US20090221544A1 - Methods for the treatment of a traumatic central nervous system injury via a tapered administration protocol - Google Patents

Methods for the treatment of a traumatic central nervous system injury via a tapered administration protocol Download PDF

Info

Publication number
US20090221544A1
US20090221544A1 US11/909,276 US90927606A US2009221544A1 US 20090221544 A1 US20090221544 A1 US 20090221544A1 US 90927606 A US90927606 A US 90927606A US 2009221544 A1 US2009221544 A1 US 2009221544A1
Authority
US
United States
Prior art keywords
progesterone
progestin
injury
administration
animals
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/909,276
Other languages
English (en)
Inventor
Donald G. Stein
Sarah Cutler
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.)
Emory University
Original Assignee
Emory University
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 Emory University filed Critical Emory University
Priority to US11/909,276 priority Critical patent/US20090221544A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT EXECUTIVE ORDER 9424, CONFIRMATORY LICENSE Assignors: EMORY UNIVERSITY
Assigned to EMORY UNIVERSITY reassignment EMORY UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CUTLER, SARAH, STEIN, DONALD G.
Publication of US20090221544A1 publication Critical patent/US20090221544A1/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
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the invention relates to methods for treating a traumatic or ischemic injury to the central nervous system.
  • progesterone As a gonadal steroid, progesterone also belongs to a family of autocrine/paracrine hormones called neurosteroids. Neurosteroids are steroids that accumulate in the brain independently of endocrine sources and which can be synthesized from sterol precursors in glial cells. These neurosteroids can potentiate GABA transmission, modulate the effects of glutamate, enhance the production of myelin, reduce the expression of inflammatory cytokines and prevent release of free radicals from activated microglia.
  • progesterone's neuroprotective effects in injured nervous systems. For example, following a contusion injury, progesterone reduces the severity of post injury cerebral edema. The attenuation of edema by progesterone is accompanied by the sparing of neurons from secondary neuronal death and improvements in cognitive outcome (Roof et al. (1994) Experimental Neurology 129:64-69). Furthermore, following ischemic injury in rats, progesterone has been shown to reduce cell damage and neurological deficit (Jiang et al. (1996) Brain Research 735:101-107). Progesterone's protective effects may be mediated thorough its interaction with GABA and/or glutamate receptors as well as its effects on inflammatory cytokines and aquaporin expression which are mediated by the intranuclear progesterone receptor.
  • progesterone metabolites have also been suggested to have neuroprotective properties.
  • progesterone metabolites allopregnanolone or epipregnanolone are positive modulators of the GABA receptor, increasing the effects of GABA in a manner that is independent of the benzodiazepines (Baulieu, E. E. (1992) Adv. Biochem. Psychopharmacol. 47:1-16; Robel et al. (1995) Crit. Rev. Neurobiol. 9:383-94; Lambert et al. (1995) Trends Pharmacol. Sci. 16:295-303; Baulieu, E. E. (1997) Recent Prog. Horm. Res. 52:1-32; Reddy et al.
  • neurosteroids act as antagonists at the sigma receptor: a receptor that can activate the NMDA channel complex (Maurice et al. (1998) Neuroscience 83:413-28; Maurice et al. (1996) J. Neurosci. Res. 46:734-43; Reddy et al. (1998) Neuroreport 9:3069-73). These neurosteroids have also been shown to reduce the stimulation of cholinergic neurons and the subsequent release of acetylcholine by excitability.
  • a cascade of physiological events leads to neuronal loss including, for example, an inflammatory immune response and excitotoxicity resulting from the initial impact disrupting the glutamate, acetylcholine, cholinergic, GABA A , and NMDA receptor systems.
  • the traumatic CNS injury is frequently followed by brain and/or spinal cord edema that enhances the cascade of injury and leads to further secondary cell death and increased patient mortality.
  • CNS injury occurs when blood flow to the CNS is interrupted.
  • consumed cellular ATP usually cannot be adequately replenished in the absence of a supply of oxygen.
  • ischemic CNS injury Other physiological events associated with ischemic CNS injury include release or overexpression of proteins such as neuron-specific enolase (NSE), myelin basic protein, glial fibrillary acidic protein (GFAP), the S-100 protein, and the gamma isoform of protein kinase C (PKCg), stimulation of membrane phospholipid degradation and subsequent free-fatty-acid accumulation, cellular acidosis, glutamate release and excitotoxicity, calcium ion influx, and free radical generation.
  • NSE neuron-specific enolase
  • GFAP glial fibrillary acidic protein
  • PLCg protein kinase C
  • the present invention provides a method of administration of a therapeutically effective amount of a progestin or progestin metabolite following a traumatic or ischemic injury to the CNS such that, prior to termination of the progestin or progestin metabolite administration is tapered to avoid withdrawal.
  • the drug taper employed can involve a linear taper, an exponential taper, progressively dividing administered doses by 50%, or can be determined based on the treating physician's assessment of the patient's response to therapy.
  • the tapered administration methods of the present invention may be used in combination with any therapeutic protocol or regimen for the administration of a therapeutically effective amount of a progestin or progestin metabolite to treat a traumatic or ischemic CNS injury.
  • FIG. 1 shows a dosage response curve for behavioral recovery following a traumatic brain injury.
  • FIGS. 1A and 113 demonstrate that following treatment with low (8 mg/kg), moderate (16 mg/kg), and high (32 mg/kg) doses of progesterone in a cyclodextrin-containing carrier, both low and moderate doses of progesterone produced consistent improvement in Morris water maze performance.
  • FIG. 2 shows the results from the “sticker removal task” following treatment with low (8 mg/kg), moderate (16 mg/kg), and high (32 mg/kg) dosages of progesterone in a cyclodextrin-containing carrier.
  • FIG. 3 shows somatosensory neglect data at one day ( FIG. 3A ) and one week ( FIG. 3B ) post-withdrawal.
  • FIG. 3A shows that at one day post-withdrawal, TWL animals showed decreased latency compared to VL and AWL animals (#, p ⁇ 0.05).
  • AWS rats demonstrated elevated sensory deficiencies compared to the TWS and VS groups (*, p ⁇ 0.05).
  • FIG. 3B shows that at one week post-withdrawal, sham animals demonstrated equivalent latency, while tapered treatment maintained decreased latency compared to acute and vehicle treatment (#, p ⁇ 0.05).
  • FIG. 4 shows center time, as determined from Digiscan Locomoter Activity Boxes, between one FIG. 4A ) and seven ( FIG. 4B ) days post-withdrawal.
  • FIG. 4A shows that one day after withdrawal, center time was increased for TWS animals compared to all other shams (#, p ⁇ 0.05), while TWL center time was increased compared to other lesion groups (**, p ⁇ 0.05).
  • AWL animals increased center time compared to vehicle animals (##, p ⁇ 0.05), and AWS animals significantly decreased center time compared to VS animals (*, p ⁇ 0.05).
  • FIG. 4B showed that TWL center time one week after withdrawal is increased over AWL (**, p ⁇ 0.05), which is increased over VL (##, p ⁇ 0.05). No difference was seen between sham groups.
  • FIG. 5 shows p53 Western blotting densitometry between experimental groups demonstrating an increase in apoptotic activity for VL animals over all other treatment groups (*, p ⁇ 0.05).
  • FIG. 6 shows HSP70 Western blotting densitometry between experimental groups demonstrating an increase for TWL animals over all other groups (*, p ⁇ 0.05).
  • FIG. 7 shows BDNF Western blot densitometry between experimental groups demonstrating an increase for TWL animals over all other groups (*, p ⁇ 0.05), followed by AWL (#, p ⁇ 0.05). VL BDNF levels were comparable to shams.
  • FIG. 8 shows representative images of selected sections anterior to bregma ( FIG. 8A ), and quantified data for each lesion group ( FIG. 8B ).
  • FIG. 8A shows representative thionin-stained sections at mm anterior to bregma for lesion animals.
  • FIG. 8B shows percent lesion volume at 3 weeks post-injury is greatest in vehicle-treated animals, followed by those with acute withdrawal (*, p ⁇ 0.05) and tapered withdrawal (#, p ⁇ 0.05).
  • FIG. 9 shows relative reactive astrocytes as determined by immunofluorescent GFAP staining at three weeks post-injury.
  • FIG. 9A shows immunofluorescent staining for GFAP in brain slices from the following groups: (A) VL; (B) AWL; (C) TWL; (D) VS; (E) AWS; and (F) TWS. Images are shown at 40 ⁇ , with 10 ⁇ m represented.
  • FIG. 9B shows quantification of luminosity for GFAP immunofluorescent staining of reactive astrocytes indicates the greatest response in VL (*, p ⁇ 0.05) animals, followed by AWL (**, p ⁇ 0.05) and TWL animals. AWS animals had significantly elevated levels of reactive astrocytes compared to other sham groups m (#, p ⁇ 0.05).
  • the present invention provides methods and compositions for the treatment or prevention of neurodegeneration following a traumatic or ischemic injury to the central nervous system.
  • the methods of the invention provide for the administration of a therapeutically effective amount of a progestin or progestin metabolite following a traumatic or ischemic injury to the CNS such that, prior to termination of administration of the progestin or progestin metabolite the administration is tapered to avoid withdrawal.
  • the present invention demonstrates that the tapered administration allows for a more beneficial CNS repair than when an abrupt termination of the progestin or progestin metabolite occurs.
  • treatment is intended any improvement in the subject having the traumatic or ischemic injury including both improved morphological (i.e., enhanced tissue viability) and/or behavioral recovery.
  • the improvement can be characterized as an increase in either the rate and/or the extent of behavioral and anatomical recovery following the traumatic or ischemic CNS injury.
  • a “positive therapeutic response” induces both a complete response and a partial response.
  • Various methods to determine if a complete or partial therapeutic response has occurred are disclosed elsewhere herein.
  • Neurodegeneration is the progressive loss of neurons in the central nervous system.
  • “neuroprotection” is the arrest and/or reverse progression of neurodegeneration following a traumatic or ischemic central nervous system injury.
  • the methods of the invention also find use in reducing and/or preventing the physiological events leading to neurodegeneration.
  • the present invention provides methods for reducing or eliminating neuronal cell death, edema, ischemia, and enhancing tissue viability following a traumatic or ischemic injury to the central nervous system.
  • the sex hormones are steroids that may be classified into functional groups according to chemical structure and physiological activity and include estrogenic hormones, progestational hormones, and androgenic hormones.
  • progestational hormones referred to herein as “progestins” or “progestogens”, and their derivatives and bioactive metabolites.
  • progestins or progestogens
  • steroid hormones disclosed in Remington's Pharmaceutical Sciences , Gennaro et al., Mack Publishing Co. (18 th ed. 1990), 990-993.
  • sterioisomerism is of fundamental importance with the sex hormones.
  • progestins i.e., progesterone
  • their derivatives are encompassed by the present invention, including both synthetic and natural products.
  • the progestin or progestin metabolite is progesterone.
  • progesterone refers to a member of the progestin family and comprises a 21 carbon steroid hormone. Progesterone is also known as D4-pregnene-3,20-dione; ⁇ 4-pregnene-3,20-dione; or pregn-4-ene-3,20-dione and it its structure is provided below as formula (I).
  • the progesterone used in the methods of the invention can be naturally occurring or synthetic.
  • synthetic progestins are a molecule whose structure is related to that of progesterone, is synthetically derived, and retains the biologically activity of progesterone (i.e., treats a traumatic CNS injury).
  • Representative synthetic progestin include, but are not limited to, modifications that produce 17a-OH esters (i.e., 17 ⁇ -hydroxyprogesterone caproate), as well as, modifications that introduce 6 ⁇ -methyl, 6-Me, 6-ene, and 6-chloro substituents onto progesterone (i.e., medroxyprogesterone acetate, megestrol acetate, and chlomadinone acetate).
  • Table 1 provides further, non-limiting examples, of synthetic progestins.
  • progestin any naturally or synthetically produced progestin that prevents or retards neurodegeneration.
  • progestin derivatives include, for example, derivatives of progesterone, such as 5-dehydroprogesterone, 6-dehydro-retroprogesterone (dydrogesterone), allopregnanolone (allopregnan-3 ⁇ , or 3 ⁇ -ol-20-one), ethynodiol diacetate, hydroxyprogesterone caproate (pregn-4-ene-3,20-dione, 17-(1-oxohexy)oxy); levonorgestrel, norethindrone, norethindrone acetate (19-norpregn-4-en-20-yn-3-one, 17-(acetyloxy)-,(17 ⁇ )-); norethynodrel, norgestrel, pregneno
  • Useful progestins also can include allopregnone-3 ⁇ or 3 ⁇ , 20 ⁇ or 20 ⁇ -diol (see Merck Index 258-261); allopregnane-3 ⁇ ,21-diol-11,20-dione; allopregnane-3 ⁇ ,17 ⁇ -diol-20-one; 3,20-allopregnanedione, allopregnane, 3 ⁇ ,11 ⁇ ,17 ⁇ ,20 ⁇ ,21-pentol; allopregnane-3 ⁇ ,17 ⁇ ,20 ⁇ ,21-tetrol; allopregnane-3 ⁇ or 3 ⁇ ,11 ⁇ ,17 ⁇ ,21-tetrol-20-one, allopregnane-3 ⁇ ,17 ⁇ or 20 ⁇ -triol; allopregnane-3 ⁇ ,17 ⁇ ,21-triol-11,20-dione; allopregnane-3 ⁇ ,11 ⁇ ,21-triol-20-one; allopregnane-3 ⁇ ,17 ⁇ ,21-triol-20-one; all
  • progestin derivatives include esters with non-toxic organic acids such as acetic acid, benzoic acid, maleic acid, malic acid, caproic acid, citric acid and the like.
  • Inorganic salts include, for example, hydrochloride, sulfate, nitrate, bicarbonate and carbonate salts.
  • compounds that may find use in the present invention include the progestin derivatives that are disclosed in U.S. Pat. No. 5,232,917, herein incorporated by reference.
  • the progestin or progestin metabolite may be administered per se or in the form of a pharmaceutically acceptable salt.
  • the salts of the progestin or progestin metabolite should be both pharmacologically and pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare the free active compound or pharmaceutically acceptable salts thereof and are not excluded from the scope of this invention.
