US20050095251A1 - Methods and compositions for the treatment of pancreatitis - Google Patents

Methods and compositions for the treatment of pancreatitis Download PDF

Info

Publication number
US20050095251A1
US20050095251A1 US11/014,795 US1479504A US2005095251A1 US 20050095251 A1 US20050095251 A1 US 20050095251A1 US 1479504 A US1479504 A US 1479504A US 2005095251 A1 US2005095251 A1 US 2005095251A1
Authority
US
United States
Prior art keywords
cck
amino acid
acid sequence
amino
polypeptide
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.)
Abandoned
Application number
US11/014,795
Inventor
Lance Steward
George Sachs
Kei Aoki
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.)
Allergan Inc
Original Assignee
Allergan Inc
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 Allergan Inc filed Critical Allergan Inc
Priority to US11/014,795 priority Critical patent/US20050095251A1/en
Publication of US20050095251A1 publication Critical patent/US20050095251A1/en
Priority to US11/845,638 priority patent/US20080025994A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6415Toxins or lectins, e.g. clostridial toxins or Pseudomonas exotoxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Definitions

  • the present invention includes methods and compositions for the treatment of acute pancreatitis.
  • the invention concerns the use of agents to reduce or prevent the secretion of pancreatic digestive enzymes within the pancreas. Such agents are targeted to pancreatic cells, and serve to prevent the exocytotic fusion of vesicles containing these enzymes with the plasma membrane.
  • the invention is also concerned with methods of treating a mammal suffering from pancreatitis through the administration of such agents.
  • Pancreatitis is a serious medical condition involving an inflammation of the pancreas.
  • the inflammation manifests itself in the release and activation of pancreatic enzymes within the organ itself, leading to autodigestion.
  • the condition can lead to death.
  • pancreas In normal mammals, the pancreas, a large gland similar in structure to the salivary gland, is responsible for the production and secretion of digestive enzymes, which digest ingested food, and bicarbonate for the neutralization of the acidic chyme produced in the stomach.
  • the pancreas contains acinar cells, responsible for enzyme production, and ductal cells, which secrete large amounts of sodium bicarbonate solution.
  • the combined secretion product is termed “pancreatic juice”; this liquid flows through the pancreatic duct past the sphincter of Oddi into the duodenum.
  • pancreatic juice The secretion of pancreatic juice is stimulated by the presence of chyme in the upper portions of the small intestine, and the precise composition of pancreatic juice appears to be influenced by the types of compounds (carbohydrate, lipid, protein, and/or nucleic acid) in the chyme.
  • compounds carbohydrate, lipid, protein, and/or nucleic acid
  • pancreatic juice includes proteases (trypsin, chymotrypsin, carboxypolypeptidase), nucleases (RNAse and DNAse), pancreatic amylase, and lipases (pancreatic lipase, cholesterol esterase and phospholipase).
  • proteases trypsin, chymotrypsin, carboxypolypeptidase
  • nucleases RNAse and DNAse
  • pancreatic amylase pancreatic amylase
  • lipases pancreatic lipase, cholesterol esterase and phospholipase
  • Trypsinogen can also be autoactivated by trypsin; thus one activation has begun, the activation process can proceed rapidly. Trypsin, in turn, activates both chymotypsinogen and procarboxypolypeptidase to form their active protease counterparts.
  • the enzymes are normally activated only when they enter the intestinal mucosa in order to prevent autodigestion of the pancreas.
  • the acinar cells also co-secrete a trypsin inhibitor that normally prevents activation of the proteolytic enzymes within the secretory cells and in the ducts of the pancreas. Inhibition of trypsin activity also prevents activation of the other proteases.
  • Pancreatitis can occur when an excess amount of trypsin saturates the supply of trypsin inhibitor. This, in turn, can be caused by underproduction of trypsin inhibitor, or the overabundance of trypsin within the cells or ducts of the pancreas. In the latter case, pancreatic trauma or blockage of a duct can lead to localized overabundance of trypsin; under acute conditions large amounts of pancreatic zymogen secretion can pool in the damaged areas of the pancreas. If even a small amount of free trypsin is available activation of all the zymogenic proteases rapidly occurs, and can lead to digestion of the pancreas (acute pancreatitis) and in particularly severe cases to the patient's death.
  • Pancreatic secretion is normally regulated by both hormonal and nervous mechanisms.
  • parasympathetic nerve impulses are relayed to the pancreas, which initially results in acetylcholine release, followed by secretion of enzymes into the pancreatic acini for temporary storage.
  • secretin is a 27 amino acid (3400 Dalton) polypeptide initially produced as the inactive form prosecretin, which is then activated by proteolytic cleavage. Secretin is then absorbed into the blood. Secretin causes the pancreas to secrete large quantities of a fluid containing bicarbonate ion. Secretin does not stimulate the acinar cells, which produce the digestive enzymes. The bicarbonate fluid serves to neutralize the chyme and to provide a slightly alkaline optimal environment for the enzymes.
  • CCK cholecystokinin
  • human CCK is synthesized as a protoprotein of 115 amino acids. Active CCK forms are quickly taken into the blood through the digestive tract, and normally stimulate the secretion of enzymes by the acinar cells. However, stimulation of the CCK receptor by the CCK analogs cerulein and CCK-octapeptide (CCK-8) appears to lead to a worsening of morbidity and mortality in mammals in whom pancreatitis is induced. See Tani et al., Pancreas 5:284-290 (1990).
  • the digestive enzymes are synthesized as zymogens; proto-enzyme synthesis occurs in the rough endoplasmic reticulum of the acinar cells.
  • the zymogens are then packaged within vesicles having a single lipid bilayer membrane.
  • the zymogens are packed within the vesicles so densely that they appear as quasi-crystalline structures when observed under light microscopy and the zymogen granules are electron-dense when observed under the electron microscope.
  • the vesicles are localized within the cytoplasm of the acinar cells. Secretion of zymogens by the acinar cells occurs through vesicle docking and subsequent fusion with the plasma membrane, resulting in the liberation of the contents into the extracellular milieu.
  • Nerve cells appear to secrete neurotransmitters and other intercellular signaling factors through a mechanism of membrane fusion that is shared with other cell types, see e.g., Rizo & Sudhof, Nature Struct. Biol. 5:839-842 (October 1998), hereby incorporated by reference herein, including the pancreatic acinar cells.
  • a vesicle first contacts the intracellular surface of the cellular membrane in a reaction called docking. Following the docking step the membrane fuses with and becomes part of the plasma membrane through a series of steps that currently remain relatively uncharacterized, but which clearly involve certain vesicle and membrane-associated proteins, as has been illustrated using neural models.
  • a protein alternatively termed synaptobrevin or VAMP vesicle-associated membrane protein
  • VAMP vesicle-associated membrane protein
  • v-SNARE vesicle-associated SNARE
  • t-SNARES target membrane-associated SNAREs
  • VAMP protein forms a core complex with syntaxin and SNAP-25; the formation of the core complex appears to be an essential step to membrane fusion.
  • SNARE system first identified in neural cells is a general model for membrane fusion in eukaryotic cells.
  • a yeast exocytotic core complex similar to that of the synaptic vesicles of mammalian neural cells has been characterized, and found to contain three proteins: Sso 1 (syntaxin 1 homolog), SncI (synaptobrevin homolog), and sec9 (SNAP-25 homolog). Rizo & Sudhof, id. These proteins share a high degree of amino acid sequence homology with their mammalian synaptosomal counterparts.
  • a synaptobrevin analog this protein is involved in the intracellular transport of vesicles, and is cleaved by TeTx, BoNT/E, BoNT/F, and BoNT/G.
  • Homologs of syntaxin have been identified in yeast (e.g., sso1p and sso2p) and mammalian non-neuronal cells (syn2p, syn3p, syn4p and syn5p).
  • yeast SNAP-25 homolog, sec9 has been identified; this protein appears to essential for vesicle fusion with the plasma membrane.
  • Intoxication of neural cells by clostridial neurotoxins exploits specific characteristics of the SNARE proteins. These neurotoxins, most commonly found expressed in Clostridium botulinum and Clostridium tetanus, are highly potent and specific poisons of neural cells. These Gram positive bacteria secrete two related but distinct toxins, each comprising two disulfide-linked amino acid chains: a light chain (L) of about 50 KDa and a heavy chain (H) of about 100 KDa, which are wholly responsible for the symptoms of botulism and tetanus, respectively.
  • L light chain
  • H heavy chain
  • the tetanus and botulinum toxins are among the most lethal substances known to man; both toxins function by inhibiting neurotransmitter release in affected neurons.
  • the tetanus neurotoxin (TeNT) acts mainly in the central nervous system, while botulinum neurotoxin (BONT) acts at the neuromuscular junction; both toxins inhibit acetylcholine release from the nerve terminal of the affected neuron into the synapse, resulting in paralysis or reduced target organ function.
  • TeNT The tetanus neurotoxin
  • BONT botulinum neurotoxins
  • BoNT/A seven different immunologically distinct serotypes
  • BoNT/G seven different immunologically distinct serotypes
  • C. botulinum two other species
  • C. baratii and C. butyricum
  • toxins similar to /F and /E respectively. See e.g., Coffield et al., The Site and Mechanism of Action of Botulinum Neurotoxin in Therapy with Botulinum Toxin 3-13 (Jankovic J. & Hallett M. eds. 1994), the disclosure of which is incorporated herein by reference.
  • the molecular mechanism of intoxication appears to be similar.
  • the toxin binds to the presynaptic membrane of the target neuron through a specific interaction between the heavy chain and a neuronal cell surface receptor; the receptor is thought to be different for each type of botulinum toxin and for TeNT.
  • the carboxy terminal (C-terminal) half of the heavy chain is required for targeting of the toxin to the cell surface.
  • the cell surface receptors while not yet conclusively identified, appear to be distinct for each neurotoxin serotype.
  • the toxin crosses the plasma membrane of the poisoned cell.
  • the toxin is first engulfed by the cell through receptor-mediated endocytosis, and an endosome containing the toxin is formed.
  • the toxin (or light chain thereof) escapes the endosome into the cytoplasm of the cell.
  • This last step is thought to be mediated by the amino terminal (N-terminal) half of the heavy chain, which triggers a conformational change of the toxin in response to a pH of about 5.5 or lower.
  • Endosomes are known to possess a proton pump that decreases intra-endosomal pH.
  • the conformational shift exposes hydrophobic residues in the toxin, which permits the toxin to embed itself in the endosomal membrane.
  • the toxin then translocates through the endosomal membrane into the cytosol.
  • the disulfide bond joining the heavy and light chain is reduced, and the light chain is released into the cytoplasm.
  • the entire toxic activity of botulinum and tetanus toxins is contained in the light chain of the holotoxin; the light chain is a zinc (Zn++) endopeptidase which selectively cleaves the SNARE proteins essential for recognition and docking of neurotransmitter-containing vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane.
  • the light chain of TxNT, BoNT/B, BoNT/D, BoNT/F, and BoNT/G cause specific proteolysis of VAMP, an integral protein.
  • BoNT/A and /E selectively cleave the plasma membrane-associated SNARE protein SNAP-25; this protein is bound to and present on the cytoplasmic surface of the plasma membrane.
  • BoNT/Cl cleaves syntaxin, which exists as an integral protein having most of its mass exposed to the cytosol. Syntaxin interacts with the calcium channels at presynaptic terminal active zones. See Tonello et al., Tetanus and Botulism Neurotoxins in Intracellular Protein Catabolism 251-260 (Suzuki K & Bond J. eds. 1996), the disclosure of which is incorporated by reference as part of this specification. Bo/NTC1 also appears to cleave SNAP-25.
  • TeNT and BONT are specifically taken up by cells present at the neuromuscular junction. BONT remains within peripheral neurons and, as indicated above, blocks release of the neurotransmitter acetylcholine from these cells.
  • TeNT through its receptor, enters vesicles that move in a retrograde manner along the axon to the soma, and is discharged into the intersynaptic space between motor neurons and the inhibitory neurons of the spinal cord.
  • TeNT binds receptors of the inhibitory neurons, is again internalized, and the light chain enters the cytosol to block the release of the inhibitory neurotransmitters 4-aminobutyric acid (GABA) and glycine from these cells.
  • GABA 4-aminobutyric acid
  • WO 96/33273 relates to derivatives of botulinum toxin designed to prevent neurotransmitter release from sensory afferent neurons to treat chronic pain. Such derivatives are targeted to nociceptive neurons using a targeting moiety that binds to a binding site of the surface of the neuron.
  • the present invention concerns methods and compositions useful for the treatment of acute pancreatitis. This condition is largely due to the defective secretion of zymogen granules by acinar cells, and by the premature co-mingling of the secreted zymogens with lysosomal hydrolysates capable of activating trypsin, thereby triggering the protease activation cascade and resulting in the destruction of pancreatic tissue.
  • the invention is a therapeutic agent comprising a chimeric protein containing an amino acid sequence-specific endopeptidase activity which will specifically cleave at least one synaptic vesicle-associated protein selected from the group consisting of SNAP-25, syntaxin or VAMP, in combination with the translocation activity of the N-terminus of a clostridial neurotoxin heavy chain, wherein the chimeric protein further comprises a recognition domain which will bind a human cholecystokinin (CCK) receptor.
  • CCK human cholecystokinin
  • the protein Upon binding of the recognition domain of the protein to the CCK receptor, the protein is specifically transported into cells containing CCK receptors (pancreatic acinar cells) through receptor-mediated endocytosis.
  • the CCK receptor is the CCK A receptor.
  • the chimeric protein functions in a manner similar to that of a clostridial neurotoxin within its target neuron.
  • the toxin moiety is translocated from the endosome into the cytoplasm, where it acts to cleave a SNARE protein identical or homologous to SNAP-25, syntaxin or VAMP.
  • the cleavage of this protein prevents formation of a core complex between the SNARE proteins and thus prevents or reduces the extent of fusion of the vesicle with the target membrane. This, in turn, results in inhibition of zymogen release from the acinar cells and of zymogen activation by lysosomal hydrolases.
  • the autodigestion of pancreatic tissue in acute pancreatitis is therefore reduced or eliminated.
  • Another embodiment of the present invention concerns a method of treating a patient suffering from acute pancreatitis by administering an effective amount of such a chimeric protein.
  • Another embodiment of the invention concerns a therapeutic composition that contains the translocation activity of a clostridial neurotoxin heavy chain in combination with a recognition domain able to bind a specific cell type and a therapeutic element having an activity other than the endopeptidase activity of a clostridial neurotoxin light chain.
  • a non-exclusive list of certain such therapeutic elements includes: hormones and hormone-agonists and antagonists, nucleic acids capable being of being used as replication, transcription, or translational templates (e.g., for expression of a protein drug having the desired biological activity or for synthesis of a nucleic acid drug as an antisense agent), enzymes, toxins, and the like.
  • the specific cell type is a pancreatic cell, most preferably a pancreatic acinar cell.
  • Another embodiment is drawn to methods for the treatment of acute pancreatitis comprising contacting an acinar cell with an effective amount of a composition comprising a chimeric protein containing an amino acid sequence-specific endopeptidase activity which will specifically cleave at least one synaptic vesicle-associated protein selected from the group consisting of SNAP-25, syntaxin or VAMP, in combination with the translocation activity of the N-terminus of a clostridial neurotoxin heavy chain, wherein the chimeric protein further comprises a recognition domain able to bind to a cell surface protein characteristic of an human pancreatic acinar cell.
  • the cell surface protein is a CCK receptor protein; most preferably the protein is the human CCK A protein.
  • CCK receptors (CCK-A receptor and CCK-B receptor) are found mainly in on the surface of pancreatic acinar cells, although they are also found in some brain cells and, to a lesser extent on the surface of gastrointestinal cells.
  • any suitable route of administration may be used in this aspect of the invention.
  • the agent is substantially specifically targeted to pancreatic cells; when the agent contains a CCK receptor-binding domain, the blood-brain barrier prevents the agent from interacting with brain cells.
  • the invention provides a composition
  • a composition comprising a drug or other therapeutic agent having an activity other than that of a clostridial neurotoxin light chain for intracellular delivery, said agent joined to the translocation domain of a clostridial neurotoxin heavy chain and a binding element able to recognize a cell surface receptor of a target cell.
  • the target cell is not a neuron.
  • the drug or other therapeutic agent has an enzymatic, catalytic, or other self-perpetuating mode of activity, so that the effective dose of drug is greater than the number of drug molecules delivered within the target cell.
  • a non-exclusive list of certain such drugs would include: hormones and hormone-agonists and antagonists, nucleic acids capable being of being used as replication, transcription, or translational templates (e.g., for expression of a protein drug having the desired biological activity or for synthesis of a nucleic acid drug as an antisense agent), enzymes, toxins (such as diphtheria toxin or ricin), and the like.
  • the drug may be cleavably linked to the remainder of the composition in such a way as to allow for the release of the drug from the composition within the target cell.
  • compositions may be provided to the patient by intravenous administration, may be administered during surgery, or may be provided parenterally.
  • WO 95/32738 which shares ownership with the present application, describes transport proteins for the therapeutic treatment of neural cells. This application is incorporated by reference herein as part of this specification.
  • the invention comprises a therapeutic polypeptide comprising three features: a binding element, a translocation element, and a therapeutic element.
  • the binding element is able to bind to a specific target cell provided that the target cell is not a motor neuron or a sensory afferent neuron.
  • the binding element comprises an amino acid chain; also an independently, it is preferably located at or near the C-terminus of a polypeptide chain.
  • binding element is meant a chemical moiety able to preferentially bind to a cell surface marker characteristic of the target cell under physiological conditions.
  • the cell surface marker may comprise a polypeptide, a polysaccharide, a lipid, a glycoprotein, a lipoprotein, or may have structural characteristics of more than one of these.
