EP1937242A2 - Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance - Google Patents

Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance

Info

Publication number
EP1937242A2
EP1937242A2 EP06826134A EP06826134A EP1937242A2 EP 1937242 A2 EP1937242 A2 EP 1937242A2 EP 06826134 A EP06826134 A EP 06826134A EP 06826134 A EP06826134 A EP 06826134A EP 1937242 A2 EP1937242 A2 EP 1937242A2
Authority
EP
European Patent Office
Prior art keywords
bumetanide
furosemide
piretanide
thioester
ester
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP06826134A
Other languages
German (de)
English (en)
Other versions
EP1937242A4 (fr
Inventor
Daryl W. Hochman
John J. Partridge
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
NeuroTherapeutics Pharma Inc
Original Assignee
NeuroTherapeutics Pharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US11/251,724 external-priority patent/US8008283B2/en
Priority claimed from US11/549,274 external-priority patent/US20070149526A1/en
Application filed by NeuroTherapeutics Pharma Inc filed Critical NeuroTherapeutics Pharma Inc
Publication of EP1937242A2 publication Critical patent/EP1937242A2/fr
Publication of EP1937242A4 publication Critical patent/EP1937242A4/fr
Withdrawn legal-status Critical Current

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
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants

Definitions

  • the present invention relates to methods and compositions for treating selected conditions of the central and peripheral nervous systems employing non-synaptic mechanisms. More specifically, the present invention relates to methods and compositions for treating neuropsychiatric and addictive disorders by administering agents that modulate expression and/or activity of sodium-potassium-chloride co- transporters.
  • Nociceptive pain occurs in response to the activation of a specific subset of peripheral sensory neurons, the nociceptors. It is generally acute (with the exception of arthritic pain), self-limiting and serves a protective biological function by acting as a warning of on-going tissue damage. It is typically well localized and often has an aching or throbbing quality. Examples of nociceptive pain include post-operative pain, sprains, bone fractures, burns, bumps, bruises, inflammation (from an infection or arthritic disorder), obstructions and myofascial pain. Nociceptive pain can usually be treated with opioids and non-steroidal anti-inflammatory drugs (NSAIDS).
  • NSAIDS non-steroidal anti-inflammatory drugs
  • 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, diabetes, infection with herpes zoster (shingles), HIV/AIDS peripheral neuropathies, 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.
  • neuropathic pain 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. Neuropathic pain is difficult to treat. Analgesic drugs that are effective against normal pain (e.g., opioid narcotics and non-steroidal anti-inflammatory drugs) are rarely effective against neuropathic pain. Similarly, drugs that have activity in neuropathic pain are not usually effective against nociceptive pain.
  • Treatment agents typically employed in the management of neuropathic pain include tricylic antidepressants (for example, amitriptyline, imipramine, desimipramine and clomipramine), systemic local anesthetics, and anti-convulsants (such as phenytoin, carbamazepine, valproic acid, clonazepam and gabapentin).
  • tricylic antidepressants for example, amitriptyline, imipramine, desimipramine and clomipramine
  • systemic local anesthetics such as phenytoin, carbamazepine, valproic acid, clonazepam and gabapentin.
  • anti-convulsants originally developed for the treatment of epilepsy and other seizure disorders have found application in the treatment of non-epileptic conditions, including neuropathic pain, mood disorders (such as bipolar affective disorder), and schizophrenia (for a review of the use of anti-epileptic drugs in the treatment of non-epileptic conditions, see Rogawski and Loscher, Nat. Medicine, 10:685- 692, 2004).
  • neuropathic pain and affective disorders have a common pathophysiological mechanism (Rogawski & Loscher, ibid; Ruscheweyh & Sandkuhler, Pain 105:327-338, 2003), namely a pathological increase in neuronal excitability, with a corresponding inappropriately high frequency of spontaneous firing of neurons.
  • antiepileptic drugs are effective in treating neuropathic pain, and furthermore such antiepileptic drugs are only effective in certain subsets of patients with neuropathic pain (McCleane, Expert. Opin. Pharmacother. 5:1299-1312, 2004).
  • Epilepsy is characterized by abnormal discharges of cerebral neurons and is typically manifested as various types of seizures.
  • Epileptiform activity is identified with spontaneously occurring synchronized discharges of neuronal populations that can be measured using electrophysiological techniques. This synchronized activity, which distinguishes epileptiform from non-epileptiform activity, is referred to as "hypersynchronization" because it describes the state in which individual neurons become increasingly likely to discharge in a time-locked manner with one another.
  • Hypersynchronized activity is typically induced in experimental models of epilepsy by either increasing excitatory or decreasing inhibitory synaptic currents, and it was therefore assumed that hyperexcitability per se was the defining feature involved in the generation and maintenance of epileptiform activity.
  • neuropathic pain was believed to involve conversion of neurons involved in pain transmission from a state of normal sensitivity to one of hypersensitivity (Costigan & Woolf, JnI. Pain 1:35-44, 2000).
  • the focus on developing treatments for both epilepsy and neuropathic pain has thus been on suppressing neuronal hyperexcitability by either: (a) suppressing action potential generation; (b) increasing inhibitory synaptic transmission; or (c) decreasing excitatory synaptic transmission.
  • CCCs cation-chloride co-transporters
  • NCCs Na + -Cl " co-transporters
  • KCCs K + -Cl " co-transporters
  • NKCCs Na + -K + -2C1 ' co-transporters
  • NKCCl is found in a wide variety of secretory epithelia and non-epithelial cells, whereas NKCC2 is principally expressed in the kidney.
  • NKCCl structure, function and regulation see, Haas and Forbush, Annu. Rev. Physiol. 62:515-534, 2000. Randall et al. have identified two splice variants of the Slcl2a2 gene that encodes NKCCl, referred to as NKCCIa and NKCCIb (Am. J. Physiol. 273 (Cell Physiol. 42):C1267-1277, 1997).
  • the NKCCl a gene has 27 exons, while the splice variant NKCCIb lacks exon 21.
  • the NKCCIb splice variant is expressed primarily in the brain. NKCCIb is believed to be more than 10% more active than NKCCIa, although it is proportionally present in a much smaller amount in the brain than is NKCCIa. It has been suggested that differential splicing of the NKCCl transcript may play a regulatory role in human tissues (Vibat et al. Anal. Biochem. 298:218-230, 2001). Na-K-Cl co- transport in all cell and tissues is inhibited by loop diuretics, including furosemide, bumetanide and benzmetanide.
  • Na-K-2C1 co-transporter knock-out mice have been shown to have impaired nociception phenotypes as well as abnormal gait and locomotion (Sung et al. JnI. Neurosci. 20:7531-7538, 2000). Delpire and Mount have suggested that NKCCl may be involved in pain perception (Ann. Rev. Physiol. 64:803-843, 2002). Laird et al. recently described studies demonstrating reduced stroking hyperalgesia in NKCCl knock-out mice compared to wild-type and heterozygous mice (Neurosci. Letts. 361:200-203, 2004). However, in this acute pain model no difference in punctuate hyperalgesia was observed between the three groups of mice.
  • Morales- Aza et al. have suggested that, in arthritis, altered expression of NKCCl and the K-Cl co-transporter KCC2 may contribute to the control of spinal cord excitability and may thus represent therapeutic targets for the treatment of inflammatory pain (Neurobiol. Dis. 17:62-69, 2004).
  • Granados-Soto et al. have described studies in rats in which formalin-induced nociception was reduced by administration of the NKCC inhibitors bumetanide, furosemide or piretanide (Pain 114:231-238, 2005). While the formalin-induced acute pain model is extensively used, it is believed to have little relevance to chronic pain conditions (Walker et al. MoI. Med. Today 5:319-321, 1999).
  • neuropathic pain As with epilepsy, the focus of pharmacological intervention in neuropathic pain has been on reducing neuronal hyperexcitability.
  • Most agents currently used to treat neuropathic pain target synaptic activity in excitatory pathways by, for example, modulating the release or activity of excitatory neurotransmitters, potentiating inhibitory pathways, blocking ion channels involved in impulse generation, and/or acting as membrane stabilizers.
  • Conventional agents and therapeutic approaches for the treatment of neuropathic pain and neuropsychiatric disorders thus reduce neuronal excitability and inhibit synaptic firing.
  • One serious drawback of these therapies is that they are nonselective and exert their actions on both normal and abnormal neuronal populations.
  • Addictive disorders such as eating disorders (including obesity), addiction to narcotics, sexual addiction, alcoholism and smoking, are a major public health problem that impacts society on multiple levels. It has been estimated that substance abuse costs the US more than $484 billion per year. Current strategies for the treatment of addictive disorders include psychological counseling and support, use of therapeutic agents or a combination of both.
  • a variety of agents known to affect the central nervous system have been used in various contexts to treat a number of indications related directly or indirectly to addictive behaviors. For example, the combination of phentermine and fenfluramine was used for many years to exert an anorectic effect to treat obesity.
  • Topiramate is an anti-convulsant that was originally developed as an anti-diabetic agent and is approved for use in the treatment of epileptic seizures in adults and children. It is a GABA-receptor agonist and has sodium channel-blocking activity. Studies on the effectiveness of topiramate in treating alcohol dependence demonstrated that oral administration of topiramate led to a decrease in heavy drinking days and alcohol craving, with a concurrent increase in abstinent days and improved liver functions (Johnson et al. Lancet, 361:1677-85, 2003). Topiramate has also been shown to be effective in treating binge eating disorder associated with obesity (McElroy et al. Am. J. Psychiatry 160:255- 261, 2003; McElroy et al. J.
  • topiramate may be an effective treatment for obesity.
  • the treatment compositions and methods of the present invention are useful for treating conditions including addictive disorders and neuropsychiatric disorders, such as bipolar disorders, anxiety disorders (including panic disorder, social anxiety disorder, obsessive compulsive disorder, posttraumatic stress disorder, generalized anxiety disorder and specific phobia (American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders, 4 th edition - Text Revision, 2000)), depression and schizophrenia, that are characterized by neuronal hypersynchrony.
  • addictive disorders and neuropsychiatric disorders such as bipolar disorders, anxiety disorders (including panic disorder, social anxiety disorder, obsessive compulsive disorder, posttraumatic stress disorder, generalized anxiety disorder and specific phobia (American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders, 4 th edition - Text Revision, 2000)), depression and schizophrenia, that are characterized by neuronal hypersynchrony.
  • the inventive compositions and methods may be employed to reduce neuronal hypersynchrony associated with addictive and/or neuropsychiatric disorders without suppressing neuronal excit
  • Addictive disorders include eating disorders (including obesity and binge eating), alcoholism, sexual addiction, addiction to narcotics and smoking, addiction to exercise and gambling.
  • eating disorders including obesity and binge eating
  • alcoholism a substance that influences the rate of compulsion
  • sexual addiction a substance that influences the rate of compulsion
  • addiction to exercise and gambling a disorder that influences the rate of compulsion
  • addictive disorder is defined as a disorder characterized by an uncontrollable compulsion to repeat a behavior regardless of its consequences.
  • inventive compositions and methods may be employed to reduce neuronal hypersynchrony associated with such conditions without suppressing neuronal excitability, thereby avoiding unwanted side effects.
  • the methods and compositions disclosed herein generally involve non-synaptic mechanisms and modulate, generally reduce, the synchronization of neuronal population activity.
  • the synchronization of neuronal population activity is modulated by manipulating anionic concentrations and gradients in the central and/or peripheral nervous systems.
  • the inventive compositions are capable of reducing the effective amount, inactivating, and/or inhibiting the activity of a Na + -K + -2C1 " (NKCC) co-transporter.
  • NKCC Na + -K + -2C1 "
  • Especially preferred treatment agents of the present invention exhibit a high degree of NKCC co-transporter antagonist activity in cells of the central and/or peripheral nervous system, e.g., glial cells, Schwann cells and/or neuronal cell populations, and exhibit a lesser degree of activity in renal cell populations.
  • the inventive compositions are capable of reducing the effective amount, inactivating, and/or inhibiting the activity of the co-transporter NKCCl.
  • NKCCl antagonists are especially preferred treatment agents for use in the inventive methods.
  • NKCC co-transporter antagonists that may be usefully employed in the inventive treatment compositions include, but are not limited to, CNS-targeted NKCC co- transporter antagonists such as furosemide, bumetanide, ethacrynic acid, torsemide, azosemide, muzolimine, piretanide, tripamide and the like, as well as thiazide and thiazide-like diuretics, such as bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methylclothiazide, polythiazide, trichlor- methiazide, chlorthalidone, indapamide, metolazone and quinethazone, together with analogs and functional derivatives of such components.
  • CNS-targeted NKCC co- transporter antagonists such as furosemide, bumetanide, ethacrynic acid
  • Analogs of CNS-targeted NKCC co-transporter antagonists such as furosemide, bumetanide, piretanide, azosemide and torsemide that may be usefully employed in the inventive compositions and methods include those provided below as Formulas I- VIII.
  • Ri is not present, H, O or S;
  • R 2 is not present, H or when R 1 is O or S, R 2 is selected from the group consisting of hydrogen, alkyl, aralkyl, aryl, alkylaminodialkyl, alkylcarbonylaminodialkyl, alkyloxycarbonylalkyl, alkylcarbonyloxyalkyl, alkylaldehyde, alkylketoalkyl, alkylamide, alkarylamide, arylamide, an alkylammonium group, alkylcarboxylic acid, alkylheteroaryl, alkylhydroxy, a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl, unsubstituted or substituted
  • R 3 is selected from the group consisting of aryl, halo, hydroxy, alkoxy, and aryloxy, unsubstituted or substituted;
  • R 4 and R 5 are each independently selected from the group consisting of hydrogen, alkylaminodialkyl, carbonylalkyl, carbonylalkaryl, carbonylaryl, and salts thereof such as sodium, potassium, calcium, ammonium, trialkylarylammonium and tetraalkylammonium salts, with the following provisos in some embodiments:
  • R 3 of formula I is not phenyloxy when Ri is O and R 2 , R 4 and R 5 are H, more specifically, in some embodiments, the compound of formula I is not bumetanide;
  • R 3 of formula III is not Cl, when Ri is O and R 2 , R 4 and R 5 are H, more specifically, in some embodiments, the compound of formula III is not furosemide;
  • R 2 of formula III is not methyl when R 1 is O, R 3 is Cl, and R 4 and R 5 are H, more specifically, in some embodiments, the compound of formula III is not furosemide methyl ester;
  • R 3 , R 4 and R 5 are defined above;
  • R 6 is selected from the group consisting of alkyloxycarbonylalkyl, alkylaminocarbonylalkyl, alkylaminodialkyl, alkylhydroxy, a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl, unsubstituted or substituted, with the proviso that, in some embodiments, R 3 of formula VII is not Cl, when R 4 , R 5 and R 6 are H, more specifically, in some embodiments, the compound of formula VII is not azosemide.
  • R 7 is not present or selected from the group consisting of hydrogen, alkyloxycarbonylalkyl, alkylaminocarbonylalkyl, alkylammodialkyl, alkylhydroxy, a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl, unsubstituted or substituted; and
  • PEG polyethylene glycol
  • PEG ester polyethylene glycol ester
  • PEG ether polyethylene glycol ether
  • X " is a halide such as bromide, chloride, fluoride, iodide or an anionic moiety such as mesylate or tosylate; alternatively, X " is not present and the compound forms an "inner” or zwitterionic salt (where R 7 is H), with the proviso that, in some embodiments, R 7 is always present and X " is not present. More specifically, in some embodiments, the compound of formula VIII is not torsemide.
  • Embodiments of the present invention provide prodrugs capable of passage across the blood-brain barrier comprising a compound of formula I, II, III, IV, V, VI, VII and/or VIII, or a pharmaceutically acceptable salt, solvate, tautomer or hydrate thereof.
  • the compound of the prodrug is provided in an amount effective for regulating a CNS disorder.
  • the CNS disorder is epilepsy, anxiety, neuropathic pain, neural function, drug addiction/physical dependence and/or migraines.
  • Embodiments of the present invention provide a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I, II, III, FV, V, VI, VII and/or VIII, or a pharmaceutically acceptable salt, solvate, tautomer, hydrate or combination thereof and a pharmaceutically acceptable carrier, excipient or diluent.
  • the compound of the pharmaceutical composition is present in an amount effective for regulating a CNS disorder.
  • the CNS disorder is an addictive disorder, a neuropsychiatric disorder or neuropathic pain.
  • Embodiments of the present invention provide uses of the compounds described herein for the preparation of a medicament for treating and/or preventing a CNS disorder selected from the group consisting of: addictive disorders, neuropsychiatric disorders and neuropathic pain.
  • the level of diuresis that occurs following administration of an effective amount of an analog provided below as Formula I- VIII is less than 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20% or 10% of that which occurs following administration of an effective amount of the parent molecule from which the analog is derived.
  • the analog may be less diuretic than the parent molecule when administered at the same mg/kg dose.
  • the analog may be more potent than the parent molecule from which it is derived, so that a smaller dose of the analog is required for effective relief of symptoms, thereby eliciting less of a diuretic effect.
  • the analog may have a longer duration of effect in treating disorders than the parent molecule, so that the analog may be administered less frequently than the parent molecule, thus leading to a lower total diuretic effect within any given period of time.
  • treatment agents that may be usefully employed in the inventive compositions and methods include, but are not limited to: antibodies, or antigen-binding fragments thereof, that specifically bind to NKCCl; soluble NKCCl ligands; small molecule inhibitors of NKCCl; anti-sense oligonucleotides to NKCCl; NKCCl -specific small interfering RNA molecules (siRNA or RNAi); and engineered soluble NKCCl molecules.
  • such antibodies, or antigen-binding fragments thereof, and small molecule inhibitors of NKCCl specifically bind to the domains of NKCCl involved in bumetanide binding, as described, for example, in Haas and Forbush II, Annu. Rev. Physiol. 62:515-534, 2000.
  • the polypeptide sequence for human NKCCl is provided in SEQ ID NO: 1, with the corresponding cDNA sequence being provided in SEQ ID NO: 2.
