US20100034802A1 - Treatment of pain - Google Patents

Treatment of pain Download PDF

Info

Publication number
US20100034802A1
US20100034802A1 US12/303,078 US30307807A US2010034802A1 US 20100034802 A1 US20100034802 A1 US 20100034802A1 US 30307807 A US30307807 A US 30307807A US 2010034802 A1 US2010034802 A1 US 2010034802A1
Authority
US
United States
Prior art keywords
protease
seq
fusion
use according
nociceptin
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
US12/303,078
Other languages
English (en)
Inventor
Keith Foster
John Chaddock
Philip Marks
Patrick Stancombe
K. Roger Aoki
Joseph Francis
Lance Steward
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.)
Syntaxin Ltd
Allergan Inc
Original Assignee
Syntaxin Ltd
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 Syntaxin Ltd, Allergan Inc filed Critical Syntaxin Ltd
Assigned to ALLERGAN, INC. reassignment ALLERGAN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STEWARD, LANCE E., AOKI, KEI ROGER, FRANICS, JOSEPH
Assigned to ALLERGAN, INC. reassignment ALLERGAN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FRANCIS, JOSEPH, AOKI, KEI ROGER, STEWARD, LANCE
Assigned to SYNTAXIN LIMITED reassignment SYNTAXIN LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHADDOCK, JOHN, FOSTER, KEITH, MARKS, PHILIP, STANCOMBE, PATRICK
Publication of US20100034802A1 publication Critical patent/US20100034802A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/24Metalloendopeptidases (3.4.24)
    • C12Y304/24069Bontoxilysin (3.4.24.69), i.e. botulinum neurotoxin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/52Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from bacteria or Archaea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • A61K38/4893Botulinum neurotoxin (3.4.24.69)
    • 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
    • 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
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/665Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans derived from pro-opiomelanocortin, pro-enkephalin or pro-dynorphin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/055Fusion polypeptide containing a localisation/targetting motif containing a signal for localisation to secretory granules (for exocytosis)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/06Fusion polypeptide containing a localisation/targetting motif containing a lysosomal/endosomal localisation signal
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site