  • the disassociation constant (K d ) of the binding element for the cell surface marker is at least one order of magnitude less than that of the binding element for any other cell surface marker.
  • the disassociation constant is at least 2 orders of magnitude less, even more preferably the disassociation constant is at least 3 orders of magnitude less than that of the binding element for any other cell surface marker to which the therapeutic polypeptide is exposed.
  • the organism to be treated is a human.
  • the cell surface receptor comprises the histamine receptor
  • the binding element comprises an variable region of an antibody which will specifically bind the histamine receptor.
  • the cell surface marker is a cholecystokinin (CCK) receptor.
  • CCK cholecystokinin
  • Cholecystokinin is a bioactive peptide that functions as both a hormone and a neurotransmitter in a wide variety of physiological settings.
  • CCK is involved in the regulation of gall bladder contraction, satiety, gastric emptying, and gut motility; additionally it is involved in the regulation of pancreatic exocrine secretion.
  • CCK receptors There are two types of CCK receptors, CCK A and CCK B; the amino acid sequences of these receptors have been determined from cloned cDNA. Despite the fact that both receptors are G protein-coupled receptors and share approximately 50% homology, there are distinct differences between their physiological activity.
  • the CCK A receptor is expressed in smooth muscle cells of the gall bladder, smooth muscle and neurons within the gastrointestinal tract, and has a much greater affinity (>10 2 times higher) for CCK than the related peptide hormone gastrin.
  • the CCK B receptor found in the stomach and throughout the CNS, has roughly equal ability to bind CCK and gastrin.
  • CCK The varied activities of CCK can be partly attributed to the fact that CCK is synthesized as procholecystokinin, a protoprotein of 115 amino acids, and is then post-translationally cleaved into a number of active fragments all sharing the same C-terminus.
  • the amino acid sequence of human procholecystokinin is shown below; amino acid residues not present in the biologically active cleavage products are in lower case. All amino acid sequences herein are shown from N-terminus to C-terminus, unless expressly indicated otherwise:
  • Human procholecystokinin having the amino acid sequence SEQ ID NO:1: mnsgvclcvlmavlaagaltqpvppadpagsglqraeeaprrqlr VSQRT DGESRAHLGA LLARYIQQAR KAPSGRMSIV KNLQNLDPSH RISDRDYMGW MDF grrsaeeyeyps
  • Biologically active cleavage products of the full length CCK chain include:
  • CCK-58 having the amino acid sequence SEQ ID NO:2: VSQRT DGESRAHLGA LLARYIQQAR KAPSGRMSIV KNLQNLDPSH RISDRDYMGW MDF;
  • CCK-39 having the amino acid sequence SEQ ID NO: 3: YIQQAR KAPSGRMSIV KNLQNLDPSH RISDRDYMGW MDF;
  • CCK-33 having the amino acid sequence SEQ ID NO: 4: KAPSGRMSIV KNLQNLDPSH RISDRDYMGW MDF;
  • CCK-12 having the amino acid sequence SEQ ID NO: 5: ISDRDYMGW MDF;
  • the biologically active polypeptides contain post-translational modifications; in the case of CCK species binding the CCK-A receptor, amidation of the C-terminal phenylalanine, and sulfatation of the tyrosine residue located seven residue from the C-terminus of the biologically active species are required for hoigh affinity binding ton the receptor.
  • CCK-B only the C-terminal amidation is necessary; sulfation of the tyrosine appears to make little diffrence in CCK-B binding.
  • These modifications appear to be necessary for full biological activity, although both the unmodified C-terminal pentapeptide and tetrapeptide of CCK retains some biological activity. Kennedy et al., J. Biol. Chem. 272: 2920-2926 (1997), hereby incorporated by reference herein.
  • the biologically active therapeutic polypeptide of the present invention comprises a CCK binding element containing the post-translational modifications described above.
  • This polypeptide can be produced by synthetic chemistry or, preferably, can be produced by a combination of recombinant and synthetic means using the “expressed protein ligation” (EPL) method. See Cotton & Muir, Chemistry & Biology 6:R247 (1999), hereby incorporated by reference herein. In this method the therapeutic polypeptide is expressed without the C-terminal binding element as a fusion protein with an “intein” polypeptide sequence positioned at the C-terminus thereof.
  • EPL expressed protein ligation
  • the intein comprises a conserved cysteine, serine, or threonine residue at its amino terminus; the carboxyl terminus of the intein contains a functional binding sequence such as chitin binding domain (CBD), poly His (6 or more consecutive histidine residues), or another amino acid sequence capable of affinity binding.
  • CBD chitin binding domain
  • poly His (6 or more consecutive histidine residues)
  • another amino acid sequence capable of affinity binding is constructed using standard recombinant DNA methods.
  • a synthetic peptide comprising a C-terminal amidated phenylalanine and the desired CCK amino acid sequence.
  • Such methods are described in e.g., Bodansky, M. and Bodansky, A. The Practice of Peptide Synthesis (2d ed. Trost B. M., ed. Springer Laboratory 1994), hereby incorporated by reference herein.
  • the synthetic peptide also contains an sulfated tyrosine at the position 7 residues from the carboxyl terminus.
  • hydroxyl-containing amino acids or cysteine as the amino terminal residue of the intein and the synthetic peptide, and either thiopheol, phenol or another nucleophile capable of creating a reactive ester or thioester linkage in accordance with the expressed protein ligation methods described herein.
  • thiol-containing amino acid residues and thipheonol or another sulfur-containing nucleophile are preferred.
  • the fusion protein is immobilized following expression by incubation under selective binding conditions with a surface to which the binding partner of the carboxyl terminal has been joined (e.g., where the binding moiety is CBP, the surface may be a resin to which chitin is conjugated).
  • the immobilized fusion protein is then permitted to react in a transthioesterification reaction with a S- or O-containing reagent (such as thiophenol or phenol) and the synthetic modified peptide described above.
  • the intein which is joined to the carboxyl terminus of the therapeutic polypeptide is cleaved at the thioester (or ester) linkage, thus liberating the protein from the surface to which it was bound.
  • the intein may be transiently replaced with the thiophenol group, and the resulting thioester is then itself attacked by the cysteine (or serine or threonine) residue of the synthetic peptide; this reaction is then spontaneously followed by a shift of the carbonyl bond from S (or O) to the N terminal nitrogen of the synthetic peptide, to form a peptide bond.
  • the resultant therapeutic polypeptide thus comprises a threapeutic domain, a translocation domain, and a binding domain comprising a CCK sequence modified to contain the naturally occuring post-translational modifications.
  • extein refers to a portion of a chimeric polypeptide that borders one or more intein, and is subsequently ligated to either another extein or a synthetic polypeptide in the EPL reaction referred to herein.
  • intein refers to a portion of a chimeric polypeptide containing an N-terminal cysteine, serine, or threonine which is excised from said polypeptide during the EPL reaction referred to herein.
  • the CCK A receptor undergoes internalization to intracellular sites within minutes after agonist exposure. Pohl et al., J. Biol. Chem. 272: 18179-18184 (1997), hereby incorporated by reference herein.
  • the CCK B receptor has also shown the same ligand-dependant internalization response in transfected NIH 3T3 cells. In the CCK B receptor, but not the CCK A receptor, the endocytotic feature of the receptor been shown to be profoundly decreased by the deletion of the C terminal 44 amino acids of the receptor chain, corresponding in both receptors to an cytoplasmic portion of the receptor chain.
  • CCK residues Trp 30 and Met 31 located at positions 4 and 3, respectively, from the C terminus of mature CCK-8) reside in a hydrophobic pocket formed by receptor residues Leu 348 , Pro 352 , Ile 353 and Ile 356 .
  • CCK residue Asp 32 located at amino acid position 2 measured from the C terminus of CCK-8) seems to be involved in an ionic interaction with receptor residue Lys 115 .
  • CCK Tyr-sulfate 27 (the CCK-8 residue 7 amino acids from C terminus) appears involved in an ionic interaction with receptor residue Lys 106 and a stacking interaction with receptor residue Phe 198 .
  • Such structural models provide detailed guidance to the person of ordinary skill in the art as to the construction of a variety of binding elements able to retain the binding characteristics of biologically active CCK peptides for the CCK-A receptor, for example, as, for example, by site directed mutagenesis of a clostridial neurotoxin heavy chain.
  • models deduced using similar methodologies have been proposed for the CCK B receptor, see e.g., Jagerschmidt, A. et al., Mol. Pharmacol. 48:783-789 (1995), and can be used as a basis for the construction of binding elements that retain binding characteristics similar to the CCK B receptor.
  • the CCK-B receptor is known to exist on the surface of neurons associated with the certal nervious system.
  • the therapeutic polypeptide may be directed (for example, by intrathecal application) to these neurons rather than to the pancreas); in such a case, the binding element may comprise a CCK containing the C terminal amidation only.
  • the binding element may be constructed using the expressed protein ligation (EPL) methods described above. Indeed, EPL methods may be used to introduce and desired or required modifications to the therapeutic element, the translocation element, and/or the binding element of the claimed therapeutic polypeptide.
  • the binding element may comprise a variable region of an antibody which will bind the CCK-A or CCK-B receptor.
  • Nucleic acids encoding polypeptides containing such a binding element may be constructed using molecular biology methods well known in the art; see e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press 2d ed. 1989), and expressed within a suitable host cell. The disclosure of this latter reference is incorporated by reference herein in its entirety.
  • the translocation element comprises a portion of a clostridial neurotoxin heavy chain having a translocation activity.
  • translocation is meant the ability to facilitate the transport of a polypeptide through a vesicular membrane, thereby exposing some or all of the polypeptide to the cytoplasm.
  • botulinum neurotoxins translocation is thought to involve an allosteric conformational change of the heavy chain caused by a decrease in pH within the endosome.
  • amino acid sequence of the translocation-mediating portion of the botulinum neurotoxin heavy chain is known to those of skill in the art; additionally, those amino acid residues within this portion that are known to be essential for conferring the translocation activity are also known.
  • the therapeutic element of the present invention may comprise, without limitation: active or inactive (i.e., modified) hormone receptors (such as androgen, estrogen, retinoid, perioxysome proliferator and ecdysone receptors etc.), and hormone-agonists and antagonists, nucleic acids capable being of being used as replication, transcription, or translational templates (e.g., for expression of a protein drug having the desired biological activity or for synthesis of a nucleic acid drug as an antisense agent), enzymes, toxins (including apoptosis-inducing agents), and the like.
  • active or inactive (i.e., modified) hormone receptors such as androgen, estrogen, retinoid, perioxysome proliferator and ecdysone receptors etc.
  • hormone-agonists and antagonists such as androgen, estrogen, retinoid, perioxysome proliferator and ecdysone receptors etc.
  • the therapeutic element is a polypeptide comprising a clostridial neurotoxin light chain or a portion thereof retaining the SNARE-protein sequence-specific endopeptidase activity of a clostridial neurotoxin light chain.
  • the amino acid sequences of the light chain of botulinum neurotoxin (BONT) subtypes A-G have been determined, as has the amino acid sequence of the light chain of the tetanus neurotoxin (TeNT).
  • Each chain contains the Zn ++ -binding motif His-Glu-x-x-His (N terminal direction at the left) characteristic of Zn ++ -dependent endopeptidases (HELIH in TeNT, BoNT/A /B and /E; HELNH in BoNT/C; and HELTH in BoNT/D).
  • the catalytic domain of BoNT/A is very specific for the C-terminus of SNAP-25 and appears to require a minimum of 16 SNAP-25 amino acids for cleavage to occur.
  • the catalytic site resembles a pocket; when the light chained is linked to the heavy chain via the disulfide bond between Cys 429 and Cys 453 , the translocation domain of the heavy chain appears to block access to the catalytic pocket until the light chain gains entry to the cytosol. When the disulfide bond is reduced, the two polypeptide chains dissociate, and the catalytic pocket is then “opened” and the light chain is fully active.
  • VAMP and syntaxin are cleaved by BoNT/B, D, F, G and TeNT, and BoNT/C 1 , respectively, while SNAP-25 is cleaved by BoNT/A and E.
  • the substrate specificities of the various clostridial neurotoxin light chains other than BoNT/A are known. Therefore, the person of ordinary skill in the art could easily determine the toxin residues essential in these subtypes for cleavage and substrate recognition (for example, by site-directed mutagenesis or deletion of various regions of the toxin molecule followed by testing of proteolytic activity and substrate specificity), and could therefore easily design variants of the native neurotoxin light chain that retain the same or similar activity.
  • the clostridial neurotoxins have three functional domains analogous to the three elements of the present invention.
  • the BoNT/A neurotoxin light chain is present in amino acid residues 1-448 of the BoNT/A prototoxin (i.e., before nicking of the prototoxin to form the disulfide-linked dichain holotoxin); this amino acid sequence is provided below as SEQ ID NO: 7.
  • BoNT/A light chain MPFVNKQFNYKDPVNGVDIAYIKIPNAGQMQPVKAFKIHNKIWV (SEQ ID NO:7) IPERDTFTNPEEGDLNPPPEAKQVPVSYYDSTYLSTDNEKDNYLKGVTKLFERIYSTD LGRMLLTSIVRGIPFWGGSTIDTELKVIDTNCINVIQPDGSYRSEELNLVIIGPSADI IQFECKSFGHEVLNLTRNGYGSTQYIRFSPDFTFGFEESLEVDTNPLLGAGKFATDPA VTLA HE LI H AGHRLYGIAINPNRVFKVNTNAYYEMSGLEVSFE E LRTFGGHDAKFIDS LQENEFRLYYYNKFKDIASTLNKAKSIVGTTASLQYMKNVFKEKYLLSEDTSGKFSVD KLKFDKLYKMLTEIYTEDNFVKFFKVLNR K TYLNFDKAVFKINIVPKVNY
  • H N The heavy chain N-terminal (H N ) translocation domain is contained in amino acid residues 449-871 of the BoNT/A amino acid sequence, shown below as SEQ ID NO: 8; a gated ion channel-forming domain probably essential for the translocation activity of this peptide is underlined (see Oblatt-Montal et al., Protein Sci. 4:1490-1497(1995), hereby incorporated by reference herein.
  • the heavy chain C-terminal neural cell binding domain is contained in amino acid residues 872-1296 (SEQ ID NO: 9) of the BoNT/A prototoxin.
  • the amino acid sequence of the BoNT/A prototoxin is encoded by nucleotides 358 to 4245 of the neurotoxin cDNA sequence, set forth herein below as SEQ ID NO: 10.
  • SEQ ID NO: 10 The amino acid sequence of the BoNT/A prototoxin is encoded by nucleotides 358 to 4245 of the neurotoxin cDNA sequence, set forth herein below as SEQ ID NO: 10.
  • the translocation element and the binding element of the compositions of the present invention are separated by a spacer moiety that facilitates the binding element's binding to the desired cell surface receptor.
  • a spacer may comprise, for example, a portion of the BONT H c sequence (so long as the portion does not retain the ability to bind to the BONT or TeNT binding site of motor neurons or sensory afferent neurons), another sequence of amino acids, or a hydrocarbon moiety.
  • the spacer moiety may also comprise a proline, serine, threonine and/or cysteine-rich amino acid sequence similar or identical to a human immunoglobulin hinge region.
  • the spacer region comprises the amino acid sequence of an immunoglobulin ⁇ 1 hinge region; such a sequence has the sequence (from N terminus to C terminus): EPKSCDKTHTCPPCP (SEQ ID NO:11)
  • An agent for the treatment of acute pancreatitis is constructed as follows.
  • RNA preparation is then passed over a oligo(dT) cellulose column, the polyadenylated messenger RNA is permitted to bind, and the column is washed with 5-10 column volumes of 20 mM Tris pH 7.6, 0.5 M NaCl, 1 mM EDTA (ethylenediamine tetraacetic acid), 0.1% (w/v)SDS (sodium dodecyl sulfate).
  • RNA Polyadenylated RNA is then eluted with 2-3 column volumes of STE (10 mM Tris (pH 7.6), 1 mM EDTA, 0.05% (w/v) SDS). The pooled MRNA is then precipitated in 2 volumes of ice cold ethanol, pelleted in a centrifuge at 10,000 ⁇ g for 15 minutes, then redissolved in a small volume of STE.
  • the BoNT/A MRNA is used as a template for DNA synthesis using Moloney murine leukemia virus reverse transcriptase (MMLV-RT), then the L chain and then H N chain of the neurotoxin is amplified from the cDNA by the polymerase chain reaction (PCR) using appropriate oligonucleotide primers whose sequences are designed based on the BoNT/A neurotoxin cDNA sequence of SEQ ID NO: 9. These procedures are performed using the standard techniques of molecular biology as detailed in, for example, Sambrook et al., already incorporated by reference herein. The primer defining the beginning of the coding region (5′ side of the L chain fragment) is given a StuI site.
  • the PCR primer defining the 3′ end of the H N -encoding domain has the following features (from 3′ to 5′): a 5′ region sufficiently complementary to the 3′ end of the HN-encoding domain to anneal thereto under amplification conditions, a nucleotide sequence encoding the human immunoglobulin hinge region ⁇ 1 (SEQ ID NO:11), a nucleotide sequence encoding the human CCK-8 octapeptide (SEQ ID NO:6), and a unique restriction endonuclease cleavage site.
  • the PCR product (termed BoNT/A L-HN- ⁇ -CCK ) is purified by agarose gel electrophoresis, and cloned into a pBluescript II SK vector.
  • the resulting plasmid is used to transform competent E. coli cells, and a preparation of the resulting plasmid is made.
  • the BoNT/A L-HN- ⁇ -CCK fragment is excised from the pBluescript vector and cloned into a mammalian expression vector immediately downstream of a strong promoter.
  • the resulting vector is used to transfect a culture of the appropriate host cell, which is then grown to confluence.
  • BoNT/A L-HN- ⁇ -CCK polypeptide Expression of the BoNT/A L-HN- ⁇ -CCK polypeptide is induced, and the cells are lysed.
  • the polypeptide is first purified by gel exclusion chromatography, the fractions containing the recombinant therapeutic agent are pooled, then the BoNT/A L-HN- ⁇ -CCK polypeptide is further purified using an anti-Ig affinity column wherein the antibody is directed to the ⁇ 1 hinge region of a human immunoglobulin.
  • a therapeutically effective amount of the BoNT/A L-HN- ⁇ -CCK agent constructed and purified as set forth in Example 1 is formulated in an acceptable infusion solution.
  • Properties of pharmacologically acceptable infusion solutions, including proper electrolyte balance, are well known in the art. This solution is provided intravenously to a patient suffering from acute pancreatitis on a single day over a period of one to two hours. Additionally, the patient is fed intravenously on a diet low in complex carbohydrates, complex fats and proteins.
  • the patient's pancreas shows signs of autodigestion, as measured by blood amylase levels.
  • autodigestion has ceased, and the patient's pancreas has stabilized.
  • Example 2 a patient suffering from acute pancreatitis is treated as in Example 2, with, the therapeutic agent given continuously over a period of two weeks. After the treatment regimen, autodigestion has ceased, and the patient's pancreas has stabilized.
  • a patient suffering from acute pancreatitis is given a single pharmacologically effective amount of the therapeutic agent of Example 1 by parenteral administration.
  • Two days after the treatment regimen autodigestion has ceased and the patient's pancreas has stabilized.

