WO2008063603A2 - Methods, compositions, and kits for treating pain and pruritis - Google Patents

Methods, compositions, and kits for treating pain and pruritis Download PDF

Info

Publication number
WO2008063603A2
WO2008063603A2 PCT/US2007/024174 US2007024174W WO2008063603A2 WO 2008063603 A2 WO2008063603 A2 WO 2008063603A2 US 2007024174 W US2007024174 W US 2007024174W WO 2008063603 A2 WO2008063603 A2 WO 2008063603A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
channels
pain
voltage
receptor
Prior art date
Application number
PCT/US2007/024174
Other languages
French (fr)
Other versions
WO2008063603A3 (en
Inventor
Bruce P. Bean
Clifford J. Woolf
Original Assignee
President And Fellows Of Harvard College
The General Hospital Corporation
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
Priority to AU2007322033A priority Critical patent/AU2007322033B2/en
Priority to CA2668652A priority patent/CA2668652C/en
Priority to JP2009537235A priority patent/JP2010510227A/en
Priority to DK07862114.1T priority patent/DK2101819T3/en
Priority to ES07862114T priority patent/ES2402789T3/en
Priority to EP07862114A priority patent/EP2101819B1/en
Priority to US12/515,429 priority patent/US20100099772A1/en
Priority to KR1020147017308A priority patent/KR20140097446A/en
Application filed by President And Fellows Of Harvard College, The General Hospital Corporation filed Critical President And Fellows Of Harvard College
Priority to EP11007949.8A priority patent/EP2446903B1/en
Publication of WO2008063603A2 publication Critical patent/WO2008063603A2/en
Publication of WO2008063603A3 publication Critical patent/WO2008063603A3/en
Priority to US14/496,629 priority patent/US9603817B2/en
Priority to US15/470,324 priority patent/US10179116B2/en
Priority to US16/851,349 priority patent/US20210186906A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/075Ethers or acetals
    • A61K31/085Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/14Quaternary ammonium compounds, e.g. edrophonium, choline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • A61P23/02Local anaesthetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening

Definitions

  • Pain may occur in patients with cancer, which may be due to multiple causes; inflammation, compression, invasion, metastatic spread into bone or other tissues.
  • Activators of TRPVl receptors include but are not limited to capsaicin, eugenol arvanil (N-arachidonoylvanillamine), anandamide, 2- aminoethoxydiphenyl borate (2 APB), AM404, resiniferatoxin, phorbol 12- phenylacetate 13-acetate 20-homovanillate (PPAHV), olvanil (NE 19550), OLDA (N-oleoyldopamine), N-arachidonyldopamine ( ⁇ ADA), 6'- iodoresiniferatoxin (6'-IRTX), Cl 8 ⁇ -acylethanolamines, lipoxygenase derivatives such as 12-hydroperoxyeicosatetraenoic acid, inhibitor cysteine knot (ICK) peptides (vanillotoxins), piperine, MSK195 ( ⁇ -[2-(3,4- dimethylbenzyl)-3-(pivaloyloxy)propyl]-2-[4-
  • the invention features a pharmaceutical composition that includes a quarternary amine derivative or other charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, articaine, bupivicaine, mepivicaine, tocainide, prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene, and a pharmaceutically acceptable excipient.
  • C 7 _i 4 alkaryl is meant an alkyl substituted by an aryl group (e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl) having from 7 to 14 carbon atoms.
  • aryl group e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl
  • hydroxyalkyl is meant a chemical moiety with the formula -(R)- OH, wherein R is selected from Ci_ 7 alkyl, C 2 _ 7 alkenyl, C 2 _ 7 alkynyl, C 2- ⁇ heterocyclyl, C 6 _i 2 aryl, C 7 _ )4 alkaryl, C 3 _i 0 alkheterocyclyl, or Ci_ 7 heteroalkyl.
  • R lv is, independently, selected from from H, Ci -4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 2-4 heteroalkyl
  • X 1 is selected from -CR 1W R 1X -, -NR !Y C(O)-, -OC(O)-, -SC(O)-, -C(O)NR 12 -, -CO 2 -, and -OC(S)-; and each of R 1W , R 1X , R l ⁇ , and R 1Z is, independently, selected from H, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 2-4 heteroalkyl; R 1D is selected from H, Q -4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 2 _ 4 heteroalkyl; and each of R 1 E , R 1 F , and R 1 G is, independently, selected from Ci -4 alky
  • each of R 8A , R 8B , and R 8C is, independently, selected from H, halogen, Ci -4 alkyl, C 2-4 alkenyl, C 2 ⁇ alkynyl, C 2 _ 4 heteroalkyl, OR 8L , NR 8M R 8N , NR 8O C(O)R 8P , S(O)R 8Q , SO 2 R 8R R 8S , SO 2 NR 8T R 8U , SO 3 R 8V , CO 2 R 8W , C(O)R 8X , and C(O)NR 8Y R 8Z ; and each of R 8L , R 8M , R 8N , R 80 , R 8P , R 8Q , R 8R , R 8S , R 8T , R 8U , R 8V , R 8W , R 8X , R 8Y
  • R 10T and R 10V combine to form an alkylene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings.
  • exemplary compounds of formula X include N-guanidyl derivatives (e.g., -C(NH)NH 2 derivatives) and methylated quaternary ammonium derivatives.
  • a visual analog scale provides a measure of a one-dimensional quantity.
  • a VAS generally utilizes a representation of distance, such as a picture of a line with hash marks drawn at regular distance intervals, e.g., ten 1- cm intervals. For example, a patient can be asked to rank a sensation of pain or itch by choosing the spot on the line that best corresponds to the sensation of pain or itch, where one end of the line corresponds to "no pain” (score of 0 cm) or "no itch” and the other end of the line corresponds to "unbearable pain” or “unbearable itch” (score of 10 cm). This procedure provides a simple and rapid approach to obtaining quantitative information about how the patient is experiencing pain or itch.
  • the Lequesne index and the Western Ontario and McMaster Universities (WOMAC) osteoarthritis index assess pain, function, and stiffness in the knee and hip of OA patients using self-administered questionnaires. Both knee and hip are encompassed by the WOMAC, whereas there is one Lequesne questionnaire for the knee and a separate one for the hip. These questionnaires are useful because they contain more information content in comparison with VAS or Likert. Both the WOMAC index and the Lequesne index questionnaires have been extensively validated in OA, including in surgical settings (e.g., knee and hip arthroplasty). Their metric characteristics do not differ significantly.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Dermatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Pain & Pain Management (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Anesthesiology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

The invention features a method for inhibiting one or more voltage-gated ion channels in a cell by contacting the cell with (i) a first compound that activates a channel-forming receptor that is present on nociceptors and/or pruriceptors; and (ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the channels but does not substantially inhibit said channels when applied to the external face of the channels, wherein the second compound is capable of entering nociceptors or pruriceptors through the channel-forming receptor when the receptor is activated. The invention also features a quarternary amine derivative or other permanently or transiently charged derivative of a compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the channels but does not substantially inhibit said channels when applied to the external face of the channels.