Definitions

  • This invention relates to the use of non-cytotoxic fusion proteins for the treatment of specific types of pain.
  • Toxins may be generally divided into two groups according to the type of effect that they have on a target cell.
  • the first group of toxins kill their natural target cells, and are therefore known as cytotoxic toxin molecules.
  • This group of toxins is exemplified inter alia by plant toxins such as ricin, and abrin, and by bacterial toxins such as diphtheria toxin, and Pseudomonas exotoxin A.
  • Cytotoxic toxins have attracted much interest in the design of “magic bullets” (e.g. immunoconjugates, which comprise a cytotoxic toxin component and an antibody that binds to a specific marker on a target cell) for the treatment of cellular disorders and conditions such as cancer. Cytotoxic toxins typically kill their target cells by inhibiting the cellular process of protein synthesis.
  • Non-cytotoxic toxins do not (as their name confirms) kill their natural target cells.
  • Non-cytotoxic toxins have attracted much less commercial interest than have their cytotoxic counterparts, and exert their effects on a target cell by inhibiting cellular processes other than protein synthesis.
  • Non-cytotoxic toxins are produced by a variety of plants, and by a variety of microorganisms such as Clostridium sp. and Neisseria sp.
  • Clostridial neurotoxins are proteins that typically have a molecular mass of the order of 150 kDa. They are produced by various species of bacteria, especially of the genus Clostridium , most importantly C. tetani and several strains of C. botulinum, C. butyricum and C. argentinense . There are at present eight different classes of the clostridial neurotoxin, namely: tetanus toxin, and botulinum neurotoxin in its serotypes A, B, C1, D, E, F and G, and they all share similar structures and modes of action.
  • Clostridial neurotoxins represent a major group of non-cytotoxic toxin molecules, and are synthesised by the host bacterium as single polypeptides that are modified post-translationally by a proteolytic cleavage event to form two polypeptide chains joined together by a disulphide bond.
  • the two chains are termed the heavy chain (H-chain), which has a molecular mass of approximately 100 kDa, and the light chain (L-chain), which has a molecular mass of approximately 50 kDa.
  • L-chains possess a protease function (zinc-dependent endopeptidase activity) and exhibit a high substrate specificity for vesicle and/or plasma membrane associated proteins involved in the exocytic process.
  • L-chains from different clostridial species or serotypes may hydrolyse different but specific peptide bonds in one of three substrate proteins, namely synaptobrevin, syntaxin or SNAP-25. These substrates are important components of the neurosecretory machinery.
  • Neisseria sp. most importantly from the species N. gonorrhoeae , produce functionally similar non-cytotoxic proteases.
  • An example of such a protease is IgA protease (see WO99/58571).
  • toxin molecules may be re-targeted to a cell that is not the toxin's natural target cell.
  • the modified toxin is capable of binding to a desired target cell and, following subsequent translocation into the cytosol, is capable of exerting its effect on the target cell.
  • Said re-targeting is achieved by replacing the natural Targeting Moiety (TM) of the toxin with a different TM.
  • the TM is selected so that it will bind to a desired target cell, and allow subsequent passage of the modified toxin into an endosome within the target cell.
  • the modified toxin also comprises a translocation domain to enable entry of the non-cytotoxic protease into the cell cytosol.
  • the translocation domain can be the natural translocation domain of the toxin or it can be a different translocation domain obtained from a microbial protein with translocation activity.
  • TM replacement may be effected by conventional chemical conjugation techniques, which are well known to a skilled person.
  • Chemical conjugation is, however, often imprecise. For example, following conjugation, a TM may become joined to the remainder of the conjugate at more than one attachment site.
  • TM may become joined to the remainder of the modified toxin at an attachment site on the protease component and/or on the translocation component. This is problematic when attachment to only one of said components (preferably at a single site) is desired for therapeutic efficacy.
  • TM replacement may be effected by recombinant preparation of a single polypeptide fusion protein (see WO98/07864).
  • This technique is based on the in vivo bacterial mechanism by which native clostridial neurotoxin (i.e. holotoxin) is prepared, and results in a fusion protein having the following structural arrangement:
  • the TM is placed towards the C-terminal end of the fusion protein.
  • the fusion protein is then activated by treatment with a protease, which cleaves at a site between the protease component and the translocation component.
  • a di-chain protein is thus produced, comprising the protease component as a single polypeptide chain covalently attached (via a disulphide bridge) to another single polypeptide chain containing the translocation component plus TM.
  • This problem is particularly relevant in the context of treating specific types of pain.
  • the present invention addresses one or more of the above-mentioned problems by providing use of a therapeutic molecule for the manufacture of a medicament for the treatment of particular types of pain, wherein the therapeutic molecule is a single chain, polypeptide fusion protein, comprising:
  • protease cleavage site at which site the fusion protein is cleavable by a protease, wherein the protease cleavage site is located between the non-cytotoxic protease or fragment thereof and the Targeting Moiety;
  • TMs require a N-terminal domain for interaction with a binding site on a nociceptive sensory afferent.
  • This problem is particularly acute with TMs that require a specific N-terminus amino acid residue or a specific sequence of amino acid residues including the N-terminus amino acid residue for interaction with a binding site on a nociceptive sensory afferent.
  • the present invention employs non-cytotoxic fusion proteins, wherein the TM component of the fusion includes the relevant binding domain in an intra domain or an amino acid sequence located towards the middle (ie. of the linear peptide sequence) of the TM, or preferably located towards the N-terminus of the TM, or more preferably at or near to the N-terminus.
  • the N-terminal domain is capable of binding to a Binding Site on a nociceptive sensory afferent, and the TM preferably has a requirement for a specific and defined sequence of amino acid residue(s) to be free at its N-terminus.
  • the compounds described here may be used to treat a patient suffering from one or more types of chronic pain including neuropathic pain, inflammatory pain, headache pain, somatic pain, visceral pain, and referred pain.
  • treat means to deal with medically. It includes, for example, administering a compound of the invention to prevent pain or to lessen its severity.
  • Pain means any unpleasant sensory experience, usually associated with a physical disorder.
  • the physical disorder may or may not be apparent to a clinician.
  • Pain is of two types: chronic and acute.
  • An “acute pain” is a pain of short duration having a sudden onset.
  • One type of acute pain for example, is cutaneous pain felt on injury to the skin or other superficial tissues, such as caused by a cut or a burn. Cutaneous nociceptors terminate just below the skin, and due to the high concentration of nerve endings, produce a well-defined, localized pain of short duration.
  • Chronic pain includes neuropathic pain, inflammatory pain, headache pain, somatic pain visceral pain and referred pain.
  • Neuroopathic pain means abnormal sensory input, resulting in discomfort, from the peripheral nervous system, central nervous systems, or both.
  • neuropathic pain can involve persistent, spontaneous pain, as well as allodynia (a painful response to a stimulus that normally is not painful), hyperalgesia (an accentuated response to a painful stimulus that usually causes only a mild discomfort, such as a pin prick), or hyperpathia (where a short discomfort becomes a prolonged severe pain).
  • allodynia a painful response to a stimulus that normally is not painful
  • hyperalgesia an accentuated response to a painful stimulus that usually causes only a mild discomfort, such as a pin prick
  • hyperpathia where a short discomfort becomes a prolonged severe pain
  • Neuropathic pain may be caused by any of the following.
  • a traumatic insult such as, for example, a nerve compression injury (e.g., a nerve crush, a nerve stretch, a nerve entrapment or an incomplete nerve transsection); a spinal cord injury (e.g., a hemisection of the spinal cord); a limb amputation; a contusion; an inflammation (e.g., an inflammation of the spinal cord); or a surgical procedure.
  • a nerve compression injury e.g., a nerve crush, a nerve stretch, a nerve entrapment or an incomplete nerve transsection
  • a spinal cord injury e.g., a hemisection of the spinal cord
  • a limb amputation e.g., a contusion
  • an inflammation e.g., an inflammation of the spinal cord
  • surgical procedure e.g., a surgical procedure.
  • An ischemic event including, for example, a stroke and heart attack.
  • a toxic agent including, for example, a drug, an alcohol, a heavy metal (e.g., lead, arsenic, mercury), an industrial agent (e.g., a solvent, fumes from a glue) or nitrous oxide.
  • a toxic agent including, for example, a drug, an alcohol, a heavy metal (e.g., lead, arsenic, mercury), an industrial agent (e.g., a solvent, fumes from a glue) or nitrous oxide.
  • a disease including, for example, an inflammatory disorder, a neoplastic tumor, an acquired immune deficiency syndrome (AIDS), Lymes disease, a leprosy, a metabolic disease, a peripheral nerve disorder, like neuroma, a mononeuropathy or a polyneuropathy.
  • AIDS acquired immune deficiency syndrome
  • Lymes disease a leprosy
  • a metabolic disease a peripheral nerve disorder, like neuroma, a mononeuropathy or a polyneuropathy.
  • a neuralgia is a pain that radiates along the course of one or more specific nerves usually without any demonstrable pathological change in the nerve structure.
  • the causes of neuralgia are varied. Chemical irritation, inflammation, trauma (including surgery), compression by nearby structures (for instance, tumors), and infections may all lead to neuralgia. In many cases, however, the cause is unknown or unidentifiable.
  • Neuralgia is most common in elderly persons, but it may occur at any age.
  • a neuralgia includes, without limitation, a trigeminal neuralgia, a post-herpetic neuralgia, a postherpetic neuralgia, a glossopharyngeal neuralgia, a sciatica and an atypical facial pain.
  • Neuralgia is pain in the distribution of a nerve or nerves. Examples are trigeminal neuralgia, atypical facial pain, and postherpetic neuralgia (caused by shingles or herpes).
  • the affected nerves are responsible for sensing touch, temperature and pressure in the facial area from the jaw to the forehead.
  • the disorder generally causes short episodes of excruciating pain, usually for less than two minutes and on only one side of the face.
  • the pain can be described in a variety of ways such as “stabbing,” “sharp,” “like lightning,” “burning,” and even “itchy”.
  • the pain can also present as severe or merely aching and last for extended periods.
  • the pain associated with TN is recognized as one the most excruciating pains that can be experienced.
  • Simple stimuli such as eating, talking, washing the face, or any light touch or sensation can trigger an attack (even the sensation of a gentle breeze).
  • the attacks can occur in clusters or as an isolated attack.
  • Symptoms include sharp, stabbing pain or constant, burning pain located anywhere, usually on or near the surface of the body, in the same location for each episode; pain along the path of a specific nerve; impaired function of affected body part due to pain, or muscle weakness due to concomitant motor nerve damage; increased sensitivity of the skin or numbness of the affected skin area (feeling similar to a local anesthetic such as a Novacaine shot); and any touch or pressure is interpreted as pain. Movement may also be painful.
  • Trigeminal neuralgia is the most common form of neuralgia. It affects the main sensory nerve of the face, the trigeminal nerve (“trigeminal” literally means “three origins”, referring to the division of the nerve into 3 branches). This condition involves sudden and short attacks of severe pain on the side of the face, along the area supplied by the trigeminal nerve on that side. The pain attacks may be severe enough to cause a facial grimace, which is classically referred to as a painful tic (tic douloureux). Sometimes, the cause of trigeminal neuralgia is a blood vessel or small tumor pressing on the nerve.
  • disorders such as multiple sclerosis (an inflammatory disease affecting the brain and spinal cord), certain forms of arthritis, and diabetes (high blood sugar) may also cause trigeminal neuralgia, but a cause is not always identified. In this condition, certain movements such as chewing, talking, swallowing, or touching an area of the face may trigger a spasm of excruciating pain.
  • a related but rather uncommon neuralgia affects the glosso-pharyngeal nerve, which provides sensation to the throat. Symptoms of this neuralgia are short, shock-like episodes of pain located in the throat.
  • Neuralgia may occur after infections such as shingles, which is caused by the varicella-zoster virus, a type of herpesvirus. This neuralgia produces a constant burning pain after the shingles rash has healed. The pain is worsened by movement of or contact with the affected area. Not all of those diagnosed with shingles go on to experience postherpetic neuralgia, which can be more painful than shingles. The pain and sensitivity can last for months or even years. The pain is usually in the form of an intolerable sensitivity to any touch but especially light touch. Postherpetic neuralgia is not restricted to the face; it can occur anywhere on the body but usually occurs at the location of the shingles rash.
  • Postherpetic neuralgia may be debilitating long after signs of the original herpes infection have disappeared.
  • Other infectious diseases that may cause neuralgia are syphilis and Lyme disease.
  • Diabetes is another common cause of neuralgia. This very common medical problem affects almost 1 out of every 20 Americans during adulthood. Diabetes damages the tiny arteries that supply circulation to the nerves, resulting in nerve fiber malfunction and sometimes nerve loss. Diabetes can produce almost any neuralgia, including trigeminal neuralgia, carpal tunnel syndrome (pain and numbness of the hand and wrist), and meralgia paresthetica (numbness and pain in the thigh due to damage to the lateral femoral cutaneous nerve). Strict control of blood sugar may prevent diabetic nerve damage and may accelerate recovery in patients who do develop neuralgia.
  • neuralgias Other medical conditions that may be associated with neuralgias are chronic renal insufficiency and porphyria—a hereditary disease in which the body cannot rid itself of certain substances produced after the normal breakdown of blood in the body. Certain drugs may also cause this problem.
  • Deafferentation indicates a loss of the sensory input from a portion of the body, and can be caused by interruption of either peripheral sensory fibres or nerves from the central nervous system.
  • a deafferentation pain syndrome includes, without limitation, an injury to the brain or spinal cord, a post-stroke pain, a phantom pain, a paraplegia, a brachial plexus avulsion injuries, lumbar radiculopathies.
  • CRPS is a chronic pain syndrome resulting from sympathetically-maintained pain, and presents in two forms.
  • CRPS 1 currently replaces the term “reflex sympathetic dystrophy syndrome”. It is a chronic nerve disorder that occurs most often in the arms or legs after a minor or major injury.
  • CRPS 1 is associated with severe pain; changes in the nails, bone, and skin; and an increased sensitivity to touch in the affected limb.
  • CRPS 2 replaces the term causalgia, and results from an identified injury to the nerve.
  • a CRPS includes, without limitation, a CRPS Type I (reflex sympathetic dystrophy) and a CRPS Type II (causalgia).
  • a neuropathy is a functional or pathological change in a nerve and is characterized clinically by sensory or motor neuron abnormalities.
  • Central neuropathy is a functional or pathological change in the central nervous system.
  • Peripheral neuropathy is a functional or pathological change in one or more peripheral nerves.
  • the peripheral nerves relay information from your central nervous system (brain and spinal cord) to muscles and other organs and from your skin, joints, and other organs back to your brain.
  • Peripheral neuropathy occurs when these nerves fail to carry information to and from the brain and spinal cord, resulting in pain, loss of sensation, or inability to control muscles.
  • the failure of nerves that control blood vessels, intestines, and other organs results in abnormal blood pressure, digestion problems, and loss of other basic body processes.
  • Risk factors for neuropathy include diabetes, heavy alcohol use, and exposure to certain chemicals and drugs. Some people have a hereditary predisposition for neuropathy.
  • Prolonged pressure on a nerve is another risk for developing a nerve injury.
  • Pressure injury may be caused by prolonged immobility (such as a long surgical procedure or lengthy illness) or compression of a nerve by casts, splints, braces, crutches, or other devices.
  • Polyneuropathy implies a widespread process that usually affects both sides of the body equally. The symptoms depend on which type of nerve is affected. The three main types of nerves are sensory, motor, and autonomic. Neuropathy can affect any one or a combination of all three types of nerves. Symptoms also depend on whether the condition affects the whole body or just one nerve (as from an injury). The cause of chronic inflammatory polyneuropathy is an abnormal immune response.
  • the specific antigens, immune processes, and triggering factors are variable and in many cases are unknown. It may occur in association with other conditions such as HIV, inflammatory bowel disease, lupus erythematosis, chronic active hepatitis, and blood cell abnormalities.
  • Peripheral neuropathy may involve a function or pathological change to a single nerve or nerve group (monneuropathy) or a function or pathological change affecting multiple nerves (polyneuropathy).
  • Polyneuropathy is a peripheral neuropathy involving the loss of movement or sensation to an area caused by damage or destruction to multiple peripheral nerves.
  • Polyneuropathic pain includes, without limitation, post-polio syndrome, postmastectomy syndrome, diabetic neuropathy, alcohol neuropathy, amyloid, toxins, AIDS, hypothyroidism, uremia, vitamin deficiencies, chemotherapy-induced pain, 2′,3′-didexoycytidine (ddC) treatment, Guillain-Barré syndrome or Fabry's disease.
  • ddC 2′,3′-didexoycytidine
  • Mononeuropathy is a peripheral neuropathy involving loss of movement or sensation to an area caused by damage or destruction to a single peripheral nerve or nerve group. Mononeuropathy is most often caused by damage to a local area resulting from injury or trauma, although occasionally systemic disorders may cause isolated nerve damage (as with mononeuritis multiplex). The usual causes are direct trauma, prolonged pressure on the nerve, and compression of the nerve by swelling or injury to nearby body structures. The damage includes destruction of the myelin sheath (covering) of the nerve or of part of the nerve cell (the axon). This damage slows or prevents conduction of impulses through the nerve. Mononeuropathy may involve any part of the body.
  • Mononeuropathic pain includes, without limitation, a sciatic nerve dysfunction, a common peroneal nerve dysfunction. a radial nerve dysfunction, an ulnar nerve dysfunction, a cranial mononeuropathy VI, a cranial mononeuropathy VII, a cranial mononeuropathy III (compression type), a cranial mononeuropathy III (diabetic type), an axillary nerve dysfunction, a carpal tunnel syndrome, a femoral nerve dysfunction, a tibial nerve dysfunction, a Bell's palsy, a thoracic outlet syndrome, a carpal tunnel syndrome and a sixth (abducent) nerve palsy
  • peripheral neuropathies are symmetrical, and usually due to various systematic illnesses and disease processes that affect the peripheral nervous system in its entirety. They are further subdivided into several categories:
  • Distal axonopathies are the result of some metabolic or toxic derangement of neurons. They may be caused by metabolic diseases such as diabetes, renal failure, deficiency syndromes such as malnutrition and alcoholism, or the effects of toxins or drugs.
  • Distal axonopathy (aka dying back neuropathy) is a type of peripheral neuropathy that results from some metabolic or toxic derangement of peripheral nervous system (PNS) neurons. It is the most common response of nerves to metabolic or toxic disturbances, and as such may be caused by metabolic diseases such as diabetes, renal failure, deficiency syndromes such as malnutrition and alcoholism, or the effects of toxins or drugs.
  • PNS peripheral nervous system
  • the most common cause of distal axonopathy is diabetes, and the most common distal axonopathy is diabetic neuropathy.
  • Myelinopathies are due to a primary attack on myelin causing an acute failure of impulse conduction.
  • the most common cause is acute inflammatory demyelinating polyneuropathy (AIDP; aka Guillain-Barré syndrome), though other causes include chronic inflammatory demyelinating syndrome (CIDP), genetic metabolic disorders (e.g., leukodystrophy), or toxins.
  • CIDP chronic inflammatory demyelinating syndrome
  • Myelinopathy is due to primary destruction of myelin or the myelinating Schwann cells, which leaves the axon intact, but causes an acute failure of impulse conduction. This demyelination slows down or completely blocks the conduction of electrical impulses through the nerve.
  • ADP acute inflammatory demyelinating polyneuropathy
  • CIDP chronic inflammatory demyelinating polyneuropathy
  • genetic metabolic disorders e.g., leukodystrophy or Charcot-Marie-Tooth disease
  • Neuronopathies are the result of destruction of peripheral nervous system (PNS) neurons. They may be caused by motor neurone diseases, sensory neuronopathies (e.g., Herpes zoster), toxins or autonomic dysfunction. Neurotoxins may cause neuronopathies, such as the chemotherapy agent vincristine. Neuronopathy is dysfunction due to damage to neurons of the peripheral nervous system (PNS), resulting in a peripheral neuropathy. It may be caused by motor neurone diseases, sensory neuronopathies (e.g., Herpes zoster), toxic substances or autonomic dysfunction. A person with neuronopathy may present in different ways, depending on the cause, the way it affects the nerve cells, and the type of nerve cell that is most affected.
  • PNS peripheral nervous system
  • a person with neuronopathy may present in different ways, depending on the cause, the way it affects the nerve cells, and the type of nerve cell that is most affected.
  • Focal entrapment neuropathies e.g., carpal tunnel syndrome
  • the compounds of the invention may be used to treat pain caused by or otherwise associated with any of the following inflammatory conditions
  • Arthritic disorders include, for example, a rheumatoid arthritis; a juvenile rheumatoid arthritis; a systemic lupus erythematosus (SLE); a gouty arthritis; a scleroderma; an osteoarthritis; a psoriatic arthritis; an ankylosing spondylitis; a Reiter's syndrome (reactive arthritis); an adult Still's disease; an arthritis from a viral infection; an arthritis from a bacterial infection, such as, e.g., a gonococcal arthritis and a non-gonococcal bacterial arthritis (septic arthritis); a Tertiary Lyme disease; a tuberculous arthritis; and an arthritis from a fungal infection, such as, e,g. a blastomycosis