  • Such pharmacologically and pharmaceutically acceptable salts can be prepared by reaction of a progestin or progestin metabolite with an organic or inorganic acid, using standard methods detailed in the literature.
  • Examples of pharmaceutically acceptable salts are organic acids salts formed from a physiologically acceptable anion, such as, tosglate, methenesulfurate, acetate, citrate, malonate, tartarate, succinate, benzoate, etc.
  • Inorganic acid salts can be formed from, for example, hydrochloride, sulfate, nitrate, bicarbonate and carbonate salts.
  • pharmaceutically acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium, or calcium salts of the carboxylic acid group.
  • a traumatic injury to the CNS is characterized by a physical impact to the central nervous system.
  • a traumatic brain injury results when the brain is subjected to a physical force that results in progressive neuronal cell damage and/or cell death.
  • a traumatic brain injury may result from a blow to the head and manifests as either an open or closed injury.
  • blast injuries are caused by the complex pressure wave generated by an explosion, and can include closed injuries such as concussion without external signs of head trauma.
  • Other physical forces that may act on the brain include increased intracranial pressure due to, for example, subarachnoid or intracranial hemorrhage, tumor growth, ventriculomegaly, or cerebral edema.
  • Severe brain damage can occur from lacerations, skull fractures, and conversely, even in the absence of external signs of head injury.
  • the physical forces resulting in a traumatic brain injury cause their effects by inducing three types of injury: skull fracture, parenchymal injury, and vascular injury.
  • Parenchymal injuries include concussion, direct parenchymal injury and diffuse axonal injury.
  • Concussions are characterized as a clinical syndrome of alteration of consciousness secondary to head injury typically resulting from a change in the momentum of the head (movement of the head arrested against a ridged surface).
  • the pathogenesis of sudden disruption of nervous activity is unknown, but the biochemical and physiological abnormalities that occur include, for example, depolarization due to excitatory amino acid-mediated ionic fluxes across cell membranes, depletion of mitochondrial adenosine triphosphate, and alteration in vascular permeability.
  • Postconcussive syndrome may show evidence of direct parenchymal injury, but in some cases there is no evidence of damage.
  • Contusion and lacerations are conditions in which direct parenchymal injury of the brain has occurred, either through transmission of kinetic energy to the brain and bruising analogous to what is seen in soft tissue (contusion) or by penetration of an object and tearing of tissue (laceration).
  • a blow to the surface of the brain leads to rapid tissue displacement, disruption of vascular channels, and subsequent hemorrhage, tissue injury and edema.
  • Morphological evidence of injury in the neuronal cell body includes pyknosis of nucleus, eosinophilia of the cytoplasm, and disintegration of the cell.
  • axonal swelling can develop in the vicinity of damage neurons and also at great distances away from the site of impact. This phenomenon can be characterized as “diffuse neuronal injury” and is caused by stretching and shearing of the axon.
  • the inflammatory response to the injured tissue follows its usual course with neutrophiles preceding the appearance of macrophages.
  • An ischemic injury to the CNS is characterized by an insufficiency or interruption in the blood supply to any locus of the brain such as, but not limited to, a locus of the cerebrum, cerebellum or brain stem.
  • the spinal cord which is also a part of the CNS, is equally susceptible to ischemia resulting from diminished blood flow.
  • An ischemic episode may be caused by a constriction or obstruction of a blood vessel, as occurs in the case of a thrombus or embolus.
  • the ischemic episode may result from any form of compromised cardiac function, including cardiac arrest.
  • the deficiency is sufficiently severe and prolonged, it can lead to disruption of physiologic function, subsequent death of neurons, and necrosis (infarction) of the affected areas.
  • the extent and type of neurologic abnormality resulting from the injury depend on the location and size of the infarct or the focus of ischemia. Where the ischemia is associated with a stroke, it can be either global or focal in extent.
  • Global ischemia refers to a condition that results from a general diminution of blood flow to the entire brain, forebrain, or spinal cord, which causes the delayed death of neurons, particularly those in metabolically active loci, throughout these tissues.
  • Focal ischemia refers to a condition that results from the blockage of a single artery that supplies blood to the brain or spinal cord, resulting in the death of all cellular elements (pan-necrosis) in the territory supplied by that artery.
  • the present invention provides a method for treating or preventing neuronal damage caused by a traumatic or ischemic injury to the CNS through the administration of a therapeutically effective amount of a progestin or progestin metabolite such that, prior to termination of administration of the progestin or progestin metabolite the administration is tapered to avoid withdrawal.
  • the present invention relates to the finding that, when stopping progesterone therapy, tapered administration of progesterone to avoid withdrawal results in greater efficacy of progesterone therapy compared to abrupt termination of administration.
  • tapered administration or “tapered administration dosing regimen” is meant successive reduced doses and eventual elimination of the progestin or progestin metabolite, either over a fixed period of time or a time determined empirically by a physician's assessment based on regular monitoring of a therapeutic response of a patient to a traumatic or ischemic CNS injury.
  • the period of the tapered progestin or progestin metabolite administration can be about 12, 24, 36, 48 hours or longer. Alternatively, the period of the tapered progestin or progestin metabolite administration can range from about 1 to 12 hours, about 12 to about 48 hours, or about 24 to about 36 hours.
  • tapered administration of a progestin or progestin metabolite involves tapered administration of progesterone.
  • the drug taper employed could involve progressively dividing administered doses by 50%. For example, such a taper from 500 mg would go 500, 250, 125, 62.5, etc.
  • the drug taper employed could be a “linear” taper. For example, a “10%” linear taper from 500 mg would go 500, 450, 400, 350, 300, 250, 200, 150, 100, 50, etc.
  • an exponential taper could be employed which, if the program outlined above is used as an example, the exponential taper would be, e.g., 500, 450, 405, 365, 329, 296, 266, 239, etc. Accordingly, about a 5%, 10%, 20%, 30%, or 40% linear or exponential taper could be employed in the methods of the invention.
  • a linear or exponential taper of about 1% to 5%, about 6% to 10%, about 11% to 15%, about 16% to 20%, about 21% to 25%, about 26% to 30%, about 31% to 35%, about 36% to 40% could be employed.
  • the taper schedule can be determined based on the treating physician's assessment of the patient's response to therapy.
  • the tapered administration methods of the present invention are used in combination with administration of progestin or progestin metabolite therapies for subjects having traumatic or ischemic CNS injury.
  • the subject can be any mammal, preferably a human or an animal, including domestic, agricultural, or exotic animals.
  • the human is an adult (over 18 years of age), while in other embodiments, the human is a child (under 18 years of age).
  • the child can be a neonate, infant, toddler, pre-pubescent or post-pubescent and range in age from about birth, 1 month to about 2 year, about 1 year to about 5 years, about 4 years to about 9 years, about 8 years to about 14, or about 13 to about 18 years of age.
  • the human can be about 55 to 60, 60 to 65, 65 to 70, 70 to 75, 75 to 80, 80 to 85, 85 to 90, 90 to 95 or older.
  • the progestin or progestin metabolite Prior to the tapered administration of the present invention, is administered at a therapeutically effective level to a subject in need thereof for the treatment of a CNS injury.
  • therapeutically effective amount is meant the concentration of a progestin or progestin metabolite that is sufficient to elicit a therapeutic effect.
  • concentration of a progestin or progestin metabolite in an administered dose unit in accordance with the present invention is effective in the treatment or prevention of neuronal damage that follows a traumatic or ischemic injury to the CNS and hence, elicits a neuroprotective effect.
  • the therapeutically effective amount will depend on many factors including, for example, the specific activity of the progestin or progestin metabolite, the severity, pattern, and type of injury (e.g., traumatic or ischemic), the resulting neuronal damage, the responsiveness of the patient, the weight of the patient along with other intraperson variability, the method of administration, and the progestin or progestin metabolite formulation used.
  • Various methods for administering a therapeutically effective amount of the progestin or progestin metabolite treat CNS injury, including determination of efficacy, dosage, and route of administration, are known in the art (see, e.g., U.S. Patent Application No. 60/664,728 filed Mar. 24, 2005, and U.S.
  • the tapered administration methods of the present invention are used in combination with administration of progestin or progestin metabolite at least once a day, including administration once or several times a day.
  • the duration of the treatment may be once per day for a period of from two to three weeks and may continue for a period of months or even years.
  • the daily dose can be administered either by a single dose in the form of an individual dosage unit or several smaller dosage units or by multiple administration of subdivided dosages at certain intervals.
  • a dosage unit can be administered from about 0 hours to about 1 hr, about 1 hr to about 24 hours, about 1 to about 72 hours, about 1 to about 120 hours, or about 24 hours to at least about 120 hours post injury.
  • the dosage unit can be administered from about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 30, 40, 48, 72, 96, 120 hours or longer post injury.
  • Subsequent dosage units can be administered any time following the initial administration such that a therapeutic effect is achieved.
  • additional dosage units can be administered to protect the patient from the secondary wave of edema that may occur over the first several days post-injury.
  • the tapered administration methods of the present invention are used in combination with a constant progesterone or synthetic progestin dosing regimen.
  • a constant progesterone or synthetic progestin dosing regimen is intended the patient undergoing therapy with the progesterone or synthetic progestin is administered a constant total hourly dose of the progesterone or synthetic progestin over the course of treatment. This hourly dose of the progesterone or synthetic progestin is partitioned into a series of equivalent doses that are administered according to an appropriate dosing schedule depending on the method of administration.
  • the duration of the constant progesterone or synthetic progestin dosing regimen is about 12, 24, 36, 60, 72, 84, or 120 hours or about 1 to 24 hours, about 12 to 36 hours, about 24 to 48 hours, about 36 to 60 hours, about 48 to 72 hours, about 60 to 96 hours, about 72 to 108 hours, about 96 to 120 hours, or about 108 to 136 hours.
  • the tapered administration methods of the present invention are used in combination with a “two-level progesterone or synthetic progestin dosing regimen”
  • a “two-level progesterone or synthetic progestin dosing regimen” is intended the patient undergoing the therapy with the progesterone or synthetic progestin is administered the progesterone or synthetic progestin during two time periods of progesterone or synthetic progestin dosing.
  • the two-time periods can have a combined duration of about 12 hours to about 7 days, including, 1, 2, 3, 4, or 5 days or about 15, 15, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, or 144 hours or about 1 to 24 hours, about 12 to 36 hours, about 24 to 48 hours, about 36 to 60 hours, about 48 to 72 hours, about 60 to 96 hours, about 72 to 108 hours, about 96 to 120 hours, or about 108 to 136 hours.
  • the two-level progesterone or synthetic progestin dosing regimen has a combined duration of about 1 day to about 5 days; in other embodiments, the two-level progesterone or synthetic progestin dosing regimen has a combined duration of about 1 day to about 3 days.
  • the total hourly dose of the progesterone or synthetic progestin that is to be administered during the first and second time periods of the two-level progesterone or synthetic progestin dosing regimen is chosen such that a higher total hourly dose of the progesterone or synthetic progestin is given during the first time period and a lower hourly dose of the progesterone or synthetic progestin is given during the second time period.
  • the duration of the individual first and second time periods of the two-level progesterone or synthetic progestin dosing regimen can vary, depending upon the health of the individual and history of the traumatic or ischemic injury.
  • the patient is administered higher total hourly dose of progesterone or synthetic progestin for at least 1, 2, 3, 4, 5, 6, 12 or 24 hours out of the 1 day to 5 day two-level progesterone or synthetic progestin dosing regimen.
  • the length of the second time period can be adjusted accordingly, and range for example, from about 12 hrs, 24 hrs, 36 hrs, 48 hrs, 60 hrs, 72 hrs, 84 hrs, 96 his, 108 hrs, 120 his or about 12 to about 36 his, about 24 to about 36 hrs, about 24 to about 48 hrs, about 36 hrs to about 60 hours, about 48 his to about 72 hrs, about 60 hrs to about 84 hours, about 72 hrs to about 96 hrs, or about 108 hrs to about 120 hrs.
  • the two-level progesterone or synthetic progestin dosing regimen has a combined duration of 3 days
  • the higher total doses of the progesterone or synthetic progestin could be administered for the first hour
  • the lower total hourly dose of the progesterone or synthetic progestin could be administered for hours 2 to 72.
  • the total hourly dose of progesterone that is to be administered during the first and second time periods of the two-level progesterone or synthetic progestin dosing regimen is chosen such that a lower total hourly dose of the progesterone or synthetic progestin is given during the first time period and a higher hourly dose of the progesterone or synthetic progestin is given during the second time period.
