US20160220515A1 - Method of reducing neuronal cell death with haloalkylamines - Google Patents

Method of reducing neuronal cell death with haloalkylamines Download PDF

Info

Publication number
US20160220515A1
US20160220515A1 US14/916,692 US201414916692A US2016220515A1 US 20160220515 A1 US20160220515 A1 US 20160220515A1 US 201414916692 A US201414916692 A US 201414916692A US 2016220515 A1 US2016220515 A1 US 2016220515A1
Authority
US
United States
Prior art keywords
haloalkylamine
tbi
hours
subject
phenoxybenzamine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/916,692
Other languages
English (en)
Inventor
David J. Poulsen
Thomas Frederick Rau
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.)
University of Montana
Original Assignee
University of Montana
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Montana filed Critical University of Montana
Priority to US14/916,692 priority Critical patent/US20160220515A1/en
Assigned to THE UNIVERSITY OF MONTANA reassignment THE UNIVERSITY OF MONTANA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: POULSEN, DAVID J., RAU, THOMAS FREDERICK
Publication of US20160220515A1 publication Critical patent/US20160220515A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/131Amines acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/14Ectoparasiticides, e.g. scabicides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • This application relates to pharmaceutical compositions comprising haloalkylamines, such as, phenoxybenzamine to treat a subject having a traumatic brain injury (TBI) and/or transient hypoxia/ischemia conditions in the central nervous system. Such conditions may lead to oxidative damage, apoptosis, or necrosis in neuronal cells.
  • TBI traumatic brain injury
  • the disclosed pharmaceutical compositions and methods reduce the occurrence of neuronal cell damage or death resulting from these conditions.
  • Stroke is clinically defined as a rapidly developing syndrome of vascular origin that manifests itself in focal loss of cerebral function. In more severe situations, the loss of cerebral function is global. Stroke can be categorized into two broad types, “ischemic stroke” (about 87%) and “hemorrhagic stroke” (about 10%). Ischemic stroke occurs when the blood supply to the brain is suddenly interrupted. Hemorrhagic stroke happens when a blood vessel located in or around the brain bursts leading to the leakage and accumulation of blood directly in the brain tissue. Additionally, a patient may experience transient ischemic attacks, which indicates a high risk for the future development of a more severe episode. Stroke also includes subarachnoid hemorrhage (about 3%).
  • stroke management is still not optimal.
  • Stroke is the third leading cause of death in the world, after only heart disease and cancer. In the United States alone, approximately 780,000 people experience a stroke each year. The cost of stroke in the US is over $43 billion, including both direct and indirect costs. The direct costs account for about 60% of the total amount and include hospital stays, physicians' fees, and rehabilitation. These costs normally reach $15,000/patient in the first three months; however, in approximately 10% of the cases, the costs are in excess of $35,000. Indirect costs account for the remaining portion and include lost productivity of the stroke victim, and lost productivity of family member caregivers (National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Md.).
  • the risk of stroke increases with age. After age 55, the risk of having a stroke doubles every decade, with approximately 40% of individuals in their 80's having strokes. Also, the risk of having a second stroke increases over time. The risk of having a second stroke is 25-40% five years after the first. As the baby boomer generation age, the total number of stroke incidents is projected to increase substantially. And with the over-65 years old portion of the population increasing as the baby boomers reach their golden years, the size of the market for stroke-related therapies will grow substantially. Also, the demand for an effective treatment will increase dramatically.
  • Traumatic brain injury is frequently caused by a sport- or recreation-related injury, and it is a national health concern in the United States.
  • CDC MMWR Weekly. Nonfatal Traumatic Brain Injuries from Sports and Adventure Activities, Jul. 27, 2007; 56(29); 733-737).
  • the highest rates of sports- and recreation-related TBI injuries are associated with males and females between the ages of 10-14 years.
  • TBIs are also common in military situations where brain damage may result from, for example, direct impact, penetrating objects such as bullets and shrapnel, and blast waves caused by explosions.
  • TBI Acute brain inflammation is most often implicated with a TBI, and the majority of TBIs are categorized as mild. However, even mild TBI can affect a person's ability to return to school or work and can result in long-term cognitive or other problems. In addition, repeated and/or severe TBIs can result in physical, cognitive, behavioral, or emotional problems and lead to various long-term, negative health effects, such as memory loss, behavioral changes, and increased risk for depression. As a result, prevention measures are desirable for TBIs. Of particular concern is the lack of effective treatments to reduce the primary or secondary phase of neuropathology induced by TBI.
  • TBI represents a heterologous injury. While it is clear there are significant differences between stroke and TBI, there are similarities in the mechanisms that lead to neuropathology. Both injuries induce the development of inflammation, reactive oxygen species (ROS), reactive nitrogen species (RNS), excitotoxicity, calcium dysregulation, and apoptosis. TBI also results in sheared blood vessels leading to impaired blood flow and ischemia.
  • ROS reactive oxygen species
  • RNS reactive nitrogen species
  • Preventative methods and pharmaceutical compositions are disclosed herein that inhibit or reduce damage and death in neuronal cells before it occurs rather than treating the damage afterwards to promote recovery.
  • the present invention relates to the treatment of a transient hypoxic and/or ischemic condition in the central nervous system with a haloalkylamine.
  • the invention provides a method of treating a transient hypoxic and/or ischemic condition in the central nervous system, the method comprising administering to a subject in need thereof a therapeutically effective amount of a haloalkylamine, such as, phenoxybenzamine or dibenamine.
  • administration of the haloalkylamine reduces the occurrence of neuronal brain cell death, for example in the striatum, hippocampus or the cortex of the subject caused by a transient hypoxic and/or ischemic condition or by a TBI event.
  • the invention provides a method of reducing the occurrence of neuronal cell death in the central nervous system.