  • the inventive treatment agents of the present invention employ "non- synaptic" mechanisms, little or no suppression of neuronal excitability occurs. More specifically, the inventive treatment agents cause little (less than a 1% change compared to pre-administration levels) or no suppression of action potential generation or excitatory synaptic transmission. In fact, a slight increase in neuronal excitability may occur upon administration of certain of the inventive treatment agents. This is in marked contrast to conventional anti-epileptic drugs currently used in the treatment of neuropathic pain, which do suppress neuronal excitability.
  • the methods and treatment agents of the present invention affect the synchronization, or relative synchrony, of neuronal population activity. Preferred methods and treatment agents modulate the extracellular anionic chloride concentration and/or the gradients in the central or peripheral nervous system to reduce neuronal synchronization, or relative synchrony, without substantially affecting neuronal excitability.
  • the present invention relates to methods and agents for relieving neuropathic pain, or the abnormal perception of pain, by affecting or modulating spontaneous hypersynchronized bursts of neuronal activity and the propagation of action potentials or conduction of impulses in certain cells and nerve fibers of the peripheral nervous system, for example, primary sensory afferent fibers, pain fibers, dorsal horn neurons, and supraspinal sensory and pain pathways.
  • the present invention related to methods and agents for treating, ameliorating and/or preventing, neuropsychiatric disorders and addictive disorders.
  • the present invention relates to methods and agents for treating or preventing neuropsychiatric disorders, and addictive and/or compulsive disorders, by affecting or modulating spontaneous hypersynchronized bursts of neuronal activity and the propagation of action potentials or conduction of impulses in certain cells and nerve fibers of the peripheral nervous system, for example, primary sensory afferent fibers, pain fibers, dorsal horn neurons, and supraspinal sensory and pain pathways.
  • inventive treatment agents may be employed in combination with other, known, treatment agents, such as those presently used in the treatment of neuropsychiatric disorders.
  • treatment agents such as those presently used in the treatment of neuropsychiatric disorders.
  • One of skill in the art will appreciate that the combination of a treatment agent of the present invention with another, known, treatment agent may involve both synaptic and non-synaptic mechanisms.
  • Treatment compositions and methods of the present invention may be used therapeutically and episodically following the onset of symptoms or prophylactically, prior to the onset of specific symptoms.
  • the inventive methods for treatment of addictive disorders involve the administration of a treatment agent comprising a diuretic (for example, a loop diuretic such as furosemide, torasemide or bumetanide, or a thiazide or thiazide-like diuretic) in combination with one or more anti-diuretic components, in order to counteract unwanted diuretic effects of the primary treatment agent.
  • a diuretic for example, a loop diuretic such as furosemide, torasemide or bumetanide, or a thiazide or thiazide-like diuretic
  • Negative side effects that can be avoided by such methods include loss of body water, and depletion of electrolytes (such as potassium, magnesium, calcium and thiamine) and B vitamins.
  • Anti-diuretic components that may be usefully employed in such methods include, for example, antidiuretic hormones, such as vasopressin, which increases water reabsorption by the kidneys; and salts and electrolytes, which act to replenish ions lost due to diuresis.
  • the diuretic treatment agent and the anti-diuretic component are combined together in a composition formulated as a liquid beverage, food or food supplement.
  • the treatment agents employed in the inventive methods are capable of crossing the blood brain barrier, and/or are administered using delivery systems that facilitate delivery of the agents to the central nervous system.
  • various blood brain barrier (BBB) permeability enhancers can be used, if desired, to transiently and reversibly increase the permeability of the blood brain barrier to a treatment agent.
  • Such BBB permeability enhancers may include leukotrienes, bradykinin agonists, histamine, tight junction disrupters (e.g., zonulin), hyperosmotic solutions (e.g., mannitol), cytoskeletal contracting agents, short chain alkylglycerols (e.g., 1-0- pentylglycerol), and others which are currently known in the art.
  • FIGS. IA, IAl, IB, IBl, 1C, ICl and ID show the effect of furosemide on stimulation evoked after discharge activity in rat hippocampal slices.
  • Figs. 2A - 2R show furosemide blockade of spontaneous epileptiform burst discharges across a spectrum of in vitro models.
  • Figs. 3 A - 3H show furosemide blockade of kainic acid-evoked electrical "status epilepticus" in urethane-anesthetized rats, with EKG recordings shown in the upper traces and cortical EEG recordings shown in the bottom traces.
  • Figs. 4 A and 4B show a schematic diagram of ion co-transport under conditions of reduced chloride concentration.
  • Fig. 5 shows that significantly less freezing was observed in animals treated with either bumetanide or furosemide than in animals receiving vehicle alone in a test of contextual fear conditioning in rats.
  • Fig. 6 shows baseline startle amplitudes in a fear potentiated startle test in rats
  • Fig. 7 shows the amplitude of response in rats on startle alone trials determined immediately following administration of either DMSO alone, bumetanide or furosemide.
  • Fig. 8 shows the difference score (startle alone - fear potentiated startle) on the test day in rats treated with either DMSO, bumetanide or furosemide
  • Fig. 9 shows the startle alone amplitude in rats one week after administration of either DMSO, bumetanide or furosemide.
  • Fig. 10 shows the difference score in rats one week after administration of either DMSO, bumetanide or furosemide.
  • Fig. 11 shows the percent difference score (startle alone - fear potentiated startle) on the test day in rats treated with one of the following bumetanide analogs: bumetanide 3-(dimethylaminoproply)ester; bumetanide benzyltrimethylammonium salt; bumetanide dibenzylamide; bumetanide cyanomethyl ester; bumetanide N,N-diethylglycolamide ester; bumetanide N,N-dimethylglycolamide ester; bumetanide morpholinodethyl ester; bumetanide pivaxetil ester; bumetanide methyl ester; bumetanide diethylamide; and bumetanide bezyl ester.
  • the vehicle was DMSO.
  • preferred treatment agents and methods of the present invention for use in treating neuropathic pain, addictive disorders and/or neuropsychiatric disorders, modulate or disrupt the synchrony of neuronal population activity in areas of heightened synchronization by reducing the activity of NKCC co-transporters.
  • movement of ions and modulation of ionic gradients by means of ion-dependent co-transporters, preferably cation-chloride dependent co-transporters is critical to regulation of neuronal synchronization.
  • Chloride co-transport function has long been thought to be directed primarily to movement of chloride out of cells.
  • the sodium independent transporter which has been shown to be neuronally localized, moves chloride ions out of neurons.
  • Blockade of this transporter leads to hyperexcitability, which is the short-term response to cation-chloride co- transporters such as furosemide.
  • the long-term response to furosemide demonstrates that the inward, sodium-dependent movement of chloride ions, mediated by the glial associated Na + -K + -2C1 " co-transporter NKCCl, plays an active role in blocking neuronal synchronization, without affecting excitability and stimulus-evoked cellular activity.
  • Haglund and Hochman have demonstrated that fuiOsemide is able to block epileptic activity in humans while not affecting normal brain activity (J. Neurophysiol. (Feb.
  • NKCCIb is more active than the NKCCIa variant.
  • a central or peripheral nervous system which expresses a few more percentage NKCCIb may thus be more prone to disorders such as neuropathic pain and epilepsy.
  • a treatment agent that is more specific for NKCCIb compared to NKCCIa may be more effective in the treatment of such disorders.
  • the inventive methods may be used for the treatment and/or prophylaxis of neuropathic pain having, for example, the following etiologies: alcohol abuse; diabetes; eosinophilia-myalgia syndrome; Guillain-Barre syndrome; exposure to heavy metals such as arsenic, lead, mercury, and thallium; HIV/AIDS; exposure to anti-HIV/AIDS drugs; malignant tumors; medications including amiodarone, aurothioglucose, cisplatinum, dapsone, stavudine, zalcitabine, didanosine, disulfiram, FK506, hydralazine, isoniazid, metronidazole, nitrofurantoin, paclitaxel, phenytoin and vincristine; monoclonal gammopathies; multiple sclerosis; post-stroke central pain, postherpetic neuralgia; trauma including carpal tunnel syndrome, cervical or lumbar radiculopathy, complex
  • Neuropsychiatry disorders that may be effectively treated using the inventive methods include, but are not limited to, bipolar disorders, anxiety disorders, panic disorders, depression, schizophrenia, obsessive-compulsive disorders and post-traumatic stress syndrome.
  • Anxiety disorders are classified into several subtypes: Panic Disorder, Social Anxiety Disorder, Obsessive Compulsive Disorder, Posttraumatic Stress Disorder, Generalized Anxiety Disorder and Specific Phobia (American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders, 4 th Editon, 2000).
  • inventive methods may be used for the treatment and/or prophylaxis of addictive and/or compulsive disorders such as: eating disorders, including obesity and binge eating; alcoholism; addiction to narcotics; and smoking.
  • addictive and/or compulsive disorders such as: eating disorders, including obesity and binge eating; alcoholism; addiction to narcotics; and smoking.
  • compositions that may be effectively employed in the inventive methods are capable of reducing the effective amount, inactivating, and/or inhibiting the activity of a Na + -K + -2C1 " (NKCC) co-transporter.
  • NKCC Na + -K + -2C1 "
  • compositions are capable of reducing the effective amount, inactivating, and/or inhibiting the activity of the co- transporter NKCCl.
  • the inventive compositions comprise at least one treatment agent selected from the group consisting of: antagonists of NKCCl (including but not limited to, small molecule inhibitors of NKCCl, antibodies, or antigen- binding fragments thereof, that specifically bind to NKCCl and soluble NKCCl ligands); anti- sense oligonucleotides to NKCCl; NKCCl -specific small interfering RNA molecules (siRNA or RNAi); and engineered soluble NKCCl molecules.
  • NKCCl including but not limited to, small molecule inhibitors of NKCCl, antibodies, or antigen- binding fragments thereof, that specifically bind to NKCCl and soluble NKCCl ligands
  • anti- sense oligonucleotides to NKCCl NKCCl -specific small interfering RNA molecules (siRNA or RNAi)
  • shRNA or RNAi small interfering RNA molecules
  • the treatment agent is selected from the group consisting of: CNS-targeted NKCC co-transporter antagonists such as furosemide, bumetanide, ethacrynic acid, torsemide, azosemide, muzolimine, piretanide, tripamide and the like; thiazide and thiazide-like diuretics, such as bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiazide, hydro-fluniethiazide, methylclothiazide, polythiazide, trichlormethiazide, chlorthalidone, indapamide, metolazone and quinethazone; and analogs and functional derivatives of such components.
  • CNS-targeted NKCC co-transporter antagonists such as furosemide, bumetanide, ethacrynic acid, torsemide, azosemide, muzolimine,
  • the methods of the present invention employ novel compounds.
  • any of the R groups as defined herein can be excluded or modified in order to exclude a known compound and/or provide a novel compound.
  • the methods disclosed herein employ compounds having the following structures:
  • R 1 is not present, H, O or S;
  • R 2 is not present, H or when R 1 is O or S, R 2 is selected from the group consisting of hydrogen, alkyl, aralkyl, aryl, alkylaminodialkyl, alkylcarbonylaminodialkyl, alkyloxycarbonylalkyl, alkylcarbonyloxyalkyl, alkylaldehyde, alkylketoalkyl, alkylamide, alkarylamide, arylamide, an alkylammonium group, alkylcarboxylic acid, alkylheteroaryl, alkylhydroxy, a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl, unsubstituted or substituted
  • R 4 and R 5 are each independently selected from the group consisting of hydrogen, alkylaminodialkyl, carbonylalkyl, carbonylalkaryl, carbonylaryl, and salts thereof such as sodium, potassium, calcium, ammonium, trialkylarylammonium and tetraalkylammonium salts, with the following provisos in some embodiments:
  • R 3 of formula I is not phenyloxy when Ri is O and R 2 , R 4 and R 5 are H, more specifically, in some embodiments, the compound of formula I is not bumetanide;
  • R 3 of formula III is not Cl, when R 1 is O and R 2 , R 4 and R 5 are H, more specifically, in some embodiments, the compound of formula III is not furosemide;
  • R 2 of formula III is not methyl when Ri is O, R 3 is Cl, and R 4 and R 5 are H, more specifically, in some embodiments, the compound of formula III is not furosemide methyl ester;
  • the compound of formula I can be bumetanide, bumetanide aldehyde, bumetanide methyl ester, bumetanide cyanomethyl ester, bumetanide ethyl ester, bumetanide isoamyl ester, bumetanide octyl ester, bumetanide benzyl ester, bumetanide dibenzylamide, bumetanide diethylamide, bumetanide morpholinoethyl ester, bumetanide 3-(dimethylaminopropyl) ester, bumetanide N,N-diethylglycolamido ester, bumetanide N,N-dimethylglycolamido ester, bumetanide pivaxetil ester, bumetanide propaxetil ester, bumetanide methoxy(polyethyleneoxy) n- i -ethyl ester, bumetanide benzyltrimethylammonium
  • the compound of formula III can be furosemide, furosemide aldehyde, furosemide methyl ester, furosemide cyanomethyl ester, furosemide ethyl ester, furosemide isoamyl ester, furosemide octyl ester, furosemide benzyl ester, furosemide morpholinoethyl ester, furosemide 3- (dimethylaminopropyl) ester, furosemide N,N-diethylglycolamido ester, furosemide N 5 N- dimethylglycolamido ester, furosemide pivaxetil ester, furosemide propaxetil ester, furosemide methoxy(polyethyleneoxy) n- i -ethyl ester, furosemide benzyltrimethylammonium acid salt and furosemide cetyltrimethylammonium acid salt.
  • the compound is not furosemide.
  • the compound of formula V can be piretanide, piretanide aldehyde piretanide methyl ester, piretanide cyanomethyl ester, piretanide ethyl ester, piretanide isoamyl ester, piretanide octyl ester, piretanide benzyl ester, piretanide dibenzylamide, piretanide diethylamide, piretanide morpholinoethyl ester, piretanide 3-(dimethylaminopropyl) ester, piretanide N,N- diethylglycolamide ester, piretanide dimethylglycolamide ester, piretanide pivaxetil ester, piretanide propaxetil ester, piretanide methoxy(polyethyleneoxy)
  • the methods disclosed herein employ compounds having the following structure:
  • R 3 , R 4 and R 5 are defined above;
  • R 6 is selected from the group consisting of alkyloxycarbonylalkyl, alkylaminocarbonylalkyl, alkylaminodialkyl, alkylhydroxy, a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl, unsubstituted or substituted, with the proviso that, in some embodiments, R 3 of formula VII is not Cl, when R 4 , R 5 and R 6 are H, more specifically, in some embodiments, the compound of formula VII is not azosemide.
  • the compounds of formula VII can be tetrazolyl- substituted azosemides (such as methoxymethyl tetrazolyl-substituted azosemides, methylthiomethyl tetrazolyl-substituted azosemides and N-mPEG350-tetrazolyl- substituted azosemides), azosemide benzyltrimethylammonium salt and/or azosemide cetyltrimethylammonium salt.
  • tetrazolyl- substituted azosemides such as methoxymethyl tetrazolyl-substituted azosemides, methylthiomethyl tetrazolyl-substituted azosemides and N-mPEG350-tetrazolyl- substituted azosemides
  • azosemide benzyltrimethylammonium salt and/or azosemide cetyl
  • R 7 is not present or selected from the group consisting of hydrogen, alkyloxycarbonylalkyl, alkylaminocarbonylalkyl, alkylaminodialkyl, alkylhydroxy, a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl, unsubstituted or substituted; and
  • a biocompatible polymer such as alkyloxy(polyalkyloxy)alkylhydroxyl, a polyethylene glycol (PEG), a polyethylene glycol ester (PEG ester) and a polyethylene glycol ether (PEG ether), methyloxyalkyl, methyloxyalkaryl, methylthioalkyl and methylthioalkaryl
  • X " is a halide such as bromide, chloride, fluoride, iodide or an anionic moiety such as mesylate or tosylate; alternatively, X " is not present and the compound forms an “inner” or zwitterionic salt (where R 7 is H), with the proviso that, in some embodiments,
  • R 7 is always present and X " is not present. More specifically, in some embodiments, the compound of formula VIII is not torsemide.
  • the compounds of formula VIII can be pyridine-substituted torsemide quaternary ammonium salts or the corresponding inner salts (zwitterions). Examples include, but are not limited to, methoxymethyl pyridinium torsemide salts, methylthiomethyl pyridinium torsemide salts and N-mPEG350-pyridinium torsemide salts.
  • Embodiments of the present invention further provide intermediate compounds formed through the synthetic methods described herein to provide the compounds of formula I, II, III, IV, V, VI, VII and/or VIII.
  • the intermediate compounds may possess utility as therapeutic agents for the range of indications described herein and/or reagents for further synthesis methods and reactions.
  • any of the R groups as defined herein can be excluded from the compounds of the present invention, particularly with reference to denoting novel compounds of the present invention.
  • aryl refers to an aromatic group, a heteroaryl group or to an optionally substituted aromatic group or heteroaryl group fused to one or more optionally substituted aromatic groups or heteroaryl groups, optionally substituted with suitable substituents including, but not limited to, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, tetrazolyl, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, acyl, aroyl, heteroaroyl, acyloxy, aroyloxy, heteroaroyloxy, alkoxycarbonyl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed.
  • suitable substituents including, but not limited to, lower alkyl,
  • halo refers to bromo, chloro, fluoro or iodo.
  • halide refers to bromide, chloride, fluoride or iodide.
  • hydroxy refers to the group -OH.
  • alkoxy refers to an alkyl group, as defined herein, appended to the parent molecular moiety through an oxy group.
  • Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, hexyloxy and the like.
  • alkaryloxy or “oxyalkaryl” as used herein refers to the group -O- alkyl-aryl wherein Ar is aryl. Examples include, but are not limited to, benzyloxy, oxybenzyl, 2-naphthyloxy and oxy-2-naphthyl.
  • aryloxy refers to the group -ArO wherein Ar is aryl or heteroaryl. Examples include, but are not limited to, phenoxy, benzyloxy and 2- naphthyloxy.
  • amino refers to -NH 2 in which one or both of the hydrogen atoms may optionally be replaced by alkyl or aryl or one of each, optionally substituted.
  • alkylthio or "thioalkyl” as used herein alone or as part of another group, refers to an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfur moiety.
  • alkylthio include, but are not limited to, methylthio, thiomethyl, ethylthio, thioethyl, n-propylthio, thio-n-propyl, isopropylthio, thio-isopropyl, n-butylthio, thio-n-butyl and the like.
  • arylthio or "thioaryl” as used herein refers to the group -ArS wherein Ar is aryl. Examples include, but are not limited to, phenylthio, thiophenyl, 2- naphthylthio and thio-2-naphthyl.
  • alkarylthio or “thioalkaryl” as used herein refers to the group -S- alkyl-aryl wherein Ar is aryl. Examples include, but are not limited to, benzyllthio, thiobenzyl, 2-naphthylthio and thio-2-naphthyl.
  • carboxy as used herein refers to the group -CO 2 H.
  • quaternary ammonium refers to a chemical structure having four bonds to the nitrogen with a positive charge on the nitrogen in the "onium” state, i.e., "R 4 N + or "quaternary nitrogen,” wherein R is an organic substituent such as alkyl or aryl.