Description

METHODS, COMPOSITIONS, AND KITS FOR TREATING PAIN AND
PRURITIS Background of the Invention
The invention features methods, compositions, and kits for selective inhibition of pain-and itch sensing neurons (nociceptors and pruriceptors) by drug molecules of small molecule weight, while minimizing effects on non- pain-sensing neurons or other types of cells. According to the method of the invention, small, hydrophilic drug molecules gain access to the intracellular compartment of pain-sensing neurons via entry through receptors that are present in pain- and itch-sensing neurons but to a lesser extent or not at all in other types of neurons or in other types of tissue.
Local anesthetics such as lidocaine and articaine act by inhibiting voltage-dependent sodium channels in neurons. These anesthetics block sodium channels and thereby the excitability of all neurons, not just pain- sensing neurons (nociceptors). Thus, while the goal of topical or regional anesthesia is to block transmission of signals in nociceptors to prevent pain, administration of local anesthetics also produces unwanted or deletrious effects such as general numbness from block of low threshold pressure and touch receptors, motor deficits from block of motor axons and other complications from block of autonomic fibers. Local anesthetics are relatively hydrophobic molecules that gain access to their blocking site on the sodium channel by diffusing into or through the cell membrane. Permanently-charged derivatives of these compounds (such as QX-314, a quaternary nitrogen derivative of lidocaine), which are not membrane-permeant, have no effect on neuronal sodium channels when applied to the external surface of the nerve membrane but can block sodium channels if somehow introduced inside the cell, for example by a micropipette used for whole-cell electrophysiological recording from isolated neurons. Pain-sensing neurons differ from other types of neurons in expressing (in most cases) the TRPVl receptor/channel, activated by painful heat or by capsaicin, the pungent ingredient in chili pepper. Other types of receptors selectively expressed in various types of pain-sensing and itch- sensing (pruriceptor) neurons include but are not limited to TRPAl, TRPM8, and P2X(2/3) receptors.
Neuropathic, inflammatory, and nociceptive pain differ in their etiology, pathophysiology, diagnosis, and treatment. Nociceptive pain occurs in response to the activation of a specific subset of peripheral sensory neurons, the nociceptors by intense or noxious stimuli. It is generally acute, self-limiting and serves a protective biological function by acting as a warning of potential or on-going tissue damage. It is typically well-localized. Examples of nociceptive pain include but are not limited to traumatic or surgical pain, labor pain, sprains, bone fractures, burns, bumps, bruises, injections, dental procedures, skin biopsies, and obstructions.
Inflammatory pain is pain that occurs in the presence of tissue damage or inflammation including postoperative, post-traumatic pain, arthritic (rheumatoid or osteoarthritis) pain and pain associated with damage to joints, muscle, and tendons as in axial low back pain.
Neuropathic pain is a common type of chronic, non-malignant pain, which is the result of an injury or malfunction in the peripheral or central nervous system and serves no protective biological function. It is estimated to affect more than 1.6 million people in the U.S. population. Neuropathic pain has many different etiologies, and may occur, for example, due to trauma, surgery, herniation of an intervertebral disk, spinal cord injury, diabetes, infection with herpes zoster (shingles), HIV/AIDS, late-stage cancer, amputation (including mastectomy), carpal tunnel syndrome, chronic alcohol use, exposure to radiation, and as an unintended side-effect of neurotoxic treatment agents, such as certain anti-HIV and chemotherapeutic drugs.
In contrast to nociceptive pain, neuropathic pain is frequently described as "burning," "electric," "tingling," or "shooting" in nature. It is often characterized by chronic allodynia (defined as pain resulting from a stimulus that does not ordinarily elicit a painful response, such as light touch) and hyperalgesia (defined as an increased sensitivity to a normally painful stimulus), and may persist for months or years beyond the apparent healing of any damaged tissues.
Pain may occur in patients with cancer, which may be due to multiple causes; inflammation, compression, invasion, metastatic spread into bone or other tissues.
There are some conditions where pain occurs in the absence of a noxious stimulus, tissue damage or a lesion to the nervous system, called dysfunctional pain and these include but are not limited to fibromyalgia, tension type headache, irritable bowel disorders and erythermalgia.
Migraine is a headache associated with the activation of sensory fibers innervating the meninges of the brain.
Itch (pruritus) is a dermatological condition that may be localized and generalized and can be associated with skin lesions (rash, atopic eczema, wheals). Itch accompanies many conditions including but not limited to stress, anxiety, UV radiation from the sun, metabolic and endocrine disorders (e.g., liver or kidney disease, hyperthyroidism), cancers (e.g., lymphoma), reactions to drugs or food, parasitic and fungal infections, allergic reactions, diseases of the blood (e.g., polycythemia vera), and dermatological conditions. Itch is mediated by a subset of small diameter primary sensory neurons, the pruriceptor, that share many features of nociceptor neurons, including but not limited to expression of TRPVl channels. Certain itch mediators — such as eicosanoids, histamine, bradykinin, ATP, and various neurotrophins have endovanilloid functions. Topical capsaicin suppresses histamine-induced itch. Pruriceptors like nociceptors are therefore a suitable target for this method of delivering ion channels blockers.
Despite the development of a variety of therapies for pain and itch, there is a need for additional agents. Summary of the Invention
In a first aspect, the invention features a method for treating pain and itch (e.g., neuropathic pain, inflammatory pain, nociceptive pain, idiopathic pain, cancer pain, migraine, dysfunctional pain or procedural pain (e.g., dental procedures, injections, setting fractures, biopsies)) as well as pruritus in a patient by administering to the patient a first compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the channels but does not substantially inhibit the channels when applied to the external face of the channels, wherein the first compound is capable of entering neurons through a membrane bound receptor/ion channel when the receptor is activated; and, optionally, a second compound that activates a receptor through which the first compound can pass. In certain embodiments, the second compound activates a receptor selected from TRPVl, P2X(2/3), TRPAl, and TRPM8 through which the first compound can pass. Treatment of pain or itch can be determined using any standard pain or itch index, such as those described herein, or can be determined based on the patient's subjective pain or itch assessment. A patient is considered "treated" if there is a reported reduction in pain or a reduced reaction to stimuli that should cause pain and a reduction in itch. In certain embodiments, it is desirable to administer the second compound in order to ensure that the receptors (e.g., the TRPVl, P2X(2/3), TRPAl, and/or TRPM8 receptors) are activated, thus allowing for entry of the first compound. In other embodiments, because the receptors (e.g., the TRPVl, P2X(2/3), TRPAl, and/or TRPM8 receptors) are already activated, the second compound is not administered. Consequently, the first compound enters only neurons having receptors that are endogenously activated. In still other embodiments, the receptors (e.g., the TRPVl , P2X(2/3), TRPAl, and/or TRPM8 receptors) are activated by indicing a physiological state that activates these receptors, thus allowing for entry of the first compound. If desired, two or more compounds that activate TRPVl, P2X(2/3), TRPAl 5 and/or TRPM8 receptors can be employed, as can two or more compounds that inhibit one or more voltage-gated ion channels. Desirably, the first compound(s) and the second compound(s) are administered to the patient within 4 hours, 2 hours, 1 hour, 30 minutes, or 15 minutes of each other, or are administered substantially simultaneously. Importantly, either compound can be administered first. Thus, in one embodiment, one or more compounds that activate TRPVl, P2X(2/3), TRPAl, and/or TRPM8 receptors are administered first, while in another embodiment, one or more compounds that inhibit one or more voltage-gated ion channels when applied to the internal face of the channels but do not substantially inhibit the channels when applied to the external face of the channels are administered first. The compounds can be co- formulated into a single composition or can be formulated separately. Each of the compounds can be administered, for example, by oral, parenteral, intravenous, intramuscular, rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intrathecal, epidural, or ocular administration, or by injection, inhalation, or direct contact with the nasal or oral mucosa.
Activators of TRPVl receptors include but are not limited to capsaicin, eugenol arvanil (N-arachidonoylvanillamine), anandamide, 2- aminoethoxydiphenyl borate (2 APB), AM404, resiniferatoxin, phorbol 12- phenylacetate 13-acetate 20-homovanillate (PPAHV), olvanil (NE 19550), OLDA (N-oleoyldopamine), N-arachidonyldopamine (ΝADA), 6'- iodoresiniferatoxin (6'-IRTX), Cl 8 Ν-acylethanolamines, lipoxygenase derivatives such as 12-hydroperoxyeicosatetraenoic acid, inhibitor cysteine knot (ICK) peptides (vanillotoxins), piperine, MSK195 (Ν-[2-(3,4- dimethylbenzyl)-3-(pivaloyloxy)propyl]-2-[4-(2-aminoethoxy)-3- methoxyphenyl]acetamide), JYL79 (N-[2-(3,4-dimethylbenzyl)-3- (pivaloyloxy)propyl]-N'-(4-hydroxy-3-methoxybenzyl)thiourea), hydroxy- alpha-sanshool, 2-aminoethoxydiphenyl borate, 10-shogaol, oleylgingerol, oleylshogaol, and SU200 (N-(4-tert-butylbenzyl)-N'-(4-hydroxy-3- methoxybenzyl)thiourea). Other activators of TRPVl receptors are described in O1DeIl et al., Bioorg Med Chem (2007) 15:6164-6149, and Sexton et al., FASEB J (2007) 21 :2695-2703.
Activators of TRPAl receptors include but are not limited to cinnamaldehyde, allyl-isothiocynanate, diallyl disulfide, icilin, cinnamon oil, wintergreen oil, clove oil, acrolein, hydroxy-alpha-sanshool, 2- aminoethoxydiphenyl borate, 4-hydroxynonenal, methyl p-hydroxybenzoate, mustard oil, and 3'-carbamoylbiphenyl-3-yl cyclohexylcarbamate (URB597). Other activators of TRPAl receptors are described in Taylor-Clark et al., MoI Pharmacol (2007) PMID: 18000030; Macpherson et al., Nature (2007) 445:541-545; and Hill et al., J Biol Chem (2007) 282:7145-7153.
Activators of P2X receptors include but are not limited to ATP, 2- methylthio-ATP, T and 3'-O-(4-benzoylbenzoyl)-ATP, and ATP5'-O-(3- thiotriphosphate).
Activators of TRPM8 receptors include but are not limited to menthol, icilin, eucalyptol, linalool, geraniol, and hydroxycitronellal.
In certain embodiments, the first compound inhibits voltage-gated sodium channels. Exemplary inhibitors of this class are QX-314, N-methyl- procaine, QX-222, N-octyl-guanidine, 9-aminoacridine, and pancuronium.
In yet other embodiments, the first compound inhibits voltage-gated calcium channels. Exemplary inhibitors of this class are D-890 (quaternary methoxyverapamil) and CERM 11888 (quaternary bepridil).
In still other embodiments, the first compound is a quarternary amine derivative or other charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, articaine, bupivicaine, mepivicaine, tocainide, prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene. Exemplary derivatives are described herein.
The invention also features a quarternary amine derivative or other charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, articaine, bupivicaine, mepivicaine, tocainide, prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene.
In a related aspect, the invention features a pharmaceutical composition that includes a quarternary amine derivative or other charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, articaine, bupivicaine, mepivicaine, tocainide, prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene, and a pharmaceutically acceptable excipient.
The invention also features a composition that includes: (i) a first compound that activates a receptor selected from TRPVl, P2X(2/3), TRPAl, and TRPM8; and (ii) a second compound that inhibits one or more voltage- gated ion channels when applied to the internal face of these channels but does not substantially inhibit the channels when applied to their external face, wherein the second compound is capable of entering pain sensing neurons through TRPVl, P2X(2/3), TRPAl, and/or TRPM8 receptors when these receptors are activated. In one embodiment, the second compound is reduced in activity or partially active when applied to the external face, but more active when applied to the internal face. The composition can be formulated, for example, for oral, intravenous, intramuscular, rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intrathecal, epidural, or ocular administration, or by injection, inhalation, or direct contact with the nasal or oral mucosa. If desired, the composition can contain two or more compounds that activate TRPVl, P2X(2/3), TRPAl, and/or TRPM8 receptors, and/or two or more compound that inhibits one or more voltage-gated ion channels. The invention also features a method for inhibiting one or more voltage- gated ion channels in a cell by contacting the cell with: (i) a first compound that activates a receptor selected from TRPVl , P2X(2/3), TRPAl, and TRPM8; and (ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the channels but does not substantially inhibit the channels when applied to the external face of the channels, wherein said second compound is capable of entering pain sensing neurons through the receptor when the receptor is activated. Suitable compounds are provided above.
The invention also features a method for identifying a compound as being useful for the treatment of pain and itch. This method includes the steps of: (a) contacting the external face of TRPVl, TRPAl, TRPM8, and/or P2X(2/3)-expressing neurons with: (i) a first compound that activates TRPVl TRPAl, TRPM8 or P2X(2/3) receptors; and (ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the channels but does not substantially inhibit the channels when applied to the external face of the channels, and (b) determining whether the second compound inhibits the voltage-gated ion channels in the neurons. Inhibition of voltage-gated ion channels by the second compound identifies the second compound as a compound that is useful for the treatment of pain and/or itch.
The methods, compositions, and kits can also be used to selectively block neuronal activity in other types of neurons that express different members of the TRPV, TRPA, TRPM, and P2X receptor families, where the first compound is an agonist of the particular TRPV, TRPA, TRPM, and P2X receptor present in those types of neurons, and the second compound is a sodium or calcium channel blocker that is normally membrane impermeant. It is understood that other receptors may exist that would permit the entry of compounds that would otherwise be incapable of entering. Coadministration of compounds that activate one or more of these receptors in combination with one or more compounds that inhibit one or more voltage- gated ion channels when applied to the internal face of the channels but does not substantially inhibit the channels when applied to the external face of the channels is also an aspect of the invention.
The methods, compositions, and kits of the invention allow for a block of pain or itch without altering light touch or motor control. For example, patients receiving an epidural will not have a complete loss of sensory input.
The term "pain" is used herein in the broadest sense and refers to all types of pain, including acute and chronic pain, such as nociceptive pain, e.g. somatic pain and visceral pain; inflammatory pain, dysfunctional pain, idiopathic pain, neuropathic pain, e.g., centrally generated pain and peripherally generated pain, migraine, and cancer pain.
The term "nociceptive pain" is used to include all pain caused by noxious stimuli that threaten to or actually injure body tissues, including, without limitation, by a cut, bruise, bone fracture, crush injury, burn, and the like. Pain receptors for tissue injury (nociceptors) are located mostly in the skin, musculoskeletal system, or internal organs.
The term "somatic pain" is used to refer to pain arising from bone, joint, muscle, skin, or connective tissue. This type of pain is typically well localized.
The term "visceral pain" is used herein to refer to pain arising from visceral organs, such as the respiratory, gastrointestinal tract and pancreas, the urinary tract and reproductive organs. Visceral pain includes pain caused by tumor involvement of the organ capsule. Another type of visceral pain, which is typically caused by obstruction of hollow viscus, is characterized by intermittent cramping and poorly localized pain. Visceral pain may be associated with inflammation as in cystitis or reflux esophagitis. The term inflammatory pain includes pain associates with active inflammation that may be caused by trauma, surgery, infection and autoimmune diseases.
The term "neuropathic pain" is used herein to refer to pain originating from abnormal processing of sensory input by the peripheral or central nervous system consequent on a lesion to these systems.
The term "procedural pain" refers to pain arising from a medical, dental or surgical procedure wherein the procedure is usually planned or associated with acute trauma.
The term "itch" is used herein in the broadest sense and refers to all types of itching and stinging sensations localized and generalized, acute intermittent and persistent. The itch may be idiopathic, allergic, metabolic, infectious, drug-induced, due to liver, kidney disease, or cancer. "Pruritus" is severe itching.
By "patient" is meant any animal. In one embodiment, the patient is a human. Other animals that can be treated using the methods, compositions, and kits of the invention include but are not limited to non- human primates (e.g., monkeys, gorillas, chimpaneees), domesticated animals (e.g., horses, pigs, goats, rabbits, sheep, cattle, llamas), and companion animals (e.g., guinea pigs, rats, mice, lizards, snakes, dogs, cats, fish, hamsters, and birds). '
Compounds useful in the invention include but are not limited to those described herein in any of their pharmaceutically acceptable forms, including isomers such as diastereomers and enantiomers, salts, esters, amides, thioesters, solvates, and polymorphs thereof, as well as racemic mixtures and pure isomers of the compounds described herein.
By "low molecular weight" is meant less than about 500 Daltons.
The term "pharmaceutically acceptable salt" represents those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid. Representative acid addition salts include but are not limited to acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphersulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, isethionate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, mesylate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3- phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include but are not limited to sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
In the generic descriptions of compounds of this invention, the number of atoms of a particular type in a substituent group is generally given as a range, e.g., an alkyl group containing from 1 to 4 carbon atoms or Ci-4 alkyl. Reference to such a range is intended to include specific references to groups having each of the integer number of atoms within the specified range. For example, an alkyl group from 1 to 4 carbon atoms includes each of Ci, C2, C3, and C4. A Ci_i2 heteroalkyl, for example, includes from 1 to 12 carbon atoms in addition to one or more heteroatoms. Other numbers of atoms and other types of atoms may be indicated in a similar manner.
As used herein, the terms "alkyl" and the prefix "alk-" are inclusive of both straight chain and branched chain groups and of cyclic groups, i.e., cycloalkyl. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 6 ring carbon atoms, inclusive. Exemplary cyclic groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups.