  • AUC Area under the curve
  • reference progesterone or synthetic progestin standard is intended the formulation of the progesterone or synthetic progestin that serves as the basis for determination of the total hourly progesterone or synthetic progestin dose to be administered to a human patient with a traumatic or ischemic central nervous system injury in accordance with the desired constant or two-level progesterone or synthetic dosing regimen to achieve the desired positive effect, i.e., a positive therapeutic response that is improved with respect to that observed without administration of the progesterone or synthetic progestin.
  • the total hourly dose of progesterone or synthetic progestin to be administered during the constant or two-level progesterone or synthetic progestin dosing regimen can therefore allow for a final serum level of the progesterone or synthetic progestin of about of about 100 ng/ml to about 1000 ng/ml, about 1100 ng/ml to about 1450 ng/ml, 100 ng/ml to about 250 ng/ml, about 200 ng/ml to about 350 ng/ml, about 300 ng/ml to about 450 ng/ml, about 400 ng/ml to about 550 ng/ml, about 500 ng/ml to about 650 ng/ml, about 600 ng/ml to about 750 ng/ml, about 700 ng/ml to about 850 ng/ml, about 800 ng/ml to about 950 ng/ml, about 900 ng/ml to about 1050 ng/ml, about 1000
  • the serum level of the progesterone or synthetic progestin comprises about 100 ng/ml, 250 ng/ml, 500 ng/ml, 750 ng/ml, 900 ng/ml, 1200 ng/ml, 1400 ng/ml, 1600 ng/ml.
  • the tapered administration methods of the present invention also contemplate embodiments where a patient undergoing a constant progesterone or synthetic progestin therapy or a two-level progesterone or synthetic dosing regimen is given a time period off from progesterone or synthetic dosing.
  • a progesterone or synthetic progestin dosing regimen is performed, the time period off from the progesterone or synthetic progestin can occur between the conclusion of the first time period of the two-level progesterone or synthetic progestin dosing regimen and the initiation of the second time period of the two-level progesterone or synthetic progestin dosing regimen.
  • the two-level progesterone or synthetic progestin dosing regimen is interrupted such that progesterone or synthetic progestin dosing is withheld for a period of about 15 minutes, 30 minutes, 1 hr, 2 his, 3 hrs, 4 hrs, 5 hrs, 6 hrs or more.
  • a patient undergoing therapy in accordance with the previously mentioned dosing regimens exhibits a partial response, or a relapse following completion of therapy
  • subsequent courses of progesterone or synthetic progestin therapy may be needed to achieve a partial or complete therapeutic response.
  • a patient may receive one or more additional treatment periods comprising either constant or two-level progesterone or synthetic progestin dosing regimens.
  • Such a period of time off between treatment periods is referred to herein as a time period of discontinuance.
  • the length of the time period of discontinuance is dependent upon the degree of patient response (i.e., complete versus partial) achieved with any prior treatment periods of the progesterone or synthetic progestin therapy. It is further recognized that prior to a period of time off or discontinuance, administration of the progesterone or synthetic progestin therapy may be tapered.
  • each maintenance cycle comprises a completed dosing regimen.
  • completed two-level dosing regimen is intended the patient has been administered both the first period and the second period of progesterone or synthetic progestin dosing.
  • the necessity for multiple maintenance cycles can be assessed by monitoring the physiological and behavioral improvement of the patient.
  • the duration between maintenance cycles can be about 1 hr, 15 hrs, 1 day, 2 day, 3 day, 4 day, 5 day, 6 day or other such time periods falling within the range of about 1 day to about 14 days.
  • progestin or progestin metabolite may further comprise an inorganic or organic, solid or liquid, pharmaceutically acceptable carrier.
  • the carrier may also contain preservatives, wetting agents, emulsifiers, solubilizing agents, stabilizing agents, buffers, solvents and salts.
  • Compositions may be sterilized and exist as solids, particulants or powders, solutions, suspensions or emulsions.
  • compositions of the invention may further include one or more accessory ingredient(s) selected from the group consisting of diluents, buffers, flavoring agents, binders, disintegrants, surface active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.
  • accessory ingredient(s) selected from the group consisting of diluents, buffers, flavoring agents, binders, disintegrants, surface active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.
  • the progestin or progestin metabolite can be formulated according to known methods to prepare pharmaceutically useful compositions, such as by admixture with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their formulation are described, for example, in Remington's Pharmaceutical Sciences (16th ed., Osol, A. (ed.), Mack, Easton Pa. (1980)). In order to form a pharmaceutically acceptable composition suitable for effective administration, such compositions will contain an effective amount of the progestin or progestin metabolite, either alone, or with a suitable amount of carrier vehicle.
  • the pharmaceutically acceptable carrier will vary depending on the method of drug administration and may be, for example, either a solid, liquid, or time release.
  • Representative solid carriers are lactose, terra alba, sucorse, talc, geletin, agar, pectin, acacia, magnesium stearate, stearic acid, microcrystalin cellulose, polymer hydrogels, and the like.
  • Typical liquid carriers include syrup, peanut oil, olive oil, cyclodextrin, and the like emulsions.
  • Those skilled in the art are familiar with appropriate carriers for each of the commonly utilized methods of administration.
  • the total amount of progestin or progestin metabolite administered as a therapeutic effective dose will depend on both the pharmaceutical composition being administered (i.e., the carrier being used) and the mode of administration.
  • compositions for use in the methods of the present invention include those suitable for oral, rectal, topical, nasal, ophthalmic, or parenteral (including intraperitoneal, intravenous, subcutaneous, or intramuscular injection) administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier or both, and then, if necessary, shaping the product into desired formulations.
  • compositions for oral administration may be presented as discrete units such as capsules, cachets, tablets, lozenges, and the like, each containing a predetermined amount of the active agent as a powder or granules; or a suspension in an aqueous liquor or non-aqueous liquid such as a syrup, an elixir, an emulsion, a draught, and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine, with the active compound being in a free-flowing form such as a powder or granules which is optionally mixed with a binder, disintegrant, lubricant, inert diluent, surface active agent or dispersing agent.
  • Molded tablets comprised with a suitable carrier may be made by molding in a suitable machine.
  • a syrup may be made by adding the active compound to a concentrated aqueous solution of a sugar, for example sucrose, to which may also be added any accessory ingredient(s).
  • a sugar for example sucrose
  • accessory ingredients may include flavorings, suitable preservatives, an agent to retard crystallization of the sugar, and an agent to increase the solubility of any other ingredient, such as polyhydric alcohol, for example, glycerol or sorbitol.
  • Formulations suitable for parental administration conveniently comprise a sterile aqueous preparation of the active compound, which can be isotonic with the blood of the recipient.
  • Nasal spray formulations comprise purified aqueous solutions of the active agent with preservative agents and isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucous membranes.
  • Formulations for rectal administration may be presented as a suppository with a suitable carrier such as cocoa butter, or hydrogenated fats or hydrogenated fatty carboxylic acids.
  • Ophthalmic formulations are prepared by a similar method to the nasal spray, except that the pH and isotonic factors are preferably adjusted to match that of the eye.
  • Topical formulations comprise the active compound dissolved or suspended in one or more media such as mineral oil, petroleum, polyhydroxy alcohols or other bases used for topical formulations.
  • media such as mineral oil, petroleum, polyhydroxy alcohols or other bases used for topical formulations.
  • the addition of other accessory ingredients as noted above may be desirable.
  • compositions for use in the methods of the present invention include liposomal formulations.
  • the technology for forming liposomal suspensions is well known in the art.
  • the progestin or progestin metabolite salt thereof is an aqueous-soluble salt
  • the same may be incorporated into lipid vesicles.
  • the compound or salt will be substantially entrained within the hydrophilic center or core of the liposomes.
  • the lipid layer employed may be of any conventional composition and may either contain cholesterol or may be cholesterol-free.
  • the salt When the compound or salt of interest is water-insoluble, again employing conventional liposome formation technology, the salt may be substantially entrained within the hydrophobic lipid bilayer that forms the structure of the liposome. In either instance, the liposomes that are produced may be reduced in size, as through the use of standard sonication and homogenization techniques.
  • the liposomal formulations containing the progestin or progestin metabolite or salts thereof may be lyophilized to produce a lyophilizate which may be reconstituted with a pharmaceutically acceptable carrier, such as water, to regenerate a liposomal suspension.
  • compositions for use in the methods of the present invention also include those which are suitable for administration as an aerosol, by inhalation. These formulations comprise a solution or suspension of the desired progestin or progestin metabolite or a salt thereof or a plurality of solid particles of the compound or salt.
  • the desired formulation may be placed in a small chamber and nebulized. Nebulization may be accomplished by compressed air or by ultrasonic energy to form a plurality of liquid droplets or solid particles comprising the compounds or salts.
  • controlled release preparations for use in the methods of the present invention include controlled release preparations.
  • Such controlled release preparations may be achieved by the use of polymers to complex or absorb the progestin or progestin metabolite.
  • the controlled delivery may be exercised by selecting appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl pyrrolidone, ethylene-vinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate).
  • the rate of drug release may also be controlled by altering the concentration of such macromolecules.
  • Another possible method for controlling the duration of action comprises incorporating the therapeutic agents into particles of a polymeric substance such as polyesters, polyamino acids, hydrogels, poly(lactic acid) or ethylene vinylacetate copolymers.
  • a polymeric substance such as polyesters, polyamino acids, hydrogels, poly(lactic acid) or ethylene vinylacetate copolymers.
  • Such teachings are disclosed in Remington's Pharmaceutical Sciences (1980).
  • compositions comprising a therapeutically effective concentration of progestin or progestin metabolite may be administered using any acceptable method known in the art.
  • the pharmaceutical composition comprising progestin or progestin metabolite can be administered methods that include intravenous (IV), intramuscular (IM), subcutaneous (SC), intraperitoneal, transdermal, buccal, vaginal, or intracerebroventricular administration.
  • IV intravenous
  • IM intramuscular
  • SC subcutaneous
  • intraperitoneal transdermal
  • buccal buccal
  • vaginal vaginal
  • intracerebroventricular administration intravenously
  • the pharmaceutical composition comprising progesterone or synthetic progestin can be administered by infusion over a period of about 1 to about 120 hours.
  • infusion of progesterone or synthetic progestin occurs over a period of about 24 to about 72 hours, over a period of about 48 to about 96 hours, or over a period of about 24 to about 120 hours.
  • An embodiment of the present invention provides for the administration of a progesterone or synthetic progestin or analogue thereof via IV administration in a dose of about 0.1 ng to about 100 g per kg of body weight, about 10 ng to about 50 g per kg of body weight, from about 100 ng to about 1 g per kg of body weight, from about 1 ⁇ g to about 100 mg per kg of body weight, from about 1 fig to about 50 mg per kg of body weight, from about 1 mg to about 500 mg per kg of body weight; and from about 1 mg to about 50 mg per kg of body weight.
  • the amount of progesterone or synthetic progestin administered to achieve a therapeutic effective dose is about 0.1 ng, 1 ng, 10 ng, 100 ng, 1 ⁇ g, 10 ⁇ g, 100 ⁇ g, 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 500 mg per kg of body weight or greater.
  • a progestin or progestin metabolite or analogue thereof via parenteral administration in a dose of about 0.1 ng to about 100 g per kg of body weight, about 10 ng to about 50 g per kg of body weight, from about 100 ng to about 1 g per kg of body weight, from about 1 ⁇ g to about 100 mg per kg of body weight, from about 1 ⁇ g to about 50 mg per kg of body weight, from about 1 mg to about 500 mg per kg of body weight; and from about 1 mg to about 50 mg per kg of body weight.
  • the amount of progestin or progestin metabolite administered to achieve a therapeutic effective dose is about 0.1 ng, 1 ng, 10 ng, 100 ng, 1 ⁇ g, 10 ⁇ g, 100 ⁇ g, 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 500 mg per kg of body weight or greater.
  • the progestin or progestin metabolite for parenteral administration is progesterone or allopregnanolone.
  • the tapered administration methods of the invention are used in combination with the use of a progestin or progestin metabolite and at least one additional neuroprotective agent to enhance neuroprotection following a traumatic or ischemic CNS injury.
  • a progestin or progestin metabolite include, for example, any combination of progestin or progestin metabolite.
  • Other neuroprotective agents of interest include, for example, compounds that reduce glutamate excitotoxicity and enhance neuronal regeneration.
  • Such agents may be selected from, but not limited to, the group comprising growth factors.