  • Autoimmune diseases include, for example, a Guillain-Barré syndrome, a Hashimoto's thyroiditis, a pernicious anemia, an Addison's disease, a type I diabetes, a systemic lupus erythematosus, a dermatomyositis, a Sjogren's syndrome, a lupus erythematosus, a multiple sclerosis, a myasthenia gravis, a Reiter's syndrome and a Grave's disease.
  • Connective tissue disorders include, for example, a spondyloarthritis a dermatomyositis, and a fibromyalgia.
  • Inflammation caused by injury including, for example, a crush, puncture, stretch of a tissue or joint, may cause chronic inflammatory pain.
  • Inflammation caused by infection including, for example, a tuberculosis or an interstitial keratitis may cause chronic inflammatory pain.
  • Neuritis is an inflammatory process affecting a nerve or group of nerves. Symptoms depend on the nerves involved, but may include pain, paresthesias, paresis, or hypesthesia (numbness).
  • Inflammation of the joint such as that caused by bursitis or tendonitis, for example, may cause chronic inflammatory pain.
  • the compounds of the invention may be used to treat pain caused by or otherwise associated with any of the following headache conditions.
  • a headache (medically known as cephalgia) is a condition of mild to severe pain in the head; sometimes neck or upper back pain may also be interpreted as a headache. It may indicate an underlying local or systemic disease or be a disorder in itself.
  • Muscular/myogenic headaches appear to involve the tightening or tensing of facial and neck muscles; they may radiate to the forehead. Tension headache is the most common form of myogenic headache.
  • a tension headache is a condition involving pain or discomfort in the head, scalp, or neck, usually associated with muscle tightness in these areas. Tension headaches result from the contraction of neck and scalp muscles. One cause of this muscle contraction is a response to stress, depression or anxiety. Any activity that causes the head to be held in one position for a long time without moving can cause a headache. Such activities include typing or use of computers, fine work with the hands, and use of a microscope. Sleeping in a cold room or sleeping with the neck in an abnormal position may also trigger this type of headache.
  • a tension-type headache includes, without limitation, an episodic tension headache and a chronic tension headache.
  • vascular headache The most common type of vascular headache is migraine.
  • Other kinds of vascular headaches include cluster headaches, which cause repeated episodes of intense pain, and headaches resulting from high blood pressure
  • a migraine is a heterogeneous disorder that generally involves recurring headaches.
  • Migraines are different from other headaches because they occur with other symptoms, such as, e.g., nausea, vomiting, or sensitivity to light. In most people, a throbbing pain is felt only on one side of the head.
  • Clinical features such as type of aura symptoms, presence of prodromes, or associated symptoms such as vertigo, may be seen in subgroups of patients with different underlying pathophysiological and genetic mechanisms.
  • a migraine headache includes, without limitation, a migraine without aura (common migraine), a migraine with aura (classic migraine), a menstrual migraine, a migraine equivalent (acephalic headache), a complicated migraine, an abdominal migraine and a mixed tension migraine.
  • Cluster headaches affect one side of the head (unilateral) and may be associated with tearing of the eyes and nasal congestion. They occurs in clusters, happening repeatedly every day at the same time for several weeks and then remitting.
  • Rebound headaches also known as medication overuse headaches, occur when medication is taken too frequently to relieve headache. Rebound headaches frequently occur daily and can be very painful.
  • Sinusitis is inflammation, either bacterial, fungal, viral, allergic or autoimmune, of the paranasal sinuses.
  • Chronic sinusitis is one of the most common complications of the common cold. Symptoms include: Nasal congestion; facial pain; headache; fever; general malaise; thick green or yellow discharge; feeling of facial ‘fullness’ worsening on bending over. In a small number of cases, chronic maxillary sinusitis can also be brought on by the spreading of bacteria from a dental infection. Chronic hyperplastic eosinophilic sinusitis is a noninfective form of chronic sinusitis.
  • Ital headaches are headaches associated with seizure activity.
  • the compounds of the invention may be used to treat pain caused by or otherwise associated with any of the following somatic pain conditions.
  • Somatic pain originates from ligaments, tendons, bones, blood vessels, and even nerves themselves. It is detected with somatic nociceptors.
  • the scarcity of pain receptors in these areas produces a dull, poorly-localized pain of longer duration than cutaneous pain; examples include sprains and broken bones. Additional examples include the following.
  • Excessive muscle tension can be caused, for example, by a sprain or a strain.
  • Repetitive motion disorders can result from overuse of the hands, wrists, elbows, shoulders, neck, back, hips, knees, feet, legs, or ankles.
  • Muscle disorders causing somatic pain include, for example, a polymyositis, a dermatomyositis, a lupus, a fibromyalgia, a polymyalgia rheumatica, and a rhabdomyolysis.
  • Myalgia is muscle pain and is a symptom of many diseases and disorders. The most common cause for myalgia is either overuse or over-stretching of a muscle or group of muscles. Myalgia without a traumatic history is often due to viral infections. Longer-term myalgias may be indicative of a metabolic myopathy, some nutritional deficiencies or chronic fatigue syndrome.
  • Infection can cause somatic pain.
  • infections include, for example, an abscess in the muscle, a trichinosis, an influenza, a Lyme disease, a malaria, a Rocky Mountain spotted fever, Avian influenza, the common cold, community-acquired pneumonia, meningitis, monkeypox, Severe Acute Respiratory Syndrome, toxic shock syndrome, trichinosis, typhoid fever, and upper respiratory tract infection.
  • Drugs can cause somatic pain.
  • Such drugs include, for example, cocaine, a statin for lowering cholesterol (such as atorvastatin, simvastatin, and lovastatin), and an ACE inhibitor for lowering blood pressure (such as enalapril and captopril)
  • the compounds of the invention may be used to treat pain caused by or otherwise associated with any of the following visceral pain conditions.
  • Visceral pain originates from body's viscera, or organs.
  • Visceral nociceptors are located within body organs and internal cavities. The even greater scarcity of nociceptors in these areas produces pain that is usually more aching and of a longer duration than somatic pain.
  • Visceral pain is extremely difficult to localise, and several injuries to visceral tissue exhibit “referred” pain, where the sensation is localised to an area completely unrelated to the site of injury. Examples of visceral pain include the following.
  • Functional visceral pain includes, for example, an irritable bowel syndrome and a chronic functional abdominal pain (CFAP), a functional constipation and a functional dyspepsia, a non-cardiac chest pain (NCCP) and a chronic abdominal pain.
  • CFAP chronic functional abdominal pain
  • NCCP non-cardiac chest pain
  • Chronic gastrointestinal inflammation includes, for example, a gastritis, an inflammatory bowel disease, like, e.g., a Crohn's disease, an ulcerative colitis, a microscopic colitis, a diverticulitis and a gastroenteritis; an interstitial cystitis; an intestinal ischemia; a cholecystitis; an appendicitis; a gastroesophageal reflux; an ulcer, a nephrolithiasis, an urinary tract infection, a pancreatitis and a hernia.
  • a gastritis an inflammatory bowel disease, like, e.g., a Crohn's disease, an ulcerative colitis, a microscopic colitis, a diverticulitis and a gastroenteritis
  • an interstitial cystitis an intestinal ischemia
  • a cholecystitis cholecystitis
  • an appendicitis a gastroesophageal reflux
  • Autoimmune pain includes, for example, a sarcoidosis and a vasculitis.
  • Organic visceral pain includes, for example, pain resulting from a traumatic, inflammatory or degenerative lesion of the gut or produced by a tumor impinging on sensory innervation.
  • Treatment-induced visceral pain includes, for example, a pain attendant to chemotherapy therapy or a pain attendant to radiation therapy.
  • the compounds of the invention may be used to treat pain caused by or otherwise associated with any of the following referred pain conditions.
  • Referred pain arises from pain localized to an area separate from the site of pain stimulation. Often, referred pain arises when a nerve is compressed or damaged at or near its origin. In this circumstance, the sensation of pain will generally be felt in the territory that the nerve serves, even though the damage originates elsewhere.
  • a common example occurs in intervertebral disc herniation, in which a nerve root arising from the spinal cord is compressed by adjacent disc material. Although pain may arise from the damaged disc itself, pain will also be felt in the region served by the compressed nerve (for example, the thigh, knee, or foot). Relieving the pressure on the nerve root may ameliorate the referred pain, provided that permanent nerve damage has not occurred.
  • Myocardial ischaemia (the loss of blood flow to a part of the heart muscle tissue) is possibly the best known example of referred pain; the sensation can occur in the upper chest as a restricted feeling, or as an ache in the left shoulder, arm or even hand.
  • the non-cytotoxic protease component of the present invention is a non-cytotoxic protease, or a fragment thereof, which protease or protease fragment is capable of cleaving different but specific peptide bonds in one of three substrate proteins, namely synaptobrevin, syntaxin or SNAP-25, of the exocytic fusion apparatus in a nociceptive sensory afferent. These substrates are important components of the neurosecretory machinery.
  • the non-cytotoxic protease component of the present invention is preferably a neisserial IgA protease or a fragment thereof or a clostridial neurotoxin L-chain or a fragment thereof.
  • a particularly preferred non-cytotoxic protease component is a botulinum neurotoxin (BoNT) L-chain or a fragment thereof.
  • the translocation component of the present invention enables translocation of the non-cytotoxic protease (or fragment thereof) into the target cell such that functional expression of protease activity occurs within the cytosol of the target cell.
  • the translocation component is preferably capable of forming ion-permeable pores in lipid membranes under conditions of low pH. Preferably it has been found to use only those portions of the protein molecule capable of pore-formation within the endosomal membrane.
  • the translocation component may be obtained from a microbial protein source, in particular from a bacterial or viral protein source.
  • the translocation component is a translocating domain of an enzyme, such as a bacterial toxin or viral protein.
  • the translocation component of the present invention is preferably a clostridial neurotoxin H-chain or a fragment thereof. Most preferably it is the H N domain (or a functional component thereof), wherein H N means a portion or fragment of the H-chain of a clostridial neurotoxin approximately equivalent to the amino-terminal half of the H-chain, or the domain corresponding to that fragment in the intact H-chain.
  • the TM component of the present invention is responsible for binding the fusion protein of the present invention to a Binding Site on a target cell.
  • the TM component is simply a ligand through which a fusion protein of the present invention binds to a selected target cell.
  • the target cell is a nociceptive sensory afferent, preferably a primary nociceptive afferent (e.g. an A-fibre such as an A6-fibre or a C-fibre).
  • a primary nociceptive afferent e.g. an A-fibre such as an A6-fibre or a C-fibre.
  • the fusion proteins of the present invention are capable of inhibiting neurotransmitter or neuromodulator [e.g. glutamate, substance P, calcitonin-gene related peptide (CGRP), and/or neuropeptide Y] release from discrete populations of nociceptive sensory afferent neurons.
  • the fusion proteins reduce or prevent the transmission of sensory afferent signals (e.g. neurotransmitters or neuromodulators) from peripheral to central pain fibres, and therefore have application as therapeutic molecules for the treatment of pain, in particular chronic pain.
  • a TM binds to a nociceptive sensory afferent.
  • tissue or cells representative of the nociceptive sensory afferent for example DRGs
  • labelled e.g. tritiated
  • the relative proportions of non-specific and specific binding may be assessed, thereby allowing confirmation that the ligand binds to the nociceptive sensory afferent target cell.
  • the assay may include one or more binding antagonists, and the assay may further comprise observing a loss of ligand binding. Examples of this type of experiment can be found in Hulme, E. C. (1990), Receptor-binding studies, a brief outline, pp. 303-311, In Receptor biochemistry, A Practical Approach, Ed. E. C. Hulme, Oxford University Press.
  • the fusion proteins of the present invention generally demonstrate a reduced binding affinity (in the region of up to 100-fold) for nociceptive sensory afferent target cells when compared with the corresponding ‘free’ TM.
  • the fusion proteins of the present invention surprisingly demonstrate good efficacy. This can be attributed to two principal features. First, the non-cytotoxic protease component is catalytic—thus, the therapeutic effect of a few such molecules is rapidly amplified. Secondly, the receptors present on the nociceptive sensory afferents need only act as a gateway for entry of the therapeutic, and need not necessarily be stimulated to a level required in order to achieve a ligand-receptor mediated pharmacological response.
  • the fusion proteins of the present invention may be administered at a dosage that is much lower that would be employed for other types of analgesic molecules such as NSAIDS, morphine, and gabapentin.
  • the latter molecules are typically administered at high microgram to milligram (even up to hundreds of milligram) quantities, whereas the fusion proteins of the present invention may be administered at much lower dosages, typically at least 10-fold lower, and more typically at 100-fold lower.
  • the TM preferably comprises a maximum of 50 amino acid residues, more preferably a maximum of 40 amino acid residues, particularly preferably a maximum of 30 amino acid residues, and most preferably a maximum of 20 amino acid residues.
  • Opioids represent a preferred group of TMs of the present invention.
  • enkephalins metal and leu
  • endomorphins 1 and 2 ⁇ -endorphin
  • dynorphin ⁇ -endorphin
  • Opioid peptides are frequently used in the clinic to modify the activity to nociceptors, and other cells involved in the pain response.
  • opioids As exemplified by the three-step World Health Organisation Analgesic Ladder, opioids have entry points into the pharmacological treatment of chronic cancer and non-cancer pain at all three stages, underlining their importance to the treatment of pain.
  • Reference to opioids embraces fragments, variants and derivatives thereof, which retain the ability to bind to nociceptive sensory afferents.
  • the TM of the invention can also be a molecule that acts as an “agonist” at one or more of the receptors present on a nociceptive sensory afferent, more particularly on a primary nociceptive afferent.
  • an agonist has been considered any molecule that can either increase or decrease activities within a cell, namely any molecule that simply causes an alteration of cell activity.
  • the conventional meaning of an agonist would include a chemical substance capable of combining with a receptor on a cell and initiating a reaction or activity, or a drug that induces an active response by activating receptors, whether the response is an increase or decrease in cellular activity.
  • an agonist is more specifically defined as a molecule that is capable of stimulating the process of exocytic fusion in a target cell, which process is susceptible to inhibition by a protease (or fragment thereof) capable of cleaving a protein of the exocytic fusion apparatus in said target cell.
  • nerve growth factor is an agonist in respect of its ability to promote neuronal differentiation via binding to a TrkA receptor.
  • NGF nerve growth factor
  • NGF is not an agonist when assessed by the above criteria because it is not a principal inducer of exocytic fusion.
  • the process that NGF stimulates i.e. cell differentiation
  • the target for the TM is the ORL 1 receptor.
  • This receptor is a member of the G-protein-coupled class of receptors, and has a seven transmembrane domain structure.
  • the properties of the ORL 1 receptor are discussed in detail in Mogil & Pasternak (2001), Pharmacological Reviews , Vol. 53, No. 3, pages 381-415.
  • the TM is a molecule that binds (preferably that specifically binds) to the ORL 1 receptor. More preferably, the TM is an “agonist” of the ORL 1 receptor.
  • the term “agonist” in this context is defined as above.
  • Inoue et al. confirm that an intraplantar injection of botulinum neurotoxin type A abolishes the nociceptin-induced responses. Since it is known that BoNT inhibits the release of substance P from primary afferent neurons (Welch et al., 2000, Toxicon, 38, 245-258), this confirms the link between nociceptin-ORL 1 interaction and subsequent release of substance P.
  • a TM can be said to have agonist activity at the ORL 1 receptor if the TM causes an induction in the release of substance P from a nociceptive sensory afferent neuron (see Example 10).
  • the TM is nociceptin—the natural ligand for the ORL 1 receptor.
  • Nociceptin targets the ORL 1 receptor with high affinity.
  • examples of other preferred TMs include:
  • TM demonstrates particularly good binding affinity (when compared with natural nociceptin) for nociceptive sensory afferents. This is surprising as the amino acid modifications occur at a position away from the N-terminus of the TM. Moreover, the modifications are almost at the C-terminus of the TM, which in turn is attached to a large polypeptide sequence (i.e. the translocation domain). Generally speaking, a TM-containing fusion protein will demonstrate an approximate 100-fold reduction in binding ability vis-à-vis the TM per se. The above-mentioned “variant” TM per se demonstrates an approximate 3- to 10-fold increase in binding ability for a nociceptive sensory afferent (e.g.
  • TM-containing fusion via the ORL1 receptor vis-à-vis natural nociceptin.
  • a “variant” TM-containing fusion might be expected to demonstrate an approximate 10-fold reduction in binding ability for a nociceptive sensory afferent (e.g. via the ORL1 receptor) vis-à-vis ‘free’ nociceptin.
  • the present inventors have demonstrated that such “variant” TM-containing fusion proteins demonstrate a binding ability that (most surprisingly) closely mirrors that of ‘free’ nociceptin—see FIG. 14 .
  • opioid or an agonist of the ORL 1 receptor (such as nociceptin, or any one of the peptides listed in the table above) embraces molecules having at least 70%, preferably at least 80%, more preferably at least 90%, and most preferably at least 95% homology with said opioid or agonist.
  • the agonist homologues retain the agonist properties of nociceptin at the ORL 1 receptor, which may be tested using the methods provided in Example 10.
  • an opioid homologue substantially retains the binding function of the opioid with which it shows high homology.
  • the invention also encompasses fragments, variants, and derivatives of any one of the TMs described above. These fragments, variants, and derivatives substantially retain the properties that are ascribed to said TMs.
  • TMs a variety of other polypeptides are suitable for targeting the fusion proteins of the present invention to nociceptive sensory afferents (e.g. to nociceptors).
  • nociceptive sensory afferents e.g. to nociceptors.
  • galanin and derivatives of galanin are found pre- and post-synaptically in DRGs (Liu & Hokfelt, (2002), Trends Pharm. Sci., 23(10), 468-74), and are enhanced in expression during neuropathic pain states.
  • PARs Proteinase-activated receptors
  • PAR-2 Proteinase-activated receptors
  • PAR2 is expressed by primary spinal afferent neurons, and PAR2 agonists stimulate release of substance P (SP) and calcitonin gene-related peptide (CGRP) in peripheral tissues
  • a particularly preferred set of TMs of the present invention includes:
  • the protease cleavage site of the present invention allows cleavage (preferably controlled cleavage) of the fusion protein at a position between the non-cytotoxic protease component and the TM component. It is this cleavage reaction that converts the fusion protein from a single chain polypeptide into a disulphide-linked, di-chain polypeptide.
  • the TM binds via a domain or amino acid sequence that is located away from the C-terminus of the TM.
  • the relevant binding domain may include an intra domain or an amino acid sequence located towards the middle (i.e. of the linear peptide sequence) of the TM.
  • the relevant binding domain is located towards the N-terminus of the TM, more preferably at or near to the N-terminus.
  • the single chain polypeptide fusion may include more than one proteolytic cleavage site. However, where two or more such sites exist, they are different, thereby substantially preventing the occurrence of multiple cleavage events in the presence of a single protease. In another embodiment, it is preferred that the single chain polypeptide fusion has a single protease cleavage site.
  • protease cleavage sequence(s) may be introduced (and/or any inherent cleavage sequence removed) at the DNA level by conventional means, such as by site-directed mutagenesis. Screening to confirm the presence of cleavage sequences may be performed manually or with the assistance of computer software (e.g. the MapDraw program by DNASTAR, Inc.).
  • protease cleavage site Whilst any protease cleavage site may be employed, the following are preferred:
  • protease cleavage site is an intein, which is a self-cleaving sequence.
  • the self-splicing reaction is controllable, for example by varying the concentration of reducing agent present.
  • the protease cleavage site is cleaved and the N-terminal region (preferably the N-terminus) of the TM becomes exposed.
  • the resulting polypeptide has a TM with an N-terminal domain or an intra domain that is substantially free from the remainder of the fusion protein. This arrangement ensures that the N-terminal component (or intra domain) of the TM may interact directly with a Binding Site on a target cell.
  • the TM and the protease cleavage site are distanced apart in the fusion protein by at most 10 amino acid residues, more preferably by at most 5 amino acid residues, and most preferably by zero amino acid residues.
  • a fusion is provided with a TM that has an N-terminal domain that is substantially free from the remainder of the fusion. This arrangement ensures that the N-terminal component of the Targeting Moiety may interact directly with a Binding Site on a target cell.
  • One advantage associated with the above-mentioned activation step is that the TM only becomes susceptible to N-terminal degradation once proteolytic cleavage of the fusion protein has occurred.
  • the selection of a specific protease cleavage site permits selective activation of the polypeptide fusion into a di-chain conformation.
  • Construction of the single-chain polypeptide fusion of the present invention places the protease cleavage site between the TM and the non-cytotoxic protease component.