By "Ci_4 alkyl" is meant a a branched or unbranched hydrocarbon group having from 1 to 4 carbon atoms. A Ci_4 alkyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. Q_4 alkyls include, without limitation, methyl, ethyl, n- propyl, isopropyl, cyclopropyl, cyclopropylmethyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, and cyclobutyl.
By "C2_4 alkenyl" is meant a branched or unbranched hydrocarbon group containing one or more double bonds and having from 2 to 4 carbon atoms. A C2_4 alkenyl may optionally include monocyclic or polycyclic rings, in which each ring desirably has from three to six members. The C2-A alkenyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. C2_4 alkenyls include, without limitation, vinyl, allyl, 2-cyclopropyl-l-ethenyl, 1-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2 -methyl- 1-propenyl, and 2-methyl-2-propenyl.
By "C2_4 alkynyl" is meant a branched or unbranched hydrocarbon group containing one or more triple bonds and having from 2 to 4 carbon atoms. A C2_4 alkynyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members. The C2_4 alkynyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. C2-4 alkynyls include, without limitation, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, and 3-butynyl.
By "C2_ό heterocyclyl" is meant a stable 5- to 7-membered monocyclic or 7- to 14-membered bicyclic heterocyclic ring which is saturated partially unsaturated or unsaturated (aromatic), and which consists of 2 to 6 carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from N, O, and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxy, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups. The nitrogen and sulfur heteroatoms may optionally be oxidized. The heterocyclic ring may be covalently attached via any heteroatom or carbon atom which results in a stable structure, e.g., an imidazolinyl ring may be linked at either of the ring-carbon atom positions or at the nitrogen atom. A nitrogen atom in the heterocycle may optionally be quaternized. Preferably when the total number of S and O atoms in the heterocycle exceeds 1 , then these heteroatoms are not adjacent to one another. Heterocycles include, without limitation, lH-indazole, 2-pyrrolidonyl, 2H,6H- 1 ,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H- quinolizinyl, 6H-l,2,5-thiadiazinyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, b-carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-l,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, moφholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinylperimidinyl, phenanthridinyl, phenanthrolinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, piperidonyl, 4-piperidonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidiny], pyrrolinyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, carbolinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-l,2,5-thiadiazinyl, 1,2,3- thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1 ,2,3-triazolyl, 1,2,4-triazolyl, 1 ,2,5- triazolyl, 1,3,4-triazolyl, xanthenyl. Preferred 5 to 10 membered heterocycles include, but are not limited to, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, tetrazolyl, benzofuranyl, benzothiofuranyl, indolyl, benzimidazolyl, lH-indazolyl, oxazolidinyl, isoxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, quinolinyl, and isoquinolinyl. Preferred 5 to 6 membered heterocycles include, without limitation, pyridinyl, pyrimidinyl, triazinyl, furanyl, thienyl, thiazolyl, pyrrolyl, piperazinyl, piperidinyl. pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, and tetrazolyl.
By "C6_i 2 aryl" is meant an aromatic group having a ring system comprised of carbon atoms with conjugated π electrons (e.g., phenyl). The aryl group has from 6 to 12 carbon atoms. Aryl groups may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has five or six members. The aryl group may be substituted or unsubstituted. Exemplary substituents include alkyl, hydroxy, alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, fluoroalkyl, carboxyl, hydroxyalkyl, carboxyalkyl, amino, aminoalkyl, monosubstituted amino, disubstituted amino, and quaternary amino groups.
By "C7_i4 alkaryl" is meant an alkyl substituted by an aryl group (e.g., benzyl, phenethyl, or 3,4-dichlorophenethyl) having from 7 to 14 carbon atoms.
By "C3^o alkheterocyclyl" is meant an alkyl substituted heterocyclic group having from 3 to 10 carbon atoms in addition to one or more heteroatoms (e.g., 3-furanylmethyl, 2-furanylmethyl, 3-tetrahydrofuranylmethyl, or 2- tetrahydrofuranylmethyl) .
By "Ci_7 heteroalkyl" is meant a branched or unbranched alkyl, alkenyl, or alkynyl group having from 1 to 7 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P. Heteroalkyls include, without limitation, tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides. A heteroalkyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members. The heteroalkyl group may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halide, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, hydroxyalkyl, carboxyalkyl, and carboxyl groups. Examples of C]_7 heteroalkyls include, without limitation, methoxymethyl and ethoxyethyl.
By "halide" is meant bromine, chlorine, iodine, or fluorine.
By "fluoroalkyl" is meant an alkyl group that is substituted with a fluorine atom. By "perfluoroalkyl" is meant an alkyl group consisting of only carbon and fluorine atoms.
By "carboxyalkyl" is meant a chemical moiety with the formula -(R)-COOH, wherein R is selected from Ci_7 alkyl, C2-7 alkenyl, C2_7 alkynyl, C2-6 heterocyclyl, C6-I2 aryl, C7_i4 alkaryl, C3_i0 alkheterocyclyl, or Ci_7 heteroalkyl.
By "hydroxyalkyl" is meant a chemical moiety with the formula -(R)- OH, wherein R is selected from Ci_7 alkyl, C2_7 alkenyl, C2_7 alkynyl, C2-^ heterocyclyl, C6_i2 aryl, C7_)4 alkaryl, C3_i0 alkheterocyclyl, or Ci_7 heteroalkyl.
By "alkoxy" is meant a chemical substituent of the formula -OR, wherein R is selected from C]_7 alkyl, C2_7 alkenyl, C2_7 alkynyl, C2^ heterocyclyl, C6_i2 aryl, C7_i4 alkaryl, C3_i0 alkheterocyclyl, or Ci_7 heteroalkyl.
By "aryloxy" is meant a chemical substituent of the formula -OR, wherein R is a C6-I2 aryl group.
By "alkylthio" is meant a chemical substituent of the formula -SR, wherein R is selected from C]_7 alkyl, C2_7 alkenyl, C2-7 alkynyl, C2-6 heterocyclyl, C6_)2 aryl, C7_i4 alkaryl, C3_i0 alkheterocyclyl, or Ci_7 heteroalkyl.
By "arylthio" is meant a chemical substituent of the formula -SR, wherein R is a C6_i2 aryl group.
By "quaternary amino" is meant a chemical substituent of the formula -(R)-N(R')(R")(R'")+, wherein R, R', R", and R'" are each independently an alkyl, alkenyl, alkynyl, or aryl group. R may be an alkyl group linking the quaternary amino nitrogen atom, as a substituent, to another moiety. The nitrogen atom, N, is covalently attached to four carbon atoms of alkyl, heteroalkyl, heteroaryl, and/or aryl groups, resulting in a positive charge at the nitrogen atom.
By "charged moiety" is meant a moiety which gains a proton at physiological pH thereby becoming positively charged (e.g., ammonium, guanidinium, or amidinium) or a moiety that includes a net formal positive charge without protonation (e.g., quaternary ammonium). The charged moiety may be either permanently charged or transiently charged.
As used herein, the term "parent" refers to a channel blocking compound which can be modified by quaternization or guanylation of an amine nitrogen atom present in the parent compound. The quaternized and guanylated compounds are derivatives of the parent compound. The guanidyl derivatives described herein are presented in their uncharged base form. These compounds can be administered either as a salt (i.e., an acid addition salt) or in their uncharged base form, which undergoes protonation in situ to form a charged moiety.
Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
Brief Description of the Drawing
Figure 1. Co-application of extracellular QX-314 (5mM) and capsaicin (1 μM) selectively blocks sodium currents in capsaicin-responsive dorsal root ganglion (DRG) sensory neurons, (a) Effect on sodium current (elicited by a step to from -70 to -5 mV) of 10 minutes wash-in of 5 mM QX-314 alone (red trace), 1 μM capsaicin alone (green trace), and co-applied 5mM QX-314 and 1 μM capsaicin (blue trace) in a small (24 μm) capsaicin-sensitive adult cultured DRG neuron. Top panel: Brief application of capsaicin induced a prolonged inward current (holding voltage of -70 mV) in this neuron, (b) Effect on sodium current of the same series of drug applications on a large (52 μm) capsaicin-insensitive neuron, (c) Peak inward current as a function of test pulse recorded in control (black symbols), in the presence of 5mM QX-314 alone (red symbols), 1 μM capsaicin alone (green symbols), and co-applied 5mM QX-314 and 1 μM capsaicin (blue symbols). Symbols show mean ± SEM for experiments on 25 small capsaicin-sensitive neurons. Currents were elicited by 20 ms depolarizing steps from a holding potential of -70 mV to a range of test potentials in 5 mV increments, (d) Time course of the effect of combination of capsaicin and QX-314 on peak sodium current. Bars plot mean ± SEM for peak sodium current normalized relative to that in control (n=25).
Figure 2. Co-application of QX-314 and capsaicin blocks excitability in nociceptive-like DRG neurons, (a) A depolarizing current step (250 pA, 4 ms) applied to a small (23 μm) DRG neuron evoked a nociceptor-like broad action potential with a prominent deflection on the falling phase (arrow). 2 minutes wash-in of QX-314 (5 mM) had no effect (second panel). Capsaicin (1 μM) reduced the action potential amplitude (third panel), probably due to a combination of the modest reduction of sodium current produced by capsaicin as in Figure 1 and inactivation of sodium current secondary to the depolarization produced by capsaicin. Co-applied QX-314 and capsaicin completely abolished action potential generation even with much larger stimulating current injection, (b) Mean ± SEM of action potential amplitudes (n=25 for QX-314, n=l 5 for capsaicin and capsaicin + QX-314).
Figure 3. Intraplantar injection of capsaicin (10 μg/10 μL) together with QX-314 (2%, 10 μL) leads to a prolonged local anesthesia to mechanical (von Frey filaments) and thermal noxious stimuli, (a) Mechanical threshold for paw withdrawal in response to von Frey hairs of increasing strength after interplantar injection of QX -314 alone (2%, 10 μL; green symbols), capsaicin alone (10 μg/10 μL; black symbols), or QX-314 and capsaicin applied together (red symbols). Number of animals that did not respond at all to the highest value (57 g, arrow) is indicated for time points with largest effects. (* = p<0.05, n=6 for each group), (b) Same for thermal (radiant heat) threshold for paw withdrawal. Arrow indicates cutoff, and numbers of animals not responding to strongest stimulus is indicated for time points with largest effects. (* = p<0.05, n=6 for each group). Figure 4. Injection of QX-314 followed by capsaicin adjacent to the sciatic nerve anesthetized the hindlimbs of the animals to noxious mechanical and thermal stimuli without producing any motor deficit, (a) Mechanical threshold for paw withdrawal in response to von Frey filaments of increasing strength after sciatic injection of QX-314 alone (0.2%, 100 μL), capsaicin alone (0.5 μg/μL, 100 μL), or QX-314 injected 10 minutes before capsaicin. Number of animals that did not respond at all to the highest value (57 g, arrow) is indicated for time points with largest effects. (* = p<0.05, ** = p< 0.01, n=6 for each group), (b) Same for thermal (radiant heat) threshold for paw withdrawal, (c). Change in motor function (score: 2 = full paralysis; 1 = partial paralysis; 0 = no impairment) evaluated after sciatic injection of lidocaine (2%; 0.2%), QX-314 (0.2%), capsaicin (5 μg/10 μL) and QX-314 followed by capsaicin injection. Numbers of animals affected by the injections are indicated above each column.
Figure 5. Voltage clamp recordings of sodium channel current in small dorsal root ganglion neurons. The data show that eugenol alone has a modest inhibitory effect on sodium current (10-20% inhibition). Co-application of eugenol and QX-314 produces progressive block that can be complete after 7 minutes. Two examples are depicted, which are representative of 10 experiments with similar results.
Figure 6. Co-application of the TRPA agonist mustard oil (MO) (50 μM) and QX-314 (5 mM). MO alone reduces sodium current by 20-30% and reaches a plateau after approximately 3 minutes. Co-application of MO and QX-314 reduced sodium current dramatically.
Detailed Description of the Invention
Voltage-dependent ion channels in pain-sensing neurons are currently of great interest in developing drugs to treat pain. Blocking voltage-dependent sodium channels in pain-sensing neurons can block pain signals by interrupting initiation and transmission of the action potential, and blocking calcium channels can prevent neurotransmission of the pain signal to the second order neuron in the spinal cord. Heretofore, a limitation in designing small organic molecules that block sodium channels or calcium channels is that they must be active when applied externally to the target cell. The vast majority of such externally-applied molecules are hydrophobic and can pass through membranes. Because of this, they will enter all cells and thus have no selectivity for affecting only pain-sensing neurons. Yet, some blockers are known, such as QX-314, that are only effective when present inside the cell. To date, such blockers have been studied primarily with electrophysiological recording techniques such as whole-cell patch clamp that permit dialysis of the inside of a cell by mechanical rupturing of the membrane. The difficulty of mechanical rupturing without killing the cell, and the difficulty of reversibly applying blockers inside the cell subsequently, has precluded development of high- throughput screening assays for drug molecules that might act from inside cells.
We have discovered a means for delivering inhibitors of voltage-gated ion channels into nociceptive neurons. By providing a way for these inhibitors to enter nociceptive neurons, the invention permits the use — both in screening and in therapy — of entire classes of molecules that are active as drug blockers from the inside of cell but need not be membrane-permeant. Moreover, confining the entry of such blockers to pain-sensing neurons under therapeutic conditions allows for the use of drugs that do not necessarily have intrinsic selectivity for ion channels in pain-sensing neurons compared to other types of cells, but rather gain their selective action on pain-sensing neurons by being allowed to enter pain-sensing neurons in preference to other cells in the nervous and cardiovascular system. Additionally, since TRPVl receptors in particular are often more active in tissue conditions associated with pain (such as inflammation), entry is favored to the particular sensory neurons most associated with tissues that are generating pain. Itch-senstive primary sensory neurons also express TRP channels, particularly TRPVl, and are also be amenable to this approach.
The invention is described in more detail below.
Inhibitors of voltage-gated ion channels
Inhibitors of voltage-gated ion channels that are suitable for use in the methods, compositions, and kits of the invention are desirably positively- charged, hydrophilic compounds. In one embodiment, the compounds are permanently charged (i.e., have a charge that is not transient). In another embodiment, the compounds are transiently charged. Suitable inhibitors of voltage-gated sodium channels include but are not limited to QX-314, N- methyl-procaine (QX-222), N-octyl-guanidine, 9-aminoacridine, and pancuronium. Suitable inhibitors of voltage-gated calcium channels include but are not limited to D-890 (quaternary methoxyverapamil) and CERM 1 1888 (quaternary bepridil).
Additionally, there are many known inhibitors of voltage-gated ion channels that would be of a suitable size to be useful in the methods of the invention (e.g., from about 100 to 4,000 Da, 100 to 3,000 Da, 100 to 2,000 Da, 150 to 1,500 Da, or even 200 to 1,200 Da) and that have amine groups, or can be modified to contain amine groups, that can be readily modified to be charged (e.g., as positively-charged quarternary amines, or as transiently charged guanylated compounds). Such inhibitors include but are not limited to riluzole, mexilitine, phenytoin, carbamazepine, procaine, tocainide, prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, articaine, bupivicaine, mepivicaine, and fluspirilene. Compounds that can be used in the compositions, kits, and methods of the invention include compounds of formulas I-X, below.
Figure imgf000023_0001
In formula I, each of R1 A, R1 B, and R1C is, independently, selected from H, halogen, Ci-4 alkyl, C2_4 alkenyl, C2-4 alkynyl, OR1 H, NRπR1J, NRI KC(O)R1 L, S(O)R1 M, SO2R1V0, SO2NRI PR1Q, SO3R1R, CO2R1 S, C(O)R1T, and C(O)NR1 UR1 V; and each of R1 H, R11, R1J, R, R1 L, R1 M, R1N, R10, R1P, R1Q, R1 R, R1 S, R1T, R! U, and Rlv is, independently, selected from from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl X1 is selected from -CR1WR1X-, -NR!YC(O)-, -OC(O)-, -SC(O)-, -C(O)NR12-, -CO2-, and -OC(S)-; and each of R1W, R1X, R, and R1Z is, independently, selected from H, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; R1D is selected from H, Q-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2_4 heteroalkyl; and each of R1 E, R1 F, and R1 G is, independently, selected from Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; or R1 D and R1G together complete a heterocyclic ring having at least one nitrogen atom. In a preferred embodiment, X1 is -NHC(O)-. Exemplary compounds of formula I include methylated quaternary ammonium derivatives of anesthetic drugs, such as N-methyl lidocaine, N,N-dimethyl prilocaine, N,N,N-trimethyl tocainide, N-methyl etidocaine, N-methyl ropivacaine, N-methyl bupivacaine, N-methyl levobupivacaine, N-methyl mepivacaine. These derivatives can be prepared using methods analogous to those described in Scheme 1. Compounds of formula I include QX-314 (CAS 21306-56-9) and QX-222 (CAS 21236-55-5) (below).
Figure imgf000024_0001
Figure imgf000024_0002
In formula II, each of R2A,R2B, and R2C is, independently, selected from H, halogen, C1-4 alkyl, C2-4 alkenyl, C2^ alkynyL OR21, NR2JR2K, NR2LC(O)R2M, S(O)R2N, SO2R2OR2P, SO2NR2QR2R, SO3R2S, CO2R2T, C(O)R2U, and C(O)NR2VR2W; and each of R2I, R2J, R2K, R2L, R2M, R2N, R20, R2P, R2Q, R2R, R2S, R2T, R2U, R2V, R2W is, independently, selected from H, C,_4 alkyl, C2-4 alkenyl, C2^ alkynyl, and C2^ heteroalkyl; X2 is selected from -CR2XR2Y-, - NR22C(O)-, -OC(O)-, -SC(O)-, -C(O)NR2AA-, -CO2-, and -OC(S)-; and each of R2X, R2Y, R2Z, and R2AA is, independently, selected from H, C^4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl ;R2D is selected from H, C]-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2_4 heteroalkyl; R2E is H or Ci-4 alkyl; and each of R2F, R2G, and R2H is, independently, selected from H, Q-4 alkyl, C2_4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; or R2F and R2G together complete a heterocyclic ring having two nitrogen atoms. Where R2F and R2G form a heterocyclic ring having two nitrogen atoms, the resulting guanidine group is, desirably, selected from
Figure imgf000024_0003
where R2H is H or CH3. Desirably, R2F and R2G combine to form an alkylene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. In a preferred embodiment, X2 is -NHC(O)-. Exemplary compounds of formula II include N-guanidyl derivatives (e.g., -C(NH)NH2 derivatives) of anesthetic drugs, such as desethyl-N-guanidyl lidocaine, N- guanidyl prilocaine, N-guanidyl tocainide, desethyl-N-guanidyl etidocaine, desbutyl-N-guanidyl ropivacaine, desbutyl-N-guanidyl bupivacaine, desbutyl- N-guanidyl levobupivacaine, desmethyl-N-guanidyl mepivacaine. These derivatives can be prepared using methods analogous to those described in Schemes 2-5.
The guanidyl derivatives described herein (e.g., the compounds of formula II) are presented in their uncharged base form. These compounds can be administered either as a salt (i.e., an acid addition salt) or in their uncharged base form, which undergoes protonation in situ to form a charged moiety.
The synthesis of parent drugs of formulas I and II are described in the literature. See, for example, U.S. Patent No. 2,441,498 (synthesis of lidocaine), U.S. Patent No. 3, 160,662 (synthesis of prilocaine), DE Patent No. 2235745 (synthesis of tocainide), DE Patent No. 2162744 (synthesis of etidocaine), PCT Publication No. WO85/00599 (synthesis of ropivacaine), U.S. Patent No. 2,955,111 (synthesis of bupivacaine and levobupivacaine), and U.S. Patent No. 2,799,679 (synthesis of mepivacaine).
Figure imgf000025_0001
(III)