  • growth factor is meant an extracellular polypeptide-signaling molecule that stimulates a cell to grow or proliferate. Preferred growth factors are those to which a broad range of cell types respond.
  • neurotrophic growth factors include, but are no limited to, fibroblast growth factor family members such as basic fibroblast growth factor (bFGF) (Abraham et al. (1986) Science 233:545-48), acidic fibroblast growth factor (aFGF) (Jaye et al. (1986) Science 233:541-45), the hst/Kfgf gene product, FGF-3 (Dickson et al. (1987) Nature 326-833), FGF-4 (Zhan et al. (1988) Mol. Cell. Biol. 8:3487-3495), FGF-6 (deLapeyriere et al.
  • bFGF basic fibroblast growth factor
  • aFGF acidic fibroblast growth factor
  • FGF-3 Dickson et al. (1987) Nature 326-833
  • FGF-4 Zhan et al. (1988) Mol. Cell. Biol. 8:3487-3495
  • FGF-6 deLapeyriere et
  • KGF keratinocyte growth factor
  • AIGF androgen-induced growth factor
  • Additional neuroprotective agents include, ciliary neurotrophic factor (CNTF), nerve growth factor (NGF) (Seiler, M. (1984) Brain Research 300:33-39; Hagg T. et al. (1988) Exp Neurol 101:303-312; Kromer L. F. (1987) Science 235:214-216; and Hagg T. et al. (1990) J. Neurosci 10(9):3087-3092), brain derived neurotrophic factor (BDNF) (Kiprianova, I. et al. (1999) J. Neurosci. Res.
  • CNTF ciliary neurotrophic factor
  • NTF nerve growth factor
  • BDNF brain derived neurotrophic factor
  • Neurotrophin 3 Neurotrophin 3
  • Neurotrophin 4 Neurotrophin 4
  • TGF- ⁇ 1 transforming growth factor- ⁇ 1
  • BMP-2 bone morphogenic protein
  • GDNF glial-cell line derived neurotrophic factor
  • ADNF activity-dependant neurotrophic factor
  • neuroprotective therapeutic agents include, for example, Clomethiazole (Zendra) (Marshal, J. W. et al. (1999) Exp. Neurol. 156:121-9); kylnurenic acid (KYNA) (Salvati, P. et al. (1999) Prog Neruopsychopharmacol Biol Psychiatry 23:741-52), Semax (Miasoedova, N. F. et al. (1999) Zh Neurol Psikhiatr Imss Korsakova 99:15-19), FK506 (tacrolimus) (Gold, B. G. et al. (1999) J. Pharmacol. Exp. Ther.
  • MK-801 Barth, A. et al. (1996) Neuro Report 7:1461-4
  • glutamate antagonist such as, NPS1506, GV1505260, MK801 (Baumgartner, W. A. et al.(1999) Ann Thorac Surg 67:1871-3), GV150526 (Dyker, A. G. et al. (1999) Stroke 30:986-92); AMPA antagonist such as NBQX (Baumgartner, W. A. (1999) et al. Ann Thorac Surg 67:1871-3, PD152247 (PNQX) (Schielke, G. P. et al.
  • tapered administration methods of the invention are used in combination with the use of a progestin or progestin metabolite and at least one additional neuroprotective agent to enhance neuroprotection following a traumatic or ischemic CNS injury, it is recognized that even less of the progestin or progestin metabolite may be required to be therapeutically effective.
  • the methods of the present invention find use in treating a traumatic or ischemic injury of the central nervous system.
  • Methods to quantify the extent of central nervous system damage (i.e., neurodegeneration) and to determine if neuronal damage was treated or prevented following the administration of a progestin or progestin metabolite are well known in the art.
  • Such neuroprotective effects can be assayed at various levels, including, for example, by promoting behavioral and morphological (i.e., enhancing tissue viability) recovery after traumatic or ischemic brain injury.
  • the neuroprotection resulting from the methods of the present invention will result in at least about a 10% to 20%, 20% to 30%, 30% to 40%, 40% to 60%, 60% to 80% or greater increase in neuronal survival and/or behavioral recovery as compared to the control groups.
  • GAP-43 Growth Associated Protein 43
  • Other histological markers can include a decrease in astrogliosis and microgliosis.
  • a delay in cellular death can be assayed using TUNEL labeling in injured tissue.
  • anatomical measures that can be used to determine an increase in neuroprotection include counting specific neuronal cell types to determine if the progestin or progestin metabolite is preferentially preserving a particular cell type (e.g., cholinergic cells) or neurons in general.
  • a particular cell type e.g., cholinergic cells
  • behavioral assays can be used to determine the rate and extent of behavior recovery in response to the treatment. Improved patient motor skills, spatial learning performance, cognitive function, sensory perception, speech and/or a decrease in the propensity to seizure may also be used to measure the neuroprotective effect.
  • Such functional/behavioral tests used to assess sensorimotor and reflex function are described in, for example, Bederson et al. (1986) Stroke 17:472-476, DeRyck et al. (1992) Brain Res. 573:44-60, Markgraf et al. (1992) Brain Res. 575:238-246, Alexis et al. (1995) Stroke 26:2336-2346; all of which are herein incorporated by reference.
  • Enhancement of neuronal survival may also be measured using the Scandinavian Stroke Scale (SSS) or the Barthl Index. Behavioral recovery can be further assessed using the recommendations of the Subcommittee of the NODS Head Injury Centers in Humans (Hannay et al. (1996) J. Head Trauma Rehabil. 11:41-50), herein incorporated by reference. Behavioral recovery can be further assessed using the methods described in, for example, Beaumont et al. (1999) Neurol. Res. 21:742-754; Becker et al. (1980) Brain Res. 200:07-320; Buresov et al. (1983) Techniques and Basic Experiments for the Study of Brain and Behavior ; Kline et al. (1994) Pharmacol.
  • SSS Scandinavian Stroke Scale
  • a traumatic injury to the CNS results in multiple physiological events that impact the extent and rate of neurodegeneration, and thus the final clinical outcome of the injury.
  • the treatment of a traumatic injury to the CNS encompasses any reduction and/or prevention in one or more of the various physiological events that follow the initial impact.
  • the methods of the invention find use in the reduction and/or prevention of physiological events leading to neurodegeneration following a traumatic injury to the central nervous system.
  • cerebral edema frequently develops following a traumatic injury to the CNS and is a leading cause of death and disability.
  • Cortical contusions for example, produce massive increases in brain tissue water content which, in turn, can cause increased intracranial pressure leading to reduced cerebral blood flow and additional neuronal loss.
  • the methods of the invention find use in reducing and/or eliminating cerebral edema and/or reducing the duration of the edemic event following a traumatic injury to the CNS.
  • Assays to determine a reduction in edema are known in the art and include, but are not limited to, a decrease in tissue water content following the administration of the progestin or progestin metabolite (Betz et al.
  • an overall improvement in behavioral recovery can also be used as a measure for a decrease in edema.
  • a decrease in edema in the effected tissue by at least about 15% to 30%, about 30% to 45%, about 45% to 60%, about 60% to 80%, or about 80% to 95% or greater will be therapeutically beneficial, as will any reduction in the duration of the edemic event.
  • Vasogenic edema following a traumatic brain injury has been associated with damage to the vasculature and disruption of the blood-brain barrier (BBB) (Duvdevani et al. (1995) J. Neurotrauma 12:65-75, herein incorporated by reference).
  • BBB blood-brain barrier
  • Progesterone has been shown to reduce the permeability of the BBB to macromolecules, but not ions, such as sodium in vitro (Betz et al. (1990) Stroke 21:1199-204; Beta et al. (1990) Acta. Neurochir. Suppl. 51:256-8; both of which are herein incorporated by reference).
  • the methods of the invention find use in reducing or eliminating vasogenic edema following a traumatic brain injury.
  • Assays to determine a decrease in vasogenic edema include, for instance, a reduction in Evans' blue extravasation after cortical contusion (Roof et al. (1994) Society for Neuroscience 20:91, herein incorporated by reference).
  • cytokines By releasing cytokines, the invading macrophages and neutrophils stimulate reactive astrocytosis. Release of different chemokines by other cell types induces these immune cells to become phagocytic, with the simultaneous release of free radicals and pro-inflammatory compounds, e.g., cytokines, prostaglandins, and excitotoxins (Arvin et al. (1996) Neurosci. Biobehav. Ref. 20:445-52; Raivich et al. (1996) Kelo J. Med. 45:239-47; Mattson et al. (1997) Brain Res. Rev. 23:47-61; all of which are herein incorporated by reference).
  • cytokines e.g., cytokines, prostaglandins, and excitotoxins
  • the methods of the invention provide a means to reduce or eliminate the inflammatory immune reactions that follow a traumatic CNS injury. Furthermore, by reducing the inflammatory response following an injury, the progestin or progestin metabolite of the present invention can substantially reduce brain swelling and intracranial pressure and reduce the amount of neurotoxic substances (e.g., free radicals and excitotoxins) that are released. Therefore, by reducing the immune/inflammatory response following a traumatic injury to the CNS, neuronal survival and/or behavioral recovery will be enhanced.
  • neurotoxic substances e.g., free radicals and excitotoxins
  • Assays that can be used to determine if the progestin or progestin metabolite of the invention is imparting an anti-inflammatory and a nonspecific suppressive effect on the immune system following a traumatic CNS injury include, for example, a reduction in cytokine induced microglial proliferation in vitro (Hoffman et al. (1994) J. Neurtotrauma 11:417-31; Garcia-Estrada et al. (1993) Brain Res. 628:271-8; both of which are herein incorporated by reference); a reduction in the generation of cytotoxic free radicals by activated macrophages (Chao et al. (1994) Am. J. Reprod. Immunol. 32:43-52; Robert et al.
  • a reduction in the inflammatory immune reactions following a traumatic brain injury can be assayed by measuring cytokine level following the injury in the sham controls versus the progestin OT progestin metabolite treated subjects.
  • Cytokines are mediators of inflammation and are released in high concentrations after brain injury.
  • the level of pro-inflammatory cytokines e.g., interleukin 1-beta, tumor necrosis factor, and interleukin 6
  • the level of anti-inflammatory cytokines e.g., interleukin 10 and transforming growth factor-beta
  • PCR polymerase chain reactions
  • ELISA ELISA
  • histological analysis for different inflammatory cell types e.g., reactive astrocytes, macrophages and microglia
  • inflammatory cell types e.g., reactive astrocytes, macrophages and microglia
  • the methods of the invention find use in reducing free radical damage and thus decreasing or eliminating lipid peroxidation. This effect may occur through an enhancement of endogenous free radical scavenging systems.
  • Assays to measure a reduction in lipid peroxidation in both brain homogenate and in mitochondria are known in the art and include, for example, the thiobarbituric acid method (Roof et al. (1997) Mol. Chem. Neuropathol. 31: 1-11; Subramanian et al. (1993) Neurosci. Lett. 155:151-4; Goodman et al. (1996) J. Neurochem. 66:1836-44; Vedder et al. (1999) J.
  • cytokine-stimulated macrophages generate nitrite, superoxide, and hydrogen peroxide. Since macrophages are known to be very active between 48 hours and seven days after a traumatic brain injury, a reduction in these reactive cells would reduce, secondary-damage to neurons. See, for example, Fulop et al. (1992) 22 nd Annual Meeting of the Society for Neuroscience 18:178; Soares et al. (1995) J. Neurosci. 15: 8223-33; Holmin et al. (1995) Acta Neurochir. 132:110-9; all of which are herein incorporated by reference.
  • an ischemic injury to the CNS results in its own set of physiological events that impact the extent and rate of neurodegeneration, and thus the final clinical outcome of the injury.
  • the treatment of an ischemic injury to the CNS encompasses any reduction and/or prevention in one or more of the various physiological events that follow the initial interruption in blood supply.
  • the methods of the invention find use in the reduction and/or prevention of physiological events leading to or associated with neurodegeneration following an ischemic injury to the central nervous system.
  • ischemic CNS injury is associated with certain physiological events leading to neurodegeneration, including, for example, release or overexpression of proteins such as NSE, myelin basic protein, GFAP, the S-100 protein, and PKCg, stimulation of membrane phospholipid degradation and subsequent free-fatty-acid accumulation, energy failure due to ATP depletion, cellular acidosis, glutamate release and excitotoxicity, calcium ion influx, and free radical generation.
  • Assays to determine a reduction and/or prevention of physiological events leading to or associated with neurodegeneration following an ischemic CNS injury may be directed toward measuring any of these physiological events.
  • assays for measuring levels of NSE, myelin basic protein, GFAP, the S-100 protein, and PKCg are well known in the art (see, e.g., Missler et al. (1997) Stroke, 28:1956-1960; Shashoua et al. (1984) J. Neurochem., 42:1536-1541; and U.S. Pat. No. 6,268,223; all of which are incorporated herein by reference).
  • Assays for measuring a decrease in serum levels of fatty acids may be determined by methods well known in the art such as taught in U.S. Pat. Nos. 4,071,413; 5,512,429; 5,449,607; and 4,369,250, all of which are incorporated herein by reference.
  • assays for determining a reduction and/or prevention of physiological events leading to or associated with neurodegeneration following an ischemic CNS injury may be directed toward clinical assessments of, for example, a decrease in infarct area, improved body weight, and improved neurological outcome. Such clinical assays are well known to those of skill in the art.
  • Bilateral contusions of the medial prefrontal cortex were created by a pneumatic impactor device as previously described [40]. Briefly, rats were given anesthetized with ketamine/xylazine (90 mg/kg; 10 mg/kg) and placed in a stereotaxic apparatus. A craniectomy (diameter 6 nm u) was made over the midline of the prefrontal cortex with its center 1.5 mm AP to bregma. After removal of the bone, the tip of the impactor (diameter 5 mm) was moved to +3.0 mm AP; 1.0 mm ML (from bregma), and checked for adequate clearance. Trauma was produced by pneumatically activating the piston to impact ⁇ 2.0 mm DV (from dura) at a velocity of 3 m/s with a brain contact time of 0.5 seconds.
  • Progesterone was dissolved in peanut oil (Sigma; 4 mg/kg) and injections were given at 1 and 6 hours post-injury and then once per day for either 3 or 5 consecutive days. Control animals received injections of vehicle at similar time-points. Animals were coded with regard to surgery and treatment to prevent experimenter bias during behavioral testing and histological examination.