  • the method typically consists of administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a haloalkylamine as at least one of the active ingredients.
  • the transient hypoxic and/or ischemic condition is often caused by low blood pressure, blood loss, a heart attack, a traumatic brain injury (TBI), a spinal cord injury (SCI), strangulation, surgery, a stroke, a spinal cord infarction, ischemic optic neuropathy, air-way blockage, or neonatal hypoxia or ischemia.
  • TBI traumatic brain injury
  • SCI spinal cord injury
  • strangulation surgery
  • surgery a stroke
  • a spinal cord infarction ischemic optic neuropathy
  • air-way blockage or neonatal hypoxia or ischemia.
  • the condition can be caused by many conditions, which can be more generally grouped as conditions that cause neuronal cell damage or death in the central nervous system due to the lack of oxygen and/or glucose reaching the neuronal cells for a temporary period of time.
  • the invention is directed to a method of treating a transient hypoxic and/or ischemic condition in the central nervous system caused by a TBI event, the method comprising administering to a subject in need thereof a therapeutically effective amount of a haloalkylamine.
  • TBI events include, for example, whiplash, a blast wave impact, and blunt force trauma, wherein the events are of sufficient force to cause neuronal cell damage or death.
  • the haloalkylamine is administered as an active ingredient with a pharmaceutically acceptable carrier.
  • the haloalkylamine may be administered with one or more additional active ingredients.
  • the haloalkylamine may also be in an extended release formulation.
  • the haloalkylamine is in unit dosage amounts of about 0.5 mg/kg body weight to about 5, 10, or 20 mg/kg body weight.
  • the haloalkylamine is administered within 24, 18, 16, 14, 12, 10, 8, 6, 4, or 2 hours after onset of the transient hypoxic and/or ischemic condition or the onset of a cause of the condition, for example, the onset of low blood pressure, blood loss, a heart attack, a TBI event, a SCI event, strangulation, surgery, a stroke, a spinal cord infarction, ischemic optic neuropathy, air-way blockage, or neonatal hypoxia or ischemia.
  • the haloalkylamine may be administered via an intravenous injection.
  • the invention relates to a pharmaceutical composition for treating a transient hypoxic and/or ischemic condition in the central nervous system, wherein the composition comprises a haloalkylamine as an active ingredient.
  • the pharmaceutical composition is for treating a transient hypoxic and/or ischemic condition in the central nervous system caused by a TBI event.
  • the pharmaceutical composition reduces the occurrence of neuronal cell death in the subject.
  • the pharmaceutical composition reduces the occurrence of neuronal cell death in neuronal cells of the striatum, hippocampus or the cortex of the subject.
  • the invention relates to a pharmaceutical composition for reducing the occurrence of neuronal cell death in the central nervous system.
  • neuronal cell death is caused by a transient hypoxic and/or ischemic condition.
  • neuronal cell death is caused by a TBI event.
  • the haloalkylamine is phenoxybenzamine, dibenamine or a combination thereof.
  • the pharmaceutical composition may comprise haloalkylamine, e.g., phenoxybenzamine, in unit dosage amounts of about 0.5 mg/kg body weight to about 5, 10, 15, or 20 mg/kg body weight.
  • the pharmaceutical composition may further comprise a pharmaceutically acceptable carrier. It may also be in an extended release formulation.
  • the pharmaceutical composition is administered within 24, 18, 16, 14, 12, 10, 8, 6, 4, or 2 hours after the onset of the transient hypoxic and/or ischemic condition or the onset of a cause of the condition, for example, the onset of low blood pressure, blood loss, a heart attack, a TBI event, a SCI event, strangulation, surgery, a stroke, a spinal cord infarction, ischemic optic neuropathy, air-way blockage, or neonatal hypoxia or ischemia.
  • the pharmaceutical composition is preferably administered via a parenteral or oral route, but other routes are contemplated and can be used depending on the condition.
  • FIG. 1 shows phenoxybenzamine provides significant neuroprotection from oxygen glucose deprivation in rat hippocampal slices cultures.
  • Representative fluorescent images of control slice cultures not exposed to OGD ( FIG. 1A ) or cultures exposed to OGD then treated with phenoxybenzamine at doses of 0.1 ⁇ M ( FIG. 1B ), 10 ⁇ M ( FIG. 1C ), 100 ⁇ M ( FIG. 1D ), 1 mM ( FIG. 1E ) or no treatment ( FIG. 1F ) are shown.
  • FIG. 1G shows neuronal loss presented as relative fluorescence intensity of propidium iodide within the CA1 (black bars), CA3 (white bars) and dentate gyrus (DG-grey bars).
  • FIG. 2 shows phenoxybenzamine provides significant neuroprotection when delivered up to 16 hours after OGD.
  • Representative fluorescent images of control slice cultures not exposed to OGD ( FIG. 2A ) or cultures exposed to OGD then treated with phenoxybenzamine at 2 hrs ( FIG. 2B ), 4 hrs ( FIG. 2C ), 8 hrs ( FIG. 2D ), or 16 hrs ( FIG. 2E ) after OGD or cultures that received no drug treatment ( FIG. 2F ) are shown.
  • FIG. 2G shows neuronal loss presented as relative fluorescence intensity of propidium iodide within the CA1 (black bars), CA3 (white bars) and dentate gyrus (DG-grey bars).
  • FIG. 3 shows phenoxybenzamine significantly improved behavioral function.
  • FIG. 3A depicts neurological severity score (NSS), and FIG. 3B depicts foot fault assessments over 30 days after TBI.
  • the dotted lines represent uninjured animals, the diamond-accented lines represent the saline-treated controls, and the circle-accented lines represent the animals treated with 1 mg/kg body weight phenoxybenzamine.
  • * represents p ⁇ 0.05, ** represents p ⁇ 0.01, and *** represents p ⁇ 0.001, all relative to saline controls.
  • the term “subject” or “patient” refers to any vertebrate including, without limitation, humans and other primates (e.g., chimpanzees and other apes and monkey species), farm animals (e.g., cattle, sheep, pigs, goats and horses), domestic mammals (e.g., dogs and cats), laboratory animals (e.g., rodents such as mice, rats, and guinea pigs), and birds (e.g., domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like).
  • the subject may be a mammal. In other implementations, the subject may be a human.
  • haloalkylamine is used herein to mean haloalkylamine a adrenergic blocking agents, which includes, for example, phenoxybenzamine, dibenamine, and related haloalkylamines and salts thereof.
  • related haloalkylamines include, but are not limited to compounds that have structural similarity to phenoxybenzamine and also share some of phenoxybenzamine's adrenergic-related effects. Such compounds are known to persons of skill in the art, e.g., Iversen et al. studied a total of 21 haloalkylamine derivatives, including phenoxybenzamine, for adrenergic-related effects (Iversen L. L. et al., Inhibition of catecholamine uptake in the isolated heart by haloalkylamines related to phenoxybenzamine, Br. J.