In formula III, n = 0-3 and m = 0-3, with (n+m) = 0-6; each of R3A, R3B, and R3C is, independently, selected from H, halogen, Q_4 alkyl, C2_4 alkenyl, C2_4 alkynyl, C2_4 heteroalkyl, OR3L, NR3MR3N, NR3OC(O)R3P, S(O)R3Q, SO2R3RR3S, SO2NR3TR3U, SO3R3V, CO2R3W, C(O)R3X, and C(O)NR3YR3Z; and each of R3L, R3M, R3N, R30, R3P, R3Q, R3R, R3S, R3T, R3U, R3V, R3W, R3X, R3Y, R3Z is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2_4 alkynyl, and C2_4 heteroalkyl; Y3 is selected from from -CR3AAR3AB-, -NR3ACC(O)-, -
OC(O)-, -SC(O)-, -C(O)NR3AD-, -CO2-, and -OC(S)-; and each of R3AA, R3AB, R3AC, and R3AD is, independently, selected from H, Ci_4 alkyl, C2-A alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; each of R3D, R3E, R3F, and R3G is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C2^ heteroalkyl, C2-^ heterocyclyl, C6_i2 aryl, C7_H alkaryl, and C3_i0 alkheterocyclyl; each of R3H, R3J, and R3K is, independently, selected from Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2_4 heteroalkyl. The quaternary nitrogen in formula TII is identified herein as N' . Exemplary compounds of formula III include methylated quaternary ammonium derivatives of anesthetic drugs, such as N'- methyl procaine, N'-methyl proparacaine, N'-methyl allocain, N'-methyl encainide, N'-methyl procainamide, N'-methyl metoclopramide, N'-methyl stovaine, N'-methyl propoxycaine, N'-methyl chloroprocaine, N',N'-dimethyl flecainide, and N'-methyl tetracaine. These derivatives can be prepared using methods analogous to those described in Scheme 1.
Figure imgf000026_0001
In formula IV, n = 0-3 and m - 0-3, with (n+m) = 0-6; each of R 4A A and R4B is, independently, selected from H, halogen, Ci-4 alkyl, C2-4 alkenyl, C2^ alkynyl, C2^ heteroalkyl, OR4L, NR4MR4N, NR4OC(O)R4P, S(O)R4Q, SO2R4RR4S, SO2NR4TR4U, SO3R4V, CO2R4W, C(O)R4X, and C(O)NR4YR4Z; and each of R4L, R4MR4N, R40, R4P, R4Q, R4R, R4S, R4T, R4U, R4V, R4W, R4X, R4Y, and R4Z is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; Y4 is selected from -CR4AAR4AB-, -NR4ACC(O)-, -OC(O)-, -SC(O)-, -C(O)NR4AD-, -CO2-, and -OC(S)-; and each of R4AA, R4AB, R4AC, and R4AD is, independently, selected from H, C]-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl;each of R4C, R4D, R4E, and R4F is, independently, selected from H, Ci_4 alkyl, C2_4 alkenyl, C2_4 alkynyl, C2-4 heteroalkyl, C2^ heterocyclyl, C6-I2 aryl, C7_i4 alkaryl, and C3_i0 alkheterocyclyl; X4 is selected from H, C\^ alkyl, C2^ alkenyl, C2^, alkynyl, and NR4JR4K; each of R4J and R4K is, independently, selected from H5 C 1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; and each of R4G, R4H, and R41 is, independently, selected from Q-4 alkyl, C2_4 alkenyl, C2-4 alkynyl, and C2_4 heteroalkyl . The quaternary nitrogen in formula IV is identified herein as N". Exemplary compounds of formula III include methylated quaternary ammonium derivatives of anesthetic drugs, such as N",N",N"-trimethyl procaine, N",N",N"-trimethyl proparacaine, N",N",N"- trimethyl procainamide, N",N",N"-trimethyl metoclopramide, N",N",N"- trimethyl propoxycaine, N",N",N"-trimethyl chloroprocaine, N",N"- dimethyl tetracaine, N",N",N"-trimethyl benzocaine, and N",N",N"-trimethyl butamben. These derivatives can be prepared using methods analogous to those described in Scheme 1.
Figure imgf000027_0001
In formula V, n = 0-3 and m - 0-3, with (n+m) = 0-6; each of R5A, R5B, and R5C is, independently, selected from H, halogen, Ci-4 alkyl, C2-4 alkenyl, C2^, alkynyl, C2-4 heteroalkyl, OR5M, NR5NR50, NR5PC(O)R5Q, S(O)R5R, SO2R5SR5T, SO2NR5UR5V, SO3R5W, CO2R5X, C(O)R5Y, and C(O)NR5ZR5AA; and each of R5M, R5N, R50, R5P, R5Q, R5R, R5S, R5T, R5U, R5V, R5W, R5X, R5Y, R5Z, and R5AA is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; Y5 is selected from -CR5ABR5AC-, -NR5ADC(O)-, -OC(O)-, -SC(O)-, -C(O)NR5AE-, -CO2-, and -OC(S)-; and each of R5AB, R5AC, R5ΛD, and R5AE is, independently, selected from H, Ci_4 alkyl, C2_4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; each of R5D, R5E, R5F, and R5G is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C2-4 heteroalkyl, C2_6 heterocyclyl, C6-I 2 aryl, C7_i4 alkaryl, and C3_i0 alkheterocyclyl; R5H is H or C]_ 4 alkyl; and each of R5J, R5K, and R5L is, independently, selected from H, Q-4 alkyl, C2-4 alkenyl, C2_4 alkynyl, and C2^ heteroalkyl; or R5J and R5K together complete a heterocyclic ring having two nitrogen atoms. Where R5J and R5K form a heterocyclic ring having two nitrogen atoms, the resulting guanidine group is, desirably, selected from
Figure imgf000028_0001
where R5L is H or CH3. Desirably, R5J and R5K combine to form an alkylene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. The guanylated nitrogen in formula V is identified herein as N'. Exemplary compounds of formula V include N-guanidyl derivatives (e.g., -C(NH)NH2 derivatives) of anesthetic drugs, such as such as desethyl-N'- guanidyl procaine, desethyl-N'-guanidyl proparacaine, desethyl-N'-guanidyl allocain, desmethyl-N'-guanidyl encainide, desethyl-N'-guanidyl procainamide, desethyl-N'-guanidyl metoclopramide, desmethyl-N'-guanidyl stovaine, desethyl-N'-guanidyl propoxycaine, desethyl-N'-guanidyl chloroprocaine, N'- guanidyl flecainide, and desethyl-N'-guanidyl tetracaine. These derivatives can be prepared using methods analogous to those described in Schemes 2-5.
In formula VI, n = 0-3 and m = 0-3, with (n+m) = 0-6; each of R6A and R6B is, independently, selected from H, halogen, Ci_4 alkyl, C2_4 alkenyl, C2-4 alkynyl, C2-4 heteroalkyl, OR6K, NR6LR6M, NR6NC(O)R60, S(O)R6P, SO2R6QR6R, SO2NR6SR6T, SO3R6U, CO2R6V, C(O)R6W, and C(O)NR6XR6Y; and each of R6K,R6L, R6M, R6N, R60, R6P, R6Q, R6R, R6S, R, R6U, R6V, R6W, R6X, and R6Y is, independently, selected from H, Q_4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; Y6 is selected from -CR6ZR6AA-, -NR6ABC(O)-, -OC(O)-, -SC(O)-, -C(O)NR6AC-, -CO2-, and -OC(S)-; and each of R6Z, R6AA, R6AB, and R6AC is, independently, selected from H, C]-4 alkyl, C2-4 alkenyl, C2_4 alkynyl, and C2-4 heteroalkyl; each of R6C, R6D, R6E, and R6F is, independently, selected from H, Ci_4 alkyl, C2-4 alkenyl, C2_4 alkynyl, C2-4 heteroalkyl, C2^ heterocyclyl, C6-^ aryl, C7_i4 alkaryl, and C3-Io alkheterocyclyl; X6 is selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and NR6ADR6AE; each of R6AD and R6AE is, independently, selected from H, Ci_4 alkyl, C2^ alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; R6G is H or C,_4 alkyl; and each of R6H, R61, and R6J is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-A heteroalkyl; or R6H and R61 together complete a heterocyclic ring having two nitrogen atoms. Where R6H and R61 form a heterocyclic ring having two nitrogen atoms, the resulting guanidine group is, desirably, selected from
Figure imgf000029_0001
where R6J is H or CH3. Desirably, R6H and R61 combine to form an alkylene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. The guanylated nitrogen in formula V is identified herein as N". Exemplary compounds of formula VI include N-guanidyl derivatives (e.g., -C(NH)NH2 derivatives) of anesthetic drugs, such as such as N"-guanidyl procaine, N"-guanidyl proparacaine, N"-guanidyl procainamide, N"-guanidyl metoclopramide, N"-guanidyl propoxycaine, N"-guanidyl chloroprocaine, N"- guanidyl tetracaine, N"-guanidyl benzocaine, and N"-guanidyl butamben. These derivatives can be prepared using methods analogous to those described in Schemes 2-5.
The synthesis of parent drugs of formulas III- VI are described in the literature. See, for example, U.S. Patent No. 812,554 (synthesis of procaine), Clinton et al., J. Am. Chem. Soc. 74:592 (1952) (synthesis of proparacaine), U.S. Patent No. 2,689,248 (synthesis of propoxycaine), Hadicke et al, Pharm. Zentralh. 94:384 (1955) (synthesis of chloroprocaine), U.S. Patent No. 1,889,645 (synthesis of tetracaine), Salkowski et al., Ber. 28: 1921 (1895) (synthesis of benzocaine), Brill et al., J. Am. Chem. Soc. 43: 1322 (1921) (synthesis of butamben), U.S. Patent No. 3,931 ,195 (synthesis of encainide), Yamazaki et al., J. Pharm. Soc. Japan 73:294 (1953) (synthesis of procainamide), U.S. Patent No. 3,177,252 (synthesis of metoclopramide), U.S. Patent No. 3,900,481 (synthesis of flecainide), and Fourneau et al., Bull. Sci. Pharmacol. 35:273 (1928) (synthesis of stovaine).
Figure imgf000030_0001
In formula VII, n - 0-3 and m = 0-3, with (n+m) = 0-6; each of R7A, R7B, and R7C is, independently, selected from H, halogen, Cj-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C2-4 heteroalkyl, OR7L, NR7MR7N, NR7OC(O)R7p, S(O)R7Q, SO2R7RR7S, SO2NR7TR7U, SO3R7V, CO2R7W, C(O)R7X, and C(O)NR7YR7Z; and each of R7L,R7M, R7N, R70, R7P, R7Q, R7R, R7S, R7T, R7U, R7V, R7W, R7X, R7Y, and R7Z is, independently, selected from H, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2_4 heteroalkyl; X7 is selected from -CR7AAR7AB-, NR7ACC(O)-, -OC(O)-, -SC(O)-, -C(O)NR7AD-, -CO2-, and -OC(S)-; and each of R7AA, R7AB, R7AC, and R7AD is, independently, selected from H, C1^ alkyl, C2_4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; each of R7D, R7E, R7F, and R7G is, independently, selected from H, Ci-4 alkyl, C2_4 alkenyl, C2-4 alkynyl, C2_4 heteroalkyl, C2_6 heterocyclyl, C6-I2 aryl, C7_)4 alkaryl, and C3_i0 alkheterocyclyl; and each of R7H, R7J, and R7K is, independently, selected from
Figure imgf000031_0001
alkynyl, and C2-4 heteroalkyl. In a preferred embodiment, X7 is -C(O)NH-. Exemplary compounds of formula VII include methylated quaternary ammonium derivatives of anesthetic drugs, such as N'- methyl dibucaine. These derivatives can be prepared using methods analogous to those described in Scheme 1.
Figure imgf000031_0002
In formula VIII, n = 0-3 and m = 0-3, with (n+m) - 0-6; each of R8A, R8B, and R8C is, independently, selected from H, halogen, Ci-4 alkyl, C2-4 alkenyl, C2^ alkynyl, C2_4 heteroalkyl, OR8L, NR8MR8N, NR8OC(O)R8P, S(O)R8Q, SO2R8RR8S, SO2NR8TR8U, SO3R8V, CO2R8W, C(O)R8X, and C(O)NR8YR8Z; and each of R8L, R8M, R8N, R80, R8P, R8Q, R8R, R8S, R8T, R8U, R8V, R8W, R8X, R8Y, and R8Z is, independently, selected from H, C-4 alkyl, C2-4 alkenyl, C2^ alkynyl, and C2-4 heteroalkyl; X8 is selected from -CR8AAR8AB-, -NR8ACC(O)-, -OC(O)-, -SC(O)-, -C(O)NR8AD-, -CO2-, and -OC(S)-; and each of R8AA, R8AB, R8AC, and R8AD is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2^ alkynyl, and C2_4 heteroalkyl; each of R8D, R8E, R8F, and R8G is, independently, selected from H, C^ alkyl, C2-4 alkenyl, C2-4 alkynyl, C2_4 heteroalkyl, C2_^ heterocyclyl, C6_i2 aryl, C7_i4 alkaryl, and C3_io alkheterocyclyl; R8H is H or C,^ alkyl; and each of R81, R8J, and R8K is, independently, selected from H, C)-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; or R81 and R8J together complete a heterocyclic ring having two nitrogen atoms. Where R81 and R8J form a heterocyclic ring having two nitrogen atoms, the resulting guanidine group is, desirably, selected from
Figure imgf000032_0001
where R8K is H or CH3. Desirably, R81 and R8J combine to form an alkylene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. The guanylated nitrogen in formula V is identified herein as N'. In a preferred embodiment, X8 is -C(O)NH-. Exemplary compounds of formula VIII include N-guanidyl derivatives (e.g., -C(NH)NH2 derivatives) of anesthetic drugs, such as such as desethyl-N-guanidyl dibucaine. These derivatives can be prepared using methods analogous to those described in Schemes 2-5.
In formula IX, n =
Figure imgf000032_0002
and Ryt is, independently, selected from H, halogen, Q-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, OR91, NR9JR9K, NR9LC(O)R9M, S(O)R9N, SO2R9OR9P, SO2NR9QR9R, SO3R95, CO2R9T, C(O)R9U, and C(O)NR9VR9W; and each of R91, R9J, R9K, R9L, R9M, R9N, R9°, R9P, R9Q, R9R, R9S, R9T, R9U, R9V, and R9W is, independently, selected from H, Ci_4 alkyl, C2_4 alkenyl, C2-4 alkynyl, and C2-4 heteroalkyl; X9 is selected from -CR9XR9Y-, -O-, -S-, and -NR9Z-; and each of R9X, R9Y, and R9Z is, independently, selected from H, Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, and C2^ heteroalkyl; Y9 is NR9AANR9ABNR9AC or NR9ADZ9; each of R9AA, R9AB, and R9AC is, independently, selected from H, C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl; R9AD is H or C1-4 alkyl; Z9 is
Figure imgf000033_0001
each of R9F, R9G, and R9H is, independently, selected from H, C1-4 alkyl, C2_4 alkenyl. and C2-4 alkynyl, or R9F and R9G together complete a heterocyclic ring having two nitrogen atoms. Where R9F and R9G form a heterocyclic ring having two nitrogen atoms, the resulting guanidine group is, desirably, selected from
Figure imgf000033_0002
where R9H is H or CH3. Desirably, R9F and R9G combine to form an alkyl ene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. In a preferred embodiment, X9 = -O-. Exemplary compounds of formula IX include N-guanidyl derivatives (e.g., -C(NH)NH2 derivatives), such as N-guanidyl fluoxetine, and methylated quaternary ammonium derivatives, such as N,N-dimethyl fluoxetine. These derivatives can be prepared using methods analogous to those described in Schemes 1-5.
Figure imgf000033_0003
In formula X, W3 is O, NH, NCH2R10J, NC(O)CH2R10J, CHCH2R10J, C=CHR10J, or C=CHR10IC; W1-W2 is S, O, OCHR10K, SCHR10K, N=CR10K, CHR10L-CHR10K, or CR10L=CR10K; each of R10A, R10B, R10C, R10D, R10E, R10F, R10G, and R10H is, independently, selected from H, OH, halide, Ci-4 alkyl, and C2-4 heteroalkyl; R10J is CH2CH2X10A or CH(CH3)CH2X10A; R10L is H or OH; R10K is H, OH, or the group: χ.0A ^ NRIOMR1ON11IOP5 or NR.0Qχ.0C. χ.0B ^ NRIORRI∞ or Nχ.0C. ^ Qf
Ri
Figure imgf000034_0001
^ independently; selected from C1-4 alkyl, C2-4 alkenyl, C2_4 alkynyl, and C2-4 heteroalkyl, or R10R, and R10S together complete a heterocyclic ring having at least one nitrogen atom; R10Q is H or Ci-4 alkyl;
X10C is
Figure imgf000034_0002
each of RIOT, RIOU, and R1OV is, independently, selected from H, CM alkyl, C2-4 alkenyl, and C2_4 alkynyl, or R10T and R1OV together complete a heterocyclic ring having two nitrogen atoms. Where RI0T and R10V form a heterocyclic ring having two nitrogen atoms, the resulting guanidine group is, desirably, selected from
Figure imgf000034_0003
where RIOU is H or CH3. Desirably, R10T and R10V combine to form an alkylene or alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. Exemplary compounds of formula X include N-guanidyl derivatives (e.g., -C(NH)NH2 derivatives) and methylated quaternary ammonium derivatives. N-guanidyl derivatives of formula X include, without limitation, N-guanidyl amoxapine, desmethyl-N-guanidyl trimipramine, desmethyl-N-guanidyl dothiepin, desmethyl-N-guanidyl doxepin, desmethyl- N-guanidyl amitriptyline, N-guanidyl protriptyline, N-guanidyl desipramine, desmethyl-N-guanidyl clomipramine, desmethyl-N-guanidyl clozapine, desmethyl-N-guanidyl loxapine, N-guanidyl nortriptyline, desmethyl-N- guanidyl cyclobenzaprine, desmethyl-N-guanidyl cyproheptadine, desmethyl- N-guanidyl olopatadine, desmethyl-N-guanidyl promethazine, desmethyl-N- guanidyl trimeprazine, desmethyl-N-guanidyl chlorprothixene, desmethyl-N- guanidyl chloφromazine, desmethyl-N-guanidyl propiomazine, desmethyl-N- guanidyl prochloφerazine, desmethyl-N-guanidyl thiethylperazine, desmethyl- N-guanidyl trifluoperazine, desethyl-N-guanidyl ethacizine, and desmethyl-N- guanidyl imipramine. Methylated quaternary ammonium derivatives of formula X include, without limitation, N,N-dimethyl amoxapine, N-methyl trimipramine, N-methyl dothiepin, N-methyl doxepin, N-methyl amitriptyline, N,N-dimethyl protriptyline, N,N-dimethyl desipramine, N-methyl clomipramine, N-methyl clozapine, N-methyl loxapine, N,N-dimethyl nortriptyline, N-methyl cyclobenzaprine, N-methyl cyproheptadine, N-methyl olopatadine, N-methyl promethazine, N-methyl trimeprazine, N-methyl chlorprothixene, N-methyl chloφromazine, N-methyl propiomazine, N-methyl moricizine, N-methyl prochloφerazine, N-methyl thiethylperazine, N-methyl fluphenazine, N-methyl peφhenazine, N-methyl flupenthixol, N-methyl acetophenazine, N-methyl trifluoperazine, N-methyl ethacizine, and N-methyl imipramine. These derivatives can be prepared using methods analogous to those described in Schemes 1-5.
Other ion channel blockers that can contain an amine nitrogen which can be guanylated or quaternized as described herein include, without limitation, oφhenadrine, phenbenzamine, bepridil, pimozide, penfluridol, flunarizine, fluspirilene, propiverine, disopyramide, methadone, tolterodine, tridihexethyl salts, tripelennamine, mepyramine, brompheniramine, chloφheniramine, dexchloφheniramine, carbinoxamine, levomethadyl acetate, gallopamil, verapamil, devapamil, tiapamil, emopamil, dyclonine, pramoxine, lamotrigine, mibefradil, gabapentin, amiloride, diltiazem, nifedipine, nimodipine, nitrendipine, cocaine, mexiletine, propafenone, quinidine, oxethazaine, articaine, riluzole, bencyclane, lifarizine, and strychnine. Still other ion channel blockers can be modified to incoφorate a nitrogen atom suitable for quaternization or guanylation. These ion channel blockers include, without limitation, fosphenytoin, ethotoin, phenytoin, carbamazepine, oxcarbazepine, topiramate, zonisamide, and salts of valproic acid.
Synthesis
The synthesis of charge-modified ion channel blockers may involve the selective protection and deprotection of alcohols, amines, ketones, sulfhydryls or carboxyl functional groups of the parent ion channel blocker, the linker, the bulky group,and/or the charged group. For example, commonly used protecting groups for amines include carbamates, such as tert-butyl, benzyl, 2,2,2-trichloroethyl, 2-trimethylsilylethyl, 9-fluorenylmethyl, allyl, and m- nitrophenyl. Other commonly used protecting groups for amines include amides, such as formamides, acetamides, trifluoroacetamides, sulfonamides, trifluoromethanesulfonyl amides, trimethylsilylethanesulfonamides, and tert- butylsulfonyl amides. Examples of commonly used protecting groups for carboxyls include esters, such as methyl, ethyl, tert-butyl, 9-fluorenylmethyl, 2- (trimethylsilyl)ethoxy methyl, benzyl, diphenylmethyl, O-nitrobenzyl, ortho- esters, and halo-esters. Examples of commonly used protecting groups for alcohols include ethers, such as methyl, methoxymethyl, methoxyethoxymethyl, methylthiomethyl, benzyloxymethyl, tetrahydropyranyl, ethoxyethyl, benzyl, 2- napthylmethyl, O-nitrobenzyl, P-nitrobenzyl, P-methoxybenzyl, 9- phenylxanthyl, trityl (including methoxy-trityls), and silyl ethers. Examples of commonly used protecting groups for sulfhydryls include many of the same protecting groups used for hydroxyls. In addition, sulfhydryls can be protected in a reduced form (e.g., as disulfides) or an oxidized form (e.g., as sulfonic acids, sulfonic esters, or sulfonic amides). Protecting groups can be chosen such that selective conditions (e,g., acidic conditions, basic conditions, catalysis by a nucleophile, catalysis by a lewis acid, or hydrogenation) are required to remove each, exclusive of other protecting groups in a molecule. The conditions required for the addition of protecting groups to amine, alcohol, sulfhydryl, and carboxyl functionalities and the conditions required for their removal are provided in detail in T.W. Green and P.G.M. Wuts, Protective Groups in Organic Synthesis (2nd Ed.), John Wiley & Sons, 1991 and P.J. Kocienski, Protecting Groups, Georg Thieme Verlag, 1994.
Charge-modified ion channel blockers can be prepared using techniques familiar to those skilled in the art. The modifications can be made, for example, by alkylation of the parent ion channel blocker using the techniques described by J. March, Advanced Organic Chemistry: Reactions, Mechanisms and Structure, John Wiley & Sons, Inc., 1992, page 617. The conversion of amino groups to guanidine groups can be accomplished using standard synthetic protocols. For example, Mosher has described a general method for preparing mono-substituted guanidines by reaction of aminoiminomethanesulfonic acid with amines (Kim et al., Tetrahedron Lett. 29:3183 (1988)). A more convenient method for guanylation of primary and secondary amines was developed by Bernatowicz employing /H-pyrazole-1-carboxamidine hydrochloride; 1 -H-pyrazole- 1 -(N5N' -b/s(tert-butoxycarbonyl)carboxami dine; or 1 -H-pyrazole- 1 -(N,N'-b/s(benzyloxycarbonyl)carboxamidine. These reagents react with amines to give mono-substituted guanidines (see Bernatowicz et al., J. Org. Chem. 57:2497 (1992); and Bernatowicz et al., Tetrahedron Lett. 34:3389 (1993)). In addition, Thioureas and S-alkyl- isothioureas have been shown to be useful intermediates in the syntheses of substituted guanidines (Poss et al., Tetrahedron Lett. 33:5933 (1992)). In certain embodiments, the guanidine is part of a heterocyclic ring having two nitrogerf atoms (see, for example, the structures below). The ring system can include an alkylene or
R I and , R / alkenylene of from 2 to 4 carbon atoms, e.g., ring systems of 5, 6, and 7- membered rings. Such ring systems can be prepared, for example, using the methods disclosed by Schlama et al., J. Org. Chem., 62:4200 (1997).