  • mice Twenty-one days after surgery, animals were perfused with 100 ml 0.1 M phosphate-buffered saline (PBS; pH 7.4) followed by 400 ml 4% paraformaldehyde in 0.1 M phosphate buffer (PB; pH 7.4). Following cryoprotection in 30% sucrose, coronal 40- ⁇ m-thick sections were cut on a freezing microtome, immediately mounted on gel-coated slides and stained for Nissl with thionine to determine placement and extent of the injury.
  • PBS 0.1 M phosphate-buffered saline
  • PB phosphate buffer
  • Mean area measurements of lesion size were quantified from sections at 15 rostral-caudal levels spaced 300 ⁇ n apart.
  • the perimeter of the necrotic cavity (including injured penumbra) was traced on digitized images using the Jandel Scientific SigmaScan software calibrated to calculate the area in mm 2 for each level traced.
  • Perimeters of the striatum and the lateral ventricles were also traced and mean areas were quantified from 7 rostral-caudat levels (300 ⁇ m apart).
  • necrotic tissue was primarily restricted to the medial prefrontal and cingulate cortex. However, in some cases, more severe tissue damage extended into the corpus callosum and the most dorsal aspects of the medial septum and striatum (Data not shown).
  • F 2,19 3.57, P ⁇ 0.05.
  • Tukey post hoc analysis revealed a dose-dependent reduction in necrotic cavity formation. Data not shown. Notably, all animals that were given progesterone tended to have smaller lesions compared to injured animals that were given vehicle injections. However, only 5 days of progesterone resulted in significant reductions in overall necrotic cavity formation (P ⁇ 0.05).
  • progesterone also protected against secondary cell loss in brain regions both proximal (e.g., STR) and distal (e.g., GP, DMN, and VMN) to the zone of injury.
  • proximal e.g., STR
  • distal e.g., GP, DMN, and VMN
  • both 3 and 5 days of progesterone treatment reduced neuronal loss in the STR, GP, and DMN, but only LP5-treatments produced significant reductions in cell loss of the VMN compared to untreated controls.
  • Example 1 Surgery to induce a traumatic brain injury was performed as outlined in Example 1. Behavior testing using the Morris Water Maize was performed as outlined in Example 1 and the methods for the tactical adhesive removal were performed.
  • FIGS. 1A and 1B demonstrate that low and moderate doses of progesterone (8 mg/kg & 16 mg/kg in a cyclodextrin-containing vehicle) produced consistent improvement in Morris water maze performance, whereas the high dose of progesterone (32 mg/kg in a cyclodextrin-containing vehicle) did not show any beneficial effect.
  • the sticker removal task is a test for sensory neglect which is a primary deficit for frontal injury. In this task all doses initially produce behavioral recovery, however, the group receiving the high dose of progesterone degraded to lesion control levels and the moderate dose, which was initially at lesion control levels improved to sham levels by day 21 post-injury. See FIG. 2 .
  • Acute withdrawal and injury interacted to increase anxiety, locomotor and sensory deficits compared to tapered progesterone withdrawal (TWI). Additionally, acute withdrawal-shams (AWS) had increased motor impairments compared to all other shams, and increased anxiety compared to tapered progesterone rats.
  • the neuroprotective factors BDNF and HSP70 increased for TWI over AWI over VI at 3 weeks post-injury. This beneficial effect of tapered hormone treatment correlated with lesion reconstruction and GFAP staining; TWI animals had the smallest lesion volume and fewest reactive astrocytes, followed by AWI, while VI had the largest lesion volume and most reactive astrocytes. Apoptosis and inflammation were decreased with TW, as demonstrated by p53, active Caspase 3, TNF ⁇ and NF ⁇ B.
  • Acute PW has a compelling effect on both behavior and tissue recovery after traumatic brain injury.
  • animals undergoing progesterone withdrawal syndrome exhibit increased anxiety, sensory deficits, and locomotor deficits; all of these are further exacerbated by injury.
  • increased behavioral impairments are still evident in AWI animals.
  • Western blotting revealed decreased expression of apoptotic and inflammatory proteins with tapered withdrawal, although all progesterone treatments led to better outcomes compared to vehicle-only controls.
  • the compound effect of lesions and acute progesterone withdrawal continued to cause behavioral deficits over those animals with a gradual decrease in progesterone treatment.
  • progesterone treatment following traumatic brain injury and stroke reduces the effects of secondary injury and necrosis (Asbury et al. (1998) Behav. Brain Res., 97:99-106; Attella et al. (1987) Behav. Neural. Biol., 48:352-367; Chen et al. (1999) J. Neurol. Sci., 171:24-30; Galani et al. (2001) Restor. Neurol. Neurosci., 18:161-166; Gibson et al. (2005) Exp. Neurol., 193:522-530; Gibson and Murphy (2004) J. Cereb. Blood Flow Metab., 24:805-813; Grossman et al.
  • AW results in an increase in apoptosis, inflammation and anxiety behaviors during the acute recovery phase after TBI compared to TW (Cutler et al. (2005) Exp. Neurol., 195(2):423-429). All animals given progesterone, regardless of their treatment regime, showed improvement over vehicle-treated animals, but those animals with TW had better recovery as evidenced by less inflammation, apoptosis and functional anxiety.
  • AW causes anxiety, depression, and increased seizure susceptibility due to a sudden decrease in GABA-A interactions with allopregnanolone, a progesterone metabolite (Foldvary-Schaefer et al. (2004) Cleve. Clin. J.
  • the p53 protein alters the permeability of mitochondrial membranes, allowing for the release of cytochrome C, which induces the activation of apoptotic proteases, including caspase-3 (Mattson (2003) Neuromolecular Med. 3:65-94). Also, to determine if neuroprotection is enhanced by TW, HSP70 and BDNF were measured, as well as necrotic lesion cavity size and reactive gliosis. Both BDNF and HSP70 act to promote synaptic plasticity and the release of trophic factors (Binder and Scharfman (2004) Growth Factors 22:123-131; Feinstein et al. (1996) J. Biol. Chem.
  • Isofluorane anesthesia was induced for four minutes and 45 seconds at 5% and maintained at 2.5%. Normal body temperature was maintained with a surgical heating pad placed beneath the sterile dressings. The scalp incision area was shaved and sterilized with iodine and isopropanol. A midline incision was made along the scalp and the fascia cleared to expose the surface of the skull. Medial, lateral, and dorsal stereotaxic coordinates were determined at bregma, and a 5-7 mm diameter bilateral craniotomy was performed mid-sagitally, 3 mm anterior to bregma.
  • Medial frontal cortex MFC injury was created with a pneumatic cortical contusion device (5 mm diameter) at a pressure of 1.7 psi, over 50 ms with a velocity of 2.25 m/s and to a depth of 2.5 mm. Sutures were used to close the incision after bleeding ceased. Animals were placed in individual heated, clean recovery cages until they awakened, and were returned to clean individual home cages with accessible moistened food pellets. Sham animals were anesthetized, and an incision was made at the top of the head. The fascia was cleared to expose bregma, then the incision was sutured closed. Sham surgeries were matched to lesion surgeries for all experimental conditions.
  • HBC 2-hydroxypropyl- ⁇ -cyclodextrin
  • Digiscan Locomoter Activity Boxes Random order, blinded testing occurred under red light in a quiet environment one day before injury, and one and seven days post-withdrawal. Up to four animals were tested using the Digiscan Activity Monitoring System (AccuScan Instruments, Inc. Columbus, Ohio) in each trial, with a total of three trials per test day. Rats were placed in the furthest left corner of the Digiscan Activity Box. At that time, the toggle switch was flipped to ‘on’. At exactly five minutes the computer ceased testing, assuring that all tests were the same length regardless of start time. Files were saved according to date and trial number, and the number of fecal boli recorded. Activity boxes were cleaned with 70% ethanol and dried between trials. Center time was defined by the computer as the amount of time the animal spent exploring the activity box away from the corners.
  • Tissue Preparation All animals were decapitated following a lethal 1 mL injection of Nembutal at three weeks post-injury. Brains for histological analysis were extracted after transcardial perfusion with 4% paraformaldehyde. After 24 hours of post-fixation in 4% paraformaldehyde, followed by 10% sucrose and then 20% sucrose solution in DI water, brains were mounted and frozen under dry ice. The forebrain was cut into 25 um sections on a cryostat and stored at ⁇ 80° C. on 1% gelatin-coated slides.
  • Brains for protein analysis were sectioned into the immediate area of the lesion and snap frozen in 2-methyl-butane chilled on dry ice. Samples were stored at ⁇ 80° C. Brain sections were weighed to assure consistency and homogenized via a glass Dounce in 800 mL Tper homogenization buffer (78510, Pierce, Rockford, Ill.) with 10 ⁇ l/ml of protease inhibitor cocktail (P8340, Sigma, St. Louis, Mo.). Homogenized tissue samples were stored at ⁇ 20° C. BCA and Coomassie protein assays (23235, Pierce) were performed in triplicate at three dilutions on each sample to determine protein concentration. The amount of brain homogenate needed to standardize samples to 2 ⁇ g/ ⁇ L for Western blotting was calculated from the results of these assays.
  • Sections used for GFAP immunofluorescence staining were rinsed in PBS, then incubated in 0.2% TritonX in PBS for 5-10 minutes and rinsed again. Sections were then incubated in 1.0% Bovine Serum Albumin (BSA) in PBS for 30 minutes, and left overnight at 4° C. under 1:2000 GFAP (MAB3402, Chemricon) in 1% BSA. After a rinse in PBS and a ten minute incubation in 1% BSA, sections were incubated in 1:1000 mouse-conjugated AlexaFluor 594 (A21125, Invitrogen, Carlsbad, Calif.) secondary antibody solution in 1% BSA overnight at 4° C.
  • BSA Bovine Serum Albumin
  • Slides were cover slipped using Vectashield Mounting Medium (H-1000, Vector Laboratories, Burlingame, Calif.). Slides were processed at 40 ⁇ magnification with a Nikon Olympus microscope equipped with epifluorescence. Prior to acquiring and analyzing images, the microscope was calibrated to 1 ⁇ m. Four separate areas directly adjacent to the injury area were analyzed per section. Luminosity was quantified for n—6 per treatment group with Adobe Photoshop v. 6.0. For each 144k+pixel image, the rating is determined and averaged per pixel over the whole.
  • Vectashield Mounting Medium H-1000, Vector Laboratories, Burlingame, Calif.
  • Proteins were then transferred onto PVDF membranes in the Criterion Western transfer module (165-6001, BioRad), blocked for several hours in milk protein diluent (50-82-00, KPL, Gaithersburg, Md.) and then incubated in primary antibody overnight at 4° C., including p53 (SC-1312, Santa Cruz Biotechnology, Santa Cruz, Calif.) BDNF (AB 1534, Chemicon, Temecula, Calif.) and HSP70 (33-3800, Zymed, Carlsbad, Calif.). HRP-conjugated secondary antibodies (4-18-18, 14-13-06, KPL) were applied the following day for 2 hours and shaken at room temperature. Blots were developed with SuperSignal West Dura substrate (34076, Pierce) using a Kodak scanner and Kodak ID software for densitometry analysis. Loading controls were performed with ⁇ -actin housekeepers.
  • FIG. 5 shows p53, a long-term marker of apoptosis.
  • p53 a long-term marker of apoptosis.
  • FIG. 8A shows representative images of the selected sections anterior to bregma, and the quantified data for each lesion group is shown in FIG. 8B .
  • FIG. 9 demonstrates relative reactive astrocytes as determined by immunofluorescent GFAP staining at three weeks post-injury.
  • This increased exploratory behavior may be due to a mild excitatory effect from the gradual withdrawal, in contrast to the more severe excitotoxic, and limiting effect of the acute withdrawal.
  • mild excitation may further enhance long term recovery of function, as delayed exercise after TBI improves function recovery (Griesbach et al. (2004) Neuroscience, 125:129-139; Kleim et al. (2003) Neurochem. Res., 28:1757-1769; Will et al. (2004) Prog. Neurobiol., 72:167-182).
  • BDNF and HSP 70 both indicators of neuroprotection, were increased for tapered compared to acute withdrawal, while all progesterone treatment resulted in increased HSP70 compared to vehicle-treated animals.
  • BDNF acts to protect tissue from insult and enable post-trauma neuronal plasticity through various mechanisms (Binder and Scharfman (2004) Growth Factors, 22:123-131; Chuang (2004) Crit. Rev. Neurobiol., 16:83-90; Gonzalez et al. (2004) Neuroscience, 125:605-614), while HSP 70 acts as a neuroprotective agent by suppressing inflammatory responses and cytotoxicity (Feinstein et al. (1996) J. Biol.
  • Immunofluorescent staining for GFAP indicated the extent of astrocyte reactivity adjacent to the injury site. While an increase in GFAP can be a hallmark of increased trophic factors, it also indicates glial scarring, inflammation, and cerebral edema (Hatten et al. (1991) Glia, 4:233-243; Leme and Chadi (2001) Arq. Neuropsiquiatr., 59:483-492). As predicted, in the present study an increased response for vehicle-treated lesion animals and a decreased GFAP reaction for acute progesterone-treated lesion animals was observed. The GFAP response was further decreased for tapered progesterone-treated lesion animals.
  • Rats received either bilateral frontal cortex contusion (L) or sham (S) surgery. Rats were injected at one and six hours post injury, then every 24 hours for six days. Vehicle (V) treated rats were given 9 injections of 22.5% cyclodextrin, while AW rats received 9 injections of 16 mg/kg progesterone and TW rats received 7 injections of progesterone at 16 mg/kg, followed by one at 8 mg/kg and one at 4 mg/kg. On day 8, sensory neglect and locomotor activity tests were initiated. Animals were dilled 22 days post-TBI and the brains prepared for either molecular or histological analysis. Western blotting revealed increased BDNF and HSP 70 in TW vs.
  • AW animals P53 was increased in VL animals, while all progesterone-treated groups were equivalent to shams. TW animals had markedly decreased sensory neglect compared to AW animals, and increased center time in locomotor activity assays. In addition, lesion reconstruction revealed a decreased lesion size for TWL over AWL over VL animals. GFAP immunofluorescent staining followed this pattern as well. In conclusion, after TBI, AW affects select behaviors and molecular markers in the chronic recovery period.

