WO2017117586A1 - Compositions and nethods for treating brain dysfunction - Google Patents

Compositions and nethods for treating brain dysfunction Download PDF

Info

Publication number
WO2017117586A1
WO2017117586A1 PCT/US2016/069636 US2016069636W WO2017117586A1 WO 2017117586 A1 WO2017117586 A1 WO 2017117586A1 US 2016069636 W US2016069636 W US 2016069636W WO 2017117586 A1 WO2017117586 A1 WO 2017117586A1
Authority
WO
WIPO (PCT)
Prior art keywords
dione
administration
amino
gwi
dihydro
Prior art date
Application number
PCT/US2016/069636
Other languages
French (fr)
Inventor
Mark O. Henry
William S. Lynn
Original Assignee
Bach Pharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bach Pharma, Inc. filed Critical Bach Pharma, Inc.
Priority to EP16882793.9A priority Critical patent/EP3397257A4/en
Priority to US16/067,542 priority patent/US20210161890A1/en
Publication of WO2017117586A1 publication Critical patent/WO2017117586A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • 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
    • A61P25/22Anxiolytics
    • 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

Definitions

  • compositions that can be used, for example, in methods of treating medical conditions and symptoms associated with brain dysfunction, including but not limited to Gulf War Illness ⁇ G I ), multiple chemical sensitivity (MCS), cognitive dysfunction (CD), multiple sclerosis (MS), and neurological disorders such as amyotrophic lateral sclerosis (AILS),
  • MCS multiple chemical sensitivity
  • CD cognitive dysfunction
  • MS multiple sclerosis
  • AILS amyotrophic lateral sclerosis
  • the compositions of the invention, whic h can be administered or prepared as a medicament for use in the treatment methods described herein, are p halazinediones or pharmaceutically acceptable salts thereof.
  • the phihaSazmedione can be 5"amino-2,3-dihydro-i ,4-phthalazinedione; an analog or variant thereof; or a salt of the specified compound, the analog, or the variant
  • the compounds described herei can be formulated as diagnostic or pharmaceutical compositions, and the in vention features kits including one or more of these compounds.
  • the present invention features methods for treating a patient who is suffering from a medical condition associated with brain dysfunction (e.g., GWI, ALS, MCS, CD, or any of the neurological disorders described herein) or any disease, condition, or syndrome that manifests as ataxia or in which ataxia is a prominent sign.
  • a medical condition associated with brain dysfunction e.g., GWI, ALS, MCS, CD, or any of the neurological disorders described herein
  • any disease, condition, or syndrome that manifests as ataxia or in which ataxia is a prominent sign.
  • Ataxia as it is conventionally understood; ataxia is the loss of full, control of one's bodily movements. While we may describe numerous embodiments of the invention as methods of treatment, it is to be understood that any of these embodiments can be presented, instead, in terms of the "use” of a composition as described herein. For example, the invention
  • dihydro- 1 ,4- hthalazinedione or a salt thereof for use in die treatment of ataxia .
  • a salt thereof e.g., a monosodmm salt thereof
  • 5-ammo-2,3-dihydiO-l ,4- phtha!azinedione which is used commonly in the scientific and industrial literature, it is to be understood (and would, be recognized by one of ordinary skill in the art), that the compound described by that term may also be described as 5-amino-2 -dihydrophti3 ⁇ 4iaz.ine-l,4-dione.
  • the invention features methods of treating a patient by administering to the patient a therapeutically effective amount of a composition comprising an agent that conforms to the following structure:
  • Ri and R_? are each, independently, hydrogen (H), lithium (Li), sodium (Na), potassium ( ), rubidium (Rb), caesium (Cs), or francium (Fx):
  • Rs is an a!kyi, alkenyl, alkynly, aryl, alkoxy!, alkenyloxy, alkynyloxy, aryloxy, alkoxy carbonyl, alkylamino, alkylthio, alkyh lfonyl, or atkylsulilnyi, each optionally substituted with an alkyf halogen, alkoxy, aryl or heteroaryl moiety:
  • Cf , Cz, C$ t G*, Cs, Cf,, C? and C& are each, independently, carbon 12 ( ! 2 C) or an isotope of u C ( .g., °C);
  • N3 ⁇ 4, N2 and N3 are each, independently, nitrogen 14 ( 54 N) or an isotope of 3 ⁇ 4 (e.g., 1> N) and Oi and O2 are each, independently, oxygen 1.6 ( K, 0) or an isotope of U O (e.g., i7 0 or s S 0).
  • the method can be employed to treat a medical condition such as a mood disorder or mood dysfunction (e.g., bipolar disorder, depression, or schizophrenia); a memory disorder or memory dysfunction (e.g., as occurs with amnesia, Alzheimer's disease, dementia, or
  • a mood disorder or mood dysfunction e.g., bipolar disorder, depression, or schizophrenia
  • a memory disorder or memory dysfunction e.g., as occurs with amnesia, Alzheimer's disease, dementia, or
  • Huntington's disease anxiety or a stress-related condition (e.g., a post-traumatic stress disorder); an acute or chronic brain injury, including an acute or chronic brain injury caused by
  • the invention features methods of treating a mood disorder or mood dysfunction m a patient who is exhibiting the signs and symptoms of such a disorder or dysfunction and who has been diagnosed with and/or who is undergoing treatment for cancer, another chronic illness, infection or substance abuse.
  • the invention also encompasses a compound of Formula 1 "for use" in the treatment of a mood disorder or mood dysfunction, or any of the conditions listed above or elsewhere herein.
  • Any of the methods described herein can include a step of identifying a patient in need of treatment.
  • a patient suspected of suffering from Gulf War Illness may be subjected to a bat tery of tests for physical and mental function, as determined by the United Sta tes Veterans' Association.
  • the tests may include assessment of neurological function, levels of fatigue (e.g., autonomic fatigue) and immune system function, geneiic testing for susceptibility to GWI, behavioral tests, and assessment of quality of life (e.g., appeti te, social interactions, sleep quality, and the like).
  • compositions described herein promote hippocampal neurogenesis, and the methods of the invention and uses of the present compositions accordingly include methods of promoting neurogenesis in the
  • compositions for use in promoting neurogenesis in the hippocampus are provided.
  • treating we mean administering a composition as described herein to a patient with an expectation that the patient will experience an improvement in the unwanted signs and/or symptoms of a condition as described herein (e.g., a less severe or less prolonged manifestation of the sign or symptom).
  • a “therapeutically effective amount” of a composition is the amount that, upon administration, brings about such an improvement in a given patient or, more generally. results in improvemen t on a verage in a population of patients.
  • many of the medical conditions described herein are recognized by a prominent sign or symptom.
  • Alzheimer's disease is firmly associated with cognitive decline
  • multiple sclerosis, ALS, and Parkinson's disease are firmly associated with ataxia
  • post-traumatic stress disorders are firmly associated with anxiety, and so on.
  • the present methods and uses of the compositions of the invention may be described equally well by reference to the condition itself or to a prominent sign or symptom thereof.
  • the methods of the in vention encompass treating a patient who has Alzheimer's disease as well as treating a patient who is experiencing cognitive decline.
  • Fig, 1A Is a flow diagram illustrating the sequence of trials, duration of trials, intervals between trials, examples of object types and floor patterns involved in a Pattern Separation Test (PST) according to one embodiment of the invention.
  • PST Pattern Separation Test
  • Figs. 1B-1G are comparison charts illustrating the results of the Pattern Separation Test (PST) in Fig, 1A.
  • Figs. 2A-2F are comparison charts illustrating the results of a Sucrose Preference Test
  • Figs. 3 A-3D are comparison charts illustrating that 5-8mino-23-dihydro- i ,4- phthalazinedione, monosodium salt treatment normalizes the expression of oxidative stress response genes prdx6, sod2, sqstml and srxnl in GWI rats according to one embodiment of the invention.
  • Fig. 4 is a clustergram showing the expression of oxidative stress response genes in various animal groups according to one embodiment of the invention.
  • the invention features, inter liu ⁇ compositions and methods fo diagnosing and treating patients who are suffering from a medical condition associated with brain dysfunction, including those conditions that manifest with ataxia.
  • the composition can be a compound that conforms to the following Formula (I) or a pharmaceutical composition or formulation containing a compound that conforms to Formula (1);
  • Rj. and Ra can each he, independently, hydrogen (11), lithium (Li), sodium (Na) s potassium (K), rubidium (Rb), caesium (Cs), or franeiura (Fr);
  • I3 ⁇ 4 can be an alkyl, alkenyl, alkynly, aryl, alkoxyl, alkenyloxy, alkynyloxy, aryloxy, aikoxy carbonyl, a!kylamino, alkylthio, alkylstiifonyl, or alkyisuifmyi, each optionally substituted with an alkyi, halogen, aikoxy, aryl or heteroaryl moiety;
  • Cs, Ci, €:3 ⁇ 4, C-4, Cs, Gs, C?, and Cm can each be * independently, carbon 12 ( 32 C) or an isotope of t2 C (e.g.
  • Ns , 2 and M3 can each be, independently, nitrogen 14 ( M N) or an isotope of i N (e.g., !5 );
  • Ot and O2 can each be, independently, oxygen i 6 ( u, 0) or an isotope of u, 0 (e.g., O or f 8 0).
  • ft is to be understood that the methods, uses, and compositions described herein can include either or both of a compound thai conforms to
  • Formula 1 or any other sal t thereof e.g., a pharmaceutically acceptable sail thereof.
  • the composition is a compound that conforms to Formula (!) with the proviso that when each of N1-N3 are j N and each of Ot and O2 are , 0, then each and every one of C2-C? cannot be C
  • the composition can be a compoimd that confbrnis to the foi lowing Formula (II):
  • the composition is a compound that conforms to Forrnul (I) with the proviso that the compound is not N-cthanoy]-5-ammo-2,3-dihydrophthalazine-i,4-dione (also known as 5-(acetoxyamino) ⁇ 2,3-dihydrophthalazine ⁇ ! ,4 ⁇ dione) or a salt thereof.
  • the compound does not conform to foilowing Formula (HI)
  • the compound does not conform to following Formula (IV :
  • N-met yl-5-anuno-2 j ⁇ dihydrophibalazine-F4-dione also known as 5-(naethyiamino ⁇ 2,3- dihydfophthalazine- ' l ,4-dione) or a salt thereof.
  • the compound does not conform to following Formula (V):
  • compositions may also be excluded from one or more of the formulations, kits, or other compositions of matter described herein. Similarly, these compositions may be excluded from the prophyiactic, diagnostic, or treatment methods described herein.
  • the compound is a sodium salt of 5 «ammo «2,3-dihydro-l ,4 « phthalazinedione (also described as 5 ⁇ amino-2,3-dihydroplitlialazme ⁇ l,4 ⁇ dione (iuminol)) as shown in following Formula (VI):
  • the agent can be 5-amino-2,3-dihydro- 1 ,4-phthalazinedione, monosodium salt, also known as 3- amiriophtlialhydrazide, sodium salt, 3-aroinophtha!.ic hydrazide, sodium salt, O-aminophthaioyi hydrazine, sodium salt, tamerit, or tamerite, or traderaarked as MSL €> and/or GVT®, In certain embodiments, the agent can be monosodiiim.5 ⁇ amino-2 !
  • diagnostic standards can include one or more of (i.e., any combination of) the compounds described herein.
  • diagnostic standards we mean formulations generated to serve as reference standards in a diagnostic or other assa conducted using mass spectrometry.
  • a medical condition treated by the pharmaceutical compositions described herein can be, or ca include as a prominent symptom, Lack of muscle control and, as a result, loss of full control over bodily movements (ataxia ⁇ , including lack of movement or of proper, controlled movement of the eye(s), difficulty walking or running in a normal, controlled manner (as can be detected and assessed by gait analysis), lack of coordination (as can be detected and assessed by any test for manual dexterity), aphasia, apraxia, or asthenia.
  • a patient experiencing ataxia may have ALS, MS, or Parkinson's disease.
  • the medical condition can also be, or can include as a prominent symptom, fatigue, Tourette's syndrome, narcolepsy, back and/ r neck pain, and headaches (including migraine headaches (e.g., visual migraines), cluster headaches, and tension headaches).
  • the patient ca exhibit signs of neuronal fatigue.
  • the patient can have multiple sclerosis.
  • the patient can have a multiple chemical sensitivity (MCS) which can, in torn, include a range of symptoms that may be attributed to exposure to chemicals tha are commonly used in building materials, industrial sites, and battle grounds, MCS has also been referred to as environmental illness, sick building syndrome, and idiopathic environmental intolerance.
  • the step of diagnosing the patient and/or monitoring the patient's treatment can include determining the rate of mitochondrial reco ver after a defined exertion (e.g., after exercise in a sample from the patient that includes, for
  • the rate of mitochondrial recovery or any other assessment of a patient can be compared to a reference standard defining a healthy/normal desired level, in other embodiments, the patient can be suffering from a mood disorder or mood dysfunction
  • bipolar disorder depression, or schizophrenia
  • a memory disorder or memory dysfunction e.g., as occurs with amnesia, Alzheimer's disease, dementia, or Huntington's disease
  • anxiety or a stress-related condition e.g., a post-traumatic stress disorder
  • an acute or chronic brain injury including an acute or chronic brain injur ⁇ ' caused by mitochondrial dysfunction or an endogenous retrovirus
  • Gulf War Illness GWI; as defined by the United States Veterans' Administration and also known as Gulf War Veterans' Illness or Gulf War Syndrome
  • acute or chronic fatigue which may or may not occur in the context of GWI; and/or ALS.
  • any of the methods of the invention can include a step of assessing a patient's weight, and in some embodiments, patients who are obese or who have been diagnosed as having obesity-induced inflammation can be excluded from treatment, in other embodiments, the excluded patient can have a metabolic syndrome or medical conditio associated with heavy metal intoxication.
  • the mood disorder or mood dysfunction can be bipolar disorder, depression, schizophrenia, or can be associated with cancer, another chronic illness, infection, or substance abuse.
  • the anxiety or stress-related disorder can be a posttraumatic stress disorder.
  • Hie memory disorder or iiiemory dysfunction can be amnesia, or is associated with a diagnosis of Alzheimer's disease, dementia, Huntington's disease, or Parkinson's disease.
  • the medical condition can. be associated with Gulf War
  • GWI Illness
  • GWI can, more generally, be a. cognitive impairment associated with mitochondrial dysfunction or with other neuronal stresses, particularly those that impair neurogenesis in the hippocampaS region of the brain.
  • the phamiaceuiicai compositions described herein can be administered to treat, cognitive impairment and/or to facilitate cognitive rehabilitation. While the present methods are intended to apply to GWI, the invention is not so limited.
  • the patient can be one who has experienced battle or worked in a battle-torn area, regardless of the precise lime or place. Further, patients who have experienced the stress of preparing for military service are amenable to treatment, as are patients who support others who are preparing for service or recovering from a deployment (e,g., family members or close friends).
  • the conditions amenable to treatment can manifest in various ways, and any given patient can be suffering from a variety of symptoms including, but not limited to, fatigue ⁇ .g., chronic fatigue syndrome), with mitochondrial dysftmction headache, memory problems, muscle or joint pain, muscle weakness (a taxia), diarrhea, dyspepsia, indigestion, or other gastrointestinal problems, other neurological problems, tumors or blood cancers, skin conditions, arthritis, or respiratory problems.
  • the patient has been diagnosed as having a post-traumatic stress disorder (PTSD), a chronic, multi-symptom illness, or a combination thereof.
  • PTSD post-traumatic stress disorder
  • the medical condition being treated by the present compositions may be caused by physical and psychological issues involving any war zone deployment (e.g., deployment during the Gulf War) or may have been caused by exposure to one or more hazards ⁇ e.g., toxins). In other words, the medical condition may have been
  • the hazard may be, but is not limited to, exposure to a nerve gas such as sarin, a pyridostigmine bromide pill, a
  • the hazard may also be exposure to radiation (e.g., electromagnetic radiation).
  • radiation e.g., electromagnetic radiation
  • the medical condition may be associated with one or more neurological disorders, and these neurological disorders may, in turn, be precipitated by preparmg for, living or working within, or attempting to cope with a stressful and/or chemical-laden environment.
  • the neurological disorder may be associated with coping mechanisms that include addiction (e.g., alcohol or substance abuse).
  • addiction e.g., alcohol or substance abuse.
  • many of these disorders manifest in ataxia, and any of the methods described herein can include a step of assessing a patient for ataxia prior to treatment.
  • the invention was developed with human patients in mind, it is not so limited.
  • the present methods can be carried out for the benefit of any vertebrate animal including domesticated mammals (e.g., dogs and cats) and birds kept as pets or in zoos.
  • the subject can also be an animal kept as livestock (e.g., cattle, sheep, chickens, horses, pigs, or goats).
  • livestock e.g., cattle, sheep, chickens, horses, pigs, or goats.
  • the present compositions can be applied to a cell, tissue, organ, organ system, organism, or a medium containing one or more of these (e.g., in a laboratory or cell culture).
  • compositions of the invention can be administered to the patient orally, topically, by inhalation, nasal delivery (at, for example, 50 to 100 mg/ml administered 5 or 6 times per day, for a daily dose of about 25- 1 ,000 mg, for about
  • compositions intended for pharmaceutical use can also be formulated for auricular administration (to or by way of the ear), conjunctival administration (to or by way of the conjunctiva), a
  • an extracorporeal administration e.g., administration outside of the body
  • a hemodialysis e.g., administration through hemodialysate fluid
  • an infiltration e.g., administration that results in substances passing into tissue spaces or into cells
  • an interstitial administration e.g., administration to or in the interstices of a tissue
  • an intra- abdominal administration e.g. administratio within the abdomen
  • administration e.g., administration within an artery or arteries
  • intra-articular administration e.g., administration within a joint
  • mtrabiliary administration e.g., administration within the bile, bile duets or gallbladder
  • intrabronehia! administration e.g., administration within a bronchus
  • intrabursai administration e.g., administration within a bursa
  • a intracardiac administration e.g.
  • an mtraeartilagmous administration e.g., administration within a cartilage
  • an intracaudal administration e.g., administration within the cauda equine
  • an intraeavernous administratio e.g., administration, within a pathologic cavity, such as occurs in the lung in tuberculosis
  • an intracavitary administration e.g., administration within a non-pathologic cavity, such as tha of the cervix, uterus, or penis, or such as that which is formed as the result of a wound
  • an intracerebral administration e.g., administration within the cerebrum
  • an intracistemal administration e.g., administration within the eisteraa magna cerehe!iomedularis
  • an intracoraeal administration e.g., administration within the cornea
  • an intracoronary administration e.g., administration within the coronary arteries
