US20200368193A1 - Deuterated analogs of d-serine and uses thereof - Google Patents

Deuterated analogs of d-serine and uses thereof Download PDF

Info

Publication number
US20200368193A1
US20200368193A1 US16/766,194 US201816766194A US2020368193A1 US 20200368193 A1 US20200368193 A1 US 20200368193A1 US 201816766194 A US201816766194 A US 201816766194A US 2020368193 A1 US2020368193 A1 US 2020368193A1
Authority
US
United States
Prior art keywords
compound
pharmaceutical composition
formula
serine
deuterium
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/766,194
Other languages
English (en)
Inventor
Dario Doller
Christopher L. Brummel
Julie F. Liu
Roger D. Tung
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sun Pharmaceutical Industries Inc
Original Assignee
Concert Pharmaceuticals 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 Concert Pharmaceuticals Inc filed Critical Concert Pharmaceuticals Inc
Priority to US16/766,194 priority Critical patent/US20200368193A1/en
Assigned to CONCERT PHARMACEUTICALS, INC. reassignment CONCERT PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DOLLER, DARIO, TUNG, ROGER D., BRUMMEL, CHRISTOPHER L., LIU, JULIE F.
Publication of US20200368193A1 publication Critical patent/US20200368193A1/en
Assigned to SUN PHARMACEUTICAL INDUSTRIES, INC. reassignment SUN PHARMACEUTICAL INDUSTRIES, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: CONCERT PHARMACEUTICALS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth 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
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • N-methyl-D-aspartate receptor is a glutamate receptor and ion channel in nerve cells. Activation of NMDAR allows positively charged ions to flow through the membrane. NMDA receptors play a role in physiological processes affecting memory and mood. See e.g., Nicholls et al., Neuron, 2008, 58(1):104-17. Binding of an agonist (such as N-methyl-D-aspartate or glutamate) and a co-agonist (such as glycine or D-serine) is required for NMDAR activation.
  • an agonist such as N-methyl-D-aspartate or glutamate
  • a co-agonist such as glycine or D-serine
  • NMDA modulators have been reported to be useful in a variety of therapeutic applications. For example, memantine is used to treat Alzheimer's disease and Lewy Body Dementia. However, treatment with NMDA modulators can have side effects such as sedation and hallucinations.
  • Anti-NMDAR encephalitis is an autoimmune encephalitis characterized by the presence of antibodies against synaptic NMDAR.
  • Anti-NMDAR encephalitis also known as NMDA receptor antibody encephalitis or NMDAR encephalitis
  • the encephalitis associated with antibodies against NMDAR predominantly affects children and young adults, occurs with or without tumor association, responds to treatment, but can relapse.
  • the exact incidence of anti-NMDAR encephalitis is unknown. Due to the rareness of the syndrome and the varied clinical presentations, anti-NMDAR syndrome may be misdiagnosed and under-recognized.
  • Schizophrenia is a chronic and devastating neuropsychiatric disorder that is ranked as a leading cause of disability worldwide.
  • the disease afflicts nearly 1% of the world's population, affecting both men and women equally, and striking all ethnic and socioeconomic groups with a similar level of prevalence.
  • the illness is characterized by multiple symptoms that are categorized into three clusters known as positive symptoms (hallucinations and delusional behaviors), negative symptoms (anhedonia, social withdrawal and apathy), and cognitive dysfunction (diminished capacity for learning, memory, and executive function).
  • positive symptoms hallucinations and delusional behaviors
  • negative symptoms anhedonia, social withdrawal and apathy
  • cognitive dysfunction diminished capacity for learning, memory, and executive function.
  • Currently available antipsychotic drugs exhibit efficacy for positive symptoms, but have been limited in their capacity to treat negative symptoms and cognitive deficits.
  • D-serine occurs naturally in the human body, although in much smaller amounts than L-serine. Only L-serine is found in proteins.
  • D-serine is an agonist of NMDA receptors. Academic studies have demonstrated that oral dosing of D-serine can result in dose-dependent improvement in positive, negative, and cognitive symptoms in schizophrenic patients when added to D2 antipsychotics (antipsychotic drugs that bind to and inhibit or block the activation of dopamine D2 receptors). However, preclinical studies have demonstrated that administration of D-serine can cause nephrotoxicity in rats. In addition, in some patients who received high doses of D-serine, clinical findings suggesting renal impairment were observed. As a result, the clinical development of D-serine has historically been limited.
  • D-D-serine may have advantageous properties, including reduced nephrotoxicity, relative to D-serine. Further, deuterated D-serine has the potential to restore NMDA receptor activity in key disease-related areas of the brain.
  • this invention relates to deuterated forms of D-serine, pharmaceutically acceptable salts thereof, analogs and prodrugs thereof, pharmaceutical compositions thereof, and methods of use.
  • the invention provides a pharmaceutical composition comprising a compound of Formula I:
  • R 1 is —OH, —OD, —O—C 1-4 alkyl, or an amino acid residue
  • R 2 is H, D, —C 1-4 alkyl, —C(O)—C 1-6 alkyl, or —C(O)—C 1-6 hydroxyalkyl;
  • R 3 is H, D, or an amino acid residue
  • R 4 is H or D
  • each of Y 1 , Y 2a Y 2b is independently H or D, provided that at least one of Y 1 ,
  • R 1 or R 3 is a D-D-serine residue (a residue of deuterated D-serine).
  • the compound is a compound of Formula II:
  • each of Y 1 , Y 2a is independently H or D, provided that at least one of Y 1 , Y 2a and Y 2b is D; or a pharmaceutically acceptable salt thereof.
  • the compound is selected from Compound 100 and Compound 103:
  • This invention also provides the use of such compounds and compositions in methods of treating diseases and conditions that are beneficially treated by administering a modulator of N-methyl-D-aspartate (NMDA) receptor function.
  • Some exemplary embodiments include a method of treating a disease or condition selected from epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive, and/or negative symptoms of schizophrenia, as well as prodromal schizophrenia), bipolar disorder, bipolar mania, bipolar depression, treatment-refractory depression, cognitive deficits in depression, major depressive disorder, generalized anxiety disorder, major depressive disorder with mixed features, and cognition deficits associated with diseases or conditions such as Huntington's disease, subjective cognitive decline, traumatic brain injury, Lewy Body Dementia, the method comprising the step of administering to a subject in need thereof an effective amount of a compound or pharmaceutical composition of
  • This invention also provides a method of treating schizophrenia (including positive, negative, and/or cognitive symptoms of schizophrenia), the method comprising administering to a subject in need thereof an effective amount of a compound or pharmaceutical composition of the present invention.
  • FIG. 1 is a graph showing the concentration of Compound 100 in rat plasma, hippocampus, and cortex after administration of a single dose of 30 mg/kg.
  • FIG. 2 is a graph showing the concentration of non-deuterated D-serine in rat plasma, hippocampus, and cortex after administration of a single dose of 300 mg/kg.
  • FIG. 3 is a graph showing the concentration of Compound 100 in rat plasma, hippocampus, and cortex after administration of a single dose of 150 mg/kg.
  • FIG. 4 is a graph showing the urea nitrogen level in rats after administration of a single dose of 150 mg/kg of either Compound 100 or non-deuterated D-serine.
  • FIG. 5 is a graph showing the creatinine level in rats after administration of a single dose of 150 mg/kg of either Compound 100 or non-deuterated D-serine.
  • FIG. 6 is a graph showing the GGAT level in rats after administration of a single dose of 150 mg/kg of either Compound 100 or non-deuterated D-serine.
  • FIG. 7 is a graph showing creatinine levels in rats following PO (oral) administration of non-deuterated D-Serine and deuterated D-Serine (Compound 100 ).
  • FIG. 8 is a graph showing mean creatinine levels in rats following PO (oral) administration of non-deuterated D-Serine and deuterated D-Serine (Compound 100).
  • FIG. 9 is a graph showing urea nitrogen (BUN) level in rats following PO (oral) administration of non-deuterated D-Serine and deuterated D-Serine (Compound 100).
  • FIG. 10 is a graph showing mean urea nitrogen (BUN) level in rats following PO (oral) administration of non-deuterated D-Serine and deuterated D-Serine (Compound 100).
  • FIG. 11 is a graph showing the urea nitrogen level in rats after administration of various dosages of non-deuterated D-serine.
  • FIG. 13 is a graph showing GGAT level in rats after administration of various dosages of non-deuterated D-serine.
  • FIG. 14 is a line plot showing NMDA receptor activity for non-deuterated (“Protio”) D-serine and compound 100 in an automated patch clamp system.
  • FIG. 16 is a plot showing compound 100 concentration in the brains of Sprague-Dawley rats after 4 days of dosing.
  • this invention relates to deuterated forms of D-serine, pharmaceutically acceptable salts thereof, analogs and prodrugs thereof, pharmaceutical compositions thereof, and methods of use.
  • the invention provides a compound of Formula I:
  • R 2 is H, D, —C 1-4 alkyl, —C(O)—C 1-6 alkyl, or —C())—C 1-6 hydroxyalkyl;
  • each of Y 1 , Y 2a and Y 2b is independently H or D, provided that at least one of Y 1 , Y 2a and Y 2b is D; wherein each position designated specifically as deuterium has at least 50.1% incorporation of deuterium;
  • R 1 or R 3 is a D-D-serine residue (a residue of deuterated D-serine).
  • R 1 or R 3 is a D-D-serine residue
  • the compound of Formula I is a dipeptide
  • both R 1 and R 3 are D-D-serine residues
  • the compound of Formula I is a tripeptide.
  • one of Y 2a Y 2b is H and the other is D.
  • the invention provides a pharmaceutical composition comprising a compound of Formula I:
  • R 2 is H, D, —C 1-4 alkyl, —C(O)—C 1-6 alkyl, or —(O)—C 1-6 hydroxyalkyl;
  • each position designated specifically as deuterium has at least 90% incorporation of deuterium.
  • the compound is a compound of Formula II:
  • Y 1 is D
  • Y 2a and Y 2b are each H.
  • the compound of Formula II is selected from Compound 100 and Compound 103:
  • Y 1 is D, and Y 2a and Y 2b are each H, and Y 1 has at least 90% incorporation of deuterium, or at least 95% incorporation of deuterium, or at least 97% incorporation of deuterium.
  • the invention provides a compound of Formula Ia:
  • R 1 is —OH, —OD, or —O—PG 1 , —O—C 1-6 cycloalkyl or an amino acid residue;
  • R 2 is H, D, —C 1-4 alkyl, —C(O)—C 1-6 alkyl, or —C(O)—C 1-6 hydroxyalkyl; and either
  • R 3 is H, D, or PG 2 ;
  • PG 1 and PG 2 are prodrug groups
  • each position designated specifically as deuterium has at least 90% incorporation of deuterium. In certain embodiments, each position designated specifically as deuterium has at least 95% incorporation of deuterium. In certain embodiments, each position designated specifically as deuterium has at least 97% incorporation of deuterium.
  • prodrug group refers to a group which can be cleaved under physiological conditions (e.g., in vivo) to provide an unprotected moiety (e.g., a carboxylate or an amino group).
  • PG 1 can be any group that is cleaved under physiological conditions to provide an unprotected carboxylate group (i.e., a compound of Formula Ia wherein R 1 is OH);
  • suitable PG 1 groups include —C 1-6 alkyl (such as methyl, ethyl, isopropyl, tert-butyl, neopentyl), —C 3-6 cycloalkyl (including cyclohexyl), or an amino acid residue.
  • PG 2 can be any group that is cleaved under physiological conditions to provide an unprotected amino group; examples of suitable PG 2 groups include an amino acid residue or a group of the formula: —C(O)OC(Z 1 Z 2 )OR 5 , in which Z 1 and Z 2 are independently selected from H, D, C 1 -C 2 alkyl, or together form a C 3 -C 5 carbocyle with the carbon atom to which they are attached; and R 5 is C 1-6 aliphatic group (including a C 1-6 alkyl or a partially or entirely unsaturated C 2-6 aliphatic group), C 3-6 cycloalkyl, or C 4-6 carbocyclyl (which can be partially or entirely unsaturated); wherein each R 5 is optionally further substituted with aryl or heterocycloalkyl.
  • the heterocyclic protecting group formed with R 3 and R 4 can be a group having the structure:
  • R 6 is methyl, ethyl, n-propyl, n-butyl, cyclohexyl, —CH 2 C 6 H 5 or —CH 2 CH 2 C 6 H 5.
  • R 1 or R 3 is a D-D-serine residue (a residue of deuterated D-serine).
  • the compound of Formula I or II is at least about 90% stereomerically pure, e.g., for a compound of Formula I, the compound comprises at least 90% of the structure
  • the compound of Formula Ia is at least about 90% stereomerically pure, e.g., for a compound of Formula Ia, the compound comprises at least 90% of the structure.
  • a compound of Formula I or II may exist as a zwitterion (e.g., a compound of Formula II can be represented by the structure:
  • the pharmaceutical composition is suitable for oral administration. In certain embodiments, the pharmaceutical composition is suitable for intravenous administration.
  • the invention provides a pharmaceutical composition comprising a compound of Formula A:
  • R 2 is H, D, —C 1-4 alkyl, —C(O)—C 1-6 alkyl, or —C(O)—C 1-6 hydroxyalkyl;
  • R 3 is H, D, or an amino acid residue
  • each of Y 1 , Y 2a and Y 2b is independently H or D, provided that at least one of Y 1 , Y 2a and Y 2b is D; wherein each position designated specifically as deuterium has at least 50.1% incorporation of deuterium;
  • composition comprises a therapeutically effective amount of the compound having the D-amino acid configuration.
  • this invention relates to deuterated forms of glycine, pharmaceutically acceptable salts thereof, analogs and prodrugs thereof, pharmaceutical compositions thereof, and methods of use thereof.
  • the invention provides a pharmaceutical composition comprising a compound of Formula III:
  • each of X 1a , X 1b , and X 2 is independently H or D;
  • each X 3 is H or D
  • X 1a is D
  • X 1b is H
  • X 1a and X 1b are each D.
  • this invention relates to deuterated forms of sarcosine, pharmaceutically acceptable salts thereof, analogs and prodrugs thereof, pharmaceutical compositions thereof, and methods of use.
  • the invention provides a pharmaceutical composition comprising a compound of Formula IV:
  • each of X 4a , X 4b , X 5 and X 6 is independently H or D;
  • X 4a , X 4b and R a comprises D
  • X 4a is D
  • X 4b is H
  • X 4a is H
  • X 4b is D
  • X 4a and X 4b are each D.
  • R a CD 3 in the compound of Formula IV, R a CD 3 .
  • the compound is selected from any one of the compounds set forth in Table D (below):
  • this invention relates to deuterated forms of D-alanine, pharmaceutically acceptable salts thereof, analogs and prodrugs thereof, pharmaceutical compositions thereof, and methods of use.
  • the invention provides a pharmaceutical composition comprising a compound of Formula V:
  • each of X 7 and X 8 is independently H or D;
  • each X 9 is H or D
  • each R b is CH 3 , CH 2 D, CD 2 H or CD 3 ;
  • At least one of X 7 and R b comprises D
  • X 7 is H and R b is CD 3 .
  • X 7 is D and R b is CD 3 .
  • X 7 is D and R b is CH 3 .
  • the compound is selected from any one of the compounds set forth in Table E (below):
  • this invention relates to deuterated forms of D-aspartic acid, pharmaceutically acceptable salts thereof, analogs and prodrugs thereof, pharmaceutical compositions thereof, and methods of use.
  • the invention provides a pharmaceutical composition comprising a compound of Formula VI:
  • each of X 10 , X 11 , X 12a , X 12b , and X 13 is independently H or D;
  • each X 14 is H or D
  • X 10 , X 12a and X 12b is D;
  • X 10 is H
  • X 12a is H
  • X 12b is D.
  • X 10 is H, X 12a is D and X 12b is D.
  • X 10 is D
  • X 12a is H
  • X 12b H
  • X 10 is D
  • X 12a is H
  • X 12b is D.
  • X 10 is D
  • X 12a is D
  • X 12b is D
  • the compound is selected from any one of the compounds set forth in Table F (below):
  • each position designated specifically as deuterium has at least 90% incorporation of deuterium.
  • any atom not designated as deuterium is present at its natural isotopic abundance.
  • the compound of Formula V or VI is at least about 90% stereomerically pure.
  • the pharmaceutical composition comprising one or more compounds of Formula III, IV, V and VI is suitable for oral administration.
  • the pharmaceutical composition comprises 0.1 g to 60 g of the compound of Formula III, IV, V or VI.
  • the pharmaceutical composition comprising one or more compounds of Formula III, IV, V and VI is suitable for intravenous administration. In certain embodiments, the pharmaceutical composition comprises 0.1 g to 60 g of the compound of Formula III, IV, V or VI.
  • Another aspect of the invention is a unit dose form comprising a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier or diluent.
  • the amount of a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof is in the range of 1 g to 10 g, or 1 g to 5 g.
  • the amount of a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof is about 1 g, about 2 g, about 3 g, about 4 g, or about 5 g, about 8 g, about 10 g, or in the range of about 5 g to about 10 g.
  • the amount of a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof is 1 g, 2 g, 3 g, 4 g, 5 g, 8 g, 10 g, or in the range of 5 to 10 g.
  • the unit dose form is a tablet. In certain embodiments, the unit dose form is a sachet.
  • Another aspect of the invention is a packaged pharmaceutical formulation comprising a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof, together with a container or package
  • the amount of a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof is in the range of 1 g to 10 g, or 1 g to 5 g.
  • the amount of a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof is about 1 g, about 2 g, about 3 g, about 4 g, or about 5 g, about 8 g, about 10 g, or in the range of about 5 g to about 10 g.
  • the amount of a compound of Formula I or Formula II, or a pharmaceutically acceptable salt thereof is 1 g, 2 g, 3 g, 4 g, 5 g, 8 g, 10 g, or in the range of 5 to 10 g.
  • the packaged pharmaceutical formulation comprises a tablet. In certain embodiments, the packaged pharmaceutical formulation comprises a sachet.
  • the invention provides therapeutic methods.
  • the invention provides a method of treating NMDAR encephalitis, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition of this invention.
  • the invention provides a method of treating epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive, and/or negative symptoms of schizophrenia, as well as prodromal schizophrenia), bipolar disorder, bipolar mania, bipolar depression, treatment-refractory depression, cognitive deficits in depression, major depressive disorder, generalized anxiety disorder, major depressive disorder with mixed features, and cognition deficits in associated with diseases or conditions such as Huntington's disease, subjective cognitive decline, traumatic brain injury, Lewy Body Dementia, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition of this invention.
  • the invention provides a method of treating Additional diseases or conditions include post-traumatic stress disorder (PTSD), ataxia, and serine deficiency disorders, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition of this invention.
  • PTSD post-traumatic stress disorder
  • ataxia ataxia
  • serine deficiency disorders the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition of this invention.
  • the invention provides a method of treating depression, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition of this invention.
  • the invention provides a method of increasing NMDA receptor function, the method comprising contacting a cell with a pharmaceutical composition of this invention, such that NMDA receptor function in the cell is increased.
  • the invention provides a method of treating schizophrenia (including positive, negative, and/or cognitive symptoms of schizophrenia), the method comprising administering to a subject in need thereof an effective amount of a compound or pharmaceutical composition of the present invention.
  • compositions or methods administration of a compound of this invention results in reduced nephrotoxicity compared to administration of an equivalent dose of (non-deuterated) D-Serine.
  • treat means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
  • a disease e.g., a disease or disorder delineated herein
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • the term “subject” includes humans and non-human mammals.
  • Non-limiting examples of non-human mammals include mice, rats, guinea pigs, rabbits, dogs, cats, monkeys, apes, pigs, cows, sheep, horses, etc.
  • the subject is a human suffering from schizophrenia.
  • alkyl refers to a monovalent saturated hydrocarbon group.
  • a C 1 -C 4 alkyl is an alkyl having from 1 to 4 carbon atoms; a C 1 -C 6 alkyl is an alkyl having from 1 to 6 carbon atoms.
  • an alkyl may be linear or branched. In some embodiments, an alkyl may be primary, secondary, or tertiary.
  • Non-limiting examples of alkyl groups include methyl; ethyl; propyl, including n-propyl and isopropyl; butyl, including n-butyl, isobutyl, sec-butyl, and t-butyl; pentyl, including, for example, n-pentyl, isopentyl, and neopentyl; and hexyl, including, for example, n-hexyl and 2-methylpentyl.
  • Non-limiting examples of primary alkyl groups include methyl, ethyl, n-propyl, n-butyl, n-pentyl, and n-hexyl.
  • Non-limiting examples of secondary alkyl groups include isopropyl, sec-butyl, and 2-methylpentyl.
  • Non-limiting examples of tertiary alkyl groups include t-butyl.
  • a “C 1 -C 6 hydroxyalkyl” group is a C 1 -C 6 alkyl group substituted with one to three hydroxyl groups.
  • D-D-serine refers to a deuterated analog of the amino acid serine in the (D)-configuration.
  • D-D-serine can be represented by the structure of Formula II:
  • each of Y 1 , y 2a and y 2b is independently H or D, provided that at least one of Y 1 , Y 2a and Y 2b is D.
  • amino acid residue refers to a group of the general formula —C(O)—CHR—NH 2 or HO—C(O)—CHR—NH—, and N-alkylated derivatives thereof (—C(O)—CHR-N(alkyl)—), wherein R is an amino acid side chain, and includes naturally occurring and synthetic amino acids in a (D)—, (L)— or racemic (D,L) configuration.
  • R 1 or R 2 of Formula I herein is an amino acid residue, the amino acid residue is linked to the rest of the molecule through an amide bond.
  • Exemplary amino acids include a residue of any naturally-occurring amino acid, including deuterated forms thereof.
  • an amino acid residue can be a residue of deuterated D-serine (D-D-serine).
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • the position is understood to have hydrogen at its natural abundance isotopic composition.
  • the position has at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% hydrogen.
  • a position when a position is designated specifically as “H” or “hydrogen”, the position incorporates ⁇ 20% deuterium, ⁇ 10% deuterium, ⁇ 5% deuterium, ⁇ 4% deuterium, ⁇ 3% deuterium, ⁇ 2% deuterium, or ⁇ 1% deuterium.
  • the position when a position is designated specifically as “D” or “deuterium”, the position is understood to have deuterium at an abundance that is at least 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1% incorporation of deuterium).
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • each designated deuterium atom has deuterium incorporation of at least 52.5%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 60%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 67.5%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 75%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 82.5%.
  • each designated deuterium atom has deuterium incorporation of at least 90%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 95%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 97.5%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 99%. In some embodiments, in a compound of this invention, each designated deuterium atom has deuterium incorporation of at least 99.5%.
  • Deuterium incorporation in a compound of the invention can be measured using a variety of techniques, some of which are known in the art.
  • 1 H NMR can be used to measure deuterium incorporation (e.g., by measuring the absence of or decrease in proton signals corresponding to deuterated positions, e.g., relative to a non-deuterated position or positions).
  • isotopologue refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof.
  • compound when referring to a compound of this invention, refers to a collection of molecules having an identical chemical structure, except that there may be isotopic variation among the constituent atoms of the molecules.
  • a compound represented by a particular chemical structure will contain molecules having deuterium at each of the positions designated as deuterium in the chemical structure, and may also contain isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure.
  • the relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound.
  • the invention also provides salts of the compounds of the invention.
  • a salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
  • the compound is a pharmaceutically acceptable acid addition salt.
  • the acid addition salt may be a deuterated acid addition salt.
  • pharmaceutically acceptable refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention.
  • pharmaceutically acceptable counterion is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
  • Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids.
  • inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1,4-dioate, hexyne-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylpropionate
  • pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid.
  • the acids commonly employed to form pharmaceutically acceptable salts include the above-listed inorganic acids, wherein at least one hydrogen is replaced with deuterium.
  • the pharmaceutically acceptable salt may also be a salt of a compound of the present invention having an acidic functional group, such as a carboxylic acid functional group, and a base.
  • exemplary bases include, but are not limited to, hydroxide of alkali metals including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydroxyl-substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-(C 1 -C 6 )-alkylamine), such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2-hydroxyethyl)amine; N-
  • Certain compounds of the present invention contain an asymmetric carbon atom (i.e., the carbon bearing the —NH2 or NR 3 R 4 and Y 1 groups in a compound of Formula I or II) and may contain one or more additional asymmetric carbon atoms.
  • a compound of Formula I or II is a deuterated D-serine analog substantially free from other possible stereoisomers, e.g., a compound of Formula I is substantially free of a compound of the structure:
  • substantially free of other stereoisomers means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers are present.
  • Methods of obtaining or synthesizing an individual stereoisomer (e.g., enantiomer or diastereomer) for a given compound are known in the art and may be applied as practicable to final compounds or to starting material or intermediates.
  • stable compounds refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
  • Stepoisomer refers to both enantiomers and diastereomers. “Tert” and “t-” each refer to tertiary. “Sec” or “s-” each refer to secondary. “n-” refers to normal. “i-” refers to iso. “US” refers to the United States of America.
  • “Substituted with deuterium” refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.
  • variable may be referred to generally (e.g.,“each R”) or may be referred to specifically (e.g., R 1 , R 2 , R 3 , etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable.
  • schizophrenia refers to a psychiatric disorder that includes at least two of the following: delusions, hallucinations, disorganized speech, grossly disorganized or catatonic behavior, or negative symptoms. Patients may be diagnosed as schizophrenic using the DSM-IV criteria (APA, 2013, Diagnostic and Statistical Manual of Mental Disorders (Fifth Edition), Washington, D.C.).
  • “Negative” symptoms of schizophrenia include affect blunting, anergia, alogia and social withdrawal, which can be measured using SANS (the Scales for the Assessment of Negative Symptoms; see Andreasen, 1983, Scales for the Assessment of Negative Symptoms (SANS), Iowa City, Iowa).
  • SANS the Scales for the Assessment of Negative Symptoms; see Andreasen, 1983, Scales for the Assessment of Negative Symptoms (SANS), Iowa City, Iowa).
  • “Positive” symptoms of schizophrenia include delusion and hallucination, which can be measured using PANSS (the Positive and Negative Syndrome Scale; see Kay et al., 1987, Schizophrenia Bulletin 13:261-276).
  • “Cognitive” symptoms of schizophrenia include impairment in obtaining, organizing, and using intellectual knowledge which can be measured by the Positive and Negative Syndrome Scale-cognitive subscale (PANSS-cognitive subscale) (Lindenmayer et al., 1994, J. Nerv. Ment. Dis. 182:631-638) or with cognitive tasks such as the Wisconsin Card Sorting Test.
  • PANSS-cognitive subscale Positive and Negative Syndrome Scale-cognitive subscale
  • cognitive tasks such as the Wisconsin Card Sorting Test.
  • the present invention provides a pharmaceutical composition comprising a compound of Formula I:
  • R 1 is —OH, —OD, —O—C 1-4 alkyl, or an amino acid residue
  • R 2 is H, D, —C 1-4 alkyl, —C(O)-C 1-6 alkyl , or —C(O)—C 1-6 hydroxyalkyl;
  • R 3 is H, D, or an amino acid residue
  • R 4 is H or D
  • each of Y 1 , Y 2a and Y 2b is independently H or D, provided that at least one of Y 1 , Y 2a and Y 2b is D;
  • R 1 or R 3 is a D-D-serine residue (the compound is a dipeptide).
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the compound is a compound of Formula II:
  • each of Y 1 , y 2a and y 2b is independently H or D, provided that at least one of Y 1 , Y 2a and Y 2b is D; or a pharmaceutically acceptable salt thereof.
  • Y 1 is D.
  • Y 2a and Y 2b are each H.
  • Y 2a and Y 2b are each D.
  • the compound of Formula II is selected from Compound 100 and Compound 103:
  • each position designated specifically as deuterium has at least 90% incorporation of deuterium.
  • Y 1 is D, and Y 2a and Y 2b are each H, and Y 1 has at least 90% incorporation of deuterium, or at least 95% incorporation of deuterium, or at least 97% incorporation of deuterium.
  • any atom not designated as deuterium is present at its natural isotopic abundance.
  • the compound of Formula I or Formula II is at least about 90% stereomerically pure.
  • the compound is selected from any one of the compounds set forth in Table A (below):
  • the compound is Compound 100:
  • the compound is Compound 103:
  • the compound is selected from any one of the compounds (Cmpd) set forth in Table B (below):
  • the compound is selected from any one of the Compounds set forth in Table A or Table B (above), or a pharmaceutically acceptable salt thereof; wherein any atom not designated as deuterium is present at its natural isotopic abundance.
  • the level of deuterium incorporation at Y 1 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
  • any atom not designated as deuterium in any of the embodiments set forth herein is present at its natural isotopic abundance.
  • deuterium incorporation at each designated deuterium atom is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
  • At least one Y 1 , Y 2a , and Y 2 b, is hydrogen.
  • Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing isotopic atoms to a chemical structure.
  • esterification of dl-serine (1) results in formation of the serine ester (2), which can be cyclized to the oxazoline (3) using benzoimidate.
  • Oxazoline (3) is then deuterated by deprotonation with a strong base (such as butyllithium) and quenching with a deuterium source (such as acetic acid O-D) to produce deuterated intermediate (4), which is resolved (e.g., using a chiral salt such as d- ⁇ -bromocamphorsulfonic acid or separating the enantiomers using SMB (simulated moving bed) chromatography) to provide intermediate (5) (as the salt). After neutralizing the salt, the oxazoline (5) is then hydrolyzed to provide a compound of Formula II (Scheme 1 illustrates preparation of Compound 100).
  • a strong base such as butyllithium
  • a deuterium source such as acetic acid O-D
  • Compound 103 can be prepared by using 2-amino-2,3,3-trideuterio-3-hydroxy-propanoic acid (which is commercially available, e.g., from Sigma-Aldrich) as the starting material in the general procedure of Scheme 1; in this instance, the deuteration step of Scheme 1 is not required and is omitted.
  • 2-amino-2,3,3-trideuterio-3-hydroxy-propanoic acid which is commercially available, e.g., from Sigma-Aldrich
  • compositions comprising an effective amount of a compound of Formula I, or II (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier.
  • the carrier(s) are “acceptable” in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
  • the invention provides a pharmaceutical composition comprising a compound of Formula Ia.
  • the invention further provides pharmaceutical compositions comprising an effective amount of a compound of Formula III, IV, V or VI (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier.
  • compositions comprising in combination an effective amount of two or more compounds selected from a compound of Formula I, II, III, IV, V and VI (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of each said compound; and a pharmaceutically acceptable carrier.
  • compositions comprising in combination an effective amount of (i) a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, and (ii) one or more compounds selected from glycine, sarcosine, (nondeuterated) D-alanine and (nondeuterated) D-aspartic acid, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • compositions comprising in combination an effective amount of two or more compounds selected from a compound of Formula III, IV, V, VI (e.g., including any of the formulae herein) and (nondeuterated) D-serine, or a pharmaceutically acceptable salt of each said compound; and a pharmaceutically acceptable carrier.
  • compositions comprising in combination an effective amount of two or more compounds selected from a compound of Formula III, IV, V, VI (e.g., including any of the formulae herein) and D-serine, or a pharmaceutically acceptable salt of each said compound; and a pharmaceutically acceptable carrier.
  • the invention provides a pharmaceutical composition comprising in combination an effective amount of a compound of Formula I and sarcosine. In a further embodiment, the invention provides a pharmaceutical composition comprising in combination an effective amount of Compound 100 and sarcosine. In a further embodiment, the invention provides a pharmaceutical composition comprising in combination an effective amount of Compound 103 and sarcosine.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as phosphat
  • solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art.
  • One method includes the use of lipid excipients in the formulation. See “Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water-Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples,” Kishor M. Wasan, ed. Wiley-Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROLTM and PLURONICTM (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See U.S. Pat. No. 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • a poloxamer such as LUTROLTM and PLURONICTM (BASF Corporation
  • compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compound of the formulae herein is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques).
  • Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, Md. (20th ed. 2000).
  • a unit dosage form can comprise, e.g., 100 mg to 1 g, or 500 mg to 2 g, of a compound of Formula I or II.
  • a unit dosage form can further include one or more second therapeutic agents, e.g., an antipsychotic agent or other agent for the treatment of schizophrenia.
  • a unit dosage form can be administered once per day, or multiple times per day (e.g., twice per day, three times per day, or four times per day). In certain embodiments, the unit dosage form is administered once per day. In other embodiments, the unit dosage form is administered twice per day. In other embodiments, the unit dosage form is administered three times per day. In other embodiments, the unit dosage form is administered four times per day.
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc.
  • Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • a composition of this invention further comprises one or more additional therapeutic agents.
  • the additional therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound having the same mechanism of action as D-serine.
  • Such agents include those indicated as being useful in combination with D-serine, including but not limited to, those described in U.S. Pat. Nos. 9,040,581 and 9,687,460.
  • the additional therapeutic agent is an agent useful in the treatment of a disease or condition selected from epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive, and/or negative symptoms of schizophrenia, as well as prodromal schizophrenia), bipolar disorder, bipolar mania, bipolar depression, treatment-refractory depression, cognitive deficits in depression, major depressive disorder, generalized anxiety disorder, major depressive disorder with mixed features, and cognition deficits in associated with diseases or conditions such as Huntington's disease, subjective cognitive decline, traumatic brain injury, Lewy Body Dementia, and the like.
  • a disease or condition selected from epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive, and/
  • a pharmaceutical composition containing a deuterated analog of D-serine can be administered to a patient suffering from schizophrenia along with, or in sequence with, an art-known additional therapeutic agent for treating schizophrenia (e.g., olanzapine, clozapine, haloperidol, and the like).
  • an art-known additional therapeutic agent for treating schizophrenia e.g., olanzapine, clozapine, haloperidol, and the like.
  • the antipsychotic therapeutic typically is administered at a dosage of 0.25-5000 mg/day (e.g., 5-1000 mg/day)).
  • Typical antipsychotics are conventional antipsychotics such as phenothiazine, butryophenones, thioxantheses, dibenzoxazepines, dihydroindolones, and diphenylbutylpiperidines.
  • “Atypical” antipsychotics are a newer generation of antipsychotics which generally act on the dopamine D 2 and 5HT 2 serotonin receptor and have high levels of efficacy and a benign extrapyramidal symptom side effect profile.
  • Examples of typical antipsychotics include chlorpromazine, thioridazine, mesoridazine, fluphenazine, perphenazine, trifluoperazine, thiothixene, haloperidol, loxapine, molindone, acetophenazine, chlorprothixene, droperidol, and pimozide.
  • Examples of atypical antipsychotics include bolanserin, clozapine, risperidone, olanzapine, cariprazine, asenapine, lurasidone, brexpiprazole, lumateperone, aripiprazole, aripiprazole lauroxil, iloperidone, paliperidone, ziprasidone, and quetiapine.
  • Depot antipsychotics also can be used, e.g., haloperidol decanoate, fluphenazine decanoate, and fluphenazine enanthate.
  • Additional antipsychotics include butaperazine, carphenazine, remoxipride, piperacetazine, and sulpiride.
  • a pharmaceutical composition containing a deuterated analog of D-serine can be administered to a patient with symptoms of schizophrenia together with, or in sequence with, one or more art-known drugs for treating schizophrenia (including antipsychotic agents, e.g., olanzapine, clozapine, haloperidol, quetiapine, risperidone, chlorpromazine and the like).
  • the pharmaceutical composition is for administration to a patient with a DSM-V diagnosis of schizophrenia for at least one year.
  • the pharmaceutical composition is for administration to a patient with a PANSS total score of 70-110.
  • the pharmaceutical composition is for administration to a patient meeting the additional PANSS criteria:
  • the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described additional therapeutic agents, wherein the compound and additional therapeutic agent are associated with one another.
  • association with one another means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
  • the compound of the present invention is present in an effective amount.
  • the term “effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
  • the dosing regimen can include one or more additional therapeutic agents (e.g., where the compound or composition of the invention is used in a combination (e.g., when a compound or composition of the invention is used as an adjunctive therapy).
  • subject in need thereof refers to a subject having or being diagnosed with a disease or condition selected from epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive, and/or negative symptoms of schizophrenia, as well as prodromal schizophrenia), bipolar disorder, bipolar mania, bipolar depression, treatment-refractory depression, cognitive deficits in depression, major depressive disorder, generalized anxiety disorder, major depressive disorder with mixed features, and cognition deficits associated with diseases or conditions such as Huntington's disease, subjective cognitive decline, traumatic brain injury, Lewy Body Dementia, and the like.
  • a disease or condition selected from epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive,
  • Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
  • the pharmaceutical composition comprises an effective amount of the compound of Formula I or II that is in the range from 0.1 g to 60 g. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 1 to 60 g/day, or from 5 to 30 g/day, or from 10 to 20 g/day. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 100 mg to 1 g/day. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 1 to 10 g/day. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 1 to 8 g/day.
  • an effective amount of Compound 100 is in the range from 1 g/day to 10 g/day, or in the range from 1 g/day to 5 g/day, or in the range from 2 g/day to 4 g/day.
  • the pharmaceutical composition comprises 1 g of Compound 100, 2 g of Compound 100, 3 g of Compound 100, 5 g of Compound 100, 8 g of Compound 100, or 10 g of Compound 100.
  • an effective amount of a compound of Formula III, IV, V or VI is in the range from 1 to 60 g/day, or from 5 to 30 g/day, or from 10 to 20 g/day.
  • an effective amount of a compound of Formula III, IV, V or VI is in the range from 30 mg/kg/day to 900 mg/kg/day, or from 60 mg/kg/day to 300 mg/kg/day, or from 150 mg/kg/day to 300 mg/kg/day.
  • Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of administration, the sex, age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician. For example, guidance for selecting an effective dose can be determined by reference to the prescribing information for Compound 1.
  • an effective amount of the additional therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent.
  • an effective amount is between about 70% and 100% of the normal monotherapeutic dose.
  • the normal monotherapeutic dosages of these additional therapeutic agents are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
  • additional therapeutic agents may act synergistically with the compounds of this invention. When this occurs, it will allow the effective dosage of the additional therapeutic agent and/or the compound of this invention to be reduced from that required in a monotherapy. This has the advantage of minimizing toxic side effects of either the additional therapeutic agent of a compound of this invention, synergistically improving efficacy, improving ease of administration or use and/or reduced overall expense of compound preparation or formulation.
  • the invention provides a method of modulating the activity of NMDAR in a cell, comprising contacting a cell with one or more compounds of Formula I or II herein, or a pharmaceutically acceptable salt thereof.
  • the cell is contacted in vitro.
  • the cell is contacted in vivo.
  • the cell is contacted ex vivo.
  • the invention provides a method of treating a disease or condition that is beneficially treated by D-serine in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound of Formula I or II or a pharmaceutical composition comprising a compound of Formula I or II, such that the disease or condition is treated.
  • the invention provides a method of treating a disease or condition that is beneficially treated by D-serine in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound of Formula Ia or a pharmaceutical composition comprising a compound of Formula Ia, such that the disease or condition is treated.
  • Such diseases include, but are not limited to, epilepsy, NMDAR encephalitis, Parkinson's disease, cognitive deficits in Parkinson's disease, Alzheimer's disease, mild cognitive impairment, amyotrophic lateral sclerosis (ALS), Huntington's disease, schizophrenia (including positive, cognitive, and/or negative symptoms of schizophrenia, as well as prodromal schizophrenia), bipolar disorder, bipolar mania, bipolar depression, treatment-refractory depression, cognitive deficits in depression, major depressive disorder, generalized anxiety disorder, major depressive disorder with mixed features, and cognition deficits in associated with diseases or conditions such as Huntington's disease, subjective cognitive decline, traumatic brain injury, Lewy Body Dementia, and the like. Additional diseases or conditions include post-traumatic stress disorder (PTSD), ataxia, and serine deficiency disorders.
  • PTSD post-traumatic stress disorder
  • ataxia and serine deficiency disorders.
  • the method of this invention is used to treat a disease or condition selected from epilepsy and NMDAR encephalitis in a subject in need thereof.
  • the method comprises administering to the subject in need thereof an effective amount of a compound of Formula I or II or a pharmaceutical composition comprising a compound of Formula I or II, such that the disease or condition is treated.
  • This invention also provides a method of treating schizophrenia (including positive, negative, and/or cognitive symptoms of schizophrenia), the method comprising administering to a subject in need thereof an effective amount of a compound or pharmaceutical composition of the present invention (e.g., a compound of Formula I or II, or a compound of Formula Ia; or a pharmaceutical composition comprising compound of Formula I or II, or a compound of Formula Ia).
  • a compound or pharmaceutical composition of the present invention e.g., a compound of Formula I or II, or a compound of Formula Ia; or a pharmaceutical composition comprising compound of Formula I or II, or a compound of Formula Ia.
  • the method further comprises administering an antipsychotic therapeutic agent to the subject.
  • the method further comprises administering a second therapeutic agent to the subject, wherein the second agent is an antipsychotic therapeutic agent.
  • negative and/or positive and/or cognitive symptom(s) of schizophrenia can be measured before and after treatment of the subject or patient. A reduction in such a symptom(s) indicates that the patient's condition has improved. Improvement in the symptoms of schizophrenia can be assessed using the Scales for the Assessment of Negative Symptoms (SANS) or Positive and Negative Syndrome Scale (PANSS) (see, e.g., Andreasen, 1983, Scales for the Assessment of Negative Symptoms (SANS), Iowa City, Iowa and Kay et al., 1987, Schizophrenia Bulletin 13:261-276). Likewise, one can measure improvement of other neuropsychiatric disorders in patients who have been treated by the methods of the invention. In certain embodiments, positive symptoms of schizophrenia are improved after treatment, relative to pre-treatment symptoms. In certain embodiments, negative symptoms of schizophrenia are improved after treatment, relative to pre-treatment symptoms. In certain embodiments, cognitive symptoms of schizophrenia are improved after treatment, relative to pre-treatment symptoms.
  • the method of treating schizophrenia includes administering a pharmaceutical composition containing a deuterated analog of D-serine (or other compound described herein) to a patient suffering from schizophrenia along with, or in sequence with, an art-known drug for treating schizophrenia (including antipsychotic agents, e.g., olanzapine, clozapine, haloperidol, and the like).
  • an art-known drug for treating schizophrenia including antipsychotic agents, e.g., olanzapine, clozapine, haloperidol, and the like.
  • the patient suffering from schizophrenia is stable on antipsychotic therapy, i.e., an existing antipsychotic therapy, prior to treatment as described herein (e.g., a Compound of Formula I or II is used as adjunctive therapy, in conjunction with an additional antipsychotic therapeutic agent).
  • the antipsychotic therapeutic typically is administered at a dosage of 0.25-5000 mg/d (e.g., 5-1000 mg/d)).
  • Typical antipsychotics are conventional antipsychotics such as phenothiazine, butryophenones, thioxantheses, dibenzoxazepines, dihydroindolones, and diphenylbutylpiperidines.
  • “Atypical” antipsychotics are a newer generation of antipsychotics which generally act on the dopamine D 2 and 5HT 2 serotonin receptor and have high levels of efficacy and a benign extrapyramidal symptom side effect profile.
  • Examples of typical antipsychotics include chlorpromazine, thioridazine, mesoridazine, fluphenazine, perphenazine, trifluoperazine, thiothixene, haloperidol, loxapine, molindone, acetophenazine, chlorprothixene, droperidol, and pimozide.
  • Examples of atypical antipsychotics include bolanserin, clozapine, risperidone, olanzapine, cariprazine, asenapine, lurasidone, brexpiprazole, lumateperone, aripiprazole, aripiprazole lauroxil, iloperidone, paliperidone, ziprasidone, and quetiapine.
  • Depot antipsychotics also can be used, e.g., haloperidol decanoate, fluphenazine decanoate, and fluphenazine enanthate.
  • Additional antipsychotics include butaperazine, carphenazine, remoxipride, piperacetazine, and sulpiride.
  • the method of treating schizophrenia includes administering a pharmaceutical composition containing a deuterated analog of D-serine (or other compound described herein) to a patient with symptoms of schizophrenia along with, or in sequence with, one or more art-known drugs for treating schizophrenia (including antipsychotic agents, e.g., olanzapine, clozapine, haloperidol, quetiapine, risperidone, chlorpromazine and the like).
  • the pharmaceutical composition is for administration to a patient with a DSM-V diagnosis of schizophrenia for at least one year.
  • the pharmaceutical composition is for administration to a patient with a PANSS total score of 70-110.
  • the pharmaceutical composition is for administration to a patient meeting the additional PANSS criteria:
  • the degree or extent of nephrotoxicity in the subject is reduced compared to treatment with an equivalent amount of D-serine (e.g., a molar equivalent amount of D-serine).
  • Nephrotoxicity can be monitored by measuring levels of markers such as serum creatinine levels or blood urea nitrogen (BUN).
  • a range of about 7 to 20 mg/dL (2.5 to 7.1 mmol/L) is considered normal for BUN.
  • a range of approximately 0.6 to 1.2 milligrams (mg) per deciliter (dL) in adult males and 0.5 to 1.1 milligrams per deciliter in adult females is considered normal for serum creatinine.
  • the serum creatinine and/or the BUN level is maintained in a normal range during and after treatment.
  • a method of treating schizophrenia comprises administering to subject in need thereof a pharmaceutical composition comprising a compound of Formula II, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, wherein the amount of the compound of Formula I or II administered per day is in the range of 10 mg/kg to 120 mg/kg (i.e., 10 mg per kilogram of body weight of the subject to 120 per kilogram of body weight of the subject), and wherein the serum creatinine level or the BUN level (or both) of the subject is maintained in the normal range (as described above).
  • the compound of Formula I or II or a pharmaceutical composition comprising a compound of Formula I or II is administered once per day. In certain embodiments, Compound 100 or a pharmaceutical composition comprising Compound 100 is administered once per day. In other embodiments, the compound of Formula I or II or a pharmaceutical composition comprising a compound of Formula I or II is administered twice per day. In certain embodiments, Compound 100 or a pharmaceutical composition comprising Compound 100 is administered twice per day. In yet other embodiments, the compound of Formula I or II or a pharmaceutical composition comprising a compound of Formula I or II is administered three times per day. In certain embodiments, Compound 100 or a pharmaceutical composition comprising Compound 100 is administered three times per day. In yet other embodiments, the compound of Formula I or II or a pharmaceutical composition comprising a compound of Formula I or II is administered four times per day. In certain embodiments, Compound 100 or a pharmaceutical composition comprising Compound 100 is administered four times per day.
  • an effective amount of a compound of Formula I or Formula II is in the range from 1 to 60 g/day, or from 5 to 30 g/day, or from 10 to 20 g/day. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 100 mg to 1 g/day. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 1 to 10 g/day.
  • an effective amount of a compound of Formula I or Formula II is in the range from 30 mg/kg/day to 900 mg/kg/day, or from 60 mg/kg/day to 300 mg/kg/day, or from 150 mg/kg/day to 300 mg/kg/day. In certain embodiments, an effective amount of a compound of Formula I or Formula II is in the range from 10 mg/kg/day to 150 mg/kg/day, or 10 mg/kg/day to 120 mg/kg/day, or 10 mg/kg/day to 90 mg/kg/day.
  • Identifying a subject in need of such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
  • any of the above methods of treatment comprises the further step of co-administering to the subject in need thereof one or more additional therapeutic agents.
  • additional therapeutic agent may be made from any additional therapeutic agent known to be useful for co-administration with a co-agonist of the NMDAR.
  • additional therapeutic agent is also dependent upon the particular disease or condition to be treated. Examples of additional therapeutic agents that may be employed in the methods of this invention are those set forth above for use in combination compositions comprising a compound of this invention and an additional therapeutic agent.
  • the combination therapies of this invention include co-administering a compound of Formula I or II and one or more additional therapeutic agents selected from a compound of Formula III, IV, V and VI to a subject in need thereof for treatment of any of the diseases or conditions described herein.
  • the method comprises administering in combination an effective amount of a compound of Formula I and sarcosine.
  • the method comprises administering in combination an effective amount of Compound 100 and sarcosine.
  • the method comprises administering in combination an effective amount of Compound 103 and sarcosine.
  • the method further comprises administering an antipsychotic therapeutic agent to the subject.
  • the combination therapies of this invention include co-administering a compound of Formula I or II to a patient suffering from schizophrenia who is stable on antipsychotic therapy.
  • the method comprises administering in combination an effective amount of a compound of Formula I or II and a “typical” antipsychotic agent.
  • the method comprises administering in combination an effective amount of a compound of Formula I or II and an “atypical” antipsychotic agent.
  • the method comprises administering in combination an effective amount of Compound 100 and an antipsychotic agent.
  • the method comprises administering in combination an effective amount of Compound 103 and an antipsychotic agent.
  • co-administered or “administering in combination” as used herein means that the additional therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an additional therapeutic agent as described above) or as separate, multiple dosage forms. Alternatively, the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the additional therapeutic agent(s) are administered by conventional methods.
  • composition of this invention comprising both a compound of the invention and an additional therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other additional therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
  • administering results in reduced nephrotoxicity compared to administration of an equivalent dose of (non-deuterated) D-Serine.
  • the effective amount of the compound of this invention is less than its effective amount would be where the additional therapeutic agent is not administered. In another embodiment, the effective amount of the additional therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized.
  • Other potential advantages including without limitation improved dosing regimens and/or reduced drug cost
  • the invention provides the use of a compound of Formula I or Formula II alone or together with one or more of the above-described additional therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment in a subject of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of Formula I or Formula II for use in the treatment in a subject of a disease, disorder or symptom thereof delineated herein.
  • the mean plasma, hippocampus, and cortex concentrations at 1 hour were 19200 ng/mL, 674 ng/g, and 754 ng/g, respectively.
  • the mean plasma, hippocampus, and cortex concentrations at 6 hours were 1780 ng/mL, 1385 ng/g, and 936 ng/g, respectively.
  • Similar concentrations of Compound 100 were seen in hippocampus and cortex at 1 hour and 6 hours. Concentrations of Compound 100 in plasma, hippocampus, and cortex were within 2-fold of each other at 6 hour.
  • the nephrotoxicity of non-deuterated D-serine was investigated in Male Sprague-Dawley Rats.
  • the rats were administered a discrete dose of 300 mg/kg PO dose of non-deuterated D-serine in 0.5% methylcellulose in water, in the fasted state.
  • the hippocampus, cortex and plasma were collected and analyzed. Additional blood was collected and were analyzed for the full clinical chemistry profile.
  • Urine was also collected and examined for glucose and total protein.
  • the average endogenous level of the non-deuterated D-serine was 155 ng/mL in rat plasma, 8970 ng/g in rat cortex, and 13450 ng/g in rat hippocampus from control group.
  • the endogenous level of non-deuterated D-serine was only subtracted from the reported plasma concentration.
  • the plasma pharmacokinetic (PK) parameters for non-deuterated D-serine are shown in Table 2.
  • the hippocampus, cortex, and plasma concentration at 8 hr for the non-deuterated D-serine are shown in Table 3 and FIG. 2 .
  • the T max , T 1/2 , C max and AUC inf for non-deuterated D-serine were 0.5 hr, 3.63 hr, 268000 ng/mL, and 500000 hr*ng/mL; respectively.
  • the mean plasma, hippocampus, and cortex concentration at 1 hour are 19200 ng/mL, 674 ng/g, and 754 ng/g; respectively.
  • the mean plasma, hippocampus, and cortex concentrations at 8 hours were 11250 ng/mL, 17725 ng/g, and 11875 ng/g, respectively.
  • Concentrations of non-deuterated D-serine in plasma, hippocampus, and cortex were within 2-fold of each other at 8 hours.
  • Nephrotoxicity of the non-deuterated D-serine at 300 mg/kg in Male Sprague-Dawley Rats was evaluated. Blood urea nitrogen and creatinine levels were elevated. Elevated levels of urea nitrogen and creatinine are suggestive of nephrotoxicity. The presence of glucose was observed in urine.
  • the pharmacokinetic profile of Compound 100 was investigated in male Sprague-Dawley rats compared with that of non-deuterated D-serine.
  • the rats were administered discrete 150 mg/kg PO doses of Compound 100 (92% D by mass spectroscopy) in 0.5% methylcellulose in water and non-deuterated D-serine, in the fasted state.
  • Plasma was collected and analyzed for Compound 100 and non-deuterated D-Serine. Additional blood was collected and analyzed for the full clinical chemistry profile.
  • the PK parameters for Compound 100 and non-deuterated D-serine are shown in Table 4.
  • the hippocampus, cortex, and plasma concentration at 4 hr, 8 hr, and 24 hr for Compound 100 are shown in Table 5 and FIG. 3 .
  • Clinical pathology data are shown in FIGS. 4, 5 and 6 .
  • the T max for Compound 100 was 2-fold greater than that of the non-deuterated D-serine.
  • the C max and AUC inf for Compound 100 were similar to the non-deuterated D-serine.
  • the T max , C max and AUC inf for non-deuterated D-serine were 0.333 hr, 180000 ng/mL, and 349000 hr*ng/mL; respectively.
  • the T max , C max and AUC inf for Compound 100 were 0.667 hr, 186000 ng/mL, and 383000 hr*ng/mL, respectively.
  • the mean plasma, hippocampus, and cortex concentrations at 4 hr were 23283 ng/mL, 6155 ng/g, and 5070 ng/g, respectively.
  • the mean plasma, hippocampus, and cortex concentrations at 8 hr were 2038 ng/mL, 4335 ng/g, and 4035 ng/g, respectively.
  • the mean plasma, hippocampus, and cortex concentrations at 24 hr were 711 ng/mL, 4200 ng/g, and 2420 ng/g, respectively.
  • Compound 100 concentrations in plasma and cortex decreased within 24 hr; however, Compound 100 concentration in hippocampus remained steady at 24 hr.
  • Rats received 150 mg/kg of either Compound 100 or non-deuterated D-serine (PO administration). Elevated levels of blood urea nitrogen (BUN) were seen in the non-deuterated D-serine at 8 hr and 24 hr serum samples. Elevated levels of creatinine and Gamma glutamyl transferase (GGT) were seen in the non-deuterated D-serine at 24 hr serum samples. Elevated levels of blood urea nitrogen, creatinine and GGT are suggestive of nephrotoxicity.
  • BUN blood urea nitrogen
  • GGT Gamma glutamyl transferase
  • FIGS. 7-10 Additional experiments were performed to compare the nephrotoxicity of Compound 100 (Compound 100) and non-deuterated D-serine at doses from 150 mg/kg to 750 mg/kg. The results are shown in FIGS. 7-10 . It can be seen from FIGS. 7-10 that minimal changes in BUN or creatinine were observed in animals dosed with Compound 100, while animals receiving comparable doses of non-deuterated D-serine showed increasingly elevated levels of BUN and creatinine. During the dose escalation, the exposure as measured by AUC and Cmax were greater for Compound 100, yet the gross signs of nephrotoxicity were only observed in rats receiving non-deuterated D-serine (not Compound 100).
  • the pharmacokinetic profile of non-deuterated D-serine was investigated in male Sprague-Dawley rats. The rats were administered a single discrete 30, 75, 100, 150, and 300 mg/kg PO doses of non-deuterated D-serine in 0.5% methylcellulose in water, in the fasted state. Plasma was collected and analyzed for the non-deuterated D-serine. Additional blood was collected and analyzed for the full clinical chemistry profile. The PK parameters for the non-deuterated D-Serine are shown in Table 6. Clinical pathology data are shown in FIGS. 11, 12, and 13 .
  • the Tmax was from 0.3 to 0.5 for all doses.
  • Increase in exposure in terms of C max and AUC inf was seen as the doses increased from 30 to 300 mg/kg.
  • the C max for 30, 75, 100, 150, and 300 mg/kg are 24300, 54300, 96100, 180000, and 216000 ng/mL, respectively.
  • the AUCf for 30, 75, 100, 150, and 300 mg/kg are 50600, 148000, 246000, 349000, and 911000 ng*hr/mL; respectively.
  • nephrotoxicity dose response of non-deuterated D-serine was evaluated in male Sprague-Dawley Rats at 30, 75, 100, 150, and 300 mg/kg PO. Elevated levels of urea nitrogen, creatine, and gamma glutamyl transferase (GGT) (which are markers indicative of nephrotoxicity) were seen in the 24 hr samples at the 150 and 300 mg/kg doses compared to control values and reference values provided by Charles River Laboratory.
  • GTT gamma glutamyl transferase
  • Non-deuterated D-serine and deuterated D-serine were administered to male Sprague Dawley rats (intravenous (IV), 5 mg/kg in phosphate-buffered saline (PBS); and orally (PO), 10 mg/kg, 0.5% methylcellulose in water). Three rats were used for each group. Blood was collected at the following time points: for IV dosing: pre-dose, 0.05, 0.167, 0.5, 1, 2, 4, 6, 8 and 12 hours post-dose; for PO dosing: pre-dose, 0.25, 0.5, 1, 2, 4, 6, 8 and 12 hours post-dose PO. Urine was collected at pre-dose (minimum 12 hours), 0-6, 6-12 and 12-24 hours. Plasma samples were analyzed and quantified for dosed compound LC-MS/MS.
  • deuterated D-serine (Compound 100) had a half-life (T 1/2 ) about 1.5-fold longer than for the non-deuterated D-serine; the deuterated and non-deuterated compounds had a similar T max .
  • Deuterated D-serine (Compound 100) had a C max about 1.3-fold greater than for the non-deuterated D-serine, and an AUC inf about 1.6-fold greater than for the non-deuterated D-serine.
  • non-deuterated D-serine and two deuterated D-serine analogs were administered to male Sprague Dawley rats.
  • Example 6 Exemplary Formulation of a D-D-Serine
  • a modified-release tablet formulation of a D-D-serine is prepared using the materials shown in the table below:
  • Tablet Strength 500 mg D-D-serine (e.g., Compound 100) Total Tablet Wt: 855 mg Amount per tablet Material Generic Name Wt % (mg) D-D-Serine N/A 58.5 500.0 Methocel K100M Hypromellose 30.0 256.5 Vivapur 101 Microcrystalline 10.0 85.7 cellulose Aerosil 200 Colloidal silicon dioxide 0.5 4.3 Magnesium stearate N/A 1.0 8.6 Total 100 855
  • the activation of the NMDA receptors by Compound 100 and by D-serine was assessed in an automated patch clamp system (ScreenPatch ⁇ ) using HEK293 cells expressing human NMDAR subunits GluN1 and GluN2A.
  • a representative graph is depicted in FIG. 14 .
  • Example 10 Evaluation of the Brain Distribution of Compound 100 in Sprague Dawley Rats after PO Administration
  • the distribution profile of Compound 100 was investigated in male Sprague-Dawley rats.
  • the rats (4) were administered a single dose of Compound 100 (oral (PO)) at 100 mg/kg.
  • oral (PO) oral
  • tissues from perfused brain and plasma were collected and analyzed by LC-MS.
  • concentration of Compound 100 in the cortex was found to be greater than in plasma or other brain locations.
  • the plasma, cortex, brain stem, and cerebellum concentration at 24 hr for Compound 100 are shown in FIG. 15 .
  • the mean concentrations at 24 hr in the plasma, cortex, brain stem, and cerebellum were 880 ng/mL, 4660 ng/g, 721 ng/g, and 290 ng/g, respectively.
  • Example 11 Evaluation of the Concentration of Compound 100 in Sprague Dawley Rats Cortex versus Plasma after 4 Days of PO Administration
  • the concentration of Compound 100 in the rat cortex versus time after 4 days of dosing 100 mg/kg is shown in FIG. 16 .
  • T 1/2 half-life of Compound 100 in the cortex (location of the target) was shown to be approximately 48 hours, in contrast to the much shorter plasma T 1/2 which was shown to be less than 12 hours.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Psychiatry (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US16/766,194 2017-11-22 2018-11-21 Deuterated analogs of d-serine and uses thereof Abandoned US20200368193A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/766,194 US20200368193A1 (en) 2017-11-22 2018-11-21 Deuterated analogs of d-serine and uses thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201762590109P 2017-11-22 2017-11-22
US201862636081P 2018-02-27 2018-02-27
US201862636427P 2018-02-28 2018-02-28
US201862755157P 2018-11-02 2018-11-02
US16/766,194 US20200368193A1 (en) 2017-11-22 2018-11-21 Deuterated analogs of d-serine and uses thereof
PCT/US2018/062263 WO2019104179A1 (fr) 2017-11-22 2018-11-21 Analogues deutérés de d-sérine et leurs utilisations

Publications (1)

Publication Number Publication Date
US20200368193A1 true US20200368193A1 (en) 2020-11-26

Family

ID=64755715

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/766,194 Abandoned US20200368193A1 (en) 2017-11-22 2018-11-21 Deuterated analogs of d-serine and uses thereof
US16/459,916 Active US10668036B2 (en) 2017-11-22 2019-07-02 Deuterated analogs of D-serine and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/459,916 Active US10668036B2 (en) 2017-11-22 2019-07-02 Deuterated analogs of D-serine and uses thereof

Country Status (12)

Country Link
US (2) US20200368193A1 (fr)
EP (1) EP3713557A1 (fr)
JP (1) JP2023123476A (fr)
KR (1) KR20200103662A (fr)
CN (1) CN111491629A (fr)
AU (1) AU2018371784A1 (fr)
BR (1) BR112020010156A2 (fr)
CA (1) CA3082834A1 (fr)
IL (1) IL274585A (fr)
MX (1) MX2020005336A (fr)
TW (1) TW201924668A (fr)
WO (1) WO2019104179A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3082834A1 (fr) 2017-11-22 2019-05-31 Concert Pharmaceuticals, Inc. Analogues deuteres de d-serine et leurs utilisations
RU2738873C2 (ru) * 2018-12-29 2020-12-17 Общество С Ограниченной Ответственностью "Сольвекс" (Ооо "Сольвекс") Препарат для магнитно-резонансной томографии, содержащий дейтерированный саркозин, и способ диагностики с использованием этого препарата
US20220169595A1 (en) * 2019-04-03 2022-06-02 Concert Pharmaceuticals, Inc. Processes for the preparation of deuterated d-serine
US20220218641A1 (en) * 2019-05-30 2022-07-14 Concert Pharmaceuticals, Inc. Methods of treatment with deuterated analogs of d-serine
JP2023507584A (ja) * 2019-12-19 2023-02-24 イントラ-セルラー・セラピーズ・インコーポレイテッド 統合失調症処置方法

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4582931A (en) * 1984-12-24 1986-04-15 Merck & Co., Inc. Preparation of 2-Deutero-D-serine
US6221335B1 (en) 1994-03-25 2001-04-24 Isotechnika, Inc. Method of using deuterated calcium channel blockers
DE69535592T2 (de) 1994-03-25 2008-06-12 Isotechnika, Inc., Edmonton Verbesserung der effektivität von arzneimitteln duren deuterierung
WO1997020552A1 (fr) 1995-12-07 1997-06-12 Albert Einstein College Of Medicine Of Yeshiva University Traitement des symptomes negatifs et cognitifs de la schizophrenie a l'aide de glycine et de ses precurseurs
PT1073432E (pt) * 1998-04-14 2007-10-22 Gen Hospital Corp Utilização da d-alanina ou da d-serina para o tratamento da esquizofrenia
US6440710B1 (en) 1998-12-10 2002-08-27 The Scripps Research Institute Antibody-catalyzed deuteration, tritiation, dedeuteration or detritiation of carbonyl compounds
US6258605B1 (en) 1999-03-26 2001-07-10 Neo Gen Screening, Inc. Clinical method for the genetic screening of newborns using tandem mass spectrometry
GB9925962D0 (en) 1999-11-02 1999-12-29 Novartis Ag Organic compounds
ES2193921T3 (es) 1999-12-03 2003-11-16 Pfizer Prod Inc Compuestos de sulfamoilheteroaril-pirazol como agentes antinflamatorios/analgesicos.
ATE282428T1 (de) 2001-05-03 2004-12-15 Hoffmann La Roche Pharmazeutische dosierungsform von amorphem nilfenavir mesylat
TW200413273A (en) 2002-11-15 2004-08-01 Wako Pure Chem Ind Ltd Heavy hydrogenation method of heterocyclic rings
IL154318A (en) * 2003-02-06 2010-05-31 Sarah Herzog Memorial Hospital Pharmaceutical compositions for the treatment of movement disorders
CN101076319A (zh) 2004-09-29 2007-11-21 科迪斯公司 稳定的非晶形雷帕霉素样化合物的药物剂型
AU2006235400A1 (en) 2005-04-11 2006-10-19 Yale University Method of treating schizophrenia prodrome
AU2006299424A1 (en) 2005-10-06 2007-04-12 Auspex Pharmaceuticals, Inc. Deuterated inhibitors of gastric H+, K+-ATPase with enhanced therapeutic properties
US7750168B2 (en) 2006-02-10 2010-07-06 Sigma-Aldrich Co. Stabilized deuteroborane-tetrahydrofuran complex
JO2630B1 (en) 2006-04-13 2012-06-17 نوفارتيس ايه جي Organic compounds
CA2661404A1 (fr) 2006-09-05 2008-03-13 Schering Corporation Compositions pharmaceutiques pour un traitement des lipides et dans le traitement de l'atherosclerose et de la steatose hepatique
IL188681A0 (en) 2008-01-09 2008-12-29 Amino Acid Solutions Inc Pharmaceutical compositions and methods utilizing a d-amino acid
US9040581B1 (en) 2013-02-21 2015-05-26 The Florida State University Research Foundation, Inc. Methods of treatment using D-serine
CA2913454C (fr) 2013-05-28 2021-09-07 Sarah Herzog Memorial Hospital-Ezrath Nashim Methodes et traitements d'un dysfonctionnement auto-immun du recepteur glutamatergique
WO2014205074A2 (fr) 2013-06-18 2014-12-24 The Trustees Of Columbia University In The City Of New York Dispositifs, compositions et procédés pour une imagerie à diffusion raman
WO2015160470A2 (fr) * 2014-03-20 2015-10-22 The Trustees Of Princeton University Production de nadph par la voie 10-formyle-thf, et son utilisation dans le diagnostic et le traitement de maladie
GB201521903D0 (en) 2015-12-11 2016-01-27 Electrophoretics Ltd Isorbaric mass labels
CN109564207B (zh) 2016-06-02 2021-07-02 梅塔博隆股份有限公司 用于检测和定量代谢物的质谱方法
CA3082834A1 (fr) 2017-11-22 2019-05-31 Concert Pharmaceuticals, Inc. Analogues deuteres de d-serine et leurs utilisations

Also Published As

Publication number Publication date
IL274585A (en) 2020-06-30
KR20200103662A (ko) 2020-09-02
JP2021504326A (ja) 2021-02-15
CA3082834A1 (fr) 2019-05-31
WO2019104179A4 (fr) 2019-08-01
CN111491629A (zh) 2020-08-04
US20200390732A1 (en) 2020-12-17
JP2023123476A (ja) 2023-09-05
MX2020005336A (es) 2020-08-13
WO2019104179A1 (fr) 2019-05-31
AU2018371784A1 (en) 2020-05-28
US20190328692A1 (en) 2019-10-31
TW201924668A (zh) 2019-07-01
US10668036B2 (en) 2020-06-02
EP3713557A1 (fr) 2020-09-30
BR112020010156A2 (pt) 2020-11-10

Similar Documents

Publication Publication Date Title
US10668036B2 (en) Deuterated analogs of D-serine and uses thereof
CN101657453B (zh) 作为PKC-θ抑制剂的嘌呤类
CA2994404A1 (fr) Acides beta-amines substitues en beta et analogues a utiliser en tant qu'agents de chimiotherapie et leurs utilisations
US20230105869A1 (en) C5aR INHIBITOR REDUCTION OF URINARY sCD163
SG178252A1 (en) Dp2 antagonist and uses thereof
US20140221492A1 (en) Deuterated 2-amino-3-hydroxypropanoic acid derivatives
US20230026438A1 (en) Protease inhibitors as antivirals
US20150166601A1 (en) Deuterated carfilzomib
US20120071554A1 (en) Deuterated 2-propylpentanoic acid compounds
US12029712B2 (en) Deuterated analogs of D-serine and uses thereof
WO2023213182A1 (fr) Sel de carebastine et son utilisation
JP7295128B2 (ja) 小分子核内受容体モジュレータのアミドプロドラッグ
CN114390924A (zh) 用于蛋白质降解的化合物、组合物和方法
US20220313640A1 (en) Deuterated analogs of d-serine and uses thereof
US20190047949A1 (en) Deuterated gft-505
JP7518765B2 (ja) D-セリンの重水素化類似体およびその使用
US20220218641A1 (en) Methods of treatment with deuterated analogs of d-serine
US10843993B2 (en) Deuterated idebenone
EP4304655A1 (fr) Conjugués de dégradation bifonctionnels à base d'acides gras et leurs procédés d'utilisation
WO2020046941A1 (fr) Formes cristallines de composés de dihydroindène-4-carboxamide substitués et leurs procédés de préparation et d'utilisation
CA2861047A1 (fr) Acide deuterie alpha-lipoique

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

AS Assignment

Owner name: CONCERT PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOLLER, DARIO;BRUMMEL, CHRISTOPHER L.;LIU, JULIE F.;AND OTHERS;SIGNING DATES FROM 20190215 TO 20190427;REEL/FRAME:052907/0299

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: SUN PHARMACEUTICAL INDUSTRIES, INC., NEW JERSEY

Free format text: MERGER;ASSIGNOR:CONCERT PHARMACEUTICALS, INC.;REEL/FRAME:064907/0514

Effective date: 20230328