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Epidemiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Steroid Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)
US11/909,276 2005-03-24 2006-03-24 Methods for the treatment of a traumatic central nervous system injury via a tapered administration protocol Abandoned US20090221544A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/909,276 US20090221544A1 (en) 2005-03-24 2006-03-24 Methods for the treatment of a traumatic central nervous system injury via a tapered administration protocol

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US66472805P 2005-03-24 2005-03-24
US72966305P 2005-10-24 2005-10-24
US11/909,276 US20090221544A1 (en) 2005-03-24 2006-03-24 Methods for the treatment of a traumatic central nervous system injury via a tapered administration protocol
PCT/US2006/010984 WO2006102644A2 (fr) 2005-03-24 2006-03-24 Methodes destinees au traitement d'une lesion au niveau du systeme nerveux central, dans lesquelles est utilise un protocole d'administration a reduction progressive

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/010984 A-371-Of-International WO2006102644A2 (fr) 2005-03-24 2006-03-24 Methodes destinees au traitement d'une lesion au niveau du systeme nerveux central, dans lesquelles est utilise un protocole d'administration a reduction progressive

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/550,148 Continuation US8614203B2 (en) 2005-03-24 2012-07-16 Methods for the treatment of a central nervous system injury via a tapered administration protocol

Publications (1)

Publication Number Publication Date
US20090221544A1 true US20090221544A1 (en) 2009-09-03

Family

ID=36987966

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/909,276 Abandoned US20090221544A1 (en) 2005-03-24 2006-03-24 Methods for the treatment of a traumatic central nervous system injury via a tapered administration protocol
US11/909,278 Abandoned US20090325920A1 (en) 2005-03-24 2006-03-24 Methods for the treatment of a traumatic central nervous system injury
US13/454,503 Abandoned US20120245133A1 (en) 2005-03-24 2012-04-24 Methods for the treatment of a traumatic central nervous system injury
US13/550,148 Active US8614203B2 (en) 2005-03-24 2012-07-16 Methods for the treatment of a central nervous system injury via a tapered administration protocol

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/909,278 Abandoned US20090325920A1 (en) 2005-03-24 2006-03-24 Methods for the treatment of a traumatic central nervous system injury
US13/454,503 Abandoned US20120245133A1 (en) 2005-03-24 2012-04-24 Methods for the treatment of a traumatic central nervous system injury
US13/550,148 Active US8614203B2 (en) 2005-03-24 2012-07-16 Methods for the treatment of a central nervous system injury via a tapered administration protocol

Country Status (16)

Country Link
US (4) US20090221544A1 (fr)
EP (5) EP1871382B1 (fr)
JP (5) JP5295755B2 (fr)
AT (2) ATE520404T1 (fr)
AU (2) AU2006226811B2 (fr)
CA (2) CA2601715C (fr)
CY (2) CY1111478T1 (fr)
DE (1) DE602006020353D1 (fr)
DK (2) DK1868614T3 (fr)
ES (2) ES2428313T3 (fr)
HK (3) HK1112579A1 (fr)
HR (2) HRP20110198T1 (fr)
PL (2) PL1868614T3 (fr)
PT (2) PT2030622E (fr)
RS (1) RS52471B (fr)
WO (2) WO2006102596A2 (fr)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080318914A1 (en) * 2005-03-24 2008-12-25 Emory University Methods for the treatment of a traumatic central nervous system injury
US20090325920A1 (en) * 2005-03-24 2009-12-31 Emory University Methods for the treatment of a traumatic central nervous system injury
US20110262502A1 (en) * 2010-02-08 2011-10-27 Prairie Pharmaceuticals LLC Pulmonary delivery of 17-hydroxyprogesterone caproate (17-hpc)
WO2012149389A3 (fr) * 2011-04-27 2014-05-08 Northwestern University Procédés de traitement de la maladie d'alzheimer, de la maladie de huntington, de l'autisme ou autres troubles
WO2013112605A3 (fr) * 2012-01-23 2015-01-22 Sage Therapeutics, Inc. Formulation de stéroïde neuroactif et procédés de traitement des troubles de snc
US9149501B2 (en) 2009-10-05 2015-10-06 Northwestern University Methods of treating depression and other related diseases
US20160045396A1 (en) * 2010-02-08 2016-02-18 Prairie Pharmaceuticals LLC Pulmonary Delivery of Progestogen
US9504670B2 (en) 2013-01-29 2016-11-29 Aptinyx, Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9512133B2 (en) 2008-09-18 2016-12-06 Northwestern University NMDA receptor modulators and uses thereof
US9708335B2 (en) 2013-01-29 2017-07-18 Apytinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9738650B2 (en) 2013-01-29 2017-08-22 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9758525B2 (en) 2013-01-29 2017-09-12 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9828384B2 (en) 2013-01-29 2017-11-28 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9925169B2 (en) 2016-05-19 2018-03-27 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9932347B2 (en) 2016-05-19 2018-04-03 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10150769B2 (en) 2016-08-01 2018-12-11 Aptinyx Inc. Spiro-lactam NMDA modulators and methods of using same
US10231976B2 (en) 2010-02-08 2019-03-19 Prairie Pharmaceuticals LLC Methods for the use of progestogen as a glucocorticoid sensitizer
US10251894B2 (en) 2012-11-30 2019-04-09 The Regents Of The University Of California Anticonvulsant activity of steroids
US10336787B2 (en) 2012-02-15 2019-07-02 Emory University Progesterone analogs and uses related thereto
US10478505B2 (en) 2011-09-23 2019-11-19 The Regents Of The University Of California Edible oils to enhance delivery of orally administered steroids
US10918637B2 (en) 2016-08-01 2021-02-16 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10940156B2 (en) 2016-03-08 2021-03-09 Sage Therapeutics, Inc. Neuroactive steroids, compositions, and uses thereof
US10961189B2 (en) 2016-08-01 2021-03-30 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10987361B2 (en) 2010-02-08 2021-04-27 Shenzhen Evergreen Therapeutics Co., Ltd. Treating auto-immune and auto-inflammatory diseases
US11028095B2 (en) 2016-08-01 2021-06-08 Aptinyx Inc. Spiro-lactam and bis-spiro-lactam NMDA receptor modulators and uses thereof
US11299495B2 (en) 2016-08-01 2022-04-12 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US11578072B2 (en) 2018-01-31 2023-02-14 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US12012413B2 (en) 2019-11-11 2024-06-18 Tenacia Biotechnology (Hong Kong) Co., Limited Methods of treating painful diabetic peripheral neuropathy