  • iutracorporus cavernosum e.g., administration within, the dilatable spaces of the corporus cavernosa of the penis
  • an ntradermal administration e.g., administration within the dermis
  • an intradiscal administration e.g., administration within a disc
  • an intraductal administration e.g.,
  • an intraduodenal administration e.g.. administration within the duodenum
  • an intradural administration e.g., administration within or beneath the dura
  • an intraepidermal administration e.g., administration within the epidermis
  • an intraduodenal administration e.g.. administration within the duodenum
  • an intradural administration e.g., administration within or beneath the dura
  • an intraepidermal administration e.g., administration within the epidermis
  • an intraduodenal administration e.g. administration within the duodenum
  • an intradural administration e.g., administration within or beneath the dura
  • an intraepidermal administration e.g., administration within the epidermis
  • intraesophageal administration e.g., administration within the esophagus
  • an intragastric administration e.g., administration within the stomach
  • an intragingival administration e.g., administration within the gingivae
  • an intraileal administration e.g., administration within or introduced directly into a localized lesion
  • an intraluminal administration e.g., administration within the lumen of a tube
  • an intralyniphatic administration e.g., administration within the lymph
  • intramedullary administration e.g. , administration within the marrow cavity of a bone
  • an intrameningeal administration e.g., administration within the meninges
  • an intramuscular administration e.g., administration within the eye
  • an intrapericardial. administration e.g., administration within the pericardium
  • an intraperitoneal administration e.g., an intrapleural administration
  • administration within the pleura an intraprostatic administration (e.g., administration within the prostate gland), an trapulraonary administration (e.g., administration within the lungs or its bronchi), an intrasinal administration (e.g., administration within the nasal or periorbital sinuses), an intraspinal administration (e.g., administration within the vertebral column), an intrasynovial administration (e.g., administration within the synovial cavity of a joint), am intratendinous administration (e.g., administration within a tendon), an intratestkular administration (e.g., administration within the testicle), an intrathecal administration (e.g.
  • administration within the uterus an intravascular administration (e.g., administration within a vessel or vessels), an intravenous administration (e.g., administration within or into a vein or veins), an intravenous bolus administration (e.g., administration within or into a vein or veins all at once), an intravenous drip administration (e.g. , administration within or into a vein or veins over a sustained period of time), a intra vesical administration (e.g., administration within the bladder), an i travitreal
  • administration e.g., administration within the vitreous body of the eye
  • an iontophoresis e.g., administration by means of an electric current where ions of soluble salts migrate into the tissues of the body
  • an irrigation e.g., administration to bathe or flush open wounds or body cavities
  • a laryngeal administration e.g., administration directl upon the larynx
  • a nasal administration e.g., administration to the nose; administered by way of the nose
  • a nasogastric administration e.g., administration through the nose and into the stomach, usually by means of a tube
  • an occlusive dressing technique e.g., administration by the topical route which is then covered by a dressing which occludes the area
  • an ophthalmic administration e.g., administration to the external, eye
  • an oral administration e.g., administration to or by way of the mouth
  • an oropharyngeal administration e.g., administration directly to the mouth
  • administration to the outside of the dura mater of the spinal cord a perineural administration (e.g., administration surrounding a nerve or nerves), aperiodontai administration (e.g., administration around. a tooth), a rectal administration (e.g.. administration to the rectum), a respiratory administration (e.g.,
  • a retrobulbar administration e.g., administration behind the pons or behind the eyeball
  • a soft tissue administration e.g. , administration into any soft tissue
  • a subarachnoid administration e.g., administration beneath the arachnoid
  • a subconjunctival administration e.g., adminisiraiion beneath the conjunctiva
  • a subcutaneous administration e.g., administration beneath the skin
  • a sublingual administration e.g., administration beneath the tongue
  • a submucosal administration e. ., administration beneath the mucous membrane
  • a topical administration e.g.,
  • a transdermal administration e.g., administration through the dermal layer of the skin to the systemic circulation by diffusion
  • a rransmucosal administration e.g., administration across the mucosa
  • a transdermal administration e.g., administration through the dermal layer of the skin to the systemic circulation by diffusion
  • a rransmucosal administration e.g., administration across the mucosa
  • transplacental administration e.g. , administration through or across the placenta
  • transtracheal administration e.g., administration through the wall of the trachea
  • transtympanic transplacental administration e.g., administration through or across the placenta
  • transtracheal administration e.g., administration through the wall of the trachea
  • transtympanic transplacental administration e.g., administration through or across the placenta
  • transtracheal administration e.g., administration through the wall of the trachea
  • transtympanic e.g., administration through or across the placenta
  • administration e.g., administration across or through the tympanic cavity
  • an ureteral administration e.g., administration into the ureter
  • an urethral e.g., administration
  • a vaginal administration e.g., administration into the vagina
  • a combination thereof e.g., administration into the vagina
  • a dose level can vary as a function of the one or more compositions, the severity of the symptoms and the susceptibility of the subject to side effects.
  • Preferred dosages for a given composition are readily determinable by those of skill in the art by a variety of means.
  • multiple doses of one or more compositions are administered.
  • the pharmaceutical compositions may be administered one to three times per month, every other week (qow), 1 -6 times per week, every other day (qod), daily (qd), twice a day (qid), or three times a day (tid), over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from, about eight months to about I. year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
  • the composition can be administrated five times a week for eight weeks. In severe eases, the patient may require administration at one or more of these delivery doses for the remainder of his or her life.
  • a composition as described herein may be administered to the patient at a dosage from about 10 .mg/kg to 500 mg kg patient body weight per day, in 1 to 5 divided doses per day.
  • the composition may be administered at a dosage of about 40 mg kg, about 80 mg/kg, or about 160 mg/kg patient body weight per day.
  • the compound can be present at between about 50-100 mg/ml (e.g. , 70 mg ml).
  • the term "about” is used herein to indicate that a value includes an inherent variation of error for the device or the method being employed to determine the value or plus-or- minus 10% of the value, whichever is greater.
  • the composition can be formulated, in the form of a pill, a capsule, a granule, a tablet, a pallet, a suspension, an injection, an infusion, a suppository, a continuous delivery system, a syrup, a tincture, an ointment, a cream, eye drops, eardrops, a flush, a lavage, a slow absorbing depot a dressing, a. lozenge, or any pharmaceutically acceptable application or as a nutritional supplement.
  • the compounds disclosed herein can be formulated with conventional carriers and
  • excipients which can be selected in accord with ordinary practice. Tablets can typically contain excipients, glidanis, fillers, binders and the like. Aqueous formulations can be prepared in sterile form, and when intended for delivery by other than oral administration generally can be isotonic.
  • Formulations can contain excipients (e.g., excipients set forth in the Handbook of Pharmaceutical
  • Excipients can include ascorbic acid or other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkyiniethylceSlulose, stearic acid or the like.
  • the pH of the t rmuiations can. range from about 3 to about 1 1 , and can be about 7 to 10. While it is possible for the active ingredients to be administered alone it can be preferable to present them as pharmaceutical formulations.
  • the formulations of the agent, as disclosed herein, can include at least one active ingredient, as above defined, together with one or more acceptable carriers thereof and optionally other therapeutic ingredients.
  • the carrieris) should be "acceptable" in the sense of being compatib le with the other ingredients of the formulation and physiologically innocuous to the recipient thereof.
  • the formulations can include those suitable for the foregoing administration routes.
  • the formulations can conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • Such methods can. include the step of bringing into assoeiation the active ingredient with the carrier thai constitutes one or more accessory ingredients.
  • the formulations can be prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers, finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the presently disclosed subject matter suitable for oral administration can be presented as discrete units such as capsules, cachets, or tablets each
  • the active ingredient can also be administered as a bolus, electuary, or paste.
  • a tablet can be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent.
  • the tablets can be coated o scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom. For administration to the eye or other externa!
  • the formulations ca be applied as a topical ointment or a cream containing the active ingredients).
  • the active ingredients can be employed with either a paraffnie or a water-miseible ointment base.
  • the active ingredients can be formulated in a cream with an oil-in-water cream base.
  • the aqueous phase of the cream base can include at least 30% w/w of a po!yhydric alcohol (e.g., an alcohol having two or more hydroxy!
  • the topical formulations can include a compound (e.g.. dimethyl sutphoxide or related analogs) that enhances absorption or penetration of the active ingredient through the ski or other affected areas.
  • the oily phase of the emulsions of this disclosed subject matter can be constituted from known ingredients in a known manner.
  • the phase cart include merely an emulsifier (otherwise blown as an emulgent) or a mixture of at least one emulsifier with, a fat or an oil or a
  • a hydrophilie emulsifier can be included together with a lipophilic emulsifier that acts as a stabilizer. It is also preferred to include both an oil and a fat Together, the emulsitier(s) with, or without stahiiizer(s) can make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base that forms the oily dispersed phase of the cream formulations, Emulgents and emulsion stabilizers suitable for use in the formulation of the agent, as disclosed herein, can include
  • the choice of suitable oils or fats tor the formulation can be based on achieving the desired cosmetic properties.
  • the cream can be a non-greasy, no -staining and washable product with suitable consistency to avoid leakage from tubes or other containers.
  • Straight or branched chain, mono- or dibasic alkyl esters e.g., diisoadipate, isocetyi stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyi oleate, isopropyl palmitate, butyl stearate, 2- ethylhexyl palmitate or a blend of branched chain esters (Crodamol CAP)
  • Iliese may be used alone or in combination depending on. the properties required.
  • high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
  • compositions can. include the agent together with one or more
  • compositions containing the active ingredient can be in any form suitable for the intended method of administration.
  • tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs can be prepared.
  • compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient that is suitable for manufacture of tablets can be acceptable.
  • excipients can be inert diluents (e.g., calcium or sodium carbonate, lactose, lactose monohyd ate, crosearraellose sodium, povidone, calcium or sodium phosphate), granulating and disintegrating agents (e.g.
  • Tablets can be uncoated or coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained, action over a longer period.
  • a time delay material such as glyceryl raonostearate or glyceryl distearate alone or with a wax can be used.
  • Formulations for oral use can be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluents (e.g., calcium phosphate or kaolin), or as soft gelatin capsule wherein the active ingredient is mixed with water or an oil medium (e.g., peanut oil liquid paraffin, or olive oil).
  • an inert solid diluents e.g., calcium phosphate or kaolin
  • an oil medium e.g., peanut oil liquid paraffin, or olive oil
  • Aqueous suspensions of the agent, as disclosed herein, can contain the active materials in admixture with excipients suitable for the manufactur of aqueous suspensions.
  • excipients can include a suspending agent (e.g., sodium carboxymethyiceilulose, mefhylceiluiose,
  • hydroxypropyl methyl.eell.uose sodium alginate, polyvinylpyrrolidone, gum iragacanih or gum acacia), a dispersing or wetting agent such as a naturally occurring phosphatide (e.g. , lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., poiyoxyethylene stearate), a condensaiion product of ethylene oxide with a long chain aliphatic alcohol (e.g..
  • aqueous suspensio can also contain one or more preservatives (e.g., ethyl or n-propy! p- hydroxy-benzoate), one or more coloring agents, one or more flavoring agents and one or more sweetening agents (e.g., sucrose or saccharin).
  • Oil suspensions can be formulated by
  • the oral suspensions can contain a thickening agent (e.g., beeswax, hard paraffin, or cetyl alcohol). Sweetening agents and/or flavoring agents can be added to provide a palatable oral preparation. These compositions can he preserved by the addition of an antioxidant such as ascorbic acid,
  • Dispersible powders and granules of the agent can be suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more
  • Suitable dispersing or wetting agents and suspending agents can be exemplified by those disclosed above. Additional excipients (e.g., sweetening, flavoring and coloring agents) can also be present,
  • compositions of the agent can also be in the form of oil -in- water emulsions.
  • the oily phase can be a vegetable oil (e.g., oliv oil or arachis oil), a mineral oil (e.g., liquid paraffin), or a mixture of these.
  • Formulations for oral use can also be presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent (e.g., calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium (e.g., peanut oil, liquid paraffin, or olive oil).
  • an inert solid diluent e.g., calcium phosphate or kaolin
  • an oil medium e.g., peanut oil, liquid paraffin, or olive oil
  • Aqueous suspensions of the agent can contain the acti ve materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients can include a suspending agent (e.g., sodium carboxymethylceilulose,
  • methykellulose hydroxypropyl methyleelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth or gum acacia
  • a dispersing or wetting agent e.g. , a naturally occurring
  • phosphatide e.g., lecithin
  • a condensation product of an alkyiene oxide with a fatty acid e.g., polyoxyethylene stearate
  • a condensation product of ethylene oxide with a long chain aliphatic alcohol e.g., heptadecaethyleneoxycetanoi
  • a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride e.g.,
  • the aqueous suspension can also contain one or .more preservatives (e.g., ethyl or n-propyl p-hydroxy ⁇ ben3 ⁇ 4oate), one or more coloring agents, one or more flavoring agents and one or more sweetening agents (e.g.) sucrose or saccharin).
  • Oil suspensions can be formulated b suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil sesame oil, or coconut oil, or in a mineral oil (e.g. , liquid paraffin).
  • the oral suspensions can contain a thickening agent (e.g., beeswax, hard paraffin, or cetyl
  • Sweetening agents or flavoring agents can be added to provide a palatable oral preparation. These compositions can be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules of the agent can be suitable tor preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture wi h a dispersing or wetting agent, a suspending agent, and one or more preservatives.
  • Suitable dispersing or wetting agents and suspending agents can be exemplified by those disclosed above. Additional excipients (e.g., sweetening, flavoring and coloring agents) can also be present.
  • the pharmaceutical compositions of the agent, as disclosed herein, can also be in the form of oil -in- water emulsions.
  • the oily phase can be a vegetable oil (e.g., olive oil or arac s oil), a mineral oil (e.g., liquid paraffin), or a mixture of these.
  • Suitable emulsifying agents can include naturally occurring gums (e.g., gum acacia and gum tragaeanih), naturally occurring phosphatides (e.g., soybean lecithin), esters or partial esters derived from fatty acids and hexitol anhydrides (e.g..).
  • sorbitan monooieate sorbitan monooieate
  • condensation products of these partial esters with ethylene oxide e.g., pol oxyethy!ene sorbitan monooieate
  • the emulsion can also contain sweetening and flavoring agents.
  • Syrups and elixirs can be formulated with sweetening agents (e.g., glycerol, sorbitol, or sucrose).
  • sweetening agents e.g., glycerol, sorbitol, or sucrose.
  • Such formulations can also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • compositions of the agent can be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension can be formulated according to the known art. using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • the sterile injectable preparation can also be a sterile injectable solution or suspensio in a non-toxic parenteraUy acceptable diluent or solvent (e.g., a solution in 1,3-butane-diol. or prepared as a Jyophi!ized powder).
  • Suitable vehicles and sol vents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils can be employed.
  • any bland fixed oil can be employed (e.g., synthetic mono- or diglycerides).
  • Fatty acids e.g., oleic acid
  • injec tables can also be used in the preparation of injec tables.
  • the amount of active ingredient that can be combined with the carrier material to produce a single dosage form should vary depending upon the host treated and the particular mode of administration.
  • administration to humans may contain approximately 1 to 1000 mg of active material
  • carrier material that may vary from about 5 to about 95% of the total compositions (weight: weight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 pg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations suitable for administration to the eye can include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient
  • a suitable carrier especially an aqueous solvent for the active ingredient
  • the active ingredient can he present in such formulations in a
  • Formulations suitable for topical administration in the mouth can include lozenges including the active ingredient in a flavored basis, typically sucrose and acacia or tragacanth; pastilles including the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes including the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration can be presented as a suppository with a suitable base including for example cocoa butter or a salicylate.