Abstract

Methods and compositions for the treatment of acute pancreatitis in a mammal. Particular compositions comprise a binding element, a translocation element, and a therapeutic element able to prevent accumulation of digestive enzymes within the pancreas.

Description

  • This application is a continuation-in-part of application Ser. No. 09/288,326, filed Apr. 8, 1999.
  • FIELD OF THE INVENTION
  • The present invention includes methods and compositions for the treatment of acute pancreatitis. In a preferred embodiment the invention concerns the use of agents to reduce or prevent the secretion of pancreatic digestive enzymes within the pancreas. Such agents are targeted to pancreatic cells, and serve to prevent the exocytotic fusion of vesicles containing these enzymes with the plasma membrane. The invention is also concerned with methods of treating a mammal suffering from pancreatitis through the administration of such agents.
  • BACKGROUND OF THE INVENTION
  • Pancreatitis is a serious medical condition involving an inflammation of the pancreas. In acute or chronic pancreatitis the inflammation manifests itself in the release and activation of pancreatic enzymes within the organ itself, leading to autodigestion. In many cases of acute pancreatitis, the condition can lead to death.
  • In normal mammals, the pancreas, a large gland similar in structure to the salivary gland, is responsible for the production and secretion of digestive enzymes, which digest ingested food, and bicarbonate for the neutralization of the acidic chyme produced in the stomach. The pancreas contains acinar cells, responsible for enzyme production, and ductal cells, which secrete large amounts of sodium bicarbonate solution. The combined secretion product is termed “pancreatic juice”; this liquid flows through the pancreatic duct past the sphincter of Oddi into the duodenum. The secretion of pancreatic juice is stimulated by the presence of chyme in the upper portions of the small intestine, and the precise composition of pancreatic juice appears to be influenced by the types of compounds (carbohydrate, lipid, protein, and/or nucleic acid) in the chyme.
  • The constituents of pancreatic juice includes proteases (trypsin, chymotrypsin, carboxypolypeptidase), nucleases (RNAse and DNAse), pancreatic amylase, and lipases (pancreatic lipase, cholesterol esterase and phospholipase). Many of these enzymes, including the proteases, are initially synthesized by the acinar cells in an inactive form as zymogens: thus trypsin is synthesized as trypsinogen, chymotrypsin as chymotypsinogen, and carboxypolypeptidase as procarboxypolypeptidase. These enzymes are activated according to a cascade, wherein, in the first step, trypsin is activated through proteolytic cleavage by the enzyme enterokinase. Trypsinogen can also be autoactivated by trypsin; thus one activation has begun, the activation process can proceed rapidly. Trypsin, in turn, activates both chymotypsinogen and procarboxypolypeptidase to form their active protease counterparts.
  • The enzymes are normally activated only when they enter the intestinal mucosa in order to prevent autodigestion of the pancreas. In order to prevent premature activation, the acinar cells also co-secrete a trypsin inhibitor that normally prevents activation of the proteolytic enzymes within the secretory cells and in the ducts of the pancreas. Inhibition of trypsin activity also prevents activation of the other proteases.
  • Pancreatitis can occur when an excess amount of trypsin saturates the supply of trypsin inhibitor. This, in turn, can be caused by underproduction of trypsin inhibitor, or the overabundance of trypsin within the cells or ducts of the pancreas. In the latter case, pancreatic trauma or blockage of a duct can lead to localized overabundance of trypsin; under acute conditions large amounts of pancreatic zymogen secretion can pool in the damaged areas of the pancreas. If even a small amount of free trypsin is available activation of all the zymogenic proteases rapidly occurs, and can lead to digestion of the pancreas (acute pancreatitis) and in particularly severe cases to the patient's death.
  • Pancreatic secretion is normally regulated by both hormonal and nervous mechanisms. When the gastric phase of stomach secretion occurs, parasympathetic nerve impulses are relayed to the pancreas, which initially results in acetylcholine release, followed by secretion of enzymes into the pancreatic acini for temporary storage.
  • When acid chyme thereafter enters the small intestine, the mucosal cells of the upper intestine release a hormone called secretin. In humans, secretin is a 27 amino acid (3400 Dalton) polypeptide initially produced as the inactive form prosecretin, which is then activated by proteolytic cleavage. Secretin is then absorbed into the blood. Secretin causes the pancreas to secrete large quantities of a fluid containing bicarbonate ion. Secretin does not stimulate the acinar cells, which produce the digestive enzymes. The bicarbonate fluid serves to neutralize the chyme and to provide a slightly alkaline optimal environment for the enzymes.
  • Another peptide hormone, cholecystokinin (CCK) is released by the mucosal cells in response to the presence of food in the upper intestine. As described in further detail below, human CCK is synthesized as a protoprotein of 115 amino acids. Active CCK forms are quickly taken into the blood through the digestive tract, and normally stimulate the secretion of enzymes by the acinar cells. However, stimulation of the CCK receptor by the CCK analogs cerulein and CCK-octapeptide (CCK-8) appears to lead to a worsening of morbidity and mortality in mammals in whom pancreatitis is induced. See Tani et al., Pancreas 5:284-290 (1990).
  • As indicated above, the digestive enzymes are synthesized as zymogens; proto-enzyme synthesis occurs in the rough endoplasmic reticulum of the acinar cells. The zymogens are then packaged within vesicles having a single lipid bilayer membrane. The zymogens are packed within the vesicles so densely that they appear as quasi-crystalline structures when observed under light microscopy and the zymogen granules are electron-dense when observed under the electron microscope. The vesicles are localized within the cytoplasm of the acinar cells. Secretion of zymogens by the acinar cells occurs through vesicle docking and subsequent fusion with the plasma membrane, resulting in the liberation of the contents into the extracellular milieu.
  • Nerve cells appear to secrete neurotransmitters and other intercellular signaling factors through a mechanism of membrane fusion that is shared with other cell types, see e.g., Rizo & Sudhof, Nature Struct. Biol. 5:839-842 (October 1998), hereby incorporated by reference herein, including the pancreatic acinar cells.
  • Although the Applicants do not wish to be bound by theory, it is believed that a vesicle first contacts the intracellular surface of the cellular membrane in a reaction called docking. Following the docking step the membrane fuses with and becomes part of the plasma membrane through a series of steps that currently remain relatively uncharacterized, but which clearly involve certain vesicle and membrane-associated proteins, as has been illustrated using neural models.
  • In neurons, neurotransmitters are packaged within synaptic vesicles, formed within the cytoplasm, then transported to the inner plasma membrane where the vesicles dock and fuse with the plasma membrane. Recent studies of nerve cells employing clostridial neurotoxins as probes of membrane fusion have revealed that fusion of synaptic vesicles with the cell membrane in nerve cells depends upon the presence of specific proteins that are associated with either the vesicle or the target membrane. See id. These proteins have been termed SNAREs. As discussed in further detail below, a protein alternatively termed synaptobrevin or VAMP (vesicle-associated membrane protein) is a vesicle-associated SNARE (v-SNARE). There are at least two isoforms of synaptobrevin; these two isoforms are differentially expressed in the mammalian central nervous system, and are selectively associated with synaptic vesicles in neurons and secretory organelles in neuroendocrine cells. The target membrane-associated SNAREs (t-SNARES) include syntaxin and SNAP-25. Following docking, the VAMP protein forms a core complex with syntaxin and SNAP-25; the formation of the core complex appears to be an essential step to membrane fusion. See Rizo & Sudhof, id. and Neimmann et al., Trends in Cell Biol. 4:179-185 (May 1994), hereby incorporated by referenced herein.
  • Recently evidence has increasingly indicated that the SNARE system first identified in neural cells is a general model for membrane fusion in eukaryotic cells. A yeast exocytotic core complex similar to that of the synaptic vesicles of mammalian neural cells has been characterized, and found to contain three proteins: Sso 1 (syntaxin 1 homolog), SncI (synaptobrevin homolog), and sec9 (SNAP-25 homolog). Rizo & Sudhof, id. These proteins share a high degree of amino acid sequence homology with their mammalian synaptosomal counterparts.
  • All mammalian non-neuronal cells appear to contain cellubrevin, a synaptobrevin analog—this protein is involved in the intracellular transport of vesicles, and is cleaved by TeTx, BoNT/E, BoNT/F, and BoNT/G. Homologs of syntaxin have been identified in yeast (e.g., sso1p and sso2p) and mammalian non-neuronal cells (syn2p, syn3p, syn4p and syn5p). Finally, as indicated above, a yeast SNAP-25 homolog, sec9 has been identified; this protein appears to essential for vesicle fusion with the plasma membrane.
  • Intoxication of neural cells by clostridial neurotoxins exploits specific characteristics of the SNARE proteins. These neurotoxins, most commonly found expressed in Clostridium botulinum and Clostridium tetanus, are highly potent and specific poisons of neural cells. These Gram positive bacteria secrete two related but distinct toxins, each comprising two disulfide-linked amino acid chains: a light chain (L) of about 50 KDa and a heavy chain (H) of about 100 KDa, which are wholly responsible for the symptoms of botulism and tetanus, respectively.
  • The tetanus and botulinum toxins are among the most lethal substances known to man; both toxins function by inhibiting neurotransmitter release in affected neurons. The tetanus neurotoxin (TeNT) acts mainly in the central nervous system, while botulinum neurotoxin (BONT) acts at the neuromuscular junction; both toxins inhibit acetylcholine release from the nerve terminal of the affected neuron into the synapse, resulting in paralysis or reduced target organ function.
  • The tetanus neurotoxin (TeNT) is known to exist in one immunologically distinct type; the botulinum neurotoxins (BONT) are known to occur in seven different immunologically distinct serotypes, termed BoNT/A through BoNT/G. While all of these latter types are produced by isolates of C. botulinum, two other species, C. baratii and C. butyricum also produce toxins similar to /F and /E, respectively. See e.g., Coffield et al., The Site and Mechanism of Action of Botulinum Neurotoxin in Therapy with Botulinum Toxin 3-13 (Jankovic J. & Hallett M. eds. 1994), the disclosure of which is incorporated herein by reference.
  • Regardless of type, the molecular mechanism of intoxication appears to be similar. In the first step of the process, the toxin binds to the presynaptic membrane of the target neuron through a specific interaction between the heavy chain and a neuronal cell surface receptor; the receptor is thought to be different for each type of botulinum toxin and for TeNT. The carboxy terminal (C-terminal) half of the heavy chain is required for targeting of the toxin to the cell surface. The cell surface receptors, while not yet conclusively identified, appear to be distinct for each neurotoxin serotype.
  • In the second step, the toxin crosses the plasma membrane of the poisoned cell. The toxin is first engulfed by the cell through receptor-mediated endocytosis, and an endosome containing the toxin is formed. The toxin (or light chain thereof) then escapes the endosome into the cytoplasm of the cell. This last step is thought to be mediated by the amino terminal (N-terminal) half of the heavy chain, which triggers a conformational change of the toxin in response to a pH of about 5.5 or lower. Endosomes are known to possess a proton pump that decreases intra-endosomal pH. The conformational shift exposes hydrophobic residues in the toxin, which permits the toxin to embed itself in the endosomal membrane. The toxin then translocates through the endosomal membrane into the cytosol.
  • Either during or after translocation the disulfide bond joining the heavy and light chain is reduced, and the light chain is released into the cytoplasm. The entire toxic activity of botulinum and tetanus toxins is contained in the light chain of the holotoxin; the light chain is a zinc (Zn++) endopeptidase which selectively cleaves the SNARE proteins essential for recognition and docking of neurotransmitter-containing vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane. The light chain of TxNT, BoNT/B, BoNT/D, BoNT/F, and BoNT/G cause specific proteolysis of VAMP, an integral protein. During proteolysis, most of the VAMP present at the cytosolic surface of the synaptic vesicle is inactivated as a result of any one of these cleavage events. Each toxin cleaves a different specific peptide bond.
  • BoNT/A and /E selectively cleave the plasma membrane-associated SNARE protein SNAP-25; this protein is bound to and present on the cytoplasmic surface of the plasma membrane. BoNT/Cl cleaves syntaxin, which exists as an integral protein having most of its mass exposed to the cytosol. Syntaxin interacts with the calcium channels at presynaptic terminal active zones. See Tonello et al., Tetanus and Botulism Neurotoxins in Intracellular Protein Catabolism 251-260 (Suzuki K & Bond J. eds. 1996), the disclosure of which is incorporated by reference as part of this specification. Bo/NTC1 also appears to cleave SNAP-25.