  • quaternary ammonium salt refers to the association of the quaternary ammonium cation with an anion.
  • substituted refers to replacement of one or more of the hydrogen atoms of the group replaced by substituents known to those skilled in the art and resulting in a stable compound as described below.
  • suitable replacement groups include, but are not limited to, alkyl, acyl, alkenyl, alkynyl cycloalkyl, aryl, alkaryl, hydroxy, thio, alkoxy, aryloxy, acyl, amino, amido, carboxy, carboxyalkyl, thiocarboxyalkyl, carboxyaryl, thiocarboxyaryl, halo, oxo, mercapto, sulfinyl, sulfonyl, sulfonamido, amidino, carbamoyl, cycloalkyl, heterocycloalkyl, dialkylaminoalkyl, carboxylic acid, carboxamido, haloalkyl, dihaloalkyl,
  • substitutions are permissible when such combinations result in compounds stable for the intended purpose.
  • substitutions are permissible when the resultant compound is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into a therapeutic or diagnostic agent or reagent.
  • an effective amount or “effective” is intended to designate a dose that causes a relief of symptoms of a disease or disorder as noted through clinical testing and evaluation, patient observation, and/or the like. "Effective amount” or “effective” further can further designate a dose that causes a detectable change in biological or chemical activity. The detectable changes may be detected and/or further quantified by one skilled in the art for the relevant mechanism or process. Moreover, “effective amount” or “effective” can designate an amount that maintains a desired physiological state, i.e., reduces or prevents significant decline and/or promotes improvement in the condition of interest. As is generally understood in the art, the dosage will vary depending on the administration routes, symptoms and body weight of the patient but also depending upon the compound being administered.
  • solvate as used herein is intended to refer to a pharmaceutically acceptable solvate form of a specified compound that retains the biological effectiveness of such compound, for example, resulting from a physical association of the compound with one or more solvent molecules.
  • solvates include compounds of the invention in combination with water, 1-propanol, 2-propanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
  • hydrate as used herein refers to the compound when the solvent is water.
  • polyalkylene glycol is a biocompatible polymer where, as used herein, polyalkylene glycol refers to straight or branched polyalkylene glycol polymers such as polyethylene glycol, polypropylene glycol, and polybutylene glycol, and further includes the monoalkylether of the polyalkylene glycol.
  • the polyalkylene glycol polymer is a lower alkyl polyalkylene glycol moiety such as a polyethylene glycol moiety (PEG), a polypropylene glycol moiety, or a polybutylene glycol moiety.
  • PEG has the formula - HO(CH2CH2 ⁇ ) n H, where n can range from about 1 to about 4000 or more. In some embodiments, n is 1 to 100, and in other embodiments, n is 5 to 30.
  • the PEG moiety can be linear or branched. In further embodiments, PEG can be attached to groups such as hydroxyl, alkyl, aryl, acyl or ester.
  • PEG can be an alkoxy PEG, such as methoxy-PEG (or mPEG), where one terminus is a relatively inert alkoxy group, while the other terminus is a hydroxyl group.
  • the compounds of formulas I- VIII can be synthesized using traditional synthesis techniques well known to those skilled in the art. More specific synthesis routes are described below.
  • the bumetanide analogs are synthesized by reacting the carboxylic acid moiety of bumetanide with various reagents.
  • bumetanide may undergo esterification via reaction with alcohols, including linear, branched, substituted, or unsubstituted alcohols.
  • Bumetanide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and aryl halides, including chloroacetonitrile, benzyl chloride, l-(dimethylamino)propyl chloride, 2-chloro-N,N-diethylacetamide, and the like.
  • PEG- type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like, or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO- PEGIOOO-OTs and the like.
  • "Axetil"-type esters may also be formed by alkylation using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Bumetanide may also undergo amidation by reaction with suitable substituted or unsubstituted alkyl amines or aryl amines, either after conversion to the acid chloride or by using an activator, such as l-ethyl-3-(3-dimethylaminopropyl)carbodiiraide (EDC). Bumetanide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonmm hydroxide or cetyltrimethylammonium hydroxide, to form bumetanide quaternary ammonium salts.
  • a cotransporter is electroneutral, moving equal amounts of oppositely charged ionic species from one side of a membrane to another.
  • a cation-chloride cotransporter refers to a cotransporter that moves one or several cations with an equal number of chloride ions.
  • exemplary cation chloride cotransporters include, but are not limited to, the loop diuretic-sensitive Na + K + 2C1 " cotransporter in the brain (NKCCl) and the thiazide-sensitive Na + Cl " cotransporter (NCC). Discussions regarding the molecular classification of cation-chloride cotransporters, their physiology, and pharmacology can be found in Mount et al. J Exp Biol 201: 2091-2102, 1998 and Russell JM. Physiol Rev. 2000 Jan; 80(l):211-76.
  • the NKCCl brain-specific cotransporter is an isoform of its kidney analog, NKCC2. Furosemide and bumetanide are classic examples of NKCC antagonists.
  • the thiazide-sensitive cotransporter is antagonized by thiazide diuretics.
  • thiazide diuretics include, but are not limited to, chlorothiazide, hydrochlorothiazide, and benzthiazide.
  • Modification of the diuretic or diuretic-like compound can include reacting the diuretic or diuretic-like compound with a functional group and/or compound selected from the group consisting of an aluminum hydride, alkyl halide, alcohol, aldehyde, alkaryl halide, mono- and dialkylamine, mono- andi dialkarylamine, mono- and diarylamine, and quaternary ammonium salt, unsubstituted or substituted, or combinations thereof.
  • a functional group and/or compound selected from the group consisting of an aluminum hydride, alkyl halide, alcohol, aldehyde, alkaryl halide, mono- and dialkylamine, mono- andi dialkarylamine, mono- and diarylamine, and quaternary ammonium salt, unsubstituted or substituted, or combinations thereof.
  • a functional group and/or compound selected from the group consisting of an aluminum hydride, alkyl halide, alcohol, aldehyde, alka
  • the thiobumetanide analogs are synthesized by reacting the carboxylic acid moiety of bumetanide with various reagents.
  • the bumetanide analogs are synthesized by reacting the carboxylic acid moiety of bumetanide with various reagents.
  • bumetanide may undergo esterification via reaction with alcohols, including linear, branched, substituted, or unsubstituted alcohols.
  • Bumetanide or thiobumetanide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and alkaryl halides, including chloroacetonitrile, benzyl chloride, l-(dimethylamino)propyl chloride, 2-chloro-N,N- diethylacetamide, and the like.
  • PEG-type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • "Axetil"-type esters may also be formed by alkylation by using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Bumetanide may also undergo amidation by reaction with suitable substituted or unsubstituted alkyl amines or aryl amines, either after conversion to the acid chloride or by using an activator, such as 1- ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC).
  • Bumetanide or thiobumetanide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonium hydroxide or cetyltrimethylammonium hydroxide, to form bumetanide or thiobemetanide quaternary ammonium salts.
  • Schemes 2, 3 and 4 present synthesis schemes of some exemplary compounds according to formula I.
  • R'" methyl, ethyl, propyl
  • Bumetanide salts, thiobumetanide and S-thiobumetanide esters should readily undergo acid- and base-catalyzed hydrolysis to produce the carboxylic acid containing molecule bumetanide by methods well known in the art (See Yang, W. and Drueckhammer, D. G. , J. Amer. Chem. Soc. 2001 , 123 (44), 11004- 11009 and references therein). (See Scheme 4). Scheme 4. Hydrolysis of Bumetanide, thiobumetanide and S-thiobumetanide esters
  • Bumetanide may undergo conversion to the corresponding thioacid by treatment with thionyl chloride to form the corresponding acid chloride followed by reaction with sodium hydroxide or sodium hydrogen sulfide to give metastable O-thiobumetanide and dithiobumetanide by the methodology of Noble, P. and Tarbell, D. S., Org. Synth., Coll. Vol. IV, John Wiley & Sons, Inc., New York, 1963, 924-927. (See Schemes 5 and 6).
  • the thiobumetanide analogs are, in turn, synthesized by reacting the thiocarboxylic acid moiety of S-thiobumetanide with various reagents.
  • S- thiobumetanide may undergo esterification via reaction with alcohols and thiols, including linear, branched, substituted, or unsubstituted alcohols and thiols.
  • S- Thiobumetanide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and alkaryl halides, including chloroacetonitrile, benzyl chloride, l-(dimethylamino)propyl chloride, 2-chloro-N,N-diethylacetamide, and the like.
  • PEG-type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • “Axetil”-type esters may also be formed by alkylation by using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • S- Thiobumetanide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonium hydroxide or cetyltrimethylammonium hydroxide, to form thiobumetanide quaternary ammonium salts. See Schemes 7, 8 and 9, which present some exemplary compounds according to formula II.
  • R benzyl, cetyl, methyl, ethyl
  • R' methyl, ethyl, propyl
  • R" methyl, ethyl, propyl
  • R'" methyl, ethyl, propyl
  • Thiobumetanide, thiobumetanide amides, O-thiobumetanide esters and dithiobumetanide esters should readily undergo acid- and base-catalyzed hydrolysis to produce the carboxylic acid containing molecule bumetanide by methods well known in the art (See Yang, W. and Drueckhammer, D. G., J. Amer. Chem. Soc. 2001, 123 (44),
  • O-thiobumetanide esters dithiobumetanide esters O-bumetanide thioesters
  • bumetanide dithioesters O-thiobumetanide esters
  • the thiofurosemide analogs are synthesized by reacting the carboxylic acid moiety of furosemide with various reagents.
  • the furosemide analogs are synthesized by methods analogous to those used . in the synthesis of the bumetanide analogs.
  • Furosemide may undergo esterification via reaction with alcohols, including linear, branched, substituted, or unsubstituted alcohols.
  • Furosemide or thiofurosemide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and alkaryl halides, including for example, chloroacetonitrile, benzyl chloride, l-(dimethylamino)propyl chloride, 2-chloro-N,N- diethylacetamide, and the like.
  • PEG-type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • "Axetil"-type esters may also be formed by alkylation by using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Furosemide may also undergo amidation by reaction with suitable substituted or unsubstituted alkyl amines or aryl amines, either after conversion to the acid chloride or by using an activator, such as 1- ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC). Furosemide or thiofurosemide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonium hydroxide or cetyltrimethylammonium hydroxide, to form furosemide or thiofurosemide quaternary ammonium salts.
  • Schemes, 11, 12 and 13 present some exemplary compounds according to formula III.
  • Scheme 11 Synthesis of Exemplary Compounds According to Formula III
  • R" methyl, ethyl, propyl,
  • R'" methyl, ethyl, propyl
  • Thiofurosemide salts and S-thiofurosemide esters should readily undergo acid- and base-catalyzed hydrolysis to produce the carboxylic acid containing molecule furosemide by methods well known in the art (See Yang, W. and Drueckhammer, D. G., J. Amer. Chem. Soc, 2001, 123 (44), 11004-11009 and references therein). (See Scheme 13). Scheme 13. Hydrolysis of Thiofurosemide salts and S-thiofurosemide Esters
  • the thiofurosemide analogs are, in turn, synthesized by reacting the thiocarboxylic acid moiety of thiofurosemide with various reagents.
  • thiofurosemide may undergo esterification via reaction with alcohols or thiols, including linear, branched, substituted, or unsubstituted alcohols and thiols.
  • S-Thiofurosemide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and alkaryl halides, including chloroacetonitrile, benzyl chloride, l-(dimethylamino)propyl chloride, 2-chloro-N,N-diethylacetamide, and the like.
  • PEG-type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • Alkyl-type esters may also be formed by alkylation by using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Thiofurosemide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonium hydroxide or cetyltrimethylammonium hydroxide, to form thiofurosemide quaternary ammonium salts.
  • Schemes 14, 15, 16, 17 and 18 present synthesis schemes of some exemplary compounds according to formula IV.
  • Thiofurosemide, thiofurosemide amides and S-thiofurosemide esters should readily undergo acid- and base-catalyzed hydrolysis to produce the carboxylic acid containing molecule furosemide by methods well known in the art (See Yang, W. and
  • piretanide analogs are synthesized by reacting the carboxylic acid moiety of piretanide with various reagents.
  • Piretanide may undergo esterification via reaction with alcohols, including linear, branched, substituted, or unsubstituted alcohols.
  • Piretanide or thiopiretanide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and alkaryl halides, including chloroacetonitrile, benzyl chloride, l-(dimethylammo)propyl chloride, 2-chloro-N,N-diethylacetamide, and the like.
  • PEG-type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like. "Axetir'-type esters may also be formed by alkylation by using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Piretanide may also undergo amidation by reaction with suitable substituted or unsubstituted alkyl amines or aryl amines, either after conversion to the acid chloride or by using an activator, such as 1- ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC).
  • EDC 1- ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • Piretanide or thiopiretanide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonium hydroxide or cetyltrimethylammonium hydroxide, to form piretanide or thiopiretanide quaternary ammonium salts.
  • Schemes 19, 20, 21 and 22 present synthesis schemes of some exemplary compounds according to formula V.
  • Thiopiretanide salts and S-thiopiretanide esters should readily undergo acid- and base-catalyzed hydrolysis to produce the carboxylic acid containing molecule bumetanide by methods well known in the art (See Yang, W., Drueckhammer D. G., J. Amer. Chem. Soc, 2001, 123 (44), 11004-11009 and references therein). (See Scheme 22). Scheme 22. Hydrolysis of Thiopiretanide salts and S-thiopiretanide Esters
  • thiopiretanide analogs are synthesized by methods analogous to those used in the synthesis of the piretanide analogs. Specifically, thiopiretanide may undergo esterification via reaction with thiols, including linear, branched, substituted, or unsubstituted thiols. Thiopiretanide may also be alkylated via reaction with suitable substituted and unsubstituted alkyl halides and alkaryl halides, including chloroacetonitrile, benzyl chloride, l-(dimethylamino)pro ⁇ yl chloride, 2-chloro-N,N- diethylacetamide, and the like.
  • PEG-type esters may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • "Axetil"-type thioesters may also be formed by alkylation by using alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Thiopiretanide may also be reacted with a quaternary ammonium hydroxide, such as benzyltrimethylammonium hydroxide or cetyltrimethylammonium hydroxide, to form thiopiretanide quaternary ammonium salts.
  • Schemes 23, 24, 25, 26 and 27 present some exemplary compounds according to formula VI.
  • Scheme 25 Synthesis of Exemplary Compounds According to Formula VI
  • R benzyl, cetyl, methyl, ethyl
  • R' methyl, ethyl, propyl
  • R H, methyl, ethyl, benzyl
  • R' H, methyl, ethyl, benzyl 5a thiopiretanide quaternary ammonium salts
  • R benzyl, cetyl, methyl, ethyl
  • Thiopiretanide, thiopiretanide amides and thiopiretanide esters should readily undergo acid- and base-catalyzed hydrolysis to produce the carboxylic acid containing molecule piretanide by methods well known in the art ⁇ See Yang, W. and Drueckhammer, D. G., J. Amer. Chem. Soc, 2001, 123 (44), 11004-11009 and references therein).
  • Thioacyl Halides "Thiocarboxylic O- Acid Esters” and “Dithiocarboxylic Acid Esters"
  • O-thiopiretanide esters dithiopiretanide ester O-piretanide thioesters (piretanide dithioester)
  • azosemide analogs are synthesized by the reaction of various reagents with the tetrazolyl moiety of azosemide.
  • Azosemide may undergo hydroxyalkylation with the addition of an aldehyde, whereby a hydroxylalkyl functionality is formed.
  • An alcohol may optionally be reacted along with the aldehyde to obtain an ether.
  • An alkyl thiol may optionally be added with the aldehyde to form a thioether.
  • Azosemide may also be alkylated by the addition of suitable alkyl halides or alkaryl halides, including alkyl or alkaryl halides comprising an ether or thioether linkage, such as methyl chloromethyl ether and benzyl chloromethyl thioether.
  • PEG-type ethers may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • Azosemide may also be reacted with a quaternary ammonium salt, such as benzyltrimethylammoniumbromide and base such as sodium hydroxide or cetyltrimethylammonium bromide and base such as sodium hydroxide, in order to form an azosemide quaternary ammonium salt.
  • Scheme 28 below presents a synthesis scheme of some exemplary compounds according to formula VII.
  • torsemide also known as torasemide
  • Torsemide may undergo alkylation by the addition of suitable alkyl or alkaryl halides, including benzyl chloride, to form N-substituted quaternary ammonium salts.
  • suitable alkyl or alkaryl halides including benzyl chloride
  • Alkyl halides and alkaryl halides comprising an ether linkage, including methyl chloromethyl ether and benzyl chloromethyl ether may be used to form N-substituted ether quaternary ammonium salts.
  • Alkyl halides and alkaryl halides comprising a thioether linkage, including methyl chloromethyl thioether and benzyl chloromethyl thioether, may be used to form N- substituted thioether quaternary ammonium salts.
  • PEG-type ether-containing quaternary ammonium salts may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like.
  • Alkyl halides such as chloromethyl pivalate or chloromethyl propionate.
  • Scheme 29 below presents a synthesis scheme of some exemplary compounds according to formula VIII.
  • the substituted benzoic acids bumetanide, piretanide and furosemide can be selectively reduced to the corresponding bumetanide aldehyde, piretanide aldehyde and furosemide aldehyde using amine-substituted ammonium hydrides such as bis(4- memylpiperazinyl)aluminum hydride by literature methods. See Muraki, M. and Mukiayama, T., Chem. Letters, 1974, 1447; Muraki, M. and Mukiayama, T., Chem. Letters, 1975, 215; and Hubert, T., D., Eyman, D. P. and Wiemer, D. F., J. Org.
  • R 3 O-aryl
  • the lipophilic thiobenzaldehydes can also be prepared from the corresponding benzaldehydes by treating agents including hydrogen sulfide and diphosphorus pentasulfide (See Smith, M. B. and March, J., March's Advanced Organic Chemistry, Fifth Edition, 2001, John Wiley & Sons, Inc., New York, Part 2, Chapter 16, pp. 1185- 1186.
  • agents including hydrogen sulfide and diphosphorus pentasulfide (See Smith, M. B. and March, J., March's Advanced Organic Chemistry, Fifth Edition, 2001, John Wiley & Sons, Inc., New York, Part 2, Chapter 16, pp. 1185- 1186.
  • C. Sulfur Nucleophiles, Section 16-10, The Addition of H 2 S and Thiols to Carbonyl Compounds.) See Scheme 31).
  • these thiobenzaldehydes are readily converted back into the corresponding benzaldehydes under hydrolytic conditions.
  • bumetanide aldehyde "bumetanide thioaldehyde"
  • R 3 O-aryl
  • the PEG-type esters of bumetanide, furosemide and piretanide may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like. (See Scheme 32).
  • piretanide PEG esters OCH 2 CH 2 (OCH 2 CH 2 V 1 -Y
  • R 3 O-aryi