  • Formulations suitable for intrapulraonary or nasal administratio can. have a particle size in the range of 0.1 to 500 microns (including particle sizes in a range between 0,1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which can be administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
  • Suitable formulations can include aqueous or oily solutions of the acti ve ingredient.
  • Formulations suitable for aerosol or dry powder administration can be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of a given condition.
  • Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams or spra formulations containing in addition to the active ingredient such carriers as are known in the ar to be appropriate.
  • Formulations suitable for parenteral administration can include aqueous and nonaqueous sterile injection solutions which can contain aiiti-o idants, buffers, bacteriostais and solutes which render the formulation isotonic wi th the blood of the in tended recipient ; and aqueous and nonaqueous sterile suspensions which can include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers (e.g. , sealed ampoules and vials) and can be stored in a freeze-dried (lyophilized) condition requiring the addition of the sterile liquid carrier (e.g., water) for injection, immediatel prior to use.
  • sterile liquid carrier e.g., water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules and tablets of the kind previously described.
  • ' Preferred uni dosage formulations can be those containing a daily dose or unit daily sub-dose, as herein above recited, or an
  • formulations of the agent can iticlode other agents conventional in the art having regard to the type of formulation in question (e.g., those suitable for oral administration can include flavoring agents).
  • the agent as disclosed herein, can also be formulated to provide controlled release of the active ingredient to allow less -frequent dosing or to improve the pharmacokinetic or
  • the agent as disclosed herein, can also be provided in compositions including one or more agents formulated for sustained or
  • An effective dose of active ingredient can depend at least, on the nature of the condi tion being treated, toxicity, whether the compound is being used prophylacticaily (typically lower doses), the method of delivery, and the pharmaceutical formulation, and is determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day, typically, from about 0.01 to about 10 mg/kg body weight per day, more typically, from about 0.01 to about 5 mg/kg body weight per clay, and more typically, from about 0.05 to about 0.5 mg/kg body weight per day.
  • the compounds of the presently di sc l osed subject matter can be appl ied in conjunction with one or more inert or inactive ingredients.
  • the agent, as disclosed herein, can be administered by any route appropriate to the condition to be treated. Suitable routes can include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intraderma!, intrathecal and epidural), and the like.
  • kits for aiding in the diagnosis and treatment of a patient who is suffering from a medical condition associated with brain dysfunction or as oihenvise described herein may include a therapeutically effective amount of one of more of the compositions described above.
  • the agent can be used in combination with other active ingredients.
  • the combinations can be selected based on the conditio to be treated, cross-reactivities of ingredients and pharmaco-properties of the combination.
  • the agent can also be combined with one or more other active .ingredients in a unitar dosage form for simultaneous or sequential administration to a patient by the same or different routes of administration.
  • the combination therapy can be administered as a simultaneous or sequential regimen. When administered sequentially, the combination can be administered in. two or more admin.istrati.ons.
  • an effective dosage of each active ingredient can be administered sequentially ( .& , serially), whereas in combination therapy, effective dosages of two or more active ingredients can be administered together.
  • the combination therapy may provide "synergy” and “synergistic effect” (i.e., the effect achieved when the active ingredients nsed together is greater than the sum of the effects thai results from using the compounds
  • a synergistic effect can be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • the synergistic effect can also be attained when the compounds are administered or delivered sequentially (e.g. , in separate tablets, pills, or capsules, or by different injections in separate syringes).
  • kits useful in the presently disclosed subject matter which can include a therapeutically effective amount of a pharmaceutical composition including (a) a compound of component and/or (b) one or more compounds of component, in one or more sterile containers, can also be within the ambit of the presently disclosed subject matter. Sterilization of the container can be carried out using conventional sterilization methodology wel l known to those skilled in the art.
  • Component (a) and/or component (b) can be in the same sterile container or in separate sterile containers.
  • the sterile containers or materials can include separate containers, or one or more multi-part containers, as desired.
  • kits can further include one or more of various conventional pharmaceutical kit components (e.g., one or more pharmaceutically acceptable carriers, additional vials for mixing the components), as should be readily apparent to those skilled in the art.
  • instructions either as inserts or as labels, indicating quantities of the components to be administered, guidelines for adraiddlingrion, and/or guidelines for mixing the components, can also be included in the kit.
  • the methods described herein may include a step to evaluate the effect of the composition (e.g., by assessing a cognitive function or mood of the patient).
  • the methods can include a Pattern Separation Test (PST) for assessing cognitive function, a Sucrose Preference Test (SPT) for assessing mood function, or both.
  • PST Pattern Separation Test
  • SPT Sucrose Preference Test
  • the evaluation step can include evaluating an oxidative stress response or determining the expression level of one or more antioxidant genes in the patient.
  • the gene may be prdx6, sad- 1, sad2, sgstml, srxnl, a cat gene (encoding catalase protein), a cisb gene (encoding eathepsin B), a dhcr24 gene (encoding 24- dehydrocholesferol reductase), a gsr gene (encoding glutathione -reductase), a gstkl gene (encoding glutathione s- transferase kappa 1), & gsipl gene (encoding glutathione s- transferase- 1 ), an idhl gene
  • ncfl encoding neutrophil cytosotic factor 1 protein
  • prdx.1-4 encoding peroxiredoxins 1-4
  • a ptgsl gene (encoding prostaglandm-eiidoperoxide synthase), an sic3Sa! gene (encoding solute carrier family 38), a txni gene (encoding ihioredoxin 1), a ixnip gene (encoding ihioredoxin inieraciing protein), a txnrdl gene (encoding thioredoxin reductase 1 ), a txnrd2 gene (encoding thioredoxin reductase 2), or an ucp2 gene (encoding uncoupling protein 2).
  • the evaluation may include a step of determining the concentration of 3-nitT lyrosine, measuring.
  • the evaluation may include a step of determining the level of endogenous retrovirus Herv-K in (Lymphoma and ALS) and other environmentally activated endogenous retroviruses (e.g., retroviruses activated by oxidative stress).
  • the evaluation may use a model system.
  • the model, system can be an animal model of disease, a cell culture system, an in vitro system, a mathematical model ⁇ e.g., a computational model), or a test carried out with a selected population of subjects (humans participating in a clinical trial).
  • the present invention features a method for preventing a patient from developing a medical condition associated with brain dysfunction (or an of the particular conditions described herein). These methods include administering to the paiient an amount of a composition described herein that is effective in reducing the likelihood thai the patient, will develop a give condition (as described herein).
  • Group I GWI-rats receiving 5-amino-2,3-dihydro- 1 ,4-phthalazinedione at 40 mg/kg b.w.
  • Group 2 GWI-rats receiving 5-amino-2,3-dihydro- 1 ,4-phthalazinedione at 80 mg/kg b.w.
  • Group 3 GWl-rats receiving 5-ammo-2,3-dihydro- 1 ,4-phthalazinedione at 160 mg/kg b.w.
  • Group 4 GWI-rats receiving vehicle fVEH); and
  • Group 5 Age-matched naive control rats.
  • Animal numbers, survival and tissue harvesting A total of 1 15 rats have been purchased so far in three different cohorts.
  • the first cohort comprised 31 animals at the beginn ing of the study. Of those, 29 animal s reached the endpoint of experiments; two animals were found dead during the four-month waiting period between the exposure of animals to GWIR chemicals and 15 minutes of restraint stress for 28 days and the commencement of 5 -amino ⁇ 2,3-di ydro- 1 ,4- phthalazinedione treatment.
  • the brain tissues from 29 animal were harvested for biochemical and molecular biological studies within these fi ve groups: ( !
  • the brain tissues from 39 animals were harvested for immunoMstochemical studies, within these five groups: (I) 5-amino-2,3-dihydRV ' 1 ,4-phthalazinedione, 40 mg/kg, n-8; (2) 5-amino ⁇ 2,3 ⁇ dihydro- ⁇ , -phthala3 ⁇ 4inedione, 80 mg kg, n ⁇ 8; (3) 5-amiiK -2J-djliydro-1 -phthalaMnedione, 160 mg kg, n-8; (4) GWI-VEH, nTM8; and (5) naive control n-7.
  • the third cohort comprised 42 animals at the beginning of the study.
  • PB pyridostigmine bromide
  • DEET 60 mg kg/day
  • permethrin 0.2 mg kg/day (via dermal application).
  • animals were subjected dally to 15 minutes of restraint stress using rat restrainers during the above 28-day period.
  • BrdU injections Subgroups of rats fr m all groups received BrdU injections in the 3rd week of drug/ vehicle treatment dally for 5 days at a dose of 100 mg/kg/day.
  • Behavioral tests for assessing cognitive and mood function We examined animals in all groups through stress- free behavioral tests. We used a Pattern Separation Test (PST) to assess cognitive function, and a Sucrose Preference Test (SPT) to assess mood function.
  • PST Pattern Separation Test
  • SPT Sucrose Preference Test
  • Euthanasia and tissue harvesting Animais belonging to cohort 1 were deeply anesthetized with isofiurane in a small chamber until respiration ceased. Deepl anesthetized animais were decapitated following thoracotomy and brain tissues were dissected rapidly for biochemical and molecular biological studies. Animals belonging to cohort 2 were first deeply anesthetized with isoflurane and then perfused through the heart with 4% paraformaldehyde solution. Fixed tissues were harvested for histological studies.
  • Analyses of oxidative stress The hippocampal tissues obtained from animals belonging to cohort 1 were used for the following .measurements .
  • lipid peroxides are unstable indicators of oxidative stress in cells that decompose to form more complex and reactive compounds such as malondialdehyde (MDA), a natural by-product of lipid peroxidation.
  • MDA malondialdehyde
  • 3-niirotyrosine Increased modification of tyrosine residues in proteins to 3-nitrotyrosine by peroxynitriie or other potential nitrating agents is seen in. tissues subjected to oxidative stress.
  • TMF-a tumor necrosis factor-alpha
  • IT- la interleukin-I alpha
  • iL- ⁇ ⁇ interleiuan- 1 beta
  • VEGF vascular endothelial growth factor
  • FGFp fibroblast growth factor beta
  • interleukin-5 interleukin-5
  • in.terleukin-6 TL-6 interleukin- 15
  • leptin monocyte chemoattractant protein- 1
  • MCP-1 monocyte chemoattractant protein- 1
  • lF >gamma-md «cible protein 10 IP- 10 or CXCLI
  • SCF stem cell factor
  • Rantes T Cell Expressed and Secreted
  • MlP-l a Macrophage inflammatory protein 1 alpha
  • TGFP transforming growth factor-bete
  • Figs, lA-l G illustrate the Pattern Separation Test (PS T) and its results.
  • Fig. 1.A illustrates the sequence of trials, duration of trials, intervals between trials, examples of object types and floor patterns in vol ved in this test.
  • Fig. 1 G compares the total object exploration time between groups in Trial-3. One-way ANOVA analysis did not show differences between groups, implying that the specificity of the novel object exploration time (NO on P2) was not influenced by differences in the total object exploration time.
  • naive rats was revealed by a greater exploration of the object from trial 1 (i.e. novel object on pattern 2 [NO on P2 j) than the object from trial 2 (i.e. iamiiiar object o pattern 2 [FO on P2], p ⁇ 0.0001 , Fig. 1).
  • GWI rats that received moderate doses of 5-arnino-2,3-dihydro- 1 ,4- phthalazinedione also remained impaired, as they spent similar amounts of time with novel and familiar objects on P2 (p>0.05, Fig. 1 ).
  • GWI rats that received higher doses of 5-amino-2,3-dihydro- 1 ,4-phthalazinedione 160 mg/kg displayed an ability for pattern separation. This was revealed by their greater exploration of the object from trial 1 (NO on P2) than the object from trial. 2 (FO on P2), p ⁇ O.05, Fig. I).
  • this study suggested thai cognitive impairment pertaining to pattern separation could be reversed through oral administration of relatively higher doses of 5-amino-2,3-dihydro ⁇ i ,4-phthalazinedione in GWI rats. It has been shown tihat this cognitive improvement is related io increased levels of dentate neurogenesis in these rats, in compariso to GWI rats that received VEH during the same period.
  • SPT Stress free test measuring anhedo a (i.e. inability to feel pleasure, a measure of depression). This test comprised four days of monitoring. On day 1, rats were housed individually and given free access to two identical bottles containing 1% sucrose solution. Rats were trained to adapt to sucrose solution for 24 hours. On day 2, one bottle was replaced with a new bottle containing regular water for 24 hours. On day 3, rats were deprived of water and food fo 23 boors, and then on day 4, rats were given free access to two bottles: one containing 100 ml of sucrose solution and another containing UK) ml of regular water. An hour later, the consumed volume in both bottles was recorded.
  • SPT Stress free test measuring anhedo a (i.e. inability to feel pleasure, a measure of depression). This test comprised four days of monitoring. On day 1, rats were housed individually and given free access to two identical bottles containing 1% sucrose solution. Rats were trained to adapt to sucrose solution for 24 hours. On day 2, one bottle was replaced with a new bottle
  • Figs. 2A-2F illustrate the results of the Sucrose Preference Test (SPT).
  • Figs. 2A- IE- compare the consumption of normal water and sucrose containing water in different animal groups (n-i 3-1.4/group in al GWI groups, n :::: 6 in naive control group). Cyan, Naive control group; red, GWI+VEH group; orange, GWR-5-amino-2.3-dihydro- J ,4-phthalazmedione
  • Fig. 2F compares the total volume (normal water + sucrose-containing water) consumed by rats in each group.
  • One-way A ' NOVA. analysis did not show differences between groups, implying that the preference for drinking sucrose-containing water observed in naive control group and 5 ⁇ amino-2,3-dihydro ⁇ 1 ,4 ⁇ phthalazinedione (80 mg kg and .1 0 mg kg groups) was not influenced by differences in the overall consumption of water during the tes ting period,
  • Fig. 4 is a ciustergrarn showing the expression of oxidative stress response genes in various animal groups.
  • GWMSL 160-5 GWI rats receiving 5-amiTK> ⁇ 2 ⁇ 3-dihydro-.i,4 ⁇ phd ala i.n.edione at .1 0 mg kg (n-5). Arrows denote genes, which show upregu!ation in GWI rats receiving VEH and
  • MSL is the monosodiuiTi salt of luminol (5-amino-2,3-dihydro-l ,4-phthalazinedione or 5-amino-2,3-dihydrophthalazine-l ,4- dione).
  • Prdx6 This gene encodes peroxiredoxin-6 protein. It is a member of the peroxiredoxin family of antioxidant enzymes (thiol-speciftc antioxidant protein family).
  • Sod2 This gene encodes mitochondria! superoxide dismutase 2 protein. It is also known as manganese-dependent superoxide dismutase (MnSOD). Sod2 protein forms a homotetramer and binds one manganese ion per sobunit. This protein binds to the superoxide byproducts of oxidative phosphorylation and converts mem to hydrogen peroxide and diatomic oxygen, which facilitates SOD2 to clear mitochondria!
  • MnSOD manganese-dependent superoxide dismutase
  • Sgstnil This gene encodes sequestosome 1 (or p62) protein. This is a multifunctional protein that binds ubiquitin and regulates activation of the nuclear factor kappa-B (NF-kB) signaling pathway. The protein functions as a scaffolding/adaptor protein in concert with TNF receptor-associated factor 6 to mediate activation of NF-kB in response to upstream signals. Studies also suggest that this protein is a common component of protein aggregates that are found in protein aggregation diseases affecting the brain (e.g.
  • GWI rats were normalized to control levels by higher doses of 5-amiTX> ⁇ 23 ⁇ dihydro-i ,4-phthalazinedione treatment (80 or 160 mg/kg, Fig, 4).
  • the genes include a cat gene, encoding cataiase protein, which is a key antioxidant enzyme that converts the reactive oxygen species hydrogen peroxide to water and oxygen; a Cisb gene, encoding cathepsin B (also called as amyloid precursor protein secrefase, which is a protein involved in the proteoiyiic processing of amyloid precursor protein; a Dhcr24 gene, encoding 24-dehydroeholesterol reductase, which is an oxidoreduciase mvolved in cholesterol biosynthesis; a Gsr gene encoding glutathione reductase.
  • cat gene encoding cataiase protein, which is a key antioxidant enzyme that converts the reactive oxygen species hydrogen peroxide to water and oxygen
  • a Cisb gene encoding cathepsin B (also called as amyloid precursor protein secrefase, which is a protein involved in the proteoiyiic processing of amyloid precursor protein
  • GSH which reduces oxidized glutathione disulfide to the sulfh dryl form GSH, which is an important cellular antioxidant
  • Gsiki gene encoding glutathione S-transferase kappa i. which functions in cellular detoxification
  • (6) a Gstpl encoding glutathione S-transferase- 1 , which plays an important role in detoxification by catalyzing the conjugation of many hydrophobic and electrophilic compounds with reduced glutathione
  • Idhl gene encoding isocitrate
  • dehydrogenase 1 which converts isocitrate to 2-ketoglutarate to produce NADPH necessary for many cellular processes and protection against ROS
  • an Ncfl gene encoding neutrophil cytosolic factor 1 protein, which is a 47 kDa cytosolic subunit of neutrophil NADPH oxidase (a nm!ticomponent enzyme thai is activated to produce superoxide anion)
  • Prdx!-4 genes encoding peroxiredoxins 1-4, which are antioxidant enzymes involved in reducing hydrogen peroxide and alkyl hydroperoxides to water and alcohol with the use of reducing equivalents derived from thiol-containing donor molecules
  • a Pmp gene encoding prion protein, which is a membrane glycosylphosphatidyl-inositol-anchored glycoprotein that tends to aggregate into rod- like structures
  • a Pigs2 gene encoding prostaglandin-endoperoxide synthase (also known as
  • uncoupling protein 2 mitochondrial, proton carrier
  • mitochondrial proton leak This protein facilitates the transfer of anions fro the inner to the outer mitochondrial membrane and the return transfer of protons from the outer to the inner mitochondrial membrane.
  • Analyses of inflammation The hippocampal tissues obtained from animals were used for measurement of the relative levels of inflammatory cytokines in different groups of animals. We employed "The Rat Cytokine Plate Array" from Signosis, which facilitated analyses of 16 rat cytokines in a high-throughput manner.
  • the cytokines included: TNF-a, IL-lo, ⁇ .,- ⁇ . VBGF, FGFp, ⁇ , IL-5, IL-6, IL-15, leprin, MCP-1. IP- 10 (or CXCL10), SCF, Rantes, MIP-la and I ' G ' p.
  • ⁇ ⁇ which showed increased expression in GWI rats receiving VEH, n comparison to naive control animals.
  • This protein is produced by macrophages believed to be involved in inflammation and is typically upregulaisd in the brain in conditions such as Alzheimer's disease, multiple sclerosis and hypoxic-ischemic brain injury. Increased expression of this chemokine is believed to enhance inflammation by attracting more leucocytes to the brain parenchyma.
  • Treatment with 5-amino-2.3-dihydro- L4-piitiiaiazi.oed.ione reduced the