  • the TM is located between the protease cleavage site and the translocation component. This ensures that the TM is attached to the translocation domain (i.e. as occurs with native clostridial holotoxin), though in the case of the present invention the order of the two components is reversed vis-à-vis native holotoxin.
  • a further advantage with this arrangement is that the TM is located in an exposed loop region of the fusion protein, which has minimal structural effects on the conformation of the fusion protein.
  • said loop is variously referred to as the linker, the activation loop, the inter-domain linker, or just the surface exposed loop (Schiavo et al 2000, Phys. Rev., 80, 717-766; Turton et al., 2002, Trends Biochem. Sci., 27, 552-558).
  • the non-cytotoxic protease component and the translocation component are linked together by a disulphide bond.
  • the polypeptide assumes a di-chain conformation, wherein the protease and translocation components remain linked together by the disulphide bond.
  • the protease and translocation components are distanced apart from one another in the single chain fusion protein by a maximum of 100 amino acid residues, more preferably a maximum of 80 amino acid residues, particularly preferably by a maximum of 60 amino acid residues, and most preferably by a maximum of 50 amino acid residues.
  • the non-cytotoxic protease component forms a disulphide bond with the translocation component of the fusion protein.
  • the amino acid residue of the protease component that forms the disulphide bond is located within the last 20, preferably within the last 10 C-terminal amino acid residues of the protease component.
  • the amino acid residue within the translocation component that forms the second part of the disulphide bond may be located within the first 20, preferably within the first 10 N-terminal amino acid residues of the translocation component.
  • the non-cytotoxic protease component and the TM may be linked together by a disulphide bond.
  • the amino acid residue of the TM that forms the disulphide bond is preferably located away from the N-terminus of the TM, more preferably towards to C-terminus of the TM.
  • the non-cytotoxic protease component forms a disulphide bond with the TM component of the fusion protein.
  • the amino acid residue of the protease component that forms the disulphide bond is preferably located within the last 20, more preferably within the last 10 C-terminal amino acid residues of the protease component.
  • the amino acid residue within the TM component that forms the second part of the disulphide bond is preferably located within the last 20, more preferably within the last 10 C-terminal amino acid residues of the TM.
  • the above disulphide bond arrangements have the advantage that the protease and translocation components are arranged in a manner similar to that for native clostridial neurotoxin.
  • the respective cysteine amino acid residues are distanced apart by between 8 and 27 amino acid residues—taken from Popoff, M R & Marvaud, J-C, 1999, Structural & genomic features of clostridial neurotoxins, Chapter 9, in The Comprehensive Sourcebook of Bacterial Protein Toxins. Ed. Alouf & Freer:
  • the fusion protein may comprise one or more purification tags, which are located N-terminal to the protease component and/or C-terminal to the translocation component.
  • His-tag e.g. 6 ⁇ histidine
  • MBP-tag maltose binding protein
  • glutthione-S-transferase a C-terminal tag binding protein
  • His-MBP-tag glutathione-S-transferase
  • His-MBP-tag preferably as an N-terminal tag His-MBP-tag
  • Thioredoxin-tag preferably as an N-terminal tag CBD-tag (Chitin Binding Domain), preferably as an N-terminal tag.
  • one or more peptide spacer molecules may be included in the fusion protein.
  • a peptide spacer may be employed between a purification tag and the rest of the fusion protein molecule (e.g. between an N-terminal purification tag and a protease component of the present invention; and/or between a C-terminal purification tag and a translocation component of the present invention).
  • a peptide spacer may be also employed between the TM and translocation components of the present invention.
  • spacer molecules may be employed in any of the fusion proteins of the present invention.
  • spacer molecules include those illustrated in FIGS. 28 and 29 . Particular mention here is made to GS15, GS20, GS25, and Hx27—see FIGS. 28 and 29 .
  • the present inventors have unexpectedly found that the fusion proteins (eg. CPNv/A) of the present invention may demonstrate an improved binding activity for nociceptive sensory afferents when the size of the spacer is selected so that (in use) the C-terminus of the TM and the N-terminus of the translocation component are separated from one another by 40-105 angstroms, preferably by 50-100 angstroms, and more preferably by 50-90 angstroms.
  • the preferred spacers have an amino acid sequence of 11-29 amino acid residues, preferably 15-27 amino acid residues, and more preferably 20-27 amino acid residues. Suitable spacers may be routinely identified and obtained according to Crasto, C. J. and Feng, J. A. (2000) May, 13(5), pp. 309-312—see also http://www.fccc./edu/research/labs/fen/limker.html.
  • a DNA sequence that encodes the above-mentioned single chain polypeptide is prepared as part of a DNA vector, wherein the vector comprises a promoter and terminator.
  • the vector has a promoter selected from:
  • the DNA construct of the present invention is preferably designed in silico, and then synthesised by conventional DNA synthesis techniques.
  • the above-mentioned DNA sequence information is optionally modified for codon-biasing according to the ultimate host cell (e.g. E. coli ) expression system that is to be employed.
  • the ultimate host cell e.g. E. coli
  • the DNA backbone is preferably screened for any inherent nucleic acid sequence, which when transcribed and translated would produce an amino acid sequence corresponding to the protease cleave site encoded by the second peptide-coding sequence. This screening may be performed manually or with the assistance of computer software (e.g. the MapDraw program by DNASTAR, Inc.).
  • a method of preparing a non-cytotoxic agent comprising:
  • di-chain polypeptide which generally mimics the structure of clostridial holotoxin.
  • the resulting di-chain polypeptide typically has a structure wherein:
  • the single chain or di-chain polypeptide of the invention treat, prevent or ameliorate pain.
  • a therapeutically effective amount of a single chain or di-chain polypeptide of the invention is administered to a patient.
  • the present invention addresses a wide range of pain conditions, in particular chronic pain conditions.
  • Preferred conditions include cancerous and non-cancerous pain, inflammatory pain and neuropathic pain.
  • the opioid-fusions of the present application are particularly suited to addressing inflammatory pain, though may be less suited to addressing neuropathic pain.
  • the galanin-fusions are more suited to addressing neuropathic pain.
  • polypeptides of the present invention are typically employed in the form of a pharmaceutical composition in association with a pharmaceutical carrier, diluent and/or excipient, although the exact form of the composition may be tailored to the mode of administration. Administration is preferably to a mammal, more preferably to a human.
  • polypeptides may, for example, be employed in the form of a sterile solution for intra-articular administration or intra-cranial administration.
  • Spinal injection e.g. epidural or intrathecal
  • the dosage ranges for administration of the polypeptides of the present invention are those to produce the desired therapeutic effect. It will be appreciated that the dosage range required depends on the precise nature of the components, the route of administration, the nature of the formulation, the age of the patient, the nature, extent or severity of the patient's condition, contraindications, if any, and the judgement of the attending physician.
  • Suitable daily dosages are in the range 0.0001-1 mg/kg, preferably 0.0001-0.5 mg/kg, more preferably 0.002-0.5 mg/kg, and particularly preferably 0.004-0.5 mg/kg.
  • the unit dosage can vary from less that 1 microgram to 30 mg, but typically will be in the region of 0.01 to 1 mg per dose, which may be administered daily or preferably less frequently, such as weekly or six monthly.
  • a particularly preferred dosing regimen is based on 2.5 ng of fusion protein (e.g. CPNv/A) as the 1 ⁇ dose.
  • preferred dosages are in the range 1 ⁇ -100 ⁇ (i.e. 2.5-250 ng). This dosage range is significantly lower (i.e. at least 10-fold, typically 100-fold lower) than would be employed with other types of analgesic molecules such as NSAIDS, morphine, and gabapentin.
  • the above-mentioned difference is considerably magnified when the same comparison is made on a molar basis—this is because the fusion proteins of the present invention have a considerably greater Mw than do conventional ‘small’ molecule therapeutics.
  • Variations in these dosage levels can be adjusted using standard empirical routines for optimisation, as is well understood in the art.
  • compositions suitable for injection may be in the form of solutions, suspensions or emulsions, or dry powders which are dissolved or suspended in a suitable vehicle prior to use.
  • Fluid unit dosage forms are typically prepared utilising a pyrogen-free sterile vehicle.
  • the active ingredients depending on the vehicle and concentration used, can be either dissolved or suspended in the vehicle.
  • the polypeptides can be dissolved in a vehicle, the solution being made isotonic if necessary by addition of sodium chloride and sterilised by filtration through a sterile filter using aseptic techniques before filling into suitable sterile vials or ampoules and sealing.
  • the solution in its sealed containers may be sterilised by autoclaving.
  • compositions such as buffering, solubilising, stabilising, preservative or bactericidal, suspending or emulsifying agents may be dissolved in the vehicle.
  • Dry powders which are dissolved or suspended in a suitable vehicle prior to use may be prepared by filling pre-sterilised drug substance and other ingredients into a sterile container using aseptic technique in a sterile area.
  • polypeptides and other ingredients may be dissolved in an aqueous vehicle, the solution is sterilized by filtration and distributed into suitable containers using aseptic technique in a sterile area. The product is then freeze dried and the containers are sealed aseptically.
  • Parenteral suspensions suitable for intramuscular, subcutaneous or intradermal injection, are prepared in substantially the same manner, except that the sterile components are suspended in the sterile vehicle, instead of being dissolved and sterilisation cannot be accomplished by filtration.
  • the components may be isolated in a sterile state or alternatively it may be sterilised after isolation, e.g. by gamma irradiation.
  • a suspending agent for example polyvinylpyrrolidone is included in the composition/s to facilitate uniform distribution of the components.
  • Targeting Moiety means any chemical structure associated with an agent that functionally interacts with a Binding Site to cause a physical association between the agent and the surface of a target cell.
  • the target cell is a nociceptive sensory afferent.
  • the term TM embraces any molecule (i.e. a naturally occurring molecule, or a chemically/physically modified variant thereof that is capable of binding to a Binding Site on the target cell, which Binding Site is capable of internalisation (e.g. endosome formation)—also referred to as receptor-mediated endocytosis.
  • the TM may possess an endosomal membrane translocation function, in which case separate TM and Translocation Domain components need not be present in an agent of the present invention.
  • the TM of the present invention binds (preferably specifically binds) to a nociceptive sensory afferent (e.g. a primary nociceptive afferent).
  • a nociceptive sensory afferent e.g. a primary nociceptive afferent
  • specifically binds means that the TM binds to a nociceptive sensory afferent (e.g. a primary nociceptive afferent) with a greater affinity than it binds to other neurons such as non-nociceptive afferents, and/or to motor neurons (i.e. the natural target for clostridial neurotoxin holotoxin).
  • TM binds to a given receptor, for example the ORL 1 receptor, with a binding affinity (Ka) of 10 6 M ⁇ 1 or greater, preferably 10 7 M ⁇ 1 or greater, more preferably 10 8 M ⁇ 1 or greater, and most preferably, 10 9 M ⁇ 1 or greater.
  • Ka binding affinity
  • an agonist is defined as a molecule that is capable of stimulating the process of exocytic fusion in a target cell, which process is susceptible to inhibition by a protease (or fragment thereof) capable of cleaving a protein of the exocytic fusion apparatus in said target cell.
  • the particular agonist definition of the present invention would exclude many molecules that would be conventionally considered as agonists.
  • nerve growth factor is an agonist in respect of its ability to promote neuronal differentiation via binding to a TrkA receptor.
  • NGF nerve growth factor
  • the process that NGF stimulates i.e. cell differentiation
  • fragment when used in relation to a protein, means a peptide having at least thirty-five, preferably at least twenty-five, more preferably at least twenty, and most preferably at least ten amino acid residues of the protein in question.
  • variant when used in relation to a protein, means a peptide or peptide fragment of the protein that contains one or more analogues of an amino acid (e.g. an unnatural amino acid), or a substituted linkage.
  • derivative when used in relation to a protein, means a protein that comprises the protein in question, and a further peptide sequence.
  • the further peptide sequence should preferably not interfere with the basic folding and thus conformational structure of the original protein.
  • Two or more peptides (or fragments, or variants) may be joined together to form a derivative.
  • a peptide (or fragment, or variant) may be joined to an unrelated molecule (e.g. a second, unrelated peptide).
  • Derivatives may be chemically synthesized, but will be typically prepared by recombinant nucleic acid methods. Additional components such as lipid, and/or polysaccharide, and/or polyketide components may be included.
  • ORL 1 receptor embraces all members of the ORL 1 receptor family.
  • Members of the ORL 1 receptor family typically have a seven transmembrane domain structure and are coupled to G-proteins of the G i and G 0 families.
  • a method for determining the G-protein-stimulating activity of ligands of the ORL 1 receptor is given in Example 12.
  • a method for measuring reduction in cellular cAMP levels following ORL 1 activation is given in Example 11.
  • a further characteristic of members of the ORL 1 receptor family is that they are typically able to bind nociceptin (the natural ligand of ORL 1 ).
  • all alternative splice variants of the ORL 1 receptor are members of the ORL 1 receptor family.
  • non-cytotoxic means that the protease molecule in question does not kill the target cell to which it has been re-targeted.
  • the protease of the present invention embraces all naturally-occurring non-cytotoxic proteases that are capable of cleaving one or more proteins of the exocytic fusion apparatus in eukaryotic cells.
  • the protease of the present invention is preferably a bacterial protease (or fragment thereof). More preferably the bacterial protease is selected from the genera Clostridium or Neisseria (e.g. a clostridial L-chain, or a neisserial IgA protease preferably from N. gonorrhoeae ).
  • the present invention also embraces modified non-cytotoxic proteases, which include amino acid sequences that do not occur in nature and/or synthetic amino acid residues, so long as the modified proteases still demonstrate the above-mentioned protease activity.
  • the protease of the present invention preferably demonstrates a serine or metalloprotease activity (e.g. endopeptidase activity).
  • the protease is preferably specific for a SNARE protein (e.g. SNAP-25, synaptobrevin/VAMP, or syntaxin).
  • protease domains of neurotoxins for example the protease domains of bacterial neurotoxins.
  • the present invention embraces the use of neurotoxin domains, which occur in nature, as well as recombinantly prepared versions of said naturally-occurring neurotoxins.
  • Exemplary neurotoxins are produced by clostridia, and the term clostridial neurotoxin embraces neurotoxins produced by C. tetani (TeNT), and by C. botulinum (BoNT) serotypes A-G, as well as the closely related BoNT-like neurotoxins produced by C. barati and C. butyricum .
  • TeNT C. tetani
  • BoNT botulinum
  • BoNT/A denotes the source of neurotoxin as BoNT (serotype A).
  • Corresponding nomenclature applies to other BoNT serotypes.
  • L-chain fragment means a component of the L-chain of a neurotoxin, which fragment demonstrates a metalloprotease activity and is capable of proteolytically cleaving a vesicle and/or plasma membrane associated protein involved in cellular exocytosis.
  • a Translocation Domain is a molecule that enables translocation of a protease (or fragment thereof) into a target cell such that a functional expression of protease activity occurs within the cytosol of the target cell. Whether any molecule (e.g. a protein or peptide) possesses the requisite translocation function of the present invention may be confirmed by any one of a number of conventional assays.
  • Shone C. (1987) describes an in vitro assay employing liposomes, which are challenged with a test molecule. Presence of the requisite translocation function is confirmed by release from the liposomes of K + and/or labelled NAD, which may be readily monitored [see Shone C. (1987) Eur. J. Biochem; vol. 167(1): pp. 175-180].
  • Blaustein R. (1987) describes a simple in vitro assay employing planar phospholipid bilayer membranes. The membranes are challenged with a test molecule and the requisite translocation function is confirmed by an increase in conductance across said membranes [see Blaustein (1987) FEBS Letts; vol. 226, no. 1: pp. 115-120].
  • the Translocation Domain is preferably capable of formation of ion-permeable pores in lipid membranes under conditions of low pH. Preferably it has been found to use only those portions of the protein molecule capable of pore-formation within the endosomal membrane.
  • the Translocation Domain may be obtained from a microbial protein source, in particular from a bacterial or viral protein source.
  • the Translocation Domain is a translocating domain of an enzyme, such as a bacterial toxin or viral protein.
  • the Translocation Domain may be of a clostridial origin, namely the H N domain (or a functional component thereof).
  • H N means a portion or fragment of the H-chain of a clostridial neurotoxin approximately equivalent to the amino-terminal half of the H-chain, or the domain corresponding to that fragment in the intact H-chain. It is preferred that the H-chain substantially lacks the natural binding function of the H C component of the H-chain.
  • the H C function may be removed by deletion of the H C amino acid sequence (either at the DNA synthesis level, or at the post-synthesis level by nuclease or protease treatment). Alternatively, the H C function may be inactivated by chemical or biological treatment.
  • the H-chain is preferably incapable of binding to the Binding Site on a target cell to which native clostridial neurotoxin (i.e. holotoxin) binds.
  • the translocation domain is a H N domain (or a fragment thereof) of a clostridial neurotoxin.
  • suitable clostridial Translocation Domains include:
  • H N embraces naturally-occurring neurotoxin H N portions, and modified H N portions having amino acid sequences that do not occur in nature and/or synthetic amino acid residues, so long as the modified H N portions still demonstrate the above-mentioned translocation function.
  • the Translocation Domain may be of a non-clostridial origin (see Table 4).
  • the Translocation Domain may mirror the Translocation Domain present in a naturally-occurring protein, or may include amino acid variations so long as the variations do not destroy the translocating ability of the Translocation Domain.
  • viral Translocation Domains suitable for use in the present invention include certain translocating domains of virally expressed membrane fusion proteins.
  • translocation i.e. membrane fusion and vesiculation
  • the translocation i.e. membrane fusion and vesiculation function of a number of fusogenic and amphiphilic peptides derived from the N-terminal region of influenza virus haemagglutinin.
  • virally expressed membrane fusion proteins known to have the desired translocating activity are a translocating domain of a fusogenic peptide of Semliki Forest Virus (SFV), a translocating domain of vesicular stomatitis virus (VSV) glycoprotein G, a translocating domain of SER virus F protein and a translocating domain of Foamy virus envelope glycoprotein.
  • SFV Semliki Forest Virus
  • VSV vesicular stomatitis virus
  • SER virus F protein a translocating domain of Foamy virus envelope glycoprotein.
  • Virally encoded Aspike proteins have particular application in the context of the present invention, for example, the E1 protein of SFV and the G protein of the G protein of VSV.
  • Translocation Domains listed in Table (below) includes use of sequence variants thereof.
  • a variant may comprise one or more conservative nucleic acid substitutions and/or nucleic acid deletions or insertions, with the proviso that the variant possesses the requisite translocating function.
  • a variant may also comprise one or more amino acid substitutions and/or amino acid deletions or insertions, so long as the variant possesses the requisite translocating function.
  • FIG. 1 Purification of a LC/A-nociceptin-H N /A fusion protein
  • FIG. 2 Purification of a nociceptin-LC/A-H N /A fusion protein
  • FIG. 3 Purification of a LC/C-nociceptin-H N /C fusion protein
  • FIG. 4 Purification of a LC/A-met enkephalin-H N /A fusion protein
  • FIG. 5 Comparison of binding efficacy of a LC/A-nociceptin-H N /A fusion protein and a nociceptin-LC/A-H N /A fusion protein
  • FIG. 6 In vitro catalytic activity of a LC/A-nociceptin-H N /A fusion protein
  • FIG. 7 Purification of a LC/A-nociceptin variant-H N /A fusion protein
  • FIG. 8 Comparison of binding efficacy of a LC/A-nociceptin-H N /A fusion protein and a LC/A-nociceptin variant-H N /A fusion protein
  • FIG. 9 Expressed/purified LC/A-nociceptin-H N /A fusion protein family with variable spacer length product(s)
  • FIG. 10 Inhibition of SP release and cleavage of SNAP-25 by CPN-A
  • FIG. 11 Inhibition of SP release and cleavage of SNAP-25 over extended time periods after exposure of DRG to CPN-A
  • FIG. 12 Cleavage of SNAP-25 by CPNv-A
  • FIG. 13 Cleavage of SNAP-25 over extended time periods after exposure of DRG to CPNv-A
  • FIG. 14 CPNv-A fusion-mediated displacement of [3H]-nociceptin binding
  • FIG. 15 Expressed/purified CPNv(Ek)-A product
  • FIG. 16 Cleavage of SNAP-25 by CPNv(Ek)-A
  • FIG. 17 Expressed/purified CPNv-C product
  • FIG. 18 Cleavage of syntaxin by CPNv-C
  • FIG. 19 CPN-A efficacy in the Acute Capsaicin-Induced Mechanical Allodynia model
  • FIG. 20 CPN-A efficacy in the Streptozotocin (STZ)-Induced Peripheral Diabetic Neuropathy (Neuropathic Pain) model
  • FIG. 21 CPNv-A efficacy in the Acute Capsaicin-Induced Mechanical Allodynia model
  • FIG. 22 Expressed/purified LC/A-CPLE-H N /A product
  • FIG. 23 Expressed/purified LC/A-CPBE-H N /A product
  • FIG. 24 Expressed/purified CPOP-A product
  • FIG. 25 Expressed/purified CPOPv-A product
  • FIG. 26 In vitro SNAP-25 cleavage in a DRG cell model
  • FIG. 27 Expressed/purified CPNv-A-FXa-HT (removable his-tag)
  • FIG. 28 In vitro efficacy of LC/A-nociceptin-H N /A fusion proteins with variable spacer length, as assessed by ligand competition assay