  • n 1 - 100
  • the PEG-type esters of thiobumetanide, thiofurosemide and thiopiretanide may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like. (See Scheme 33).
  • Scheme 33 Synthesis of Exemplary Polyethylene Glycol Thioesters of Thiobumetanide, Thiofurosemide and Thiopiretanide
  • R 2 OCH z CH 2 (OCH2CH 2 ) n -rY
  • R 3 O-aryl
  • n 1 - 100
  • R 2 OCH 2 CH 2 (OCH 2 CH 2 J n-1 -Y thiofurosemide
  • R 3 chloride
  • n 1 - 100
  • R 3 O-aryl
  • n 1 - 100
  • the PEG-type esters of dithiobumetanide, dithioftirosemide and dithiopiretanide may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO- PEG350-C1 and the like or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO- PEGIOOO-OTs and the like. (See Scheme 34). Scheme 34. Synthesis of Exemplary Polyethylene Glycol Dithioesters of Dithiobumetanide, Dithiofurosemide and Dithiopiretanide
  • R 2 OCH 2 CH 2 (OGH 2 CHa) n -I-Y
  • R 3 chloride
  • n 1 - 100
  • R 3 O-aryl
  • n 1 - 100
  • PEG-X is X-(CH 2 ) ni (OCH a CH 2 )n-rY.
  • X halo or other leaving group (mesylate "OMs", tosylate "OTs”)
  • H PEG-Type Analogs of Azosemide and Torsemide
  • the PEG-type ethers of azosemide and torsemide may be formed by alkylation using alkyloxy(polyalkyloxy)alkyl halides such as MeO-PEG350-Cl and the like, or alkyloxy(polyalkyloxy)alkyl tosylates such as MeO-PEGlOOO-OTs and the like. (See Scheme 35).
  • torsemide torsemide methyl PEG ether quaternary ammonium salts
  • R 7 (CH 2 ) m OCH 2 CH 2 (OCH 2 CH 2 ) n .
  • Starting materials for synthesizing compounds of the present invention can further include compounds described in U.S. Patent No. 3,634,583 to Feit; U.S. Patent No.
  • Compounds of the present invention can include isomers, tautomers, zwitterions, enantiomers, diastereomers, racemates or stereochemical mixtures thereof.
  • isomers refers to compounds having the same number and kind of atoms, and hence the same molecular weight, but differing with respect to the arrangement or configuration of the atoms in space. Additionally, the term “isomers” includes stereoisomers and geometric isomers.
  • stereoisomer or “optical isomer” as used herein refer to a stable isomer that has at least one chiral atom or restricted rotation giving rise to perpendicular dissymmetric planes (e.g., certain biphenyls, allenes, and spiro compounds) and can rotate plane-polarized light. Because asymmetric centers and other chemical structure can exist in some of the compounds of the present invention which may give rise to stereoisomerism, the invention contemplates stereoisomers and mixtures thereof.
  • the compounds of the present invention and their salts can include asymmetric carbon atoms and may therefore exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers.
  • such compounds will be prepared as a racemic mixture. If desired, however, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures.
  • Tautomers are readily interconvertible constitutional isomers and there is a change in connectivity of a ligand, as in the keto and enol forms of ethyl acetoacetate (The present invention includes tautomers of any said compounds.)
  • Zwitterions are inner salts or dipolar compounds possessing acidic and basic groups in the same molecule. At neutral pH, the cation and anion of most zwitterions are equally ionized.
  • the present invention further provides prodrugs comprising the compounds described herein.
  • prodrug is intended to refer to a compound that is converted under physiological conditions, by solvolysis or metabolically, to a specified compound that is pha ⁇ naceutically/pharmacologically active.
  • the prodrug can be a compound of the present invention that has been chemically derivatized such that: (i) it retains some, all or none of the bioactivity of its parent drug compound, and (ii) it is metabolized in a subject to yield the parent drug compound.
  • the prodrug of the present invention may also be a "partial prodrug" in that the compound has been chemically derivatized such that: (i) it retains some, all or none of the bioactivity of its parent drug compound, and (ii) it is metabolized in a subject to yield a biologically active derivative of the compound.
  • the prodrugs can be formed utilizing a hydrolyzable coupling to the compounds described herein. A further discussion of prodrugs can be found in Ettmayer et al. J. Med. Chem. 47(10):2394-2404 (2004).
  • Prodrugs of the present invention are capable of passage across the blood-brain barrier and may undergo hydrolysis by CNS esterases to provide the active compound. Further, the prodrugs provided herein may also exhibit improved bioavailability, improved aqueous solubility, improved passive intestinal absorption, improved transporter-mediated intestinal absorption, protection against accelerated metabolism, tissue-selective delivery and/or passive enrichment in the target tissue.
  • Prodrugs of the present invention can include compounds according to formula I, II, III, IV and/or V described herein.
  • Prodrugs of the present invention can further include bumetanide, bumetanide dibenzylamide, bumetanide diethylamide, bumetanide morpholinoethyl ester, bumetanide 3-(dimethylaminopropyl) ester, bumetanide N 5 N- diethylglycolamide ester, bumetanide dimethylglycolamide ester, bumetanide pivaxetil ester, furosemide, furosemide ethyl ester, furosemide cyanomethyl ester, furosemide benzyl ester, furosemide morpholinoethyl ester, furosemide 3-(dimethylaminopropyl) ester, furosemide N,N-diethylglycolamide ester, furosemide dibenzylamide, furosemide benzyltri
  • prodrugs can be formed by attachment of biocompatible polymers, such as those previously described including polyethylene glycol (PEG), to compounds of the present invention using linkages degradable under physiological conditions.
  • PEG polyethylene glycol
  • linkages degradable under physiological conditions See also Schacht, E.H. et al. Poly(ethylene glycol) Chemistry and Biological Applications, American Chemical Society, San Francisco, CA 297-315 (1997). Attachment of PEG to proteins can be employed to reduce immunogenicity and/or extend the half-life of the compounds provided herein. Any conventional PEGylation method can be employed, provided that the PEGylated agent retains pharmaceutical activity.
  • compositions of the subject invention are suitable for human and veterinary applications and are preferably delivered as pharmaceutical compositions.
  • Pharmaceutical compositions comprise one or more treatment agents, or a pharmaceutically acceptable salt thereof, and a physiologically acceptable carrier.
  • a pharmaceutically acceptable salt refers to a salt form of a compound permitting its use or formulation as a pharmaceutical and which retains the biological effectiveness of the free acid and base of the specified compound and is not biologically or otherwise undesirable. Examples of such salts are described in Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Wermuth, CG. and Stahl, P.H. (eds.), Wiley- Verlag Helvetica Acta, Zurich, 2002 [ISBN 3-906390-26-8].
  • salts include alkali metal salts and addition salts of free acids and bases.
  • pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzo
  • compositions of the present invention may also contain other compounds, which may be biologically active or inactive.
  • one or more treatment agents of the present invention may be combined with another agent, in a treatment combination, and administered according to a treatment regimen of the present invention.
  • Such combinations may be administered as separate compositions, combined for delivery in a complementary delivery system, or formulated in a combined composition, such as a mixture or a fusion compound.
  • the aforementioned treatment combination may include a BBB permeability enhancer and/or a hyperosmotic agent.
  • the carriers and additives used for such pharmaceutical compositions can take a variety of forms depending on the anticipated mode of administration.
  • compositions for oral administration may be, for example, solid preparations such as tablets, sugar-coated tablets, hard capsules, soft capsules, granules, powders and the like, with suitable carriers and additives being starches, sugars, binders, diluents, granulating agents, lubricants, disintegrating agents and the like. Because of their ease of use and higher patient compliance, tablets and capsules represent advantageous oral dosage forms for many medical conditions.
  • compositions for liquid preparations include solutions, emulsions, dispersions, suspensions, syrups, elixirs, and the like with suitable carriers and additives being water, alcohols, oils, glycols, preservatives, flavoring agents, coloring agents, suspending agents, and the like.
  • suitable carriers and additives being water, alcohols, oils, glycols, preservatives, flavoring agents, coloring agents, suspending agents, and the like.
  • Typical preparations for parenteral administration comprise the active ingredient with a carrier such as sterile water or parenterally acceptable oil including polyethylene glycol, polyvinyl pyrrolidone, lecithin, arachis oil or sesame oil, with other additives for aiding solubility or preservation may also be included.
  • a carrier such as sterile water or parenterally acceptable oil including polyethylene glycol, polyvinyl pyrrolidone, lecithin, arachis oil or sesame oil, with other additives for aiding solubility or preservation may
  • compositions according to embodiments of the present invention include those suitable for oral, rectal, topical, nasal, inhalation (e.g., via an aerosol) buccal (e.g., sub-lingual), vaginal, topical (i.e., both skin and mucosal surfaces, including airway surfaces), transdermal administration and parenteral (e.g., subcutaneous, intramuscular, intradermal, intraarticular, intrapleural, intraperitoneal, intrathecal, intracerebral, intracranially, intraarterial, or intravenous), although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular active agent which is being used.
  • Pharmaceutical compositions of the present invention are particularly suitable for oral, sublingual, parenteral, implantation, nasal and inhalational administration.
  • compositions for injection will include the active ingredient together with suitable carriers including propylene glycol-alcohol-water, isotonic water, sterile water for injection (USP), emulPhorTM-alcohol-water, cremophor-ELTM or other suitable carriers known to those skilled in the art. These carriers may be used alone or in combination with other conventional solubilizing agents such as ethanol, a glycol, or other agents known to those skilled in the art. Where the compounds of the present invention are to be applied in the form of solutions or injections, the compounds may be used by dissolving or suspending in any conventional diluent.
  • the diluents may include, for example, physiological saline, Ringer's solution, an aqueous glucose solution, an aqueous dextrose solution, an alcohol, a fatty acid ester, glycerol, a glycol, an oil derived from plant or animal sources, a paraffin and the like. These preparations may be prepared according to any conventional method known to those skilled in the art.
  • compositions for nasal administration may be formulated as aerosols, drops, powders and gels.
  • Aerosol formulations typically comprise a solution or fine suspension of the active ingredient in a physiologically acceptable aqueous or non-aqueous solvent.
  • Such formulations are typically presented in single or multidose quantities in a sterile form in a sealed container.
  • the sealed container can be a cartridge or refill for use with an atomizing device.
  • the sealed container may be a unitary dispensing device such as a single use nasal inhaler, pump atomizer or an aerosol dispenser fitted with a metering valve set to deliver a therapeutically effective amount, which is intended for disposal once the contents have been completely used.
  • the dosage form comprises an aerosol dispenser, it will contain a propellant such as a compressed gas, air as an example, or an organic propellant including a fluorochlorohydrocarbon or fluorohydrocarbon.
  • compositions suitable for buccal or sublingual administration include tablets, lozenges and pastilles, wherein the active ingredient is formulated with a carrier such as sugar and acacia, tragacanth or gelatin and glycerin.
  • a carrier such as sugar and acacia, tragacanth or gelatin and glycerin.
  • compositions for rectal administration include suppositories containing a conventional suppository base such as cocoa butter.
  • compositions suitable for transdermal administration include ointments, gels and patches.
  • compositions known to those skilled in the art can also be applied for percutaneous or subcutaneous administration, such as plasters.
  • compositions comprising the active ingredient or ingredients in admixture with components necessary for the formulation of the compositions
  • other conventional pharmacologically acceptable additives may be incorporated, for example, excipients, stabilizers, antiseptics, wetting agents, emulsifying agents, lubricants, sweetening agents, coloring agents, flavoring agents, isotonicity agents, buffering agents, antioxidants and the like.
  • additives there may be mentioned, for example, starch, sucrose, fructose, dextrose, lactose, glucose, mannitol, sorbitol, precipitated calcium carbonate, crystalline cellulose, earboxymethylcellulose, dextrin, gelatin, acacia, EDTA, magnesium stearate, talc, hydroxypropylmethylcellulose, sodium metabisulfite, and the like.
  • kits including one or more containers comprising pharmaceutical dosage units comprising an effective amount of one or more compounds of the present invention.
  • the essential active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if desired, emulsifying or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin and combinations thereof.
  • compositions described herein may be administered as part of a sustained release formulation.
  • sustained release formulations may generally be prepared using well-known technology and administered by, for example, oral, rectal or transdermal delivery systems, or by implantation of a formulation or therapeutic device at one or more desired target site(s).
  • Sustained-release formulations may contain a treatment composition comprising an inventive treatment agent alone, or in combination with a second treatment agent, dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane.
  • Carriers for use within such formulations are biocompatible, and may also be biodegradable.
  • the sustained release formulation provides a relatively constant level of active composition release.
  • the sustained release formulation is contained in a device that may be actuated by the subject or medical personnel, upon onset of certain symptoms, for example, to deliver predetermined dosages of the treatment composition.
  • the amount of the treatment composition contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition to be treated or prevented.
  • compositions of the present invention for treatment of neuropathic pain and neuropsychiatric disorders are administered using a formulation and a route of administration that facilitates delivery of the treatment composition(s) to the central nervous system.
  • Treatment compositions such as NKCCl antagonists, may be formulated to facilitate crossing of the blood brain barrier as described above, or may be co-administered with an agent that crosses the blood brain barrier.
  • Treatment compositions may be delivered in liposome formulations, for example, that cross the blood brain barrier, or may be co-administered with other compounds, such as bradykinins, bradykinin analogs or derivatives, or other compounds, such as SERAPORTTM, that cross the blood brain barrier.
  • treatment compositions of the present invention may be delivered using a spinal tap that places the treatment composition directly in the circulating cerebrospinal fluid.
  • a spinal tap that places the treatment composition directly in the circulating cerebrospinal fluid.
  • a bolus iv injection of 20 mg furosemide reduces or abolishes both spontaneous interictal activity and electrical stimulation-evoked epileptiform activity in human patients who are refractory to antiepileptic drugs (AEDs) (Haglund & Hochman J Neurophysiol. (Feb. 23, 2005) doi:10.1152/ jn.00944.2004).
  • AEDs antiepileptic drugs
  • dosages vary according to the indication, and from individual to individual, and may be readily determined by a physician from information that is generally available, and by monitoring patients and adjusting the dosages and treatment regimen accordingly using standard techniques.
  • appropriate dosages and treatment regimen provide the active composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit. Dosages and treatment regimen may be established by monitoring improved clinical outcomes in treated patients as compared to non-treated patients.
  • an effective amount or “effective” is intended to designate a dose that causes a relief of symptoms of a disease or disorder as noted through clinical testing and evaluation, patient observation, and/or the like. "Effective amount” or “effective” further can further designate a dose that causes a detectable change in biological or chemical activity. The detectable changes may be detected and/or further quantified by one skilled in the art for the relevant mechanism or process. Moreover, “effective amount” or “effective” can designate an amount that maintains a desired physiological state, i.e., reduces or prevents significant decline and/or promotes improvement in the condition of interest. Therapeutically effective dosages and treatment regimen will depend on the condition, the severity of the condition, and the general state of the patient being treated.
  • a preferred method for determining a therapeutically effective dosage in a patient is to gradually escalate the dosage and monitor the clinical and laboratory indicia.
  • the two or more agents are coadministered such that each of the agents is present in a therapeutically effective amount for sufficient time to produce a therapeutic or prophylactic effect.
  • coadministration is intended to encompass simultaneous or sequential administration of two or more agents in the same formulation or unit dosage form or in separate formulations. Appropriate dosages and treatment regimen for treatment of acute episodic conditions, chronic conditions, or prophylaxis will necessarily vary to accommodate the condition of the patient.
  • furosemide may be administered orally to a patient in amounts of 10-40 ing at a frequency of 1-3 times per day, preferably in an amount of 40 mg three times per day.
  • bumetanide may be administered orally for the treatment of neuropathic pain in amounts of 1-10 mg at a frequency of 1-3 times per day.
  • smaller doses may be employed, for example, in pediatric applications.
  • bumetanide analogs according to the present invention may be administered in amounts of 1.5 to 6 mg daily, for example 1 tablet or capsule three times a day.
  • furosemide analogs according to the present invention may be administered in amounts of 60 to 240 mg/day, for example 1 tablet or capsule three times a day.
  • piretanide analogs according to the present invention may be administered in amounts of 10 to 20 mg daily, for example 1 tablet or capsule once a day.
  • azosemide analogs according to the present invention may be administered in an amount of 60 mg per day.
  • torsemide analogs according to the present invention may be administered in amounts of 10 to 20 mg daily, for example 1 tablet or capsule once a day. It should be noted that lower doses may be administered, particularly for IV administration. Methods and systems of the present invention may also be used to evaluate candidate compounds and treatment regimen for the treatment and/or prophylaxis of neuropathic pain and neuropsychiatric disorders. Various techniques for generating candidate compounds potentially having the desired NKCCl cotransporter antagonist activity may be employed. Candidate compounds may be generated using procedures well known to those skilled in the art of synthetic organic chemistry. Structure-activity relationships and molecular modeling techniques are useful for the purpose of modifying known NKCCl antagonists, such furosemide, bumetanide, ethacrinic acid and related compounds, to confer the desired activities and specificities. Methods for screening