Charge-modified ion channel blockers can be prepared by alkylation of an amine nitrogen in the parent compound as shown in Scheme 1.
Scheme 1
Figure imgf000038_0001
Alternatively, charge-modified ion channel blockers can be prepared by introduction of a guanidine group. The parent compound can be reacted with a cynamide, e.g., methylcyanamide, as shown in Scheme 2 or pyrazole-1- carboxamidine derivatives as shown in Scheme 3 where Z is H or a suitable protecting group. Alternatively, the parent compound can be reacted with cyanogens bromide followed by reaction with methylchloroaluminum amide as shown in Scheme 4. Reagents such as 2-(methylthio)-2-imidazoline can also be used to prepare suitably functionalized derivatives (Scheme 5).
Scheme 2
Figure imgf000038_0002
Scheme 3
Figure imgf000039_0001
Z = protecting group amide
Scheme 4
Figure imgf000039_0002
Scheme 5
Figure imgf000039_0003
Any ion channel blocker containing an amine nitrogen atom can be modified as shown in Schemes 1-5. TRPVl agonists
TRPVl agonists that can be employed in the methods, compositions, and kits of the invention include but are not limited to any that activates TRPVl receptors on nociceptors and allows for entry of at least one inhibitor of voltage-gated ion channels. Suitable TRPVl agonists include but are not limited to capsaicin, eugenol, arvanil (N-arachidonoylvanillamine), anandamide, 2-aminoethoxydiphenyl borate (2 APB), AM404, resiniferatoxin, phorbol 12-phenylacetate 13-acetate 20-homovanillate (PPAHV), olvanil (NE 19550), OLDA (N-oleoyldopamine), N-arachidonyldopamine (NAD A), 6'- iodoresiniferatoxin (6'-IRTX), Cl 8 N-acylethanolamines, lipoxygenase derivatives such as 12-hydroperoxyeicosatetraenoic acid, inhibitor cysteine knot (ICK) peptides (vanillotoxins), piperine, MSK195 (N-[2-(3,4- dimethylbenzyl)-3-(pivaloyloxy)propyl]-2-[4-(2-aminoethoxy)-3- methoxyphenyl]acetamide), JYL79 (N-[2-(3,4-dimethylbenzyl)-3- (pivaloyloxy)propyl]-N'-(4-hydroxy-3-methoxybenzyl)thiourea), hydroxy- alpha-sanshool, 2-aminoethoxydiphenyl borate, 10-shogaol, oleylgingerol, oleylshogaol, and SU200 (N-(4-tert-butylbenzyl)-N'-(4-hydroxy-3- methoxybenzyl)thiourea) .
TRPlA agonists
TRPlA agonists that can be employed in the methods, compositions, and kits of the invention include any that activates TRPlA receptors on nociceptors or pruriceptors and allows for entry of at least one inhibitor of voltage-gated ion channels. Suitable TRPlA agonists include but are not limited to cinnamaldehyde, allyl-isothiocynanate, diallyl disulfide, icilin, cinnamon oil, wintergreen oil, clove oil, acrolein, hydroxy-alpha-sanshool, 2- aminoethoxydiphenyl borate, 4-hydroxynonenal, methyl p-hydroxybenzoate, mustard oil, and 3'-carbamoylbiphenyl-3-yl cyclohexylcarbamate (URB597). P2X agonists
P2X agonists that can be employed in the methods, compositions, and kits of the invention include any that activates P2X receptors on nociceptors or pruriceptors and allows for entry of at least one inhibitor of voltage-gated ion channels. Suitable P2X agonists include but are not limited to 2-methylthio- ATP, T and 3'-O-(4-benzoylbenzoyl)-ATP, and ATP5'-C>-(3-thiotriphosphate).
TRPM8 agonists
TRPM8 agonists that can be employed in the methods, compositions, and kits of the invention include any that activates TRPM8 receptors on nociceptors or pruriceptors and allows for entry of at least one inhibitor of voltage-gated ion channels. Suitable TRPM8 agonists include but are not limited to menthol, iciclin, eucalyptol, linalool, geraniol, and hydroxycitronellal.
Additional agents
The methods, compositions, and kits of the invention may be used for the treatment of pain (e.g., neuropathic pain, nociceptive pain, idiopathic pain, inflammatory pain, dysfunctional pain, migraine, or procedural pain) and itch (e.g. dermatological conditions like atopic eczema or psoriasis, pruritis in parasitic and fungal infections, drug-induced, allergic, metabolic, in cancer or liver and kidney failure). If desired, one or more additional agents typically used to treat pain may be used in conjunction with a combination of the invention in the methods, compositions, and kits described. herein. Such agents include but are not limited to NSAIDs, opioids, tricyclic antidepressants, amine transporter inhibitors, anticonvulsants. If desired, one or more additional agents typically used to treat itch may be used in conjunction with a combination of the invention in the methods, compositions, and kits described herein. Such agents include topical or oral steroids and antihistamines. Formulation of compositions
The administration of a combination of the invention may be by any suitable means that results in the reduction of pain sensation at the target region. The inhibitor(s) of voltage-gated ion channels and the TRPVl /TRPA 1/P2X/TRPM8 receptor agonist(s) may be contained in any appropriate amount in any suitable carrier substance, and are generally present in amounts totaling 1-95% by weight of the total weight of the composition. The composition may be provided in a dosage form that is suitable for oral, parenteral (e.g., intravenous, intramuscular), rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intrathecal, epidural, or ocular administration, or by injection, inhalation, or direct contact with the nasal or oral mucosa.
Thus, the composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, or aerosols. The compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy, 20th edition, 2000, ed. A.R. Gennaro, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
Each compound of the combination may be formulated in a variety of ways that are known in the art. For example, the first and second agents may be formulated together or separately. Desirably, the first and second agents are formulated together for the simultaneous or near simultaneous administration of the agents.
The individually or separately formulated agents can be packaged together as a kit. Non-limiting examples include but are not limited to kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc. The kit can include optional components that aid in the administration of the unit dose to patients, such as vials for reconstituting powder forms, syringes for injection, customized IV delivery systems, inhalers, etc. Additionally, the unit dose kit can contain instructions for preparation and administration of the compositions.
The kit may be manufactured as a single use unit dose for one patient, multiple uses for a particular patient (at a constant dose or in which the individual compounds may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple patients ("bulk packaging"). The kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.
Solid dosage forms for oral use
Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose and sorbitol), lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc).
Two or more compounds may be mixed together in a tablet, capsule, or other vehicle, or may be partitioned. In one example, the first compound is contained on the inside of the tablet, and the second compound is on the outside; such that a substantial portion of the second compound is released prior to the release of the first compound.
Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium. Generally, when administered to a humankind oral dosage of any of the compounds of the combination of the invention will depend on the nature of the compound, and can readily be determined by one skilled in the art. Typically, such dosage is normally about 0.001 mg to 2000 mg per day, desirably about 1 mg to 1000 mg per day, and more desirably about 5 mg to 500 mg per day. Dosages up to 200 mg per day may be necessary. It may be useful to administer the minimum therapeutic dose required to activate the TRPVl /TRPA 1/P2X/TRPM8 receptor, which can be determined using standard techniques.
Administration of each drug in the combination can, independently, be one to four times daily for one day to one year, and may even be for the life of the patient. Chronic, long-term administration will be indicated in many cases.
Topical formulations
Compositions can also be adapted for topical use with a topical vehicle containing from between 0.0001% and 25% (w/w) or more of active ingredient(s).
In a preferred combination, the active ingredients are preferably each from between 0.0001% to 10% (w/w), more preferably from between 0.0005% to 4% (w/w) active agent. The cream can be applied one to four times daily, or as needed. For example, for prednisolone adapted for topical administration, a topical vehicle will contain from between 0.01% to 5% (w/w), preferably from between 0.01% to 2% (w/w), more preferably from between 0.01% to 1% (w/w).
Performing the methods described herein, the topical vehicle containing the combination of the invention is preferably applied to the site of discomfort on the subject. For example, a cream may be applied to the hands of a subject suffering from arthritic fingers. Conjugates
If desired, the drugs used in any of the combinations described herein may be covalently attached to one another to form a conjugate of formula (XI).
(A)-(L)-(B) (XI)
In formula (XI), (A) is a compound that activates a channel-forming receptor that is present on nociceptors and/or pruriceptors; (L) is a linker; and (B) is a compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the channels but does not substantially inhibit the channels when applied to the external face of the channels, and is capable of entering nociceptors or pruriceptors through the channel- forming receptor when the receptor is activated.
The conjugates of the invention can be prodrugs, releasing drug (A) and drug (B) upon, for example, cleavage of the conjugate by intracellular and extracellular enzymes (e.g., amidases, esterases, and phosphatases). The conjugates of the invention can also be designed to largely remain intact in vivo, resisting cleavage by intracellular and extracellular enzymes, so long as the conjugate and is capable of entering nociceptors or pruriceptors through the channel-forming receptor when the receptor is activated. The degradation of the conjugate in vivo can be controlled by the design of linker (L) and the covalent bonds formed with compound (A) and compound (B) during the synthesis of the conjugate.
Conjugates can be prepared using techniques familiar to those skilled in the art. For example, the conjugates can be prepared using the methods disclosed in G. Hermanson, Bioconjugate Techniques, Academic Press, Inc., 1996. The synthesis of conjugates may involve the selective protection and deprotection of alcohols, amines, ketones, sulfhydryls or carboxyl functional groups of drug (A), the linker, and/or drug (B). For example, commonly used protecting groups for amines include carbamates, such as tert-butγl, benzyl, 2,2,2-trichloroethyl, 2-trimethylsilylethyl, 9-fluorenylmethyl, allyl, and m- nitrophenyl. Other commonly used protecting groups for amines include amides, such as formamides, acetamides, trifluoroacetamides, sulfonamides, trifluoromethanesulfonyl amides, trimethylsilylethanesulfonamides, and tert- butylsulfonyl amides. Examples of commonly used protecting groups for carboxyls include esters, such as methyl, ethyl, tert-butyl, 9-fluorenylmethyl, 2- (trimethylsilyl)ethoxy methyl, benzyl, diphenylmethyl, O-nitrobenzyl, ortho- esters, and halo-esters. Examples of commonly used protecting groups for alcohols include ethers, such as methyl, methoxymethyl, methoxyethoxymethyl, methylthiomethyl, benzyloxymethyl, tetrahydropyranyl, ethoxyethyl, benzyl, 2- napthylmethyl, O-nitrobenzyl, P-nitrobenzyl, P-methoxybenzyl, 9- phenylxanthyl, trityl (including methoxy-trityls), and silyl ethers. Examples of commonly used protecting groups for sulfhydryls include many of the same protecting groups used for hydroxyls. In addition, sulfhydryls can be protected in a reduced form (e.g., as disulfides) or an oxidized form (e.g., as sulfonic acids, sulfonic esters, or sulfonic amides). Protecting groups can be chosen such that selective conditions (e.g., acidic conditions, basic conditions, catalysis by a nucleophile, catalysis by a lewis acid, or hydrogenation) are required to remove each, exclusive of other protecting groups in a molecule. The conditions required for the addition of protecting groups to amine, alcohol, sulfhydryl, and carboxyl functionalities and the conditions required for their removal are provided in detail in T.W. Green and P.G.M. Wuts, Protective Groups in Organic Synthesis (2nd Ed.), John Wiley & Sons, 1991 and PJ. Kocienski, Protecting Groups, Georg Thieme Verlag, 1994. Additional synthetic details are provided below.
Linkers
The linker component of the invention is, at its simplest, a bond between compound (A) and compound (B), but typically provides a linear, cyclic, or branched molecular skeleton having pendant groups covalently linking compound (A) to compound (B). Thus, linking of compound (A) to compound (B) is achieved by covalent means, involving bond formation with one or more functional groups located on compound (A) and compound (B). Examples of chemically reactive functional groups which may be employed for this purpose include, without limitation, amino, hydroxyl, sulfhydryl, carboxyl, carbonyl, carbohydrate groups, vicinal diols, thioethers, 2-aminoalcohols, 2-aminothiols, guanidinyl, imidazolyl, and phenolic groups.
The covalent linking of compound (A) and compound (B) may be effected using a linker which contains reactive moieties capable of reaction with such functional groups present in compound (A) and compound (B). For example, an amine group of compound (A) may react with a carboxyl group of the linker, or an activated derivative thereof, resulting in the formation of an amide linking the two.
Examples of moieties capable of reaction with sulfhydryl groups include α-haloacetyl compounds of the type XCH2CO- (where X=Br, Cl or I), which show particular reactivity for sulfhydryl groups, but which can also be used to modify imidazolyl, thioether, phenol, and amino groups as described by Gurd, Methods Enzymol. 11 :532 (1967). N-Maleimide derivatives are also considered selective towards sulfhydryl groups, but may additionally be useful in coupling to amino groups under certain conditions. Reagents such as 2- iminothiolane (Traut et al., Biochemistry 12:3266 (1973)), which introduce a thiol group through conversion of an amino group, may be considered as sulfhydryl reagents if linking occurs through the formation of disulphide bridges.
Examples of reactive moieties capable of reaction with amino groups include, for example, alkylating and acylating agents. Representative alkylating agents include: (i) α-haloacetyl compounds, which show specificity towards amino groups in the absence of reactive thiol groups and are of the type XCH2CO- (where X=Cl, Br or I), for example, as described by Wong Biochemistry 24:5337 (1979);
(ii) N-maleimide derivatives, which may react with amino groups either through a Michael type reaction or through acylation by addition to the ring carbonyl group, for example, as described by Smyth et al., J. Am. Chem. Soc. 82:4600 (1960) and Biochem. J. 91 :589 (1964);
(iii) aryl halides such as reactive nitrohaloaromatic compounds;
(iv) alkyl halides, as described, for example, by McKenzie et al., J. Protein Chem. 7:581 (1988);
(v) aldehydes and ketones capable of Schiff s base formation with amino groups, the adducts formed usually being stabilized through reduction to give a stable amine;
(vi) epoxide derivatives such as epichlorohydrin and bisoxiranes, which may react with amino, sulfhydryl, or phenolic hydroxyl groups;
(vii) chlorine-containing derivatives of s-triazines, which are very reactive towards nucleophiles such as amino, sufhydryl, and hydroxyl groups;
(viii) aziridines based on s-triazine compounds detailed above, e.g., as described by Ross, J. Adv. Cancer Res. 2: 1 (1954), which react with nucleophiles such as amino groups by ring opening;
(ix) squaric acid diethyl esters as described by Tietze. Chem. Ber. 124: 1215 (1991); and
(x) α-haloalkyl ethers, which are more reactive alkylating agents than normal alkyl halides because of the activation caused by the ether oxygen atom, as described by Benneche et al., Eur. J. Med. Chem. 28:463 (1993).
Representative amino-reactive acylating agents include:
(i) isocyanates and isothiocyanates, particularly aromatic derivatives, which form stable urea and thiourea derivatives respectively; (ii) sulfonyl chlorides, which have been described by Herzig et al., Biopolymers 2:349 (1964);
(iii) acid halides;
(iv) active esters such as nitrophenylesters or N-hydroxysuccinimidyl esters;
(v) acid anhydrides such as mixed, symmetrical, or N- carboxyanhydrides ;
(vi) other useful reagents for amide bond formation, for example, as described by M, Bodansky, Principles of Peptide Synthesis, Springer- Verlag, 1984;
(vii) acylazides, e.g. wherein the azide group is generated from a preformed hydrazide derivative using sodium nitrite, as described by Wetz et ύ., Anal. Biochem. 58:347 (1974); and
(viii) imidoesters, which form stable amidines on reaction with amino groups, for example, as described by Hunter and Ludwig, J. Am. Chem. Soc. 84:3491 (1962).
Aldehydes and ketones may be reacted with amines to form Schiff s bases, which may advantageously be stabilized through reductive amination. Alkoxylamino moieties readily react with ketones and aldehydes to produce stable alkoxamines, for example, as described by Webb et al., in Bioconjugate Chem. 1 :96 (1990).
Examples of reactive moieties capable of reaction with carboxyl groups include diazo compounds such as diazoacetate esters and diazoacetamides, which react with high specificity to generate ester groups, for example, as described by Herriot, Adv. Protein Chem. 3: 169 (1947). Carboxyl modifying reagents such as carbodiimides, which react through O-acylurea formation followed by amide bond formation, may also be employed.
It will be appreciated that functional' groups in compound (A) and/or compound (B) may, if desired, be converted to other functional groups prior to reaction, for example, to confer additional reactivity or selectivity. Examples of methods useful for this purpose include conversion of amines to carboxyls using reagents such as dicarboxylic anhydrides; conversion of amines to thiols using reagents such as N-acetylhomocysteine thiolactone, S- acetylmercaptosuccinic anhydride, 2-iminothiolane, or thiol-containing succinimidyl derivatives; conversion of thiols to carboxyls using reagents such as α -haloacetates; conversion of thiols to amines using reagents such as ethylenimine or 2-bromoethylamine; conversion of carboxyls to amines using reagents such as carbodiimides followed by diamines; and conversion of alcohols to thiols using reagents such as tosyl chloride followed by transesterification with thioacetate and hydrolysis to the thiol with sodium acetate.
So-called zero-length linkers, involving direct covalent joining of a reactive chemical group of compound (A) with a reactive chemical group of compound (B) without introducing additional linking material may, if desired, be used in accordance with the invention.
Most commonly, however, the linker will include two or more reactive moieties, as described above, connected by a spacer element. The presence of such a spacer permits bifunctional linkers to react with specific functional groups within compound (A) and compound (B), resulting in a covalent linkage between the two. The reactive moieties in a linker may be the same (homobifunctional linker) or different (heterobifunctional linker, or, where several dissimilar reactive moieties are present, heteromultifunctional linker), providing a diversity of potential reagents that may bring about covalent attachment between compound (A) and compound (B).
Spacer elements in the linker typically consist of linear or branched chains and may include a Ci_io alkyl, C2-Io alkenyl, C2-Io alkynyl, C2-^ heterocyclyl, C6_i2 aryl, C7_)4 alkaryl, C3_i0 alkheterocyclyl, or Ci_i0 heteroalkyl. In some instances, the linker is described by formula (XII):
G1-(Z1)0-(Y1)u-(Z2)s-(R3o)-(Z3)r(Y2)v-(Z4)p-G2 (XII)
In formula (XII), G1 is a bond between compound (A) and the linker; G2 is a bond between the linker and compound (B); Z1, Z2, Z3, and Z4 each, independently, is selected from O, S, and NR31; R3i is hydrogen, Ci-^ alkyl, C2- 4 alkenyl, C2-A alkynyl, C2_6 heterocyclyl, C6-I2 aryl, C7_i4 alkaryl, C3_i0 alkheterocyclyl, or Ci_7 heteroalkyl; Y1 and Y2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; o, p, s, t, u, and v are each, independently, 0 or 1 ; and R30 is a C1-I0 alkyl, C2_i0 alkenyl, C2_i0 alkynyl, C2^ heterocyclyl, C6-I2 aryl, C7_i4 alkaryl, C3_i0 alkheterocyclyl, or Ci_ 10 heteroalkyl, or a chemical bond linking G1-(Z')O-(Y1)U-(Z2)S- to -(Z3)t-(Y2)V- (Z4)p-G2.