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to compositions that can be used, for example, in methods of treating medical conditions and symptoms associated with brain dysfunction, including but not limited to Gulf War illness (GWI), multiple chemical sensitivity (MCS), cognitive dysfunction (CD), multiple sclerosis (MS), and neurological disorders such as amyotrophic lateral sclerosis (ALS). in various embodiments, the compositions of the invention, which can. be administered or prepared as a medicament for use in the treatment methods described herein, are phthalazinediones or pharmaceutically acceptable salts thereof. As described further below, the phthalazinedione can be 5-amino-2,3-dihydro-1;4-phthala2:inedion.e; an analog or variant thereof; or a salt of the specified compound, the analog, or the variant. The compounds described herein can be formulated as diagnostic or pharmaceutical compositions, and the invention features kits including one or more of these compounds.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of the filin date of U.S. provisional application No. 62/273,913, filed December 31 , 2015, the entire content of which is hereby incorporated by reference in lis entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
This invention was made with government support under grant number W81X WH- 14-1- 0572, awarded by the Uni ted States Army Medical Research and Material Command . The government has certain rights in the invention.
FIELD OF THE INVENTION
The present invention relates to compositions that can be used, for example, in methods of treating medical conditions and symptoms associated with brain dysfunction, including but not limited to Gulf War Illness {G I ), multiple chemical sensitivity (MCS), cognitive dysfunction (CD), multiple sclerosis (MS), and neurological disorders such as amyotrophic lateral sclerosis ( AILS), In various embodiments, the compositions of the invention, whic h can be administered or prepared as a medicament for use in the treatment methods described herein, are p halazinediones or pharmaceutically acceptable salts thereof. As described further below, the phihaSazmedione can be 5"amino-2,3-dihydro-i ,4-phthalazinedione; an analog or variant thereof; or a salt of the specified compound, the analog, or the variant The compounds described herei can be formulated as diagnostic or pharmaceutical compositions, and the in vention features kits including one or more of these compounds. SUMMARY OF THE INVENTION
In a first aspect, the present invention features methods for treating a patient who is suffering from a medical condition associated with brain dysfunction (e.g., GWI, ALS, MCS, CD, or any of the neurological disorders described herein) or any disease, condition, or syndrome that manifests as ataxia or in which ataxia is a prominent sign. We use the term
"ataxia" as it is conventionally understood; ataxia is the loss of full, control of one's bodily movements. While we may describe numerous embodiments of the invention as methods of treatment, it is to be understood that any of these embodiments can be presented, instead, in terms of the "use" of a composition as described herein. For example, the invention
encompasses a method of treating ataxia with 5~ammo-2,3-dihydro-i,4~phthalazinedione or a salt thereof (e.g., a monosodium salt thereof), and it likewise encompasses 5-amino-2,3~
dihydro- 1 ,4- hthalazinedione or a salt thereof (e.g., a monosodmm salt thereof) for use in die treatment of ataxia . While we tend to use the term "5-ammo-2,3-dihydiO-l ,4- phtha!azinedione," which is used commonly in the scientific and industrial literature, it is to be understood (and would, be recognized by one of ordinary skill in the art), that the compound described by that term may also be described as 5-amino-2 -dihydrophti¾iaz.ine-l,4-dione. The latter descriptor was used in the provisional application from which this application claims the benefit of priority and ma be the preferred iUPAC name, in case of doubt, we use the terms "5~amino-2,3-dihydro~ 1 ,4-phthalazinedione" and "5-amino-2,3-dihydrophthala2ine- 1 ,4- dlone" interchangeably.
In one embodiment, the invention features methods of treating a patient by administering to the patient a therapeutically effective amount of a composition comprising an agent that conforms to the following structure:
Figure imgf000003_0001
wherein, Ri and R_? are each, independently, hydrogen (H), lithium (Li), sodium (Na), potassium ( ), rubidium (Rb), caesium (Cs), or francium (Fx):
Rs is an a!kyi, alkenyl, alkynly, aryl, alkoxy!, alkenyloxy, alkynyloxy, aryloxy, alkoxy carbonyl, alkylamino, alkylthio, alkyh lfonyl, or atkylsulilnyi, each optionally substituted with an alkyf halogen, alkoxy, aryl or heteroaryl moiety:
Cf , Cz, C$t G*, Cs, Cf,, C? and C& are each, independently, carbon 12 (! 2C) or an isotope of uC ( .g., °C);
N¾, N2 and N3 are each, independently, nitrogen 14 (54N) or an isotope of ¾ (e.g., 1>N) and Oi and O2 are each, independently, oxygen 1.6 (K,0) or an isotope of UO (e.g., i70 or s S0).
The method can be employed to treat a medical condition such as a mood disorder or mood dysfunction (e.g., bipolar disorder, depression, or schizophrenia); a memory disorder or memory dysfunction (e.g., as occurs with amnesia, Alzheimer's disease, dementia, or
Huntington's disease); anxiety or a stress-related condition (e.g., a post-traumatic stress disorder); an acute or chronic brain injury, including an acute or chronic brain injury caused by
mitochondrial dysfunction or dysfunction of an endogenous retrovirus (e.g., a HERV (human endogenous retrovirus) such as Herv-K); Gulf War illness (GWI; as defined by the United States Veterans' Administration and also known as Gulf War Veterans' Illness or Gulf War Syndrome); acute or chronic fatigue, which may or may not occur in the context of GWI; and/or ALS. The mood disorder or mood dysfunction may also occur in connection with cancer, another chronic illness, infection, or substance abuse. Accordingly, and in case of any doubt, the invention features methods of treating a mood disorder or mood dysfunction m a patient who is exhibiting the signs and symptoms of such a disorder or dysfunction and who has been diagnosed with and/or who is undergoing treatment for cancer, another chronic illness, infection or substance abuse. As indicated above, the invention also encompasses a compound of Formula 1 "for use" in the treatment of a mood disorder or mood dysfunction, or any of the conditions listed above or elsewhere herein.
Any of the methods described herein can include a step of identifying a patient in need of treatment. For example, a patient suspected of suffering from Gulf War Illness (GWI) may be subjected to a bat tery of tests for physical and mental function, as determined by the United Sta tes Veterans' Association. The tests may include assessment of neurological function, levels of fatigue (e.g., autonomic fatigue) and immune system function, geneiic testing for susceptibility to GWI, behavioral tests, and assessment of quality of life (e.g., appeti te, social interactions, sleep quality, and the like).
While the invention is not limited to compositions that achieve a positive treatment outcome by any particular physiological mechanism, we believe the compositions described herein promote hippocampal neurogenesis, and the methods of the invention and uses of the present compositions accordingly include methods of promoting neurogenesis in the
hippocampus and compositions for use in promoting neurogenesis in the hippocampus.
By "treating" we mean administering a composition as described herein to a patient with an expectation that the patient will experience an improvement in the unwanted signs and/or symptoms of a condition as described herein (e.g., a less severe or less prolonged manifestation of the sign or symptom). A "therapeutically effective amount" of a composition is the amount that, upon administration, brings about such an improvement in a given patient or, more generally. results in improvemen t on a verage in a population of patients. As will be understood in the art, many of the medical conditions described herein are recognized by a prominent sign or symptom. For example, Alzheimer's disease is firmly associated with cognitive decline, multiple sclerosis, ALS, and Parkinson's disease are firmly associated with ataxia, post-traumatic stress disorders are firmly associated with anxiety, and so on. Accordingly, the present methods and uses of the compositions of the invention may be described equally well by reference to the condition itself or to a prominent sign or symptom thereof. For example, the methods of the in vention encompass treating a patient who has Alzheimer's disease as well as treating a patient who is experiencing cognitive decline.
Other aspects and embodiments of the invention are described ilsriher below,
BRIEF DESCRIPTION OF THE DRAWINGS
Fig, 1A Is a flow diagram illustrating the sequence of trials, duration of trials, intervals between trials, examples of object types and floor patterns involved in a Pattern Separation Test (PST) according to one embodiment of the invention.
Figs. 1B-1G are comparison charts illustrating the results of the Pattern Separation Test (PST) in Fig, 1A.
Figs. 2A-2F are comparison charts illustrating the results of a Sucrose Preference Test
( SPT) according to one embodiment of the invention.
Figs. 3 A-3D are comparison charts illustrating that 5-8mino-23-dihydro- i ,4- phthalazinedione, monosodium salt treatment normalizes the expression of oxidative stress response genes prdx6, sod2, sqstml and srxnl in GWI rats according to one embodiment of the invention.
Fig. 4 is a clustergram showing the expression of oxidative stress response genes in various animal groups according to one embodiment of the invention,
DETAILED DESCRIPTION
As described further herein, the invention features, inter liu^ compositions and methods fo diagnosing and treating patients who are suffering from a medical condition associated with brain dysfunction, including those conditions that manifest with ataxia. The composition can be a compound that conforms to the following Formula (I) or a pharmaceutical composition or formulation containing a compound that conforms to Formula (1);
Figure imgf000006_0001
Within Formula (I), Rj. and Ra can each he, independently, hydrogen (11), lithium (Li), sodium (Na)s potassium (K), rubidium (Rb), caesium (Cs), or franeiura (Fr); I¾ can be an alkyl, alkenyl, alkynly, aryl, alkoxyl, alkenyloxy, alkynyloxy, aryloxy, aikoxy carbonyl, a!kylamino, alkylthio, alkylstiifonyl, or alkyisuifmyi, each optionally substituted with an alkyi, halogen, aikoxy, aryl or heteroaryl moiety; Cs, Ci,€:¾, C-4, Cs, Gs, C?, and Cm can each be* independently, carbon 12 (32C) or an isotope of t2C (e.g.* i3Cv); Ns , 2 and M3 can each be, independently, nitrogen 14 (MN) or an isotope of i N (e.g., !5 ); Ot and O2 can each be, independently, oxygen i 6 (u,0) or an isotope of u,0 (e.g., O or f 80). ft is to be understood that the methods, uses, and compositions described herein can include either or both of a compound thai conforms to
Formula 1 or any other sal t thereof (e.g., a pharmaceutically acceptable sail thereof).
hi some embodiments (e.g., where the invention is the compound per e), the composition is a compound that conforms to Formula (!) with the proviso that when each of N1-N3 are j N and each of Ot and O2 are ,0, then each and every one of C2-C? cannot be C For example, the composition can be a compoimd that confbrnis to the foi lowing Formula (II):
Figure imgf000006_0002
in other embodiments, the composition is a compound that conforms to Forrnul (I) with the proviso that the compound is not N-cthanoy]-5-ammo-2,3-dihydrophthalazine-i,4-dione (also known as 5-(acetoxyamino)~2,3-dihydrophthalazine~! ,4~dione) or a salt thereof. For example, in some embodiments, the compound does not conform to foilowing Formula (HI)
Figure imgf000007_0001
5-(hexanoyl oxyan«no)~2,3-dihydrop halazine-l ,4~d5oae (also known as monosodium luxninoi henonyl) or a salt thereof. For example, in some embodiments, the compound does not conform to following Formula (IV :
Figure imgf000007_0002
or N-met yl-5-anuno-2 j~dihydrophibalazine-F4-dione (also known as 5-(naethyiamino}~2,3- dihydfophthalazine-'l ,4-dione) or a salt thereof. For example, in some embodiments, the compound does not conform to following Formula (V):
Figure imgf000007_0003
These compounds may also be excluded from one or more of the formulations, kits, or other compositions of matter described herein. Similarly, these compositions may be excluded from the prophyiactic, diagnostic, or treatment methods described herein.
In some embodiments, the compound is a sodium salt of 5«ammo«2,3-dihydro-l ,4« phthalazinedione (also described as 5~amino-2,3-dihydroplitlialazme~l,4~dione (iuminol)) as shown in following Formula (VI):
Figure imgf000008_0001
6~ammo-2,3-d ydrophthalazine-l ,4-dione, 5-amino-2)3-dihydrophthalazine-l,4-dion-8-yl (iuminyl), ' -bromo«5«amino 2^^i ydrophthala7ine«1 ,4-dione, -chloro-5«amiao»23- dihydrophihalazine-l ,4-dione, N~iiiKm -5-amiBO-2r3-d¾ydrophthaiaziiits- 1 ,4-dione, N-iodo-5- amlT -2?3-dihydrophdialazine-l,4-diooe, N-methyl-5-ammo-2 ^¾ydfophthalazine- 1 ,4-dione, N»eihyi-5"amino-2,3-dihydrophi¾alazifie~l ,4-dioi , N-propyl-5~amino-2 -dihydrophihalazine- 1 ,4-dione, N~isopropyi~5-amino-2,3-dihydropht alazine- 1 ,4-dione, N-methanoy.I-5~amino~2,3- dihydrophthalazine- ,4-dione, N-et1iaooyI-5-ara:rao-23- jhydrophthalazine- 1 ,4-dione, N- p.ropanoyI~5-ammo~2,3~dihydrophthaiazine-l. ,4-dione, N-hydroxyl~5~ammo~2,3- dihydrophthalazine- 1 ,4-dione, -earbox I -5-arabx>-23-dihydrophmalazine- 1 ,4-dione, N- methanol-5-amino-2,3-dihydrophthaia2dine- 1 ,4-dione, N-ethanoi-5-ammo-23^ihydix>phthaia2ane- 1 ,4-dione, N-propanol-5-amino-2,3-dihydrophthalazine- 1 ,4-dione, N-methenyi-5-amino-2,3- dibydrophthalazine-l ,4-dione, N~ethen>i-5-aniino-23-dihydrophihaia ine- 1 ,4-dione, N»propenyl- 5-amino-2,3-dibydrophtha!a2me- 1 ,4-dione, N-memoxy-5~amino-23-dihydmphthalazine-l,4- dione, N-et oxy-5-ainioo-23-dihydiOphtlialazine- 1 ,4-dione, N-pK>poxy-5-ammQ-2,3- dihydrophthalazme-l ,4-dione, N,N-dimethyI-5-amino-2 -dihydrophthaia2-ine- 1 ,4-dione, H- acetyIcysteme-5-ainino-2,3-dihy<irophfhalazine- 1 ,4-dione, N-acetylghiiathione-5-ammo~23~ dihydropht¾aI.azine-1 ,4-dione, 5-(hexanoyl oxyaniim>)-2,3~diIiydrophdialazrae- 1 ,4-dione, 5- (methy!amino)-2,3-dihydrophtbalazme- 1 ,4-dione, or 5-{acetoxyammo)-2,3-d.ihydropliiliakzine- ! ,4-dione, a pharmaceutically acceptable salt thereof (/. e. , of an of me foregoing), or an isotopic derivative thereof (i.e.. an isotopic derivative of any of the foregoing). For example, the agent can be 5-amino-2,3-dihydro- 1 ,4-phthalazinedione, monosodium salt, also known as 3- amiriophtlialhydrazide, sodium salt, 3-aroinophtha!.ic hydrazide, sodium salt, O-aminophthaioyi hydrazine, sodium salt, tamerit, or tamerite, or traderaarked as MSL€> and/or GVT®, In certain embodiments, the agent can be monosodiiim.5~amino-2!3-c1ihydro~ l. ,4~pbtha!a inedione (as of Formula (VI)), monosodiiim 5-meth.yIamiiK -2,3-dihydiO-I,4-plii:haiazinedione (as of Formula (V)), monosodium 5-aeetoxyamino-2,3-dihydro-l,4~ph.thalavi.inedk>ne (as of Formula (HI)), or monosodiiim 5-(hexanoyl oxylaraino)-23-dihydro-l ,4-ph(halazinedione (as of Formula (IV)). The diagnostic standards, pharmaceutical compositions, and kits of the invention can include one or more of (i.e., any combination of) the compounds described herein. By diagnostic standards, we mean formulations generated to serve as reference standards in a diagnostic or other assa conducted using mass spectrometry.
Patient's amenable to treatment: As described above, a medical condition treated by the pharmaceutical compositions described herein can be, or ca include as a prominent symptom, Lack of muscle control and, as a result, loss of full control over bodily movements (ataxia}, including lack of movement or of proper, controlled movement of the eye(s), difficulty walking or running in a normal, controlled manner (as can be detected and assessed by gait analysis), lack of coordination (as can be detected and assessed by any test for manual dexterity), aphasia, apraxia, or asthenia. A patient experiencing ataxia may have ALS, MS, or Parkinson's disease. The medical condition can also be, or can include as a prominent symptom, fatigue, Tourette's syndrome, narcolepsy, back and/ r neck pain, and headaches (including migraine headaches (e.g., visual migraines), cluster headaches, and tension headaches). In some embodiments, the patient ca exhibit signs of neuronal fatigue. In some embodiments, the patient can have multiple sclerosis. In some embodiments, the patient can have a multiple chemical sensitivity (MCS) which can, in torn, include a range of symptoms that may be attributed to exposure to chemicals tha are commonly used in building materials, industrial sites, and battle grounds, MCS has also been referred to as environmental illness, sick building syndrome, and idiopathic environmental intolerance. Where a patient is experiencing fatigue, the step of diagnosing the patient and/or monitoring the patient's treatment can include determining the rate of mitochondrial reco ver after a defined exertion (e.g., after exercise in a sample from the patient that includes, for
example, white blood cells). The rate of mitochondrial recovery or any other assessment of a patient can be compared to a reference standard defining a healthy/normal desired level, in other embodiments, the patient can be suffering from a mood disorder or mood dysfunction
bipolar disorder, depression, or schizophrenia); a memory disorder or memory dysfunction (e.g., as occurs with amnesia, Alzheimer's disease, dementia, or Huntington's disease); anxiety or a stress-related condition (e.g., a post-traumatic stress disorder); an acute or chronic brain injury, including an acute or chronic brain injur}' caused by mitochondrial dysfunction or an endogenous retrovirus; Gulf War Illness (GWI; as defined by the United States Veterans' Administration and also known as Gulf War Veterans' Illness or Gulf War Syndrome); acute or chronic fatigue, which may or may not occur in the context of GWI; and/or ALS. Any of the methods of the invention can include a step of assessing a patient's weight, and in some embodiments, patients who are obese or who have been diagnosed as having obesity-induced inflammation can be excluded from treatment, in other embodiments, the excluded patient can have a metabolic syndrome or medical conditio associated with heavy metal intoxication. In some embodiments, the mood disorder or mood dysfunction can be bipolar disorder, depression, schizophrenia, or can be associated with cancer, another chronic illness, infection, or substance abuse. The anxiety or stress-related disorder can be a posttraumatic stress disorder. Hie memory disorder or iiiemory dysfunction can be amnesia, or is associated with a diagnosis of Alzheimer's disease, dementia, Huntington's disease, or Parkinson's disease.
As noted, in some embodiments, the medical condition can. be associated with Gulf War
Illness (GWI) or can, more generally, be a. cognitive impairment associated with mitochondrial dysfunction or with other neuronal stresses, particularly those that impair neurogenesis in the hippocampaS region of the brain. Thus, the phamiaceuiicai compositions described herein can be administered to treat, cognitive impairment and/or to facilitate cognitive rehabilitation. While the present methods are intended to apply to GWI, the invention is not so limited. The patient can be one who has experienced battle or worked in a battle-torn area, regardless of the precise lime or place. Further, patients who have experienced the stress of preparing for military service are amenable to treatment, as are patients who support others who are preparing for service or recovering from a deployment (e,g., family members or close friends). The conditions amenable to treatment can manifest in various ways, and any given patient can be suffering from a variety of symptoms including, but not limited to, fatigue { .g., chronic fatigue syndrome), with mitochondrial dysftmction headache, memory problems, muscle or joint pain, muscle weakness (a taxia), diarrhea, dyspepsia, indigestion, or other gastrointestinal problems, other neurological problems, tumors or blood cancers, skin conditions, arthritis, or respiratory problems. In one embodiment; the patient has been diagnosed as having a post-traumatic stress disorder (PTSD), a chronic, multi-symptom illness, or a combination thereof.
As described above, the medical condition being treated by the present compositions may be caused by physical and psychological issues involving any war zone deployment (e.g., deployment during the Gulf War) or may have been caused by exposure to one or more hazards {e.g., toxins). In other words, the medical condition may have been
precipitated by chemical or environmental factors, particularly in die context of experiencing or preparing for a stressful situation such as .military deployment. The hazard may be, but is not limited to, exposure to a nerve gas such as sarin, a pyridostigmine bromide pill, a
depleted uranium munition, an anthrax vaccine, a botulinura vaccine, oil smoke from
burning oil, pesticides, or microwaves. The hazard may also be exposure to radiation (e.g., electromagnetic radiation).
in some embodiments, the medical condition may be associated with one or more neurological disorders, and these neurological disorders may, in turn, be precipitated by preparmg for, living or working within, or attempting to cope with a stressful and/or chemical-laden environment. For example, the neurological disorder may be associated with coping mechanisms that include addiction (e.g., alcohol or substance abuse). As noted above, many of these disorders manifest in ataxia, and any of the methods described herein can include a step of assessing a patient for ataxia prior to treatment.
Although the invention was developed with human patients in mind, it is not so limited. The present methods can be carried out for the benefit of any vertebrate animal including domesticated mammals (e.g., dogs and cats) and birds kept as pets or in zoos. The subject can also be an animal kept as livestock (e.g., cattle, sheep, chickens, horses, pigs, or goats). In other embodiments, the present compositions can be applied to a cell, tissue, organ, organ system, organism, or a medium containing one or more of these (e.g., in a laboratory or cell culture).
Formulations and dosages: As described above, a composition of the invention can be administered to the patient orally, topically, by inhalation, nasal delivery (at, for example, 50 to 100 mg/ml administered 5 or 6 times per day, for a daily dose of about 25- 1 ,000 mg, for about
20 days) to the brain or by an injection. Our work to date suggests that administration to the nasal passages, buccal mucosal (at, e.g. , about 200 to 500 mg daily for about 20 days) and/or sublingual tissue (at, e.g., about 25 mg to 50 rag four or five times per day) may be preferred. The
compositions intended for pharmaceutical use can also be formulated for auricular administration (to or by way of the ear), conjunctival administration (to or by way of the conjunctiva), a
cutaneous administration to the skin, dental or s racoronal administration {administration to a tooth or teeth, including to the portion of a tooth covered, by enamel), an electro-osmosis administration (e.g., administration through the diffusion of a substance through a membrane in an electric field), an endoeervical administration (e.g., administration within the canal of the cervix uteri), an endosinusial administration (e.g. , administration within the nasal sinuses of the head), an endotracheal administration (e.g., administration directly into the trachea), an enteral administration (e.g. , administration directly into the intestines), an epidural administration (e.g. , administration upon or over the dura mater), an extracorporeal administration (e.g., administration outside of the body), a hemodialysis (e.g., administration through hemodialysate fluid), an infiltration {e.g., administration that results in substances passing into tissue spaces or into cells), an interstitial administration (e.g., administration to or in the interstices of a tissue), an intra- abdominal administration (e.g. administratio within the abdomen), an mtra-arterial
administration (e.g., administration within an artery or arteries), an intra-articular administration (e.g., administration within a joint), an mtrabiliary administration (e.g., administration within the bile, bile duets or gallbladder), an intrabronehia! administration (e.g., administration within a bronchus), an intrabursai administration (e.g., administration within a bursa), a intracardiac administration (e.g. administration with the heart), an mtraeartilagmous administration (e.g., administration within a cartilage), an intracaudal administration (e.g., administration within the cauda equine), an intraeavernous administratio (e.g., administration, within a pathologic cavity, such as occurs in the lung in tuberculosis), an intracavitary administration (e.g., administration within a non-pathologic cavity, such as tha of the cervix, uterus, or penis, or such as that which is formed as the result of a wound), an intracerebral administration, (e.g., administration within the cerebrum), an intracistemal administration (e.g., administration within the eisteraa magna cerehe!iomedularis), an intracoraeal administration (e.g., administration within the cornea), an intracoronary administration (e.g., administration within the coronary arteries), an. iutracorporus cavernosum (e.g., administration within, the dilatable spaces of the corporus cavernosa of the penis), an ntradermal administration (e.g., administration within the dermis), an intradiscal administration (e.g., administration within a disc), an intraductal administration (e.g.,
administration within the duct of a gland), an intraduodenal administration (e.g.. administration within the duodenum), an intradural administration (e.g., administration within or beneath the dura), an intraepidermal administration (e.g., administration within the epidermis), an
intraesophageal administration (e.g., administration within the esophagus), an intragastric administration (e.g., administration within the stomach), an intragingival administration (e.g., administration within the gingivae), an intraileal administration, an intralesional administration (e.g., administration within or introduced directly into a localized lesion), an intraluminal administration (e.g., administration within the lumen of a tube), an intralyniphatic administration (e.g., administration within the lymph), an intramedullary administration (e.g. , administration within the marrow cavity of a bone), an intrameningeal administration (e.g., administration within the meninges), an intramuscular administration, an intraocular administration (e.g. , administration within the eye), an intrapericardial. administration (e.g., administration within the pericardium), an intraperitoneal administration, an intrapleural administration (e.g.,
administration within the pleura), an intraprostatic administration (e.g., administration within the prostate gland), an trapulraonary administration (e.g., administration within the lungs or its bronchi), an intrasinal administration (e.g., administration within the nasal or periorbital sinuses), an intraspinal administration (e.g., administration within the vertebral column), an intrasynovial administration (e.g., administration within the synovial cavity of a joint), am intratendinous administration (e.g., administration within a tendon), an intratestkular administration (e.g., administration within the testicle), an intrathecal administration (e.g. , administration within the cerebrospinal fluid at any level of the cerebrospinal axis, including injection into the cerebral ventricles), an intrathoracic administration (e.g., administration within the thorax), an in ratubuiar administration (e.g., administration within the tubules of an organ), an intratumor administration (e.g., administration within a tumor), an intratympanic administration (e.g., administration withi the aurus media), an intrauterine administration (e.g. , administration within the uterus), an intravascular administration (e.g., administration within a vessel or vessels), an intravenous administration (e.g., administration within or into a vein or veins), an intravenous bolus administration (e.g., administration within or into a vein or veins all at once), an intravenous drip administration (e.g. , administration within or into a vein or veins over a sustained period of time), a intra vesical administration (e.g., administration within the bladder), an i travitreal
administration (e.g., administration within the vitreous body of the eye), an iontophoresis (e.g., administration by means of an electric current where ions of soluble salts migrate into the tissues of the body), an irrigation (e.g., administration to bathe or flush open wounds or body cavities), a laryngeal administration (e.g., administration directl upon the larynx), a nasal administration (e.g., administration to the nose; administered by way of the nose), a nasogastric administration (e.g., administration through the nose and into the stomach, usually by means of a tube), an occlusive dressing technique (e.g., administration by the topical route which is then covered by a dressing which occludes the area), an ophthalmic administration (e.g., administration to the external, eye), an oral administration (e.g., administration to or by way of the mouth), an oropharyngeal administration (e.g., administration directly to the mouth and pharynx), a parenteral administration (e.g., administration by injection, infusion, or implantation), a percutaneous administration (e.g., administration through the skin), a periarticular administration (e.g, administration around a joint), a peridural administration (e.g. , administration to the outside of the dura mater of the spinal cord), a perineural administration (e.g., administration surrounding a nerve or nerves), aperiodontai administration (e.g., administration around. a tooth), a rectal administration (e.g.. administration to the rectum), a respiratory administration (e.g.,
administration within the respiratory tract by inhaling orally or nasally for local or systemic effect), a retrobulbar administration (e.g., administration behind the pons or behind the eyeball), a soft tissue administration (e.g. , administration into any soft tissue), a subarachnoid administration (e.g., administration beneath the arachnoid), a subconjunctival administration (e.g., adminisiraiion beneath the conjunctiva), a subcutaneous administration (e.g., administration beneath the skin), a sublingual administration (e.g., administration beneath the tongue), a submucosal administration (e. ., administration beneath the mucous membrane), a topical administration (e.g.,
administration to a particular spot on the outer surface of the body), a transdermal administration (e.g., administration through the dermal layer of the skin to the systemic circulation by diffusion), a rransmucosal administration (e.g., administration across the mucosa), a
transplacental administration (e.g. , administration through or across the placenta), a transtracheal administration (e.g., administration through the wall of the trachea), a transtympanic
administration (e.g., administration across or through the tympanic cavity), an ureteral administration {e.g., administration into the ureter), an urethral, administration (e.g.,
administration into the urethra), a vaginal administration (e.g., administration into the vagina), or a combination thereof.
In considering the formulations and dosages to be administered, one can invoke principles of personalized medicine, and such regimens are encompassed by the present methods. During the administration of the composition, a dose level can vary as a function of the one or more compositions, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given composition are readily determinable by those of skill in the art by a variety of means. In many embodiments, multiple doses of one or more compositions are administered.
The pharmaceutical compositions may be administered one to three times per month, every other week (qow), 1 -6 times per week, every other day (qod), daily (qd), twice a day (qid), or three times a day (tid), over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from, about eight months to about I. year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more. For example, the composition can be administrated five times a week for eight weeks. In severe eases, the patient may require administration at one or more of these delivery doses for the remainder of his or her life.
In connection with the above-described methods, a composition as described herein may be administered to the patient at a dosage from about 10 .mg/kg to 500 mg kg patient body weight per day, in 1 to 5 divided doses per day. For example, the composition may be administered at a dosage of about 40 mg kg, about 80 mg/kg, or about 160 mg/kg patient body weight per day.
Where liquid or powdered t rmuiations are prepared for administration to a mucous membrane
(e.g. , within the nose or mouth), the compound can be present at between about 50-100 mg/ml (e.g. , 70 mg ml). The term "about" is used herein to indicate that a value includes an inherent variation of error for the device or the method being employed to determine the value or plus-or- minus 10% of the value, whichever is greater.
In various embodiments, the composition can be formulated, in the form of a pill, a capsule, a granule, a tablet, a pallet, a suspension, an injection, an infusion, a suppository, a continuous delivery system, a syrup, a tincture, an ointment, a cream, eye drops, eardrops, a flush, a lavage, a slow absorbing depot a dressing, a. lozenge, or any pharmaceutically acceptable application or as a nutritional supplement.
The compounds disclosed herein can be formulated with conventional carriers and
excipients, which can be selected in accord with ordinary practice. Tablets can typically contain excipients, glidanis, fillers, binders and the like. Aqueous formulations can be prepared in sterile form, and when intended for delivery by other than oral administration generally can be isotonic.
Formulations can contain excipients (e.g., excipients set forth in the Handbook of Pharmaceutical
Excipients, 5th Ed.; Rowe, Sheskey, and Owen, Eds.; American Pharmacists Association;
Pharmaceutical Press: Washington, DC, 2006).
Excipients can include ascorbic acid or other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkyiniethylceSlulose, stearic acid or the like.
The pH of the t rmuiations can. range from about 3 to about 1 1 , and can be about 7 to 10. While it is possible for the active ingredients to be administered alone it can be preferable to present them as pharmaceutical formulations. The formulations of the agent, as disclosed herein, can include at least one active ingredient, as above defined, together with one or more acceptable carriers thereof and optionally other therapeutic ingredients. The carrieris) should be "acceptable" in the sense of being compatib le with the other ingredients of the formulation and physiologically innocuous to the recipient thereof. The formulations can include those suitable for the foregoing administration routes. The formulations can conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally can be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Easion, PA, (1985). Such methods can. include the step of bringing into assoeiation the active ingredient with the carrier thai constitutes one or more accessory ingredients. In general the formulations can be prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers, finely divided solid carriers, or both, and then, if necessary, shaping the product. Formulations of the presently disclosed subject matter suitable for oral administration can be presented as discrete units such as capsules, cachets, or tablets each
containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil -in- water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient can also be administered as a bolus, electuary, or paste.
A tablet can be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets can be coated o scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom. For administration to the eye or other externa! tissues (e.g., mouth and skin), the formulations ca be applied as a topical ointment or a cream containing the active ingredients). When formulated in an ointment, the active ingredients can be employed with either a paraffnie or a water-miseible ointment base. Alternatively, the active ingredients can be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase of the cream base can include at least 30% w/w of a po!yhydric alcohol (e.g., an alcohol having two or more hydroxy! groups such as propylene glycol, butane 1 ,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof). The topical formulations can include a compound (e.g.. dimethyl sutphoxide or related analogs) that enhances absorption or penetration of the active ingredient through the ski or other affected areas. The oily phase of the emulsions of this disclosed subject matter can be constituted from known ingredients in a known manner. The phase cart include merely an emulsifier (otherwise blown as an emulgent) or a mixture of at least one emulsifier with, a fat or an oil or a
combination thereof. Preferably, a hydrophilie emulsifier can be included together with a lipophilic emulsifier that acts as a stabilizer. It is also preferred to include both an oil and a fat Together, the emulsitier(s) with, or without stahiiizer(s) can make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base that forms the oily dispersed phase of the cream formulations, Emulgents and emulsion stabilizers suitable for use in the formulation of the agent, as disclosed herein, can include
TWEEN 60, SPAN 80, cetostearyl alcohol, benzyl alcohol, myristy.1 alcohol, glyceryl mono- stearate, or sodium lauryl sulfate.
The choice of suitable oils or fats tor the formulation can be based on achieving the desired cosmetic properties. The cream can be a non-greasy, no -staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters (e.g., diisoadipate, isocetyi stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyi oleate, isopropyl palmitate, butyl stearate, 2- ethylhexyl palmitate or a blend of branched chain esters (Crodamol CAP)) may be used. Iliese may be used alone or in combination depending on. the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
Pharmaceutical formulations can. include the agent together with one or more
pharmaceutically acceptable carriers or exeipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingredient can be in any form suitable for the intended method of administration.
When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs can be prepared.
Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient that is suitable for manufacture of tablets can be acceptable. These excipients can be inert diluents (e.g., calcium or sodium carbonate, lactose, lactose monohyd ate, crosearraellose sodium, povidone, calcium or sodium phosphate), granulating and disintegrating agents (e.g. , maize starch or algtnle acid), binding agents (e.g., cellulose, mieroeryslallme cellulose, starch, gelatin or acacia), or lubricating agents (e.g. , magnesium stearate, stearic acid or talc). Tablets can be uncoated or coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained, action over a longer period. For example, a time delay material such as glyceryl raonostearate or glyceryl distearate alone or with a wax can be used.
Formulations for oral use can be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluents (e.g., calcium phosphate or kaolin), or as soft gelatin capsule wherein the active ingredient is mixed with water or an oil medium (e.g., peanut oil liquid paraffin, or olive oil).
Aqueous suspensions of the agent, as disclosed herein, can contain the active materials in admixture with excipients suitable for the manufactur of aqueous suspensions. Such excipients can include a suspending agent (e.g., sodium carboxymethyiceilulose, mefhylceiluiose,
hydroxypropyl methyl.eell.uose, sodium alginate, polyvinylpyrrolidone, gum iragacanih or gum acacia), a dispersing or wetting agent such as a naturally occurring phosphatide (e.g. , lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., poiyoxyethylene stearate), a condensaiion product of ethylene oxide with a long chain aliphatic alcohol (e.g..
hepiadecaethyleneoxyceianol), or a condensaiion product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g. , poiyoxyethylene sofbitan monoo!eate). The aqueous suspensio can also contain one or more preservatives (e.g., ethyl or n-propy! p- hydroxy-benzoate), one or more coloring agents, one or more flavoring agents and one or more sweetening agents (e.g., sucrose or saccharin). Oil suspensions can be formulated by
suspending the active ingredient in a vegetable oil (e.g., araehis oil, olive oil, sesame oil, or coconut oil), or in a mineral oil (e.g., liquid paraffin). The oral suspensions can contain a thickening agent (e.g., beeswax, hard paraffin, or cetyl alcohol). Sweetening agents and/or flavoring agents can be added to provide a palatable oral preparation. These compositions can he preserved by the addition of an antioxidant such as ascorbic acid,
Dispersible powders and granules of the agent, as disclosed herein, can be suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more
preservatives. Suitable dispersing or wetting agents and suspending agents can be exemplified by those disclosed above. Additional excipients (e.g., sweetening, flavoring and coloring agents) can also be present,
The pharmaceutical compositions of the agent, as disclosed herein, can also be in the form of oil -in- water emulsions. The oily phase can be a vegetable oil (e.g., oliv oil or arachis oil), a mineral oil (e.g., liquid paraffin), or a mixture of these.
Formulations for oral use can also be presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent (e.g., calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium (e.g., peanut oil, liquid paraffin, or olive oil).
Aqueous suspensions of the agent, as disclosed herein, can contain the acti ve materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such
excipients can include a suspending agent (e.g., sodium carboxymethylceilulose,
methykellulose, hydroxypropyl methyleelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth or gum acacia), a dispersing or wetting agent (e.g. , a naturally occurring
phosphatide (e.g., lecithin)), a condensation product of an alkyiene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanoi), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g.,
polyoxyethylene sorbitan monooieaie). The aqueous suspension can also contain one or .more preservatives (e.g., ethyl or n-propyl p-hydroxy~ben¾oate), one or more coloring agents, one or more flavoring agents and one or more sweetening agents (e.g.) sucrose or saccharin). Oil suspensions can be formulated b suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil sesame oil, or coconut oil, or in a mineral oil (e.g. , liquid paraffin). The oral suspensions can contain a thickening agent (e.g., beeswax, hard paraffin, or cetyl
alcohol). Sweetening agents or flavoring agents can be added to provide a palatable oral preparation. These compositions can be preserved by the addition of an antioxidant such as ascorbic acid.
Dispersible powders and granules of the agent, as discl osed herein, can be suitable tor preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture wi h a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents can be exemplified by those disclosed above. Additional excipients (e.g., sweetening, flavoring and coloring agents) can also be present.
The pharmaceutical compositions of the agent, as disclosed herein, can also be in the form of oil -in- water emulsions. The oily phase can be a vegetable oil (e.g., olive oil or arac s oil), a mineral oil (e.g., liquid paraffin), or a mixture of these. Suitable emulsifying agents can include naturally occurring gums (e.g., gum acacia and gum tragaeanih), naturally occurring phosphatides (e.g., soybean lecithin), esters or partial esters derived from fatty acids and hexitol anhydrides (e.g.. sorbitan monooieate), and condensation products of these partial esters with ethylene oxide (e.g., pol oxyethy!ene sorbitan monooieate). The emulsion can also contain sweetening and flavoring agents. Syrups and elixirs can be formulated with sweetening agents (e.g., glycerol, sorbitol, or sucrose). Such formulations can also contain a demulcent, a preservative, a flavoring or a coloring agent.
The pharmaceutical compositions of the agent, as disclosed herein, can be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
This suspension can be formulated according to the known art. using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation can also be a sterile injectable solution or suspensio in a non-toxic parenteraUy acceptable diluent or solvent (e.g., a solution in 1,3-butane-diol. or prepared as a Jyophi!ized powder).
Among the acceptable vehicles and sol vents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils can be
conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed (e.g., synthetic mono- or diglycerides). Fatty acids (e.g., oleic acid) can also be used in the preparation of injec tables.
The amount of active ingredient that can be combined with the carrier material to produce a single dosage form should vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral
administration to humans may contain approximately 1 to 1000 mg of active material
compounded with an appropriate and convenient amount of carrier material that may vary from about 5 to about 95% of the total compositions (weight: weight).
The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 pg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
Formulations suitable for administration to the eye can include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient The active ingredient can he present in such formulations in a
concentration of 0.5 to 20%, 0.5 to 10%, or 1.5% to 8% (w/w).
Formulations suitable for topical administration in the mouth can include lozenges including the active ingredient in a flavored basis, typically sucrose and acacia or tragacanth; pastilles including the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes including the active ingredient in a suitable liquid carrier.
Formulations for rectal administration can be presented as a suppository with a suitable base including for example cocoa butter or a salicylate. Formulations suitable for intrapulraonary or nasal administratio can. have a particle size in the range of 0.1 to 500 microns (including particle sizes in a range between 0,1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which can be administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations can include aqueous or oily solutions of the acti ve ingredient. Formulations suitable for aerosol or dry powder administration can be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of a given condition. Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams or spra formulations containing in addition to the active ingredient such carriers as are known in the ar to be appropriate.
Formulations suitable for parenteral administration can include aqueous and nonaqueous sterile injection solutions which can contain aiiti-o idants, buffers, bacteriostais and solutes which render the formulation isotonic wi th the blood of the in tended recipient ; and aqueous and nonaqueous sterile suspensions which can include suspending agents and thickening agents.
The formulations can be presented in unit-dose or multi-dose containers (e.g. , sealed ampoules and vials) and can be stored in a freeze-dried (lyophilized) condition requiring the addition of the sterile liquid carrier (e.g., water) for injection, immediatel prior to use.
Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules and tablets of the kind previously described. 'Preferred uni dosage formulations can be those containing a daily dose or unit daily sub-dose, as herein above recited, or an
appropriate fraction thereof, of the active ingredient.
In addition to the ingredients particularly mentioned above, the formulations of the agent, as disclosed herein, can iticlode other agents conventional in the art having regard to the type of formulation in question (e.g., those suitable for oral administration can include flavoring agents).
The agent, as disclosed herein, can also be formulated to provide controlled release of the active ingredient to allow less -frequent dosing or to improve the pharmacokinetic or
toxicity profile of the active ingredient. Accordingly, the agent, as disclosed herein, can also be provided in compositions including one or more agents formulated for sustained or
controlled release.
An effective dose of active ingredient can depend at least, on the nature of the condi tion being treated, toxicity, whether the compound is being used prophylacticaily (typically lower doses), the method of delivery, and the pharmaceutical formulation, and is determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day, typically, from about 0.01 to about 10 mg/kg body weight per day, more typically, from about 0.01 to about 5 mg/kg body weight per clay, and more typically, from about 0.05 to about 0.5 mg/kg body weight per day.
If desired, the compounds of the presently di sc l osed subject matter can be appl ied in conjunction with one or more inert or inactive ingredients. The agent, as disclosed herein, can be administered by any route appropriate to the condition to be treated. Suitable routes can include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intraderma!, intrathecal and epidural), and the like.
Diagnostic ami Pharmaceutical Kits: As described further herein, the invention also features kits for aiding in the diagnosis and treatment of a patient who is suffering from a medical condition associated with brain dysfunction or as oihenvise described herein. The kit may include a therapeutically effective amount of one of more of the compositions described above.
In some embodiment, the agent can be used in combination with other active ingredients. The combinations can be selected based on the conditio to be treated, cross-reactivities of ingredients and pharmaco-properties of the combination. The agent can also be combined with one or more other active .ingredients in a unitar dosage form for simultaneous or sequential administration to a patient by the same or different routes of administration. The combination therapy can be administered as a simultaneous or sequential regimen. When administered sequentially, the combination can be administered in. two or more admin.istrati.ons.
In genera!, during alternation therapy, an effective dosage of each active ingredient can be administered sequentially ( .& , serially), whereas in combination therapy, effective dosages of two or more active ingredients can be administered together. The combination therapy may provide "synergy" and "synergistic effect" (i.e., the effect achieved when the active ingredients nsed together is greater than the sum of the effects thai results from using the compounds
separately). In certain embodiments, a synergistic effect can be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. In alternation therapy, the synergistic effect can also be attained when the compounds are administered or delivered sequentially (e.g. , in separate tablets, pills, or capsules, or by different injections in separate syringes).
In some embodiments, pharmaceutical kits useful in the presently disclosed subject matter, which can include a therapeutically effective amount of a pharmaceutical composition including (a) a compound of component and/or (b) one or more compounds of component, in one or more sterile containers, can also be within the ambit of the presently disclosed subject matter. Sterilization of the container can be carried out using conventional sterilization methodology wel l known to those skilled in the art. Component (a) and/or component (b) can be in the same sterile container or in separate sterile containers. The sterile containers or materials can include separate containers, or one or more multi-part containers, as desired. Component (a) and/or component (b) can be separate, or physically combined into a single dosage form or unit The kits can further include one or more of various conventional pharmaceutical kit components (e.g., one or more pharmaceutically acceptable carriers, additional vials for mixing the components), as should be readily apparent to those skilled in the art. instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for adrainisterion, and/or guidelines for mixing the components, can also be included in the kit.
Evaluation Methods: The methods described herein may include a step to evaluate the effect of the composition (e.g., by assessing a cognitive function or mood of the patient). For example, the methods can include a Pattern Separation Test (PST) for assessing cognitive function, a Sucrose Preference Test (SPT) for assessing mood function, or both.
Alternatively or in addition, the evaluation step can include evaluating an oxidative stress response or determining the expression level of one or more antioxidant genes in the patient. The gene may be prdx6, sad- 1, sad2, sgstml, srxnl, a cat gene (encoding catalase protein), a cisb gene (encoding eathepsin B), a dhcr24 gene (encoding 24- dehydrocholesferol reductase), a gsr gene (encoding glutathione -reductase), a gstkl gene (encoding glutathione s- transferase kappa 1), & gsipl gene (encoding glutathione s- transferase- 1 ), an idhl gene
(encoding isocitrate dehydrogenase 1), an ncfl gene (encoding neutrophil cytosotic factor 1 protein), one or more of the prdx.1-4 genes (encoding peroxiredoxins 1-4), a prnp gene
(encoding prion protein), a ptgsl gene (encoding prostaglandm-eiidoperoxide synthase), an sic3Sa! gene (encoding solute carrier family 38), a txni gene (encoding ihioredoxin 1), a ixnip gene (encoding ihioredoxin inieraciing protein), a txnrdl gene (encoding thioredoxin reductase 1 ), a txnrd2 gene (encoding thioredoxin reductase 2), or an ucp2 gene (encoding uncoupling protein 2). Alternatively or in addition, the evaluation may include a step of determining the concentration of 3-nitT lyrosine, measuring. The evaluation may include a step of determining the level of endogenous retrovirus Herv-K in (Lymphoma and ALS) and other environmentally activated endogenous retroviruses (e.g., retroviruses activated by oxidative stress).
hi some embodiments, the evaluation may use a model system. The model, system can be an animal model of disease, a cell culture system, an in vitro system, a mathematical model {e.g., a computational model), or a test carried out with a selected population of subjects (humans participating in a clinical trial).
Preventative Methods: As another respect, the present invention features a method for preventing a patient from developing a medical condition associated with brain dysfunction (or an of the particular conditions described herein). These methods include administering to the paiient an amount of a composition described herein that is effective in reducing the likelihood thai the patient, will develop a give condition (as described herein).
It is to be understood thai this invention is not limited to particular variations set forth and may, of course, vary. Various changes may be made to the inventio described, and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process aci(s) or step(s), to the objective(s), spirit or scope of the present invention. All such modifications are intended to be within the scope of the claims made herein.
EXAMPLES We have quantified the efficacy of oral administration of different doses of (via oral gavage) for: ( 1) suppressing oxidative stress and inflammation and (2) stimulating the
proliferation of hippocampai neural stem cel ls ( SCs) and increasing the extent of net neurogenesis in rats exposed to GWI-related (GWIR) chemicals and moderate levels of stress four months earlier (GWI-rats). We also aimed to identify an optimal dose of 5-amino~2,3~ dthydro-i .,4-phthalazinedione or the monosodium salt thereof mat greatly regulates inflammation and oxidative stress and normalizes hippocampai neurogenesis in rats exposed to GWIR- chemicals and moderate levels of stress. In our studies neuronal fatigue was shown by
determining the rate of mitochondrial recovery in the white blood cells after exercise,
Description of Experimental .Procedures: The following narrative describes studies that have been accomplished. The experiments comprises 5 major groups of rats:
Group I; GWI-rats receiving 5-amino-2,3-dihydro- 1 ,4-phthalazinedione at 40 mg/kg b.w.; Group 2: GWI-rats receiving 5-amino-2,3-dihydro- 1 ,4-phthalazinedione at 80 mg/kg b.w.; Group 3: GWl-rats receiving 5-ammo-2,3-dihydro- 1 ,4-phthalazinedione at 160 mg/kg b.w.; Group 4: GWI-rats receiving vehicle fVEH); and Group 5: Age-matched naive control rats.
Animal numbers, survival and tissue harvesting: A total of 1 15 rats have been purchased so far in three different cohorts. The first cohort comprised 31 animals at the beginn ing of the study. Of those, 29 animal s reached the endpoint of experiments; two animals were found dead during the four-month waiting period between the exposure of animals to GWIR chemicals and 15 minutes of restraint stress for 28 days and the commencement of 5 -amino~2,3-di ydro- 1 ,4- phthalazinedione treatment. The brain tissues from 29 animal were harvested for biochemical and molecular biological studies within these fi ve groups: ( ! ) 5-amino-2,3-dihydro- 1 ,4- phthalazinedione, 40 mg/kg, n::::6; (2) 5-araino-23~dihydro~ll«4~phthalazinedione, 80 mg/kg, tv::6; (3) 5-amino-2,3-dihydro- 1 ,4-phthalazmedione, 160 mg kg, n~5; (4) GWI-vehicle (VEH), n-:6; and (5) naive control, n=6. The second cohort comprised 42 animals at the beginning of the study. Of these, 39 animals reached the endpoint of experiments. Two animals were found dead during the four-month waiting period between the exposure of animals to GWIR chemicals and stress and the commencement of 5-ammo-23^ihydro-l ,4-phthalazinedione treatment. An additional animal was euthanized in this period because it developed uncontrolled seizures, typified by continuous Stage- V seizures (bilateral forelimb clonus with rearing and falling). The brain tissues from 39 animals were harvested for immunoMstochemical studies, within these five groups: (I) 5-amino-2,3-dihydRV'1 ,4-phthalazinedione, 40 mg/kg, n-8; (2) 5-amino~2,3~dihydro- Ϊ , -phthala¾inedione, 80 mg kg, n~8; (3) 5-amiiK -2J-djliydro-1 -phthalaMnedione, 160 mg kg, n-8; (4) GWI-VEH, n™8; and (5) naive control n-7. The third cohort comprised 42 animals at the beginning of the study. Of these, 34 animals were exposed to GWIR-cbemicals and stress and 8 animals were maintained in a control group. Animals exposed to GWIR-chemicals and stress were assigned to VEH or 5-amiH -253-dihydro~l?4-phtha!.axhiedione treatment groups after they completed a 4-month waiting period,
'rime-line of various procedures for animais in cohorts ί and 2: Animals were exposed daily to GWIR-chemicals and stress for 28 days, and were subjected to a survival period of
4 months between exposure and treatment. The animals were then treated 5 times a week with 5-amiiu -2,3-dihydro-l ,4-phthaiazinedione or VBH for 8 weeks, 5'-bramodeoxyuridme (BrdU) is injected for 5 days in the 3rd week of treatment. The animals are then subjected to cognitive and mood function tests starting from the fifth week of treatment Finally, euthanasia and tissue harvesting was performed after 8 weeks of treatment.
Exposure of animals to GWIR-chemicals and stress: Animals were exposed daily to the following chemicals for 28 days: pyridostigmine bromide (PB) at 2 mg/kg/da (via oral gavage), DEET at 60 mg kg/day (via dermal application) and permethrin at 0.2 mg kg/day (via dermal application). In addition, animals were subjected dally to 15 minutes of restraint stress using rat restrainers during the above 28-day period.
Survival period between exposure and treatment: Following the exposure to GWIR chemicals and stress, animais were maintained in the vivarium for four months in regular cages (two per cage) with ad libitum access to food and water. Administration of 5-amino-2s3- dihydro- 1 ,4-phthalazinedione or VEH: Treatment was given for 8 weeks (5 times/week) via oral gavage, commencing in the 5th month after exposure to GWIR chemicals and stress. The doses of 5-amino-2,3-dihydro- 1 ,4-phthalazinedione employed were 40 mg kg, 80 mg/kg and 160 mg/kg.
BrdU injections: Subgroups of rats fr m all groups received BrdU injections in the 3rd week of drug/ vehicle treatment dally for 5 days at a dose of 100 mg/kg/day.
Behavioral tests for assessing cognitive and mood function: We examined animals in all groups through stress- free behavioral tests. We used a Pattern Separation Test (PST) to assess cognitive function, and a Sucrose Preference Test (SPT) to assess mood function. Euthanasia and tissue harvesting: Animais belonging to cohort 1 were deeply anesthetized with isofiurane in a small chamber until respiration ceased. Deepl anesthetized animais were decapitated following thoracotomy and brain tissues were dissected rapidly for biochemical and molecular biological studies. Animals belonging to cohort 2 were first deeply anesthetized with isoflurane and then perfused through the heart with 4% paraformaldehyde solution. Fixed tissues were harvested for histological studies.
Analyses of oxidative stress: The hippocampal tissues obtained from animals belonging to cohort 1 were used for the following .measurements . First, we analyzed the oxidati ve stress response and the expression of antioxidant genes using the Rat Oxidative Stress Response PGR Array from Qiagen. We analyzed the expression of 84 key genes involved in oxidative stress response and antioxidant activity in the hippocampus of animals belonging to different groups using quantitative real time PGR (qR'f-PCR.) to ascertain the effects of 5-amino-2s3-dihydro-L4- phthaiazinedione treatment on oxidative stress. Second, we measured lipid peroxidation through quantification of malondiakleh de. lipid peroxides are unstable indicators of oxidative stress in cells that decompose to form more complex and reactive compounds such as malondialdehyde (MDA), a natural by-product of lipid peroxidation. Hence, we .measured DA in hippocampal tissue extracts from different groups of animals using a TEARS Assa Kit, Third, we quantified 3-niirotyrosine. Increased modification of tyrosine residues in proteins to 3-nitrotyrosine by peroxynitriie or other potential nitrating agents is seen in. tissues subjected to oxidative stress. Hence, we quantified 3-nitrotyrosine in. hippocampal tissue extracts from different groups, using the nitrotyrosine EL ISA. Kit,
Analyses of inflammation: The hippocampal tissues obtained from animals belonging to cohort 1 were also used to measure the relative levels of inflammatory cytokines in different groups of animals. We employe the Rat Cytokine Plate Array from Signosis, which facilitated analyses of 16 rat cytokines in a high-throughput manner. The cytokines included: tumor necrosis factor-alpha (TMF-a), interleukin-I alpha (IT- la), interleiuan- 1 beta (iL-Ι β), vascular endothelial growth factor (VEGF), fibroblast growth factor beta (FGFp), interferon gamma
(IFNy), interleukin-5 (IL-5), in.terleukin-6 TL-6), interleukin- 15 (IL-15), leptin, monocyte chemoattractant protein- 1 (MCP-1), lF >gamma-md«cible protein 10 (IP- 10 or CXCLI ), stem cell factor (SCF), Regulated on Activation, Normal. T Cell Expressed and Secreted (Rantes), Macrophage inflammatory protein 1 alpha (MlP-l a) and transforming growth factor-bete (TGFP).
Inimunohistocheniical studies: Fixed brain tissues obtained from animals belonging to cohort 2 were processed for cryostat sectioning. Serial sections (every 1.5 til or 20th) through the entire hippocampus are currently being processed for immunohistochemicai detection of BrdU* eells (i.e. newly born cells), doublecortin (OCX, a marker of newly born neurons), glial fibrillary acidic protein (GFAP, a marker of astrocytes), 3BA-1 (a marker of all microglia) and ED-1 a marker of activated microglia. These multiple cell types will be quantified using stereologv in the coming year (second year of the project).
Dose-response studies conducted so far using S-amino-23-dihydro- i .4- phthaiazmedione in GWI rats suggest the following.
5^mino*2 ^ihydra'I,4'phthaIazmediane treatment at higher doses improves cognitive fijtncthm in GWI rats: We examined the cognitive ability of GWI rats belonging to different groups (n:;;;l 1 34/giOup) through a pattern separation test (PST). Pattern separation function reflects proficiency for discriminating analogous experiences through storage of similar
representations in a non-overlapping manner (Leutgeb ei a/.. Science, 315:961-966, 2007; Yassa and Stark, Trends Neurosci, 34:515-525). In this test, each rat successively explored two different sets of Identical objects (object types 1. and 2) placed on distinct types of floor patterns (Patterns 1 and 2 [PI and P2J) for 5 minutes each in the two acquisition trials (separated by
30 minutes). Thirty minutes later, in the testing phase (Trial-3), each rat explored an object from trial 2 (which is now a familiar object) and an object from Trial- 1 (which is now a novel object) placed on the floor pattern employed in trial 2 (P2).
Figs, lA-l G illustrate the Pattern Separation Test (PS T) and its results. Fig. 1.A illustrates the sequence of trials, duration of trials, intervals between trials, examples of object types and floor patterns in vol ved in this test. Figs. IB- 1 F compare percentages of object exploration time spent with the familiar object on pattern 2 (Fa on P2) and the novel object on pattern 2 (NO of P2) in different animal groups (n=l 1 -14/group). Cyan, Naive control group; red, GWi+VEH group; orange, 5-amino-2,3-dihydro-l,4-phthalazinedione 40 mg/Kg group; yellow, G'WJ+5-amino-2,3- dihydro- 1 ,4-phthalazinedione, 80 mg/Kg group; green, GWI+5-amko-2,3-dihydro-l ,4- phthalazinedione, 160 mg/Kg group. Fig. 1 G compares the total object exploration time between groups in Trial-3. One-way ANOVA analysis did not show differences between groups, implying that the specificity of the novel object exploration time (NO on P2) was not influenced by differences in the total object exploration time.
(i) Excellent pattern separation ability (i.e. , ability to distinguish between similar
experiences) in naive rats was revealed by a greater exploration of the object from trial 1 (i.e. novel object on pattern 2 [NO on P2 j) than the object from trial 2 (i.e. iamiiiar object o pattern 2 [FO on P2], p<0.0001 , Fig. 1).
(it) GWI rats that received VEH (GWI+VEH) showed no preference for the NO on P2, as they spent nearly similar amounts of time with novel and familiar objects on P2 (Fig. 1 ), implying loss of ability for pattern separation. Previous studies have shown that t is task requires normal levels of dentate neurogenesis (Jain ei /., PLoS One, 7:e46340, 2012; cAvoy ei a/., Front. Syst. Neurosel^ 9:120, eColleciion, 201 S; Oomen ei ah, Wiley Inteniiscip, Rev. Cogn. Set. 5:573-587, 2014). However, it remains to be examined whether these rats display decreased levels of dentate neurogenesis.
(iii) GWI rats thai received lower doses of 5 5-amino~2,3~dihydro- 1 ,4-phthalazinedione (40 mg/kg) remained impaired, which was evidenced by their greater exploration of the object from trial 2 (FO on P2|) than the object from trial 1 (NO on P2, p<Q,05, Fig. 1), suggesting that a low dose of 5~ainino-2J3-dihydro~l,4~phthalazinedione is not efficacious for reversing pattern separation dysfunction. GWI rats that received moderate doses of 5-arnino-2,3-dihydro- 1 ,4- phthalazinedione (80 mg/kg) also remained impaired, as they spent similar amounts of time with novel and familiar objects on P2 (p>0.05, Fig. 1 ). In contrast, GWI rats that received higher doses of 5-amino-2,3-dihydro- 1 ,4-phthalazinedione (160 mg/kg) displayed an ability for pattern separation. This was revealed by their greater exploration of the object from trial 1 (NO on P2) than the object from trial. 2 (FO on P2), p<O.05, Fig. I). Taken together, this study suggested thai cognitive impairment pertaining to pattern separation could be reversed through oral administration of relatively higher doses of 5-amino-2,3-dihydro~i ,4-phthalazinedione in GWI rats. It has been shown tihat this cognitive improvement is related io increased levels of dentate neurogenesis in these rats, in compariso to GWI rats that received VEH during the same period.
5-amino-2,3-dihydro- 1 ,4-phthaJazmedione treatment at moderate to higher doses improves mood function in GWI rats: We examined mood function (or the extent of depressive- like behavior) in GWI rats belonging to different groups (n~:l ϊ-14/group) through a Sucrose
Preference Test (SPT), which is a stress free test measuring anhedo a (i.e. inability to feel pleasure, a measure of depression). This test comprised four days of monitoring. On day 1, rats were housed individually and given free access to two identical bottles containing 1% sucrose solution. Rats were trained to adapt to sucrose solution for 24 hours. On day 2, one bottle was replaced with a new bottle containing regular water for 24 hours. On day 3, rats were deprived of water and food fo 23 boors, and then on day 4, rats were given free access to two bottles: one containing 100 ml of sucrose solution and another containing UK) ml of regular water. An hour later, the consumed volume in both bottles was recorded.
Figs. 2A-2F illustrate the results of the Sucrose Preference Test (SPT). Figs. 2A- IE- compare the consumption of normal water and sucrose containing water in different animal groups (n-i 3-1.4/group in al GWI groups, n::::6 in naive control group). Cyan, Naive control group; red, GWI+VEH group; orange, GWR-5-amino-2.3-dihydro- J ,4-phthalazmedione
(40 mg/kg) group; yellow, GWH -5-amino-2,3-dihydro-l }4-phthaiazinedione (80 mg/kg) group; green, GWf†5-ami.oo-2,3-dihydro-I,4-phthaiazinedione ( .160 mg/kg) group.
Fig. 2F compares the total volume (normal water + sucrose-containing water) consumed by rats in each group. One-way A'NOVA. analysis did not show differences between groups, implying that the preference for drinking sucrose-containing water observed in naive control group and 5~amino-2,3-dihydro~ 1 ,4~phthalazinedione (80 mg kg and .1 0 mg kg groups) was not influenced by differences in the overall consumption of water during the tes ting period,
(i) 'Naive control rats clearly showed a preference for drinking sucrose-containing water over regular water (Fig. 2 ) .
(ii) GWI rats that received VEH did not exhibit such preference as they consumed normal and sucrose-containing water in equal proportions (Fig, 2), implying the presence of anhedoni in G WI rats.
(Hi.) GWI rats that received lower doses of 5-amino-2,3-dihydro- 1 ,4-pltthalazinedione (40 mg/kg) also remained impaired, as they consumed sucrose-containing water and regular water in almost equal proportions (Fig. 2), suggesting that lower dose of 5~amino-253-dilr dro~ L4- phthalazinedione does not have a positive effect on mood function in GWI rats.
(iv) GWI rats that received moderate and higher doses of 5-amin.o-2,3-dihydro- 11 ,4- phthalazinedione (80 or 160 mg/kg) exhibited a clear preference for drinking sucrose-containing water over regular water (Fig, 2), Calculation of sucrose preference rate using the formula, sucrose consumption (water consumption + sucrose consumption) x 100% also showed similar results (data not illustrated).
Thus, these results demonstrate that mood impairment, particularly anhedonia, may be reversed with oral administration of moderate to higher doses of 5-ara.ino-2,3-d:ihydro-l ,4- phthalazmedione in GWI rats. The study shows that improved mood with 5-amino~2,3~dihydro- 1 ,4-phthalazinedione treatment is related to increased levels of dentate neurogenesis in these rats, in comparison to GWI rats that received VEH during the same period. This relationship has been examined quantitatively and in other studies we have been examining the effects of 5-ammo~2,3~ dihydro- 1 ,4-phthalazinedione in severs! mood function tests.
5-amino-2,3-dihydro- 1 ,4-phthalazinedione treatment at higher doses mod Jates oxidative stress in the hippocampus ofGWI rats: Figs. 3A-3D demonstrate that 5-ammo-2,3-dihydro-l ,4- phthaiazmedione treatment normalizes the expression of oxi dati ve stress response genes prdx6, sod2„ sqstml and sr l in GWI rats.
Fig. 4 is a ciustergrarn showing the expression of oxidative stress response genes in various animal groups. N2- 5, naive control animals (n-4); GWMV2-GWIV1V5, GWI rats receiving vehicle (n~4); GWMSL40 to GWMSL40-S, GWI rats receiving 5« rnino-2,3- dihydro-?.,4-phthaiazinedione at 40 mg/kg (n-5); GW SL80-.1 to GWMSL80-5, GWI rats receiving 5«amino-2,3-dihydro- 1 ,4-phthalazinedione at 80 mg kg (n~S); GWMSL160-I to
GWMSL 160-5, GWI rats receiving 5-amiTK>~2}3-dihydro-.i,4~phd ala i.n.edione at .1 0 mg kg (n-5). Arrows denote genes, which show upregu!ation in GWI rats receiving VEH and
normalization in GWI rats receiving 5-amino-2,3-dihydro-l,4-phthalaz.inedione particularly obvious in GWI rats receiving 80 rag/Kg orl 0mgKg doses. MSL is the monosodiuiTi salt of luminol (5-amino-2,3-dihydro-l ,4-phthalazinedione or 5-amino-2,3-dihydrophthalazine-l ,4- dione).
(j) Expression of oxidative stress response and antioxidant genes; We analyzed the expression of 84 key genes involved in oxidative stress response and antioxidant activity in the hippocampus of animals belonging to different groups using quantitative real time PGR
(qRTPCR), Among 84 genes related to oxidative stress response examined in this experiment, GWI rats receiving VEH exhibited increased expression of 24 genes, in comparison to age- matched naive control animal (Figs, 3 and 4), implying the presence of significant oxida tive stress in the hippocampus of G WI rats. Among these 24 genes, the expression of 4 genes was completely normalized by 5-amrao-2,3-dihydro- 1 ,4-phthalazinedione treatment (Fig. 3). This was evidenced through statistics (one-way ANOVA with Newman-Keuls multiple comparison test), which showed that GWI rats receiving 5~amino-2,3-dihydro~l,4~phthalazi»edione exhibited reduced expression of these genes, in compariso to GWI rats receiving VEH. The genes comprise the following: (1) Prdx6: This gene encodes peroxiredoxin-6 protein. It is a member of the peroxiredoxin family of antioxidant enzymes (thiol-speciftc antioxidant protein family). 1 1 is involved in redox regulation of the cell, as it can reduce hydrogen peroxide and short chain organic, fatty acid, and phospholipid hydroperoxides, it is also believed to play a role in the regulation of phospholipid turnover as well as in. protection against oxidati ve injury; (2) Sod2: This gene encodes mitochondria! superoxide dismutase 2 protein. It is also known as manganese-dependent superoxide dismutase (MnSOD). Sod2 protein forms a homotetramer and binds one manganese ion per sobunit. This protein binds to the superoxide byproducts of oxidative phosphorylation and converts mem to hydrogen peroxide and diatomic oxygen, which facilitates SOD2 to clear mitochondria! reaction oxygen species (ROS) and thereby provides protection against ceil death; (3) Sgstnil: This gene encodes sequestosome 1 (or p62) protein. This is a multifunctional protein that binds ubiquitin and regulates activation of the nuclear factor kappa-B (NF-kB) signaling pathway. The protein functions as a scaffolding/adaptor protein in concert with TNF receptor-associated factor 6 to mediate activation of NF-kB in response to upstream signals. Studies also suggest that this protein is a common component of protein aggregates that are found in protein aggregation diseases affecting the brain (e.g.
Parkinson's and Alzheimer's diseases); and (4) Srxiil : This gene encodes sulfsredoxin-l
protein. This protein binds to peroxiredoxins and reduces over oxidized peroxiredoxins in the presence ofcofactors including magnesium and ATP. Elevated expression of this protein has been associated with different types of malignant tumors. Thus, sulfiredoxin along with peroxiredoxins. play an important tole in protecting tissues from oxidative stress- Furthermore, 20 genes that displayed increased expression in. GWI rats were normalized to control levels by higher doses of 5-amiTX>~23~dihydro-i ,4-phthalazinedione treatment (80 or 160 mg/kg, Fig, 4). Their expression was greater in GWI rats receiving VEH than in naive control animals (p<0.O5-0.Ol) but did not differ from expression in GWI rats receiving higher doses of 5-amino-2,3-dihydrO" 1 ,4-phthalazinedione (p>0.05). Yet, as the overall reductions were moderate, their expression in GWI rats receiving 5-a ino-2*3-dihydro- 1 ,4-phthalazinedione did not differ statistically from GWI rats receiving VEH. The genes include a cat gene, encoding cataiase protein, which is a key antioxidant enzyme that converts the reactive oxygen species hydrogen peroxide to water and oxygen; a Cisb gene, encoding cathepsin B (also called as amyloid precursor protein secrefase, which is a protein involved in the proteoiyiic processing of amyloid precursor protein; a Dhcr24 gene, encoding 24-dehydroeholesterol reductase, which is an oxidoreduciase mvolved in cholesterol biosynthesis; a Gsr gene encoding glutathione reductase. which reduces oxidized glutathione disulfide to the sulfh dryl form GSH, which is an important cellular antioxidant; a Gsiki gene, encoding glutathione S-transferase kappa i. which functions in cellular detoxification; (6) a Gstpl encoding glutathione S-transferase- 1 , which plays an important role in detoxification by catalyzing the conjugation of many hydrophobic and electrophilic compounds with reduced glutathione; an Idhl gene, encoding isocitrate
dehydrogenase 1, which converts isocitrate to 2-ketoglutarate to produce NADPH necessary for many cellular processes and protection against ROS; and an Ncfl gene, encoding neutrophil cytosolic factor 1 protein, which is a 47 kDa cytosolic subunit of neutrophil NADPH oxidase (a nm!ticomponent enzyme thai is activated to produce superoxide anion); four Prdx!-4 genes, encoding peroxiredoxins 1-4, which are antioxidant enzymes involved in reducing hydrogen peroxide and alkyl hydroperoxides to water and alcohol with the use of reducing equivalents derived from thiol-containing donor molecules; a Pmp gene, encoding prion protein, which is a membrane glycosylphosphatidyl-inositol-anchored glycoprotein that tends to aggregate into rod- like structures; a Pigs2 gene encoding prostaglandin-endoperoxide synthase (also known as cyciooxygenase), which is a key enzyme in prostaglandin biosynthesis and acts both as a dioxygenase and as a peroxidase; a SlcSSai gene, encoding so hue carrier family 38, member 1 protein, which is an important transporter of glutamine, an intermediate in the detoxification of ammonia and the production of urea; a Txnl gene, encoding thioredoxin 1, which participates in various redox reactions through the reversible oxidation of its active center di thiol to a disulfide and catalyzes dimioldisulf.de exchange reactions; a Txnip gene, encoding thioredoxin interacting protein, which is believed to act as an oxidative stress mediator b inhibiting thioredoxin activity or by limiting its bioavailability; a Txnrd.1 gene encoding thioredoxin reductase 1 , which reduces thioredoxins as well as other substrates, and plays a role in selenium metabolism and protection against oxidative stress; a Txnrd2 gene, encoding thioredoxin reductase 2, which is a
selenocysteine-containing flavoenzyme that maintains thioredoxins in a reduced state and thereby plays a key role in regulating the cellular redox environment; and a Vcp2 gene, encoding
uncoupling protein 2 (mitochondrial, proton carrier), which separates oxidative phosphorylation from ATP synthesis wi th energy dissipated as heat, also referred to as the mitochondrial proton leak. This protein facilitates the transfer of anions fro the inner to the outer mitochondrial membrane and the return transfer of protons from the outer to the inner mitochondrial membrane. One or more of these genes can be assessed in the methods of the present invention.
Taken together, our qRT-PCR analyses suggest that 5-araino-2;3-dihyd:n>! ,4- phthalazmedione treatment considerably alleviates oxidative stress in G'WJ rats, which was evidenced through nor malization of the expression of multiple genes (that are typically upregulated in conditions such as increased oxidative stress) to levels seen in naive control animals. (i) Levels of roalondiaidehyde (a measure of lipid peroxidation): Lipid peroxides are unstable indicators of oxidative stress in cells that decompose to form more complex and reactive compounds such as roalondiaidehyde (MDA), a natural by-product of lipid
peroxidation. Hence, we measured MDA in bippocampai tissue extracts from different groups using TBARS Assay Kit. One-way ANOVA analyses showed significant differences between groups (p<0.01, F-5.6). Post-hoc analyses were done using Newman- euls multiple comparison test, which revealed the following findings: (!) MDA concentration in GWI rats receiving VEH was higher than naive control rats but the difference was not significant statistically; (2) GWI rats receiving lower dose of 5~aroioo~2 -di hydro- ί ,4-phthalazinedione (40 mg/kg) displayed higher concentration of MD A than nai ve control rats (p<0.05), GWI rats receiving 80 mg/kg 5-amino-2,3-dihydro~l ,4-phthalazinedione (p 0.05) and GWI rats receiving 160 mg kg 5-amino-2 -dihydro~l ,4-phtha!azinedione (p<0.01); and (3) GWI rats receiving higher dose of 5~ainino-2s3- ihydro-L4-phthalaziiiedioiie (160 mg/kg) displayed reduced MDA concentration than GWI rats receiving VEH, Thus, administration of a higher concentrati n of 5-amino-2,3-dihydro- 1.,4-phthalazinedione ( 160 mg/kg) decreases MD.A concentration in GWI rats.
(ii) Concentration of 3-mrrotyrosme: increased modification of tyrosine residues in proteins to 3-oitroiyrosioe by peroxyni trite or other potential nitrating agents is seen in tissues subjected to oxidative stress. Hence, we quantified 3-mtrotyros.ine in. hippocampal tissue extracts from different groups using the nitroryrosine ELISA Kit. However, 3-NT levels did not differ between groups (one-way ANOVA. p>0.05). Naive control animals and GWI rats that received VEH, and GWI rats that received 5-amino-2,3-dihydro- 1 ,4-phthalazinedione exhibited similar levels of 3-NT ( Fig. 4). Thus, 3-NT levels are not altered in the hippocampus of GWI rats.
Analyses of inflammation: The hippocampal tissues obtained from animals were used for measurement of the relative levels of inflammatory cytokines in different groups of animals. We employed "The Rat Cytokine Plate Array" from Signosis, which facilitated analyses of 16 rat cytokines in a high-throughput manner. The cytokines included: TNF-a, IL-lo, ΙΙ.,-ίβ. VBGF, FGFp, ΙΡΝγ, IL-5, IL-6, IL-15, leprin, MCP-1. IP- 10 (or CXCL10), SCF, Rantes, MIP-la and I'G 'p. This study revealed no significant differences in the concentration of these cytokines between naive control animals, GWI rats receiving VEH, and GWI rats receiving different doses of 5~amino~2,3-dihydro~l ,4-phthalazinedione (see Table I below). Tabie 1
Figure imgf000035_0001
Nai e eofitioi wrssys GWf+V H, p«O.OS (twM&i!ed, unpaired West)
The only exception is Π α, which showed increased expression in GWI rats receiving VEH, n comparison to naive control animals. This protein is produced by macrophages believed to be involved in inflammation and is typically upregulaisd in the brain in conditions such as Alzheimer's disease, multiple sclerosis and hypoxic-ischemic brain injury. Increased expression of this chemokine is believed to enhance inflammation by attracting more leucocytes to the brain parenchyma. Treatment with 5-amino-2.3-dihydro- L4-piitiiaiazi.oed.ione reduced the
concentration of ΜΓΡ-ία, although the decreases were not significant statistically.
What is claimed is:

Claims

1. Use of a compound of Formula I or a pharmaceutically acceptable composition comprising the compound of Formula Ϊ in the treatment of a medical condition associated with brain dysfunctio :
Figure imgf000036_0001
wherein, Ri and R2 are each, independently, hydrogen (H), lithium (Li), sodium (Na), potassium ( ), rubidium (Rb)» caesium (Cs), or {ranciurn (Fr);
R3 is an alkyl, a kenyl, alkynly, aryl, alkoxyi, alkenySoxy, alkynybxy, aryloxy, alkoxy carbonyl, alkylamino, alkyl thio, alkylsulfonyl, or alkylsuliinyl, each optionally substituted with an alkyl, halogen, alkoxy, aryl or heteroaryl moiety;
Ci , C2, C3, C- . Cs, C«, CT and Cs are each, independently, carbon 12 (r2C) or an isotope of i2C (e.g.. !3C);
Nf, N2 and Ns are each, independently, nitrogen 14 (1 N) or an isotope of ] N (e.g., i3N); and Oi and O2 are each, independently, oxygen 16 (i60) or an isotope of l60 (e.g., "O or i80).
2. The use of claim 1, wherein the medical condi tion is a mood disorder or mood dysfunction, a memory disorder or memory dysfunction, anxiety or a stress-related condition, or an acute or chronic brain injury.
3. The use of claim 2, wherein the acute or chronic brain injury is caused by trauma, mitochondrial dysfunction, or dysfunction of an endogenous retrovirus.
4. The use of claim 2, wherein the mood disorder or mood dysfunction is bipolar disorder, depression (or a depressive illness), schizophrenia, or is associated with cancer, another chronic illness, infection, or substance abuse.
5. The use of claim 2, wherein the stress-related condition is a post-traumatic stress disorder. 6, The use of claim i, wherein the medical condition is Gulf War Illness (GWl).
7, The use of claim 1, wherein the medical condition is, or has as a prominent symptom, S amnesia, Alzheimer's disease (AD), dementia, Huntington's disease (HD), ataxia, Parkinson's disease (PD), Tourette's syndrome, migraine headache, multiple sclerosis (MS), multiple chemical sensitivity (MCS), cognitive dysfunction (CD), multiple sclerosis (MS), or amyotrophic lateral sclerosis (ALS). 0 8, The use of claim 1, wherein the compound of Formula I or the pharmaceutically
acceptable composition comprising the compound of Formula I is formulated for oral
administration or administration to a mucous membrane.
9. The use of claim 1, wherein the compound of formula I is 5-amino-2,3~
5 dihydrophthatazme- 1 ,4-dione (!uminol), 6-am½o-2,3-d¾ydrophthala20ne- 1 ,4-dione, 5-amino-2,3- dihydrophthalazine-l,4-dion-8-yi (luminyl), N-biOmo-5-amino-2 -dmydrophthalazine-l,4-dione, N-clik ro-5-amif -2,3-diiiydrophthaiazine-i ,4-djon,e, N-t1iK>ro-5-amino-2,3-dihydrophiha!azine- 1 ,4-dione, N-iodo-5-amino~2,3~dihydrophthalazine~ 1, ,4-dione, N-.methyl-5-amino-2,3- dihydrophthalazine-1 ,4-dione, N-etttyl-5-amifto-2,3-dihydrophthalazme-l ,4-dione, N-propyl-5-0 amino~2s3-dihydrophmaIazine- 1 ,4-dione, N~isopiOpyl-5-ami»o-2,3~dihydrophthalazine- 1 ,4-dione, N-methanoyl-5-amino-2,3'dihydrophthalazme- 1 ,4-dione, N-etliauoyl~5~amino~2,3- dihvdrophthalazine-1 ,4-dione, N-propanoyl-5-amino-2,3-dihydTOphthalazine- 1 ,4-dione, N- hydroxyl-5-amino~2,3>dthydrophthalazine~l,4-dione, N~carhoxy|~5~amino~2,3- dmydrophthalasriiie-l ,4-dione, N-methanol-5-ami«o-2,3-dihydrop thalazine-l.,4-dione, N-ethanol-5 5-aniino-23-dihydrophihala2;ine- 1 ,4-dione, N-pK>panol-5-aniino-2,3-dihydropht:halazine- i ,4- dione, -methenyl-5-ai¾ino-2,3-dihydrophmalazine- 1 ,4-dione, N-e eiiy !~5-amino-2,3- dihydropMhalazine- 1 ,4-dione, N-pR penyl~5-amino-2,3-dihydrophi:haSazine- ,4-dione, H- methoxy-5-amino-2,3-dihydrophthala2ine- 1 ,4-dione, N-ethoxy-5-amino-2,3-dihydrophthalazine- 1 ,4-dione, N-propoxy-5-aramo-2,3-dihydroplitliakzioe-l ,4-dione, N,N-diraeihyl-5-amino-2,3-0 dihydrophthalazine-l ,4-dione, N-aceiylcysieine-5~amino-253-dih droplithala2ine- 1 ,4-dione, ~ acet5'lgiuiatbione-5~aiiiioo-2 ,3 -dihydrophdialazine- 1 ,4-dione, 5~(hexaaoyl oxyamino)-2,3 - dihydrophthalazine-ί ,4-dione, S-(raethylamino)-2,3-dihydrophlijalazine-l ,4-dione, or 5- (acetoxyam »o)-2,3-<iibydrophtlialazine- 1 ,4-dione, a pharmaceutically acceptable salt thereof, or an isotopic derivative thereof.
10. The use of claim 1 , wherein the compound of formula I is;
monosodium 5~amino-253-dihydro-.1 ,4-phthalaJiinedione;
monosodium 5-meihaffiino-2 -dibydro-l,4-phthalazinedione;
monosodium 5-acet0xyamino-253~dihydro- 1 ,4-phthalazinedione; or
monosodium 5-(bexanoyl oxylamkro)-2 -dihydr -pht¾alazinedione.
PCT/US2016/069636 2015-12-31 2016-12-31 Compositions and nethods for treating brain dysfunction WO2017117586A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP16882793.9A EP3397257A4 (en) 2015-12-31 2016-12-31 Compositions and nethods for treating brain dysfunction
US16/067,542 US20210161890A1 (en) 2015-12-31 2016-12-31 Compositions and methods for treating brain dysfunction

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562273913P 2015-12-31 2015-12-31
US62/273,913 2015-12-31

Publications (1)

Publication Number Publication Date
WO2017117586A1 true WO2017117586A1 (en) 2017-07-06

Family

ID=59225526

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/069636 WO2017117586A1 (en) 2015-12-31 2016-12-31 Compositions and nethods for treating brain dysfunction

Country Status (3)

Country Link
US (1) US20210161890A1 (en)
EP (1) EP3397257A4 (en)
WO (1) WO2017117586A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020084348A1 (en) * 2018-10-26 2020-04-30 Immunopharma Plus D.O.O. Oral aminodihydrophthalazinedione compositions and their use the treatment of non-viral hepatitis
RU2774615C1 (en) * 2020-12-29 2022-06-21 Адмир Мусаевич Абидов Method for producing a medicinal product

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2222327C2 (en) * 2002-03-22 2004-01-27 Общество с ограниченной ответственностью "Абидофарма" Method for preparing medicinal preparation
US20120164243A1 (en) * 2010-12-23 2012-06-28 Amazentis Sa Compositions and Methods for Improving Mitochondrial Function and Treating Neurodegenerative Diseases and Cognitive Disorders
US20140030317A1 (en) * 2002-10-30 2014-01-30 Mark O. Henry Modulation of cell fates and activities by phthalazinediones
US20140107140A1 (en) * 2011-06-24 2014-04-17 K-Pax Pharmaceuticals, Inc. Compositions and methods for treatment of gulf war illness

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100086531A1 (en) * 2008-09-23 2010-04-08 Bach Pharma Inc. METHODS OF MODULATING PROTEIN HOMEOSTASIS, METABOLIC SYNDROME, HEAVY METAL INTOXICATION AND Nrf2 TRANSCRIPTION FACTORS
CA2546395A1 (en) * 2003-11-21 2005-06-09 Memory Pharmaceutical Corporation Compositions and methods of treatment using l-type calcium channel blockers and cholinesterase inhibitors
PL231885B1 (en) * 2009-01-16 2019-04-30 Abidopharma Spolka Z Ograniczona Odpowiedzialnoscia New method of manufacturing the 5-amino-2,3-dihydrophtalazine-1,4-dione salts with alkali metals and their application in medicine
ES2918548T3 (en) * 2016-11-07 2022-07-18 Metriopharm Ag Use of 5-amino-2,3-dihydro-1,4-phthalazindione in the treatment of chronic progressive multiple sclerosis

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2222327C2 (en) * 2002-03-22 2004-01-27 Общество с ограниченной ответственностью "Абидофарма" Method for preparing medicinal preparation
US20140030317A1 (en) * 2002-10-30 2014-01-30 Mark O. Henry Modulation of cell fates and activities by phthalazinediones
US20120164243A1 (en) * 2010-12-23 2012-06-28 Amazentis Sa Compositions and Methods for Improving Mitochondrial Function and Treating Neurodegenerative Diseases and Cognitive Disorders
US20140107140A1 (en) * 2011-06-24 2014-04-17 K-Pax Pharmaceuticals, Inc. Compositions and methods for treatment of gulf war illness

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3397257A4 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020084348A1 (en) * 2018-10-26 2020-04-30 Immunopharma Plus D.O.O. Oral aminodihydrophthalazinedione compositions and their use the treatment of non-viral hepatitis
US20210379063A1 (en) * 2018-10-26 2021-12-09 Immunopharma Plus D.O.O. Oral aminodihydrophthalazinedione compositions and their use in the treatment of non-viral hepatitis
RU2774615C1 (en) * 2020-12-29 2022-06-21 Адмир Мусаевич Абидов Method for producing a medicinal product
RU2774597C1 (en) * 2020-12-29 2022-06-21 Адмир Мусаевич Абидов Method for producing a medicinal product

Also Published As

Publication number Publication date
EP3397257A1 (en) 2018-11-07
US20210161890A1 (en) 2021-06-03
EP3397257A4 (en) 2019-11-13

Similar Documents

Publication Publication Date Title
Peruzzaro et al. Transplantation of mesenchymal stem cells genetically engineered to overexpress interleukin-10 promotes alternative inflammatory response in rat model of traumatic brain injury
ES2640777T3 (en) Anaplerotic therapy for Alzheimer&#39;s disease
ES2750728T3 (en) Use of cannabinoids in combination with Aripriprazole
ES2382334T3 (en) Use of selenium yeasts in the treatment of Alzheimer&#39;s disease
JP5616631B2 (en) Methods and compositions for altering cell function
Kong et al. Effects of gestational isoflurane exposure on postnatal memory and learning in rats
ES2733929T3 (en) A pharmaceutical combination for the treatment of melanoma
JP2005533812A (en) Pharmaceutical use of nitric oxide, heme oxygenase-1 and heme degradation products
US20200206244A1 (en) Treatment of demyelinating diseases
JP2005531534A (en) How to treat ileus
JP2005532351A (en) Methods of treating angiogenesis, tumor growth, and metastasis
Buford et al. Angiotensin (1–7) delivered orally via probiotic, but not subcutaneously, benefits the gut-brain axis in older rats
Wu et al. The microglial innate immune receptors TREM-1 and TREM-2 in the anterior cingulate cortex (ACC) drive visceral hypersensitivity and depressive-like behaviors following DSS-induced colitis
TWI798320B (en) Therapeutic agent for nervous system disease
WO2017117586A1 (en) Compositions and nethods for treating brain dysfunction
Takei et al. High-functioning autistic disorder with Ehlers-Danlos syndrome.
US20190091207A1 (en) Treatment Of CDKL5 Disorders With GSK3B Inhibitor Tideglusib
ES2525337T3 (en) Treatment of diseases or neural states
WO2013017136A1 (en) Treatment of cognitive impairment
ES2377381B1 (en) USE OF A COMBINATION OF N-ACETYL-CYSTEINE AND LIPOIC ACID FOR THE PREPARATION OF A USEFUL MEDICINAL PRODUCT FOR THE TREATMENT OF A DISEASE WITH AXONAL DAMAGE AND COMPATIENT OXIDATIVE INJURY.
Li et al. Middle aged CAMKII-Cre: Cbsfl/fl mice: a new model for studying perioperative neurocognitive disorders
US20150031765A1 (en) Treatment of cognitive impairment
JP2016523975A (en) Use of N-acetylcysteine amide in the treatment of penetrating head trauma
RU2660344C2 (en) Diamine oxidase for use in the treatment or prevention of attention deficit hyperactivity disorder (adhd)
CN116949169B (en) Application of SMEK1 in diagnosis and treatment of cerebral arterial thrombosis

Legal Events

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

Ref document number: 16882793

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2016882793

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016882793

Country of ref document: EP

Effective date: 20180731