  • Both TeNT and BONT are specifically taken up by cells present at the neuromuscular junction. BONT remains within peripheral neurons and, as indicated above, blocks release of the neurotransmitter acetylcholine from these cells.
  • By contrast TeNT, through its receptor, enters vesicles that move in a retrograde manner along the axon to the soma, and is discharged into the intersynaptic space between motor neurons and the inhibitory neurons of the spinal cord. At this point, TeNT binds receptors of the inhibitory neurons, is again internalized, and the light chain enters the cytosol to block the release of the inhibitory neurotransmitters 4-aminobutyric acid (GABA) and glycine from these cells. Id.
  • International Patent Publication No. WO 96/33273 relates to derivatives of botulinum toxin designed to prevent neurotransmitter release from sensory afferent neurons to treat chronic pain. Such derivatives are targeted to nociceptive neurons using a targeting moiety that binds to a binding site of the surface of the neuron.
  • International Patent Publication No. 98/07864 discusses the production of recombinant toxin fragments that have domains that enable the polypeptide to translocate into a target cell or which increase the solubility of the polypeptide, or both.
  • SUMMARY OF THE INVENTION
  • The present invention concerns methods and compositions useful for the treatment of acute pancreatitis. This condition is largely due to the defective secretion of zymogen granules by acinar cells, and by the premature co-mingling of the secreted zymogens with lysosomal hydrolysates capable of activating trypsin, thereby triggering the protease activation cascade and resulting in the destruction of pancreatic tissue.
  • In one embodiment of this aspect, the invention is a therapeutic agent comprising a chimeric protein containing an amino acid sequence-specific endopeptidase activity which will specifically cleave at least one synaptic vesicle-associated protein selected from the group consisting of SNAP-25, syntaxin or VAMP, in combination with the translocation activity of the N-terminus of a clostridial neurotoxin heavy chain, wherein the chimeric protein further comprises a recognition domain which will bind a human cholecystokinin (CCK) receptor. Upon binding of the recognition domain of the protein to the CCK receptor, the protein is specifically transported into cells containing CCK receptors (pancreatic acinar cells) through receptor-mediated endocytosis. In a preferred embodiment, the CCK receptor is the CCK A receptor.
  • Once inside the acinar cell, the chimeric protein functions in a manner similar to that of a clostridial neurotoxin within its target neuron. The toxin moiety is translocated from the endosome into the cytoplasm, where it acts to cleave a SNARE protein identical or homologous to SNAP-25, syntaxin or VAMP. The cleavage of this protein prevents formation of a core complex between the SNARE proteins and thus prevents or reduces the extent of fusion of the vesicle with the target membrane. This, in turn, results in inhibition of zymogen release from the acinar cells and of zymogen activation by lysosomal hydrolases. The autodigestion of pancreatic tissue in acute pancreatitis is therefore reduced or eliminated.
  • Another embodiment of the present invention concerns a method of treating a patient suffering from acute pancreatitis by administering an effective amount of such a chimeric protein.
  • Another embodiment of the invention concerns a therapeutic composition that contains the translocation activity of a clostridial neurotoxin heavy chain in combination with a recognition domain able to bind a specific cell type and a therapeutic element having an activity other than the endopeptidase activity of a clostridial neurotoxin light chain. A non-exclusive list of certain such therapeutic elements includes: hormones and hormone-agonists and antagonists, nucleic acids capable being of being used as replication, transcription, or translational templates (e.g., for expression of a protein drug having the desired biological activity or for synthesis of a nucleic acid drug as an antisense agent), enzymes, toxins, and the like.
  • In a preferred embodiment, the specific cell type is a pancreatic cell, most preferably a pancreatic acinar cell.
  • Another embodiment is drawn to methods for the treatment of acute pancreatitis comprising contacting an acinar cell with an effective amount of a composition comprising a chimeric protein containing an amino acid sequence-specific endopeptidase activity which will specifically cleave at least one synaptic vesicle-associated protein selected from the group consisting of SNAP-25, syntaxin or VAMP, in combination with the translocation activity of the N-terminus of a clostridial neurotoxin heavy chain, wherein the chimeric protein further comprises a recognition domain able to bind to a cell surface protein characteristic of an human pancreatic acinar cell. Preferably the cell surface protein is a CCK receptor protein; most preferably the protein is the human CCK A protein. CCK receptors (CCK-A receptor and CCK-B receptor) are found mainly in on the surface of pancreatic acinar cells, although they are also found in some brain cells and, to a lesser extent on the surface of gastrointestinal cells.
  • Any suitable route of administration may be used in this aspect of the invention. Applicants currently prefer to administer the therapeutic agent in an intravenous infusion solution; however methods such as ingestion (particularly when associated with neurotoxin-associated proteins (NAPs); see Sharma et al., J. Nat. Toxins 7:239-253(1998), incorporated by reference herein), direct delivery to the pancreas, injection and the like may also be used. The agent is substantially specifically targeted to pancreatic cells; when the agent contains a CCK receptor-binding domain, the blood-brain barrier prevents the agent from interacting with brain cells.
  • In yet another embodiment the invention provides a composition comprising a drug or other therapeutic agent having an activity other than that of a clostridial neurotoxin light chain for intracellular delivery, said agent joined to the translocation domain of a clostridial neurotoxin heavy chain and a binding element able to recognize a cell surface receptor of a target cell. In a preferred embodiment, the target cell is not a neuron. Also, in this embodiment it is preferred that the drug or other therapeutic agent has an enzymatic, catalytic, or other self-perpetuating mode of activity, so that the effective dose of drug is greater than the number of drug molecules delivered within the target cell. A non-exclusive list of certain such drugs would include: hormones and hormone-agonists and antagonists, nucleic acids capable being of being used as replication, transcription, or translational templates (e.g., for expression of a protein drug having the desired biological activity or for synthesis of a nucleic acid drug as an antisense agent), enzymes, toxins (such as diphtheria toxin or ricin), and the like.
  • In this embodiment the drug may be cleavably linked to the remainder of the composition in such a way as to allow for the release of the drug from the composition within the target cell.
  • The presently claimed compositions may be provided to the patient by intravenous administration, may be administered during surgery, or may be provided parenterally.
  • WO 95/32738, which shares ownership with the present application, describes transport proteins for the therapeutic treatment of neural cells. This application is incorporated by reference herein as part of this specification.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • In a basic and presently preferred form, the invention comprises a therapeutic polypeptide comprising three features: a binding element, a translocation element, and a therapeutic element.
  • The binding element is able to bind to a specific target cell provided that the target cell is not a motor neuron or a sensory afferent neuron. Preferably, the binding element comprises an amino acid chain; also an independently, it is preferably located at or near the C-terminus of a polypeptide chain. By “binding element” is meant a chemical moiety able to preferentially bind to a cell surface marker characteristic of the target cell under physiological conditions. The cell surface marker may comprise a polypeptide, a polysaccharide, a lipid, a glycoprotein, a lipoprotein, or may have structural characteristics of more than one of these. By “preferentially interact” is meant that the disassociation constant (Kd) of the binding element for the cell surface marker is at least one order of magnitude less than that of the binding element for any other cell surface marker. Preferably, the disassociation constant is at least 2 orders of magnitude less, even more preferably the disassociation constant is at least 3 orders of magnitude less than that of the binding element for any other cell surface marker to which the therapeutic polypeptide is exposed. Preferably, the organism to be treated is a human.
  • In one embodiment the cell surface receptor comprises the histamine receptor, and the binding element comprises an variable region of an antibody which will specifically bind the histamine receptor.
  • In an especially preferred embodiment, the cell surface marker is a cholecystokinin (CCK) receptor. Cholecystokinin is a bioactive peptide that functions as both a hormone and a neurotransmitter in a wide variety of physiological settings. Thus, CCK is involved in the regulation of gall bladder contraction, satiety, gastric emptying, and gut motility; additionally it is involved in the regulation of pancreatic exocrine secretion.
  • There are two types of CCK receptors, CCK A and CCK B; the amino acid sequences of these receptors have been determined from cloned cDNA. Despite the fact that both receptors are G protein-coupled receptors and share approximately 50% homology, there are distinct differences between their physiological activity. The CCK A receptor is expressed in smooth muscle cells of the gall bladder, smooth muscle and neurons within the gastrointestinal tract, and has a much greater affinity (>102 times higher) for CCK than the related peptide hormone gastrin. The CCK B receptor, found in the stomach and throughout the CNS, has roughly equal ability to bind CCK and gastrin.
  • The varied activities of CCK can be partly attributed to the fact that CCK is synthesized as procholecystokinin, a protoprotein of 115 amino acids, and is then post-translationally cleaved into a number of active fragments all sharing the same C-terminus. The amino acid sequence of human procholecystokinin is shown below; amino acid residues not present in the biologically active cleavage products are in lower case. All amino acid sequences herein are shown from N-terminus to C-terminus, unless expressly indicated otherwise:
  • Human procholecystokinin, having the amino acid sequence SEQ ID NO:1:
    mnsgvclcvlmavlaagaltqpvppadpagsglqraeeaprrqlr
    VSQRT DGESRAHLGA LLARYIQQAR KAPSGRMSIV
    KNLQNLDPSH RISDRDYMGW MDF grrsaeeyeyps
  • Biologically active cleavage products of the full length CCK chain include:
  • CCK-58, having the amino acid sequence SEQ ID NO:2:
    VSQRT DGESRAHLGA LLARYIQQAR KAPSGRMSIV KNLQNLDPSH
    RISDRDYMGW MDF;
  • CCK-39, having the amino acid sequence SEQ ID NO: 3:
    YIQQAR KAPSGRMSIV KNLQNLDPSH RISDRDYMGW MDF;
  • CCK-33, having the amino acid sequence SEQ ID NO: 4:
    KAPSGRMSIV KNLQNLDPSH RISDRDYMGW MDF;
  • CCK-12, having the amino acid sequence SEQ ID NO: 5:
    ISDRDYMGW MDF;
  • and CCK-8, having the amino acid sequence SEQ ID NO: 6:
    RDYMGW MDF.
  • In each case, the biologically active polypeptides contain post-translational modifications; in the case of CCK species binding the CCK-A receptor, amidation of the C-terminal phenylalanine, and sulfatation of the tyrosine residue located seven residue from the C-terminus of the biologically active species are required for hoigh affinity binding ton the receptor. In the case of CCK-B, only the C-terminal amidation is necessary; sulfation of the tyrosine appears to make little diffrence in CCK-B binding. These modifications appear to be necessary for full biological activity, although both the unmodified C-terminal pentapeptide and tetrapeptide of CCK retains some biological activity. Kennedy et al., J. Biol. Chem. 272: 2920-2926 (1997), hereby incorporated by reference herein.
  • In a preferred embodiment, the biologically active therapeutic polypeptide of the present invention comprises a CCK binding element containing the post-translational modifications described above. This polypeptide can be produced by synthetic chemistry or, preferably, can be produced by a combination of recombinant and synthetic means using the “expressed protein ligation” (EPL) method. See Cotton & Muir, Chemistry & Biology 6:R247 (1999), hereby incorporated by reference herein. In this method the therapeutic polypeptide is expressed without the C-terminal binding element as a fusion protein with an “intein” polypeptide sequence positioned at the C-terminus thereof. The intein comprises a conserved cysteine, serine, or threonine residue at its amino terminus; the carboxyl terminus of the intein contains a functional binding sequence such as chitin binding domain (CBD), poly His (6 or more consecutive histidine residues), or another amino acid sequence capable of affinity binding. The coding sequence of this recombinantly expressed polypeptide is constructed using standard recombinant DNA methods.