  • a “salt” refers to salts derived from a variety of organic and inorganic counter ions well known in the art and includes, by way of example only, sodium, potassium, calcium, magnesium, ammonium, and tetraalkylammonium.
  • its salt is made with the addition of acid salts of organic or inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic
  • haloalkylamines such as phenoxybenzamine may be used to treat transient hypoxia/ischemia conditions in the central nervous system. This treatment reduces the occurrence of neuronal cell damage or death resulting from transient hypoxia/ischemia conditions.
  • the haloalkylamines are unique a adrenergic receptor blocking agents; they form covalent (irreversible) bonds with a adrenergic receptors, thereby causing prolonged blockage of adrenergic transmission to the vasculature of the treated tissue or organ. Transmission is blocked until there is resynthesis of receptors in the vasculature, which appears to take several days, a week, or more.
  • phenoxybenzamine (Dibenzyline; Wellspring Pharmaceutical) is a haloalkylamine that blocks both ⁇ -1 and ⁇ -2 adrenergic receptors, but has a higher affinity for the ⁇ -1 receptor.
  • receptor antagonism achieves a peak effect at approximately 1 hour.
  • Phenoxybenzamine exerts a long-term effect in the brain with a half-life of approximately 24 hours. Previous research has shown that reversing the effect of phenoxybenzamine is dependent on the synthesis of new receptors and not the half-life of the drug. Hamilton et al.
  • the present invention is directed to a method of treating a transient hypoxic and/or ischemic condition in the central nervous system by administering to a subject in need thereof a therapeutically effective amount of a haloalkylamine.
  • the haloalkylamine may be phenoxybenzamine, dibenamine, or a related haloalkylamine.
  • phenoxybenzamine is one of a class of compounds known as haloalkylamine a adrenergic blocking agents
  • the use of other haloalkylamine a adrenergic blocking agents for the treatment of a transient hypoxic and/or ischemic condition in the central nervous system (CNS) is also within the scope of the present invention.
  • a TBI event may further be defined as any event in which the individual's normal activity level (basal functioning) is interrupted by an impact event.
  • TBI can be identified by a chart or device showing impact forces for different impact events, e.g., blast, car collision at 30 miles an hour, etc.
  • An example of a device for measuring impact force is a device worn by a soldier (e.g., helmet attachable) or part of a vehicle that can measure the pressure difference cause by a blast wave or blunt force impact, see for example U.S. patent application Ser. No. 12/154,166, entitled “Soft tissue impact assessment device and system,” which incorporated by reference herein.
  • a loss of consciousness is not required in order to find that a subject has suffered a TBI.
  • Significant research into the field of TBIs clearly demonstrates that TBI can cause neuronal cell damage or death even through the subject did not lose consciousness when he or she received the TBI.
  • a subject may be found to have suffered TBI without demonstrating a physical presentation of neurological symptoms depending on the source of the physical force or torsion.
  • a solider subject to concussive blast wave energy in the filed is preferably immediately identified and administered a low dose of the haloalkylamine. Any individual that has been exposed to a significant amount of physical force or torsion applied to the upper torso, neck, or head area would preferably be administered the haloalkylamine in an amount sufficient to reduce the occurrence of neuronal cell damage or death.
  • Traumatic forces may also damage the spinal cord indirectly, for example, damage induced by a blow to the head or a fall on the feet. Aside from the interruption from outside physical forces, bleeding, swelling, inflammation in and around the spinal cord subsequent to damage from traumatic forces may continue the interruption of the axons or nerve fibers of the spinal cord. For example, epidural hemorrhage and spinal subdural hematoma can result in progressive paraparesis due to pressure on the spinal cord.
  • Spinal cord injury may also be caused without traumatic forces.
  • arthritis, cancer, inflammation, infection, or disk degeneration of the spinal cord results in interruption in the axons and nerve fibers of the spinal cord.
  • Intramedullary injury can be the result of direct pressure on the cord or the passage of a pressure wave through the cord, laceration of the cord by bone, or the rupture of a blood vessel during the passage of a pressure wave through the cord with a hemorrhage into the cord.
  • Intramedullary bleeding and hematoma formation can also be caused by rupture of a weakened blood vessel.
  • the method may comprise the steps of identifying that a subject has a transient hypoxic/ischemic condition, or a TBI event, and, within 24 hours, 16 hours, 12 hours, 8 hours, 6 hours, 4 hours, or 2 hours of the onset of the condition, administering a haloalkylamine to the subject in an amount sufficient to treat the transient hypoxic and/or ischemic condition or TBI.
  • the haloalkylamine can be administered after a TBI event or SCI event even prior to the physical manifestation of neurological symptoms of neuronal cell damage or death. Slight to moderate TBI events have even been shown to induce neurological damage that may take months to manifest as physical symptoms. Therefore, in one embodiment, haloalkylamine is administered to a subject as quickly as possible after the TBI event or SCI event.
  • the method may comprises the steps of identifying that a subject suffered a TBI or SCI and, within 24 hours, 22 hours, 20 hours, 18 hours, 16 hours, 14 hours, 12 hours, 10 hours, 9 hours, 8 hours, 7 hours, 6 hours, 5 hours, 4 hours, 3 hours, 2 hours, or 1 hour of having received the injury, administering a haloalkylamine to the subject in an amount sufficient to treat the transient hypoxic and/or ischemic condition (e.g., to reduce the occurrence of neuronal cell damage or death caused by the TBI or SCI).
  • a haloalkylamine e.g., to reduce the occurrence of neuronal cell damage or death caused by the TBI or SCI.
  • the dose regimes disclosed herein are preferably used in this specific TBI and SCI embodiment as well.
  • the step of administering the haloalkylamine to the subject having a TBI event or SCI event comprises one or more intravenous injections of the haloalkylamine. It is also preferable that administration begins as soon as possible after the condition or event.
  • the methods of the invention advantageously typically reduce the occurrence of neuronal cell damage in the hippocampus, striatum, or cortex of the brain.