  • FIG. 29 In vitro efficacy of LC/A-nociceptin-H N /A fusion proteins with variable spacer length, as assessed by in vitro SNAP-25 cleavage
  • FIG. 1 Purification of a LC/A-Nociceptin-H N /A Fusion Protein
  • LC/A-nociceptin-H N /A fusion protein was purified from E. coli BL21 cells. Briefly, the soluble products obtained following cell disruption were applied to a nickel-charged affinity capture column. Bound proteins were eluted with 100 mM imidazole, treated with Factor Xa to activate the fusion protein and remove the maltose-binding protein (MBP) tag, then re-applied to a second nickel-charged affinity capture column. Samples from the purification procedure were assessed by SDS-PAGE (Panel A) and Western blotting (Panel B). Anti-nociceptin antisera (obtained from Abcam) were used as the primary antibody for Western blotting. The final purified material in the absence and presence of reducing agent is identified in the lanes marked [ ⁇ ] and [+] respectively.
  • FIG. 2 Purification of a Nociceptin-LC/A-H N /A Fusion Protein
  • a nociceptin-LC/A-H N /A fusion protein was purified from E. coli BL21 cells. Briefly, the soluble products obtained following cell disruption were applied to a nickel-charged affinity capture column. Bound proteins were eluted with 100 mM imidazole, treated with Factor Xa to activate the fusion protein and remove the maltose-binding protein (MBP) tag, then re-applied to a second nickel-charged affinity capture column. Samples from the purification procedure were assessed by SDS-PAGE (Panel A) and Western blotting (Panel B). Anti-nociceptin antisera (obtained from Abcam) were used as the primary antibody for Western blotting. The final purified material in the absence and presence of reducing agent is identified in the lanes marked [ ⁇ ] and [+] respectively.
  • FIG. 3 Purification of a LC/C-Nociceptin-H N /C Fusion Protein
  • an LC/C-nociceptin-H N /C fusion protein was purified from E. coli BL21 cells. Briefly, the soluble products obtained following cell disruption were applied to a nickel-charged affinity capture column. Bound proteins were eluted with 100 mM imidazole, treated with Factor Xa to activate the fusion protein and remove the maltose-binding protein (MBP) tag, then re-applied to a second nickel-charged affinity capture column. Samples from the purification procedure were assessed by SDS-PAGE (Panel A) and Western blotting (Panel B). Anti-nociceptin antisera (obtained from Abcam) were used as the primary antibody for Western blotting. The final purified material in the absence and presence of reducing agent is identified in the lanes marked H and [+] respectively.
  • MBP maltose-binding protein
  • FIG. 4 Purification of a LC/A-met Enkephalin-H N /A Fusion Protein
  • an LC/A-met enkephalin-H N /A fusion protein was purified from E. coli BL21 cells. Briefly, the soluble products obtained following cell disruption were applied to a nickel-charged affinity capture column. Bound proteins were eluted with 100 mM imidazole, treated with Factor Xa to activate the fusion protein and remove the maltose-binding protein (MBP) tag, then re-applied to a second nickel-charged affinity capture column. Samples from the purification procedure were assessed by SDS-PAGE. The final purified material in the absence and presence of reducing agent is identified in the lanes marked [ ⁇ ] and [+] respectively.
  • MBP maltose-binding protein
  • FIG. 5 Comparison of Binding Efficacy of a LC/A-Nociceptin-H N /A Fusion Protein and a Nociceptin-LC/A-H N /A Fusion Protein
  • nociceptin fusions to bind to the ORL 1 receptor was assessed using a simple competition-based assay.
  • Primary cultures of dorsal root ganglia (DRG) were exposed to varying concentrations of test material in the presence of 1 nM [3H]-nociceptin.
  • the reduction in specific binding of the radiolabelled ligand was assessed by scintillation counting, and plotted in comparison to the efficacy of unlabelled ligand (Tocris nociceptin). It is clear that the LC/A-nociceptin-H N /A fusion is far superior to the nociceptin-LC/A-H N /A fusion at interacting with the ORL 1 receptor.
  • FIG. 6 In Vitro Catalytic Activity of a LC/A-Nociceptin-H N /A Fusion Protein
  • FIG. 7 Purification of a LC/A-Nociceptin Variant-H N /A Fusion Protein
  • an LC/A-nociceptin variant-H N /A fusion protein was purified from E. coli BL21 cells. Briefly, the soluble products obtained following cell disruption were applied to a nickel-charged affinity capture column. Bound proteins were eluted with 100 mM imidazole, treated with Factor Xa to activate the fusion protein and remove the maltose-binding protein (MBP) tag, then re-applied to a second nickel-charged affinity capture column. Samples from the purification procedure were assessed by SDS-PAGE. The final purified material in the absence and presence of reducing agent is identified in the lanes marked [ ⁇ ] and [+] respectively.
  • MBP maltose-binding protein
  • FIG. 8 Comparison of Binding Efficacy of a LC/A-Nociceptin-H N /A Fusion Protein and a LC/A-Nociceptin Variant-H N /A Fusion Protein
  • nociceptin fusions to bind to the ORL 1 receptor was assessed using a simple competition-based assay.
  • Primary cultures of dorsal root ganglia (DRG) were exposed to varying concentrations of test material in the presence of 1 nM [3H]-nociceptin.
  • the reduction in specific binding of the radiolabelled ligand was assessed by scintillation counting, and plotted in comparison to the efficacy of unlabelled ligand (Tocris nociceptin). It is clear that the LC/A-nociceptin variant-H N /A fusion (CPNv-LHA) is superior to the LC/A-nociceptin variant-H N /A fusion (CPN-LHA) at interacting with the ORL 1 receptor.
  • FIG. 9 Expressed/Purified LC/A-Nociceptin-H N /A Fusion Protein Family with Variable Spacer Length Product(s)
  • variants of the LC/A-CPN-H N /A fusion consisting of GS10, GS30 and HX27 are purified from E. coli cell paste.
  • Samples from the purification of LC/A-CPN(GS10)-H N /A, LC/A-CPN(GS15)-H N /A, LC/A-CPN(GS25)-H N /A, LC/A-CPN(GS30)-H N /A and LC/A-CPN(HX27)-H N /A were assessed by SDS-PAGE prior to staining with Coomassie Blue.
  • the electrophoresis profile indicates purification of a disulphide-bonded di-chain species of the expected molecular mass of CPBE-A.
  • FIG. 10 Inhibition of SP Release and Cleavage of SNAP-25 by CPN-A
  • DRG dorsal root ganglia
  • FIG. 11 Inhibition of SP Release and Cleavage of SNAP-25 Over Extended Time Periods after Exposure of DRG to CPN-A
  • FIG. 12 Cleavage of SNAP-25 by CPNv-A
  • DRG dorsal root ganglia
  • FIG. 13 Cleavage of SNAP-25 Over Extended Time Periods after Exposure of DRG to CPNv-A
  • CPNv-A dorsal root ganglia
  • FIG. 14 CPNv-A Fusion-Mediated Displacement of [3H]-Nociceptin Binding
  • nociceptin fusions to bind to the ORL 1 receptor was assessed using a simple competition-based assay.
  • Primary cultures of dorsal root ganglia (DRG) were exposed to varying concentrations of test material in the presence of 1 nM [3H]-nociceptin.
  • the reduction in specific binding of the radiolabelled ligand was assessed by scintillation counting, and plotted in comparison to the efficacy of unlabelled ligand (Tocris nociceptin).
  • FIG. 15 Expressed/Purified CPNv(Ek)-A Product
  • Lane 3 purified material following initial capture on Ni 2+ -charged Sepharose
  • Lane 4 purified final material post activation with enterokinase (5 ⁇ l)
  • Lane 5 purified final material post activation with enterokinase (10 ⁇ l)
  • Lane 6 purified final material post activation with enterokinase (20 ⁇ l)
  • Lane 7 purified final material post activation with enterokinase+DTT (5 ⁇ l)
  • Lane 8 purified final material post activation with enterokinase+DTT (10 ⁇ l)
  • Lane 9 purified final material post activation with enterokinase+DTT (20 ⁇ l).
  • FIG. 16 Cleavage of SNAP-25 by CPNv(Ek)-A
  • CPNv(Ek)-A Primary cultures of dorsal root ganglia (DRG) were exposed to varying concentrations of CPNv(Ek)-A for 24 hours. Cellular proteins were separated by SDS-PAGE, Western blotted, and probed with anti-SNAP-25 to facilitate an assessment of SNAP-25 cleavage. The percentage of cleaved SNAP-25 was calculated by densitometric analysis. CPNv-A as prepared in Example 9 was used for comparison purposes. The percentage cleavage of SNAP-25 by CPNv(Ek)-A (labelled as En activated) and CPNv-A (labelled as Xa activated) are illustrated.
  • FIG. 17 Expressed/Purified CPNv-C Product
  • FIG. 18 Cleavage of Syntaxin by CPNv-C
  • DRG dorsal root ganglia
  • FIG. 19 CPN-A Efficacy in the Acute Capsaicin-Induced Mechanical Allodynia Model
  • LC/A-nociceptin-H N /A fusion (CPN/A) to inhibit capsaicin-induced mechanical allodynia was evaluated following subcutaneous intraplantar injection in the rat hind paw.
  • Test animals were evaluated for paw withdrawal frequency (PWF %) in response to a 10 g Von Frey filament stimulus series (10 stimuli ⁇ 3 trials) prior to recruitment into the study (Pre-Treat); after subcutaneous intraplantar treatment with CPN/A but before capsaicin (Pre-CAP); and following capsaicin challenge post-injection of CPN/A (average of responses at 15′ and 30′; CAP).
  • Capsaicin challenge was achieved by injection of 10 ⁇ L of a 0.3% solution. Sample dilutions were prepared in 0.5% BSA/saline.
  • FIG. 20 CPN-A Efficacy in the Streptozotocin (STZ)-Induced Peripheral Diabetic Neuropathy (Neuropathic Pain) Model
  • Test materials (20-25 ⁇ l) are injected subcutaneously as a single injection (except gabapentin) and the PWT is measured at 1 day post-treatment and periodically thereafter over a 2 week period.
  • Gabapentin (30 mg/kg i.p. @ 3 ml/kg injection volume) is injected daily, 2 hours prior to the start of PWT testing.
  • FIG. 21 CPNv-A Efficacy in the Acute Capsaicin-Induced Mechanical Allodynia Model
  • LC/A-nociceptin variant-H N /A fusion (CPNv/A) to inhibit capsaicin-induced mechanical allodynia was evaluated following subcutaneous intraplantar injection in the rat hind paw.
  • Test animals were evaluated for paw withdrawal frequency (PWF %) in response to a 10 g Von Frey filament stimulus series (10 stimuli ⁇ 3 trials) prior to recruitment into the study (Pre-Treat), after subcutaneous intraplantar treatment with CPNv/A but before capsaicin (Pre-CAP), and following capsaicin challenge post-injection of CPNv/A (average of responses at 15′ and 30′; CAP).
  • Capsaicin challenge was achieved by injection of 10 ⁇ L of a 0.3% solution.
  • FIG. 22 Example/Purified LC/A-CPLE-H N /A Product
  • FIG. 23 Example/Purified LC/A-CPBE-H N /A Product
  • FIG. 24 Example/Purified CPOP-A Product
  • FIG. 25 Example/Purified CPOPv-A Product
  • FIG. 26 In Vitro SNAP-25 Cleavage in a DRG Cell Model
  • DRG dorsal root ganglia
  • FIG. 27 Expressed/Purified CPNv-A-FXa-HT (Removable His-Tag)
  • FIG. 28 In Vitro Efficacy of LC/A-Nociceptin-H N /A Fusion Proteins with Variable Spacer Length, as Assessed by Ligand Competition Assay
  • DRG dorsal root ganglia
  • the reduction in specific binding of the radiolabelled ligand was assessed by scintillation counting, and plotted in comparison to the efficacy of unlabelled ligand (Tocris nociceptin).
  • the upper panel illustrates the displacement characteristics of the GS0, GS20, GS30 and Hx27 spacers, whilst the lower panel illustrates the displacement achieved by the GS10, GS15 and GS25 spaced fusion proteins. It is concluded that the GS0 and GS30 spacers are ineffective, and the GS10 is poorly effective, at displacing nociceptin from the ORL1 receptor.
  • FIG. 29 In Vitro Efficacy of LC/A-Nociceptin-H N /A Fusion Proteins with Variable Spacer Length, as Assessed by In Vitro SNAP-25 Cleavage
  • DRG dorsal root ganglia
  • CPN-A of variable spacer length
  • Cellular proteins were separated by SDS-PAGE, Western blotted, and probed with anti-SNAP-25 to facilitate an assessment of SNAP-25 cleavage.
  • the percentage of cleaved SNAP-25 was calculated by densitometric analysis.
  • the poorly effective binding characteristics of the GS10 spaced fusion protein (see FIG. 28 ) are reflected in the higher concentrations of fusion required to achieve cleavage of intracellular SNAP-25.
  • GS0 and GS30 spaced fusion proteins were completely ineffective (date not shown).
  • GS15, 20 and 25 spaced fusion proteins were similarly effective.
  • A) SEQ ID67 Protein sequence of the LC/C-CPNv-H N /C fusion (act. A) SEQ ID68 DNA sequence of the LC/A-CPLE-H N /A fusion SEQ ID69 Protein sequence of the LC/A-CPLE-H N /A fusion SEQ ID70 DNA sequence of the LC/A-CPOP-H N /A fusion SEQ ID71 Protein sequence of the LC/A-CPOP-H N /A fusion SEQ ID72 DNA sequence of the LC/A-CPOPv-H N /A fusion SEQ ID73 Protein sequence of the LC/A-CPOPv-H N /A fusion SEQ ID74 DNA sequence of the IgA protease SEQ ID75 DNA sequence of the IgA-CPNv-H N /A fusion SEQ ID76 Protein sequence of the IgA-CPNv-H N /A fusion SEQ ID77 DNA sequence of the FXa-HT SEQ ID78 DNA sequence of the C
  • the following procedure creates the LC and H N fragments for use as the component backbone for multidomain fusion expression.
  • This example is based on preparation of a serotype A based clone (SEQ ID1 and SEQ ID2), though the procedures and methods are equally applicable to the other serotypes [illustrated by the sequence listing for serotype B (SEQ ID3 and SEQ ID4) and serotype C (SEQ ID5 and SEQ ID6)].
  • pCR 4 (Invitrogen) is the chosen standard cloning vector, selected due to the lack of restriction sequences within the vector and adjacent sequencing primer sites for easy construct confirmation.
  • the expression vector is based on the pMAL (NEB) expression vector, which has the desired restriction sequences within the multiple cloning site in the correct orientation for construct insertion (BamHI-SalI-PstI-HindIII). A fragment of the expression vector has been removed to create a non-mobilisable plasmid and a variety of different fusion tags have been inserted to increase purification options.
  • the LC/A (SEQ ID1) is created by one of two ways:
  • the DNA sequence is designed by back translation of the LC/A amino acid sequence [obtained from freely available database sources such as GenBank (accession number P10845) or Swissprot (accession locus BXA1_CLOBO) using one of a variety of reverse translation software tools (for example EditSeq best E. coli reverse translation (DNASTAR Inc.), or Backtranslation tool v2.0 (Entelechon)].
  • GenBank accession number P10845
  • Swissprot accession locus BXA1_CLOBO
  • BamHI/SalI recognition sequences are incorporated at the 5′ and 3′ ends respectively of the sequence, maintaining the correct reading frame.
  • the DNA sequence is screened (using software such as MapDraw, DNASTAR Inc.) for restriction enzyme cleavage sequences incorporated during the back translation.
  • E. coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example GenBank Release 143, 13 Sep. 2004).
  • This optimised DNA sequence containing the LC/A open reading frame (ORF) is then commercially synthesized (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector.
  • the alternative method is to use PCR amplification from an existing DNA sequence with BamHI and SalI restriction enzyme sequences incorporated into the 5′ and 3′ PCR primers respectively.
  • Complementary oligonucleotide primers are chemically synthesised by a supplier (for example MWG or Sigma-Genosys), so that each pair has the ability to hybridize to the opposite strands (3′ ends pointing “towards” each other) flanking the stretch of Clostridium target DNA, one oligonucleotide for each of the two DNA strands.
  • the pair of short oligonucleotide primers specific for the Clostridium DNA sequence are mixed with the Clostridium DNA template and other reaction components and placed in a machine (the ‘PCR machine’) that can change the incubation temperature of the reaction tube automatically, cycling between approximately 94° C. (for denaturation), 55° C. (for oligonucleotide annealing), and 72° C. (for synthesis).
  • reagents required for amplification of a PCR product include a DNA polymerase (such as Taq or Pfu polymerase), each of the four nucleotide dNTP building blocks of DNA in equimolar amounts (50-200 ⁇ M) and a buffer appropriate for the enzyme optimised for Mg 2+ concentration (0.5-5 mM).
  • a DNA polymerase such as Taq or Pfu polymerase
  • each of the four nucleotide dNTP building blocks of DNA in equimolar amounts (50-200 ⁇ M)
  • a buffer appropriate for the enzyme optimised for Mg 2+ concentration 0.5-5 mM.
  • the amplification product is cloned into pCR 4 using either, TOPO TA cloning for Taq PCR products or Zero Blunt TOPO cloning for Pfu PCR products (both kits commercially available from Invitrogen).
  • the resultant clone is checked by sequencing. Any additional restriction sequences which are not compatible with the cloning system are then removed using site directed mutagenesis [for example, using Quickchange (Stratagene Inc.)].
  • the H N /A (SEQ ID2) is created by one of two ways:
  • the DNA sequence is designed by back translation of the H N /A amino acid sequence [obtained from freely available database sources such as GenBank (accession number P10845) or Swissprot (accession locus BXA1_CLOBO)] using one of a variety of reverse translation software tools [for example EditSeq best E. coli reverse translation (DNASTAR Inc.), or Backtranslation tool v2.0 (Entelechon)].
  • a PstI restriction sequence added to the N-terminus and XbaI-stop codon-HindIII to the C-terminus ensuring the correct reading frame is maintained.
  • the DNA sequence is screened (using software such as MapDraw, DNASTAR Inc.) for restriction enzyme cleavage sequences incorporated during the back translation.
  • E. coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example GenBank Release 143, 13 Sep. 2004).
  • This optimised DNA sequence is then commercially synthesized (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector.
  • the alternative method is to use PCR amplification from an existing DNA sequence with PstI and XbaI-stop codon-HindIII restriction enzyme sequences incorporated into the 5′ and 3′ PCR primers respectively.
  • the PCR amplification is performed as described above.
  • the PCR product is inserted into pCR 4 vector and checked by sequencing. Any additional restriction sequences which are not compatible with the cloning system are then removed using site directed mutagenesis [for example using Quickchange (Stratagene Inc.)].
  • the LC-H N linker can be designed from first principle, using the existing sequence information for the linker as the template.
  • the serotype A linker in this case defined as the inter-domain polypeptide region that exists between the cysteines of the disulphide bridge between LC and H N
  • the sequence VRGIITSKTKSLDKGYNKALNDL is 23 amino acids long and has the sequence VRGIITSKTKSLDKGYNKALNDL.
  • proteolytic activation in nature leads to an H N domain that has an N-terminus of the sequence ALNDL.
  • This sequence information is freely available from available database sources such as GenBank (accession number P10845) or Swissprot (accession locus BXA1_CLOBO).
  • E. coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example, GenBank Release 143, 13 Sep. 2004). This optimised DNA sequence is then commercially synthesized (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector.
  • the pCR 4 vector encoding the linker (SEQ ID7) is cleaved with BamHI+SalI restriction enzymes.
  • This cleaved vector then serves as the recipient vector for insertion and ligation of the LC/A DNA (SEQ ID1) cleaved with BamHI+SalI.
  • the resulting plasmid DNA is then cleaved with PstI+XbaI restriction enzymes and serves as the recipient vector for the insertion and ligation of the H N /A DNA (SEQ ID2) cleaved with PstI+XbaI.
  • the final construct contains the LC-linker-nociceptin-spacer-H N ORF (SEQ ID13) for transfer into expression vectors for expression to result in a fusion protein of the sequence illustrated in SEQ ID14.
  • the LC/A-H N /A backbone is constructed as described in Example 2 using the synthesised A serotype linker with the addition of a Factor Xa site for activation, arranged as BamHI-SalI-linker-protease site-linker-PstI-XbaI-stop codon-Hind III (SEQ ID8).
  • the LC/A-H N /A backbone and the synthesised N-terminal presentation nociceptin insert (SEQ ID9) are cleaved with BamHI+HindIII restriction enzymes, gel purified and ligated together to create a nociceptin-spacer-LC-linker-H N .
  • the ORF (SEQ ID15) is then cut out using restriction enzymes AvaI+XbaI for transfer into expression vectors for expression to result in a fusion protein of the sequence illustrated in SEQ ID16.
  • the LC/C (SEQ ID5) and H N /C (SEQ ID6) are created and inserted into the C serotype linker arranged as BamHI-SalI-linker-protease site-nociceptin-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID10).
  • the final construct contains the LC-linker-nociceptin-spacer-H N ORF (SEQ ID17) for expression as a protein of the sequence illustrated in SEQ ID18.
  • the LC/C (SEQ ID5) and H N /C (SEQ ID6) are created and inserted into the A serotype linker arranged as BamHI-SalI-linker-protease site-nociceptin-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID7).
  • the final construct contains the LC-linker-nociceptin-spacer-H N ORF (SEQ ID19) for expression as a protein of the sequence illustrated in SEQ ID20.
  • LC/A-met enkephalin-H N /A fusion Due to the small, five-amino acid, size of the met-enkephalin ligand the LC/A-met enkephalin-H N /A fusion is created by site directed mutagenesis [for example using Quickchange (Stratagene Inc.)] using the LC/A-nociceptin-H N /A fusion (SEQ ID13) as a template. Oligonucleotides are designed encoding the YGGFM met-enkephalin peptide, ensuring standard E.
  • SDM product is checked by sequencing and the final construct containing the LC-linker-met enkephalin-spacer-H N ORF (SEQ ID21) for expression as a protein of the sequence illustrated in SEQ ID22.
  • the LC/A (SEQ ID1) and H N /A (SEQ ID2) are created and inserted into the A serotype ⁇ endorphin linker arranged as BamHI-Sa/1-linker-protease site- ⁇ endorphin-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID11).
  • the final construct contains the LC-linker- ⁇ endorphin-spacer-H N ORF (SEQ ID23) for expression as a protein of the sequence illustrated in SEQ ID24.
  • the LC/A (SEQ ID1) and H N /A (SEQ ID2) are created and inserted into the A serotype nociceptin variant linker arranged as BamHI-SalI-linker-protease site-nociceptin variant-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID12).
  • the final construct contains the LC-linker-nociceptin variant-spacer-H N ORF (SEQ ID25) for expression as a protein of the sequence illustrated in SEQ ID26.
  • Defrost falcon tube containing 25 ml 50 mM HEPES pH 7.2, 200 mM NaCl and approximately 10 g of E. coli BL21 cell paste.
  • Make the thawed cell paste up to 80 ml with 50 mM HEPES pH 7.2, 200 mM NaCl and sonicate on ice 30 seconds on, 30 seconds off for 10 cycles at a power of 22 microns ensuring the sample remains cool.
  • Spin the lysed cells at 18 000 rpm, 4° C. for 30 minutes. Load the supernatant onto a 0.1 M NiSO 4 charged Chelating column (20-30 ml column is sufficient) equilibrated with 50 mM HEPES pH 7.2, 200 mM NaCl.
  • Substance P EIA is obtained from R&D Systems, UK.
  • the amount of Substance P released by the neuronal cells in the presence of the TM of interest is compared to the release obtained in the presence and absence of 100 mM KCl. Stimulation of Substance P release by the TM of interest above the basal release, establishes that the TM of interest is an “agonist ligand” as defined in this specification. If desired the stimulation of Substance P release by the TM of interest can be compared to a standard Substance P release-curve produced using the natural ORL-1 receptor ligand, nociceptin (Tocris).
  • test is conducted essentially as described previously by Meunier et al. [Isolation and structure of the endogenous agonist of opioid receptor-like ORL 1 receptor. Nature 377: 532-535, 1995] in intact transfected-CHO cells plated on 24-well plastic plates.
  • [3H]adenine (1.0 ⁇ Ci) in 0.4 ml of culture medium.