  • Candidate compounds may be screened for NKCCl antagonist activity using screening methods of the present invention with various types of cells in culture such as glial cells, neuronal cells, renal cells, and the like, or in situ in animal models. Screening techniques to identify chloride cotransporter antagonist activity, for example, may involve altering the ionic balance of the extracellular space in the tissue culture sample, or in situ in an animal model, by producing a higher than "normal" anionic chloride concentration. The geometrical and/or optical properties of the cell or tissue sample subject to this altered ionic balance are determined, and candidate agents are administered.
  • the corresponding geometrical and/or optical properties of the cell or tissue sample are monitored to determine whether the ionic imbalance remains, or whether the cells responded by altering the ionic balances in the extracellular and intracellular space. If the ionic imbalance remains, the candidate agent is likely a chloride cotransporter antagonist.
  • candidate compounds having a high level of glial cell chloride cotransporter antagonist activity and having a reduced level of neuronal cell and renal cell chloride cotransporter antagonist activity may be identified. Similarly, effects on different types of cells and tissue systems may be assessed.
  • the efficacy of candidate compounds may be assessed by simulating or inducing a condition, such as neuropathic pain, in situ in an animal model, monitoring the geometrical and/or optical properties of the cell or tissue sample during stimulation of the condition, administering the candidate compound, then monitoring the geometrical and/or optical properties of the cell or tissue sample following administration of the candidate compound, and comparing the geometrical and/or optical properties of the cell or tissue sample to determine the effect of the candidate compound.
  • Testing the efficacy of treatment compositions for relief of neuropathic pain can be carried using well known methods and animal models, such as that described in Bennett, Hosp. Pract. (Off Ed). 33:95-98, 1998.
  • compositions for use in the inventive methods may comprise a treatment agent selected from the group consisting of: antibodies, or antigen-binding fragments thereof, that specifically bind to NKCCl; soluble ligands that bind to NKCCl;
  • si anti-sense oligonucleotides to NKCCl si anti-sense oligonucleotides to NKCCl; and small interfering RNA molecules (siRNA or RNAi) that are specific for NKCCl.
  • siRNA or RNAi small interfering RNA molecules
  • Antibodies that specifically bind to NKCCl are known in the art and include those available from Alpha Diagnostic International, Inc. (San Antonio, TX 78238).
  • An "antigen-binding site,” or “antigen-binding fragment” of an antibody refers to the part of the antibody that participates in antigen binding.
  • the antigen binding site is formed by amino acid residues of the N-terminal variable ("V") regions of the heavy ("H") and light (“L”) chains.
  • V N-terminal variable
  • H heavy
  • L light chains.
  • Three highly divergent stretches within the V regions of the heavy and light chains are referred to as "hypervariable regions" which are interposed between more conserved flanking stretches known as "framework regions,” or "FRs”.
  • FR refers to amino acid sequences which are naturally found between and adjacent to hypervariable regions in immunoglobulins.
  • the three hypervariable regions of a light chain and the three hypervariable regions of a heavy chain are disposed relative to each other in three dimensional space to form an antigen- binding surface.
  • the antigen-binding surface is complementary to the three-dimensional surface of a bound antigen, and the three hypervariable regions of each of the heavy and light chains are referred to as "complementarity-determining regions,” or "CDRs.”
  • a number of molecules are known in the art that comprise antigen-binding sites capable of exhibiting the binding properties of an antibody molecule.
  • the proteolytic enzyme papain preferentially cleaves IgG molecules to yield several fragments, two of which (the "F(ab)" fragments) each comprise a covalent heterodimer that includes an intact antigen-binding site.
  • the enzyme pepsin is able to cleave IgG molecules to provide several fragments, including the "F(ab') 2 " fragment, which comprises both antigen-binding sites.
  • An "Fv” fragment can be produced by preferential proteolytic cleavage of an IgM, IgG or IgA immunoglobulin molecule, but are more commonly derived using recombinant techniques known in the art.
  • the Fv fragment includes a non-covalent V H "V L heterodimer including an antigen-binding site which retains much of the antigen recognition and binding capabilities of the native antibody molecule (Inbar et al. Proc. Natl. Acad. Sd. USA 69:2659-2662, 1972; Hochman et al. Biochem 75:2706-2710, 1976; and Ehrlich et al. Biochem 79:4091-4096, 1980).
  • Humanized antibodies that specifically bind to NKCCl may also be employed in the inventive methods.
  • a number of humanized antibody molecules comprising an antigen-binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent V regions and their associated CDRs fused to human constant domains (Winter et al. Nature 349:293-299, 1991; Lobuglio et al. Proc. Natl. Acad. Sci. USA 86:4220-4224, 1989; Shaw et al. J Immunol. 755:4534-4538, 1987; and Brown et al. Cancer Res.
  • Modulating the activity of NKCCl may alternatively be accomplished by reducing or inhibiting expression of the polypeptide, which can be achieved by interfering with transcription and/or translation of the corresponding polynucleotide.
  • Polypeptide expression may be inhibited, for example, by introducing anti-sense expression vectors, anti-sense oligodeoxyribonucleotides, anti-sense phosphorothioate oligodeoxy- ribonucleotides, anti-sense oligoribonucleotides or anti-sense phosphorothioate oligoribonucleotides; or by other means well known in the art. All such anti-sense polynucleotides are referred to collectively herein as "anti-sense oligonucleotides”.
  • the anti-sense oligonucleotides for use in the inventive methods are sufficiently complementary to the NKCCl polynucleotide to bind specifically to the polynucleotide.
  • the sequence of an anti-sense oligonucleotide need not be 100% complementary to the of the polynucleotide in order for the anti-sense oligonucleotide to be effective in the inventive methods.
  • an anti-sense oligonucleotide is sufficiently complementary when binding of the anti-sense oligonucleotide to the polynucleotide interferes with the normal function of the polynucleotide to cause a loss of utility, and when non-specific binding of the oligonucleotide to other, non-target sequences is avoided.
  • the design of appropriate anti-sense oligonucleotides is well known in the art. Oligonucleotides that are complementary to the 5' end of the message, for example the 5' untranslated sequence up to and including the AUG initiation codon, should work most efficiently at inhibiting translation.
  • oligonucleotides complementary to either the 5'- or 3'-non- translated, non-coding, regions of the targeted polynucleotide may also be employed.
  • Cell permeation and activity of anti-sense oligonucleotides can be enhanced by appropriate chemical modifications, such as the use of phenoxazine-substituted C-5 propynyl uracil oligonucleotides (Flanagan et al., Nat. Biotechnol. 17:48-52, 1999) or 2'- O-(2-methoxy) ethyl (2'-MOE)-oligonucleotides (Zhang et al, Nat. Biotechnol.
  • RNA interference RNA interference
  • RNAi also operates on a post-translational level and is sequence specific, but suppresses gene expression far more efficiently.
  • Exemplary methods for controlling or modifying gene expression are provided in WO 99/49029, WO 99/53050 and WO01/75164, the disclosures of which are hereby incorporated by reference. In these methods, post-transcriptional gene silencing is brought about by a sequence-specific RNA degradation process which results in the rapid degradation of transcripts of sequence-related genes.
  • RNA may act as a mediator of sequence-specific gene silencing (see, for example, Montgomery and Fire, Trends in Genetics, 14:255-258, 1998). Gene constructs that produce transcripts with self-complementary regions are particularly efficient at gene silencing.
  • RNAi specifically bind to and cleave double-stranded RNA into short fragments.
  • the ribonuclease(s) remains associated with these fragments, which in turn specifically bind to complementary mRNA, i.e. specifically bind to the transcribed rnRNA strand for the gene of interest.
  • the mRNA for the gene is also degraded by the ribonuclease(s) into short fragments, thereby obviating translation and expression of the gene.
  • an RNA- polymerase may act to facilitate the synthesis of numerous copies of the short fragments, which exponentially increases the efficiency of the system.
  • silencing is not limited to the cells where it is initiated. The gene-silencing effects may be disseminated to other parts of an organism.
  • the NKCCl polynucleotide may thus be employed to generate gene silencing constructs and/or gene-specific self-complementary, double-stranded RNA sequences that can be employed in the inventive methods using delivery methods known in the art.
  • a gene construct may be employed to express the self-complementary RNA sequences.
  • cells may be contacted with gene-specific double-stranded RNA molecules, such that the RNA molecules are internalized into the cell cytoplasm to exert a gene silencing effect.
  • the double-stranded RNA must have sufficient homology to the NKCCl gene to mediate RNAi without affecting expression of non-target genes.
  • the double-stranded DNA is at least 20 nucleotides in length, and is preferably 21-23 nucleotides in length.
  • the double-stranded RNA corresponds specifically to a polynucleotide of the present invention.
  • siRNA small interfering RNA
  • RNAi technique employs genetic constructs within which sense and anti- sense sequences are placed in regions flanking an intron sequence in proper splicing orientation with donor and acceptor splicing sites. Alternatively, spacer sequences of various lengths may be employed to separate self-complementary regions of sequence in the construct.
  • intron sequences are spliced-out, allowing sense and anti-sense sequences, as well as splice junction sequences, to bind forming double-stranded RNA.
  • Select ribonucleases then bind to and cleave the double-stranded RNA, thereby initiating the cascade of events leading to degradation of specific mRNA gene sequences, and silencing specific genes.
  • a genetic construct, anti-sense oligonucleotide or RNA molecule may be administered by various art-recognized procedures (see, e.g., Rolland, CHt. Rev. Therap. Drug Carrier Systems /5:143-198, 1998, and cited references). Both viral and non- viral delivery methods have been used for gene therapy.
  • Useful viral vectors include, for example, adenovirus, adeno-associated virus (AAV), retrovirus, vaccinia vims and avian poxvirus.
  • Improvements have been made in the efficiency of targeting genes to tumor cells with adenoviral vectors, for example, by coupling adenovirus to DNA- polylysine complexes and by strategies that exploit receptor-mediated endocytosis for selective targeting (see, e.g., Curiel et al., Hum. Gene Then, 5: 147-154, 1992; and Cristiano & Curiel, Cancer Gene Ther. 3:49-57, 1996).
  • Non- viral methods for delivering polynucleotides are reviewed in Chang & Seymour, (Eds) Curr. Opin. MoI. Ther., vol. 2, 2000.
  • Liposomes can be modified by incorporation of ligands that recognize cell-surface receptors and allow targeting to specific receptors for uptake by receptor-mediated endocytosis (see, for example, Xu et al., MoI. Genet. Metab., 64:193-197; 1998; and Xu et al., Hum. Gene Ther., 10:2941-2952, 1999).
  • Tumor-targeting bacteria such as Salmonella
  • Bacteria can be engineered ex vivo to penetrate and to deliver DNA with high efficiency into, for example, mammalian epithelial cells in vivo (see, e.g., Grillot- Courvalin et al., Nat. Biotechnol. ./6:862-866, 1998).
  • Degradation-stabilized oligonucleotides may be encapsulated into liposomes and delivered to patients by injection either intravenously or directly into a target site (for example, the origin of neuropathic pain).
  • retroviral or adenoviral vectors, or naked DNA expressing anti-sense RNA for the inventive polypeptides may be administered to patients. Suitable techniques for use in such methods are well known in the art.
  • Bumetanide can be reacted thionyl chloride to make the corresponding acid chloride which can then be reacted with sodium hydrogen sulfide to give 3- aminosulfonyl-5-butylamino-4-phenoxythiobenzoic acid (thiobumetanide, S-bumetanide thioacid) by the methodology of Noble, P. and Tarbell, D. S., Ore. Synth., Coll. Vol. IV,
  • Bumetanide methyl ester can be reacted with hydrogen sulfide or sodium hydrogen sulfide to give, following acidification, 3-aminosulfonyl-5-butylamino-4- phenoxythiobenzoic acid (thiobumetanide, bumetanide thioacid).
  • bumetanide can be reacted with a catalytic amount of thionyl chloride in methanethiol (methyl mercaptan) to give thiomethyl 3- aminosulfonyl-5-butylamino-4-phenoxybenzoate.
  • bumetanide S-ethyl thioester Using similar methodology with bumetanide and the corresponding thiols, bumetanide S-ethyl thioester, bumetanide S- isoamyl thioester, bumetanide S-octyl thioester and bumetanide S-benzyl thioester, can be prepared.
  • bumetanide O-ethyl thioester Using similar methodology with dithiobumetanide and the corresponding alcohols, bumetanide O-ethyl thioester, bumetanide O-isoamyl thioester, bumetanide O- octyl thioester and bumetanide O-benzyl thioester, can be prepared.
  • dithiobumetanide can be reacted with a catalytic amount of thionyl chloride in methanethiol (methyl mercaptan) to give methyl 3- aminosulfonyl-5-butylammo-4-phenoxydithiobenzoate.
  • bumetanide ethyl dithioester, bumetanide isoamyl dithioester, bumetanide octyl dithioester and bumetanide benzyl dithioester can be prepared.
  • Bumetanide (l.Og, 2.7mmol) was dissolved in dimethylformamide (DMF) and chloroacetonitrile (195uL, 2.7mmol) was added followed by triethylamine (465uL). The reaction was heated to 100°C for 12 hours, TLC and liquid chromatography-coupled mass spectrometry (LC/MS) indicated the reaction was complete. The reaction was cooled to room temperature brought up in dichloromethane and washed with water, saturated ammonium chloride and reduced to a slurry. To the slurry was added water (25mL) and crude product precipitated as an off white solid.
  • DMF dimethylformamide
  • LC/MS liquid chromatography-coupled mass spectrometry
  • Bumetanide (1.2g, 3.29mmol) was dissolved in dimethylformamide (DMF, 12mL) and 4-(2-chloroethyl)morpholine hydrochloride (675mg, 3.62mmol) was added followed by triethylamine (ImL) and sodium iodide (500mg 3.33mmol).
  • DMF dimethylformamide
  • ImL triethylamine
  • sodium iodide 500mg 3.33mmol.
  • the reaction was heated to 95°C for 8 hours, TLC and LC/MS indicated the reaction was complete.
  • the reaction was cooled to room temperature brought up in dichloromethane and washed with water, saturated ammonium chloride and concentrated to dryness.
  • dithiobumetanide can be reacted with 3- (dimefhylamino)propyl chloride hydrochloride, triethylamine and sodium iodide in dimethylformamide (DMF) to yield 3-(N,N-dimethylaminopropyl 3-aminosulfonyl-5- butylamino-4-phenoxy-dithiobenzoate.
  • DMF dimethylformamide
  • Bumetanide (1.2g, 3.29mmol) was dissolved in dimethylformamide (12mL) and 2-chloro-N,N-diethylacetamide (500mg, 3.35mmol) was added followed by triethylamine
  • Bumetanide (1.2g, 3.29mmol) was dissolved in dimethylformamide (DMF, 1OmL) and 2-chloro-N,N-dimethylacetamide (41OuL, 3.9mmol) was added followed by triethylamine (0.7OmL) and sodium iodide (545mg, 3.6mmol). The reaction was heated to 50°C for 10 hours, TLC and LC/MS indicated the reaction was complete. The solvent was removed under reduced pressure and the residue was dissolved in ethyl acetate and washed with saturated sodium bicarbonate, water, and brine and dried over anhydrous magnesium sulfate.
  • DMF dimethylformamide
  • 2-chloro-N,N-dimethylacetamide 41OuL, 3.9mmol
  • sodium iodide 545mg, 3.6mmol
  • Bumetanide (1.2g, 3.29mmol) was dissolved in dimethylformamide (DMF, 1OmL) and chloromethyl pivalate (575uL, 3.9mmol) was added followed by triethylamine (0.7OmL) and sodium iodide (545mg, 3.6mmol). The reaction was heated to 50°C for 10 hours, TLC and LC/MS indicated the reaction was complete. The solvent was removed under reduced pressure and the residue was dissolved in ethyl acetate and washed with saturated sodium bicarbonate, water, and brine and dried over anhydrous magnesium sulfate.
  • dithiobumetanide can be reacted with chloromethyl pivalate, triethylamine and sodium iodide in dimethylformamide (DMF) to yield t-butylcarbonyloxymethyl 3-aminosulfonyl-5-butylamino-4-phenoxy-dithio- benzoate.
  • DMF dimethylformamide
  • bumetanide can be reacted with chloromethyl propionate, triethylamine and sodium iodide in dimethylformamide (DMF) to yield ethylcarbonyloxymethyl 3-aminosulfonyl-5-butylamino-4-pllenoxybenzoate.
  • DMF dimethylformamide
  • dithiobumetanide can be reacted with chloromethyl propionate, triethylamine and sodium iodide in dimethylformamide (DMF) to yield ethylcarbonyloxymethyl 3-aminosulfonyl-5-butylamino-4-phenoxy- dithiobenzoate.
  • DMF dimethylformamide
  • piretanide in similar manner to Example 1, piretanide can be reacted with thionyl chloride and methanol to yield methyl 3-aminosulfonyl-4- ⁇ henoxy-5-(l-pyrrolidinyl)benzoate.
  • piretanide ethyl ester piretanide isoamyl ester, piretanide octyl ester and piretanide benzyl ester can be prepared.
  • Piretanide can be reacted thionyl chloride to make the corresponding acid chloride which can then be reacted with sodium hydrogen sulfide to give 3-aminosulfonyl-4- phenoxy-5-(l-pyrrolidinyl)-thiobenzoic acid (thiopiretanide, S-piretanide thioacid) by the methodology of Noble, P. and Tarbell, D. S., Org. Synth., Coll. Vol. IV, John Wiley &
  • Piretanide methyl ester can be reacted with hydrogen sulfide or sodium hydrogen sulfide to give 3-aminosulfonyl-4-phenoxy-5-(l-pyrrolidinyl)-thiobenzoic acid (thiopiretanide, S-piretanide thioacid).
  • piretanide S-ethyl thioester piretanide S-isoamyl thioester
  • piretanide S-octyl thioester piretanide S-benzyl thioester
  • piretanide O- ethyl thioester piretanide O-isoamyl thioester
  • piretanide O-octyl thioester piretanide O-benzyl thioester
  • Thiopiretanide can be reacted thionyl chloride to make the corresponding thioacid chloride which can then be reacted with sodium hydrogen sulfide to give 3- aminosulfonyl-4-phenoxy-5-( 1 -pyrrolidinyl)-dithiobenzoic acid (dithiopiretanide, piretanide dithioacid) by the methodology of Noble, P. and Tarbell, D. S., Qrg. Synth.,
  • dithiopiretanide can be reacted with a catalytic amount of thionyl chloride in methanethiol (methyl mercaptan) to give methyl 3- ammosulfonyl-4-phenoxy-5-(l-pyrrolidinyl)-dithiobenzoate.
  • thionyl chloride methanethiol (methyl mercaptan)
  • methanethiol methyl mercaptan
  • piretanide ethyl dithioester piretanide isoamyl dithioester
  • piretanide octyl dithioester piretanide benzyl dithioester
  • piretanide can be reacted with chloroacetonitrile and triethylamine in DMF to yield cyanomethyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)benzoate.
  • piretanide can be reacted with benzyl chloride and triethylamine in DMF to yield benzyl 3-arninosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)benzoate.
  • piretanide can be reacted with 4-(2- chloroethyl)morpholine hydrochloride, triethylamine and sodium iodide in DMF to yield 2-(4-morpholino)ethyl 3-aminosulfonyl-4-phenoxy-5-(l-pyrrolidmyl)benzoate.
  • piretanide can be reacted with 3- (dimethylamino)propyl chloride hydrochloride, triethylamine and sodium iodide in dimethylformamide (DMF) to yield 3-(N,N-dimethylaminopropyl 3-aminosulfonyl-4- phenoxy-5-(l -pyrrolidmyl)benzoate.
  • DMF dimethylformamide
  • dithiopiretanide can be reacted with 3- (dimethylamino)pro ⁇ yl chloride hydrochloride, triethylamine and sodium iodide in dimethylformamide (DMF) to yield 3-(N,N-dimethylaminopropyl 3-aminosulfonyl-4- phenoxy-5-(l-pyrrolidinyl)dithiobenzoate.
  • DMF dimethylformamide