Examples of homobifunctional linkers useful in the preparation of conjugates of the invention include, without limitation, diamines and diols selected from ethylenediamine, propylenediamine and hexamethylenediamine, ethylene glycol, diethylene glycol, propylene glycol, 1 ,4-butanediol, 1 ,6- hexanediol, cyclohexanediol, and polycaprolactone diol.
Exemplary uses
The methods, compositions, and kits of the invention can be used to treat pain associated with any of a number of conditions, including back and neck pain, cancer pain, gynecological and labor pain, fibromyalgia, arthritis and other rheumatological pains, orthopedic pains, post herpetic neuralgia and other neuropathic pains, sickle cell crises, interstitial cystitis, urethritis and other urological pains, dental pain, headaches, postoperative pain, and procedural pain (i.e., pain associated with injections, draining an abcess, surgeiy, dental procedures, opthalmic procedures, arthroscopies and use of other medical instrumentation, cosmetic surgical procedures, dermatological procedures, setting fractures, biopsies, and the like).
Since a subclass of nociceptors mediate itch sensation the methods, compositions, and kits of the invention can also be used to treat itch in patients with conditions like dermatitis, infections, parasites, insect bites, pregnancy, metabolic disorders, liver or renal failure, drug reactions, allergic reactions, eczema, and cancer.
Pain and function indices
In order to measure the efficacy of any of the methods, compositions, or kits of the invention, a measurement index may be used. Indices that are useful in the methods, compositions, and kits of the invention for the measurement of pain associated with musculoskeletal, immunoinflammatory and neuropathic disorders include a visual analog scale (VAS), a Likert scale, categorical pain scales, descriptors, the Lequesne index, the WOMAC index, and the AUSCAN index, each of which is well known in the art. Such indices may be used to measure pain, itch, function, stiffness, or other variables.
A visual analog scale (VAS) provides a measure of a one-dimensional quantity. A VAS generally utilizes a representation of distance, such as a picture of a line with hash marks drawn at regular distance intervals, e.g., ten 1- cm intervals. For example, a patient can be asked to rank a sensation of pain or itch by choosing the spot on the line that best corresponds to the sensation of pain or itch, where one end of the line corresponds to "no pain" (score of 0 cm) or "no itch" and the other end of the line corresponds to "unbearable pain" or "unbearable itch" (score of 10 cm). This procedure provides a simple and rapid approach to obtaining quantitative information about how the patient is experiencing pain or itch. VAS scales and their use are described, e.g., in U.S. Patent Nos. 6,709,406 and 6,432,937. A Likert scale similarly provides a measures f a one-dimensional quantity. Generally, a Likert scale has discrete integer values ranging from a low value (e.g., O, meaning no pain) to a high value (e.g., 7, meaning extreme pain). A patient experiencing pain is asked to choose a number between the low value and the high value to represent the degree of pain experienced. Likert scales and their use are described, e.g., in U.S. Patent Nos. 6,623,040 and 6,766,319.
The Lequesne index and the Western Ontario and McMaster Universities (WOMAC) osteoarthritis index assess pain, function, and stiffness in the knee and hip of OA patients using self-administered questionnaires. Both knee and hip are encompassed by the WOMAC, whereas there is one Lequesne questionnaire for the knee and a separate one for the hip. These questionnaires are useful because they contain more information content in comparison with VAS or Likert. Both the WOMAC index and the Lequesne index questionnaires have been extensively validated in OA, including in surgical settings (e.g., knee and hip arthroplasty). Their metric characteristics do not differ significantly.
The AUSCAN (Australian-Canadian hand arthritis) index employs a valid, reliable, and responsive patient self-reported questionnaire. In one instance, this questionnaire contains 15 questions within three dimensions (Pain, 5 questions; Stiffness, 1 question; and Physical function, 9 questions). An AUSCAN index may utilize, e.g.. a Likert or a VAS scale.
Indices that are useful in the methods, compositions, and kits of the invention for the measurement of pain include the Pain Descriptor Scale (PDS), the Visual Analog Scale (VAS), the Verbal Descriptor Scales (VDS), the Numeric Pain Intensity Scale (NPIS), the Neuropathic Pain Scale (NPS), the Neuropathic Pain Symptom Inventory (NPSI), the Present Pain Inventory (PPI), the Geriatric Pain Measure (GPM), the McGiIl Pain Questionnaire (MPQ), mean pain intensity (Descriptor Differential Scale), numeric pain scale (NPS) global evaluation score (GES) the Short-Form McGiIl Pain Questionnaire, the Minnesota Multiphasic Personality Inventory, the Pain Profile and Multidimensional Pain Inventory, the Child Heath Questionnaire, and the Child Assessment Questionnaire.
Itch can be measured by subjective measures (VAS, Lickert, descriptors). Another approach is to measure scratch which is an objective correlate of itch using a vibration transducer or movement-sensitive meters.
Screening
Our discovery that certain channels expressed by and present on nociceptors and pruriceptors allow entry of compounds that inhibit voltage- gated ion channels into the target cells provides a method for identifying compounds as being useful for the treatment of pain and itch. In one example, a nociceptor or pruriceptor is contacted with a one, two, or more compounds that activate TRPV l , TRPAl, TRPM8 and/or P2X(2/3) receptors. The same nociceptor or pruriceptor is also contacted with a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of the nociceptor (e.g., by intracellular application via micropipette in the whole-cell patch-clamp technique) but not when applied to the external face of the cell (because of the inability of the compound to cross the cell membrane). Inhibition of the ion channels in the nociceptor or pruriceptor will inhibit the cell from propagating an action potential and/or signalling to the second order neuron, in either case blocking the transmission of the pain signal, thus, the ability of the second compound to inhibit voltage-gated ion channels in the nociceptor identifies that compound as one that can be used in combination with compounds that activate TRPVl, TRPAl, TRPM8 and/or P2X(2/3) receptors to treat pain or itch. The following examples are intended to illustrate the invention, and is not intended to limit it.
Example 1
We recorded current through voltage-dependent sodium channels using whole-cell voltage clamp recordings from adult rat DRG neurons. To select for nociceptors, we recorded from small (24 ± 5 μm; n=25) neurons and tested the neurons for the expression of TRPVl receptors by a short (1-sec) application of 1 μM capsaicin. In 25/25 of small neurons tested, capsaicin produced a prolonged (10 ± 3 sec) inward current (Fig. IA, upper panel), consistent with the neurons being nociceptors. Sodium currents were elicited by depolarizing steps from a holding potential of -70 mV. Bath application of 5 mM QX-314 alone had a minimal effect on sodium current (decrease by 3 ± 0.5% after a 5- minute application, n=25) (Fig. IA, left; b). Application of capsaicin alone (1 μM for 1-10 minutes) reduced sodium current moderately (31 ± 9% inhibition (n=25). However, when QX-314 was applied together with capsaicin, sodium current was nearly totally abolished (inhibition by 98 ± 0.4%, n=25) (Fig. IA, left; b). As expected if the block of sodium current resulted from gradual entry of QX-314 through TRPVl receptors, inhibition developed over several minutes and was nearly complete after 15 minutes (Fig. 1C).
To test whether the ability of co-applied capsaicin and QX-314 to inhibit sodium current is selective for cells that express TRPVl receptors, we also recorded from large DRG neurons (soma diameter > 40 μm) (Fig. IA, right). In these neurons, capsaicin did not elicit an inward current (10 of 10). As for small diameter neurons, QX-314 applied alone had little or no effect on sodium current (current increased by 8 ± 4% after a 10-minute application, n=10). Unlike small diameter neurons, capsaicin had no effect on sodium current in large diameter neurons (average increase by 3 ± 2% after a 10-minute application, n=10). Most notably, co-application of QX-314 and capsaicin had little or no effect on sodium current in the large diameter neurons (decrease by 9 ± 5% after a 10-minute application, n=10). Thus, the ability of co-applied QX-314 and capsaicin to inhibit sodium current is highly selective for neurons expressing TRPVl receptors, as expected if QX-314 enters the neurons through TRPVl receptors.
We also examined the effect of co-applied QX-314 and capsaicin in current clamp using physiological internal and external solutions. As expected from the voltage clamp results, co-application of QX-314 and capsaicin inhibited the excitability of small diameter neurons, completely blocking action potential generation (Fig. 2, 15 of 15 neurons).
We next examined if the combination of capsaicin and QX-314 can reduce pain behavior in vivo. Injection of QX-314 alone (10 μL of 2% solution) into the hindpaw of adult rats had no significant effect on the mechanical threshold for eliciting a withdrawal response, as determined by von Frey hairs (p=0.33) (Fig. 3A). Capsaicin alone (10 μg/lOμL) elicited spontaneous flinching (40 ± 6 flinches in 5 min), reflecting the direct irritant action of the capsaicin on nociceptors and after 15 and 30 minutes significantly reduced the mechanical threshold (p<0.05) (Fig. 3a), as expected. Injection of capsaicin and QX-314 together did not significantly change the number of flinches during the first 5 minutes after the injection (30 ± 7, p = 0.24). However, the combination completely abolished the later reduction in mechanical threshold normally produced by capsaicin alone (p = 0.14, measured at 15 minutes). Moreover, 60 minutes after the combined injection of capsaicin and QX-314, mechanical threshold actually increased to reach twice the baseline value, two hours after injection (46 ± 5 g vs. 24 ± 3 g, p<0.05). In three animals the paw was insensitive to even the highest value von Frey filament (57 g). The elevated mechanical threshold lasted for about three hours and then gradually returned back to basal levels by four hours (Fig. 3A). Similar effects were seen examining sensitivity to a standardized noxious radiant heat stimulus. Unexpectedly, QX-314 alone transiently reduced the thermal response latency at 30 min after the injection (p<0.01 at 30 min; p> 0.05 for all other time points) (Fig. 3B). Capsaicin (10 μg/lOμL) alone also reduced as expected the thermal response latency (p<0.01 15 and 30 min) (Fig. 3B). However, while both QX-314 and capsaicin alone increased heat sensitivity, the co-application of QX-314 and capsaicin together progressively anesthetized the animals to noxious heat, such that 2 hours after the injection no animal reacted to the radiant noxious heat applied for 25 seconds. This effect remained for 4 hours after the injection (Fig. 3B).
We next tested if capsaicin and QX-314 co-administration can be used to produce regional nerve block without the motor effects seen when local anesthesia is produced by lidocaine. Motor effects were scored according to a scale of 0 (no effect; normal gait and limb placement), 1 (limb movement but with abnormal limb placement and movement) or 2 (complete loss of limb movement). Injection of 2% lidocaine (a standard concentration for local nerve block) in close proximity to the sciatic nerve caused complete paralysis of the lower limb when assayed at 15 minutes (6 of 6 animals) and complete or partial paralysis was still present at 30 minutes (mean motor score 1.67 ± 0.2, p<0.01 ; Fig. 4C). There was a complete loss of the tactile stimulus-evoked placing reflex lasting for at least 30 minutes in all animals with full recovery of these sensory and motor deficits by 45 minutes (Fig. 4). During the period of paralysis, it was not possible to assay sensory sensitivity. In pilot experiments with QX-314, it became clear that much lower concentrations of QX-314 than lidocaine could be used to produce effective local anesthesia when applied with capsaicin. Injection of QX-314 (0.2%, 100 μL) alone had no effect on motor function (6 of 6 animals; Fig. 4C) and also had no significant effect on either mechanical threshold (p=0.7) or thermal response latency (p=0.66) (Fig. 4A, 4B). Capsaicin alone (0.5μg/μL, 100 μL) injected near the nerve reduced both mechanical threshold (p<0.05) and thermal latency (p<0.05) for 30 min after injection (Fig. 4A, 4B). During this period 4 out of the 6 animals demonstrated a sustained flexion of the injected limb leading to a slight impairment of locomotion (mean motor score 0.7 ± 0.2, pO.Ol) but movement of the knee and hip as well as the placing reflex were unchanged. We interpret the sensitivity and motor changes as reflecting activation of nociceptor axons producing a sustained flexion reflex. For co-application of QX- 314 and capsaicin into the para-sciatic nerve region, we injected QX-314 first, followed 10 minutes later by capsaicin, with the idea that QX-314 would be present extracellularly and ready to enter TRPVl channels as soon as they were activated. Indeed, there was little or no behavioral response to the capsaicin injection when preceded by QX-314 injection, and the behavioral responses indicated that there was effective anesthesia to noxious stimuli. There was a very marked increase in mechanical threshold such that all animals showed no response to the stiffest von Frey hair (57 g; vs. pre-injection withdrawal to stimuli averaging 15.2± 3.4; pO.Ol, n=6) and also in the thermal response latency (22.3 ± 2.3 s vs. 14.9 ± 0.4 s, p<0.05, n=6). These changes were evident at 15 min after the capsaicin injection for the mechanical stimuli and at 30 rain for the thermal stimuli and lasted for 90 minutes (Fig. 4A, 4B). Five of six animals had no motor deficit whatsoever (mean motor score 0. 17 ± 0.17, p=0.34) (Fig. 4C) and no change in the placing reflex. One animal demonstrated sustained flexion similar to that observed when capsaicin was injected alone, but more transient.
Methods
Electrophysiology
Dorsal root ganglia from 6-8 week old Sprague-Dawley rats were removed and placed into Dulbecco's Minimum Essential Medium containing 1% penicillin-streptomycin (Sigma), then treated for 90 minutes with 5 mg/ml collagenase, lrng/ml Dispase II (Roche, Indianapolis, IN) and for 7 minutes with 0.25% trypsin, followed by addition of 2.5% trypsin inhibitor. Cells were triturated in the presence of DNAase I inhibitor (50 U), centrifuged through 15% BSA (Sigma), resuspended in ImI Neurobasat medium (Sigma), 10 μM AraC, NGF (50 ng/ml) and GDNF (2 ng/ml) and plated onto poly-lysine (500 μg/ml) and laminin (5 mg/ml) coated 35 mm tissue culture dishes (Becton Dickinson) at 8000-9000 per well. Cultures were incubated at 37 0C, 5% carbon dioxide. Recordings were made within 48 hours after plating. Average size of small neurons chosen as likely nociceptors was 23 ± 6 μm (n=50) and that of large neurons was 48 ± 8 μm (n=10).
Whole-cell voltage-clamp or current-clamp recordings were made using an Axopatch 200A amplifier (Axon Instruments, Union CJ ty, CA) and patch pipettes with resistances of 1-2 MΩ. For voltage-clamp recordings pipette capacitance was reduced by wrapping the shank by Parafilm or coating the shank with Sylgard (Dow Corning, Midland, MI). Cell capacitance was compensated for using the amplifier circuitry, and linear leakage currents subtracted using a P/4 procedure. Series resistance (usually 3-7 MΩ and always less than 10 MΩ) was compensated by ~80%. Voltage clamp recordings used solutions designed to isolate sodium currents by blocking potassium and calcium currents and with reduced external sodium to improve voltage clamp. Pipette solution was 110 mM CsCl, ImM CaCl2, 2 mM MgCl2, 1 ImM EGTA, and 10 mM HEPES, pH adjusted to 7.4 with -25 mM CsOH. External solution was 60 mM NaCl, 60 mM choline chloride, 4 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 0.1 mM CdCl2, 15 mM tetraethylammonium chloride, 5 mM 4-aminopyridine, 10 mM glucose, and 10 mM HEPES, pH adjusted to 7.4 with NaOH. No correction was made for the small liquid junction potential (-2.2 mV). Current clamp recordings were made using the fast current clamp mode of the Axopatch 200A amplifier Pipette solution was 135 mM K gluconate; 2 mM MgCl2; 6 mM KCl; 10 mM HEPES; 5 mM Mg ATP; 0.5 mM Li2GTP; (pH = 7.4 with KOH). External solution was 145 mM NaCl; 5 mM KCl; 1 mM MgCl2; 2 mM CaCl2; 10 mM HEPES; 10 mM glucose; (pH adjusted to 7.4 with NaOH). Membrane potential was corrected for a liquid junction potential of -15 mV.
Command protocols were generated and data digitized using a Digidata 1200 A/D interface with pCLAMP 8.2 software (Axon Instruments, Union City, CA). Voltage-clamp current records were low pass filtered at 2 kHz and current clamp recordings at 10 kHz (-3 dB, 4 pole Bessel filter).
QX-314 (5 mM), capsaicin (1 μM or 500 nM), or their combination was applied using custom-designed multibarrel fast drug delivery system placed about 200-250 μm from the neuron. Solution exchange was complete in less than a second.
Behavior
For intraplantar injections, rats were first habituated to handling and tests performed with the experimenter blind to the treatment. Intraplantar injections of vehicle (20 % ethanol, 5% Tween 20 in saline, 10 μL) capsaicin (1 μg/μL), QX-314 (2%) or mixture of capsaicin and QX-314 into the left hindpaw were made and mechanical and thermal sensitivities determined using von Frey hairs and radiant heat respectively.
For sciatic nerve injections, animals were first habituated to handling for 10 days. Lidocaine (0.2% or 2%, 100 μL); QX-314 (0.2%, 100 μL) alone; capsaicin (50 μg in 100 μL) alone, or QX-314 followed by capsaicin (10 minutes interval) were injected into the area of sciatic nerve below the hip joint. Mechanical and thermal thresholds were determined using von Frey filaments and radiant heat. Motor function of the injected leg was assessed every 15 minutes using the following grading score: 0 = none; 1 = partially blocked; and 2 = fully blocked. Walking, climbing, walking on the rod and placing reflex were examined. Motor blockade was graded as none when gait was normal and there was no visible limb weakness; as partially blocked when the limb could move but movements were abnormal and could not support the normal posture; and as completely blocked when the limb was flaccid and without resistance to extension of the limb. All experiments were done with the experimenter blinded.
Statistical analysis
Statistics were analyzed using Students t test or one-way ANOVA, followed by Dunnett's test as appropriate. For the motor scoring the data obtained after injection of lidocaine 0.2% used as a control for the Dunnett's test. Data represented as mean + SEM.
Example 2
We have also shown that eugenol (Ci0H12O2), an allyl chain-substituted guaiacol, 2-methoxy-4-(2-propenyl)phenol (active ingredient in oil of clove, and a non-pungent agonist of TRPVl receptors) promotes entry of QX-314 into dorsal root ganglion neurons by activating TRPVl channels. Fig. 5 depicts voltage clamp recordings of sodium channel current in small dorsal root ganglion neurons. The data show that eugenol alone has a modest inhibitory effect on sodium current (10-20% inhibition). Co-application of eugenol and QX-314 produces progressive block that can be complete after 7 minutes. Two examples are depicted, which are representative of 10 experiments with similar results. As is demonstrated above, external QX-314 alone has no effect while internal QX-314 blocks sodium channels. Thus, these experiments indicate that eugenol promotes entry of QX-314 into dorsal root ganglion neurons by activating TRPVl channels. Example 3
Fig. 6 shows the results of co-application of the TRPA agonist mustard oil (MO) (50 μM) and QX-314 (5 mM). MO alone reduces sodium current by 20-30% and reaches a plateau after approximately 3 minutes. Co-application of MO and QX-314 reduced sodium current dramatically.
Other Embodiments
Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific desired embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the fields of medicine, immunology, pharmacology, endocrinology, or related fields are intended to be within the scope of the invention.
AU publications mentioned in this specification are herein incorporated by reference to the same extent as if each independent publication was specifically and individually incorporated by reference.
What is claimed is:

Claims

Claims
1. A method for treating pain or itch in a patient, said method comprising administering to said patient:
(i) a first compound that activates a channel-forming receptor that is present on nociceptors and/or pruriceptors; and
(ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of said channels but does not substantially inhibit said channels when applied to the external face of said channels, wherein said second compound is capable of entering nociceptors or pruriceptors through said channel-forming receptor when said receptor is activated.
2. The method of claim I3 wherein said first compound activates a channel-forming receptor selected from TRPVl, P2X(2/3), TRPAl3 and TRPM8.
3. The method of claim 2, wherein said first compound is an activator of TRPVl receptors, said activator selected from capsaicin, eugenol, arvanil (N-arachidonoylvanillamine), anandamide, 2-aminoethoxydiphenyl borate (2APB)3 AM404, resiniferatoxin, phorbol 12-phenylacetate 13-acetate 20- homovanillate (PPAHV), olvanil (NE 19550), OLDA (N-oleoyldopamine), N- arachidonyldopamine (NADA)3 6'-iodoresiniferatoxin (6'-IRTX), Cl 8 N- acylethanolamines, lipoxygenase derivatives such as 12- hydroperoxyeicosatetraenoic acid, inhibitor cysteine knot (ICK) peptides (vanillotoxins), pipeline, MSKl 95 (N-[2-(3,4-dimethylbenzyl)-3- (pivaloyloxy)propyl]-2-[4-(2-aminoethoxy)-3-methoxyphenyl]acetamide), JYL79 (N-[2-(3,4-dimethylbenzyl)-3-(pivaloyloxy)propyl]-N'-(4-hydroxy-3- methoxybenzyl)thiourea), hydroxy-alpha-sanshool, 2-aminoethoxydiphenyl borate, 10-shogaol, oleylgingerol, oleylshogaol, and SU200 (N-(4-tert- butylbenzyl)-N'-(4-hydroxy-3-methoxybenzyl)thiourea).
4. The method of claim 2, wherein said first compound is an activator of TRPAl receptors, said activator selected from cinnamaldehyde, allyl- isothiocynanate, diallyl disulfide, icilin, cinnamon oil, wintergreen oil, clove oil, acrolein, hydroxy-alpha-sanshool, 2-aminoethoxydiphenyl borate, 4- hydroxynonenal, methyl p-hydroxybenzoate, mustard oil, and 3'- carbamoylbiphenyl-3-yl cyclohexylcarbamate (URB597).
5. The method of claim 2, wherein said first compound is an activator of P2X receptors, said activator selected from ATP, 2-methylthio-ATP, 2' and 3'-<9-(4-benzoylbenzoyl)-ATP, and ATP5'-(9-(3-thiotriphosphate).
6. The method of claim 2, wherein said first compound is an activator of TRPM8 receptors, said activator selected from menthol, iciclin, eucalyptol, linalool, geraniol, and hydroxycitronellal.
7. The method of any of claims 1-6, wherein said second compound inhibits voltage-gated sodium channels.
8. The method of claim 7, wherein said second compound is QX-314, N-methyl-procaine, QX-222, N-octyl-guanidine, 9-aminoacridine, pancuronium, or another low molecular weight, charged molecule that inhibits voltage-gated sodium channels when present inside of the cell.
9. The method of any of claims 1-6, wherein said second compound inhibits voltage-gated calcium channels.
10. The method of claim 9, wherein said compound is D-890 (quaternary methoxyverapamil), CERM 11888 (quaternary bepridil), or another low molecular weight, charged molecule that inhibits voltage-gated calcium channels when present inside of the cell.
1 1. The method of any one of claims 1-6, wherein said second compound is a quarternary amine derivative or other charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, tocainide, prilocaine, articaine, bupivicaine, mepivicine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene.
12. The method of any one of claims 1-11 , wherein said pain is neuropathic pain.
13. The method of any one of claims 1-11 , wherein said pain is inflammatory pain.
14. The method of any one of claims 1 -11, wherein said pain is nociceptive pain.
15. The method of any one of claims 1-11 , wherein said pain is procedural pain.
16. A composition comprising:
(i) a first compound that activates a channel-forming receptor that is present on nociceptors and/or pruriceptors; and
(ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of said channels but does not substantially inhibit said channels when applied to the external face of said channels, wherein said second compound is capable of entering nociceptors or pruriceptors through said channel-forming receptor when said receptor is activated.
17. The composition of claim 16, wherein said first compound activates a receptor selected from TRPVl , P2X(2/3), TRPAl, and TRPM8.
18. The composition of claim 16 or 17, said composition formulated for oral, parenteral (e.g., intravenous, intramuscular), rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intrathecal, epidural, or ocular administration, or by injection, inhalation, or direct contact with the nasal or oral mucosa.
19. A method for inhibiting one or more voltage-gated ion channels in a cell, said method comprising contacting said cell with:
(i) a first compound that activates a channel-forming receptor that is present on nociceptors and/or pruriceptors; and
(ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of said channels but does not substantially inhibit said channels when applied to the external face of said channels, wherein said second compound is capable of entering nociceptors or pruriceptors through said channel-forming receptor when said receptor is activated.
20. The method of claim 20, wherein said first compound activates a receptor selected from TRPVl, P2X(2/3), TRPAl, and TRPM8.
21. A method for identifying a compound as being useful for the treatment of pain or itch, said method comprising the steps of:
(a) contacting the external face of TRPVl, TRPAl, TRPM8, or P2X(2/3)-expressing neurons with:
(i) a first compound that activates TRPVl, TRPAl, TRPM8 or P2X(2/3) receptors; and
(ii) a second compound that inhibits one or more voltage-gated ion channels when applied to the internal face of said channels but does not substantially inhibit said channels when applied to the external face of said channels, and
(b) determining whether said second compound inhibits said voltage- gated ion channels in said neurons, wherein inhibition of said voltage-gated ion channels by said second compound identifies said second compound as a compound that is useful for the treatment of pain or itch.
22. A quarternary amine derivative or other permanently or transiently charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, articaine, bupivicaine, mepivicaine, tocainide, prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene.
23. The quarternary amine derivative or other charged derivative of claim 22, wherein said compound has the formula of any one of formulas (I)- (X).
24. A pharmaceutical composition comprising (i) a quarternary amine derivative or other permanently or transiently charged derivative of a compound selected from riluzole, mexilitine, phenytoin, carbamazepine, procaine, aiticaine, bupivicaine, mepivicaine, tocainide. prilocaine, diisopyramide, bencyclane, quinidine, bretylium, lifarizine, lamotrigine, flunarizine, and fluspirilene, and (ii) a pharmaceutically acceptable excipient.
25. The composition of claim 24, wherein said compound has the formula of any one of formulas (I)-(X).
PCT/US2007/024174 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis WO2008063603A2 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
US12/515,429 US20100099772A1 (en) 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis
JP2009537235A JP2010510227A (en) 2006-11-20 2007-11-19 Pain and itching treatment methods, compositions and kits
DK07862114.1T DK2101819T3 (en) 2006-11-20 2007-11-19 Methods, compositions and kits for the treatment of pain and pruritis
ES07862114T ES2402789T3 (en) 2006-11-20 2007-11-19 Methods, compositions and kits to treat pain and pruritus
EP07862114A EP2101819B1 (en) 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis
AU2007322033A AU2007322033B2 (en) 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis
KR1020147017308A KR20140097446A (en) 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis
CA2668652A CA2668652C (en) 2006-11-20 2007-11-19 Inhibitors of voltage-gated ion channels for use in treating pain and pruritis
EP11007949.8A EP2446903B1 (en) 2006-11-20 2007-11-19 Compositions for treating itch
US14/496,629 US9603817B2 (en) 2006-11-20 2014-09-25 Methods, compositions, and kits for treating pain and pruritis
US15/470,324 US10179116B2 (en) 2006-11-20 2017-03-27 Methods, compositions, and kits for treating pain and pruritis
US16/851,349 US20210186906A1 (en) 2006-11-20 2020-04-17 Methods, compositions, and kits for treating pain and pruritis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US86012406P 2006-11-20 2006-11-20
US60/860,124 2006-11-20
US95859407P 2007-07-06 2007-07-06
US60/958,594 2007-07-06
US99751007P 2007-10-03 2007-10-03
US60/997,510 2007-10-03

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/515,429 A-371-Of-International US20100099772A1 (en) 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis
US14/496,629 Continuation US9603817B2 (en) 2006-11-20 2014-09-25 Methods, compositions, and kits for treating pain and pruritis

Publications (2)

Publication Number Publication Date
WO2008063603A2 true WO2008063603A2 (en) 2008-05-29
WO2008063603A3 WO2008063603A3 (en) 2008-12-11

Family

ID=39430358

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/024174 WO2008063603A2 (en) 2006-11-20 2007-11-19 Methods, compositions, and kits for treating pain and pruritis

Country Status (10)

Country Link
US (4) US20100099772A1 (en)
EP (3) EP2425858A3 (en)
JP (4) JP2010510227A (en)
KR (2) KR20090082507A (en)
CN (1) CN103933572B (en)
AU (1) AU2007322033B2 (en)
CA (2) CA3002443A1 (en)
DK (1) DK2101819T3 (en)
ES (1) ES2402789T3 (en)
WO (1) WO2008063603A2 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009143175A2 (en) * 2008-05-19 2009-11-26 Children's Medical Center Corporation Sensory-specific local anesthesia and prolonged duration local anesthesia
WO2009114139A3 (en) * 2008-03-11 2009-12-23 President And Fellows Of Harvard College Methods, compositions, and kits for treating pain and pruritis
EP2142182A1 (en) * 2007-02-06 2010-01-13 Origin BioMed Inc. Composition comprising terpene compounds and methods for inhibiting nerve transmission
WO2011006073A1 (en) * 2009-07-10 2011-01-13 President And Fellows Of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
US20110144191A1 (en) * 2008-08-13 2011-06-16 Mclellan Alexander Compositions comprising terpene compounds for treating negative sensory phenomena
KR101188945B1 (en) 2009-01-29 2012-10-08 서울대학교산학협력단 Composition containing sulforaphane and its analog for treating pain
KR101243430B1 (en) * 2012-04-23 2013-03-13 서울대학교산학협력단 Composition of health-promoting food containing sulforaphane and its analog for relieving pain
WO2013063127A1 (en) * 2011-10-24 2013-05-02 Endo Pharmaceuticals Inc. Cyclohexylamines
US20130150455A1 (en) * 2011-12-07 2013-06-13 Snu R&Db Foundation Method for treating mechanical allodynia comprising administration of eugenol
US8865741B2 (en) 2011-02-18 2014-10-21 Asana Biosciences, Llc Aminoindane compounds and use thereof in treating pain
US8952152B2 (en) 2009-03-24 2015-02-10 Proteus S.A. Methods for purifying phycotoxins, pharmaceutical compositions containing purified phycotoxins, and methods of use thereof
US8975268B2 (en) 2013-03-15 2015-03-10 The Children's Medical Center Corporation Neosaxitoxin combination formulations for prolonged local anesthesia
US9044482B2 (en) 2012-08-15 2015-06-02 Asana Biosciences, Llc Use of aminoindane compounds in treating overactive bladder and interstitial cystitis
US10179116B2 (en) 2006-11-20 2019-01-15 President And Fellows Of Harvard College Methods, compositions, and kits for treating pain and pruritis
US10780083B1 (en) 2019-03-11 2020-09-22 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10786485B1 (en) 2019-03-11 2020-09-29 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10842798B1 (en) 2019-11-06 2020-11-24 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10927096B2 (en) 2019-03-11 2021-02-23 Nocion Therapeutics, Inc. Ester substituted ion channel blockers and methods for use
US10934263B2 (en) 2019-03-11 2021-03-02 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10933055B1 (en) 2019-11-06 2021-03-02 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10968179B2 (en) 2019-03-11 2021-04-06 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US11021443B2 (en) 2015-08-03 2021-06-01 President And Fellows Of Harvard College Charged ion channel blockers and methods for use
US11332446B2 (en) 2020-03-11 2022-05-17 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
EP4428132A1 (en) * 2023-03-08 2024-09-11 Johann-Wolfgang-Goethe-Universität Frankfurt am Main Slack-activating compounds and their medical use
WO2024184543A1 (en) * 2023-03-08 2024-09-12 Johann Wolfgang Goethe-Universität Frankfurt am Main Slack-activating compounds and their medical use

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2229391B1 (en) 2007-12-19 2014-08-06 Cancer Research Technology Limited Pyrido[2,3-b]pyrazine-8-substituted compounds and their use
WO2010096496A2 (en) 2009-02-17 2010-08-26 Memorial Sloan-Kettering Cancer Center Methods of neural conversion of human embryonic stem cells
EP2577318B1 (en) * 2010-05-25 2019-06-26 Memorial Sloan-Kettering Cancer Center Method of nociceptor differentiantion of human embryonic stem cells and uses thereof
EP3295794A1 (en) 2010-07-27 2018-03-21 Flex Pharma, Inc. Methods and compositions for preventing and relieving muscle cramps and for recovery from neuromuscular irritability and fatigue following exercise
IL295556A (en) 2011-11-04 2022-10-01 Memorial Sloan Kettering Cancer Center Midbrain dopamine (da) neurons for engraftment
JP4979830B1 (en) * 2011-11-10 2012-07-18 重憲 青木 Painless topical injection
ES2936634T3 (en) 2013-02-08 2023-03-21 Gen Mills Inc Reduced Sodium Food Products
EP3498824A1 (en) 2013-04-26 2019-06-19 Memorial Sloan-Kettering Cancer Center Cortical interneurons and other neuronal cells produced by the directed differentiation of pluripotent and multipotent cells
EP2842428B1 (en) * 2013-08-27 2018-08-08 Symrise AG Oral composition
KR101856589B1 (en) 2015-07-30 2018-05-10 (주)에이엔티랩스 Methods for predicting cutaneous adverse drug reaction of lamotrigine
JP6970691B2 (en) * 2016-05-19 2021-11-24 グラクソスミスクライン、インテレクチュアル、プロパティー、(ナンバー2)、リミテッドGlaxosmithkline Intellectual Property (No.2) Limited TRPV4 antagonist
EP3570861A4 (en) 2017-01-23 2020-11-18 Flex Pharma, Inc. Compositions and methods affecting exercise performance
WO2019191200A1 (en) * 2018-03-27 2019-10-03 American Genomics, Llc Method and formulation for producing anesthesia of internal aspect of eye wall by topical application
WO2020008261A2 (en) * 2018-07-05 2020-01-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Methods and compositions for treating pain and itch
US11617727B2 (en) 2019-04-30 2023-04-04 Bayer Healthcare Llc Topical analgesic gel compositions
CN115243683A (en) * 2020-03-04 2022-10-25 纽克尔斯有限公司 Use of QX314 to prevent sympathetic excitation associated with administration of TRPV1 modulators
US20230372330A1 (en) * 2020-10-13 2023-11-23 New York Blood Center, Inc. Compounds for treatment of hemolysis-and inflammasome-associated diseases
WO2022099284A1 (en) * 2020-11-05 2022-05-12 Young Biopharma, Llc Novel capsaicin analogs and uses thereof

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2441498A (en) 1943-07-15 1948-05-11 Astra Apotekarnes Kem Fab Alkyl glycinanilides
US2799679A (en) 1955-04-28 1957-07-16 Bofors Ab Process of preparing amides of heterocyclic carboxylic acids
US2955111A (en) 1957-01-28 1960-10-04 Bofors Ab Synthesis of n-alkyl-piperidine and n-alkyl-pyrrolidine-alpha-carboxylic acid amides
US3160662A (en) 1957-06-26 1964-12-08 Astra Apotekarnes Kem Fab Lower alkylaminoacyl amide anesthetics
DE2162744A1 (en) 1970-12-22 1972-07-06 Astra Pharmaceutical Products Inc., Worcester, Mass. (V.St.A.) 2-alkyl-2-alkylamino-2,6-acetoxylidides, processes for their preparation and their use
DE2235745A1 (en) 1971-07-28 1973-02-22 Astra Pharma Prod ANTIARRHYTHMIC COMPOUND, PROCESS FOR THEIR PRODUCTION AND THEIR USE
WO1985000599A1 (en) 1983-08-01 1985-02-14 A/S Apothekernes Laboratorium For Specialpraeparat L-N-n-PROPYLPIPECOLIC ACID-2,6-XYLIDIDE AND METHOD FOR PREPARING THE SAME
US6432937B1 (en) 1996-12-20 2002-08-13 Astra Aktiebolag Treatment for joint inflammation
US6623040B1 (en) 1997-09-03 2003-09-23 Recot, Inc. Method for determining forced choice consumer preferences by hedonic testing
US6709406B2 (en) 1996-01-02 2004-03-23 Cefar Matcher Ab Measurement of pain
US6766319B1 (en) 2000-10-31 2004-07-20 Robert J. Might Method and apparatus for gathering and evaluating information