  • the cells remain at 37° C. for 2 h to allow the adenine to incorporate into the intracellular ATP pool.
  • the cells are washed once with incubation buffer containing: 130 mM NaCl, 4.8 mM KCl, 1.2 mM KH 2 PO 4 , 1.3 mM CaCl 2 , 1.2 mM MgSO 4 , 10 mM glucose, 1 mg/ml bovine serum albumin and 25 mM HEPES pH 7.4, and replaced with buffer containing forskolin (10 ⁇ M) and isobutylmethylxanthine (50 ⁇ M) with or without the TM of interest.
  • the medium is aspirated and replaced with 0.5 ml, 0.2 M HCl.
  • Approximately 1000 cpm of [ 14 C]cAMP is added to each well and used as an internal standard.
  • the contents of the wells are then transferred to columns of 0.65 g dry alumina powder.
  • the columns are eluted with 4 ml of 5 mM HCl, 0.5 ml of 0.1 M ammonium acetate, then two additional millilitres of ammonium acetate.
  • the final eluate is collected into scintillation vials and counted for 14 C and tritium. Amounts collected are corrected for recovery of [ 14 C]cAMP.
  • TMs that are agonists at the ORL 1 receptor cause a reduction in the level of cAMP produced in response to forskolin.
  • SPA agglutinin-coated
  • This assay is carried out essentially as described by Traynor and Nahorski [Modulation by ⁇ -opioid agonists of guanosine-5-O-(3-[ 35 S]thio)triphosphate binding to membranes from human neuroblastoma SH-SY5Y cells. Mol. Pharmacol. 47: 848-854, 1995].
  • Cells are scraped from tissue culture dishes into 20 mM HEPES, 1 mM ethylenediaminetetraacetic acid, then centrifuged at 500 ⁇ g for 10 min. Cells are resuspended in this buffer and homogenized with a Polytron Homogenizer.
  • the homogenate is centrifuged at 27,000 ⁇ g for 15 min, and the pellet resuspended in buffer A, containing: 20 mM HEPES, 10 mM MgCl 2 , 100 mM NaCl, pH 7.4.
  • the suspension is recentrifuged at 20,000 ⁇ g and suspended once more in buffer A.
  • membranes (8-15 ⁇ g protein) are incubated with [ 35 S]GTP S (50 pM), GDP (10 ⁇ M), with and without the TM of interest, in a total volume of 1.0 ml, for 60 min at 25° C. Samples are filtered over glass fibre filters and counted as described for the binding assays.
  • the linker-nociceptin-spacer insert is prepared as described in Example 2.
  • the pCR 4 vector encoding the linker (SEQ ID7) is cleaved with BamHI+SalI restriction enzymes. This cleaved vector then serves as the recipient for insertion and ligation of the LC/A DNA (SEQ ID1) also cleaved with BamHI+SalI.
  • the resulting plasmid DNA is then cleaved with BamHI+HindIII restriction enzymes and the LC/A-linker fragment inserted into a similarly cleaved vector containing a unique multiple cloning site for BamHI, SalI, PstI, and HindIII such as the pMAL vector (NEB).
  • the H N /A DNA (SEQ ID2) is then cleaved with PstI+HindIII restriction enzymes and inserted into the similarly cleaved pMAL-LC/A-linker construct.
  • the final construct contains the LC-linker-nociceptin-spacer-H N ORF (SEQ ID13) for expression as a protein of the sequence illustrated in SEQ ID14.
  • an A serotype linker with the addition of a Factor Xa site for activation, arranged as BamHI-SalI-linker-protease site-linker-PstI-XbaI-stop codon-HindIII (SEQ ID8) is synthesised as described in Example 13.
  • the pCR 4 vector encoding the linker is cleaved with BamHI+SalI restriction enzymes. This cleaved vector then serves as the recipient for insertion and ligation of the LC/A DNA (SEQ ID1) also cleaved with BamHI+SalI.
  • the resulting plasmid DNA is then cleaved with BamHI+HindIII restriction enzymes and the LC/A-linker fragment inserted into a similarly cleaved vector containing the synthesised N-terminal presentation nociceptin insert (SEQ ID9).
  • This construct is then cleaved with AvaI+HindIII and inserted into an expression vector such as the pMAL plasmid (NEB).
  • the H N /A DNA (SEQ ID2) is then cleaved with PstI+HindIII restriction enzymes and inserted into the similarly cleaved pMAL-nociceptin-LC/A-linker construct.
  • the final construct contains the nociceptin-spacer-LC/A-H N /A ORF (SEQ ID51) for expression as a protein of the sequence illustrated in SEQ ID52.
  • Restriction sites are then incorporated into the DNA sequence and can be arranged as BamHI-SalI-linker-protease site-nociceptin-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID53 to SEQ ID57). It is important to ensure the correct reading frame is maintained for the spacer, nociceptin and restriction sequences and that the XbaI sequence is not preceded by the bases, TC which would result on DAM methylation. The DNA sequence is screened for restriction sequence incorporation and any additional sequences are removed manually from the remaining sequence ensuring common E. coli codon usage is maintained. E.
  • coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example GenBank Release 143, 13 Sep. 2004).
  • This optimised DNA sequence is then commercially synthesized (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector.
  • the pCR 4 vector encoding the linker (SEQ ID54) is cleaved with BamHI+SalI restriction enzymes.
  • This cleaved vector then serves as the recipient vector for insertion and ligation of the LC/A DNA (SEQ ID1) also cleaved with BamHI+SalI.
  • the resulting plasmid DNA is then cleaved with BamHI+HindIII restriction enzymes and the LC/A-linker fragment inserted into a similarly cleaved vector containing a unique multiple cloning site for BamHI, SalI, PstI, and HindIII such as the pMAL vector (NEB).
  • the H N /A DNA (SEQ ID2) is then cleaved with PstI+HindIII restriction enzymes and inserted into the similarly cleaved pMAL-LC/A-linker construct.
  • the final construct contains the LC/A-CPN(GS15)-H N /A ORF (SEQ ID58) for expression as a protein of the sequence illustrated in SEQ ID59.
  • the pCR 4 vector encoding the linker (SEQ ID55) is cleaved with BamHI+SalI restriction enzymes.
  • This cleaved vector then serves as the recipient vector for insertion and ligation of the LC/A DNA (SEQ ID1) cleaved with BamHI+SalI.
  • the resulting plasmid DNA is then cleaved with BamHI+HindIII restriction enzymes and the LC/A-linker fragment inserted into a similarly cleaved vector containing a unique multiple cloning site for BamHI, SalI, PstI, and HindIII such as the pMAL vector (NEB).
  • the H N /A DNA (SEQ ID2) is then cleaved with PstI+HindIII restriction enzymes and inserted into the similarly cleaved pMAL-LC/A-linker construct.
  • the final construct contains the LC/A-CPN(GS25)-H N /A ORF (SEQ ID60) for expression as a protein of the sequence illustrated in SEQ ID61.
  • FIG. 9 illustrates the purified product obtained in the case of LC/A-CPN(GS10)-H N /A, LC/A-CPN(GS15)-H N /A, LC/A-CPN(GS25)-H N /A, LC/A-CPN(GS30)-H N /A and LC/A-CPN(HX27)-H N /A.
  • Fusion protein prepared according to Examples 2 and 9 was assessed in the eDRG neuronal cell model.
  • dorsal root ganglia neurons are harvested from 15-day-old fetal Sprague-Dawley rats and dissociated cells plated onto 24-well plates coated with Matrigel at a density of 1 ⁇ 10 6 cells/well. One day post-plating the cells are treated with 10 ⁇ M cytosine ⁇ -D-arabinofuranoside for 48 h.
  • Cells are maintained in Dulbecco's minimal essential medium supplemented with 5% heat-inactivated fetal bovine serum, 5 mM L-glutamine, 0.6% D-glucose, 2% B27 supplement, and 100 ng/ml 2.5 S mouse nerve growth factor. Cultures are maintained for 2 weeks at 37° C. in 95% air/5% CO 2 before addition of test materials.
  • eDRG Release of substance P from eDRG is assessed by enzyme-linked immunosorbent assay. Briefly, eDRG cells are washed twice with low potassium-balanced salt solution (BSS: 5 mM KCl, 137 mM NaCl, 1.2 mM MgCl 2 , 5 mM glucose, 0.44 mM KH 2 PO 4 , 20 mM HEPES, pH 7.4, 2 mM CaCl 2 ). Basal samples are obtained by incubating each well for 5 min. with 1 ml of low potassium BSS.
  • BSS potassium-balanced salt solution
  • the cells are stimulated to release by incubation with 1 ml of high potassium buffer (BSS as above with modification to include 100 mM KCl isotonically balanced with NaCl) for 5 min. All samples are removed to tubes on ice prior to assay of substance P. Total cell lysates are prepared by addition of 250 ⁇ l of 2 M acetic acid/0.1% trifluoroacetic acid to lyse the cells, centrifugal evaporation, and resuspension in 500 ⁇ l of assay buffer. Diluted samples are assessed for substance P content. Substance P immunoreactivity is measured using Substance P Enzyme Immunoassay Kits (Cayman Chemical Company or R&D Systems) according to manufacturers' instructions. Substance P is expressed in pg/ml relative to a standard substance P curve run in parallel.
  • BSS high potassium buffer
  • All samples are removed to tubes on ice prior to assay of substance P.
  • Total cell lysates are prepared by addition of 250 ⁇ l of
  • SDS-PAGE and Western blot analysis were performed using standard protocols (Novex). SNAP-25 proteins were resolved on a 12% Tris/glycine polyacrylamide gel (Novex) and subsequently transferred to nitrocellulose membrane. The membranes were probed with a monoclonal antibody (SMI-81) that recognises cleaved and intact SNAP-25. Specific binding was visualised using peroxidase-conjugated secondary antibodies and a chemiluminescent detection system. Cleavage of SNAP-25 was quantified by scanning densitometry (Molecular Dynamics Personal SI, ImageQuant data analysis software). Percent SNAP-25 cleavage was calculated according to the formula: (Cleaved SNAP-25/(Cleaved+Intact SNAP-25)) ⁇ 100.
  • CPN-A LC/A-nociceptin-H N /A fusion
  • FIG. 11 illustrates the prolonged duration of action of the CPN-A fusion protein, with measurable activity still being observed at 28 days post exposure.
  • Fusion protein prepared according to Examples 8 and 9 was assessed in the eDRG neuronal cell mode using the method described in Example 16.
  • CPNv-A LC/A-nociceptin variant-H N /A fusion
  • FIG. 13 illustrates the prolonged duration of action of the CPN-A fusion protein, with measurable activity still being observed at 24 days post exposure.
  • FIG. 14 illustrates the results of a competition experiment to determine binding efficacy at the ORL-1 receptor.
  • CPNv-A is demonstrated to displace [3H]-nociceptin, thereby confirming that access to the receptor is possible with the ligand in the central presentation format.
  • the LC/A (SEQ ID1) and H N /A (SEQ ID2) are created and inserted into the A serotype nociceptin variant linker arranged as BamHI-Sa/1-linker-enterokinase protease site-nociceptin variant-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID62).
  • the final construct contains the LC-linker-nociceptin variant-spacer-H N ORF sequences (SEQ ID63) for expression as a protein of the sequence illustrated in SEQ ID64.
  • the fusion protein is termed CPNv(Ek)-A.
  • FIG. 15 illustrates the purification of CPNv(Ek)-A from E. coli following the methods used in Example 9 but using Enterokinase for activation at 0.00064 ⁇ g per 100 ⁇ g of fusion protein.
  • Example 18 The CPNv(Ek)-A prepared in Example 18 is obtained in a purified form and applied to the eDRG cell model to assess cleavage of SNAP-25 (using methodology from Example 16).
  • FIG. 16 illustrates the cleavage of SNAP-25 following 24 h exposure of eDRG to CPNv(Ek)-A. The efficiency of cleavage is observed to be similar to that achieved with the Factor Xa-cleaved material, as recorded in Example 17.
  • the LC/C (SEQ ID5) and H N /C (SEQ ID6) are created and inserted into the A serotype nociceptin variant linker arranged as BamHI-SalI-linker-nociceptin variant-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID65).
  • the final construct contains the LC-linker-nociceptin variant-spacer-H N ORF sequences (SEQ ID66) for expression as a protein of the sequence illustrated in SEQ ID67.
  • the fusion protein is termed CPNv-C (act. A).
  • FIG. 17 illustrates the purification of CPNv-C (act. A) from E. coli following the methods used in Example 9.
  • Example 18 illustrates the cleavage of syntaxin following 24 h exposure of eDRG to CPNv-C (act. A).
  • LC/A-nociceptin-H N /A fusion (CPN/A) to inhibit acute capsaicin-induced mechanical allodynia is evaluated following subcutaneous intraplantar injection in the rat hind paw.
  • Test animals are evaluated for paw withdrawal frequency (PWF %) in response to a 10 g Von Frey filament stimulus series (10 stimuli ⁇ 3 trials) prior to recruitment into the study, after subcutaneous treatment with CPN/A but before capsaicin, and following capsaicin challenge post-injection of CPN/A (average of responses at 15′ and 30′).
  • Capsaicin challenge is achieved by injection of 10 ⁇ L of a 0.3% solution. Sample dilutions are prepared in 0.5% BSA/saline.
  • FIG. 19 illustrates the reversal of mechanical allodynia that is achieved by pre-treatment of the animals with a range of concentrations of LC/A-nociceptin-H N /A fusion.
  • STZ-induced mechanical allodynia in rats is achieved by injection of streptozotocin (i.p. or i.v.) which yields destruction of pancreatic ⁇ -cells leading to loss of insulin production, with concomitant metabolic stress (hyperglycemia and hyperlipidemia).
  • streptozotocin i.p. or i.v.
  • STZ induces Type I diabetes.
  • STZ treatment leads to progressive development of neuropathy, which serves as a model of chronic pain with hyperalgesia and allodynia that may reflect signs observed in diabetic humans (peripheral diabetic neuropathy).
  • Test materials (20-25 ⁇ l) are injected subcutaneously as a single injection (except gabapentin) and the PWT is measured at 1 day post-treatment and periodically thereafter over a 2-week period.
  • Gabapentin (30 mg/kg i.p. @ 3 ml/kg injection volume) is injected daily, 2 hours prior to the start of PWT testing.
  • FIG. 20 illustrates the reversal of allodynia achieved by pre-treatment of the animals with 750 ng of CPN/A. Data were obtained over a 2-week period after a single injection of CPN/A
  • LC/A-nociceptin variant-H N /A fusion (CPNv/A) to inhibit capsaicin-induced mechanical allodynia is evaluated following subcutaneous intraplantar injection in the rat hind paw.
  • Test animals are evaluated for paw withdrawal frequency (PWF %) in response to a 10 g Von Frey filament stimulus series (10 stimuli ⁇ 3 trials) prior to recruitment into the study (Pre-Treat); after subcutaneous intraplantar treatment with CPNv/A but before capsaicin (Pre-CAP); and following capsaicin challenge post-injection of CPNv/A (average of responses at 15′ and 30′; CAP).
  • Capsaicin challenge is achieved by injection of 10 ⁇ L of a 0.3% solution. Sample dilutions are prepared in 0.5% BSA/saline.
  • FIG. 21 illustrates the reversal of allodynia that is achieved by pre-treatment of the animals with a range of concentrations of LC/A-nociceptin variant-H N /A fusion in comparison to the reversal achieved with the addition of LC/A-nociceptin-H N /A fusion.
  • These data are expressed as a normalized paw withdrawal frequency differential, in which the difference between the peak response (post-capsaicin) and the baseline response (pre-capsaicin) is expressed as a percentage.
  • LC/A-leu enkephalin-H N /A fusion Due to the small, five-amino acid, size of the leu-enkephalin ligand the LC/A-leu enkephalin-H N /A fusion is created by site directed mutagenesis [for example using Quickchange (Stratagene Inc.)] using the LC/A-nociceptin-H N /A fusion (SEQ ID13) as a template. Oligonucleotides are designed encoding the YGGFL leu-enkephalin peptide, ensuring standard E.
  • FIG. 22 illustrates the purification of CPLE-A from E. coli following the methods used in Example 9.
  • Example 7 Following the methods used in Example 9, and with the LC/A-beta-endorphin-H N /A fusion protein (termed CPBE-A) created in Example 7, the CPBE-A is purified from E. coli .
  • FIG. 23 illustrates the purified protein as analysed by SDS-PAGE.
  • the LC/A-nociceptin mutant-H N /A fusion is created by site directed mutagenesis [for example using Quickchange (Stratagene Inc.)] using the LC/A-nociceptin-H N /A fusion (SEQ ID13) as a template. Oligonucleotides are designed encoding tyrosine at position 1 of the nociceptin sequence, ensuring standard E.
  • FIG. 24 illustrates the purification of CPOP-A from E. coli following the methods used in Example 9.
  • the LC/A-nociceptin variant mutant-H N /A fusion is created by site directed mutagenesis [for example using Quickchange (Stratagene Inc.)] using the LC/A-nociceptin variant-H N /A fusion (SEQ ID25) as a template. Oligonucleotides are designed encoding tyrosine at position 1 of the nociceptin sequence, ensuring standard E.
  • FIG. 25 illustrates the purification of CPOPv-A from E. coli following the methods used in Example 9.
  • FIG. 26 illustrates that CPOPv-A is able to cleave SNAP-25 in the eDRG model, achieving cleavage of 50% of the maximal SNAP-25 after exposure of the cells to approximately 5.9 nM fusion for 24 h.
  • the IgA protease amino acid sequence was obtained from freely available database sources such as GenBank (accession number P09790). Information regarding the structure of the N. Gonorrhoeae IgA protease gene is available in the literature (Pohlner et al., Gene structure and extracellular secretion of Neisseria gonorrhoeae IgA protease, Nature, 1987, 325(6103), 458-62). Using Backtranslation tool v2.0 (Entelechon), the DNA sequence encoding the IgA protease modified for E. coli expression was determined.
  • a BamHI recognition sequence was incorporated at the 5′ end and a codon encoding a cysteine amino acid and SalI recognition sequence were incorporated at the 3′ end of the IgA DNA.
  • the DNA sequence was screened using MapDraw, (DNASTAR Inc.) for restriction enzyme cleavage sequences incorporated during the back translation. Any cleavage sequences that are found to be common to those required for cloning were removed manually from the proposed coding sequence ensuring common E. coli codon usage is maintained. E. coli codon usage was assessed Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables. This optimised DNA sequence (SEQ ID74) containing the IgA open reading frame (ORF) is then commercially synthesized.
  • the IgA (SEQ ID74) is inserted into the LC-linker-nociceptin variant-spacer-H N ORF (SEQ ID25) using BamHI and SalI restriction enzymes to replace the LC with the IgA protease DNA.
  • the final construct contains the IgA-linker-nociceptin variant-spacer-H N ORF (SEQ ID75) for expression as a protein of the sequence illustrated in SEQ ID76.
  • DNA was prepared that encoded a Factor Xa removable his-tag (his6), although it is clear that alternative proteases site such as Enterokinase and alternative purification tags such as longer histidine tags are also possible.
  • reverse translation software tools for example EditSeq best E. coli reverse translation (DNASTAR Inc.), or Backtranslation tool v2.0 (Entelechon)]
  • the DNA sequence encoding the Factor Xa removable his-tag region is determined. Restriction sites are then incorporated into the DNA sequence and can be arranged as NheI-linker-SpeI-PstI-H N /A-XbaI-LEIEGRSGHHHHHHStop codon-HindIII (SEQ ID77).
  • E. coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example GenBank Release 143, 13 Sep. 2004).
  • This optimised DNA sequence is then commercially synthesized (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector.
  • CPNv-A-FXa-HT (SEQ ID78, removable his-tag construct)
  • the pCR 4 vector encoding the removable his-tag is cleaved with NheI and HindIII.
  • the NheI-HindIII fragment is then inserted into the LC/A-CPNv-H N /A vector (SEQ ID25) that has also been cleaved by NheI and HindIII.
  • the final construct contains the LC/A-linker-nociceptin variant-spacer-H N -FXa-Histag-HindIII ORF sequences (SEQ ID78) for expression as a protein of the sequence illustrated in SEQ ID79.
  • FIG. 27 illustrates the purification of CPNv-A-FXa-HT from E. coli following the methods used in Example 9.
  • the DNA sequence is designed by back translation of the amino acid sequence of the translocation domain of the diphtheria toxin (obtained from freely available database sources such as GenBank (accession number 1XDTT) using one of a variety of reverse translation software tools [for example EditSeq best E. coli reverse translation (DNASTAR Inc.), or Backtranslation tool v2.0 (Entelechon)]. Restriction sites are then incorporated into the DNA sequence and can be arranged as NheI-Linker-SpeI-PstI-diphtheria translocation domain-XbaI-stop codon-HindIII (SEQ ID80).
  • PstI/XbaI recognition sequences are incorporated at the 5′ and 3′ ends of the translocation domain respectively of the sequence maintaining the correct reading frame.
  • the DNA sequence is screened (using software such as MapDraw, DNASTAR Inc.) for restriction enzyme cleavage sequences incorporated during the back translation. Any cleavage sequences that are found to be common to those required by the cloning system are removed manually from the proposed coding sequence ensuring common E. coli codon usage is maintained. E. coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example GenBank Release 143, 13 Sep. 2004).
  • This optimised DNA sequence containing the diphtheria translocation domain is then commercially synthesized as NheI-Linker-SpeI-PstI-diphtheria translocation domain-XbaI-stop codon-HindIII (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector (Invitrogen).
  • the pCR 4 vector encoding the diphtheria translocation domain is cleaved with NheI and XbaI.
  • the NheI-XbaI fragment is then inserted into the LC/A-CPLE-H N /A vector (SEQ ID68) that has also been cleaved by NheI and XbaI.
  • the final construct contains the LC/A-leu-enkephalin-spacer-diphtheria translocation domain ORF sequences (SEQ ID81) for expression as a protein of the sequence illustrated in SEQ ID82.
  • the DNA sequence is designed by back translation of the tetanus toxin LC amino acid sequence (obtained from freely available database sources such as GenBank (accession number X04436) using one of a variety of reverse translation software tools [for example EditSeq best E. coli reverse translation (DNASTAR Inc.), or Backtranslation tool v2.0 (Entelechon)].
  • BamHI/SalI recognition sequences are incorporated at the 5′ and 3′ ends respectively of the sequence maintaining the correct reading frame (SEQ ID83).
  • the DNA sequence is screened (using software such as MapDraw, DNASTAR Inc.) for restriction enzyme cleavage sequences incorporated during the back translation.
  • E. coli codon usage is assessed by reference to software programs such as Graphical Codon Usage Analyser (Geneart), and the % GC content and codon usage ratio assessed by reference to published codon usage tables (for example GenBank Release 143, 13 Sep. 2004).
  • This optimised DNA sequence containing the tetanus toxin LC open reading frame (ORF) is then commercially synthesized (for example by Entelechon, Geneart or Sigma-Genosys) and is provided in the pCR 4 vector (invitrogen).
  • the pCR 4 vector encoding the TeNT LC is cleaved with BamHI and SalI.
  • the BamHI-SalI fragment is then inserted into the LC/A-CPNv-H N /A vector (SEQ ID25) that has also been cleaved by BamHI and SalI.
  • the final construct contains the TeNT LC-linker-nociceptin variant-spacer-H N ORF sequences (SEQ ID84) for expression as a protein of the sequence illustrated in SEQ ID85.
  • the LC/C (SEQ ID5) and H N /C (SEQ ID6) are created and inserted into the C serotype nociceptin variant linker arranged as BamHI-SalI-linker-nociceptin variant-NheI-spacer-SpeI-PstI-XbaI-stop codon-HindIII (SEQ ID86).
  • the final construct contains the LC-linker-nociceptin variant-spacer-H N ORF sequences (SEQ ID87) for expression as a protein of the sequence illustrated in SEQ ID88.
  • the fusion protein is termed CPNv-C (act. C).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Neurology (AREA)
  • Pain & Pain Management (AREA)
  • Neurosurgery (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Rheumatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US12/303,078 2006-06-01 2007-06-01 Treatment of pain Abandoned US20100034802A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB0610867.4A GB0610867D0 (en) 2006-06-01 2006-06-01 Treatment of pain
GB0610867.4 2006-06-01
PCT/GB2007/002049 WO2007138339A2 (en) 2006-06-01 2007-06-01 Treatment of pain by use of a single chain polypeptide fusion protein

