792B1 (en) Novel recombinant clostridial neurotoxins with increased duration of effect
US11357821B2 (en) Recombinant clostridial neurotoxins with increased duration of effect
US20090018081A1 (en) Activatable clostridial toxins
EP3335719A1 (en) Novel recombinant botulinum neurotoxins with a stabilized light chain
US20150322118A1 (en) Recombinant clostridial neurotoxins with enhanced membrane localization
US11952601B2 (en) Recombinant botulinum toxin with increased duration of effect
US11078472B2 (en) Recombinant clostridial neurotoxins with increased duration of effect
US20210008156A1 (en) Novel recombinant botulinum neurotoxins with increased duration of effect
US20200354706A1 (en) Novel recombinant botulinum toxin with increased duration of effect
WO2016180533A1 (en) Novel recombinant clostridial neurotoxins with increased duration of effect
EP3312193A1 (en) Novel recombinant botulinum neurotoxins with accelerated onset of effect
EP3333179A1 (en) Novel recombinant botulinum toxin with accelarated onset of effect
EP3290437A1 (en) Novel recombinant clostridial neurotoxins with decreased duration of effect
US20150232828A1 (en) Method for the manufacturing of recombinant proteins harbouring an n-terminal lysine

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERZ PHARMA GMBH & CO. KGAA, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FREVERT, JUERGEN;HOFMANN, FRED;JURK, MARCEL;AND OTHERS;SIGNING DATES FROM 20190804 TO 20190823;REEL/FRAME:057597/0420

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

ZAAB Notice of allowance mailed

Free format text: ORIGINAL CODE: MN/=.