  • the haloalkylamine exerts a neuroprotective effect on neuronal cells by reduction of inflammation, antagonism of the ⁇ -1 and ⁇ -2 adrenergic receptors and blocking of norepinephrine signaling, and/or inhibition of calmodulin (CaM)/CaMKII activity. See Example 4.
  • the methods of the invention significantly reduce neurological and cognitive dysfunction.
  • the haloalkylamine is administered along with one or more additional ⁇ -blockers.
  • an “ ⁇ -blocker” is an agent that acts as a receptor antagonist of ⁇ -adrenergic receptors.
  • An ⁇ 1 -blocker acts against ⁇ 1 -adrenergic receptors, and an ⁇ 2 -blocker acts against ⁇ 2 -adrenergic receptors.
  • the haloalkylamine is administered with a non-selective ⁇ -blocker (e.g., phentolamine, tolazoline, trazodone), an ⁇ 1 -blocker (e.g., alfuzosin, prazosin, doxazosin, tamsulosin, terazosin, silodosin), and/or an ⁇ 2 -blocker (atipamezole, idazoxan, yohimbine).
  • a non-selective ⁇ -blocker e.g., phentolamine, tolazoline, trazodone
  • an ⁇ 1 -blocker e.g., alfuzosin, prazosin, doxazosin, tamsulosin, terazosin, silodosin
  • an ⁇ 2 -blocker atipamezole, idazoxan, y
  • the haloalkylamine is administered with one or more anti-inflammatory agents such as a non-steroidal anti-inflammatory drug (NSAID).
  • NSAID non-steroidal anti-inflammatory drug
  • the haloalkylamine is administered with a COX-2 inhibitor.
  • the COX-2 inhibitor may be rofecoxib, celecoxib, cimicoxib, valdecoxib, etoricoxib, parecoxib, lumiracoxib, or diclofenac.
  • the present invention is also directed to pharmaceutical compositions comprising a haloalkylamine as an active ingredient for the treatment of a transient hypoxic and/or ischemic condition in the central nervous system.
  • the transient hypoxic and/or ischemic condition may be caused by one or more of low blood pressure, blood loss, a heart attack, TBI, SCI, strangulation, surgery, a stroke, a spinal cord infarction, ischemic optic neuropathy, air-way blockage, or neonatal hypoxia or ischemia.
  • the pharmaceutical composition comprises a haloalkylamine, an NSAID, and a pharmaceutically acceptable carrier.
  • the NSAID may be a COX-2 inhibitor such as, for example, rofecoxib, celecoxib, cimicoxib, valdecoxib, etoricoxib, parecoxib, lumiracoxib, and diclofenac.
  • COX-2 inhibitor such as, for example, rofecoxib, celecoxib, cimicoxib, valdecoxib, etoricoxib, parecoxib, lumiracoxib, and diclofenac.
  • compositions of the present invention comprise a haloalkylamine as an active ingredient for reducing the occurrence of neuronal cell death in the central nervous system.
  • the occurrence of neuronal cell death may be caused by a transient hypoxic and/or ischemic condition.
  • the occurrence of neuronal cell death may also be caused by one or more of low blood pressure, blood loss, a heart attack, a TBI event, a SCI event, strangulation, surgery, a stroke, a spinal cord infarction, ischemic optic neuropathy, air-way blockage, or neonatal hypoxia or ischemia.
  • the pharmaceutical composition comprises a haloalkylamine, an NSAID, and a pharmaceutically acceptable carrier.
  • the NSAID may be a COX-2 inhibitor such as, for example, rofecoxib, celecoxib, cimicoxib, valdecoxib, etoricoxib, parecoxib, lumiracoxib, and diclofenac.
  • COX-2 inhibitor such as, for example, rofecoxib, celecoxib, cimicoxib, valdecoxib, etoricoxib, parecoxib, lumiracoxib, and diclofenac.
  • compositions and dosage forms of the invention can further comprise a pharmaceutically acceptable carrier.
  • the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which an active ingredient is administered.
  • Such pharmaceutical carriers can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • the pharmaceutical carriers can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like.
  • other excipients can be used.
  • Single unit dosage forms of the invention are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial), or transdermal administration to a patient.
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial
  • transdermal administration to a patient.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • the invention is preferably administered via a parenteral
  • the active ingredient may be administered in unit dosage amounts of about 0.1 mg/kg body weight to about 20 mg/kg body weight; e.g., any range within about 0.1 mg/kg body weight to about 5, 10, 15, or 20 mg/kg body weight such as 0.2 mg/kg body weight to about 5, 10, or 15 mg/kg body weight, about 0.5 mg/kg body weight to about 7.5 or 1o mg/kg body weight, about 0.5 mg/kg body weight to about 5 mg/kg body weight, about 1 mg/kg body weight to about 5 mg/kg body weight, about 2.5 mg/kg body weight to about 5 mg/kg body weight, etc.
  • the active ingredient is administered in a unit dosage amount of about 0.5 mg/kg body weight to about 5 mg/kg body weight.
  • the active ingredient may be administered in a unit dosage amount of less than about 20, 15, or 10 mg/kg body weight, less than about 9 mg/kg body weight, less than about 8 mg/kg body weight, less than about 7 mg/kg body weight, less than about 6 mg/kg body weight, less than about 5 mg/kg body weight, less than about 4 mg/kg body weight, less than about 3 mg/kg body weight, less than about 2 mg/kg body weight, or less than about 1 mg/kg body weight. In one implementation, the active ingredient is administered in a unit dosage amount of less than about 5 mg/kg body weight.
  • the active ingredient may be administered in unit dosage amounts of about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, or about 100 mg.
  • These unit dosages may be administered once, twice, or three times daily.
  • the active ingredient is administered within 24 hours, 22 hours, 20 hours, 18 hours, 16 hours, 14 hours, 12 hours, 10 hours, 9 hours, 8 hours, 7 hours, 6 hours, 5 hours, 4 hours, 3 hours, 2 hours, or 1 hour of the onset of the transient hypoxic and/or ischemic condition in the central nervous system.
  • the active ingredient is administered within 24 hours, 22 hours, 20 hours, 18 hours, 16 hours, 14 hours, 12 hours, 10 hours, 9 hours, 8 hours, 7 hours, 6 hours, 5 hours, 4 hours, 3 hours, 2 hours, or 1 hour of the onset low blood pressure, blood loss, a heart attack, a TBI event, a SCI event, strangulation, surgery, a stroke, a spinal cord infarction, ischemic optic neuropathy, or air-way blockage.
  • the active ingredient in administered continuously, for example by a continuous IV infusion.
  • the active ingredient e.g., the haloalkylamine
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can be an immediate or extended release formulation depending on the condition and likelihood of reoccurrence.
  • the pharmaceutical composition for treating a transient hypoxic condition may be different than that for treating a transient ischemic condition.
  • the pharmaceutical composition for treating a transient hypoxic and/or ischemic condition may also vary based on the causes for the condition, for example if the condition were caused by strangulation compared to if the condition were caused by stroke.