  • piretanide can be reacted with 2-chloro-N,N- diethylacetamide, triethylamine and sodium iodide in dimethylformamide (DMF) to yield N,N-diethylaminocarbonylmethyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)benzoate.
  • DMF dimethylformamide
  • piretanide can be reacted with EDC, HOBt and diethylamine in DMF to yield N,N-diethyl 3-aminosulfbnyl-4- ⁇ henoxy-5-(l- pyrrolidinyl)benzamide.
  • piretanide can be reacted with EDC, HOBt and dibenzylamine in DMF to yield N,N-dibenzyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)benzamide.
  • BenzvItrimethylammonium 3-Aminosulfonyl-4-phenoxy-5-(l-pyrrolidinyI)benzoate (Piretanide Benzylltrimethylammonium Salt)
  • piretanide can be reacted with benzyltrimethylammonium hydroxide to yield benzyltrimethylammonium 3- aminosulfonyl-4-phenoxy-5-( 1 -pyrrolidmyl)benzoate.
  • piretanide can be reacted with cetyltrimethylammonium hydroxide in water to yield cetyltrimethylar ⁇ monium 3- aminosulfonyl-4-phenoxy-5 -( 1 -pyrrolidinyl)benzoate.
  • piretanide can be reacted with chloromethyl pivalate, triethylamine and sodium iodide in DMF to yield t-butylcarbonyloxymethyl 3- aminosulfonyl-4-phenoxy-5-(l-pyrrolidinyl)benzoate.
  • dithiopiretanide can be reacted with chloromethyl pivalate, triethylamine and sodium iodide in DMF to yield t- butylcarbonyloxymethyl 3-aminosulfonyl-4-phenoxy-5-(l-pyrrolidinyl)dithiobenzoate.
  • piretanide can be reacted with chloromethyl propionate, triethylamine and sodium iodide in DMF to yield ethylcarbonyloxymethyl 3- aminosulfbnyl-4-phenoxy-5-( 1 -pyrrolidinyl)benzoate.
  • Furosemide can be reacted thionyl chloride to make the corresponding acid chloride which can then be reacted with sodium hydrogen sulfide to give 5- aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]thiobenzoic acid (thiofurosemide, S- furosemide thioacid) by the methodology of Noble, P. and Tarbell, D. S., Org. Synth., Coll. Vol. IV. John Wiley & Sons, Inc., New York, 1963, 924-927.
  • Furosemide methyl ester can be reacted with hydrogen sulfide or sodium hydrogen sulfide to give, following acidification, 3-aminosulfonyl-5-butyIamino-4- phenoxythiobenzoic acid (thiofurosemide, S-furosemide thioacid).
  • bumetanide can be reacted with a catalytic amount of thionyl chloride in methanethiol (methyl mercaptan) to 'give thiomethyl 5- aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]benzoate.
  • furosemide S-ethyl thioester furosemide S- isoamyl thioester, furosemide S-octyl thioester and furosemide S-benzyl thioester, can be prepared.
  • furosemide O-ethyl thioester Using similar methodology with dithio furosemide and the corresponding alcohols, furosemide O-ethyl thioester, furosemide O-isoamyl thioester, furosemide O- octyl thioester and furosemide O-benzyl thioester, can be prepared.
  • Thiofurosemide can be reacted thionyl chloride to make the corresponding thioacid chloride which can then be reacted with sodium hydrogen sulfide to give 5- aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]-dithiobenzoic aciid
  • dithiofurosemide can be reacted with a catalytic amount of tm ' onyl chloride in methanethiol (methyl mercaptan) to give methyl 5- aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]dithiobenzoate.
  • furosemide ethyl dithioester furosemide Ssoamyl dithioester, furosemide octyl dithioester and furosemide benzyl dithioester can be prepared.
  • furosemide can be reacted with chloroacetonitrile and triethylamine in DMF to yield cyanomethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]benzoate.
  • furosemide can be reacted with benzyl chloride and triethylamine in DMF to yield benzyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]benzoate.
  • Pharmaceutics 1990, 60, 163-169, can be employed to prepare 3 -(N 5 N- dimethylaminopropyl 5-aminosulfonyl-4-chloro-2-[(2-faranylmethyl)amino]benzoate, m.p. 212 - 213 0 .
  • dithiofurosemide can be reacted with 3- (dimethylamino)propyl chloride hydrochloride, triethylamine and sodium iodide in dimethylformarnide (DMF) to yield 3-(N,N-dimethylaminopropyl 5-aminosulfonyl-4- chloro-2-[(2-furanylmethyl)amino]dithiobenzoate.
  • DMF dimethylformarnide
  • dithiofurosemide can be reacted with EDC, HOBt and diethylamine in DMF to yield N,N-diethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]thiobenzarnide.
  • furosemide can be reacted with benzyltrimethylammonium hydroxide to yield benzyltrimethylammonium 5- aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]benzoate.
  • dithiofurosemide can be reacted with chloromethyl pivalate, triethylamine and sodium iodide in dimethylformamide (DMF) to yield t-butylcarbonyloxymethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino] dithiobenzoate.
  • DMF dimethylformamide
  • Pharmaceutics 1990, 60, 163-169, can be employed to prepare ethylcarbonyloxymethyl 5-aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]benzoate, m.p. 141 - 142°.
  • azosemide can be reacted with chloromethyl propionate, triethylamine and sodium iodide in DMF to yield 2-chloro-5-[l- (ethylcarbonyloxymethyl)- 1 H-tetrazol-5 -yl] -4- [(2- thienylmethyl)ammo]benzenesulfonamide.
  • Azosemide can be reacted with formaldehyde in methylene chloride, methylene chloride-DMF mixtures or DMF to yield 2-chloro-5-[l-(hydroxymethyl)-lH-tetrazol-5- yl]-4-[(2-thienylmethyl)amino]benzenesulfonamide.
  • Azosemide can be reacted with formaldehyde, methanol and a strong acid in methylene chloride, methylene chloride-DMF mixtures or DMF to yield 2-chloro-5-[l- (methoxymethyl)-lH-tetrazol-5-yl]-4-[(2-thienylmethyl)amino]benzenesulfonamide.
  • Azosemide can be reacted with formaldehyde, methanethiol and a strong acid in methylene chloride, methylene chloride-DMF mixtures or DMF to yield 2-chloro-5-[l- (methylthiomethyI)-lH-tetrazol-5-yl]-4-[(2-thienylmethyl)amino]benzenesulfonamide.
  • Azosemide can be reacted with benzyl chloromethyl ether, triethylamine and sodium iodide in DMF to yield 5-[l-(benzyloxymethyl)-lH-tetrazol-5-yl]- 2-chloro-4-[(2- thienylmethyl)amino]benzenesulfonamide.
  • azosemide can be reacted with benzyltrimethylammonium hydroxide in water to yield the benzyltrimethylammonium salt of 2 -chloro-5-( 1 H-tetrazol-5-yl)-4- [(2-thienylmethyl)amino]benzenesulfonamide .
  • torsemide can be reacted with chloromethyl pivalate, triethylamine and sodium iodide in DMF to yield 3-isopropylcarbamylsulfonamido-4-(3'- methylphenyl)ammopyridinium t-butylcarbonyloxymethochloride and some 3- isopropylcarbamylsulfonamido-4-(3'-r ⁇ ethylphenyl)aminopyridinium t-butylcarbonyloxymethoiodide .
  • EXAMPLE 75 EXAMPLE 75
  • torsemide can be reacted with chloromethyl propionate, triethylamine and sodium iodide in DMF to yield 3- isopropylcarbamylsulfonamido-4-(3'-methylphenyl)aminopyridinium ethylcarbonyloxymethochloride and some 3-isopropylcarbamylsulfbnamido-4-(3'- methylphenyl)aminopyridinium ethylcarbonyloxymethoiodide.
  • torsemide can be reacted with benzyl chloromethyl ether and triethylamine in DMF to yield 3-isopropylcarbarnylsulfonamido- 4-(3 '-methylphenyl)aminopyridinium benzyloxymethochloride.
  • torsemide can be reacted with methyl chloromethyl ether and triethylamine and in DMF to yield 3- isopropylcarbamylsulfonamido-4-(3'-methylphenyl)aminopyridinium methoxymethochloride.
  • torsemide can be reacted with benzyl chloromethyl thioether and triethylamine in DMF to yield 3- isopropylcarbamylsulfonamido-4-(3'-methylphenyl)aminopyridinium benzylthiamethochloride.
  • torsemide can be reacted with methyl chloromethyl thioether and triethylamine and in DMF to yield 3- isopropylcarbamylsulfonamido-4-(3'-methylphenyl)aminopyridinium methylthiamethochloride.
  • bumetanide can be reacted with MeO-PEG350- Cl (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n-1 -ethyl 3-aminosulfonyl-5-butylammo-4- phenoxybenzoate where n is in the 7-8 range.
  • MeO-PEG350- Cl Biolink Life Sciences, Inc., Cary, NC, BLS-106-350
  • thiobumetanide can be reacted with MeO- PEG350-C1 (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF to yield methoxyCporyethyleneoxy):,-! -ethyl 3-aminosulfonyl-5-butylamino-4- phenoxy-thiobenzoate where n is in the 7-8 range.
  • MeO- PEG350-C1 Biolink Life Sciences, Inc., Cary, NC, BLS-106-350
  • triethylamine in DMF to yield methoxyCporyethyleneoxy
  • bumetanide in a manner similar to Example 8, bumetanide can be reacted with MeO- PEGIOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamme in DMF to yield methoxy(polyethyleneoxy) n- i -ethyl 3-aminosulfonyI-5-butylamino-4- phenoxybenzoate where n is in the 19-24 range.
  • S-bumetanide mPEGlOOO thiosters can be formed with S-thiobumetanide, MeO-PEGlOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamine in DMF.
  • MethoxyfpolvethyIeneoxy)n ⁇ -ethy ⁇ 3-AminosulfonvL-5-butylammo-4- phenoxybenzoate (Bumetanide mPEGlOOO Dithioesters)
  • dithiobumetanide can be reacted with MeO- PEGIOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n- i -ethyl 3-aminosulfonyl-5-butylammo-4- phenoxy-dithiobenzoate where n is in the 19-24 range.
  • MeO- PEGIOOO-OTs Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000
  • piretanide can be reacted with MeO-PEG350-Cl (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n -i-ethyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)benzoate where n is in the 7-8 range.
  • bumetanide mPEG350 dithiosters can be formed with dithiobumetanide, MeO-PEG350-Cl (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF.
  • thiopiretanide can be reacted with MeO-PEG350- Cl (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n-1 -ethyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)thiobenzoate where n is in the 7-8 range.
  • MeO-PEG350- Cl Biolink Life Sciences, Inc., Cary, NC, BLS-106-350
  • piretanide can be reacted with MeO-PEGlOOO- OTs (Biolink Life Sciences, Inc., Cary, NC, BLS- 107- 1000) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n- i-ethyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)benzoate where n is in the 19-24 range.
  • MeO-PEGlOOO- OTs Biolink Life Sciences, Inc., Cary, NC, BLS- 107- 1000
  • S-piretanide mPEGlOOO thiosters can be formed with S-thiopiretanide, MeO-PEGlOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamine in DMF.
  • dithiopiretanide can be reacted with MeO- PEGIOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n- i -ethyl 3-aminosulfonyl-4-phenoxy-5-(l- pyrrolidinyl)dithiobenzoate where n is in the 19-24 range.
  • MeO- PEGIOOO-OTs Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000
  • piretanide mPEGlOOO dithiosters can be formed with dithiopiretanide, MeO-PEGlOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamine in DMF.
  • furosemide can be reacted with MeO-PEG350-Cl (Biolink Life Sciences, Inc., Cary, NC, BLS- 106-350) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n -i-ethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]benzoate where n is in the 7-8 range.
  • thiofurosemide can be reacted with MeO- PEG350-C1 (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n- i -ethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]thiobenzoate where n is in the 7-8 range.
  • MeO- PEG350-C1 Biolink Life Sciences, Inc., Cary, NC, BLS-106-350
  • furosemide can be reacted with MeO-PEGlOOO- OTs (Biolink Life Sciences, Inc., Cary, NC, BLS- 107- 1000) and triethylamine in DMF to yield methoxy(polyethyleneoxy) n-1 -ethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]benzoate where n is in the 19-24 range.
  • MeO-PEGlOOO- OTs Biolink Life Sciences, Inc., Cary, NC, BLS- 107- 1000
  • triethylamine in DMF to yield methoxy(polyethyleneoxy) n-1 -ethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]benzoate where n is in the 19-24 range.
  • dithiofurosemide can be reacted with MeO- PEGIOOO-OTs (Biolink Life Sciences, Inc., Cary, NC, BLS-107-1000) and triethylamine in DMF to yield methoxy(poryethyleneoxy) n- rethyl 5-aminosulfonyl-4-chloro-2-[(2- furanylmethyl)amino]dithiobenzoate where n is in the 19-24 range.
  • furosemide mPEG350 dithiosters can be formed with dithiofurosemide, MeO-PEG350-Cl (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF.
  • azosemide can be reacted with MeO-PEG350-Cl (Biolink Life Sciences, Inc., Cary, NC, BLS-106-350) and triethylamine in DMF to yield 5-[l-[methoxy(polyethyleneoxy) n -i-ethylJ-lH-tetrazol-5-yl]-2-chloro-4-[(2- thienylmethyl)amino]benzenesulfonamides where n is in the 7-8 range.
  • torsemide can be reacted with MeO-PEG350-Cl (Biolink Life Sciences, Inc., Gary, NC, BLS-106-350) and triethylamine in DMF to yield 3 -isopropylcarbamylsulfonamido-4-(3 ' -methylphenyl)aminopyridinium methoxy(polyethyleneoxy)n-i-ethochlorides where n is in the 7-8 range.
  • torsemide can be reacted with MeO-PEGlOOO- OTs (Biolink Life Sciences, Inc., Cary, NC, BLS- 107- 1000) and triethylamine in DMF to yield 3-isopropylcarbamylsulfonamido-4-(3 '-methylphenyl)aminopyridinium methoxy(polyethyleneoxy) n-1 -ethochlorides where n is in the 19-24 range.
  • MeO-PEGlOOO- OTs Biolink Life Sciences, Inc., Cary, NC, BLS- 107- 1000
  • bumetanide can be reacted with bis(4-methylpiperazinyl)aluminum hydride to yield 3-aminosulfonyl-5-butylamino-4-phenoxybenzaldehyde.
  • piretanide can be reacted with bis(4-methylpiperazinyl)aluminum hydride to yield 3-aminosulfonyl-4-phenoxy-5-(l-pyrrolidinyl)benzaldehyde.
  • furosemide can be reacted with bis(4-methylpiperazinyl)aluminum hydride to yield 5-aminosulfonyl-4-chloro-2-[(2-furanylmethyl)amino]benzaldehyde.
  • the slicing medium was a sucrose-based artificial cerebrospinal fluid (sACSF) consisting of 220 mM sucrose, 3 niM KCI, 1.25 itiM NaH 2 PO 4 , 2 mM MgSO 4 , 26 mM NaHCO 3 , 2 mM CaCl 2 , and 10 mM dextrose (295-305 mOsm).
  • sACSF sucrose-based artificial cerebrospinal fluid
  • a hemisphere of brain containing hippocampus was blocked and glued (cyanoacrylic adhesive) to the stage of a Vibroslicer (Frederick Haer, Brunsick, ME). Horizontal or transverse slices 400 ⁇ m thick were cut in 4° C, oxygenated (95% O 2 ; 5% CO 2 ) slicing medium.
  • the slices were immediately transferred to a holding chamber where they remained submerged in oxygenated bathing medium (ACSF) consisting of 124 mM NaCl, 3 mM KCl, 1.25 mM NaH 2 PO 4 , 2 mM MgSO 4 , 26 mM NaHCO 3 , 2 mM CaCl 2 , and 10 mM dextrose (295-305 mOsm).
  • ACSF oxygenated bathing medium
  • the slices were held at room temperature for at least 45 minutes before being transferred to a submersion-style recording chamber (all other experiments). In the recording chamber, the slices were perfused with oxygenated recording medium at 34-35° C. All animal procedures were conducted in accordance with NIH and University of Washington animal care guidelines.
  • Spontaneous interictal-like bursts were observed in slices treated by the following modifications or additions to the bathing medium: 10 mM potassium (6 slices; 4 animals; average - 81 bursts/min.); 200-300 ⁇ M 4-aminopyridine (4 slices; 2 animals; average — 33 burst/min.); 50-100 ⁇ M bicuculline (4 slices; 3 animals; average - 14 bursts/min); M Mg ++ (1 hour of perfusion - 3 slices; 2 animals; average - 20 bursts/min. or 3 hours of perfusion - 2 slices; 2 animals); zero calcium/6 mM KCI and 2 mM EGTA (4 slices; 3 animals).
  • the tissue was placed in a perfusion chamber located on the stage of an upright microscope and illuminated with a beam of white light (tungsten filament light and lens system; Dedo Inc.) directed through the microscope condenser.
  • the light was controlled and regulated (power supply - Lamda Inc.) to minimize fluctuations and filtered (695 nm longpass) so that the slice was transilluminated with long wavelengths (red).
  • Field of view and magnification were determined by the choice of microscope objectives (4X for monitoring the entire slice).
  • Image-frames were acquired with a charge-coupled device (CCD) camera (Dage MTI Inc.) at 30 HZ and were digitized at 8 bits with a spatial resolution of 512 x 480 pixels using an Imaging Technology Inc.
  • CCD charge-coupled device
  • Noise was defined as the maximum standard deviation of fluctuations of AR/R of the sequence of control images within a given acquisition series, where AR/R represented the magnitude of the change in light-transmission through the tissue.
  • Delta R/R was calculated by taking all the difference-images and dividing by the first control image: (subsequent image - first-control-image)/first-control-image. The noise was always ⁇ 0.01 for each of the chosen image sequences.
  • the absolute change in light transmission through the tissue was estimated during some experiments by acquiring images after placing neutral density filters between the camera and the light source. On average, the camera electronics and imaging system electronics amplified the signal 10- fold prior to digitization so that the peak absolute changes in light transmission through the tissue were usually between 1% and 2%.
  • Fig. ID is a video image of a typical hippocampal slice in the recording chamber.
  • the fine gold-wire mesh that was used to hold the tissue in place can be seen as dark lines running diagonally across the slice.
  • a stimulating electrode can be seen in the upper right on the stratum radiatum of CAl .
  • the recording electrode (too thin to be seen in the photo) was inserted at the point indicated by the white arrow.
  • Fig. IA illustrates that two seconds of stimulation at 60 Hz elicited after discharge activity and shows a typical after discharge episode recorded by the extracellular electrode.
  • the inset of Fig. IA shows the CAl field response to a single 200 sec test pulse (artifact at arrow) delivered to the Schaffer collaterals.
  • IAl shows a map of the peak change in optical transmission through the tissue evoked by Schaffer collateral stimulation.
  • the region of maximum optical change corresponds to the apical and basal dendritic regions of CAl on either side of the stimulating electrode.
  • Fig. IB illustrates sample traces showing responses to stimulation after 20 minutes of perfusion with medium containing 2.5 mM furosemide. Both the electrical after discharge activity (shown in Fig. IB) and the stimulation-evoked optical changes (shown in Fig. IBl) were blocked. However, there was a hyper-excitable field response (multiple population spikes) to the test pulse (inset).
  • Figs 1C and ICl illustrate that restoration of initial response patterns was seen after 45 minutes of perfusion with normal bathing medium.
  • EXAMPLE 101 The effects of furosemide on epileptiform discharges in hippocampal slices perfused with high-K + (10 mM) bathing medium
  • EXAMPLE 102 The effects of furosemide on epileptiform activity induced by i.v. injection of kainic acid in anesthetized rats
  • This example illustrates an in vivo model in which epileptiform activity was induced by i.v. injection of kainic acid (KA) into anesthetized rats (Lothman et al., Neurology 31:806, 1981). The results are illustrated in Figs. 3 A - 3H.
  • KA kainic acid
  • Sprague-Dawley rats (4 animals; weights 250-270 g) were anesthetized with urethane (1.25 g/kg i.p.) and anesthesia maintained by additional urethane injections (0.25 g/kg i.p.) as needed.
  • Body temperature was monitored using a rectal temperature probe and maintained at 35-37° C with a heating pad; heart rate (EKG) was continuously monitored.
  • the jugular vein was cannulated on one side for intravenous drug administration.
  • Rats were placed in a Kopf stereotaxic device (with the top of the skull level), and a bipolar stainless-steel microelectrode insulated to 0.5 mm of the tip was inserted to a depth of 0.5-1.2 mm from the cortical surface to record electroencephalographic (EEG) activity in the fronto-parietal cortex.
  • EEG electroencephalographic
  • a 2M NaCl-containing pipette was lowered to a depth of 2.5-3.0 mm to record hippocampal EEG.
  • Data were stored on VHS videotape and analyzed off-line. Following the surgical preparation and electrode placement, animals were allowed to recover for 30 minutes before the experiments were initiated with an injection of kainic acid (10-12 mg/kg i.v.).
  • Intense seizure activity an increased heart rate, and rapid movements of the vibrissae were induced with a latency of about 30 minutes. Once stable electrical seizure was evident, furosemide was delivered in 20 mg/kg boluses every 30 minutes to a total of 3 injections. Experiments were terminated with the intravenous administration of urethane. Animal care was in accordance with NIH guidelines and approved by the University of Washington Animal Care Committee.