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2052724A1 (fr) * 2007-10-26 2009-04-29 sanofi-aventis Utilisation de norgestimate en tant qu'inhibiteur sélectif de canal d'ion TRPC3, TRPC6 et TRPC6 et TRPC7
EP2260052A4 (fr) 2008-02-26 2013-01-16 Univ Emory Analogues stéroïdiens utilisés en neuroprotection
WO2010088409A2 (fr) * 2009-01-30 2010-08-05 Emory University Procédés de neuroprotection utilisant des stéroïdes neuroprotecteurs et une vitamine d
TW201138782A (en) 2010-04-26 2011-11-16 Besins Healthcare Lu Sarl Low-oil pharmaceutical emulsion compositions comprising progestogen
EP2819650A1 (fr) 2012-02-29 2015-01-07 B. Braun Melsungen AG Émulsion contenant des hormones comprenant des phospholipides de krill
RU2498826C1 (ru) * 2012-05-14 2013-11-20 Федеральное государственное бюджетное учреждение науки Комплексный научно-исследовательский институт им. Х.И. Ибрагимова Российской академии наук Способ лечения больных с черепно-мозговой травмой
EP2887944B1 (fr) * 2012-08-21 2021-10-06 Sage Therapeutics, Inc. Allopregnanolone pour le traitement du status épileptique réfractaire
DK3033350T3 (da) 2013-08-12 2021-08-30 Univ Emory Progesterone fosfatanaloger og anvendelser relateret dertil
US10231979B2 (en) * 2015-08-07 2019-03-19 Indiana University Research And Technology Corporation Modulation of inflammation using progesterone metabolites
WO2020180424A1 (fr) 2019-03-04 2020-09-10 Iocurrents, Inc. Compression et communication de données à l'aide d'un apprentissage automatique
US20230190709A1 (en) * 2021-12-02 2023-06-22 MarvelBiome, Inc. Methods and uses of microbiome compositions, components, or metabolites for treating vagus nerve associated diseases, disorders, and conditions
WO2024059056A1 (fr) * 2022-09-12 2024-03-21 Emory University Méthodes de gestion d'une lésion cérébrale traumatique

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4404366A (en) * 1980-05-06 1983-09-13 Miles Laboratories, Inc. Beta-galactosyl-umbelliferone-labeled hapten conjugates
US5114388A (en) * 1991-07-26 1992-05-19 True Fitness Technology, Inc. Stair simulator exerciser with adjustable incline
US5120723A (en) * 1987-08-25 1992-06-09 University Of Southern California Method, compositions, and compounds for modulating brain excitability
US5206415A (en) * 1991-12-20 1993-04-27 Washington University Tricyclic steroid analogs
US5212167A (en) * 1991-09-12 1993-05-18 Trustees Of Boston University Modulation of receptor-mediated ion transport
US5232917A (en) * 1987-08-25 1993-08-03 University Of Southern California Methods, compositions, and compounds for allosteric modulation of the GABA receptor by members of the androstane and pregnane series
US5550120A (en) * 1993-08-20 1996-08-27 Jackson; Meyer B. Regulating neuropeptide hormone secretion
USRE35517E (en) * 1987-08-25 1997-05-20 University Of Southern California Method, compositions, and compounds for modulating brain excitability
US5633011A (en) * 1994-08-04 1997-05-27 Alza Corporation Progesterone replacement therapy
US5728688A (en) * 1993-01-19 1998-03-17 Endoreoherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5763431A (en) * 1993-08-20 1998-06-09 Jackson; Meyer B. Method for regulating neuropeptide hormone secretion
US5763492A (en) * 1996-10-01 1998-06-09 Duguesne University Of The Holy Ghost Methods for effecting memory enhancement mediated by non-steroidal sulfatase inhibitors
US5767117A (en) * 1994-11-18 1998-06-16 The General Hospital Corporation Method for treating vascular headaches
US5798347A (en) * 1993-01-19 1998-08-25 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5837544A (en) * 1995-02-02 1998-11-17 Cell Genesys, Inc. Method of inducing a cell to proliferate using a chimeric receptor comprising janus kinase
US5888996A (en) * 1995-07-26 1999-03-30 Trustees Of Boston University Inhibition of NMDA receptor activity and modulation of glutamate-mediated synaptic activity
US5925630A (en) * 1995-06-06 1999-07-20 Cocensys, Inc. Neuroactive steroids of the androstane and pregnane series
US5939545A (en) * 1994-02-14 1999-08-17 Cocensys, Inc. Method, compositions, and compounds for allosteric modulation of the gaba receptor by members of the androstane and pregnane series
US5942241A (en) * 1995-06-09 1999-08-24 Euro-Celtique, S.A. Formulations and methods for providing prolonged local anesthesia
US20010001280A1 (en) * 1998-09-09 2001-05-17 Liang-Chang Dong Dosage form comprising liquid formulation
US6245757B1 (en) * 1997-10-03 2001-06-12 Research Corporation Technologies, Inc. Use of progestins to treat ischemic event
US20080318914A1 (en) * 2005-03-24 2008-12-25 Emory University Methods for the treatment of a traumatic central nervous system injury
US20090325920A1 (en) * 2005-03-24 2009-12-31 Emory University Methods for the treatment of a traumatic central nervous system injury

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5217085A (en) 1975-07-30 1977-02-08 Ono Pharmaceut Co Ltd Method of quantitative determination of free fatty acids in serum usin g fatty acid activating enzymes
US4369250A (en) 1981-07-31 1983-01-18 Sherwood Medical Industries Inc. Fatty acid determination
AU609927B2 (en) 1987-08-25 1991-05-09 Kelvin W. Gee Compositions and methods for alleviating stress, anxiety and seizure activity
US5512429A (en) 1989-09-19 1996-04-30 British Technology Group Limited Assay for enzyme activity
GB9105707D0 (en) 1991-03-18 1991-05-01 Wilton David C Assay
DE4311870C2 (de) 1993-04-10 1998-07-30 Altramed Holdings Ltd Verwendung eines Anti-Östrigens zur Therapie und Prophylaxe von Demenz-Erkrankungen
FR2727117A1 (fr) * 1994-11-18 1996-05-24 Geffard Michel Utilisation de conjugues de la polylysine pour la preparation de medicaments utiles dans le traitement des maladies neurodegeneratives et des affections degeneratives a caractere autoimmun
AU3136897A (en) 1996-05-22 1997-12-09 Diversified Pharmaceuticals, Inc. Compositions, methods and devices for the transdermal delivery of drugs
DE69809223T2 (de) 1997-05-02 2003-07-24 Wyeth Corp 5-alpha-pregnan-3-beta-ol-20-one 3-sulfate für di behandlung von tumoren und zns krankheiten
US6268223B1 (en) 1999-08-27 2001-07-31 Viatech Imagin, Llc Assay for detecting damage to the central nervous system
US20020072509A1 (en) * 2000-10-11 2002-06-13 Stein Donald Gerald Methods for the treatment of a traumatic central nervous system injury

Patent Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4404366A (en) * 1980-05-06 1983-09-13 Miles Laboratories, Inc. Beta-galactosyl-umbelliferone-labeled hapten conjugates
USRE35517E (en) * 1987-08-25 1997-05-20 University Of Southern California Method, compositions, and compounds for modulating brain excitability
US5120723A (en) * 1987-08-25 1992-06-09 University Of Southern California Method, compositions, and compounds for modulating brain excitability
US5232917A (en) * 1987-08-25 1993-08-03 University Of Southern California Methods, compositions, and compounds for allosteric modulation of the GABA receptor by members of the androstane and pregnane series
US5114388A (en) * 1991-07-26 1992-05-19 True Fitness Technology, Inc. Stair simulator exerciser with adjustable incline
US5212167A (en) * 1991-09-12 1993-05-18 Trustees Of Boston University Modulation of receptor-mediated ion transport
US5366968A (en) * 1991-09-12 1994-11-22 Trustees Of Boston University Modulation of receptor-mediated ion transport
US5292906A (en) * 1991-12-20 1994-03-08 Washington University Tricyclic steroid analogs
US5344826A (en) * 1991-12-20 1994-09-06 Washington University Tricycle steroid analogs
US5206415A (en) * 1991-12-20 1993-04-27 Washington University Tricyclic steroid analogs
US5824671A (en) * 1993-01-19 1998-10-20 Endorecherche Inc Therapeutic methods and delivery systems utilizing sex steroid precursors
US5728688A (en) * 1993-01-19 1998-03-17 Endoreoherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5955455A (en) * 1993-01-19 1999-09-21 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5922700A (en) * 1993-01-19 1999-07-13 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5872114A (en) * 1993-01-19 1999-02-16 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5843932A (en) * 1993-01-19 1998-12-01 Endorcaherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5780460A (en) * 1993-01-19 1998-07-14 Endoreoherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5798347A (en) * 1993-01-19 1998-08-25 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5807849A (en) * 1993-01-19 1998-09-15 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5837700A (en) * 1993-01-19 1998-11-17 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5550120A (en) * 1993-08-20 1996-08-27 Jackson; Meyer B. Regulating neuropeptide hormone secretion
US5763431A (en) * 1993-08-20 1998-06-09 Jackson; Meyer B. Method for regulating neuropeptide hormone secretion
US5939545A (en) * 1994-02-14 1999-08-17 Cocensys, Inc. Method, compositions, and compounds for allosteric modulation of the gaba receptor by members of the androstane and pregnane series
US5633011A (en) * 1994-08-04 1997-05-27 Alza Corporation Progesterone replacement therapy
US5767117A (en) * 1994-11-18 1998-06-16 The General Hospital Corporation Method for treating vascular headaches
US5929061A (en) * 1994-11-18 1999-07-27 The General Hospital Corporation Method for treating vascular headaches
US5837544A (en) * 1995-02-02 1998-11-17 Cell Genesys, Inc. Method of inducing a cell to proliferate using a chimeric receptor comprising janus kinase
US5925630A (en) * 1995-06-06 1999-07-20 Cocensys, Inc. Neuroactive steroids of the androstane and pregnane series
US5942241A (en) * 1995-06-09 1999-08-24 Euro-Celtique, S.A. Formulations and methods for providing prolonged local anesthesia
US5888996A (en) * 1995-07-26 1999-03-30 Trustees Of Boston University Inhibition of NMDA receptor activity and modulation of glutamate-mediated synaptic activity
US5763492A (en) * 1996-10-01 1998-06-09 Duguesne University Of The Holy Ghost Methods for effecting memory enhancement mediated by non-steroidal sulfatase inhibitors
US6245757B1 (en) * 1997-10-03 2001-06-12 Research Corporation Technologies, Inc. Use of progestins to treat ischemic event
US20010001280A1 (en) * 1998-09-09 2001-05-17 Liang-Chang Dong Dosage form comprising liquid formulation
US20080318914A1 (en) * 2005-03-24 2008-12-25 Emory University Methods for the treatment of a traumatic central nervous system injury
US7473687B2 (en) * 2005-03-24 2009-01-06 Emory University Methods for the treatment of a traumatic central nervous system injury
US20090325920A1 (en) * 2005-03-24 2009-12-31 Emory University Methods for the treatment of a traumatic central nervous system injury
US7915244B2 (en) * 2005-03-24 2011-03-29 Emory University Methods for the treatment of a traumatic central nervous injury
US20110224181A1 (en) * 2005-03-24 2011-09-15 Emory University Methods for the treatment of a traumatic central nervous system injury