  • Additionally, standard solid phase peptide synthesis methods are employed to construct a synthetic peptide comprising a C-terminal amidated phenylalanine and the desired CCK amino acid sequence. Such methods are described in e.g., Bodansky, M. and Bodansky, A. The Practice of Peptide Synthesis (2d ed. Trost B. M., ed. Springer Laboratory 1994), hereby incorporated by reference herein. The synthetic peptide also contains an sulfated tyrosine at the position 7 residues from the carboxyl terminus. This can be done either by incorporation of commercially available Fmoc-Tyr(OSO3 )—OH into the peptide chain at the 7th amino acid position prior to cleavage of the synthetic peptide from the solid support hereby incorporated by reference herein), or by standard peptide synthesis using tyrosine at position 7, followed by a sulfation reaction of the peptide resulting in tyrosine sulfate at the 7 position. See e.g., Koeller, K. M., J. Am. Chem. Soc. 122:742-743 (2000). The synthetic peptide is constructed with a cysteine (or serine or threonine) residue at the amino terminus.
  • It will be understood that one can use either hydroxyl-containing amino acids or cysteine as the amino terminal residue of the intein and the synthetic peptide, and either thiopheol, phenol or another nucleophile capable of creating a reactive ester or thioester linkage in accordance with the expressed protein ligation methods described herein. However, thiol-containing amino acid residues and thipheonol or another sulfur-containing nucleophile are preferred.
  • Thus, according to one embodiment of the expressed protein ligation method, the fusion protein is immobilized following expression by incubation under selective binding conditions with a surface to which the binding partner of the carboxyl terminal has been joined (e.g., where the binding moiety is CBP, the surface may be a resin to which chitin is conjugated). The immobilized fusion protein is then permitted to react in a transthioesterification reaction with a S- or O-containing reagent (such as thiophenol or phenol) and the synthetic modified peptide described above. In this step, the intein which is joined to the carboxyl terminus of the therapeutic polypeptide is cleaved at the thioester (or ester) linkage, thus liberating the protein from the surface to which it was bound. The intein may be transiently replaced with the thiophenol group, and the resulting thioester is then itself attacked by the cysteine (or serine or threonine) residue of the synthetic peptide; this reaction is then spontaneously followed by a shift of the carbonyl bond from S (or O) to the N terminal nitrogen of the synthetic peptide, to form a peptide bond. The resultant therapeutic polypeptide thus comprises a threapeutic domain, a translocation domain, and a binding domain comprising a CCK sequence modified to contain the naturally occuring post-translational modifications.
  • As intended herein, the term “extein” refers to a portion of a chimeric polypeptide that borders one or more intein, and is subsequently ligated to either another extein or a synthetic polypeptide in the EPL reaction referred to herein.
  • As intended herein, the term “intein” refers to a portion of a chimeric polypeptide containing an N-terminal cysteine, serine, or threonine which is excised from said polypeptide during the EPL reaction referred to herein.
  • Of course, the Applicants contemplate that this method of producing a CCK-containing therapeutic polypeptide is exemplary only, and that variations and modification of the above-described method will be well within the ability and knowledge of those of ordinary skill in the art in light of the present patent application.
  • While it will be understood that the applicants do not wish to be bound by theory, the following findings may assist an understanding the nature of the interaction between CCK and the CCK receptors, and thus between the CCK receptor binding element of an embodiment of the present invention and its CCK receptor target.
  • In pancreatic acinar cells the CCK A receptor undergoes internalization to intracellular sites within minutes after agonist exposure. Pohl et al., J. Biol. Chem. 272: 18179-18184 (1997), hereby incorporated by reference herein. The CCK B receptor has also shown the same ligand-dependant internalization response in transfected NIH 3T3 cells. In the CCK B receptor, but not the CCK A receptor, the endocytotic feature of the receptor been shown to be profoundly decreased by the deletion of the C terminal 44 amino acids of the receptor chain, corresponding in both receptors to an cytoplasmic portion of the receptor chain.
  • Recent studies of the interaction between the CCK A receptor and CCK have shown that the primary receptor sequence region containing amino acid residues 38 through 42 is involved in the binding of CCK. Residues Trp39 and Gln40 appear to be essential for the binding of a synthetic CCK C-terminal nonapeptide (in which the methionine residues located at residue 3 and 6 from the C-terminus are substituted by norleucine and threonine respectively) to the receptor. Kennedy et al., supra. These residues do not appear to be essential for the binding of CCK analogs JMV 180 (corresponding the synthetic C-terminal heptapeptide of CCK in which the phenylalanylamide residue is substituted by a phenylethyl ester and the threonine is substituted with norleucine), and JMV 179 (in which the phenylalanylamide residue and the L-tryptophan residues of the synthetic CCK nonapeptide are substituted by a phenylethyl ester and D-tryptophan, respectively and the threonine is substituted with norleucine). Id.
  • These and similar studies have shed light on the structure of the CCK A receptor active site. Based on receptor binding experiments, a current structural model indicates that CCK residues Trp30 and Met31 (located at positions 4 and 3, respectively, from the C terminus of mature CCK-8) reside in a hydrophobic pocket formed by receptor residues Leu348, Pro352, Ile353 and Ile356. CCK residue Asp32 (located at amino acid position 2 measured from the C terminus of CCK-8) seems to be involved in an ionic interaction with receptor residue Lys115. CCK Tyr-sulfate27 (the CCK-8 residue 7 amino acids from C terminus) appears involved in an ionic interaction with receptor residue Lys106 and a stacking interaction with receptor residue Phe198. Ji, et al., 272 J. Biol. Chem. 24393-24401 (1997).
  • Such structural models provide detailed guidance to the person of ordinary skill in the art as to the construction of a variety of binding elements able to retain the binding characteristics of biologically active CCK peptides for the CCK-A receptor, for example, as, for example, by site directed mutagenesis of a clostridial neurotoxin heavy chain. Similarly, models deduced using similar methodologies have been proposed for the CCK B receptor, see e.g., Jagerschmidt, A. et al., Mol. Pharmacol. 48:783-789 (1995), and can be used as a basis for the construction of binding elements that retain binding characteristics similar to the CCK B receptor.
  • It will be appreciated that the CCK-B receptor is known to exist on the surface of neurons associated with the certal nervious system. In one alternative embodiment of the present invention the therapeutic polypeptide may be directed (for example, by intrathecal application) to these neurons rather than to the pancreas); in such a case, the binding element may comprise a CCK containing the C terminal amidation only. Such a binding element may be constructed using the expressed protein ligation (EPL) methods described above. Indeed, EPL methods may be used to introduce and desired or required modifications to the therapeutic element, the translocation element, and/or the binding element of the claimed therapeutic polypeptide.
  • Additionally, the binding element may comprise a variable region of an antibody which will bind the CCK-A or CCK-B receptor.
  • Nucleic acids encoding polypeptides containing such a binding element may be constructed using molecular biology methods well known in the art; see e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press 2d ed. 1989), and expressed within a suitable host cell. The disclosure of this latter reference is incorporated by reference herein in its entirety.
  • The translocation element comprises a portion of a clostridial neurotoxin heavy chain having a translocation activity. By “translocation” is meant the ability to facilitate the transport of a polypeptide through a vesicular membrane, thereby exposing some or all of the polypeptide to the cytoplasm.
  • In the various botulinum neurotoxins translocation is thought to involve an allosteric conformational change of the heavy chain caused by a decrease in pH within the endosome.
  • This conformational change appears to involve and be mediated by the N terminal half of the heavy chain and to result in the formation of pores in the vesicular membrane; this change permits the movement of the proteolytic light chain from within the endosomal vesicle into the cytoplasm. See e.g., Lacy, et al., Nature Struct. Biol. 5:898-902 (October 1998).
  • The amino acid sequence of the translocation-mediating portion of the botulinum neurotoxin heavy chain is known to those of skill in the art; additionally, those amino acid residues within this portion that are known to be essential for conferring the translocation activity are also known.
  • It would therefore be well within the ability of one of ordinary skill in the art, for example, to employ the naturally occurring N-terminal peptide half of the heavy chain of any of the various Clostridium tetanus or Clostridium botulinum neurotoxin subtypes as a translocation element, or to design an analogous translocation element by aligning the primary sequences of the N-terminal halves of the various heavy chains and selecting a consensus primary translocation sequence based on conserved amino acid, polarity, steric and hydrophobicity characteristics between the sequences. The therapeutic element of the present invention may comprise, without limitation: active or inactive (i.e., modified) hormone receptors (such as androgen, estrogen, retinoid, perioxysome proliferator and ecdysone receptors etc.), and hormone-agonists and antagonists, nucleic acids capable being of being used as replication, transcription, or translational templates (e.g., for expression of a protein drug having the desired biological activity or for synthesis of a nucleic acid drug as an antisense agent), enzymes, toxins (including apoptosis-inducing agents), and the like.
  • In a preferred embodiment, the therapeutic element is a polypeptide comprising a clostridial neurotoxin light chain or a portion thereof retaining the SNARE-protein sequence-specific endopeptidase activity of a clostridial neurotoxin light chain. The amino acid sequences of the light chain of botulinum neurotoxin (BONT) subtypes A-G have been determined, as has the amino acid sequence of the light chain of the tetanus neurotoxin (TeNT). Each chain contains the Zn++-binding motif His-Glu-x-x-His (N terminal direction at the left) characteristic of Zn++-dependent endopeptidases (HELIH in TeNT, BoNT/A /B and /E; HELNH in BoNT/C; and HELTH in BoNT/D).
  • Recent studies of the BoNT/A light chain have revealed certain features important for the activity and specificity of the toxin towards its target substrate, SNAP-25. Thus, studies by Zhou et al. Biochemistry 34:15175-15181 (1995) have indicated that when the light chain amino acid residue His227 is substituted with tyrosine, the resulting polypeptide is unable to cleave SNAP-25; Kurazono et al., J. Biol. Chem. 14721-14729 (1992) performed studies in the presynaptic cholinergic neurons of the buccal ganglia of Aplysia californica using recombinant BoNT/A light chain that indicated that the removal of 10 N-terminal or 32 C-terminal residues did not abolish toxicity, but that removal of 10 N-terminal or 57 C-terminal residues abolished toxicity in this system. Most recently, the crystal structure of the entire BoNT/A holotoxin has been solved; the active site is indicated as involving the participation of His222, Glu223, His226, Glu261 and Tyr365. Lacy et al., supra. (These residues correspond to His223, Glu224, His227, Glu262 and Tyr366 of the BoNT/A L chain of Kurazono et al., supra.) Interestingly, an alignment of BoNT/A through E and TeNT light chains reveals that every such chain invariably has these residues in positions analogous to BoNT/A. Kurazono et al., supra.
  • The catalytic domain of BoNT/A is very specific for the C-terminus of SNAP-25 and appears to require a minimum of 16 SNAP-25 amino acids for cleavage to occur. The catalytic site resembles a pocket; when the light chained is linked to the heavy chain via the disulfide bond between Cys429 and Cys453, the translocation domain of the heavy chain appears to block access to the catalytic pocket until the light chain gains entry to the cytosol. When the disulfide bond is reduced, the two polypeptide chains dissociate, and the catalytic pocket is then “opened” and the light chain is fully active.
  • As described above, VAMP and syntaxin are cleaved by BoNT/B, D, F, G and TeNT, and BoNT/C1, respectively, while SNAP-25 is cleaved by BoNT/A and E.
  • The substrate specificities of the various clostridial neurotoxin light chains other than BoNT/A are known. Therefore, the person of ordinary skill in the art could easily determine the toxin residues essential in these subtypes for cleavage and substrate recognition (for example, by site-directed mutagenesis or deletion of various regions of the toxin molecule followed by testing of proteolytic activity and substrate specificity), and could therefore easily design variants of the native neurotoxin light chain that retain the same or similar activity.
  • Additionally, construction of the therapeutic agents set forth in this specification would be easily constructed by the person of skill in the art. It is well known that the clostridial neurotoxins have three functional domains analogous to the three elements of the present invention. For example, and without limitation, the BoNT/A neurotoxin light chain is present in amino acid residues 1-448 of the BoNT/A prototoxin (i.e., before nicking of the prototoxin to form the disulfide-linked dichain holotoxin); this amino acid sequence is provided below as SEQ ID NO: 7. Active site residues are underlined:
    BoNT/A light chain
    MPFVNKQFNYKDPVNGVDIAYIKIPNAGQMQPVKAFKIHNKIWV (SEQ ID NO:7)
    IPERDTFTNPEEGDLNPPPEAKQVPVSYYDSTYLSTDNEKDNYLKGVTKLFERIYSTD
    LGRMLLTSIVRGIPFWGGSTIDTELKVIDTNCINVIQPDGSYRSEELNLVIIGPSADI
    IQFECKSFGHEVLNLTRNGYGSTQYIRFSPDFTFGFEESLEVDTNPLLGAGKFATDPA
    VTLAHELIHAGHRLYGIAINPNRVFKVNTNAYYEMSGLEVSFEELRTFGGHDAKFIDS
    LQENEFRLYYYNKFKDIASTLNKAKSIVGTTASLQYMKNVFKEKYLLSEDTSGKFSVD
    KLKFDKLYKMLTEIYTEDNFVKFFKVLNRKTYLNFDKAVFKINIVPKVNYTIYDGFNL
    RNTNLAANFNGQNTEINNMNFTKLKNFTGLFEFYKLLCVRGIITSKTKSLDKGYNK;
  • The heavy chain N-terminal (HN) translocation domain is contained in amino acid residues 449-871 of the BoNT/A amino acid sequence, shown below as SEQ ID NO: 8; a gated ion channel-forming domain probably essential for the translocation activity of this peptide is underlined (see Oblatt-Montal et al., Protein Sci. 4:1490-1497(1995), hereby incorporated by reference herein.
    ALNDLCIKVNNWDLFFSPSEDNFTNDLNKGEEITSDTNIEAAEENISLDLIQQYYLTFNF
    DNEPENISIENLSSDIIGQLELMPNIERFPNGKKYELDKYTMFHYLRAQEFEHGKSRI
    ALTNSVNEALLNPSRVYTFFSSDYVKKVNKATEAAMFLGWVEQLVYDFTDETSEVSTT
    DKIADITIIIPYIGPALNIGNMLYKDDFVGALIFSGAVILLEFIPEIAIPVLGTFALV
    SYIANKVLTVQTIDNALSKRNEKWDEVYKYIVTNWLAKVNTQIDLIRKKMKEALENQA
    EATKAIINYQYNQYTEEEKNNINFNIDDLSSKLNESINKAMININKFLNQCSVSYLMN
    SMIPYGVKRLEDFDASLKDALLKYIYDNRGTLIGQVDRLKDKVNNTLSTDIPFQLSKY
    VDNQRLLSTFTEYIK;
  • The heavy chain C-terminal neural cell binding domain is contained in amino acid residues 872-1296 (SEQ ID NO: 9) of the BoNT/A prototoxin.
    NIINTSILNLRYESNHLIDLSRYASKINIGSKVNFDPIDKNQI
    QLFNLESSKIEVILKNAIVYNSMYENFSTSFWIRIPKYFNSISLNNEYTIINCMENNS
    GWKVSLNYGEIIWTLQDTQEIKQRVVFKYSQMINISDYINRWIFVTITNNRLNNSKIY
    INGRLIDQKPISNLGNIHASNNIMFKLDGCRDTHRYIWIKYFNLFDKELNEKEIKDLY
    DNQSNSGILKDFWGDYLQYDKPYYMLNLYDPNKYVDVNNVGIRGYMYLKGPRGSVMTT
    NIYLNSSLYRGTKFIIKKYASGNKDNIVRNNDRVYINVVVKNKEYRLATNASQAGVEK
    ILSALEIPDVGNLSQVVVMKSKNDQGITNKCKMNLQDNNGNDIGFIGFHQFNNIAKLV
    ASNWYNRQIERSSRTLGCSWEFIPVDDGWGERPL
  • The amino acid sequence of the BoNT/A prototoxin is encoded by nucleotides 358 to 4245 of the neurotoxin cDNA sequence, set forth herein below as SEQ ID NO: 10.
    aagcttctaa atttaaatta ttaagtataa atccaaataa acaatatgtt caaaaacttg
    atgaggtaat aatttctgta ttagataata tggaaaaata tatagatata tctgaagata
    atagattgca actaatagat aacaaaaata acgcaaagaa gatgataatt agtaatgata
    tatttatttc caattgttta accctatctt ataacggtaa atatatatgt ttatctatga
    aagatgaaaa ccataattgg atgatatgta ataatgatat gtcaaagtat ttgtatttat
    ggtcatttaa ataattaata atttaattaa ttttaaatat tataagaggt gttaaatatg
    ccatttgtta ataaacaatt taattataaa gatcctgtaa atggtgttga tattgcttat
    ataaaaattc caaatgcagg acaaatgcaa ccagtaaaag cttttaaaat tcataataaa
    atatgggtta ttccagaaag agatacattt acaaatcctg aagaaggaga tttaaatcca
    ccaccagaag caaaacaagt tccagtttca tattatgatt caacatattt aagtacagat
    aatgaaaaag ataattattt aaagggagtt acaaaattat ttgagagaat ttattcaact
    gatcttggaa gaatgttgtt aacatcaata gtaaggggaa taccattttg gggtggaagt
    acaatagata cagaattaaa agttattgat actaattgta ttaatgtgat acaaccagat
    ggtagttata gatcagaaga acttaatcta gtaataatag gaccctcagc tgatattata
    cagtttgaat gtaaaagctt tggacatgaa gttttgaatc ttacgcgaaa tggttatggc
    tctactcaat acattagatt tagcccagat tttacatttg gttttgagga gtcacttgaa
    gttgatacaa atcctctttt aggtgcaggc aaatttgcta cagatccagc agtaacatta
    gcacatgaac ttatacatgc tggacataga ttatatggaa tagcaattaa tccaaatagg
    gtttttaaag taaatactaa tgcctattat gaaatgagtg ggttagaagt aagctttgag
    gaacttagaa catttggggg acatgatgca aagtttatag atagtttaca ggaaaacgaa
    tttcgtctat attattataa taagtttaaa gatatagcaa gtacacttaa taaagctaaa
    tcaatagtag gtactactgc ttcattacag tatatgaaaa atgtttttaa agagaaatat
    ctcctatctg aagatacatc tggaaaattt tcggtagata aattaaaatt tgataagtta
    tacaaaatgt taacagagat ttacacagag gataattttg ttaagttttt taaagtactt
    aacagaaaaa catatttgaa ttttgataaa gccgtattta agataaatat agtacctaag
    gtaaattaca caatatatga tggatttaat ttaagaaata caaatttagc agcaaacttt
    aatggtcaaa atacagaaat taataatatg aattttacta aactaaaaaa ttttactgga
    ttgtttgaat tttataagtt gctatgtgta agagggataa taacttctaa aactaaatca
    ttagataaag gatacaataa ggcattaaat gatttatgta tcaaagttaa taattgggac
    ttgtttttta gtccttcaga agataatttt actaatgatc taaataaagg agaagaaatt
    acatctgata ctaatataga agcagcagaa gaaaatatta gtttagattt aatacaacaa
    tattatttaa cctttaattt tgataatgaa cctgaaaata tttcaataga aaatctttca
    agtgacatta taggccaatt agaacttatg cctaatatag aaagatttcc taatggaaaa
    aagtatgagt tagataaata tactatgttc cattatcttc gtgctcaaga atttgaacat
    ggtaaatcta ggattgcttt aacaaattct gttaacgaag cattattaaa tcctagtcgt
    gtttatacat ttttttcttc agactatgta aagaaagtta ataaagctac ggaggcagct
    atgtttttag gctgggtaga acaattagta tatgatttta ccgatgaaac tagcgaagta
    agtactacgg ataaaattgc ggatataact ataattattc catatatagg acctgcttta
    aatataggta atatgttata taaagatgat tttgtaggtg ctttaatatt ttcaggagct
    gttattctgt tagaatttat accagagatt gcaatacctg tattaggtac ttttgcactt
    gtatcatata ttgcgaataa ggttctaacc gttcaaacaa tagataatgc tttaagtaaa
    agaaatgaaa aatgggatga ggtctataaa tatatagtaa caaattggtt agcaaaggtt
    aatacacaga ttgatctaat aagaaaaaaa atgaaagaag ctttagaaaa tcaagcagaa
    gcaacaaagg ctataataaa ctatcagtat aatcaatata ctgaggaaga gaaaaataat
    attaatttta atattgatga tttaagttcg aaacttaatg agtctataaa taaagctatg
    attaatataa ataaattttt gaatcaatgc tctgtttcat atttaatgaa ttctatgatc
    ccttatggtg ttaaacggtt agaagatttt gatgctagtc ttaaagatgc attattaaag
    tatatatatg ataatagagg aactttaatt ggtcaagtag atagattaaa agataaagtt
    aataatacac ttagtacaga tatacctttt cagctttcca aatacgtaga taatcaaaga
    ttattatcta catttactga atatattaag aatattatta atacttctat attgaattta
    agatatgaaa gtaatcattt aatagactta tctaggtatg catcaaaaat aaatattggt
    agtaaagtaa attttgatcc aatagataaa aatcaaattc aattatttaa tttagaaagt
    agtaaaattg aggtaatttt aaaaaatgct attgtatata atagtatgta tgaaaatttt
    agtactagct tttggataag aattcctaag tattttaaca gtataagtct aaataatgaa
    tatacaataa taaattgtat ggaaaataat tcaggatgga aagtatcact taattatggt
    gaaataatct ggactttaca ggatactcag gaaataaaac aaagagtagt ttttaaatac
    agtcaaatga ttaatatatc agattatata aacagatgga tttttgtaac tatcactaat
    aatagattaa ataactctaa aatttatata aatggaagat taatagatca aaaaccaatt
    tcaaatttag gtaatattca tgctagtaat aatataatgt ttaaattaga tggttgtaga
    gatacacata gatatatttg gataaaatat tttaatcttt ttgataagga attaaatgaa
    aaagaaatca aagatttata tgataatcaa tcaaattcag gtattttaaa agacttttgg
    ggtgattatt tacaatatga taaaccatac tatatgttaa atttatatga tccaaataaa
    tatgtcgatg taaataatgt aggtattaga ggttatatgt atcttaaagg gcctagaggt
    agcgtaatga ctacaaacat ttatttaaat tcaagtttgt atagggggac aaaatttatt
    ataaaaaaat atgcttctgg aaataaagat aatattgtta gaaataatga tcgtgtatat
    attaatgtag tagttaaaaa taaagaatat aggttagcta ctaatgcatc acaggcaggc
    gtagaaaaaa tactaagtgc attagaaata cctgatgtag gaaatctaag tcaagtagta
    gtaatgaagt caaaaaatga tcaaggaata acaaataaat gcaaaatgaa tttacaagat
    aataatggga atgatatagg ctttatagga tttcatcagt ttaataatat agctaaacta
    gtagcaagta attggtataa tagacaaatag aaagatcta gtaggacttt gggttgctca
    tgggaattta ttcctgtaga tgatggatgg ggagaaaggc cactgtaatt aatctcaaac
    tacatgagtc tgtcaagaat tttctgtaaa catccataaa aattttaaaa ttaatatgtt
    taagaataac tagatatgag tattgtttga actgcccctg tcaagtagac aggtaaaaaa
    ataaaaatta agatactatg gtctgatttc gatattctat cggagtcaga ccttttaact
    tttcttgtat cctttttgta ttgtaaaact ctatgtattc atcaattgca agttccaatt
    agtcaaaatt atgaaacttt ctaagataat acatttctga ttttataatt tcccaaaatc
    cttccatagg accattatca atacatctac caactcgaga catactttga gttgcgccta
    tctcattaag tttattcttg aaagatttac ttgtatattg aaaaccgcta tcactgtgaa
    aaagtggact agcatcagga ttggaggtaa ctgctttatc aaaggtttca aagacaagga
    cgttgttatt tgattttcca agtacatagg aaataatgct attatcatgc aaatcaagta
    tttcactcaa gtacgccttt gtttcgtctg ttaac
  • Of course, three distinct domains analogous to those described above for BoNT/A exist for all the BONT subtypes as well as for TeNT neurotoxin; an alignment of the amino acid sequences of these holotoxins will reveal the sequence coordinates for these other neurotoxin species. Additionally, while sequence information is given above for BoNT/A, the amino acid sequences of all BONT species and tetanus toxin TeNT are known and can easily be obtained from, for example, the NCBI Gen-Bank Web site: www.ncbi.nlm.nih.gov. The Clostrdial neurotoxin nucleotide and amino acid sequences disclosed at this site are expressly incorporated by reference herein.
  • Preferably, the translocation element and the binding element of the compositions of the present invention are separated by a spacer moiety that facilitates the binding element's binding to the desired cell surface receptor. Such a spacer may comprise, for example, a portion of the BONT Hc sequence (so long as the portion does not retain the ability to bind to the BONT or TeNT binding site of motor neurons or sensory afferent neurons), another sequence of amino acids, or a hydrocarbon moiety. The spacer moiety may also comprise a proline, serine, threonine and/or cysteine-rich amino acid sequence similar or identical to a human immunoglobulin hinge region. In a preferred embodiment, the spacer region comprises the amino acid sequence of an immunoglobulin γ1 hinge region; such a sequence has the sequence (from N terminus to C terminus):
    EPKSCDKTHTCPPCP  (SEQ ID NO:11)
  • It will be understood that none of the examples or embodiments described herein are to be construed as limiting the scope of the invention, which is defined solely by the claims that conclude this specification.
  • EXAMPLE 1
  • An agent for the treatment of acute pancreatitis is constructed as follows.
  • A culture of Clostridium botulinum is permitted to grown to confluence. The cells are then lysed and total RNA is extracted according to conventional methods and in the presence of an RNAse inhibitor. The RNA preparation is then passed over a oligo(dT) cellulose column, the polyadenylated messenger RNA is permitted to bind, and the column is washed with 5-10 column volumes of 20 mM Tris pH 7.6, 0.5 M NaCl, 1 mM EDTA (ethylenediamine tetraacetic acid), 0.1% (w/v)SDS (sodium dodecyl sulfate). Polyadenylated RNA is then eluted with 2-3 column volumes of STE (10 mM Tris (pH 7.6), 1 mM EDTA, 0.05% (w/v) SDS). The pooled MRNA is then precipitated in 2 volumes of ice cold ethanol, pelleted in a centrifuge at 10,000×g for 15 minutes, then redissolved in a small volume of STE.
  • The BoNT/A MRNA is used as a template for DNA synthesis using Moloney murine leukemia virus reverse transcriptase (MMLV-RT), then the L chain and then H N chain of the neurotoxin is amplified from the cDNA by the polymerase chain reaction (PCR) using appropriate oligonucleotide primers whose sequences are designed based on the BoNT/A neurotoxin cDNA sequence of SEQ ID NO: 9. These procedures are performed using the standard techniques of molecular biology as detailed in, for example, Sambrook et al., already incorporated by reference herein. The primer defining the beginning of the coding region (5′ side of the L chain fragment) is given a StuI site. The PCR primer defining the 3′ end of the HN-encoding domain has the following features (from 3′ to 5′): a 5′ region sufficiently complementary to the 3′ end of the HN-encoding domain to anneal thereto under amplification conditions, a nucleotide sequence encoding the human immunoglobulin hinge region γ1 (SEQ ID NO:11), a nucleotide sequence encoding the human CCK-8 octapeptide (SEQ ID NO:6), and a unique restriction endonuclease cleavage site.
  • The PCR product (termed BoNT/AL-HN-γ-CCK) is purified by agarose gel electrophoresis, and cloned into a pBluescript II SK vector. The resulting plasmid is used to transform competent E. coli cells, and a preparation of the resulting plasmid is made. The BoNT/AL-HN-γ-CCK fragment is excised from the pBluescript vector and cloned into a mammalian expression vector immediately downstream of a strong promoter. The resulting vector is used to transfect a culture of the appropriate host cell, which is then grown to confluence. Expression of the BoNT/AL-HN-γ-CCK polypeptide is induced, and the cells are lysed. The polypeptide is first purified by gel exclusion chromatography, the fractions containing the recombinant therapeutic agent are pooled, then the BoNT/AL-HN-γ-CCK polypeptide is further purified using an anti-Ig affinity column wherein the antibody is directed to the γ1 hinge region of a human immunoglobulin.
  • EXAMPLE 2
  • Method of Treating a Patient Suffering from Acute Pancreatitis
  • A therapeutically effective amount of the BoNT/AL-HN-γ-CCK agent constructed and purified as set forth in Example 1 is formulated in an acceptable infusion solution. Properties of pharmacologically acceptable infusion solutions, including proper electrolyte balance, are well known in the art. This solution is provided intravenously to a patient suffering from acute pancreatitis on a single day over a period of one to two hours. Additionally, the patient is fed intravenously on a diet low in complex carbohydrates, complex fats and proteins.
  • At the beginning of treatment, the patient's pancreas shows signs of autodigestion, as measured by blood amylase levels. After the treatment regimen, autodigestion has ceased, and the patient's pancreas has stabilized.
  • EXAMPLE 3
  • Alternative Treatment Method
  • In this example, a patient suffering from acute pancreatitis is treated as in Example 2, with, the therapeutic agent given continuously over a period of two weeks. After the treatment regimen, autodigestion has ceased, and the patient's pancreas has stabilized.
  • EXAMPLE 4
  • Alternative Treatment Method
  • In this example, a patient suffering from acute pancreatitis is given a single pharmacologically effective amount of the therapeutic agent of Example 1 by parenteral administration. Two days after the treatment regimen, autodigestion has ceased and the patient's pancreas has stabilized.
  • It will be understood that the present invention is not to be limited by the embodiments and examples described herein, and that the invention is defined solely by the claims that conclude this specification.