  • composition, shape, and type of dosage forms of the invention will typically vary depending on their route of administration and subject being treated.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease.
  • Typical pharmaceutical compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient.
  • oral dosage forms such as tablets may contain excipients not suited for use in parenteral dosage forms.
  • the suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • the dosage is determined empirically, using known methods, and will depend upon facts such as the biological activity of the particular compound employed, the means of administrations, the age, health and body weight of the host; the nature and extent of the symptoms; the frequency of treatment; the administration of other therapies and the effect desired.
  • the dosage is determined empirically, using known methods, and will depend upon facts such as the biological activity of the particular compound employed, the means of administrations, the age, health and body weight of the host; the nature and extent of the symptoms; the frequency of treatment; the administration of other therapies and the effect desired.
  • the actual dosages and method of administration or delivery may be determined by one of skill in the art. Frequency of dosage may also vary depending on the compound used and whether an extended release formulation is used. However, for treatment of most disorders, a single dose is preferred.
  • compositions of the invention that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990).
  • Typical oral dosage forms of the invention are prepared by combining the active ingredients in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, Natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101, AVICEL-PH-103 AVICEL RC-581, AVICEL-PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pa.), and mixtures thereof.
  • An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103 and Starch 1500 LM.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions of the invention is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Disintegrants are used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention.
  • the amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, preferably from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g., peanut oil, cottonseed oil
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Plano, Tex.), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • AEROSIL 200 a syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Plano, Tex.
  • CAB-O-SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.
  • a preferred solid oral dosage form of the invention comprises an active ingredient, anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
  • Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous, bolus injection, intramuscular, and intraarterial.
  • the parenteral dosage form is suitable for intravenous delivery.
  • the parenteral dosage forms of the invention are preferably sterile or capable of being sterilized prior to administration to a patient.
  • Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art. Examples include, but are not limited to: water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • water for Injection USP water for Injection USP
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride
  • a rat model was used.
  • Male Wistar rats (350-500 g) were obtained from Charles River Laboratories (Wilmington, Mass.) and housed with a 12-hour light/dark cycle and ad libitum access to food and water.
  • Severe TBI was induced using the lateral fluid percussion (LFP) procedure as previously described (Rau et al., 2012, J Trauma and Acute Care Surgery 73:S165). Briefly, a 5 mm trephination was made over the right hemisphere equidistant from the lambda and the bregma Animals were given a fluid pulse to the brain at 1.9-2.3 atm of pressure for 20 msec.
  • LFP lateral fluid percussion
  • mice experienced apnea and were manually ventilated until normal breathing occurred. Animals had an average righting time of 24 minutes and a 25% mortality rate was observed. At 8 hours post TBI, phenoxybenzamine (1 mg/kg) was injected into the tail vein of randomly selected rats. Saline treated animals underwent the same tail vein injection procedure receiving only pre-warmed saline. Sham operated animals received a trephination but did not receive a TBI.
  • Phenoxybenzamine Prevents Neuronal Cell Death in Rat Hippocampal Slice Culture-Oxygen Glucose Deprivation
  • Hippocampal slice cultures were prepared from the brains of 7-day-old Sprague-Dawley rat pups as previously described (Selkirk et al., 2005, Eur J Neur 21:2291). After 7 days in culture, slices were exposed to oxygen-glucose deprivation (OGD).
  • OGD oxygen-glucose deprivation
  • a glucose free balanced salt solution composed of 120 mM NaCl, 5 mM KCl, 1.25 mM NaH 2 PO 4 , 2 mM MgSO 4 , 2 mM CaCl 2 , 25 mM NaHCO 3 , 20 mM HEPES, 25 mM sucrose; pH 7.3 was bubbled for one hour with 5% CO 2 /95% N 2 at 10 L/hour.
  • Slices were washed 6 times in deoxygenated SBSS to remove residual glucose, transferred into deoxygenated SBSS, and placed in a 37° C. chamber (Pro-Ox) with an oxygen feedback sensor that maintained gas levels at 0.1% O 2 , 5% CO 2 , 94.4% Nitrogen for 60 min.
  • Propidium iodide (PI) was added to the media at a concentration of 2 ⁇ M (Noraberg et al., 1999, Brain Research Protocols 3:278), 4 hours prior to OGD. Images were taken of the hippocampal slices prior to OGD to establish baseline fluorescence. After OGD, slices were placed in normal media containing 2 ⁇ M PI and imaged again at 24 hours post-OGD using fluorescence optics with an Olympus IMT-2 microscope and a Hamamatsu camera. The total fluorescent intensity in each slice was determined using ImagePro Plus software (Media Cybernetics, Silver Springs, Md.) and all values were expressed as the percent change from untreated slices exposed to OGD.
  • ImagePro Plus software Media Cybernetics, Silver Springs, Md.
  • a middle dose (100 ⁇ M) was selected and added to the cultures at 2, 4, 8, or 16 hours post-OGD.
  • phenoxybenzamine prevented neuronal death from OGD in all regions of the hippocampus when delivered at 2, 4, and 8 hours post-OGD.
  • phenoxybenzamine prevented neuronal death only in the CA1 region of the hippocampus ( FIG. 2 ).
  • Neurological severity scoring was performed as previously described (Rau et al., 2011, Neuropharmacology 61:677; Rau et al., 2012 J Trauma and Acute Care Surgery 73:S165). Assessments were conducted on days 1, 7, 14, 21, and 30 by a blinded observer. Animals were scored from 0-16 with 0 indicating no impairment, and 16 indicating maximal impairment. Scoring criteria for a severe TBI was 16-10, a moderate TBI was 9-5 and a mild TBI was 4-1. Animals scoring and NSS of 9 or less on day 1 were identified as having a moderate/mild injury and were excluded.