  • Figs. 3A-3H show furosemide blockade of kainic acid-evoked electrical "status epilepticus" in urethane-anesthetized rats. EKG recordings are shown as the top traces and EEG recordings are shown as the bottom traces. In this model, intense electrical discharge (electrical "status epilepticus") was recorded from the cortex (or from depth hippocampal electrodes) 30-60 minutes after KA injection (10-12 mg/kg) (Figs. 3C and 3D). Control experiments (and previous reports, Lothman et al., Neurology, 31:806, 1981) showed that this status-like activity was maintained for well over 3 hours.
  • Hippocampal slices were prepared from Sprague-Dawley adult rats as described previously. Transverse hippocampal slices 100 ⁇ m thick were cut with a vibrating cutter.
  • Slices typically contained the entire hippocampus and subiculum. After cutting, slices were stored in an oxygenated holding chamber at room temperature for at least one hour before recording. AU recordings were acquired in an interface type chamber with oxygenated (95% O 2 , 5%CO 2 ) artificial cerebral spinal fluid (ACSF) at 34°-35°C. Normal ACSF contained (in mmol/1): 124 NaCl, 3 KCl, 1.25 NaH 2 PO 4 , 1.2 MgSO 4 , 26
  • normal or low-chloride medium was used containing bicuculline (20 ⁇ M), 4-amino pyridine (4-AP) (100 ⁇ M), or high-K + (7.5 or 12 mM).
  • low-chloride solutions (7, and 21 mM [C1 " ]O) were prepared by equimolar replacement of NaCl with Na + -gluconate (Sigma). All solutions were prepared so that they had a pH of approximately 7.4 and an osmolality of 290-300 mOsm at 35°C and at equilibrium from carboxygenation with 95%O 2 / 5%CO 2 .
  • the relative contributions of the factors that modulate synchronized activity vary between areas CAl and CA3. These factors include differences in the local circuitry and region-specific differences in cell packing and volume fraction of the extracellular spaces. If the anti-epileptic effects of anion or chloride-cotransport antagonism are due to a desynchronization in the timing of neuronal discharge, chloride-cotransport blockade might be expected to differentially affect areas CAl and CA3. To test this, a series of experiments was performed to characterize differences in the timing of the blockade of spontaneous epileptiform activity in areas CAl and CA3.
  • Example 103 Effect of chloride-cotransport antagonism on the synchronization of CAl and CA3 field population discharges
  • the observation from Example 103 suggested a temporal relationship between the exposure time to 1 OW-[CF]O or furosemide-containing medium and the characteristics of the spontaneous burst activity. Further, this relationship was different between areas CAl and C A3.
  • we compared the occurrences of CAl action potentials and the population spike events in the field responses of CAl and C A3 subfields during spontaneous and stimulation-evoked burst discharge.
  • Intracellular recordings were obtained from CAl pyramidal cells, with the intracellular electrode placed close ( ⁇ 100 ⁇ M) to the CAl field electrode. The slice was stimulated every 20 seconds with single stimuli delivered to the Schaffer collaterals. After continuous spontaneous bursting was established for at least 20 minutes, the bathing medium was switched to bicuculline-containing low-[Cl " ]O (21 mM) medium. After approximately 20 minutes, the burst frequency and amplitude was at its greatest. Simultaneous field and intracellular recordings during this time showed that the CAl field and intracellular recordings were closely synchronized with the CA3 field discharges. During each spontaneous discharge, the CA3 field response preceded the CAl discharge by several milliseconds.
  • the time interval during which these data were acquired corresponds to the time immediately prior to the cessation of spontaneous bursting in CAl. After 40-50 minutes perfusion with low-[Cl " ]O medium, the spontaneous bursts were nearly abolished in CAl but were unaffected in CA3.
  • Schaffer collateral stimulation during this time showed that monosynaptically-triggered responses of CAl pyramidal cells occurred without any significant increase in latency, but that stimulation- evoked field responses were almost abolished.
  • the time interval during which these data were acquired corresponds to the moments immediately prior to the cessation of spontaneous bursting in CA3.
  • [CT]O was reduced by equimolar replacement of NaCl with Na + -gluconate.
  • Gluconate was used rather than other anion replacements for several reasons.
  • patch-clamp studies have demonstrated that gluconate appears to be virtually impermeant to chloride channels, whereas other anions (including sulfate, isethionate, and acetate) are permeable to varying degrees.
  • Sprague-Dawley adult rats were prepared as previously described. Briefly, transverse hippocampal slices, 400 ⁇ m thick, were cut using a vibrating cutter. Slices typically contained the entire hippocampus and subiculum. After cutting, slices were stored in an oxygenated holding chamber for at least one hour prior to recording. All recordings were acquired in an interface type chamber with oxygenated (95% 02/5% CO 2 ) artificial cerebral spinal fluid (ACSF) at 34°-35°C. Normal ACSF contained (in mmol/1): 124 NaCl, 3 KCl, 1.25 NaH 2 PO 4 , 1.2 MgSO 4 , 26 NaHCO 3 , 2 CaCl 2 , and 10 dextrose.
  • normal or low-chloride medium was used containing bicuculline (20 ⁇ M), 4-AP (100 ⁇ M), or high-K + (12 mM).
  • Low-chloride solutions (7, 16, and 21 mM [CF]O) were prepared by equimolar replacement of NaCl with Na + - gluconate (Sigma Chemical Co., St. Louis, MO). All solutions were prepared so that they had a pH of approximately 7.4 and an osmolarity of 290-300 mOsm at 35°C and at equilibrium from carboxygenation with 95% O 2 / 5% CO 2 .
  • Sharp-electrodes filled with 4 M potassium acetate were used for intracellular recordings from CAl pyramidal cells.
  • Field recordings from the CAl or C A3 cell body layers were acquired with low-resistance glass electrodes filled with NaCl (2 M).
  • a small monopolar electrode was placed on the surface of the slice midway between areas CAl and C A3.
  • Spontaneous and stimulation-evoked activities from field and intracellular recordings were digitized (Neurocorder, Neurodata Instruments, New York, NY), and stored on video tape.
  • AxoScope software (Axon Instruments Inc.) on a PC-computer was used for off-line analyses of data.
  • Ion-selective microelectrodes were fabricated according to standard methods well known in the art. Double-barreled pipettes were pulled and broken to a tip diameter of approximately 3.0 ⁇ m.
  • the reference barrel was filled with ACSF and the other barrel was sylanized and the tip back-filled with a resin selective for K + (Corning 477317). The remainder of the sylanized barrel was filled with KCl (140 mM). Each barrel was led, via Ag/ AgCl wires, to a high impedance dual-differential amplifier (WPI FD223).
  • Each ion- selective microelectrode was calibrated by the use of solutions of known ionic composition and was considered suitable if it was characterized by a near-Nernstian slope response and if it remained stable throughout the duration of the experiment. After placement in the interface chamber, slices were superfused at approximately
  • the field and intracellular electrodes were always placed in close proximity to one another ( ⁇ 200 ⁇ m).
  • spontaneous bursting developed, first at the cellular level, and then in the field.
  • This spontaneous field activity representing synchronized burst discharge in a large population of neurons, lasted from 5 - 10 minutes, after which time the field recording became silent.
  • the field first became silent the cell continued to discharge spontaneously.
  • This result suggests that population activity has been "desynchronized" while the ability of individual cells to discharge has not been impaired.
  • intracellular recording showed that cells continued to discharge spontaneously even though the field remained silent.
  • the K + -selective microelectrode and a field electrode were placed in the CAl pyramidal layer close to one another ( ⁇ 200 ⁇ m), and a stimulation pulse was delivered to the Schaffer collaterals every 20 seconds so that the magnitude of the population spike could be monitored.
  • Multiple spontaneously occurring negative field shifts were evoked by perfusion with Iow-[C1O] (7 mM) medium.
  • Each event was associated with a significant increase in [K + ]O, with the [K + ]O increase starting several seconds prior to the onset of negative field shift.
  • a slow 1.5-2.0 mM increase in [K + ]O occurred over a time interval of approximately 1-2 minute seconds prior to the onset of each event.
  • the stimulation-evoked field responses slowly increased in amplitude over time, along with the increasing [K + ]O, until just before the negative field shift.
  • Sprague-Dawley adult rats were prepared as previously described. Transverse hippocampal slices, 400 ⁇ m thick, were cut with a vibrating cuter and stored in an oxygenated holding chamber for 1 hour before recording. A submersion-type chamber was used for K + -selective microelectrode recordings. Slices were perfused with oxygenated (95% O 2 /5% CO 2 ) artificial cerebrospinal fluid (ACSF) at 34-35°C.
  • oxygenated (95% O 2 /5% CO 2 ) artificial cerebrospinal fluid (ACSF) at 34-35°C.
  • Normal ACSF contained 10 mM dextrose, 124 niM NaCl, 3 mM KCl, 1.25 mM NaH 2 PO 4 , 1.2 mM MgSO 4 , 26 mM NaHCO 3 and 2 mM CaCl 2 .
  • normal or low- chloride medium was used containing 4-aminopyridine (4-AP) at lOO ⁇ M.
  • Low-chloride solutions 21 mM [Cl]o) were prepared by equimolar replacement of NaCl with Na+- gluconate (Sigma Chemical Co.).
  • K + selective microelectrodes were fabricated according to standard methods. Briefly, the reference barrel of a double-barreled pipette was filled with ACSF, and the other barrel was sylanized and the tip back-filled with KCl with K + -selective resin (Coming 477317). Ion-selective microelectrodes were calibrated and considered suitable if they had a Nernstian slope response and remained stable throughout the duration of the experiment.
  • Exposure of hippocampal slices to low-[Cl-]o medium has been shown to include a temporally-dependent sequence of changes on the activity of CAl pyramidal cells, with three characteristics phases, as described above.
  • exposure to low-[Cl-]o medium results in a brief period of increased hyperexcitability and spontaneous epileptiform discharge.
  • spontaneous epileptiform activity is blocked, but cellular hyperexcitability remains, and action potential firing times become less synchronized with one another.
  • the action potential firing times become sufficiently desynchronized so that stimulation-evoked field responses completely disappear, yet individual cells continue to show monosynapticlly- evoked responses to Schaffer collateral stimulation.
  • Antagonists of the anion/chloride-dependent cotransporter may affect extracellular pH transients that might contribute to the maintenance of synchronized population activity.
  • Rat hippocampal brain slices were prepared as described in Example 80, except the NaHCO 3 was substituted by equimolar amount of HEPES (26 nM) and an interface-type chamber was used.
  • FIG. 4 illustrates a schematic model of ion cotransport under conditions of reduced [Cl " ].
  • Fig. 4A left panel, shows that the chloride gradient necessary for the generation of IPSPs in neurons is maintained by efflux of ions through a furosemide- sensitive K ,Cl " cotransporter.
  • a high concentration of intracellular potassium (maintained by the 3Na + , 2K + -ATPase pump) serves as the driving force for the extrusion of Cl " against its concentration gradient.
  • the movement of ions through the furosemide-sensitive NKCC co-transporter is from extracellular to intracellular spaces.
  • transmembrane sodium cycle sodium ions taken into glial cells through NKCC cotransport are continuously extruded by the 3Na + ,2K + ,-ATPase pump so that a low intracellular sodium concentration is maintained.
  • the rate and direction of ion-flux through the furosemide-dependent cotransporters are functionally proportional to their ion-product differences written as [K + Ji x [Cl “ ] i - [K + ]o x [Cl “ ] o) for neuronal K + , Cl " cotransport and as [Na + ] i x [K + ]i x [Cl " ] 2 i - [Na + ] o x [K + Jo x [Cl " ] 2 o) for glial NKCC cotransport.
  • the sign of these ion- product differences show the direction of ion transport with positive being from intracellular to extracellular spaces.
  • Figure 4B shows a schematic phenomenological model that explains the emergence of the late-occurring spontaneous field events that arise as a result of prolonged low -[Cl " ]o exposure.
  • the ion-product differences for neurons and glia as QN and QG, respectively.
  • the differences of the ion- products for neurons are such that K + and Cl " are cotransported from intracellular to extracellular spaces (QN > 0);
  • the differences in ion-products for glial cells are such that Na + , K + and Cl " are cotransported from the ECS to intracellular compartments (QG ⁇ 0).
  • furosemide The ability of furosemide to alleviate pain will be examined in rodents using the Chung model of neuropathic pain (see, for example, Walker et al. MoI. Med. Today 5:319-321, 1999). Sixteen adult male Long-Evans rats will be used in this study. All rats will receive spinal ligation of the L5 nerve as detailed below. Eight of the sixteen rats will receive an injection (intravenous) of furosemide and the remaining eight will receive intravenous injection of vehicle only. Pain threshold will be assessed immediately using the mechanical paw withdrawal test. Differences in pain thresholds between the two groups will be compared. If furosemide alleviates pain, the group with the furosemide treatment will exhibit a higher pain threshold than the group that received vehicle.
  • the chamber is positioned on top of a mesh screen so that mechanical stimuli can be administered to the plantar surface of both hindpaws.
  • Mechanical threshold measurements for each hindpaw are obtained using an up/down method with eight von Frey monofilaments (5, 7, 13, 26, 43, 64, 106, and 202 niN). Each trial begins with a von Frey force of 13 mN delivered to the right hindpaw for approximately 1 sec, and then the left hindpaw. If there is no withdrawal response, the next higher force is delivered. If there is a response, the next lower force is delivered. This procedure is performed until no response is made at the highest force (202 mN) or until four stimuli are administered following the initial response.
  • Contextual fear conditioning involves pairing an aversive event, in this case moderate foot shock, with a distinctive environment.
  • the strength of the fear memory is assessed using freezing, a species-typical defensive reaction in rats, marked by complete immobility, except for breathing. If rats are placed into a distinctive environment and are immediately shocked they do not leam to fear the context. However, if they are allowed to explore the distinctive environment sometime before the immediate shock, they show intense anxiety and fear when placed back into the same environment.
  • mice received a 30 min period of habituation to the FPS apparatus. Twenty-four hours later, baseline startle amplitudes were collected. The rats were then divided into three matched groups based on baseline startle amplitudes. One of the rats exhibited a significantly higher baseline startle than the others and was excluded from the experiments. Groups 1 and 2 therefore consisted of 8 rats each, with Group 3 consisting of 7 rats. Following baseline startle amplitude collection, 20 light/shock pairings were delivered on two sessions over two consecutive days (i.e., 10 light/shock pairings per day).
  • Groups 2 and 3 received an injection (i.v.) of either furosemide (100 mg/kg) or bumetanide (70 mg/kg) in vehicle (DMSO) and Group 1 received vehicle alone.
  • DMSO vehicle
  • startle amplitudes were assessed during startle alone trials and startle plus fear (light followed by startle) trials.
  • Fear potentiated startle was compared between the treatment groups. Animals were trained and tested in four identical stabilimeter devices (Med-
  • each rat was placed in a small Plexiglas cylinder.
  • the floor of each stabilimeter consisted of four 6-mm-diameter stainless steel bars spaced 18 mm apart through which shock can be delivered. Cylinder movements result in displacement of an accelerometer where the resultant voltage is proportional to the velocity of the cage displacement. Startle amplitude is defined as the maximum accelerometer voltage that occurs during the first 0.25 sec after the startle stimulus is delivered.
  • the analog output of the accelerometer was amplified, digitized on a scale of 0-4096 units and stored on a microcomputer.
  • Each stabilimeter was enclosed in a ventilated, light-, and sound- attenuating box. All sound level measurements were made with a Precision Sound Level Meter.
  • the noise of a ventilating fan attached to a sidewall of each wooden box produced an overall background noise level of 64 dB.
  • the startle stimulus was a 50 ms burst of white noise (5 ms rise-decay time) generated by a white noise generator.
  • the visual conditioned stimulus employed was illumination of a light bulb adjacent to the white noise source.
  • the unconditioned stimulus was a 0.6 mA foot shock with duration of 0.5 sec, generated by four constant-current shockers located outside the chamber.
  • the presentation and sequencing of all stimuli were under the control of the microcomputer.
  • FPS procedures consisted of 5 days of testing; during days 1 and 2 baseline startle responses were collected, days 3 and 4 light/shock pairings were delivered, day 5 testing for fear potentiated startle was conducted.
  • Matching On the first two days all rats were placed in the Plexiglas cylinders and
  • Fig. 6 shows the baseline startle amplitudes for each group of rats determined prior to the test day.
  • Fig. 7 shows the amplitude of response on startle alone trials determined on the test day immediately following injection of either DMSO alone, bumetanide or furosemide, with Fig. 8 showing the difference score (startle alone - fear potentiated startle) on the test day.
  • a statistically significantly lower difference score was observed in rats treated with either furosemide or bumetanide than in rats treated with vehicle alone, indicating that both furosemide and bumetanide are effective in reducing anxiety.
  • Figs. 9 and 10 show the startle alone amplitude and the difference score, respectively, one week after treatment with either furosemide or bumetanide. Animals treated with either furosemide or bumetanide were found to have a higher difference score than animals treated with vehicle alone. However, as the error bars are so large for the vehicle-treated animals, the data does not imply any statistically significant difference between vehicle and bumetanide, with possibly a small difference between vehicle and furosemide.
  • Fig. 11 shows the percent difference score (startle alone - fear potentiated startle) on the test day in rats treated with one of the following bumetanide analogs: bumetanide
  • the percent difference score obtained after administration of most of the bumetanide analogs was significantly lower than that observed following administration of either vehicle alone or bumetanide, demonstrating that these analogs may be effectively employed to reduce anxiety.
  • several of the bumetanide analogs were observed to have significantly lower diuretic effects than those generally associated with either furosemide or bumetanide.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des procédés et des compositions pour traiter une douleur neuropathique et des troubles neuropsychiatriques, comprenant l'administration d'agents qui permettent de réduire la quantité efficace, d'inactiver et/ou d'inhiber l'activité d'un co-transporteur Na+-K+-2Cl- (NKCC). Dans certains modes de réalisation, le co-transporteur Na+-K+-2Cl- est NKCC1.
EP06826134A 2005-10-17 2006-10-17 Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance Withdrawn EP1937242A4 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US72731805P 2005-10-17 2005-10-17
US72756405P 2005-10-17 2005-10-17
US11/251,724 US8008283B2 (en) 1998-12-23 2005-10-17 Methods and compositions for the treatment of neuropsychiatric disorders
US11/549,274 US20070149526A1 (en) 2005-10-17 2006-10-13 Diuretic and diuretic-like compound analogs
PCT/US2006/040598 WO2007047698A2 (fr) 2005-10-17 2006-10-17 Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance

Publications (2)

Publication Number Publication Date
EP1937242A2 true EP1937242A2 (fr) 2008-07-02
EP1937242A4 EP1937242A4 (fr) 2011-06-15

Family

ID=37963225

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06826134A Withdrawn EP1937242A4 (fr) 2005-10-17 2006-10-17 Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance

Country Status (2)

Country Link
EP (1) EP1937242A4 (fr)
WO (1) WO2007047698A2 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017042314A1 (fr) * 2015-09-11 2017-03-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et composition pharmaceutique pour le traitement d'épilepsies post-traumatiques
US11666547B2 (en) * 2019-06-28 2023-06-06 Northwestern University Methods for treating cognitive deficits in a subject having schizophrenia, bipolar disorder, or psychiatric depression, and exhibiting cognitive deficits by administering an antagonist of the Na+-K+-2Cl-cation-chloride cotransporter isoform 1 (NKCC1)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020082252A1 (en) * 1998-12-23 2002-06-27 Cytoscan Sciences L.L.C. Methods and compositions for treating conditions of the central and peripheral nervous systems using non-synaptic mechanisms
WO2007047447A2 (fr) * 2005-10-17 2007-04-26 Neurotherapeutics Pharma, Inc. Analogues de compose diuretique ou de type diuretique

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5128327A (en) * 1991-03-25 1992-07-07 Merck & Co., Inc. Angiotensin II antagonists incorporating a nitrogen containing six membered ring heterocycle
US5162325A (en) * 1991-05-07 1992-11-10 Merck & Co., Inc. Angiotensin ii antagonists incorporating a substituted benzyl element

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020082252A1 (en) * 1998-12-23 2002-06-27 Cytoscan Sciences L.L.C. Methods and compositions for treating conditions of the central and peripheral nervous systems using non-synaptic mechanisms
WO2007047447A2 (fr) * 2005-10-17 2007-04-26 Neurotherapeutics Pharma, Inc. Analogues de compose diuretique ou de type diuretique

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
M. A. COLLINS ET AL.: "Brain damage due to episodic alcohol exposure in vivo and in vitro: furosemide neuroprotection implicates edema-based mechanism", THE FASEB JOURNAL, vol. 12, 1998, pages 221-230, XP002634773, *
See also references of WO2007047698A2 *

Also Published As

Publication number Publication date
WO2007047698A3 (fr) 2007-07-05
EP1937242A4 (fr) 2011-06-15
WO2007047698A2 (fr) 2007-04-26

Similar Documents

Publication Publication Date Title
US8008283B2 (en) Methods and compositions for the treatment of neuropsychiatric disorders
US8722668B2 (en) Methods and compositions for the treatment of neuropathic pain and neuropsychiatric disorders
US20060025387A1 (en) Compositions and methods for the treatment of disorders of the central and peripheral nervous systems
US20070149526A1 (en) Diuretic and diuretic-like compound analogs
TW518338B (en) Phosphinic acid amides as matrix metalloprotease inhibitors
US11033518B2 (en) Pharmaceutical composition containing niclosamide for treating axin-GSK3 interaction-related diseases
MX2011007759A (es) Analogos de bumetanida, furosemida, piretanida, azosemida y torsemida, composiciones y metodos de uso.
CN101820848B (zh) 抑制n-酰基乙醇胺水解性酸酰胺酶的组合物和方法
JP2011231094A (ja) ブメタニド、フロセミド、ピレタニド、アゾセミド、およびトルセミドのアナログ、組成物および使用方法
DE10295684T9 (de) Verfahren zum spezifischen Hemmen von Histon-Deacetylase-4
KR20170132333A (ko) 글루타미나제 억제제에 의한 폐암의 치료
KR20180059544A (ko) 글루타미나제 억제제와 면역-종양학적 약제에 의한 병용 요법
JP2009102330A (ja) 不安障害の治療のための方法および組成物
KR20050085542A (ko) 피라진계 튜불린 억제제
KR20180120746A (ko) 이카리인 및 이카리틴 유도체
JP2008535836A5 (fr)
AU2019200353B2 (en) Sigma-2 receptor ligand drug conjugates as antitumor compounds, methods of synthesis and uses thereof
EP1937242A2 (fr) Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance
US9670236B2 (en) Class of HDAC inhibitors expands the renal progenitor cells population and improves the rate of recovery from acute kidney injury
CA2625362A1 (fr) Procedes et compositions pour traiter des troubles neuropsychiatriques et des troubles lies a la dependance
CN111759827A (zh) 一种用于预防或治疗关节炎或炎性疾病的药物组合物
EA022196B1 (ru) СОЕДИНЕНИЯ 4-ЗАМЕЩЕННОГО-3-БЕНЗИЛОКСИБИЦИКЛО[3.1.0]ГЕКСАНА В КАЧЕСТВЕ АНТАГОНИСТОВ mGluR 2/3
CA2604446A1 (fr) Methodes et compositions pour le traitement de troubles anxieux
MX2008004951A (en) Methods and compositions for the treatment of neuropsychiatric and addictive disorders
WO2013090991A1 (fr) Thérapie par le tgf-bêta

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080331

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Free format text: PREVIOUS MAIN CLASS: A61K0031195000

Ipc: C07D0307520000

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 25/08 20060101ALI20110502BHEP

Ipc: A61K 31/41 20060101ALI20110502BHEP

Ipc: A61K 31/381 20060101ALI20110502BHEP

Ipc: A61K 31/341 20060101ALI20110502BHEP

Ipc: C07D 207/06 20060101ALI20110502BHEP

Ipc: C07C 311/00 20060101ALI20110502BHEP

Ipc: C07D 213/34 20060101ALI20110502BHEP

Ipc: C07D 257/04 20060101ALI20110502BHEP

Ipc: C07D 333/10 20060101ALI20110502BHEP

Ipc: C07D 307/52 20060101AFI20110502BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20110513

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20111213