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2007/002049 A-371-Of-International WO2007138339A2 (en) 2006-06-01 2007-06-01 Treatment of pain by use of a single chain polypeptide fusion protein

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/360,565 Continuation US9072736B2 (en) 2006-06-01 2012-01-27 Treatment of pain
US13/419,381 Continuation-In-Part US9243301B2 (en) 2006-06-01 2012-03-13 Fusion proteins

Publications (1)

Publication Number Publication Date
US20100034802A1 true US20100034802A1 (en) 2010-02-11

Family

ID=36694795

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/303,078 Abandoned US20100034802A1 (en) 2006-06-01 2007-06-01 Treatment of pain
US12/868,510 Abandoned US20110091437A1 (en) 2006-06-01 2010-08-25 Fusion proteins
US13/360,565 Active US9072736B2 (en) 2006-06-01 2012-01-27 Treatment of pain
US13/419,381 Expired - Fee Related US9243301B2 (en) 2006-06-01 2012-03-13 Fusion proteins

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/868,510 Abandoned US20110091437A1 (en) 2006-06-01 2010-08-25 Fusion proteins
US13/360,565 Active US9072736B2 (en) 2006-06-01 2012-01-27 Treatment of pain
US13/419,381 Expired - Fee Related US9243301B2 (en) 2006-06-01 2012-03-13 Fusion proteins

Country Status (8)

Country Link
US (4) US20100034802A1 (es)
EP (1) EP2046370B1 (es)
CN (2) CN101495135B (es)
BR (1) BRPI0713355E8 (es)
ES (1) ES2607789T3 (es)
GB (1) GB0610867D0 (es)
MX (1) MX337729B (es)
WO (1) WO2007138339A2 (es)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090162341A1 (en) * 2004-12-01 2009-06-25 Keith Foster Non-Cytotoxic Protein Conjugates
US20110027256A1 (en) * 2004-12-01 2011-02-03 Syntaxin Ltd. Fusion proteins
WO2012051447A1 (en) 2010-10-14 2012-04-19 Allergan, Inc. Targeted delivery of targeted exocytosis modulators to the sphenopalatine ganglion for treatment of headache disorders
WO2012134904A1 (en) 2011-03-28 2012-10-04 Allergan, Inc. Endopeptidase treatment of involuntary movement disorders
WO2012135448A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Endopeptidase treatment of smooth muscle disorders
WO2012135343A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Endopeptidase treatment of sexual dysfunction disorders
WO2012135304A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Vagal nerve-based disorders
WO2012134897A1 (en) 2011-03-25 2012-10-04 Allergan, Inc. Treatment of sensory disturbance disorders
WO2012134900A1 (en) 2011-03-28 2012-10-04 Allergan, Inc. Endopeptidase treatment of neuroendocrine disorders
WO2012134902A1 (en) 2011-03-28 2012-10-04 Allergan, Inc. Endopeptidase and neurotoxin combination treatment of dystonia, cerebral palsy and migraine
WO2012134901A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Re-targeted endopeptidases for use in the treatment of skin diseases
WO2012174123A1 (en) 2011-06-13 2012-12-20 Allergan, Inc. Treatment of psychological trauma
WO2013102063A1 (en) 2011-12-29 2013-07-04 Allergan, Inc. Endopeptidase and neurotoxin combination treatment of bladder disorders
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
US8697090B2 (en) 2011-05-05 2014-04-15 Allergan, Inc. Method of treating persistent genital arousal disorder with a neurotoxin
WO2014100019A1 (en) 2012-12-18 2014-06-26 Allergan, Inc. Prophylactic treatment of herpes recurrence
US8778634B2 (en) 2004-12-01 2014-07-15 Syntaxin, Ltd. Non-cytotoxic protein conjugates
WO2014117148A1 (en) 2013-01-28 2014-07-31 New York University Treatment methods using atoxic neurotoxin derivatives
US9144600B2 (en) 2011-07-14 2015-09-29 Allergan, Inc. Methods for treatment of incontinence associated with sexual activity
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 (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7192596B2 (en) 1996-08-23 2007-03-20 The Health Protection Agency Ipsen Limited Recombinant toxin fragments
GB9617671D0 (en) * 1996-08-23 1996-10-02 Microbiological Res Authority Recombinant toxin fragments
US20050191321A1 (en) * 2004-02-26 2005-09-01 Allergan, Inc. Methods for treating headache
US20100266638A1 (en) 2004-02-26 2010-10-21 Allergan, Inc. Headache treatment method
AU2011202219B2 (en) * 2005-12-01 2015-05-28 Allergan Inc. Fusion proteins
GB0610867D0 (en) 2006-06-01 2006-07-12 Syntaxin Ltd Treatment of pain
KR20100088683A (ko) 2007-10-23 2010-08-10 알러간, 인코포레이티드 변형 클로스트리듐 독소를 사용한 만성 신경성 염증의 치료 방법
CN101918021A (zh) 2007-10-23 2010-12-15 阿勒根公司 使用经修饰的梭菌毒素治疗泌尿生殖系统神经疾病的方法
EP3473643A1 (en) 2008-06-12 2019-04-24 Ipsen Bioinnovation Limited Fusion proteins for use in the treatemnt of cancer
US20100303757A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Interleukin Retargeted Endopepidases
US11224659B2 (en) 2010-10-29 2022-01-18 Infirst Healthcare Limited Solid solution compositions and use in severe pain
US9744132B2 (en) 2010-10-29 2017-08-29 Infirst Healthcare Limited Solid solution compositions and use in chronic inflammation
US9271950B2 (en) 2010-10-29 2016-03-01 Infirst Healthcare Limited Compositions for treating chronic inflammation and inflammatory diseases
US10695431B2 (en) 2010-10-29 2020-06-30 Infirst Healthcare Limited Solid solution compositions and use in cardiovascular disease
US10695432B2 (en) 2010-10-29 2020-06-30 Infirst Healthcare Limited Solid solution compositions and use in severe pain
US8895537B2 (en) 2010-10-29 2014-11-25 Infirst Healthcare Ltd. Compositions and methods for treating cardiovascular diseases
US9504664B2 (en) 2010-10-29 2016-11-29 Infirst Healthcare Limited Compositions and methods for treating severe pain
US9308213B2 (en) 2010-10-29 2016-04-12 Infirst Healthcare Limited Solid solution compositions and use in chronic inflammation
US11730709B2 (en) 2010-10-29 2023-08-22 Infirst Healthcare Limited Compositions and methods for treating severe pain
US11202831B2 (en) 2010-10-29 2021-12-21 Infirst Healthcare Limited Solid solution compositions and use in cardiovascular disease
GB201108108D0 (en) 2011-05-16 2011-06-29 Syntaxin Ltd Therapeutic fusion proteins
US8992941B2 (en) 2011-07-08 2015-03-31 Allergan, Inc. Method for treatment of esophageal spasm
US9005628B2 (en) * 2012-10-04 2015-04-14 Dublin City University Biotherapy for pain
CN104870468B (zh) * 2012-11-21 2020-11-10 益普生生物创新有限公司 用于制备经蛋白水解处理的多肽的方法
JP6474732B2 (ja) * 2013-01-14 2019-02-27 インファースト ヘルスケア リミテッド 激痛治療用組成物と方法
JP6389190B2 (ja) * 2013-01-14 2018-09-12 インファースト ヘルスケア リミテッド 固溶体組成物および慢性炎症における使用
GB201312295D0 (en) * 2013-07-09 2013-08-21 Syntaxin Ltd Suppression of itch
WO2016025626A2 (en) * 2014-08-12 2016-02-18 Biomadison, Inc. Botulinum neurotoxins with modified light chain specifity and methods for producing same
KR20180070563A (ko) 2015-08-27 2018-06-26 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 통증 치료용 조성물 및 방법
ES2914590T3 (es) 2015-11-09 2022-06-14 Tufts College Compuestos y métodos de tratamiento del dolor
TW201814045A (zh) 2016-09-16 2018-04-16 英商艾普森生物製藥有限公司 製造雙鏈梭狀芽孢桿菌神經毒素之方法
JP7118055B2 (ja) 2016-09-29 2022-08-15 イプセン バイオファーム リミテッド ハイブリッド神経毒
EP3312290A1 (en) 2016-10-18 2018-04-25 Ipsen Biopharm Limited Cellular vamp cleavage assay
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
EP3796929A1 (en) * 2018-05-21 2021-03-31 Ipsen Biopharm Limited Suppression of bone cancer-induced allodynia
GB201900621D0 (en) * 2019-01-16 2019-03-06 Ipsen Biopharm Ltd Labelled polypeptides

Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136564A (en) * 1995-11-16 2000-10-24 Roche Diagnostics Gmbh Process for the production of peptides by way of streptavidin fusion proteins
US6395513B1 (en) * 1995-04-21 2002-05-28 The Speywood Laboratory, Ltd. Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US6461617B1 (en) * 1996-08-23 2002-10-08 Microbiological Research Authority Recombinant toxin fragments
US20040115727A1 (en) * 2002-12-11 2004-06-17 Allergan, Inc., A Corporation Evolved clostridial toxins with altered protease specificity
US6776990B2 (en) * 1999-04-08 2004-08-17 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
US7056729B2 (en) * 2000-01-19 2006-06-06 Allergan, Inc. Botulinum neurotoxin-substance P conjugate or fusion protein for treating pain
US7132259B1 (en) * 1999-08-25 2006-11-07 Allergan, Inc. Activatable recombinant neurotoxins
US20070010475A1 (en) * 2002-12-09 2007-01-11 Peter Richardson Use of spongosine (2-methoxyadenosein) for the treatment of pain, in particular hyperalgesia
US20070066559A1 (en) * 2003-03-07 2007-03-22 Peter Richardson Compounds for the treatment of pain
US20080032931A1 (en) * 1999-08-25 2008-02-07 Steward Lance E Activatable clostridial toxins
US20090018081A1 (en) * 1999-08-25 2009-01-15 Allergan, Inc. Activatable clostridial toxins
US7494661B2 (en) * 2000-06-28 2009-02-24 Ira Sanders Methods for using tetanus toxin for beneficial purposes in animals (mammals)
US7514088B2 (en) * 2005-03-15 2009-04-07 Allergan, Inc. Multivalent Clostridial toxin derivatives and methods of their use
US20090104234A1 (en) * 2007-10-23 2009-04-23 Allergan, Inc. Methods of treating chronic neurogenic inflammation using modified clostridial toxins
US20090117157A1 (en) * 2007-10-23 2009-05-07 Allergan, Inc. Methods of treating urogenital-neurological disorders using modified clostridial toxins
US20090162341A1 (en) * 2004-12-01 2009-06-25 Keith Foster Non-Cytotoxic Protein Conjugates
US7659092B2 (en) * 2004-12-01 2010-02-09 Syntaxin, Ltd. Fusion proteins
US20100055761A1 (en) * 2006-07-20 2010-03-04 The General Hospital Corporation Methods, compositions, and kits for the selective activation of protoxins through combinatoral targeting
US7740868B2 (en) * 1999-08-25 2010-06-22 Allergan, Inc. Activatable clostridial toxins
US20100196421A1 (en) * 2004-11-22 2010-08-05 New York University Genetically engineered clostridial genes, proteins encoded by the engineered genes, and uses thereof
US20100209955A1 (en) * 2009-01-20 2010-08-19 Oyler George A Methods For The Delivery Of Toxins Or Enzymatically Active Portions Thereof
US20100303789A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Neurotrophin Retargeted Endopepidases
US20100303791A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Glucagon Like Hormone Retargeted Endopepidases
US20100303757A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Interleukin Retargeted Endopepidases
US20110027256A1 (en) * 2004-12-01 2011-02-03 Syntaxin Ltd. Fusion proteins

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7192596B2 (en) 1996-08-23 2007-03-20 The Health Protection Agency Ipsen Limited Recombinant toxin fragments
GB9721189D0 (en) 1997-10-08 1997-12-03 Speywood Lab The Limited Analgesic conjugates
DK1234043T3 (da) 1999-12-02 2004-07-19 Health Prot Agency Konstruktion for afgivelse af terapeutiske midler til neuroceller
AU2002343489A1 (en) * 2001-10-09 2003-04-22 Synvax, Inc. Nociceptin-based analgesics
ITMI20022022A1 (it) 2002-09-24 2004-03-25 Girolamo Calo' Analoghi di nocicettina.
WO2006026780A1 (en) 2004-09-01 2006-03-09 Allergan, Inc. Degradable clostridial toxins
GB0426397D0 (en) * 2004-12-01 2005-01-05 Health Prot Agency Fusion proteins
CA2595115C (en) * 2004-12-01 2014-01-21 Health Protection Agency Fusion proteins for treating, preventing or ameliorating pain
US8778634B2 (en) * 2004-12-01 2014-07-15 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8603779B2 (en) * 2004-12-01 2013-12-10 Syntaxin, Ltd. Non-cytotoxic protein conjugates
AU2005311098B2 (en) 2004-12-01 2011-08-11 Allergan, Inc. Non-cytotoxic protein conjugates
US8512984B2 (en) * 2004-12-01 2013-08-20 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US7817957B2 (en) 2005-07-08 2010-10-19 Ricoh Company, Ltd. Double feed sensing device, double feed determining method and image forming apparatus
GB0610867D0 (en) 2006-06-01 2006-07-12 Syntaxin Ltd Treatment of pain
EP2568285B1 (en) 2009-03-13 2019-12-25 Allergan, Inc. Immuno-based retargeted endopeptidase activity assays
GB201108108D0 (en) 2011-05-16 2011-06-29 Syntaxin Ltd Therapeutic fusion proteins
US20140056870A1 (en) * 2012-08-27 2014-02-27 Allergan, Inc. Fusion proteins

Patent Citations (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6395513B1 (en) * 1995-04-21 2002-05-28 The Speywood Laboratory, Ltd. Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US7892560B2 (en) * 1995-04-21 2011-02-22 Syntaxin, Ltd Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US20060051356A1 (en) * 1995-04-21 2006-03-09 Health Protection Agency Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US7887810B2 (en) * 1995-04-21 2011-02-15 Syntaxin, Ltd Clostridial toxin derivatives able to modify peripheral sensory afferent functions
US6136564A (en) * 1995-11-16 2000-10-24 Roche Diagnostics Gmbh Process for the production of peptides by way of streptavidin fusion proteins
US6461617B1 (en) * 1996-08-23 2002-10-08 Microbiological Research Authority Recombinant toxin fragments
US7276473B2 (en) * 1999-04-08 2007-10-02 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
US6776990B2 (en) * 1999-04-08 2004-08-17 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
US6843998B1 (en) * 1999-04-08 2005-01-18 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
US20050095251A1 (en) * 1999-04-08 2005-05-05 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
US20080025994A1 (en) * 1999-04-08 2008-01-31 Allergan, Inc. Methods and compositions for the treatment of pancreatitis
US7749514B2 (en) * 1999-08-25 2010-07-06 Allergan, Inc. Activatable clostridial toxins
US20090069238A1 (en) * 1999-08-25 2009-03-12 Allergan, Inc. Activatable clostridial toxins
US7132259B1 (en) * 1999-08-25 2006-11-07 Allergan, Inc. Activatable recombinant neurotoxins
US7740868B2 (en) * 1999-08-25 2010-06-22 Allergan, Inc. Activatable clostridial toxins
US7709228B2 (en) * 1999-08-25 2010-05-04 Allergan, Inc. Activatable recombinant neurotoxins
US7897157B2 (en) * 1999-08-25 2011-03-01 Allergan, Inc. Activatable clostridial toxins
US20080032931A1 (en) * 1999-08-25 2008-02-07 Steward Lance E Activatable clostridial toxins
US20080182294A1 (en) * 1999-08-25 2008-07-31 Allergan, Inc. Activatable recombinant neurotoxins
US20090087458A1 (en) * 1999-08-25 2009-04-02 Allergan, Inc. Activatable recombinant neurotoxins
US7419676B2 (en) * 1999-08-25 2008-09-02 Allergan, Inc. Activatable recombinant neurotoxins
US7422877B2 (en) * 1999-08-25 2008-09-09 Allergan, Inc. Activatable recombinant neurotoxins
US20080311622A1 (en) * 1999-08-25 2008-12-18 Allergan, Inc. Activatable recombinant neurotoxins
US20090004224A1 (en) * 1999-08-25 2009-01-01 Allergan, Inc. Activatable clostridial toxins
US20090005313A1 (en) * 1999-08-25 2009-01-01 Steward Lance E Activatable clostridial toxins
US20090018081A1 (en) * 1999-08-25 2009-01-15 Allergan, Inc. Activatable clostridial toxins
US20090030182A1 (en) * 1999-08-25 2009-01-29 Allergan, Inc. Activatable recombinant neurotoxins
US20090030188A1 (en) * 1999-08-25 2009-01-29 Allergan, Inc. Activatable recombinant neurotoxins
US20090042270A1 (en) * 1999-08-25 2009-02-12 Allergan, Inc. Activatable recombinant neurotoxins
US20090081730A1 (en) * 1999-08-25 2009-03-26 Allergan, Inc. Activatable recombinant neurotoxins
US7736659B2 (en) * 2000-01-19 2010-06-15 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7413742B2 (en) * 2000-01-19 2008-08-19 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7056729B2 (en) * 2000-01-19 2006-06-06 Allergan, Inc. Botulinum neurotoxin-substance P conjugate or fusion protein for treating pain
US7244437B2 (en) * 2000-01-19 2007-07-17 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7833535B2 (en) * 2000-01-19 2010-11-16 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7780968B2 (en) * 2000-01-19 2010-08-24 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7244436B2 (en) * 2000-01-19 2007-07-17 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7262291B2 (en) * 2000-01-19 2007-08-28 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7494661B2 (en) * 2000-06-28 2009-02-24 Ira Sanders Methods for using tetanus toxin for beneficial purposes in animals (mammals)
US20070010475A1 (en) * 2002-12-09 2007-01-11 Peter Richardson Use of spongosine (2-methoxyadenosein) for the treatment of pain, in particular hyperalgesia
US20040115727A1 (en) * 2002-12-11 2004-06-17 Allergan, Inc., A Corporation Evolved clostridial toxins with altered protease specificity
US20070066559A1 (en) * 2003-03-07 2007-03-22 Peter Richardson Compounds for the treatment of pain
US20100196421A1 (en) * 2004-11-22 2010-08-05 New York University Genetically engineered clostridial genes, proteins encoded by the engineered genes, and uses thereof
US7785606B2 (en) * 2004-11-22 2010-08-31 New York University Genetically engineered clostridial genes, proteins encoded by the engineered genes, and uses thereof
US7659092B2 (en) * 2004-12-01 2010-02-09 Syntaxin, Ltd. Fusion proteins
US20110027256A1 (en) * 2004-12-01 2011-02-03 Syntaxin Ltd. Fusion proteins
US7658933B2 (en) * 2004-12-01 2010-02-09 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US20100247509A1 (en) * 2004-12-01 2010-09-30 Keith Foster Fusion Proteins
US20090162341A1 (en) * 2004-12-01 2009-06-25 Keith Foster Non-Cytotoxic Protein Conjugates
US7514088B2 (en) * 2005-03-15 2009-04-07 Allergan, Inc. Multivalent Clostridial toxin derivatives and methods of their use
US20100055761A1 (en) * 2006-07-20 2010-03-04 The General Hospital Corporation Methods, compositions, and kits for the selective activation of protoxins through combinatoral targeting
US20090104234A1 (en) * 2007-10-23 2009-04-23 Allergan, Inc. Methods of treating chronic neurogenic inflammation using modified clostridial toxins
US20090117157A1 (en) * 2007-10-23 2009-05-07 Allergan, Inc. Methods of treating urogenital-neurological disorders using modified clostridial toxins
US20100209955A1 (en) * 2009-01-20 2010-08-19 Oyler George A Methods For The Delivery Of Toxins Or Enzymatically Active Portions Thereof
US20100303757A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Interleukin Retargeted Endopepidases
US20100303791A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Glucagon Like Hormone Retargeted Endopepidases
US20100303789A1 (en) * 2009-05-29 2010-12-02 Allergan, Inc. Methods of Treating Chronic Neurogenic Inflammation Using Neurotrophin Retargeted Endopepidases

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10619146B2 (en) * 2004-12-01 2020-04-14 Ipsen Bioinnovation Limited Non-cytotoxic protein conjugates
US8067200B2 (en) 2004-12-01 2011-11-29 Syntaxin Ltd. Fusion proteins
US8399400B2 (en) 2004-12-01 2013-03-19 Syntaxin, Ltd. Fusion proteins
US20110177053A1 (en) * 2004-12-01 2011-07-21 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US9139635B2 (en) 2004-12-01 2015-09-22 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8778634B2 (en) 2004-12-01 2014-07-15 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8187834B2 (en) 2004-12-01 2012-05-29 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US20090162341A1 (en) * 2004-12-01 2009-06-25 Keith Foster Non-Cytotoxic Protein Conjugates
US20110027256A1 (en) * 2004-12-01 2011-02-03 Syntaxin Ltd. Fusion proteins
US20100247509A1 (en) * 2004-12-01 2010-09-30 Keith Foster Fusion Proteins
US9012195B2 (en) 2004-12-01 2015-04-21 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8603779B2 (en) 2004-12-01 2013-12-10 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8512984B2 (en) 2004-12-01 2013-08-20 Syntaxin, Ltd. Non-cytotoxic protein conjugates
US8940870B2 (en) 2004-12-01 2015-01-27 Syntaxin, Ltd. Fusion proteins
US20160369257A1 (en) * 2004-12-01 2016-12-22 Ipsen Bioinnovation Ltd. Non-cytotoxic protein conjugates
US9474807B2 (en) 2004-12-01 2016-10-25 Ipsen Bioinnovation Limited Non-cytotoxic protein conjugates
US8399401B2 (en) 2004-12-01 2013-03-19 Syntaxin, Ltd. Fusion proteins
AU2011315962B2 (en) * 2010-10-14 2016-06-02 Allergan, Inc. Targeted delivery of Targeted Exocytosis Modulators to the sphenopalatine ganglion for treatment of headache disorders
EP3173095A1 (en) 2010-10-14 2017-05-31 Allergan, Inc. Targeted delivery of targeted exocytosis modulators to the sphenopalatine ganglion for treatment of headache disorders
WO2012051447A1 (en) 2010-10-14 2012-04-19 Allergan, Inc. Targeted delivery of targeted exocytosis modulators to the sphenopalatine ganglion for treatment of headache disorders
WO2012134897A1 (en) 2011-03-25 2012-10-04 Allergan, Inc. Treatment of sensory disturbance disorders
WO2012134902A1 (en) 2011-03-28 2012-10-04 Allergan, Inc. Endopeptidase and neurotoxin combination treatment of dystonia, cerebral palsy and migraine
WO2012134900A1 (en) 2011-03-28 2012-10-04 Allergan, Inc. Endopeptidase treatment of neuroendocrine disorders
WO2012134904A1 (en) 2011-03-28 2012-10-04 Allergan, Inc. Endopeptidase treatment of involuntary movement disorders
WO2012135304A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Vagal nerve-based disorders
WO2012134901A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Re-targeted endopeptidases for use in the treatment of skin diseases
WO2012135448A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Endopeptidase treatment of smooth muscle disorders
WO2012135343A1 (en) 2011-03-29 2012-10-04 Allergan, Inc. Endopeptidase treatment of sexual dysfunction disorders
US8697090B2 (en) 2011-05-05 2014-04-15 Allergan, Inc. Method of treating persistent genital arousal disorder with a neurotoxin
US10456455B2 (en) 2011-06-13 2019-10-29 Allergan, Inc. Treatment of psychological trauma
WO2012174123A1 (en) 2011-06-13 2012-12-20 Allergan, Inc. Treatment of psychological trauma
US9764009B2 (en) 2011-06-13 2017-09-19 Allergan, Inc. Treatment of psychological trauma
US11077174B2 (en) 2011-06-13 2021-08-03 Allergan, Inc. Treatment of psychological trauma
US9480732B2 (en) 2011-07-14 2016-11-01 Allergan, Inc. Methods for treatment of incontinence associated with sexual activity
US9144600B2 (en) 2011-07-14 2015-09-29 Allergan, Inc. Methods for treatment of incontinence associated with sexual activity
US10034923B2 (en) 2011-07-14 2018-07-31 Allergan, Inc. Methods for treatment of incontinence associated with sexual activity
WO2013102063A1 (en) 2011-12-29 2013-07-04 Allergan, Inc. Endopeptidase and neurotoxin combination treatment of bladder disorders
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
WO2014100019A1 (en) 2012-12-18 2014-06-26 Allergan, Inc. Prophylactic treatment of herpes recurrence
EP2948174A4 (en) * 2013-01-28 2016-08-31 Univ New York TREATMENT PROCEDURE USING ATOXIC NEUROTOXIN DERIVATIVES
WO2014117148A1 (en) 2013-01-28 2014-07-31 New York University Treatment methods using atoxic neurotoxin derivatives

Also Published As

Publication number Publication date
WO2007138339A3 (en) 2008-11-27
CN101495135A (zh) 2009-07-29
US9243301B2 (en) 2016-01-26
MX337729B (es) 2016-03-16
CN103602650A (zh) 2014-02-26
US9072736B2 (en) 2015-07-07
BRPI0713355A2 (pt) 2012-08-21
EP2046370A2 (en) 2009-04-15
BRPI0713355E8 (pt) 2016-01-05
US20120230975A1 (en) 2012-09-13
CN101495135B (zh) 2013-10-30
BRPI0713355E2 (pt) 2015-09-08
EP2046370B1 (en) 2016-08-10
MX2008015227A (es) 2009-04-22
GB0610867D0 (en) 2006-07-12
CN103602650B (zh) 2018-03-27
US20120189610A1 (en) 2012-07-26
US20110091437A1 (en) 2011-04-21
ES2607789T3 (es) 2017-04-04
WO2007138339A2 (en) 2007-12-06

Similar Documents

Publication Publication Date Title
US9072736B2 (en) Treatment of pain
US7659092B2 (en) Fusion proteins
US11248219B2 (en) 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
US9012195B2 (en) Non-cytotoxic protein conjugates
US8603779B2 (en) Non-cytotoxic protein conjugates
EP2335718A1 (en) Non-cytotoxic protein conjugates
US10619146B2 (en) Non-cytotoxic protein conjugates
WO2007138336A2 (en) Treatment of pain by single chain, polypeptide fusion protein
AU2011202219B2 (en) Fusion proteins
US20170327810A1 (en) Fusion proteins and methods for treating, preventing or ameliorating pain
AU2011202225B2 (en) Non-cytotoxic protein conjugates

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALLERGAN, INC.,CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AOKI, KEI ROGER;FRANICS, JOSEPH;STEWARD, LANCE E.;SIGNING DATES FROM 20071109 TO 20071115;REEL/FRAME:020118/0327

AS Assignment

Owner name: ALLERGAN, INC.,CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AOKI, KEI ROGER;FRANCIS, JOSEPH;STEWARD, LANCE;SIGNING DATES FROM 20090302 TO 20090304;REEL/FRAME:022585/0699

AS Assignment

Owner name: SYNTAXIN LIMITED,UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FOSTER, KEITH;CHADDOCK, JOHN;MARKS, PHILIP;AND OTHERS;REEL/FRAME:022707/0855

Effective date: 20090304

STCB Information on status: application discontinuation

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