  • Foot fault assessments were conducted as previously described (Rau et al., 2011, Neuropharmacology 61:677; Rau et al., 2012 J Trauma and Acute Care Surgery 73:S165). Briefly, rats were set on an elevated grid. With each weight-bearing step, the paw may fall or slip off the wire grid. Each time the left forelimb (affected by damage to the right hemisphere) missed a placement on the wire rack it was recorded as a foot fault. The total number of steps (movement of each forelimb) that the rat used to cross the grid was counted, and the total numbers of foot faults for each forelimb was recorded.
  • rat lateral fluid percussion injury LFP
  • phenoxybenzamine rat lateral fluid percussion injury
  • NSS Neurological severity score
  • foot fault assessments were used to test the hypothesis that phenoxybenzamine treatment could improve behavioral outcomes. Animals were assessed 24 hours after injury and again on 7, 14, 21, and 30 days post-TBI. We found no significant differences in NSS or foot fault scoring between the saline and phenoxybenzamine treated animals at 24 hours or 7 days after the TBI. These data indicate that all animals in both treatment groups experienced injuries of similar severity. However, phenoxybenzamine treated animals showed significant improvements in NSS and foot fault scoring on days 14, 21, and 30 ( FIG. 3 ). It is worth noting that phenoxybenzamine treated rats had foot fault values similar to uninjured rats on days 21 and 30 of testing.
  • the Morris water maze (MWM) was used to assess the impact of phenoxybenzamine on cognitive function (learning and memory) following TBI.
  • the assessment procedure was performed as previously published (Rau et al., 2011, Neuropharmacology 61:677; Rau et al., 2012 J Trauma and Acute Care Surgery 73:S165). Pre-acclimation began on day 24 post-TBI.
  • the training phase began on day 25 post-injury, and the probe trial was conducted on day 30 post-injury.
  • the Morris water maze (MWM) was used to assess cognitive function beginning 25 days after injury.
  • Administration of phenoxybenzamine 8 hours after TBI resulted in a significant improvement in learning on days 2, 3, 4, and 5 of the training phase ( FIG. 4A ).
  • phenoxybenzamine treated animals were not significantly different from the un-injured sham control animals on any of the training days. These data suggest that phenoxybenzamine dramatically improves learning after severe TBI.
  • a probe trial was conducted to assess spatial memory function. During the probe trial the phenoxybenzamine treated animals displayed significantly greater spatial memory capacity than the saline treated controls ( FIG. 4B ).
  • the phenoxybenzamine treated animals spent approximately 28% of their time searching the target quadrant for the removed escape platform compared to saline treated animals which only 10% of the time in the target quadrant.
  • the phenoxybenzamine treated TBI injured animals did not differ from un-injured sham controls, which also spent approximately 25% of their time searching the target quadrant ( FIG. 4B ).
  • Severe TBI increases the activity of the sympathetic nervous system resulting in the excessive release of epinephrine and norepinephrine (Tran et al., 2008).
  • Previous research indicates a direct correlation between the severity of TBI, plasma epinephrine and norepinephrine levels, and recovery rates (Tran et al., 2008).
  • Patients remaining in a persistent coma have epinephrine and norepinephrine plasma levels several-fold higher than controls. Furthermore, these catecholamine levels remain elevated for the duration of the comatose state.
  • CaM/CaMKII Another potential mechanism of neuroprotection associated with phenoxybenzamine is a reduction in calmodulin (CaM)/CaMKII activity.
  • Phenoxybenzamine is a potent inhibitor of CaM/CaMKII activity (Cimino and Weiss, 1988).
  • CaMKII is a major mediator of glutamate signaling, however, under acute injury conditions, CaM/CaMKII interacts with the NR2B subunit of NMDA receptors leading to excitotoxic death (Vest et al., 2010).
  • CaM/CaMKII increases the trafficking of AMPA receptors to the cell surface leading to greater excitotoxic death during acute injury.
  • Zhang et al found that TBI increased the expression of CaMKII ⁇ . Pre-treating rats with a CaMKII ⁇ inhibitor before TBI resulted in a significant decrease in lesion volume and a significant increase in neuromotor function. Zhang et al went on to elucidate a mechanism in which CaMKII actively promotes apoptosis in neurons by increasing the pro-apoptotic protein BAX and subsequent caspase 3 activation (Zhang et al., 2012).
  • phenoxybenzamine appears to block critical gene changes that occur after TBI.
  • Genes involved in inflammation such as CCl2, IL-1 ⁇ , and MyD88 were all significantly elevated in TBI animals, but phenoxybenzamine treated animals did not differ from uninjured controls. This is a crucial finding because inflammation contributes to the formation of edema, a loss of neurons, and negatively affects patient recovery.
  • a key component of brain inflammation is the recruitment of neutrophils and monocytes, which are toxic to neurons (Semple et al., 2010). Recruitment of monocytes into the brain is primarily controlled by monocyte chemotactic protein 1 (MCP-1) otherwise known as CCl2, which is expressed by astrocytes, macrophages, and reactive microglia.
  • MCP-1 monocyte chemotactic protein 1
  • CCl2 actively recruits monocytes to areas of brain damage leading to inflammation, edema and neuronal damage (Ziebell and Morganti-Kossmann, 2012).
  • Rhodes et al (2009) found a rapid increase in CCl2 in human spinal fluid following severe TBI. Furthermore, CCl2 levels remained significantly elevated for up to 10 days post-injury.
  • Rhodes et al. (2009) also reported that increased levels of CCl2 were detected in the serum of patients who died after TBI.
  • IL-1 ⁇ interleukin-1 ⁇
  • IL-1 ⁇ further contributes to immunoexcitoxicty by enhancing the sensitivity of NMDA receptors and tumor necrosis factor alpha (TNF-alpha) leading to increased brain inflammation and excitotoxicity (Arand et al., 2001; Block et al., 2007; Brown and Neher, 2010).
  • TNF-alpha tumor necrosis factor alpha
  • Phenoxybenzamine may further reduce post-traumatic inflammation by reducing expression of myeloid differentiation primary response protein 88 (Myd88).
  • Myd88 is a key adaptor protein involved in Toll-like receptor and pro-inflammatory cytokine signal transduction (Li et al., 2011; Ling et al., 2013).
  • Myd88 was significantly upregulated over uninjured controls.
  • MyD88 expression levels were equivalent to uninjured controls in phenoxybenzamine treated rats after TBI.
US14/916,692 2013-09-06 2014-09-08 Method of reducing neuronal cell death with haloalkylamines Abandoned US20160220515A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/916,692 US20160220515A1 (en) 2013-09-06 2014-09-08 Method of reducing neuronal cell death with haloalkylamines