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080318914A1 (en) * 2005-03-24 2008-12-25 Emory University Methods for the treatment of a traumatic central nervous system injury
US20090325920A1 (en) * 2005-03-24 2009-12-31 Emory University Methods for the treatment of a traumatic central nervous system injury
US7915244B2 (en) 2005-03-24 2011-03-29 Emory University Methods for the treatment of a traumatic central nervous injury
US20110224181A1 (en) * 2005-03-24 2011-09-15 Emory University Methods for the treatment of a traumatic central nervous system injury
US8455468B2 (en) 2005-03-24 2013-06-04 Emory University Methods for the treatment of a traumatic central nervous system injury
US9512133B2 (en) 2008-09-18 2016-12-06 Northwestern University NMDA receptor modulators and uses thereof
US9802946B2 (en) 2008-09-18 2017-10-31 Northwestern University NMDA receptor modulators and uses thereof
US10906913B2 (en) 2008-09-18 2021-02-02 Northwestern University NMDA receptor modulators and uses thereof
US9340576B2 (en) 2009-10-05 2016-05-17 Northwestern University Methods of treating depression and other related diseases
US9198948B2 (en) 2009-10-05 2015-12-01 Northwestern University Methods of treating depression and other related diseases
US9149501B2 (en) 2009-10-05 2015-10-06 Northwestern University Methods of treating depression and other related diseases
US10167314B2 (en) 2009-10-05 2019-01-01 Northwestern University Methods of treating depression and other related diseases
US9796755B2 (en) 2009-10-05 2017-10-24 Northwestern University Methods of treating depression and other related diseases
US20160045396A1 (en) * 2010-02-08 2016-02-18 Prairie Pharmaceuticals LLC Pulmonary Delivery of Progestogen
US10993879B2 (en) * 2010-02-08 2021-05-04 Shenzhen Evergreen Therapeutics Co., Ltd. Pulmonary delivery of progestogen
US10231976B2 (en) 2010-02-08 2019-03-19 Prairie Pharmaceuticals LLC Methods for the use of progestogen as a glucocorticoid sensitizer
US20110262502A1 (en) * 2010-02-08 2011-10-27 Prairie Pharmaceuticals LLC Pulmonary delivery of 17-hydroxyprogesterone caproate (17-hpc)
US10987361B2 (en) 2010-02-08 2021-04-27 Shenzhen Evergreen Therapeutics Co., Ltd. Treating auto-immune and auto-inflammatory diseases
EP3679931A3 (fr) * 2011-04-27 2020-10-14 Northwestern University Procédés de traitement de la maladie d'alzheimer, de la maladie de huntington, de l'autisme ou d'autres troubles
EA029999B1 (ru) * 2011-04-27 2018-06-29 Нортвестерн Юниверсити Способы лечения аутизма
WO2012149389A3 (fr) * 2011-04-27 2014-05-08 Northwestern University Procédés de traitement de la maladie d'alzheimer, de la maladie de huntington, de l'autisme ou autres troubles
AU2012249397B2 (en) * 2011-04-27 2017-03-30 Northwestern University Methods of treating Alzheimer's disease, Huntington's disease, autism, or other disorders
US10478505B2 (en) 2011-09-23 2019-11-19 The Regents Of The University Of California Edible oils to enhance delivery of orally administered steroids
EP3650027A1 (fr) * 2012-01-23 2020-05-13 Sage Therapeutics, Inc. Formulations stéroïdiennes neuroactives comprenant un complexe d'alloprégnanolone et d'éther sulfobutylique bêta-cyclodextrine
EP2806877A4 (fr) * 2012-01-23 2016-02-17 Sage Therapeutics Inc Formulation de stéroïde neuroactif et procédés de traitement des troubles de snc
AU2018201307B2 (en) * 2012-01-23 2020-05-14 Sage Therapeutics, Inc. Neuroactive Steroid Formulations and Methods of Treating CNS Disorders
EP4295908A3 (fr) * 2012-01-23 2024-03-20 Sage Therapeutics, Inc. Formulations stéroïdiennes neuroactives comprenant un complexe d'alloprégnanolone et d'éther sulfobutylique bêta-cyclodextrine
US11426417B2 (en) 2012-01-23 2022-08-30 Sage Therapeutics, Inc. Neuroactive steroid formulations and methods of treating CNS disorders
WO2013112605A3 (fr) * 2012-01-23 2015-01-22 Sage Therapeutics, Inc. Formulation de stéroïde neuroactif et procédés de traitement des troubles de snc
EP2806877B1 (fr) 2012-01-23 2019-10-09 Sage Therapeutics, Inc. Formulations de stéroïdes neuroactifs comprenant un complexe d'allopregnanolone et de sulfobutyl ether beta-cyclodextrine
RU2681835C2 (ru) * 2012-01-23 2019-03-13 Сейдж Терапьютикс, Инк. Лекарственные формы нейроактивных стероидов и способы лечения нарушений цнс
CN112472814A (zh) * 2012-01-23 2021-03-12 萨奇治疗股份有限公司 神经活性类固醇制剂和治疗中枢神经系统障碍的方法
US10322139B2 (en) 2012-01-23 2019-06-18 Sage Therapeutics, Inc. Neuroactive steroid formulations and methods of treating CNS disorders
US11274118B2 (en) 2012-02-15 2022-03-15 Emory University Progesterone analogs and uses related thereto
US10336787B2 (en) 2012-02-15 2019-07-02 Emory University Progesterone analogs and uses related thereto
US10251894B2 (en) 2012-11-30 2019-04-09 The Regents Of The University Of California Anticonvulsant activity of steroids
US9738650B2 (en) 2013-01-29 2017-08-22 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9758525B2 (en) 2013-01-29 2017-09-12 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10253032B2 (en) 2013-01-29 2019-04-09 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US11077094B2 (en) 2013-01-29 2021-08-03 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10316041B2 (en) 2013-01-29 2019-06-11 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10441571B2 (en) 2013-01-29 2019-10-15 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10273239B2 (en) 2013-01-29 2019-04-30 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9512134B2 (en) 2013-01-29 2016-12-06 Aptinyx, Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10052308B2 (en) 2013-01-29 2018-08-21 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9708335B2 (en) 2013-01-29 2017-07-18 Apytinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10441572B2 (en) 2013-01-29 2019-10-15 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9828384B2 (en) 2013-01-29 2017-11-28 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9504670B2 (en) 2013-01-29 2016-11-29 Aptinyx, Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9579304B2 (en) 2013-01-29 2017-02-28 Aptinyx, Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10940156B2 (en) 2016-03-08 2021-03-09 Sage Therapeutics, Inc. Neuroactive steroids, compositions, and uses thereof
US11554125B2 (en) 2016-03-08 2023-01-17 Sage Therapeutics, Inc. Neuroactive steroids, compositions, and uses thereof
US9925169B2 (en) 2016-05-19 2018-03-27 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US9932347B2 (en) 2016-05-19 2018-04-03 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10196401B2 (en) 2016-05-19 2019-02-05 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10195179B2 (en) 2016-05-19 2019-02-05 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US11370790B2 (en) 2016-08-01 2022-06-28 Aptinyx Inc. Spiro-lactam NMDA modulators and methods of using same
US11299495B2 (en) 2016-08-01 2022-04-12 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US11028095B2 (en) 2016-08-01 2021-06-08 Aptinyx Inc. Spiro-lactam and bis-spiro-lactam NMDA receptor modulators and uses thereof
US11376250B2 (en) 2016-08-01 2022-07-05 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US11427585B2 (en) 2016-08-01 2022-08-30 Aptinyx Inc. Spiro-lactam NMDA modulators and methods of using same
US11512051B2 (en) 2016-08-01 2022-11-29 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US11530223B2 (en) 2016-08-01 2022-12-20 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10961189B2 (en) 2016-08-01 2021-03-30 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10918637B2 (en) 2016-08-01 2021-02-16 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US10150769B2 (en) 2016-08-01 2018-12-11 Aptinyx Inc. Spiro-lactam NMDA modulators and methods of using same
US11578072B2 (en) 2018-01-31 2023-02-14 Aptinyx Inc. Spiro-lactam NMDA receptor modulators and uses thereof
US12012413B2 (en) 2019-11-11 2024-06-18 Tenacia Biotechnology (Hong Kong) Co., Limited Methods of treating painful diabetic peripheral neuropathy

Also Published As

Publication number Publication date
PL2030622T3 (pl) 2011-07-29
HK1128242A1 (en) 2009-10-23
ATE499104T1 (de) 2011-03-15
DE602006020353D1 (de) 2011-04-07
JP2014055183A (ja) 2014-03-27
WO2006102644A9 (fr) 2006-11-23
HK1112579A1 (en) 2008-09-12
EP1871382A2 (fr) 2008-01-02
ES2392454T3 (es) 2012-12-10
EP1868614B1 (fr) 2012-08-08
JP2011225629A (ja) 2011-11-10
US20090325920A1 (en) 2009-12-31
PT2030622E (pt) 2011-06-02
WO2006102596A3 (fr) 2007-01-18
ATE520404T1 (de) 2011-09-15
AU2006226811A1 (en) 2006-09-28
CY1111478T1 (el) 2015-08-05
DK2030622T3 (da) 2011-05-02
PT1868614E (pt) 2012-11-23
HRP20110198T1 (hr) 2011-04-30
WO2006102644A3 (fr) 2007-03-01
EP2431042B1 (fr) 2013-06-26
WO2006102596A2 (fr) 2006-09-28
CY1113253T1 (el) 2016-04-13
US8614203B2 (en) 2013-12-24
CA2602950A1 (fr) 2006-09-28
JP5394059B2 (ja) 2014-01-22
CA2601715A1 (fr) 2006-09-28
DK1868614T3 (da) 2012-11-12
JP5496407B2 (ja) 2014-05-21
JP2008534512A (ja) 2008-08-28
AU2006226774A1 (en) 2006-09-28
HRP20120688T1 (hr) 2012-09-30
EP2030622A1 (fr) 2009-03-04
US20120289491A1 (en) 2012-11-15
CA2601715C (fr) 2012-10-16
JP2013234191A (ja) 2013-11-21
EP2431042A1 (fr) 2012-03-21
WO2006102644A2 (fr) 2006-09-28
ES2428313T3 (es) 2013-11-07
HK1112581A1 (en) 2008-09-12
EP1871382B1 (fr) 2011-08-17
AU2006226811B2 (en) 2010-11-25
RS52471B (en) 2013-02-28
EP1868614A2 (fr) 2007-12-26
EP2494976A1 (fr) 2012-09-05
EP2030622B1 (fr) 2011-02-23
PL1868614T3 (pl) 2013-01-31
JP5295755B2 (ja) 2013-09-18
AU2006226774B2 (en) 2010-12-02
CA2602950C (fr) 2012-10-30
US20120245133A1 (en) 2012-09-27
JP2008534515A (ja) 2008-08-28

Similar Documents

Publication Publication Date Title
US8614203B2 (en) Methods for the treatment of a central nervous system injury via a tapered administration protocol
US20050187188A1 (en) Methods for the treatment of a traumatic central nervous system injury
JP2004532796A5 (fr)
Sherwin Estrogen and cognitive functioning in women: lessons we have learned.
DE69825906T2 (de) Verwendung von angiostatische steroide in photodynamischer therapie
JP7362870B2 (ja) 抗菌剤及び抗炎症剤を含む眼内投与のための医薬組成物
US20140193523A1 (en) Pharmaceutical formulations for the treatment and prevention of trauma-induced neuropathology and neurodegeneration
EP3154551B1 (fr) Utilisation de sulfate de dextrane ayant une masse moléculaire moyenne inférieure à 10 000 da pour induire l'angiogenèse chez un sujet
KR20180018730A (ko) 신경활성 스테로이드 용액 및 그의 사용 방법
WO2018014770A1 (fr) Dérivé de phénol et composition pharmaceutique d'étomidate, préparation pharmaceutique et leur application
JP2023154024A (ja) 眼病態を治療するための方法および組成物
Fréchou et al. Dose-dependent and long-term cerebroprotective effects of intranasal delivery of progesterone after ischemic stroke in male mice
Stein et al. Progesterone and recovery after traumatic brain injury: an overview
ES2371930T3 (es) Método para el tratamiento de una herida del sistema nervioso central por medio de administración reducida de progesterona.
EP1977752A1 (fr) Composition pharmaceutique destinée à réduire l'endométriose
US20030050295A1 (en) Use of non-feminizing estrogens as treatment for retinal vascular diseases
EP3585367A1 (fr) Composition renfermant du cinéol utilisée pour traiter des maladies nasales
US20240115662A1 (en) Methods of treating hypogonadotropic hypogonadism and cognition impairment following a traumatic brain injury
TWI620566B (zh) 三萜混合物用以治療多發性硬化的用途
Chavan et al. An Initial Evaluation of (Aristocort®) Triamcinolone in the Therapy of Ocular Inflammation: Part I: Experimental Studies
Chu et al. Optimum pharmacologic management of an acute spinal cord injury in the setting of a lumbar burst fracture
DE102007008231A1 (de) Pharmazeutische Zubereitung zur Behandlung der Migräne
EP1964567A1 (fr) Préparation pharmaceutique destinée au traitement de la migraine

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: EXECUTIVE ORDER 9424, CONFIRMATORY LICENSE;ASSIGNOR:EMORY UNIVERSITY;REEL/FRAME:020892/0805

Effective date: 20080501

AS Assignment

Owner name: EMORY UNIVERSITY, GEORGIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STEIN, DONALD G.;CUTLER, SARAH;REEL/FRAME:022677/0714;SIGNING DATES FROM 20090412 TO 20090428

STCB Information on status: application discontinuation

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