Claims (33)

1-13. (canceled)
14. A method for making a polypeptide comprising:
a) expressing within a host cell a recombinant chimeric polypeptide comprising an amino terminal side extein and a carboxyl terminal side intein
 wherein said extein comprises able to facilitate the transfer of a polypeptide across a vesicular membrane and a therapeutic element able, when present in the cytoplasm of a pancreatic cell, to inhibit enzymatic secretion by said pancreatic cell, and
 wherein said intein comprises a binding element capable of affinity binding under selective conditions with a binding partner and an amino terminal end first amino acid selected from the group consisting of cysteine, serine or threonine.
b) contacting said chimeric polypeptide with a synthetic peptide and a nucleophilic reagent
 wherein said synthetic peptide comprises a CCK binding element able to selectively bind a pancreatic cell surface marker under physiological conditions, a carboxyl terminal end amidated phenylalanine modification and an amino terminal end second amino acid selected from the group consisting of cysteine, serine or threonine,
 wherein said nucleophilic reagent is able to cause cleavage of said intein from the said extein, and
 wherein subsequent formation of a peptide bond occurs between carboxyl terminal end of said extein and amino terminal end of said synthetic peptide.
15. The method of claim 14 wherein said first and second amino acids are cysteine.
16. The method of claim 15 wherein said nucleophilic reagent is selected from the group consisting of phenol or thiphenol.
17. The method of claim 14 wherein said synthetic polypeptide further comprises a sulfated tyrosine at the position 7 amino acids from a natural C terminus of said sequence, and said therapeutic polypeptide preferentially binds a CCK-A receptor.
18. The method of claim 17 wherein said first and second amino acids are cysteine.
19. The method of claim 18 wherein said nucleophilic reagent is selected from the group consisting of phenol or thiphenol.
20. The method of claim 14 wherein said first and second amino acids are serine.
21. The method of claim 14 wherein said first and second amino acids are threonine.
22. The method of claim 17 wherein said first and second amino acids are serine.
23. The method of claim 17 wherein said first and second amino acids are threonine.
24. A method for making a polypeptide comprising:
a) expressing within a host cell a recombinant chimeric polypeptide comprising an amino terminal side extein and a carboxyl terminal side intein
 wherein said extein comprises a translocation element able to facilitate the transfer of a polypeptide across a vesicular membrane and a therapeutic element able, when present in the cytoplasm of a pancreatic cell, to inhibit enzymatic secretion by said pancreatic cell, and
 wherein said intein comprises a binding element capable of affinity binding under selective conditions with a binding partner and an amino terminal end first amino acid selected from the group consisting of cysteine, serine or threonine.
b) contacting said chimeric protein with a synthetic peptide and a nucleophilic reagent
 wherein said synthetic peptide comprises a binding element able to selectively bind a pancreatic cell surface marker under physiological conditions, a carboxyl terminal end amidated phenylalanine modification, and an amino terminal end second amino acid selected from the group consisting of cysteine, serine or threonine,
 wherein said nucleophilic reagent is able to cause cleavage of said intein from the said extein, and
 wherein subsequent formation of a peptide bond occurs between carboxyl terminal end of said extein and amino terminal end of said synthetic peptide.
25. The method of claim 24 wherein said therapeutic element will cleave a SNARE protein.
26. The method of claim 25 wherein said SNARE protein is selected from the group consisting of syntaxin, SNAP-25 and VAMP.
27. The method of claim 24 wherein said binding element of said synthetic peptide comprises a CCK sequence.
28. The method of claim 27 wherein said CCK sequence comprises a human CCK A amino acid sequence.
29. The method of claim 28 wherein said CCK A amino acid sequence comprises SEQ ID NO: 6.
30. The method of claim 28 wherein said CCK A amino acid sequence comprises SEQ ID NO: 5.
31. The method of claim 28 wherein said CCK A amino acid sequence comprises SEQ ID NO: 4.
32. The method of claim 28 wherein said CCK A amino acid sequence comprises SEQ ID NO: 3.
33. The method of claim 28 wherein said CCK A amino acid sequence comprises SEQ ID NO: 2.
34. The method of claim 24 wherein said first and second amino acids are cysteine.
35. The method of claim 24 wherein said nucleophilic reagent is selected from the group consisting of phenol or thiophenol.
36. The method of claim 24 wherein said first and second amino acids are serine.
37. The method of claim 24 wherein said first and second amino acids are threonine.
38. The method of claim 20 wherein said binding element of said synthetic peptide further comprises a sulfated tyrosine at the position 7 residues from the carboxyl terminal end.
39. The method of claim 38 wherein said binding element of said synthetic peptide comprises a CCK sequence.
40. The method of claim 39 wherein said CCK sequence comprises a human CCK A amino acid sequence.
41. The method of claim 39 wherein said CCK A amino acid sequence comprises SEQ ID NO: 6.
42. The method of claim 39 wherein said CCK A amino acid sequence comprises SEQ ID NO: 5.
43. The method of claim 39 wherein said CCK A amino acid sequence comprises SEQ ID NO: 4.
44. The method of claim 39 wherein said CCK A amino acid sequence comprises SEQ ID NO: 3.
45. The method of claim 39 wherein said CCK A amino acid sequence comprises SEQ ID NO: 2.
US11/014,795 1999-04-08 2004-12-15 Methods and compositions for the treatment of pancreatitis Abandoned US20050095251A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/014,795 US20050095251A1 (en) 1999-04-08 2004-12-15 Methods and compositions for the treatment of pancreatitis
US11/845,638 US20080025994A1 (en) 1999-04-08 2007-08-27 Methods and compositions for the treatment of pancreatitis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/288,326 US6776990B2 (en) 1999-04-08 1999-04-08 Methods and compositions for the treatment of pancreatitis
US09/548,409 US6843998B1 (en) 1999-04-08 2000-04-13 Methods and compositions for the treatment of pancreatitis
US11/014,795 US20050095251A1 (en) 1999-04-08 2004-12-15 Methods and compositions for the treatment of pancreatitis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/548,409 Division US6843998B1 (en) 1999-04-08 2000-04-13 Methods and compositions for the treatment of pancreatitis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/845,638 Continuation US20080025994A1 (en) 1999-04-08 2007-08-27 Methods and compositions for the treatment of pancreatitis

Publications (1)

Publication Number Publication Date
US20050095251A1 true US20050095251A1 (en) 2005-05-05

Family

ID=23106629

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/288,326 Expired - Fee Related US6776990B2 (en) 1999-04-08 1999-04-08 Methods and compositions for the treatment of pancreatitis
US09/548,409 Expired - Lifetime US6843998B1 (en) 1999-04-08 2000-04-13 Methods and compositions for the treatment of pancreatitis
US10/831,435 Expired - Fee Related US7276473B2 (en) 1999-04-08 2004-04-23 Methods and compositions for the treatment of pancreatitis
US11/014,795 Abandoned US20050095251A1 (en) 1999-04-08 2004-12-15 Methods and compositions for the treatment of pancreatitis
US11/845,638 Abandoned US20080025994A1 (en) 1999-04-08 2007-08-27 Methods and compositions for the treatment of pancreatitis

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US09/288,326 Expired - Fee Related US6776990B2 (en) 1999-04-08 1999-04-08 Methods and compositions for the treatment of pancreatitis
US09/548,409 Expired - Lifetime US6843998B1 (en) 1999-04-08 2000-04-13 Methods and compositions for the treatment of pancreatitis
US10/831,435 Expired - Fee Related US7276473B2 (en) 1999-04-08 2004-04-23 Methods and compositions for the treatment of pancreatitis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/845,638 Abandoned US20080025994A1 (en) 1999-04-08 2007-08-27 Methods and compositions for the treatment of pancreatitis

Country Status (3)

Country Link
US (5) US6776990B2 (en)
AU (1) AU4205100A (en)
WO (1) WO2000061192A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070179281A1 (en) * 2003-04-11 2007-08-02 Atassi M Zouhair BoNT/A Peptides and Methods of Predicting and Reducing Immunoresistance to Botulinum Toxin Therapy
US20090162341A1 (en) * 2004-12-01 2009-06-25 Keith Foster Non-Cytotoxic Protein Conjugates
WO2009131435A1 (en) * 2008-04-23 2009-10-29 Erasmus University Medical Center Rotterdam Linker containing bungarotoxin and a binding peptide
US20100034802A1 (en) * 2006-06-01 2010-02-11 Syntaxin Limited Treatment of pain
US20110027256A1 (en) * 2004-12-01 2011-02-03 Syntaxin Ltd. Fusion proteins
US20110028691A1 (en) * 2002-09-12 2011-02-03 The Health Protection Agency Recombinant toxin fragments
US8512984B2 (en) 2004-12-01 2013-08-20 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8603779B2 (en) 2004-12-01 2013-12-10 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8778634B2 (en) 2004-12-01 2014-07-15 Syntaxin, Ltd. Non-cytotoxic protein conjugates
WO2019126502A1 (en) * 2017-12-20 2019-06-27 Allergan, Inc. Botulinum toxin cell binding domain polypeptides and methods of use for skin rejuvenation
US11248219B2 (en) 2012-08-27 2022-02-15 Ipsen Bioinnovation Limited Fusion proteins comprising a non-cytotoxic protease, a translocation domain, and a targeting moiety that binds a galanin receptor and methods for treating, preventing or ameliorating pain using such fusion proteins