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361874865P 2013-09-06 2013-09-06
US14/916,692 US20160220515A1 (en) 2013-09-06 2014-09-08 Method of reducing neuronal cell death with haloalkylamines
PCT/US2014/054569 WO2015035308A2 (en) 2013-09-06 2014-09-08 Method of reducing neuronal cell death with haloalkylamines

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/054569 A-371-Of-International WO2015035308A2 (en) 2013-09-06 2014-09-08 Method of reducing neuronal cell death with haloalkylamines

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/148,825 Continuation US10849865B2 (en) 2013-09-06 2018-10-01 Method of reducing neuronal cell death with haloalkylamines

Publications (1)

Publication Number Publication Date
US20160220515A1 true US20160220515A1 (en) 2016-08-04

Family

ID=52629091

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/916,692 Abandoned US20160220515A1 (en) 2013-09-06 2014-09-08 Method of reducing neuronal cell death with haloalkylamines
US16/148,825 Active US10849865B2 (en) 2013-09-06 2018-10-01 Method of reducing neuronal cell death with haloalkylamines
US17/082,161 Abandoned US20210093586A1 (en) 2013-09-06 2020-10-28 Method of reducing neuronal cell death with haloalkylamines

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/148,825 Active US10849865B2 (en) 2013-09-06 2018-10-01 Method of reducing neuronal cell death with haloalkylamines
US17/082,161 Abandoned US20210093586A1 (en) 2013-09-06 2020-10-28 Method of reducing neuronal cell death with haloalkylamines

Country Status (7)

Country Link
US (3) US20160220515A1 (es)
EP (1) EP3041465B1 (es)
JP (3) JP6475733B2 (es)
CN (2) CN110251496A (es)
CA (1) CA2923476C (es)
MX (1) MX2016002655A (es)
WO (1) WO2015035308A2 (es)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10849865B2 (en) 2013-09-06 2020-12-01 The University Of Montana Method of reducing neuronal cell death with haloalkylamines

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6009875A (en) 1993-10-14 2000-01-04 Berlex Laboratories, Inc. Use of sympathetic antagonists for treatment of chronic muscle pain
US5513661A (en) 1993-10-14 1996-05-07 Hubbard; David R. Use of sympathetic blockade for treatment of chronic muscle pain
US5898035A (en) 1995-08-29 1999-04-27 New York Medical College Formulations of haloalkylamines and local anesthetic and methods for the treatment of reflex sympathetic dystrophy (RSD)
US6284763B1 (en) 1998-08-26 2001-09-04 Queen's University At Kingston Methods for remodeling neuronal and cardiovascular pathways
ATE480246T1 (de) 2000-07-19 2010-09-15 Univ Northwest Transport von nukleinsäuresubstanzen
US6730667B2 (en) 2001-11-26 2004-05-04 William R. Deagle Iontophoresis disc pain blocker
US7172614B2 (en) * 2002-06-27 2007-02-06 Advanced Cardiovascular Systems, Inc. Support structures for embolic filtering devices
AU2003272719A1 (en) * 2002-09-26 2004-04-19 Carbomer, Inc. Inhibitors of the nitrix oxide synthase iii (nos iii) as neuroprotective agents
US7600349B2 (en) * 2003-02-26 2009-10-13 Unirac, Inc. Low profile mounting system
CA2588017A1 (en) * 2004-12-13 2006-06-22 Boehringer Ingelheim Pharmaceuticals, Inc. Use of alpha-adrenergic blockers for the treatment of dysmenorrhea
WO2007057508A2 (en) 2005-11-18 2007-05-24 Orion Corporation Treatment of pain with a combination of an alpha2 -adrenoceptor antagonist such as atipemezole or fipamezoiie and an opioid receptor agonist, such as tramadol
WO2007062862A2 (en) 2005-12-02 2007-06-07 Ludwig Maximilians Universität München Use of calmodulin inhibitors for the treatment of neurodegenerative disorders
US20070249725A1 (en) 2006-02-17 2007-10-25 Hubbard David R Pharmaceutical formulations of sympathetic antagonist for the treatment of chronic muscle pain
US20110105621A1 (en) * 2006-08-23 2011-05-05 The University Of Montana Method of reducing brain cell damage, inflammation or death
EP2053919B1 (en) 2006-08-23 2013-12-25 The University Of Montana Method of reducing neuronal cell damage
US20090197969A1 (en) * 2006-08-23 2009-08-06 Poulsen David J Method of reducing brain cell damage or death
WO2008091555A2 (en) 2007-01-22 2008-07-31 Gtx, Inc. Nuclear receptor binding agents
US8292830B2 (en) 2007-05-22 2012-10-23 University Of Louisville Research Foundation, Inc. Soft tissue impact assessment device and system
US20110053913A1 (en) 2009-06-25 2011-03-03 Khem Jhamandas Methods and Therapies for Alleviating Pain
US20130266663A1 (en) * 2010-04-30 2013-10-10 Arthur Brown Sox9 inhibitors
WO2012074561A2 (en) 2010-11-30 2012-06-07 Columbia Northwest Pharmaceuticals Llc Methods and compositions for the treatment of anxiety disorders, including post traumatic stress disorder (ptsd) and related central nervous system (cns) disorders
EP2466311A1 (en) 2010-12-17 2012-06-20 Roche Diagnostics GmbH sFlt1 in patients with ischemic stroke
CA2768796A1 (en) 2011-02-22 2012-08-22 Wellspring Pharmaceutical Corporation Phenoxybenzamine assay
WO2012162364A1 (en) * 2011-05-23 2012-11-29 New York Medical College Phenoxybenzamine for pain
US20140371322A1 (en) 2013-06-13 2014-12-18 Professional Compounding Centers Of America Phenoxybenzamine Transdermal Composition
CN110251496A (zh) 2013-09-06 2019-09-20 蒙大拿大学 用卤代烷基胺减少神经元细胞死亡的方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Gustafson et al. Journal of Cerebral Blood Flow and Metabolism 1989, 9, 171-174 *
Hamilton et al. Naunyn-Schmiedeberg’s Arch Pharmacol 1984, 325, 34-41 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10849865B2 (en) 2013-09-06 2020-12-01 The University Of Montana Method of reducing neuronal cell death with haloalkylamines

Also Published As

Publication number Publication date
JP6865806B2 (ja) 2021-04-28
CA2923476A1 (en) 2015-03-12
US10849865B2 (en) 2020-12-01
CA2923476C (en) 2021-03-16
WO2015035308A3 (en) 2015-10-29
US20190083423A1 (en) 2019-03-21
US20210093586A1 (en) 2021-04-01
EP3041465B1 (en) 2020-11-11
JP2016529320A (ja) 2016-09-23
JP2019077718A (ja) 2019-05-23
WO2015035308A2 (en) 2015-03-12
JP6636660B2 (ja) 2020-01-29
MX2016002655A (es) 2016-06-06
JP6475733B2 (ja) 2019-02-27
CN105530924A (zh) 2016-04-27
EP3041465A4 (en) 2017-03-08
EP3041465A2 (en) 2016-07-13
JP2020059746A (ja) 2020-04-16
CN110251496A (zh) 2019-09-20

Similar Documents

Publication Publication Date Title
Sikiric et al. Brain-gut axis and pentadecapeptide BPC 157: Theoretical and practical implications
IL194987A (en) Use of PIF peptide to produce a drug to regulate the immune system
TW201038544A (en) Anti-neurodegenerative diseases agents
JP2021526507A (ja) 発作により誘発される突然死を処置するための組成物および方法
KR101494675B1 (ko) 신경 세포 손상을 감소시키는 방법
JP2017526647A (ja) Ang−(1−7)誘導体オリゴペプチド、ならびにそれを使用および作製するための方法
JP2022504944A (ja) Nad+またはnad+前駆体と組み合わせたsarm1の阻害
US20160008336A1 (en) Hdac inhibitors for the treatment of traumatic brain injury
US20210093586A1 (en) Method of reducing neuronal cell death with haloalkylamines
US20220280474A1 (en) Kv1.3 antagonists for use in the treatment of chronic and acute pain
US20210236594A1 (en) Methods for improving frailty and aging
JP2009531423A (ja) 神経親和性、神経調節性、脳血管作用性および抗発作活性を呈する薬剤
US20220110953A1 (en) Methods and compositions for treating human papillomavirus (hpv)-induced cancers
ES2943585T3 (es) Remielinización estimulada por IgG de nervios periféricos
JP2017109987A (ja) ErbB4+炎症性マクロファージによって媒介される疾患の治療方法
CN113069458A (zh) 哌唑嗪用于制备治疗和/或预防脑血管疾病药物中的应用
ES2808667T3 (es) Derivados de diarilmetilideno piperidina y su uso como agonistas del receptor opioide delta
CN110882240A (zh) 作为急性缺血性中风的治疗剂的多酚衍生物化合物6-cepn
KR20150119439A (ko) 통증 치료를 위한 na(v) 1.9 채널 활성의 억제제 및 이의 용도
CN113712965B (zh) N、n、n′、n′-四基(2-苯甲基)-乙酰胺在制备治疗uc的药物中的应用
US11306122B2 (en) Compositions and methods of inhibiting the binding of plasma IgG autoantibodies to serotonin 2A receptor
JP2018173307A (ja) 末梢神経又は脊髄に存在する膜に特異的に結合して鎮痛作用を有する候補化合物のスクリーニング方法
Rosa Exploring the effect of NLRP3 Inflammasome inhibition in the ex vivo model of epileptogenesis
Korimová et al. Protective effects of glucosamine-kynurenic acid after compression-induced spinal cord injury in the rat
WO2019211783A1 (en) Indoleamine 2,3-dioxygenase signalling modulator and therapeutic use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNIVERSITY OF MONTANA, MONTANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:POULSEN, DAVID J.;RAU, THOMAS FREDERICK;REEL/FRAME:038114/0424

Effective date: 20140911

STCB Information on status: application discontinuation

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