Family Cites Families (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US812554A (en) 1905-11-08 1906-02-13 Hoechst Ag Alkamin esters of para-aminobenzoic acid.
US1889645A (en) 1930-12-22 1932-11-29 Winthrop Chem Co Inc Beta-dimethylaminoethyl ester of para-butylamino-benzoic acid
US2689248A (en) 1950-06-17 1954-09-14 Sterling Drug Inc Tertiary-aminoalkyl 4-amino-2-alkoxybenzoates and their synth esis
NL281394A (en) 1961-07-25
DE1251764B (en) * 1965-08-18 1967-10-12 C H Boehnnger Sohn Ingelheim/ Rhein Process for the production of antimicrobially active 12 3,4-Tetra hydro 9 -ammo acridmiumverbmdungen
US3931195A (en) 1971-03-03 1976-01-06 Mead Johnson & Company Substituted piperidines
US4069309A (en) * 1972-09-19 1978-01-17 Avon Products, Inc. Cationic skin substantive sunscreen composition and method
US3900481A (en) 1974-04-01 1975-08-19 Riker Laboratories Inc Derivatives of pyrrolidine and piperidine
US5591317A (en) 1994-02-16 1997-01-07 Pitts, Jr.; M. Michael Electrostatic device for water treatment
US5716622A (en) 1995-01-06 1998-02-10 The Rockefeller University Functionally active regions of signal transducer and activators of transcription
US5783683A (en) 1995-01-10 1998-07-21 Genta Inc. Antisense oligonucleotides which reduce expression of the FGFRI gene
US5747470A (en) 1995-06-07 1998-05-05 Gen-Probe Incorporated Method for inhibiting cellular proliferation using antisense oligonucleotides to gp130 mRNA
US6413961B1 (en) * 1995-12-12 2002-07-02 Omeros Medical Systems, Inc. Irrigation solution and method for inhibition of pain and inflammation
WO1998024428A1 (en) * 1996-12-02 1998-06-11 Brigham & Women's Hospital, Inc. Long-acting local anesthetics
US5837713A (en) 1997-02-26 1998-11-17 Mayo Foundation For Medical Education And Research Treatment of eosinophil-associated pathologies by administration of topical anesthetics and glucocorticoids
TW536401B (en) * 1997-09-03 2003-06-11 Cardiome Pharma Corp A pharmaceutical composition of N,N-bis(phenylcarbamoylmethyl)dimethylammomum chloride and derivatives for the treatment of pain
CN1311673A (en) * 1998-06-09 2001-09-05 诺特兰药品有限公司 Compositions and methods for treatment of cough
AU7839100A (en) 1999-10-01 2001-05-10 Advanced Medicine, Inc. Quinazolinones and analogues and their use as local anesthetics
EP1278736A1 (en) * 1999-12-15 2003-01-29 Ucb Farchim S.A. Quaternary salts of n-substituted cyclic or acyclic amines as pharmaceuticals
BR0016431A (en) 1999-12-15 2002-10-01 Ucb Farchim Sa Method for the treatment and / or prevention of cough in a warm-blooded animal, use of a compound, compound, and pharmaceutical composition
AU3162001A (en) * 1999-12-21 2001-07-03 Id-Pharma Gmbh Medicament, a method for its production and the use thereof
AU2001259561A1 (en) 2000-05-04 2001-11-12 Dzgenes, L.L.C. Tgfbeta-rii promoter polymorphisms
WO2002000218A2 (en) 2000-06-23 2002-01-03 Mayo Foundation For Medical Education And Research Methods of treating neutrophil-related diseases with topical anesthetics
DE10039449A1 (en) * 2000-08-11 2003-07-24 Id Pharma Gmbh I Ins Pharmaceutical composition, useful for the treatment or prevention of hyperalgesia, comprises an infusion solution of local anesthetic adjusted to alkaline pH
US6884782B2 (en) 2000-11-08 2005-04-26 Amgen Inc. STAT modulators
US20040146590A1 (en) * 2001-03-22 2004-07-29 Iadarola Michael J Molecular neurochirurgie for pain control administering locally capsaicin or resinferatoxin
WO2002089849A1 (en) * 2001-05-07 2002-11-14 Corium International Compositions and delivery systems for administration of a local anesthetic agent
JP4541695B2 (en) 2001-06-15 2010-09-08 バーテックス ファーマシューティカルズ インコーポレイテッド 5- (2-Aminopyrimidin-4-yl) benzisoxazole as a protein kinase inhibitor
US7585511B2 (en) 2002-08-02 2009-09-08 Universite Libre De Bruxelles Apolipoprotein L-I and/or derivated polypeptide for the treatment of and/or the prevention of diseases induced by trypanosoma
CA2522028C (en) 2003-04-10 2011-11-01 Neurogesx, Inc. Methods and compositions for administration of trpv1 agonists
WO2004093814A2 (en) 2003-04-22 2004-11-04 Pharmacia Corporation Compositions of a cyclooxygenase-2 selective inhibitor and a sodium channel blocker
ES2223277B1 (en) 2003-06-19 2006-03-01 Fernando Bouffard Fita ANESTHETIC COMPOSITION FOR TOPICAL ADMINISTRATION.
US20070149469A1 (en) 2003-10-02 2007-06-28 Christian Korherr Medical use of tbk-1 or of inhibitors thereof
US20050142596A1 (en) 2003-11-14 2005-06-30 Krolewski Andrzej S. Methods of diagnosing renal and cardiovascular disease
WO2005058829A1 (en) 2003-12-11 2005-06-30 Board Of Regents, The University Of Texas System Compounds for treatment of cell proliferative diseases
US20070196866A1 (en) * 2004-03-13 2007-08-23 Irm Llc Modulators of ion channel trpa1
WO2005105814A1 (en) 2004-04-28 2005-11-10 Incyte Corporation Tetracyclic inhibitors of janus kinases
WO2005117981A1 (en) * 2004-06-02 2005-12-15 Sri International Formulations comprising a capsaicinoid a local anesthetic and/or an antipruritic agent for the treatment of pain
JP2008507489A (en) * 2004-07-24 2008-03-13 ラボラトリオス・デル・ドクトル・エステベ・ソシエダッド・アノニマ Use of sigma receptor active compounds for the treatment of mechanical allodynia
WO2006036181A1 (en) 2004-09-20 2006-04-06 Corus Pharma, Inc. Inhalable lidocaine formulation for treatment of asthma and for reducing the need for corticosteroids in asthmatic patients
US7947682B2 (en) 2004-12-29 2011-05-24 University Of Southern California Substituted N′-pyrrolo[1,2-a]quinoxalin-4-yl-hydrazides as anti-cancer agents
JP2008514648A (en) 2004-12-16 2008-05-08 アドバンスト インハレーション リサーチ,インコーポレイテッド Compositions and methods for lung disease
AR054416A1 (en) 2004-12-22 2007-06-27 Incyte Corp PIRROLO [2,3-B] PIRIDIN-4-IL-AMINAS AND PIRROLO [2,3-B] PIRIMIDIN-4-IL-AMINAS AS INHIBITORS OF THE JANUS KINASES. PHARMACEUTICAL COMPOSITIONS.
EP2270014A1 (en) 2005-09-22 2011-01-05 Incyte Corporation Azepine inhibitors of janus kinases
CN101287742B (en) 2005-10-12 2016-01-06 艾德拉药物股份有限公司 Based on immunomodulatory oligonucleotide (IRO) compound of variation response modulation Toll-like receptor
MY159449A (en) 2005-12-13 2017-01-13 Incyte Holdings Corp Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
KR20080089416A (en) 2005-12-21 2008-10-06 페인셉터 파마 코포레이션 Compositions and methods for modulating gated ion channels
GB0526244D0 (en) 2005-12-22 2006-02-01 Novartis Ag Organic compounds
EP1867338A1 (en) 2006-05-30 2007-12-19 Université Libre De Bruxelles Pharmaceutical composition comprising apolipoproteins for the treatment of human diseases
KR20090082507A (en) 2006-11-20 2009-07-30 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Methods, compositions, and kits for treating pain and pruritis
CA2673038C (en) 2006-12-22 2015-12-15 Incyte Corporation Substituted tricyclic heteroaryl compounds as janus kinase inhibitors
US7705004B2 (en) 2007-08-17 2010-04-27 Portola Pharmaceuticals, Inc. Protein kinase inhibitors
CN101156851A (en) 2007-09-27 2008-04-09 刘全胜 Nicardipine hydrochloride dispersion piece and method for making same
KR20110044166A (en) 2008-03-11 2011-04-28 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Methods, compositions, and kits for treating pain and pruritis
US8138339B2 (en) 2008-04-16 2012-03-20 Portola Pharmaceuticals, Inc. Inhibitors of protein kinases
AU2009238590A1 (en) 2008-04-22 2009-10-29 Portola Pharmaceuticals, Inc. Inhibitors of protein kinases
KR20110033922A (en) 2008-07-08 2011-04-01 더 보드 오브 리전츠 오브 더 유니버시티 오브 텍사스 시스템 Novel inhibitors of proliferation and activation of signal transducer and activator of transcription(stats)
DE102008037682A1 (en) 2008-08-14 2010-04-08 Strackharn, Klaus, Dr.med. Use of equipotent doses of local anesthetics or derivatives thereof for the treatment of chronic pain
CN101347427A (en) 2008-09-22 2009-01-21 北京理工大学 Compound of losartan compound or its medical salt and calcium channel blocker or its medical salt
CL2009001884A1 (en) 2008-10-02 2010-05-14 Incyte Holdings Corp Use of 3-cyclopentyl-3- [4- (7h-pyrrolo [2,3-d] pyrimidin-4-yl) -1h-pyrazol-1-yl) propanonitrile, janus kinase inhibitor, and use of a composition that understands it for the treatment of dry eye.
KR20110071108A (en) 2008-10-06 2011-06-28 이데라 파마슈티칼즈, 인코포레이티드 Use of inhibitors of toll-like receptors in the prevention and treatment of hypercholesterolemia and hyperlipidemia and diseases related thereto
EP2411019A2 (en) 2009-03-27 2012-02-01 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 1 (STAT1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
CA3027255C (en) 2009-07-10 2022-06-21 The General Hospital Corporation Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
WO2011133474A2 (en) 2010-04-18 2011-10-27 Beth Israel Deaconess Medical Center Methods of predicting predisposition to or risk of kidney disease
US9023355B2 (en) 2010-04-13 2015-05-05 Beth Israel Deaconess Medical Center, Inc. Compositions and methods for treating renal disease
EP2611448A1 (en) 2010-09-01 2013-07-10 Ambit Biosciences Corporation 7-cyclylquinazoline derivatives and methods of use thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2441498A (en) 1943-07-15 1948-05-11 Astra Apotekarnes Kem Fab Alkyl glycinanilides
US2799679A (en) 1955-04-28 1957-07-16 Bofors Ab Process of preparing amides of heterocyclic carboxylic acids
US2955111A (en) 1957-01-28 1960-10-04 Bofors Ab Synthesis of n-alkyl-piperidine and n-alkyl-pyrrolidine-alpha-carboxylic acid amides
US3160662A (en) 1957-06-26 1964-12-08 Astra Apotekarnes Kem Fab Lower alkylaminoacyl amide anesthetics
DE2162744A1 (en) 1970-12-22 1972-07-06 Astra Pharmaceutical Products Inc., Worcester, Mass. (V.St.A.) 2-alkyl-2-alkylamino-2,6-acetoxylidides, processes for their preparation and their use
DE2235745A1 (en) 1971-07-28 1973-02-22 Astra Pharma Prod ANTIARRHYTHMIC COMPOUND, PROCESS FOR THEIR PRODUCTION AND THEIR USE
WO1985000599A1 (en) 1983-08-01 1985-02-14 A/S Apothekernes Laboratorium For Specialpraeparat L-N-n-PROPYLPIPECOLIC ACID-2,6-XYLIDIDE AND METHOD FOR PREPARING THE SAME
US6709406B2 (en) 1996-01-02 2004-03-23 Cefar Matcher Ab Measurement of pain
US6432937B1 (en) 1996-12-20 2002-08-13 Astra Aktiebolag Treatment for joint inflammation
US6623040B1 (en) 1997-09-03 2003-09-23 Recot, Inc. Method for determining forced choice consumer preferences by hedonic testing
US6766319B1 (en) 2000-10-31 2004-07-20 Robert J. Might Method and apparatus for gathering and evaluating information

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
G. HERMANSON: "Bioconjugate Techniques", 1996, ACADEMIC PRESS, INC.
HILL ET AL., J BIOL CHEM, vol. 282, 2007, pages 7145 - 7153
MACPHERSON ET AL., NATURE, vol. 445, 2007, pages 541 - 545
O'DELL ET AL., BIOORG MED CHEM, vol. 15, 2007, pages 6164 - 6149
P.J. KOCIENSKI: "Protecting Groups", 1994, GEORG THIEME VERLAG
SEXTON ET AL., FASEB J, vol. 21, 2007, pages 2695 - 2703
T.W..GREEN; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
TAYLOR-CLARK ET AL., MOL PHARMACOL, 2007

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10179116B2 (en) 2006-11-20 2019-01-15 President And Fellows Of Harvard College Methods, compositions, and kits for treating pain and pruritis
JP2016121169A (en) * 2007-02-06 2016-07-07 ニューロクエスト インク Compositions for treating neurolopathic pain
EP2142182A4 (en) * 2007-02-06 2010-03-03 Origin Biomed Inc Composition comprising terpene compounds and methods for inhibiting nerve transmission
JP2010518030A (en) * 2007-02-06 2010-05-27 オリジン バイオメッド インク Composition containing terpene compound and method for inhibiting neurotransmission
EP2142182A1 (en) * 2007-02-06 2010-01-13 Origin BioMed Inc. Composition comprising terpene compounds and methods for inhibiting nerve transmission
US9999601B2 (en) 2007-02-06 2018-06-19 Neuroquest Inc. Composition and method for inhibition of nerve transmission
WO2009114139A3 (en) * 2008-03-11 2009-12-23 President And Fellows Of Harvard College Methods, compositions, and kits for treating pain and pruritis
US8609733B2 (en) 2008-05-19 2013-12-17 Massachusetts Institute Of Technology Sensory-specific local anesthesia and prolonged duration local anesthesia
WO2009143175A3 (en) * 2008-05-19 2010-10-28 Children's Medical Center Corporation Sensory-specific local anesthesia and prolonged duration local anesthesia
WO2009143175A2 (en) * 2008-05-19 2009-11-26 Children's Medical Center Corporation Sensory-specific local anesthesia and prolonged duration local anesthesia
US9415023B2 (en) * 2008-08-13 2016-08-16 Neuroquest Inc. Compositions comprising terpene compounds for treating negative sensory phenomena
US20110144191A1 (en) * 2008-08-13 2011-06-16 Mclellan Alexander Compositions comprising terpene compounds for treating negative sensory phenomena
KR101188945B1 (en) 2009-01-29 2012-10-08 서울대학교산학협력단 Composition containing sulforaphane and its analog for treating pain
US8952152B2 (en) 2009-03-24 2015-02-10 Proteus S.A. Methods for purifying phycotoxins, pharmaceutical compositions containing purified phycotoxins, and methods of use thereof
US8957207B2 (en) 2009-03-24 2015-02-17 Proteus S.A. Methods for producing phycotoxins
US9249150B2 (en) 2009-03-24 2016-02-02 Proteus S.A. Methods for purifying phycotoxins, pharmaceutical compositions containing purified phycotoxins, and methods of use thereof
JP2012532887A (en) * 2009-07-10 2012-12-20 プレジデント アンド フェロウズ オブ ハーバード カレッジ Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
US10729664B2 (en) 2009-07-10 2020-08-04 President And Fellows Of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
EP3485881B1 (en) * 2009-07-10 2024-03-13 President and Fellows of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
EP2451944A4 (en) * 2009-07-10 2012-11-28 Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
CN102725401A (en) * 2009-07-10 2012-10-10 哈佛大学校长及研究员协会 Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
JP2017178948A (en) * 2009-07-10 2017-10-05 プレジデント アンド フェローズ オブ ハーバード カレッジ Permanently Charged Sodium and Calcium Channel Blockers as Anti-Inflammatory Agents
AU2010271269B2 (en) * 2009-07-10 2016-09-08 Childrens' Medical Center Corporation Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
EP2451944A1 (en) * 2009-07-10 2012-05-16 President and Fellows of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
WO2011006073A1 (en) * 2009-07-10 2011-01-13 President And Fellows Of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
EP2995303A1 (en) * 2009-07-10 2016-03-16 President and Fellows of Harvard College Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
US8865741B2 (en) 2011-02-18 2014-10-21 Asana Biosciences, Llc Aminoindane compounds and use thereof in treating pain
WO2013063127A1 (en) * 2011-10-24 2013-05-02 Endo Pharmaceuticals Inc. Cyclohexylamines
US9180115B2 (en) 2011-10-24 2015-11-10 Asana Biosciences, Llc Cyclohexylamines
US8685418B2 (en) 2011-10-24 2014-04-01 Endo Pharmaceuticals Inc. Cyclohexylamines
US20130150455A1 (en) * 2011-12-07 2013-06-13 Snu R&Db Foundation Method for treating mechanical allodynia comprising administration of eugenol
KR101243430B1 (en) * 2012-04-23 2013-03-13 서울대학교산학협력단 Composition of health-promoting food containing sulforaphane and its analog for relieving pain
US9044482B2 (en) 2012-08-15 2015-06-02 Asana Biosciences, Llc Use of aminoindane compounds in treating overactive bladder and interstitial cystitis
US9375423B2 (en) 2012-08-15 2016-06-28 Asana Biosciences, Llc Use of aminoindane compounds in treating overactive bladder and interstitial cystitis
US10881647B2 (en) 2013-03-15 2021-01-05 The Children's Medical Center Corporation Neosaxitoxin combination formulations for prolonged local anesthesia
US8975268B2 (en) 2013-03-15 2015-03-10 The Children's Medical Center Corporation Neosaxitoxin combination formulations for prolonged local anesthesia
US8975281B2 (en) 2013-03-15 2015-03-10 The Children's Medical Center Corporation Neosaxitoxin combination formulations for prolonged local anesthesia
US10314833B2 (en) 2013-03-15 2019-06-11 The Children's Medical Center Corporation Neosaxitoxin combination formulations for prolonged local anesthesia
US11021443B2 (en) 2015-08-03 2021-06-01 President And Fellows Of Harvard College Charged ion channel blockers and methods for use
US11603355B2 (en) 2019-03-11 2023-03-14 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10828287B2 (en) 2019-03-11 2020-11-10 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10927096B2 (en) 2019-03-11 2021-02-23 Nocion Therapeutics, Inc. Ester substituted ion channel blockers and methods for use
US10934263B2 (en) 2019-03-11 2021-03-02 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10780083B1 (en) 2019-03-11 2020-09-22 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10968179B2 (en) 2019-03-11 2021-04-06 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10786485B1 (en) 2019-03-11 2020-09-29 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US11643404B2 (en) 2019-03-11 2023-05-09 Nocion Therapeutics, Inc. Ester substituted ion channel blockers and methods for use
US11377422B2 (en) 2019-03-11 2022-07-05 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US11512058B2 (en) 2019-03-11 2022-11-29 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10933055B1 (en) 2019-11-06 2021-03-02 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US11696912B2 (en) 2019-11-06 2023-07-11 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US10842798B1 (en) 2019-11-06 2020-11-24 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
US11332446B2 (en) 2020-03-11 2022-05-17 Nocion Therapeutics, Inc. Charged ion channel blockers and methods for use
EP4428132A1 (en) * 2023-03-08 2024-09-11 Johann-Wolfgang-Goethe-Universität Frankfurt am Main Slack-activating compounds and their medical use
WO2024184543A1 (en) * 2023-03-08 2024-09-12 Johann Wolfgang Goethe-Universität Frankfurt am Main Slack-activating compounds and their medical use

Also Published As

Publication number Publication date
JP2018039788A (en) 2018-03-15
CA3002443A1 (en) 2008-05-29
JP2016185955A (en) 2016-10-27
JP5926301B2 (en) 2016-05-25
US20100099772A1 (en) 2010-04-22
EP2446903A3 (en) 2012-11-14
US9603817B2 (en) 2017-03-28
WO2008063603A3 (en) 2008-12-11
ES2402789T3 (en) 2013-05-08
CN103933572A (en) 2014-07-23
US20210186906A1 (en) 2021-06-24
JP2010510227A (en) 2010-04-02
EP2101819A2 (en) 2009-09-23
JP2014111626A (en) 2014-06-19
EP2446903B1 (en) 2019-10-09
US10179116B2 (en) 2019-01-15
DK2101819T3 (en) 2013-04-29
AU2007322033B2 (en) 2013-07-11
CA2668652A1 (en) 2008-05-29
US20170319517A1 (en) 2017-11-09
US20150087714A1 (en) 2015-03-26
EP2425858A3 (en) 2012-03-28
EP2101819B1 (en) 2013-01-09
AU2007322033A2 (en) 2009-07-16
AU2007322033A1 (en) 2008-05-29
CA2668652C (en) 2018-06-12
EP2425858A2 (en) 2012-03-07
KR20140097446A (en) 2014-08-06
KR20090082507A (en) 2009-07-30
EP2446903A2 (en) 2012-05-02
CN103933572B (en) 2017-10-13

Similar Documents

Publication Publication Date Title
US20210186906A1 (en) Methods, compositions, and kits for treating pain and pruritis
US20110086818A1 (en) Methods, compositions, and kits for treating pain and pruritus
JP6845280B2 (en) Permanently charged sodium and calcium channel blockers as anti-inflammatory agents
AU2016301282B2 (en) Charged ion channel blockers and methods for use
CN101610789A (en) Method, compositions and the test kit of treatment pain and pruritus

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780050131.9

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07862114

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2007322033

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2668652

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2009537235

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007322033

Country of ref document: AU

Date of ref document: 20071119

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020097012908

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2007862114

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12515429

Country of ref document: US