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7214787B1 (en) * 1993-09-21 2007-05-08 United States Of America As Represented By The Secretary Of The Army Recombinant vaccine against botulinum neurotoxin
GB9617671D0 (en) * 1996-08-23 1996-10-02 Microbiological Res Authority Recombinant toxin fragments
US8012491B2 (en) * 1996-08-23 2011-09-06 Syntaxin, Ltd. Recombinant toxin fragments
US6776990B2 (en) * 1999-04-08 2004-08-17 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
DE60032367T3 (en) 1999-08-25 2011-03-10 Allergan, Inc., Irvine ACTIVE RECOMBINANT NEUROTOXINE
GB9922554D0 (en) * 1999-09-23 1999-11-24 Microbiological Res Authority Inhibition of secretion from non-neuronal cells
AU2005227383B2 (en) * 1999-09-23 2008-08-21 Ipsen Bioinnovation Limited Inhibition of secretion from non-neuronal cells
US20080038274A1 (en) 1999-09-23 2008-02-14 Foster Keith A Inhibition of secretion from non-neuronal cells
US7273722B2 (en) * 2000-11-29 2007-09-25 Allergan, Inc. Neurotoxins with enhanced target specificity
US6936551B2 (en) * 2002-05-08 2005-08-30 Applied Materials Inc. Methods and apparatus for E-beam treatment used to fabricate integrated circuit devices
US8168233B2 (en) * 2002-10-18 2012-05-01 Amylin Pharmaceuticals, Inc. Treatment of pancreatitis with amylin
US20040226556A1 (en) 2003-05-13 2004-11-18 Deem Mark E. Apparatus for treating asthma using neurotoxin
US20050169942A1 (en) * 2003-10-07 2005-08-04 Allergan, Inc. Novel DNA sequences of the botulinum neurotoxin complex of Clostridium botulinum type A-Hall (Allergan) strain for production of therapeutics
US7381698B2 (en) * 2003-12-12 2008-06-03 Chirhoclin, Inc. Methods for treatment of acute pancreatitis
US7947285B2 (en) * 2003-12-12 2011-05-24 Fein Seymour H Methods for preventing post endoscopic retrograde cholangiopancreatography pancreatitis
US7514088B2 (en) * 2005-03-15 2009-04-07 Allergan, Inc. Multivalent Clostridial toxin derivatives and methods of their use
US7811584B2 (en) * 2004-06-30 2010-10-12 Allergan, Inc. Multivalent clostridial toxins
DE102004035606A1 (en) * 2004-07-22 2006-03-30 Biotecon Therapeutics Gmbh Carrier for drugs for obtaining oral bioavailability
US7659092B2 (en) * 2004-12-01 2010-02-09 Syntaxin, Ltd. Fusion proteins
AU2006227816B2 (en) * 2005-03-15 2012-04-05 Allergan, Inc. Modified clostridial toxins with enhanced targeting capabilities for endogenous clostridial toxin receptor systems
US8021859B2 (en) * 2005-03-15 2011-09-20 Allergan, Inc. Modified clostridial toxins with altered targeting capabilities for clostridial toxin target cells
US10052465B2 (en) 2005-07-22 2018-08-21 The Foundry, Llc Methods and systems for toxin delivery to the nasal cavity
US7655243B2 (en) 2005-07-22 2010-02-02 The Foundry, Llc Methods and systems for toxin delivery to the nasal cavity
EP1906923B1 (en) 2005-07-22 2018-01-24 The Foundry, LLC Systems and methods for delivery of a therapeutic agent
ATE463506T1 (en) * 2005-09-19 2010-04-15 Allergan Inc CLOSTRIDIAL TOXINS ACTIVATED WITH CLOSTRIDIAL TOXINS
US8168206B1 (en) 2005-10-06 2012-05-01 Allergan, Inc. Animal protein-free pharmaceutical compositions
EP2001902B1 (en) * 2006-03-14 2013-03-27 Allergan, Inc. Modified clostridial toxins with altered targeting capabilities for clostridial toxin target cells
AU2007347781B2 (en) 2006-07-11 2013-10-03 Allergan, Inc. Modified clostridial toxins with enhanced translocation capability and enhanced targeting activity
EP2038298A2 (en) * 2006-07-11 2009-03-25 Allergan, Inc. Modified clostridial toxins with enhanced translocation capabilities and altered targeting activity for clostridial toxin target cells
US8483831B1 (en) 2008-02-15 2013-07-09 Holaira, Inc. System and method for bronchial dilation
CA2723806C (en) 2008-05-09 2022-05-31 Innovative Pulmonary Solutions, Inc. Systems, assemblies, and methods for treating a bronchial tree
EP2310028B1 (en) 2008-06-12 2016-11-16 Ipsen Bioinnovation Limited Fusion proteins for use in the treatment of acromegaly
WO2009150470A2 (en) 2008-06-12 2009-12-17 Syntaxin Limited Suppression of cancers
GB0820970D0 (en) 2008-11-17 2008-12-24 Syntaxin Ltd Suppression of cancer
RU2535003C2 (en) 2008-12-10 2014-12-10 Аллерган, Инк. Pharmaceutical compositions of clostridia toxin
MX2011009606A (en) 2009-03-13 2011-09-28 Allergan Inc Immuno-based retargeted endopeptidase activity assays.
US8440204B2 (en) * 2009-04-30 2013-05-14 Wisconsin Alumni Research Foundation Subtype of Closteridium botulinum neurotoxin type A and uses thereof
EP2467477B1 (en) 2009-08-17 2014-05-07 East Carolina University Fast acting snare-cleaving enzymes
US20120196935A1 (en) * 2009-09-18 2012-08-02 The Regents Of The University Of California Methods for detecting autodigestion
EP4193948A1 (en) 2009-10-27 2023-06-14 Nuvaira, Inc. Delivery devices with coolable energy emitting assemblies
US8911439B2 (en) 2009-11-11 2014-12-16 Holaira, Inc. Non-invasive and minimally invasive denervation methods and systems for performing the same
EP4111995A1 (en) 2009-11-11 2023-01-04 Nuvaira, Inc. Device for treating tissue and controlling stenosis
CN102753681A (en) 2009-12-16 2012-10-24 阿勒根公司 Modified clostridial toxins comprising an integrated protease cleavage site-binding domain
KR101930962B1 (en) 2010-01-25 2018-12-19 알러간, 인코포레이티드 Methods of intracellular conversion of single-chain proteins into their di-chain form
CA2788544C (en) 2010-02-18 2019-03-05 Effimune Anti-cd28 humanized antibodies
ES2600463T3 (en) 2010-05-20 2017-02-09 Allergan, Inc. Degradable Clostridial Toxins
CA2813467A1 (en) 2010-10-02 2012-04-05 The Regents Of The University Of California Minimizing intestinal dysfunction
US9005628B2 (en) 2012-10-04 2015-04-14 Dublin City University Biotherapy for pain
US9398933B2 (en) 2012-12-27 2016-07-26 Holaira, Inc. Methods for improving drug efficacy including a combination of drug administration and nerve modulation
GB201312317D0 (en) 2013-07-09 2013-08-21 Syntaxin Ltd Cationic neurotoxins
US9216210B2 (en) 2013-12-23 2015-12-22 Dublin City University Multiprotease therapeutics for chronic pain
US10647750B2 (en) 2015-01-09 2020-05-12 Ipsen Bioinnovation Limited Cationic neurotoxins
GB201517450D0 (en) 2015-10-02 2015-11-18 Ipsen Biopharm Ltd Method
EP3263710A1 (en) 2016-07-01 2018-01-03 Ipsen Biopharm Limited Production of activated clostridial neurotoxins
ES2755815T3 (en) 2016-09-13 2020-04-23 Allergan Inc Stabilized non-protein Clostridium toxin compositions
GB201815817D0 (en) 2018-09-28 2018-11-14 Ispen Biopharm Ltd Clostridial neurotoxins comprising and exogenous activation loop
US20220017577A1 (en) * 2018-11-21 2022-01-20 The Regents Of The University Of Colorado, A Body Corporate Proteins for Blocking Neurotransmitter Release
GB201900621D0 (en) 2019-01-16 2019-03-06 Ipsen Biopharm Ltd Labelled polypeptides
GB201914034D0 (en) 2019-09-30 2019-11-13 Ipsen Biopharm Ltd Treatment of neurological disorders
GB202100566D0 (en) 2021-01-15 2021-03-03 Ipsen Biopharm Ltd Treatment of brain damage
GB202104294D0 (en) 2021-03-26 2021-05-12 Ipsen Biopharm Ltd Clostridial neurotoxins comprising an exogenous activation loop
EP4297773A1 (en) 2021-03-30 2024-01-03 Ipsen Biopharm Limited Catalytically inactive clostridial neurotoxins for the treatment of pain & inflammatory disorders
JP2024513191A (en) 2021-03-30 2024-03-22 イプセン バイオファーム リミテッド Treatment of pain and inflammatory disorders
GB202116795D0 (en) 2021-11-22 2022-01-05 Ipsen Biopharm Ltd Treatment of visceral pain
WO2023105289A1 (en) 2021-12-06 2023-06-15 Dublin City University Methods and compositions for the treatment of pain
GB202214232D0 (en) 2022-09-28 2022-11-09 Ispen Biopharm Ltd Clostridial neurotoxins comprising an activating exogenous protease cleavage site
GB202214229D0 (en) 2022-09-28 2022-11-09 Ipsen Biopharm Ltd Clostridial neurotoxins comprising an activating endosomal protease cleavage site

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919665A (en) * 1989-10-31 1999-07-06 Ophidian Pharmaceuticals, Inc. Vaccine for clostridium botulinum neurotoxin
US6875594B2 (en) * 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US14795A (en) * 1856-05-06 Peters
US831435A (en) * 1906-05-19 1906-09-18 Charles Joseph Hohmann Non-refillable bottle.
IE902295A1 (en) 1989-07-07 1991-01-16 Abbott Lab Amino acid analog cck antagonists
US6203794B1 (en) 1994-05-31 2001-03-20 Allergan Sales, Inc. Modification of clostridial toxins for use as transport proteins
GB9508204D0 (en) 1995-04-21 1995-06-07 Speywood Lab Ltd A novel agent able to modify peripheral afferent function
GB9617671D0 (en) 1996-08-23 1996-10-02 Microbiological Res Authority Recombinant toxin fragments
US6776990B2 (en) * 1999-04-08 2004-08-17 Allergan, Inc. Methods and compositions for the treatment of pancreatitis

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919665A (en) * 1989-10-31 1999-07-06 Ophidian Pharmaceuticals, Inc. Vaccine for clostridium botulinum neurotoxin
US6875594B2 (en) * 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110028691A1 (en) * 2002-09-12 2011-02-03 The Health Protection Agency Recombinant toxin fragments
US9006395B2 (en) * 2002-09-12 2015-04-14 The Secretary Of State For Health Recombinant toxin fragments
US20070179281A1 (en) * 2003-04-11 2007-08-02 Atassi M Zouhair BoNT/A Peptides and Methods of Predicting and Reducing Immunoresistance to Botulinum Toxin Therapy
US7635484B2 (en) * 2003-04-11 2009-12-22 Allergan, Inc. BoNT/A peptides and methods of predicting and reducing immunoresistance to botulinum toxin therapy
US8603779B2 (en) 2004-12-01 2013-12-10 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8778634B2 (en) 2004-12-01 2014-07-15 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US20110027256A1 (en) * 2004-12-01 2011-02-03 Syntaxin Ltd. Fusion proteins
US10619146B2 (en) 2004-12-01 2020-04-14 Ipsen Bioinnovation Limited Non-cytotoxic protein conjugates
US20110177053A1 (en) * 2004-12-01 2011-07-21 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8067200B2 (en) 2004-12-01 2011-11-29 Syntaxin Ltd. Fusion proteins
US8187834B2 (en) 2004-12-01 2012-05-29 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8399401B2 (en) 2004-12-01 2013-03-19 Syntaxin, Ltd. Fusion proteins
US8399400B2 (en) 2004-12-01 2013-03-19 Syntaxin, Ltd. Fusion proteins
US8512984B2 (en) 2004-12-01 2013-08-20 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US9474807B2 (en) 2004-12-01 2016-10-25 Ipsen Bioinnovation Limited Non-cytotoxic protein conjugates
US20100247509A1 (en) * 2004-12-01 2010-09-30 Keith Foster Fusion Proteins
US8940870B2 (en) 2004-12-01 2015-01-27 Syntaxin, Ltd. Fusion proteins
US20090162341A1 (en) * 2004-12-01 2009-06-25 Keith Foster Non-Cytotoxic Protein Conjugates
US9012195B2 (en) 2004-12-01 2015-04-21 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US9139635B2 (en) 2004-12-01 2015-09-22 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US9072736B2 (en) 2006-06-01 2015-07-07 Allergan, Inc. Treatment of pain
US9243301B2 (en) 2006-06-01 2016-01-26 Allergan, Inc. Fusion proteins
US20100034802A1 (en) * 2006-06-01 2010-02-11 Syntaxin Limited Treatment of pain
WO2009131435A1 (en) * 2008-04-23 2009-10-29 Erasmus University Medical Center Rotterdam Linker containing bungarotoxin and a binding peptide
US11248219B2 (en) 2012-08-27 2022-02-15 Ipsen Bioinnovation Limited Fusion proteins comprising a non-cytotoxic protease, a translocation domain, and a targeting moiety that binds a galanin receptor and methods for treating, preventing or ameliorating pain using such fusion proteins
WO2019126502A1 (en) * 2017-12-20 2019-06-27 Allergan, Inc. Botulinum toxin cell binding domain polypeptides and methods of use for skin rejuvenation
WO2019126542A1 (en) * 2017-12-20 2019-06-27 Allergan, Inc. Botulinum toxin cell binding domain polypeptides and methods of use for treatments of fibrosis associated disorders
US11149262B2 (en) 2017-12-20 2021-10-19 Alleergan, Inc. Botulinum toxin cell binding domain polypeptides and methods of use for skin rejuvenation

Also Published As

Publication number Publication date
US20010018049A1 (en) 2001-08-30
US20040176299A1 (en) 2004-09-09
WO2000061192A3 (en) 2001-03-08
US20080025994A1 (en) 2008-01-31
US6843998B1 (en) 2005-01-18
US7276473B2 (en) 2007-10-02
AU4205100A (en) 2000-11-14
WO2000061192A2 (en) 2000-10-19
US6776990B2 (en) 2004-08-17

Similar Documents

Publication Publication Date Title
US6843998B1 (en) Methods and compositions for the treatment of pancreatitis
JP7402916B2 (en) Novel botulinum neurotoxin and its derivatives
AU2007226657B2 (en) Multivalent Clostridial toxins
US7811584B2 (en) Multivalent clostridial toxins
US8273865B2 (en) Multivalent clostridial toxins
ES2562425T3 (en) Therapeutic fusion proteins
TW201130974A (en) Modified clostridial toxins comprising an integrated protease cleavage site-binding domain
AU2005205597A1 (en) Chimera botulinum toxin type E
US20200407702A1 (en) Botulinum Neurotoxin Biohybrid
US20030147895A1 (en) Constructs for delivery of threrapeutic agents to neuronal cells
US11118170B2 (en) Engineered clostridium botulinum toxin adapted to deliver molecules into selected cells
KR102353086B1 (en) Novel Method for Preparing Immunotoxin
A Wilson et al. Cargo-delivery platforms for targeted delivery of inhibitor cargos against botulism
Saelinger Receptors for bacterial toxins
Foran 9 Use of Neurotoxins as Drug Delivery Systems

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION