US20140206667A1 - Methods and compositions for treating schizophrenia - Google Patents

Methods and compositions for treating schizophrenia Download PDF

Info

Publication number
US20140206667A1
US20140206667A1 US14/080,531 US201314080531A US2014206667A1 US 20140206667 A1 US20140206667 A1 US 20140206667A1 US 201314080531 A US201314080531 A US 201314080531A US 2014206667 A1 US2014206667 A1 US 2014206667A1
Authority
US
United States
Prior art keywords
pat
methyl
oxo
hydrogen
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/080,531
Inventor
Michela Gallagher
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.)
Johns Hopkins University
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US14/080,531 priority Critical patent/US20140206667A1/en
Publication of US20140206667A1 publication Critical patent/US20140206667A1/en
Priority to US15/629,314 priority patent/US10624875B2/en
Assigned to THE JOHNS HOPKINS UNIVERSITY reassignment THE JOHNS HOPKINS UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GALLAGHER, MICHELA
Assigned to THE JOHNS HOPKINS UNIVERSITY reassignment THE JOHNS HOPKINS UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KOH, MING TENG
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
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4015Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having oxo groups directly attached to the heterocyclic ring, e.g. piracetam, ethosuximide
    • 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
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/554Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one sulfur as ring hetero atoms, e.g. clothiapine, diltiazem
    • 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
    • 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the invention relates to methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania).
  • schizophrenia or bipolar disorder in particular, mania
  • it relates to the use of a combination of a synaptic vesicle protein 2A (SV2A) inhibitor and an antipsychotic in treating a subject having or at risk for schizophrenia or bipolar disorder (in particular, mania).
  • SV2A synaptic vesicle protein 2A
  • Schizophrenia is a chronic psychiatric disorder, characterized by a wide spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g., hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalities in the brain are proposed to underlie the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely ineffective in treating cognitive impairments in schizophrenia patients.
  • Cognitive impairments in schizophrenia involve both frontal and temporal lobe functions that include memory, attention, processing speed, and executive control.
  • Recent observations, drawn from preclinical animal models and human neuroimaging studies, indicate that altered brain activity/excitability in the medial temporal lobe memory system may contribute to cognitive impairment and may also play a role in augmenting psychotic symptoms due to disinhibition of dopaminergic neurons.
  • Cognitive deficits are increasingly recognized as a key clinical feature that can be detected in a prodromal phase and in remission, as well as during full expression of the illness but are not effectively treated by available antipsychotics. Because untreated features of schizophrenia, especially impaired cognition, predict long-term disability in patients (Green et al., Schizophr. Res. 2004, 72, 41-45), it is critical to develop effective therapies for the spectrum of this illness.
  • a method for treating a subject suffering from schizophrenia or bipolar disorder (in particular, mania), or at risk thereof comprising the step of administering to said subject a therapeutically effective amount of a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof in combination with a therapeutically effective amount of an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.
  • the methods of the present invention treat one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia.
  • the methods of the present invention treat one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania).
  • the methods of this invention prevent or slow the progression of cognitive impairment or bipolar disorder (in particular, mania) of schizophrenia in said subject.
  • the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this aspect of the invention include those disclosed in, for example, United States (U.S.) patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, and U.S. Patent Application 61/175,536.
  • any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of this aspect of the invention.
  • the SV2A inhibitor is selected from the group of SV2A inhibitors referred to in International Patent Applications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S.
  • the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof.
  • the SV2A inhibitor is levetiracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.
  • the SV2A inhibitor is brivaracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2A inhibitor is seletracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.
  • the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this invention include both typical and atypical antipsychotics.
  • the antipsychotics suitable for use in the present invention are selected from atypical antipsychotics, including, but not limited to, those disclosed in, for example, U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
  • atypical antipsychotics including, but not limited to, those disclosed in, for example, U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,0
  • atypical antipsychotics suitable for use in the present invention include, but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
  • the antipsychotic of this invention is selected from aripiprazole (Bristol-Myers Squibb), olanzapine (Lilly) and ziprasidone (Pfizer), and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
  • the antipsychotics suitable for use in the present invention are typical antipsychotics, including, but not limited to, acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothip
  • the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be selected from compounds that are dopaminergic agents (such as dopamine D1 receptor antagonists or agonists, dopamine D 2 receptor antagonists or partial agonists, dopamine D3 receptor antagonists or partial agonists, dopamine D4 receptor antagonists), glutamatergic agents, N-methyl-D-aspartate (NMDA) receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonist
  • dopaminergic agents such
  • the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this invention include compounds that may be used to treat at least one sign or symptom of schizophrenia or bipolar disorder (in particular, mania).
  • the antipsychotic is administered at a dose that is subtherapeutic as compared to the dose at which it is therapeutically effective when administered in the absence of the SV2A inhibitor.
  • the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof can be administered at doses as disclosed, for example, in U.S. patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, U.S. Patent Application 61/175,536, and U.S. Patent Application 61/441,251.
  • the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.001 mg/kg to 5 mg/kg.
  • the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 5 mg/kg, or about 1 to 2 mg/kg, or about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.01 to 1 mg/kg, or about 0.001 to 1 mg/kg, or about 0.5 mg/kg to 5 mg/kg, or about 0.05 mg/kg to 0.5 mg/kg.
  • the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 2.0 to 4.0 mg/kg, or about 2.0 to 3.0 mg/kg, or about 3.0 to 4.0 mg/kg, or about 0.2 to 0.4 mg/kg, or about 0.2 to 0.3 mg/kg, or about 0.3 to 0.4 mg/kg, or about 0.001-5 mg/kg, or about 0.001-0.5 mg/kg, or about 0.01-0.5 mg/kg.
  • the SV2A inhibitor may be selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof, said SV2A inhibitor being administered every 12 or 24 hours at a daily dose selected from any of the above.
  • the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs are administered simultaneously, or sequentially, or in a single formulation, or in separate formulations packaged together.
  • the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs are administered via different routes.
  • “combination” includes administration by any of these formulations or routes of administration.
  • the combined treatment has a longer or improved therapeutic effect in the subject than is attained by administering the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the absence of the SV2A inhibitor or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof by at least about 1.5 ⁇ , or 2.0 ⁇ , or 2.5 ⁇ , or 3.0 ⁇ , or 3.5 ⁇ , or 4.0 ⁇ , or 4.5 ⁇ , or 5.0 ⁇ , or 5.5 ⁇ , or 6.0 ⁇ , or 6.5 ⁇ , or 7.0 ⁇ , or 7.5 ⁇ , or 8.0 ⁇ , or 8.5 ⁇ , or 9.0 ⁇ , or 9.5 ⁇ , or 10 ⁇ , or greater than about 10 ⁇ .
  • the combined treatment has a longer or improved therapeutic effect in the subject than is attained by administering the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the absence of the antipsychotic or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof by at least about 1.5 ⁇ , or 2.0 ⁇ , or 2.5 ⁇ , or 3.0 ⁇ , or 3.5 ⁇ , or 4.0 ⁇ , or 4.5 ⁇ , or 5.0 ⁇ , or 5.5 ⁇ , or 6.0 ⁇ , or 6.5 ⁇ , or 7.0 ⁇ , or 7.5 ⁇ , or 8.0 ⁇ , or 8.5 ⁇ , or 9.0 ⁇ , or 9.5 ⁇ , or 10 ⁇ , or greater than about 10 ⁇ .
  • a method of increasing the therapeutic index of an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in a method of treating schizophrenia or bipolar disorder (in particular, mania) in a subject in need or at risk thereof comprising administering a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in combination with the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof to said subject.
  • the increase in the therapeutic index of the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is greater than the therapeutic index of the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof when administered in the absence of the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof by at least about 1.5 ⁇ , or 2.0 ⁇ , or 2.5 ⁇ , or 3.0 ⁇ , or 3.5 ⁇ , or 4.0 ⁇ , or 4.5 ⁇ , or 5.0 ⁇ , or 5.5 ⁇ , or 6.0 ⁇ , or 6.5 ⁇ , or 7.0 ⁇ , or 7.5 ⁇ , or 8.0 ⁇ , or 8.5 ⁇ , or 9.0 ⁇ , or 9.5 ⁇ , or 10 ⁇ , or greater than about 10 ⁇ .
  • a pharmaceutical composition for treating a subject suffering from schizophrenia or bipolar disorder (in particular, mania), or at risk thereof comprising a SV2A inhibitor and an antipsychotic or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof.
  • the composition of this invention is for treating one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia.
  • the composition of this invention is for treating one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania).
  • the composition is in a liquid form.
  • the composition is in an aqueous solution.
  • the composition is in a suspension form.
  • the composition is in a sustained release form, or a controlled release form, or a delayed release form, or an extended release form. In some embodiments, the composition is in a unit dosage form. In other embodiments, the two components of the compositions are in separate delivery forms packaged together.
  • the composition comprises levetiracetam, brivaracetam, seletracetam, or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and an antipsychotic or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.
  • the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the composition is present in an amount of 0.07-350 mg, or 50-250 mg, or 3-50 mg. In some embodiments, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is present in an amount less than 350 mg, less than 250 mg, less than 200 mg, less than 150 mg, less than 100 mg, less than 50 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0.5 mg, less than 0.1 mg, or less than 0.07 mg.
  • the SV2A inhibitor is present in an amount of 0.07-60 mg, 0.07-350 mg, 25-60 mg, 25-125 mg, 50-250 mg, 5-140 mg, 0.7-180 mg, 125-240 mg, 3-50 mg, or 3-60 mg. In other embodiments of this aspect of the invention, the SV2A inhibitor is present in an amount of 0.05-35 mg. In some embodiments of the composition of this invention, the SV2A inhibitor may be selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof, said SV2A inhibitor being present in an amount selected from any of the above.
  • FIG. 1 depicts increased mRNA expression of the gene encoding SV2A in the dentate gyrus of the hippocampus of aged-impaired rats (AI) as compared to young rats (Y) and aged-unimpaired rats (AU).
  • Normalized Affymetrix GeneChip probe set signal values (Y-axis), as a measure of mRNA expression, are plotted against learning indices of different rats, as a measure of cognitive impairment.
  • FIG. 2 depicts the effects of administering levetiracetam on the spatial memory retention of six aged-impaired rats (AI) in a Morris Water Maze (MWM) test.
  • AI aged-impaired rats
  • MLM Morris Water Maze
  • Three treatment conditions were employed: vehicle control, levetiracetam (5 mg/kg/day) and levetiracetam (10 mg/kg/day).
  • the AI rats were trained for two consecutive days, with a one-time treatment prior to the training trials per day. 24 hours later, the AI rats were tested. The time the AI rats, 24 hours after treatment with the different conditions and two days of training, spent swimming in the target quadrant or the target annulus in a memory retention trial is used as a measure of spatial memory retention.
  • the target quadrant refers to the quadrant of the maze (which is a circular pool) where the escape platform is placed during the training trials.
  • the target annulus refers to the exact location of the escape platform during the training trials.
  • FIG. 3 depicts the effects of administering levetiracetam on the spatial memory retention of ten aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test.
  • Six treatment conditions were employed: vehicle control, levetiracetam (1.25 mg/kg), levetiracetam (2.5 mg/kg), levetiracetam (5 mg/kg), levetiracetam (10 mg/kg) and levetiracetam (20 mg/kg).
  • vehicle control levetiracetam (1.25 mg/kg
  • levetiracetam 2.5 mg/kg
  • levetiracetam 5 mg/kg
  • levetiracetam (10 mg/kg) levetiracetam
  • levetiracetam (20 mg/kg levetiracetam
  • Rats were pre-treated 30-40 minutes before daily trials with a one-time drug/control treatment.
  • the number of errors made by the rats after the delay was used as a measure of spatial memory retention. Errors were defined as instances when rats entered an arm from which food had already been retrieved in the pre-delay component of the trial or when rats re-visited an arm in the post-delay session that had already been visited. Paired t-tests were used to compare the number of errors between different doses of levetiracetam and vehicle control.
  • FIG. 4 depicts the effects of administering levetiracetam or valproate separately on the spatial memory retention of ten aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test.
  • FIG. 5 depicts the effects of administering levetiracetam or valproate in combination on the spatial memory retention often aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test.
  • FIG. 6 shows an isobologram plotting levetiracetam dose against valproate dose.
  • the diagonal straight line is the line of additivity, anchored on each axis by the lowest effective doses of valproate and levetiracetam when assessed individually.
  • FIG. 7 depicts the experimental design of the human trials for levetiracetam treatment.
  • FIG. 8A depicts the average activity in the left CA3 of aMCI subjects with placebo treatment and age-matched control subjects with placebo treatment during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 8B depicts the average activity in the left CA3 of aMCI subjects with placebo treatment or levetiracetam treatment (125 mg twice a day for two weeks) during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 8C is a table of the data represented in FIGS. 8A and 8B .
  • FIG. 9A depicts the average activity in the left entorhinal cortex of age-matched control subjects with placebo treatment and aMCI subjects with placebo treatment during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 9B depicts the average activity in the left entorhinal cortex of the same aMCI subjects with placebo treatment or levetiracetam treatment (125 mg twice a day for two weeks) during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 9C is a table of the data represented in FIGS. 9A and 9B .
  • FIG. 10A depicts an example of the sequence of images shown to subjects in the explicit 3-alternative forced choice task described in Example 2.
  • FIG. 10B shows sample pairs of similar (“lure”) images.
  • FIG. 11 shows the difference between the aMCI (placebo) subjects and age-matched control (placebo) subjects in their performance of the explicit 3-alternative forced choice task described in Example 2.
  • Each bar represents the proportion of the subject responses (old, similar, or new) when presented with a lure image.
  • FIG. 12 shows the difference between the same aMCI subjects with placebo treatment or with levetiracetam treatment (125 mg twice a day for two weeks) in their performance of the explicit 3-alternative forced choice task described in Example 2.
  • Each bar represents the proportion of the subjects responses (old, similar, or new) when presented with a lure image.
  • FIG. 13 is a table of the data represented in FIGS. 11 and 12 .
  • FIG. 14A shows the difference between the age-matched control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Buschke Selective reminding Test—Delayed Recall.
  • FIG. 14B is a table of the data represented in FIG. 14A .
  • FIG. 15A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Benton Visual Retention Test.
  • FIG. 15B is a table of the data represented in FIG. 15A .
  • FIG. 16A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Verbal Paired Associates Test—Recognition.
  • FIG. 16B is a table of the data represented in FIG. 16A .
  • FIG. 17A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Verbal Paired Associates Test—Delayed Recall.
  • FIG. 17B is a table of the data represented in FIG. 17A .
  • FIG. 18A is a table showing the subject selection process for the human levetiracetam trial described in Example 2.
  • FIG. 18B is a table showing the characteristics of the subjects selected for the human levetiracetam trial described in Example 2.
  • agent is used herein to denote a chemical compound (such as an organic or inorganic compound, a mixture of chemical compounds), a biological macromolecule (such as a nucleic acid, an antibody, including parts thereof as well as humanized, chimeric and human antibodies and monoclonal antibodies, a protein or portion thereof, e.g., a peptide, a lipid, a carbohydrate), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues.
  • Agents include, for example, agents which are known with respect to structure, and those which are not known with respect to structure.
  • a “patient”, “subject”, or “individual” are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (including bovines, porcines, etc.), companion animals (e.g., canines, felines, etc.) and rodents (e.g., mice and rats).
  • “Cognitive function” or “cognitive status” refers to any higher order intellectual brain process or brain state, respectively, involved in learning and/or memory including, but not limited to, attention, information acquisition, information processing, working memory, short-term memory, long-term memory, anterograde memory, retrograde memory, memory retrieval, discrimination learning, decision-making, inhibitory response control, attentional set-shifting, delayed reinforcement learning, reversal learning, the temporal integration of voluntary behavior, expressing an interest in one's surroundings and self-care, speed of processing, reasoning and problem solving and social cognition.
  • cognitive function may be measured, for example and without limitation, by the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Selective Reminding Test (Buschke and Fuld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest; the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997); the Benton Visual Retention Test, or the explicit 3-alternative forced choice task, or MATRICS consensus neuropsychological test battery.
  • CBIIC-plus scale the Mini Mental State Exam
  • NPI Neuropsychiatric Inventory
  • CDR Clinical Dementia Rating Scale
  • CDR Clinical Dementia Rating Scale
  • CANTAB Cambridge Neuropsychological Test Automated Battery
  • cognitive function may be measured in various conventional ways known in the art, including using a Morris Water Maze (MWM), Barnes circular maze, elevated radial arm maze, T maze or any other mazes in which the animals use spatial information.
  • MMM Morris Water Maze
  • Cognitive function can be assessed by reversal learning, extradimensional set shifting, conditional discrimination learning and assessments of reward expectancy.
  • Other tests known in the art may also be used to assess cognitive function, such as novel object recognition and odor recognition tasks.
  • Cognitive function may also be measured using imaging techniques such as Positron Emission Tomography (PET), functional magnetic resonance imaging (fMRI), Single Photon Emission Computed Tomography (SPECT), or any other imaging technique that allows one to measure brain function.
  • PET Positron Emission Tomography
  • fMRI functional magnetic resonance imaging
  • SPECT Single Photon Emission Computed Tomography
  • electrophysiological techniques any other imaging technique that allows one to measure brain function.
  • “Promoting” cognitive function refers to affecting impaired cognitive function so that it more closely resembles the function of a normal, unimpaired subject.
  • Cognitive function may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject.
  • Preserving cognitive function refers to affecting normal or impaired cognitive function such that it does not decline or does not fall below that observed in the subject upon first presentation or diagnosis, or delays such decline.
  • “Improving” cognitive function includes promoting cognitive function and/or preserving cognitive function in a subject.
  • Cognitive impairment refers to cognitive function in subjects that is not as robust as that expected in a normal, unimpaired subject. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in a normal, unimpaired subject. In other cases, “cognitive impairment” in subjects affected by schizophrenia or bipolar disorder (in particular, mania) refers to cognitive function in subjects that is not as robust as that expected in normal, unimpaired subject.
  • “Schizophrenia” refers to a chronic debilitating disorder, characterized by a spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g. hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalities in the brain are proposed to underlie the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely ineffective in treating cognitive impairments in patients.
  • BP Bipolar disorder
  • manic depressive disorder or “manic depressive illness” refers to a chronic psychological/mood disorder which can be characterized by significant mood changes including periods of depression and euphoric manic periods.
  • BP may be diagnosed by a skilled physician based on personal and medical history, interview consultation and physical examinations.
  • mania or “manic periods” or other variants refers to periods where an individual exhibits some or all of the following characteristics: racing thoughts, rapid speech, elevated levels of activity and agitation as well as an inflated sense of self-esteem, euphoria, poor judgment, insomnia, impaired concentration and aggression.
  • Treating” a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results.
  • Beneficial or desired clinical results include, but are not limited to, preventing or slowing the progression of the disease or disorder, or alleviation, amelioration, or slowing the progression, of one or more symptoms associated with CNS disorders with cognitive impairment, such as schizophrenia or bipolar disorder (in particular, mania).
  • Treating cognitive impairment refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject's performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline.
  • that subject's cognitive function after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject.
  • Treatment of cognitive impairment in humans may improve cognitive function to any detectable degree, but is preferably improved sufficiently to allow the impaired subject to carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject.
  • “treating cognitive impairment” refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject's performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline.
  • that subject's cognitive function, after treatment of cognitive impairment more closely resembles the function of a normal, unimpaired subject.
  • “treating cognitive impairment” in a subject affecting by schizophrenia or bipolar disorder (in particular, mania) refers to takings steps to improve cognitive function in the subject so that the subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject.
  • administering or “administration of” a substance, a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art.
  • a compound or an agent can be administered, intravenously, arterially, intradermally, intramuscularly, intraperitonealy, intravenously, subcutaneously, ocularly, sublingually, orally (by ingestion), intranasally (by inhalation), intraspinally, intracerebrally, and transdermally (by absorption, e.g. through a skin duct).
  • a compound or agent can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, or controlled release of the compound or agent.
  • Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
  • the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug.
  • a physician who instructs a patient to self-administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is administering the drug to the patient.
  • a compound or an agent is administered orally, e.g., to a subject by ingestion, or intravenously. e.g., to a subject by injection.
  • the orally administered compound or agent is in an extended release or slow release formulation, or administered using a device for such slow or extended release.
  • SV2A inhibitor refers to any agent, substance or compound that binds to SV2A and reduces synaptic function by reducing pre-synaptic vesicle release (See, e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006; Custer et al., 2006; Smedt et al., 2007; Yang et al., 2007; Meehan, “Levetiracetam has an activity-dependent effect on inhibitory transmission,” Epilepsia, 2012 Jan.
  • a substance, or a compound or an agent is an SV2A inhibitor even if it does not itself bind to SV2A, as long as it causes, or affects the ability of, another compound or agent to bind SV2A or reduce synaptic function by reducing pre-synaptic vesicle release.
  • SV2A inhibitors include pharmaceutically acceptable salts of the inhibitors thereof. They also include hydrates, polymorphs, prodrugs, salts, and solvates of these inhibitors.
  • Antipsychotic refers to (1) a typical or an atypical antipsychotic; (2) an agent that is selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid
  • Typical antipsychotics refer to conventional antipsychotics, which produce antipsychotic effects as well as movement related adverse effects related to disturbances in the nigrostriatal dopamine system.
  • extrapyramidal side effects include Parkinsonism, akathisia, tardive dyskinesia and dystonia. See Baldessarini and Tarazi in Goodman & Gilman's The Pharmacological Basis of Therapeutics 10 Edition, 2001, pp. 485-520.
  • “Atypical antipsychotics”, as used herein, refer to antipsychotic drugs that produce antipsychotic effects with little or no EPS and include, but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone. “Atypical” antipsychotics differ from conventional antipsychotics in their pharmacological profiles. While conventional antipsychotics are characterized principally by D 2 dopamine receptor blockade, atypical antipsychotics show antagonist effects on multiple receptors including the 5HT a and 5HT c serotonin receptors and varying degrees of receptor affinities.
  • Atypical antipsychotic drugs are commonly referred to as serotoninidopamine antagonists, reflecting the influential hypothesis that greater affinity for the 5-HTT 2 receptor than for the D 2 receptor underlies “atypical” antipsychotic drug action or “second generation” antipsychotic drugs.
  • the atypical antipsychotics often display side effects, including, but not limited to, weight gain, diabetes (e.g., type II diabetes mellitus), hyperlipidemia, QTc interval prolongation, myocarditis, sexual side effects, extrapyramidal side effects and cataract.
  • atypical antipsychotics do not represent a homogeneous class, given their differences in the context of both alleviation of clinical symptoms and their potential for inducing side effects such as the ones listed above.
  • the common side effects of the atypical antipsychotics as described above often limit the antipsychotic doses that can be used for these agents.
  • SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs are administered with a time separation of no more than about 15 minutes, and in some embodiments no more than about 10 minutes.
  • the SV2A inhibitor and the antipsychotic, or their salts, hydrates, solvates, or polymorphs may be contained in the same dosage (e.g., a unit dosage form comprising both the SV2A inhibitor and the antipsychotic) or in discrete dosages (e.g., the SV2A inhibitor or its salt, hydrate, solvate, or polymorph is contained in one dosage form and the antipsychotic or its salt, hydrate, solvate, or polymorph is contained in another dosage form).
  • sequential administration means that the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, are administered with a time separation of more than about 15 minutes, and in some embodiments more than about one hour, or up to 12-24 hours. Either the SV2A inhibitor or the antipsychotic may be administered first.
  • the SV2A inhibitor and the antipsychotic, or their salts, hydrates, solvents, or polymorphs, for sequential administration may be contained in discrete dosage forms, optionally contained in the same container or package.
  • a “therapeutically effective amount” of a drug or agent is an amount of a drug or an agent that, when administered to a subject will have the intended therapeutic effect. e.g. improving cognitive function in a subject in a subject suffering from a disease or disorder (e.g., schizophrenia or bipolar disorder (in particular, mania)), preventing or slowing the progression of a disease or disorder (e.g., schizophrenia or bipolar disorder (in particular, mania)), and/or alleviating, ameliorating, or slowing the progression of one or more symptoms associated with the disease or disorder (e.g., schizophrenia or bipolar disorder (in particular, mania)).
  • a disease or disorder e.g., schizophrenia or bipolar disorder (in particular, mania)
  • preventing or slowing the progression of a disease or disorder e.g., schizophrenia or bipolar disorder (in particular, mania)
  • alleviating, ameliorating, or slowing the progression of one or more symptoms associated with the disease or disorder e.g., schizophrenia or bipolar disorder (in particular,
  • a therapeutically effective amount may be administered in one or more administrations.
  • the precise effective amount needed for a subject will depend upon, for example, the subject's size, health and age, the nature and extent of the cognitive impairment, and the therapeutics or combination of therapeutics selected for administration, and the mode of administration. The skilled worker can readily determine the effective amount for a given situation by routine experimentation.
  • “Subtherapeutic amount” refers to an amount administered of an agent or compound of the invention that is less than the therapeutic amount, that is, less than the amount normally used when said agent or compound is administered alone (i.e., individually and in the absence of other therapeutic agents or compounds) to treat disorders, such as schizophrenia or bipolar disorder (in particular, mania).
  • Analog is used herein to refer to a compound which functionally resembles another chemical entity, but does not share the identical chemical structure.
  • an analog is sufficiently similar to a base or parent compound such that it can substitute for the base compound in therapeutic applications, despite minor structural differences.
  • “Derivative” is used herein to refer to the chemical modification of a compound. Chemical modifications of a compound can include, for example, replacement of hydrogen by an alkyl, acyl, or amino group. Many other modifications are also possible.
  • prodrug is art-recognized and is intended to encompass compounds or agents which, under physiological conditions, are converted into a SV2A inhibitor or an antipsychotic.
  • a common method for making a prodrug is to select moieties which are hydrolyzed or metabolized under physiological conditions to provide the desired compound or agent.
  • the prodrug is converted by, for example, an enzymatic activity of the host animal to a SV2A inhibitor or an antipsychotic.
  • aliphatic as used herein means a straight chained or branched alkyl, alkenyl or alkynyl. It is understood that alkenyl or alkynyl embodiments need at least two carbon atoms in the aliphatic chain. Aliphatic groups typically contains from 1 (or 2) to 12 carbons, such as from 1 (or 2) to 4 carbons.
  • aryl as used herein means a monocyclic or bicyclic carbocyclic aromatic ring system.
  • aryl as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic carbocyclic aromatic ring system.
  • Phenyl is an example of a monocyclic aromatic ring system.
  • Bicyclic aromatic ring systems include systems wherein both rings are aromatic, e.g., naphthyl, and systems wherein only one of the two rings is aromatic, e.g., tetralin.
  • heterocyclic as used herein means a monocyclic or bicyclic non-aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH, S, SO, or SO 2 in a chemically stable arrangement.
  • heterocyclic as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic non-aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH, S, SO, or SO 2 in a chemically stable arrangement.
  • a bicyclic non-aromatic ring system embodiment of “heterocyclyl” one or both rings may contain said heteroatom or heteroatom groups.
  • one of the two rings may be aromatic.
  • a non-aromatic heterocyclic ring may optionally be fused to an aromatic carbocycle.
  • heterocyclic rings examples include 3-1H-benzimidazol-2-one, 3-(1-alkyl)-benzimidazol-2-one, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholino, 3-morpholino.
  • heteroaryl as used herein means a monocyclic or bicyclic aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH or S in a chemically stable arrangement.
  • heteroaryl as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH or S in a chemically stable arrangement.
  • heteroaryl as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH or S in a chemically stable arrangement.
  • one or both rings may contain said heteroatom or heteroatom groups.
  • heteroaryl rings examples include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, benzimidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl.
  • 2-pyrazolyl isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, purinyl, pyrazinyl, 1,3,5-triazinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl).
  • quinolinyl e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl
  • isoquinolinyl e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinoliny
  • cycloalkyl or cycloalkenyl refers to a monocyclic or fused or bridged bicyclic carbocyclic ring system that is not aromatic.
  • cycloalkyl or cycloalkenyl as used herein can be a C5-C10 monocyclic or fused or bridged C8-C12 bicyclic carbocyclic ring system that is not aromatic.
  • Cycloalkenyl rings have one or more units of unsaturation.
  • Preferred cycloalkyl or cycloalkenyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, norbornyl, adamantyl and decalinyl.
  • the carbon atom designations may have the indicated integer and any intervening integer.
  • the number of carbon atoms in a (C1-C4)-alkyl group is 1, 2, 3, or 4. It should be understood that these designation refer to the total number of atoms in the appropriate group.
  • the total number of carbon atoms and heteroatoms is 3 (as in aziridine). 4, 5, 6 (as in morpholine), 7, 8, 9, or 10.
  • “Pharmaceutically acceptable salt” is used herein to refer to an agent or a compound according to the invention that is a therapeutically active, non-toxic base and acid salt form of the compounds.
  • the acid addition salt form of a compound that occurs in its free form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclic, salicylic, p-aminosalicylic, pamoic and the like. See, e.g., WO 01/
  • Compounds containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt form, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases.
  • Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts. e.g., lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • said salt forms can be converted into the free forms by treatment with an appropriate base or acid.
  • Compounds and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like. See, e.g., WO 01/062726.
  • hydrate refers to a combination of water with a compound wherein the water retains its molecular state as water and is either absorbed, adsorbed or contained within a crystal lattice of the substrate compound.
  • polymorph refers to different crystalline forms of the same compound and other solid state molecular forms including pseudo-polymorphs, such as hydrates (e.g., bound water present in the crystalline structure) and solvates (e.g., bound solvents other than water) of the same compound.
  • pseudo-polymorphs such as hydrates (e.g., bound water present in the crystalline structure) and solvates (e.g., bound solvents other than water) of the same compound.
  • pseudo-polymorphs such as hydrates (e.g., bound water present in the crystalline structure) and solvates (e.g., bound solvents other than water) of the same compound.
  • hydrates e.g., bound water present in the crystalline structure
  • solvates e.g., bound solvents other than water
  • Crystalline polymorphic forms are of interest to the pharmaceutical industry and especially to those involved in the development of suitable dosage forms. If the polymorphic form is not held constant during clinical or stability studies, the exact dosage form used or studied may not be comparable from one lot to another. It is also desirable to have processes for producing a compound with the selected polymorphic form in high purity when the compound is used in clinical studies or commercial products since Impurities present may produce undesired toxicological effects. Certain polymorphic forms may exhibit enhanced thermodynamic stability or may be more readily manufactured in high purity in large quantities, and thus are more suitable for inclusion in pharmaceutical formulations. Certain polymorphs may display other advantageous physical properties such as lack of hygroscopic tendencies, improved solubility, and enhanced rates of dissolution due to different lattice energies.
  • stereogenic center in their structure.
  • This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem. (1976), 45, 11-30.
  • the invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds or mixtures thereof (including all possible mixtures of stereoisomers). See, e.g., WO 01/062726.
  • certain compounds which contain alkenyl groups may exist as Z (zusammen) or E (entussi) isomers.
  • the invention includes both mixture and separate individual isomers.
  • Multiple substituents on a piperidinyl or the azepanyl ring can also stand in either cis or trans relationship to each other with respect to the plane of the piperidinyl or the azepanyl ring.
  • Some of the compounds may also exist in tautomeric forms. Such forms, although not explicitly indicated in the formulae described herein, are intended to be included within the scope of the present invention.
  • the methods of this invention comprise administration of a SV2A inhibitor or a pharmaceutically acceptable salt thereof in combination with administration of an antipsychotic or a pharmaceutically acceptable salt thereof.
  • the agents or compounds of the SV2A inhibitor or the antipsychotic and their pharmaceutically acceptable salts also include hydrates, solvates, polymorphs, and prodrugs of those agents, compounds, and salts.
  • Animal models serve as an important resource for developing and evaluating treatments for CNS disorders with cognitive impairment.
  • Features that characterize cognitive impairment in animal models typically extend to cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be predictive of efficacy in humans.
  • the extent of cognitive impairment in an animal model for a CNS disorder, and the efficacy of a method of treatment for said CNS disorder may be tested and confirmed with the use of a variety of cognitive tests.
  • a Radial Arm Maze (RAM) behavioral task is one example of a cognitive test, specifically testing spacial memory (Chappell et al. Neuropharmacology 37: 481-487, 1998).
  • the RAM apparatus consists of, e.g., eight equidistantly spaced arms. A maze arm projects from each facet of a center platform. A food well is located at the distal end of each arm. Food is used as a reward. Blocks can be positioned to prevent entry to any arm. Numerous extra maze cues surrounding the apparatus may also be provided. After habituation and training phases, spatial memory of the subjects may be tested in the RAM under control or test compound-treated conditions.
  • subjects are pretreated before trials with a vehicle control or one of a range of dosages of the test compound.
  • a subset of the arms of the eight-arm maze is blocked.
  • Subjects are allowed to obtain food on the unblocked arms to which access is permitted during this initial “information phase” of the trial.
  • Subjects are then removed from the maze for a delay period, e.g., a 60 second delay, a 15 minute delay, a one-hour delay, a two-hour delay, a six hour delay, a 24 hour delay, or longer) between the information phase and the subsequent “retention test,” during which the barriers on the maze are removed, thus allowing access to all eight arms.
  • a cognitive test that may be used to assess the effects of a test compound on the cognitive impairment of a CNS disorder model animal is the Morris water maze.
  • a water maze is a pool surrounded with a novel set of patterns relative to the maze.
  • the training protocol for the water maze may be based on a modified water maze task that has been shown to be hippocampal-dependent (de Hoz et al., Eur. J. Neurosci., 22:745-54, 2005; Steele and Morris, Hippocampus 9:118-36, 1999).
  • the subject is trained to locate a submerged escape platform hidden underneath the surface of the pool.
  • a subject is released in the maze (pool) from random starting positions around the perimeter of the pool.
  • the starting position varies from trial to trial. If the subject does not locate the escape platform within a set time, the experimenter guides and places the subject on the platform to “teach” the location of the platform. After a delay period following the last training trial, a retention test in the absence of the escape platform is given to assess spatial memory.
  • the subject's level of preference for the location of the (now absent) escape platform as measured by, e.g., the time spent in that location or the number of crossings of that location made by the mouse, indicates better spatial memory, i.e., treatment of cognitive impairment.
  • the preference for the location of the escape platform under different treatment conditions can then be compared for efficacy of the test compound in treating CNS disorders with cognitive impairment.
  • This invention provides methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania) using a SV2A inhibitor or a pharmaceutically acceptable salt thereof in combination with an antipsychotic or a pharmaceutically acceptable salt thereof.
  • treatment comprises preventing or slowing the progression of schizophrenia or bipolar disorder (in particular, mania).
  • Schizophrenia is characterized by a wide spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g., hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment.
  • treatment comprises alleviation, amelioration or slowing the progression of one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia.
  • psychiatric diseases such as schizotypical and schizoaffective disorder, other acute- and chronic psychoses and bipolar disorder (in particular, mania), which have an overlapping symptomatology with schizophrenia.
  • treatment comprises alleviation, amelioration or slowing the progression of one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania).
  • the methods and compositions may be used for human patients in clinical applications in treating schizophrenia or bipolar disorder (in particular, mania).
  • the dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
  • Cognitive impairments are associated with schizophrenia. They precede the onset of psychosis and are present in non-affected relatives. The cognitive impairments associated with schizophrenia constitute a good predictor for functional outcome and are a core feature of the disorder. Cognitive features in schizophrenia reflect dysfunction in frontal cortical and hippocampal circuits. Patients with schizophrenia also present hippocampal pathologies such as reductions in hippocampal volume, reductions in neuronal size and dysfunctional hyperactivity. An imbalance in excitation and inhibition in these brain regions has also been documented in schizophrenic patients suggesting that drugs targeting inhibitory mechanisms could be therapeutic. See, e.g., Guidotti et al., Psychopharmacology 180: 191-205, 2005; Zierhut. Psych. Res. Neuroimag.
  • Animal models serve as an important resource for developing and evaluating treatments for schizophrenia.
  • Features that characterize schizophrenia in animal models typically extend to schizophrenia in humans.
  • efficacy in such animal models is expected to be predictive of efficacy in humans.
  • Various animal models of schizophrenia are known in the art.
  • Methionine-treated mice exhibit deficient expression of GAD67 in frontal cortex and hippocampus, similar to those reported in the brain of postmortem schizophrenia patients. They also exhibit prepulse inhibition of startle and social interaction deficits (Tremonlizzo et al., PNAS, 99: 17095-17100, 2002).
  • Another animal model of schizophrenia is methylaoxymethanol acetate (MAM)-treatment in rats. Pregnant female rats are administered MAM (20 mg/kg, intraperitoneal) on gestational day 17. MAM-treatment recapitulate a pathodevelopmental process to schizophrenia-like phenotypes in the offspring, including anatomical changes, behavioral deficits and altered neuronal information processing.
  • MAM-treated rats display a decreased density of parvalbumin-positive GABAergic interneurons in portions of the prefrontal cortex and hippocampus.
  • MAM-treated rats display reduced latent inhibition.
  • Latent inhibition is a behavioral phenomenon where there is reduced learning about a stimulus to which there has been prior exposure with any consequence. This tendency to disregard previously benign stimuli, and reduce the formation of association with such stimuli is believed to prevent sensory overload. Low latent inhibition is indicative of psychosis.
  • Latent inhibition may be tested in rats in the following manner. Rats are divided into two groups. One group is pre-exposed to a tone over multiple trials. The other group has no tone presentation.
  • Both groups are then exposed to an auditory fear conditioning procedure, in which the same tone is presented concurrently with a noxious stimulus, e.g. an electric shock to the foot. Subsequently, both groups are presented with the tone, and the rats' change in locomotor activity during tone presentation is monitored. After the fear conditioning the rats respond to the tone presentation by strongly reducing locomotor activity. However, the group that has been exposed to the tone before the conditioning period displays robust latent inhibition: the suppression of locomotor activity in response to tone presentation is reduced. MAM-treated rats, by contrast show impaired latent inhibition. That is, exposure to the tone previous to the fear conditioning procedure has no significant effect in suppressing the fear conditioning. (see Lodge et al., J. Neurosci., 29:2344-2354, 2009) Such animal models of schizophrenia may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
  • a noxious stimulus e.g. an electric shock to the foot.
  • MAM-treated rats display a significantly enhanced locomotor response (or aberrant locomotor activity) to low dose D-amphetamine administration.
  • the MAM-treated rats also display a significantly greater number of spontaneously firing ventral tegmental dopamine (DA) neurons.
  • DA ventral tegmental dopamine
  • MAM-treated rats in the above study may be suitable for use to assay the effectiveness of the methods and compositions of the present invention in treating schizophrenia or bipolar disorder (in particular, mania).
  • the methods and compositions of this invention maybe evaluated, using MAM-treated animals, for their effects on the central hippocampus (vHipp) regulation, on the elevated DA neuron population activity and on the hyperactive locomotor response to amphetamine in the MAM-treated animals.
  • vHipp central hippocampus
  • HPC hippocampal
  • PAM benzodiazepine-positive allosteric modulator
  • the ⁇ 5GABAAR PAM reduces the number of spontaneously active DA neurons in the ventral tegmental area (VTA) of MAM rats to levels observed in saline-treated rats (control group), both when administered systemically and when directly infused into the ventral HPC. Moreover, HPC neurons in both saline-treated and MAM-treated animals show diminished cortical-evoked responses following the ⁇ 5GABAAR PAM treatment. In addition, the increased locomotor response to amphetamine observed in MAM-treated rats is reduced following the ⁇ 5GABA A R PAM treatment. See Gill K. M et al. Neuropsychopharmacology (2011), 1-9.
  • MAM-treated rats in the above study may be suitable for use in the present invention to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
  • the methods and compositions of this invention maybe evaluated, using MAM-treated animals, for their effects on the output of the hippocampal (HPC) and on the hyperactive locomotor response to amphetamine in the MAM-treated animals.
  • HPC hippocampal
  • Apomorphine-induced climbing (AIC) and stereotype (AIS) in mice is another animal model useful in this invention.
  • Agents are administered to mice at a desired dose level (e.g., via intraperitoneal administration).
  • a desired dose level e.g., via intraperitoneal administration.
  • experimental mice are challenges with apomorphine (e.g., with 1 mg/kg sc).
  • apomorphine e.g., with 1 mg/kg sc
  • the sniffing-licking-gnawing syndrome stereotyped behavior
  • climbing behavior induced by apomorphine are scored and recorded for each animal. Readings can be repeated every 5 min during a 30-min test session. Scores for each animal are totaled over the 30-min test session for each syndrome (stereotyped behavior and climbing).
  • the efficacy of the methods and compositions of this invention in treating schizophrenia may also be assessed in animal models of schizophrenia or bipolar disorder (in particular, mania), as well as human subjects with schizophrenia, using a variety of cognitive tests known in the art, as discussed above.
  • SV2A inhibitors are also be assessed in animal models of schizophrenia or bipolar disorder (in particular, mania), as well as human subjects with schizophrenia, using a variety of cognitive tests known in the art, as discussed above.
  • SV2A synaptic vesicle protein-2
  • SV2B synaptic vesicle protein-2
  • SV2C synaptic vesicle protein-2
  • the proteins are integral membrane proteins and have a low-level homology (20-30%) to the twelve transmembrane family of bacterial and fungal transporter proteins that transport sugar, citrate, and xenobiotics (Bajjalieh et al., Science. 257: 1271-1273. (1992)).
  • SV2 family proteins are present in the brain and endocrine cells, and further are present in all synaptic and endocrine vesicles.
  • SV2 proteins are reported to play a role in normal synaptic function, and functions in a maturation step of primed vesicles that converts the vesicles into a Cat( 2+ )- and synaptotagmin-responsive state (Sudhof et al., 2009). Functionally, SV2 proteins are reported to enhance synaptic currents and increase the probability of transmitter release by maintaining the size of the readily releasable pool of vesicles (Custer et al., 2006).
  • SV2A inhibitor refers to any agent, substance or compound that binds to SV2A and reduces synaptic function by reducing pre-synaptic vesicle release (See, e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006; Custer et al., 2006; Smedt et al., 2007; Yang et al., 2007; Meehan, “Levetiracetam has an activity-dependent effect on inhibitory transmission,” Epilepsia, 2012 Jan.
  • a substance, or a compound or an agent is an SV2A inhibitor even if it does not itself bind to SV2A, as long as it causes, or affects the ability of, another compound or agent to bind SV2A or reduce synaptic function by reducing pre-synaptic vesicle release.
  • SV2A inhibitors include pharmaceutically acceptable salts of the inhibitors thereof. They also include hydrates, polymorphs, prodrugs, salts, and solvates of these inhibitors.
  • SV2A inhibitors or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof that are useful in the methods and compositions of this invention are those disclosed, for example, U.S. patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, and U.S. Patent Application 61/175,536.
  • any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of the invention.
  • the SV2A inhibitor is selected from the group of SV2A inhibitors referred to in International Patent Applications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S.
  • Other SV2A inhibitors may also be used in this invention.
  • Applicants also refer to methods of preparing these compounds found in the documents cited above.
  • Other synthetic methods may also be used. These methods are well known to those skilled in the art.
  • the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, or prodrugs thereof.
  • the SV2A inhibitor is levetiracetam or salts, solvates, hydrates, polymorphs or prodrugs thereof.
  • Levetiracetam refers to the International Union of Pure and Applied Chemistry (IUPAC) name of the compound (2S)-2-(2-oxopyrrolidin-1-yl) butanamide).
  • Levetiracetam is a widely used antiepileptic drug.
  • Levetiracetam binds to a specific site in the CNS: the synaptic vesicle protein 2A (SV2A) (See. e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006) and has further been shown to directly inhibit synaptic activity and neurotransmission by inhibiting presynaptic neurotransmitter release (Yang et al. 2007).
  • SV2A synaptic vesicle protein 2A
  • SV2A inhibitors useful for the methods and compositions of this invention are the following:
  • X is —CA 1 NR 5 R 6 or —CA 1 OR 7 or —CA 1 -R 8 or CN;
  • a 1 and A 2 are independently oxygen, sulfur or —NR 9 ;
  • R 1 is hydrogen, alkyl, aryl or —CH 2 —R 1a wherein R 1a is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
  • R 2 , R 3 and R 4 are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative:
  • R 2a , R 3a and R 4a are the same or different and each is independently hydrogen, halogen, alkyl, alkenyl, alkynyl or aryl;
  • R 8 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative
  • R 2 , R 3 , R 4 , R 2 , R 3a and R 4a is other than hydrogen; and that when the compound is a mixture of all possible isomers, X is —CONR 5 R 6 , A 2 is oxygen and R 1 is hydrogen, methyl, ethyl or propyl then substitution on the pyrollidine ring is other than mono-, di-, or tri-methyl or mono-ethyl; and that when R 1 , R 2 , R 4 , R 2a , R 3a and R 4a are each hydrogen, A2 is oxygen and X is CONR 5 R 6 then R 3 is different from carboxy, ester, amido, substituted oxo-pyrrolidine, hydroxy, oxy derivative, amino, amino derivatives, methyl, naphthyl, phenyl optionally substituted by oxy derivatives or in the para position by an halogen atom.
  • R 11 and R 12 are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfinyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.
  • oxy derivative is defined as including —O—R 11 groups wherein R 11 is as defined above except for “oxy derivative”.
  • Non-limiting examples are alkoxy, alkenyloxy, alkynyloxy, acyloxy, oxyester, oxyamido, alkylsulfonyloxy, alkylsulfinyloxy, arylsulfonyloxy, arylsulfinyloxy, aryloxy, aralkoxy or heterocyclooxy such as pentyloxy, allyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methylenedioxy, carbonate.
  • thio derivative as used herein, is defined as including —S—R 11 groups wherein R 11 is as defined above except for “thio derivative”.
  • Non-limiting examples are alkylthio, alkenylthio, alkynylthio and arylthio.
  • amino derivative as used herein, is defined as including —NHR 11 or —NR 11 R 12 groups wherein R 11 and R 12 are as defined above.
  • Non-limiting examples are mono- or di-alkyl-, alkenyl-, alkynyl- and arylamino or mixed amino.
  • acyl derivative represents a radical derived from carboxylic acid and thus is defined as including groups of the formula R 11 —CO—, wherein R 11 is as defined above and may also be hydrogen.
  • Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl, heptanedioyl, cyclohexanecarbonyl, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxalyl, ethoxalyl, cysteinyl, oxamoyl.
  • sulfonyl derivative as used herein, is defined as including a group of the formula —SO 2 —R 11 , wherein R 11 is as defined above except for “sulfonyl derivative”.
  • Non-limiting examples are alkylsulfonyl, alkenylsulfonyl, alkynylsulfonyl and arylsulfonyl.
  • sulfinyl derivative as used herein, is defined as including a group of the formula —SO—R 11 , wherein R 11 is as defined above except for “sulfinyl derivative”.
  • Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl and arylsulfinyl.
  • alkyl is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and containing 1-20 carbon atoms, preferably 1-6 carbon atoms for non-cyclic alkyl and 3-6 carbon atoms for cycloalkyl (in these two preferred cases, unless otherwise specified, “lower alkyl”).
  • Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cycloalkyl, sulfonic acid, sulfonamide, thio derivative, oxyester, oxyamido, heterocycle, vinyl, C1-5-alkoxy, C6-10-aryloxy and C6-10-aryl.
  • substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido
  • Preferred alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, and 2,2,2-trimethylethyl each optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro and cyano, such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • alkyl, alkenyl and alkynyl represent straight- or branched chains, C1-12, preferably C1-4-alkylene or C2-12-, preferably C2-4-alkenylene or -alkynylene moieties respectively.
  • branched derivatives are conventionally qualified by prefixes such as “n”, “sec”, “iso” and the like (e.g., “n-propyl”, “sec-butyl”) are in the n-form unless otherwise stated.
  • aryl as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of 1-3 rings and containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfinyl, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, alkylthio, oxyester, oxyamido, aryl, C1-6-alkoxy, C6-10-aryloxy, C1-6-alkyl, C1-6-haloalkyl.
  • Aryl radicals are preferably monocyclic containing 6-10 carbon atoms.
  • Preferred aryl groups are phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, nitro, amino, azido, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkyl, C1-6-haloalkyl and phenyl.
  • halogen includes an atom of Cl, Br, F, I.
  • hydroxy represents a group of the formula —OH.
  • thiol represents a group of the formula —SH.
  • cyano represents a group of the formula —CN.
  • nitro represents a group of the formula —NO 2 .
  • nitrooxy represents a group of the formula —ONO 2 .
  • amino represents a group of the formula —NH 2 .
  • azido represents a group of the formula —N 3 .
  • sulfonic acid represents a group of the formula —SO 3 H.
  • sulfonamide represents a group of the formula —SO 2 NH 2 .
  • esters as used herein is defined as including a group of formula —COO—R 11 wherein R 11 is as defined above except oxy derivative, thio derivative or amino derivative.
  • ether is defined as including a group selected from C1-50-straight or branched alkyl, or C2-50-straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.
  • heterocycle as used herein is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl.
  • Non-limiting examples of aromatic heterocycles are pyridyl, furyl, pyrrolyl, thienyl, isothiazolyl, imidazolyl, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, pyrazolyl, indolyl, indolizinyl, purinyl, isoindolyl, carbazolyl, thiazolyl, 1,2,4-thiadiazolyl, thieno (2,3-b) furanyl, furopyranyl, benzofuranyl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl, benzothiazolyl, or benzox
  • Non-limiting examples of non aromatic heterocycles are tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i.e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) or the same which can optionally be substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups.
  • heterocycle also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1)heptanyl, 7- oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1)octanyl.
  • alkylene group such as quinuclidinyl, 7-azabicyclo (2.2.1)heptanyl, 7- oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1)octanyl.
  • X are —COO R 7 or —CONR 5 R 6 , wherein R 5 , R 6 and R 1 are preferably hydrogen, C1-4-alkyl, phenyl or alkylphenyl.
  • X is carboxy or —CONR 5 R 6 , wherein R 5 and R 6 are preferably hydrogen, C1-4-alkyl, phenyl or alkylphenyl, especially —CONH 2 .
  • a 1 and A 2 are each oxygen.
  • R 1 is hydrogen, alkyl, especially C1-12 alkyl, particularly lower alkyl or aryl especially phenyl.
  • R 1 groups are methyl, ethyl, propyl, isopropyl, butyl, iso- or ter-butyl.
  • 2,2,2-trimethylethyl each optionally attached via a methylene bridge or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • R 1 as ethyl is especially preferred.
  • R 2 and R 2a are independently hydrogen, halogen or alkyl, especially lower alkyl.
  • R 1 and R 2a groups are independently hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl. 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • R 2 and R 2a are hydrogen.
  • R 1a , R 4 and R 4a are independently hydrogen, alkyl, especially methyl or ethyl or aryl especially phenyl or aralkyl, especially benzyl.
  • R 3a , R 4 and R 4a groups are independently hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl. 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • R 4 and R 4a are hydrogen.
  • R 3a is particularly hydrogen or alkyl, especially lower alkyl and is most preferably hydrogen.
  • R 3 is hydrogen, C1-12-alkyl, especially C1-6-alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato or alkoxy and attached to the ring either directly or via a thio, sulfinyl, sulfonyl, carbonyl or oxycarbonyl group and optionally, a C1-4-alkylene bridge, particularly methylene; C2-6-alkenyl or -alkynyl, especially C2-3-alkenyl or -alkynyl each optionally substituted by one or more halogens; azido; cyano; amido: carboxy; triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl,
  • R 3 is C1-6-alkyl optionally substituted by one or more substituents selected from halogen, thiocyanato, azido, alkoxy, alkylthio, phenylsulfonyl; nitrooxy; C2-3-alkenyl or -alkynyl each optionally substituted by one or more halogens or by acetyl; tetrazolyl, pyridyl, furyl, pyrrolyl, thiazolyl or thienyl; or phenyl or phenylalkyl each optionally substituted by one or more substituents selected from halogen, C1-6-alkyl, C1-6 haloalkyl, C1-6-alkoxy, amino, azido, phenyl and nitro and each attached to the ring either directly or via a sulfonyloxy and optionally additionally a C1-4-alkylene bridge, particularly methylene.
  • substituents selected from halogen,
  • R 3 groups are hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • R 3 is especially C1-4-alkyl optionally substituted by one or more substituents selected from halogen, thiocyanato or azido; C2-5-alkenyl or -alkynyl, each optionally substituted by one or more halogens; thienyl; or phenyl optionally substituted by one or more substituents selected from halogen, C1-6-alkyl, C1-6 haloalkyl or azido.
  • R 3 groups are C1-6 alkyl and C2-6 haloalkenyl.
  • R 5 and R 6 are independently hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl, especially hydrogen or methyl.
  • R 5 and R 6 are hydrogen.
  • R 7 is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or tert-butyl, 2,2,2-trimethylethyl, methoxy, ethoxy, phenyl, benzyl or the same substituted by at least one halogen atom such as trifluoromethyl, chlorophenyl.
  • R 7 is hydrogen, methyl or ethyl especially hydrogen.
  • R 8 is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl. 2,2,2-trimethylethyl, phenyl, benzyl or the same substituted by at least one halogen atom such as trifluoromethyl, chlorobenzyl.
  • R S is hydrogen or methyl.
  • Combinations of one or more of these preferred compound groups are especially preferred.
  • a particular group of compounds of formula I comprises those wherein,
  • a 2 is oxygen
  • X is —CONR 5 R 6 or —COOR 7 or —CO—R 8 or CN;
  • R 1 is hydrogen or alkyl, aryl, halogen, hydroxy, amino, nitro, cyano;
  • R 2 , R 3 , R 4 are the same or different and each is independently hydrogen or halogen, hydroxy, amino, nitro, cyano, acyl, acyloxy, a sulfonyl derivative, a sulfinyl derivative, an amino derivative, carboxy, ester, ether, amido, sulfonic acid, sulfonamide, alkoxycarbonyl, a thio derivative, alkyl, alkoxy, oxyester, oxyamido, aryl, an oxy derivative, heterocycle, vinyl and R 3 may additionally represent C2-5 alkenyl, C2-5 alkynyl or azido each optionally substituted by one or more halogen, cyano, thiocyano, azido, cyclopropyl, acyl and/or phenyl; or phenylsulfonyloxy whereby any phenyl moiety may be substituted by one or more halogen, alkyl, acy
  • R 2a , R 3a and R 4a are hydrogen
  • R 4 , R 6 , R 7 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or oxy derivative;
  • R 8 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle, alkylthio or thio derivative.
  • R 1 is preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl; most preferably methyl, ethyl or n-propyl.
  • R 2 and R 4 are preferably independently hydrogen or halogen or methyl, ethyl, propyl, isopropyl, butyl, isobutyl; and, most preferably, are each hydrogen.
  • R 3 is preferably C1-5 alkyl, C2-5 alkenyl, C2-C5 alkynyl, cyclopropyl, azido, each optionally substituted by one or more halogen, cyano, thiocyano, azido, alkylthio, cyclopropyl, acyl and/or phenyl; phenyl; phenylsulfonyl; phenylsulfonyloxy, tetrazole, thiazole, thienyl, furyl, pyrrole, pyridine, whereby any phenyl moiety may be substituted by one or more halogen, alkyl, haloalkyl, alkoxy, nitro, amino, and/or phenyl; most preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl.
  • X is preferably —COOH or —COOMe or —COOEt or —CONH 2 ; most preferably —CONH 2 .
  • a further particular group of compounds of formula I comprises those wherein,
  • X is —CA 1 NH 2 ,—CA 1 NHCH 3 or —CA 1 N(CH 3 ) 2 ;
  • R 1 is alkyl or phenyl
  • R 3 is alkyl, alkenyl, alkynyl, cyano, isothiocyanato, ether, carboxyl, amido, aryl, heterocycle; or
  • R 3 is CH 2 R 10 wherein R 10 is hydrogen, cycloalkyl, oxyester, oxyalkylsulfonyl, oxyarylsulfonyl, aminoalkylsulfonyl, aminoarylsulfonyl, nitrooxy, cyano, isothiocyanato, azido, alkylthio, arylthio, alkylsulfinyl, alkylsulfonyl, heterocycle, aryloxy, alkoxy or trifluoroethyl;
  • R 3a is hydrogen, alkyl or aryl (especially with the proviso that when R 3a is hydrogen, R 1 other than methyl);
  • R 3 R 3a form a cycloalkyl
  • R 2 , R 2a , R 4 and R 4a are each hydrogen.
  • R 1 is preferably alkyl especially C1-12- more particularly C1-6-alkyl and is most preferably ethyl;
  • R 2 , R 2a , R 3a and R 4a are preferably hydrogen
  • R 3 is preferably selected from hydrogen; C1-12-alkyl, especially C1-6-alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato or alkoxy and attached to the ring either directly or via a thio, sulfinyl, sulfonyl, carbonyl or oxycarbonyl group and optionally additionally a C1-4-alkylene bridge, particularly methylene; C2-6-alkenyl or -alkynyl, especially C2-3-alkenyl or -alkynyl, each optionally substituted by one or more halogens; azido; cyano; amido; carboxy; triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidiny
  • R 3a is preferably hydrogen or C1-4-alkyl
  • R 4 and R 4a are preferably, independently hydrogen, C1-4-alkyl, phenyl or benzyl.
  • a further group of compounds of formula I comprises those in racemic form wherein, when X is —CONR 5 R 6 and R 1 is hydrogen, methyl, ethyl or propyl, then substitution on the pyrrolidine ring is other than mono-, di-, or tri-methyl or mono-ethyl.
  • a further group of compound of formula I comprises those in racemic form wherein, when X is —CONR 5 R 6 and R 1 is hydrogen or C1-6-alkyl, C2-6-alkenyl or -alkynyl or cycloalkyl, each unsubstituted, then substitution in the ring is other than by alkyl, alkenyl or alkynyl, each unsubstituted.
  • a further particular group of compounds of formula I comprises those wherein,
  • X is —CA 1 NH 2 ;
  • R 1 is H
  • R 3 is azidomethyl, iodomethyl, ethyl optionally substituted by 1 to 5 halogen atoms, n-propyl optionally substituted by 1 to 5 halogen atoms, vinyl optionally substituted by one or two methyl, and/or 1 to 3 halogen atoms, acetylene optionally substituted by C1-4-alkyl, phenyl or halogen;
  • R 3a is hydrogen or halogen, preferably fluorine
  • R 2 , R 2a , R 4 and R 4a are each hydrogen;
  • a further particular group of compounds of formula I comprises those wherein,
  • X is —CA 1 NH 2 ;
  • R 1 is H
  • R 3 is C1-6-alkyl, C2-6-alkenyl or C2-6-alkynyl optionally substituted by azido, oxynitro, 1 to 6 halogen atoms;
  • R 3a is hydrogen or halogen, preferably fluorine
  • R 1 , R 2 , R 4 and R 4a are each hydrogen; as their racemates or in enantiomerically enriched form, preferably the pure enantiomers.
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of:
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of:
  • a 1 represents an oxygen or a sulfur atom
  • X is —CONR 17 R 8 , —COOR 9 , —CO—R 10 or CN;
  • R 1 when existent, R 2 , R 3 , R 4 and R 5 are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative,
  • R 1 when existent, R 2 , R 3 , R 4 or R 5 is not hydrogen;
  • R 6 is hydrogen, alkyl, aryl or —CH 2 —R 6 wherein R is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
  • R 7 , R 8 and R 9 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or an oxy derivative
  • R 10 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative
  • At least one substituent R 1 to R 5 is different from hydrogen.
  • Some non-substituted compounds are referred to in U.S. Pat. Nos. 5,468,733 and 5,516,759.
  • U.S. Pat. No. 5,468,733 refers to non-ring substituted 2-oxo-1-pyrrolidinyl and 2-oxo-1-piperidinyl derivatives as inhibitors of the oncogene Ras protein.
  • these compounds block the ability of Ras to transform normal cells to cancer cells, and therefore can be included in several chemotherapeutic compositions for treating cancer.
  • U.S. Pat. No. 5,516,759 refers to non-ring substituted 2-oxo-1-pyrrolidinyl, 2-oxo-1-piperidinyl and azepanyl derivatives present at the N-terminus of dodecapeptides possessing LHRH (luteinizing hormone-releasing hormone) antagonistic activity.
  • LHRH antagonists are useful in the treatment of a variety of conditions in which suppression of sex steroids plays a key role including contraception, delay of puberty, treatment of benign prostatic hyperplasia a. o.
  • R 11 and R 12 are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfinyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.
  • oxy derivative is defined as including —O—R 11 groups wherein R 11 is as defined above except for “oxy derivative”.
  • Non-limiting examples are alkoxy, alkenyloxy, alkynyloxy, acyloxy, oxyester, oxyamido, alkylsulfonyloxy, alkylsulfinyloxy, arylsulfonyloxy, arylsulfinyloxy, aryloxy, aralkoxy or heterocyclooxy such as pentyloxy, allyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methylenedioxy, carbonate.
  • thio derivative is defined as including —S—R 11 groups wherein R 11 is as defined above except for “thio derivative”.
  • Non-limiting examples are alkylthio, alkenylthio, alkynylthio and arylthio.
  • amino derivative is defined as including —NHR 11 or —NR 11 R 12 groups wherein R 11 and R 12 are as defined above.
  • Non-limiting examples are mono- or di-alkyl-, alkenyl-, alkynyl- and arylamino or mixed amino.
  • acyl derivative represents a radical derived from carboxylic acid and thus is defined as including groups of the formula R—CO—, wherein R 11 is as defined above and may also be hydrogen. Preferred are acyl derivatives of formula —COR 11 wherein R 11 is selected from hydrogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkenyl, heterocyle and aryl.
  • Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl, heptanedioyl, cyclohexanecarbonyl, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxalyl, ethoxalyl, cysteinyl, oxamoyl.
  • sulfonyl derivative is defined as including a group of the formula —SO 2 —R 11 , wherein R 11 is as defined above except for “sulfonyl derivative”.
  • Non-limiting examples are alkylsulfonyl, alkenylsulfonyl, alkynylsulfonyl and arylsulfonyl.
  • sulfinyl derivative is defined as including a group of the formula —SO—R 11 , wherein R 11 is as defined above except for “sulfinyl derivative”.
  • Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl and arylsulfinyl.
  • alkyl is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and generally containing 1-20 carbon atoms, most often 1 to 12 carbon atoms, preferably 1-7 carbon atoms for non-cyclic alkyl and 3-7 carbon atoms for cycloalkyl (in these two preferred cases, unless otherwise specified, “lower alkyl”), each optionally substituted by, preferably 1 to 5, substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cycloalkyl, sulfonic acid, sulfonamide, thio derivative, alkylthio, oxyester, oxyamido, hetero
  • alkyl groups containing 1 to 7 carbon atoms each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl.
  • substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl.
  • C1-4 alkyl and C3-7 cycloalkyl each optionally substituted by one or more hydroxy, halogen, lower alkyl or/and azido.
  • alkyl groups are hydroxymethyl, propyl, butyl, 2,2,2-trifluoroethyl, 2-bromo-2,2-difluoroethyl, 2-chloro-2,2-difluoroethyl, 3,3,3-trifluoropropyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2,2-difluoropropyl, 2-iodo-2,2-difluoroethyl.
  • lower alkyl refers to C 1 to C 7 saturated straight, branched or cyclic hydrocarbon.
  • Non limiting examples are methyl, ethyl, propyl, isopropyl, butyl, tertiobutyl, pentyl, cyclopropyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methypentyl, 2,2-dimethylbutyl, optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • lower alkyl is methyl.
  • alkenyl is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, thiocyanato, azido, alkylthio, cycloalkyl, acyl, nitro, cyano, aryl and heterocycle.
  • Most preferred is vinyl, optionally substituted by one or more halogen or/and lower alkyl, and especially 2,2-difluorovinyl, 2,2-dibromovinyl and 2,2-dichloroviny
  • Preferred alkynyl groups are C2-12 alkynyl, especially C2-6 alkynyl, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, acyl, aryl such as phenyl and alkyl, preferably cycloalkyl.
  • ethynyl, propynyl and butynyl are preferred, optionally substituted by lower alkyl or/and halogen, and especially 1-propynyl, cyclopropylethynyl, 3-methyl-1-butynyl and 3,3,3-trifluoro-1-propynyl.
  • alkyl, alkenyl and alkynyl represent straight- or branched chains, C1-12, preferably C1-4-alkylene or C2-12-, preferably C2-4-alkenylene or -alkynylene moieties respectively.
  • branched derivatives are conventionally qualified by prefixes such as “n”, “sec”, “iso” and the like (e.g. “n-propyl”, “sec-butyl”) are in the n-form unless otherwise stated.
  • aryl is defined as including an organic radical derived from an aromatic hydrocarbon consisting of at least one ring, most often 1 to 3 rings and generally containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl, each optionally substituted by one or more substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfinyl, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, C1-6-alkylthio, oxyester, oxyamido, aryl, C1-6-alkoxy, C6-10-aryloxy, C1-6-alkyl, C1-6-haloalkyl.
  • Aryl radicals are preferably monocyclic or bicyclic containing 6-10 carbon atoms.
  • Preferred aryl groups are phenyl and naphthyl each optionally substituted by one or more substituents independently selected from halogen, nitro, amino, azido, C1-6-alkoxy, C1-6-alkyl, C1-6-haloalkyl, sulfonyl and phenyl.
  • Preferred aryl is phenyl, optionally substituted by one or more halogen, lower alkyl, azido or nitro, such as 3-chlorophenyl and 3-azidophenyl.
  • halogen includes an atom of Cl, Br, F, I.
  • hydroxy represents a group of the formula —OH.
  • thiol represents a group of the formula —SH.
  • cyano represents a group of the formula —CN.
  • nitro represents a group of the formula —NO 2 .
  • nitrooxy represents a group of the formula —ONO 2 .
  • amino represents a group of the formula —NH 2 .
  • azido represents a group of the formula —N 3 .
  • sulfonic acid represents a group of the formula —SO 3 H.
  • sulfonamide represents a group of the formula —SO 2 NH 2 .
  • esters as used herein, is defined as including a group of formula —COO—R 11 wherein R 11 is as defined above except oxy derivative, thio derivative or amino derivative.
  • esters of formula —COOR 11 wherein R 11 is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl and aryl. Most preferred are esters where R 11 is a lower alkyl, especially methyl.
  • ether is defined as including a group selected from C1-50-straight or branched alkyl, or C2-50-straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.
  • heterocycle is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl, and optionally being substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups.
  • heterocycles are pyridyl, furyl, pyrrolyl, thienyl, isothiazolyl, triazolyl, imidazolyl, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, pyrazolyl, indolyl, indolizinyl, purinyl, isoindolyl, carbazolyl, thiazolyl, 1,2,4-thiadiazolyl, thiomorpholinyl, thieno (2,3-b) furanyl, furopyranyl, benzofuranyl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl,
  • 5-azauracilyl triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i.e.
  • heterocycle also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1) heptanyl, 7-oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1) octanyl.
  • alkylene group such as quinuclidinyl, 7-azabicyclo (2.2.1) heptanyl, 7-oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1) octanyl.
  • the heterocycle is preferably selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl.
  • the heterocycle is selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl, and especially from 2- and 3-thienyl, optionally substituted by one or more halogen, acyl such as formyl, cyano and/or lower alkyl, such as methyl.
  • R substituent refers to R 1 , R 2 , R 3 , R 4 or R 5 , independently.
  • a compound of formula I is as defined above wherein n represents 0.
  • the compound of formula I is as defined above wherein n represents 1.
  • R substituent(s) is/are hydrogen.
  • compounds of formula I are as defined above wherein A 1 represents an oxygen atom.
  • a 1 represents an oxygen atom.
  • compounds of formula I are as defined above wherein X is CONR 7 R 8 , especially CONH 2 .
  • X is CONR 7 R 8 , especially CONH 2 .
  • compounds of formula I are as defined above wherein R 6 represents hydrogen, C1-4 alkyl, or a CH 2 —R 6a group wherein R 6a represents a heterocycle. Most preferably R 6 is a C1-4 alkyl, especially ethyl.
  • R 6 is ethyl we refer to 2-(2-oxo (or thioxo)-1-piperidinyl) butanamide or 2-(2-oxo (or thioxo)-1-azepanyl) butanamide derivatives.
  • compounds of formula I are as defined above wherein the carbon atom to which R 6 is attached is of the S configuration.
  • R 6 is ethyl
  • A is oxygen
  • X is CONR 7 R 5 we refer then to (2S)-2-(2-oxo-1-piperidinyl) butanamide or (2S)-2-(2-oxo-1-azepanyl) butanamide derivatives.
  • R 3 and R 4 are the same or different and each is independently hydrogen, halogen, nitro, nitrooxy, cyano, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, acyl derivative, sulfonyl derivative or sulfinyl derivative;
  • R 1 when existent, R 2 when n 0 and R 5 are hydrogen;
  • R 6 is hydrogen, alkyl, aryl or —CH 2 —R 6a wherein R 6a is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
  • C1-12 alkyl each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkyltio, cycloalkyl, acyl, aryl and heterocycle;
  • C2-12 alkenyl each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl;
  • C2-12 alkynyl each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl; acyl derivative of formula —CO—R 11 , wherein R 11 is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, heterocycle and aryl;
  • heterocycle selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl;
  • aryl each optionally substituted by one or more substituents selected from C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 alkylthio, amino, azido, sulfonyl, aryl and nitro.
  • C1-7 alkyl each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl;
  • C2-6 alkenyl each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl;
  • C2-6 alkynyl each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl;
  • heterocycle selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl;
  • phenyl each optionally substituted by one or more substituents selected from C1-6 alkyl, halogen substituted alkyl, halogen, alkoxy, amino, azido, sulfonyl, phenyl and nitro.
  • the compound is as defined above wherein R 1 , R 2 , R 4 and R 5 are hydrogen.
  • the compound is as defined above wherein R 1 , R 2 , R 3 and R 5 are hydrogen.
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of:
  • R 1 is hydrogen
  • R 2 is hydrogen or C1-20-alkyl
  • R 3 is hydrogen, C1-20-alkyl, C 4-8 -cycloalkyl, C5-8-cycloalkenyl, aryl, aromatic or non aromatic heterocycle, C1-20-alkoxy, or a group of formula —W—R 8 , R 3a is hydrogen, C1-20-alkyl or a group of formula:
  • NR 3 R 3a is a group of formula
  • R 4 is hydrogen
  • R 5 is hydrogen; nitro; halogen; azido; cyano; —S—C1-4-alkyl; —SO—C1-4-alkyl; —SO 2 —C1-4-alkyl: —SONH 2 ; C1-20-alkyl unsubstituted or substituted by halogen; or C1-20-alkoxy unsubstituted or substituted by halogen,
  • R 6 is hydrogen, C1-20-alkyl or halogen
  • R 7 is hydrogen, C1-20-alkyl or halogen
  • W is C1-12-alkylene, —NH— or —NHC( ⁇ O)—
  • X is O, S or NH
  • Y is O, S, —CR 12 R 13 —, —NR 14 — or —C(—O)—,
  • R 8 is aryl or heterocycle
  • R 9 , R 10 , R 10a and R 11 are independently selected from hydrogen, C1-4-alkyl, halogen, hydroxy or methoxycarbonyl,
  • R 10 and R 10a together form a C3-6-alkylene
  • R 12 is hydrogen, C1-4-alkyl, halogen or hydroxy
  • R 13 is hydrogen
  • CR 12 R 13 is dioxolanyl
  • R 14 is aryl, heterocycle or a group of formula —V—R 15 ,
  • V is C 1-12 -alkylene
  • R 15 is aryl or heterocycle
  • m 1 to 4.
  • n 0 or 1
  • R 5 , R 6 or R 7 is different from hydrogen when R 2 is hydrogen, R 3 is H or 2,6-diisopropylphenyl, and R 3a is H.
  • the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • R 1 is hydrogen
  • R 2 is hydrogen or C1-20-alkyl
  • R 3 is hydrogen, C1-20-alkyl, C 4-8 -cycloalkyl, C5-8-cycloalkenyl, aryl, aromatic or non aromatic heterocycle, C1-20-alkoxy, or a group of formula —W—R S ,
  • R 3a is hydrogen, C1-20-alkyl or a group of formula:
  • NR 3 R 3a is a group of formula
  • R 4 is hydrogen
  • R 7 is hydrogen; nitro; halogen; C1-20-alkyl unsubstituted or substituted by halogen; or C1-20-alkoxy unsubstituted or substituted by halogen,
  • R 6 is hydrogen, C1-20-alkyl or halogen
  • R 7 is hydrogen, C1-20-alkyl or halogen
  • W is C1-12-alkylene, —NH— or —NHC( ⁇ O)—.
  • X is O, S or NH.
  • Y is O, S, —CR 12 R 13 —, —NR 14 — or —C( ⁇ O)—,
  • R 8 is aryl or heterocycle
  • R 9 , R 10 , R 10a and R 11 are independently selected from hydrogen, C1-4-alkyl, halogen, hydroxy or methoxycarbonyl.
  • R 10 and R 10a together form a C3-6-alkylene
  • R 12 is hydrogen, C1-4-alkyl, halogen or hydroxy
  • R 13 is hydrogen
  • CR 12 R 13 is dioxolanyl
  • R 14 is aryl, heterocycle or a group of formula —V—R 15 ,
  • V is C1-12-alkylene
  • R 15 is aryl or heterocycle
  • n 1 to 4
  • n 0 or 1
  • R 5 , R 6 or R 7 is different from hydrogen when R 2 is hydrogen, R 3 is H or 2,6-diisopropylphenyl, and R 3a is H.
  • alkyl is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and containing 1-20 carbon atoms, preferably 1-6 carbon atoms and more preferably 1-4 carbon atoms for non-cyclic alkyl and 3-8 carbon atoms for cycloalkyl. Alliyl moieties may optionally be substituted by 1 to 5 substituents independently selected from halogen, hydroxy, alkoxy, alkoxycarbonyl, ester or alkylamino.
  • Preferred alkyl groups are methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, n-butyl, 2-fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2,2-dimethylpropyl, 1-(hydroxymethyl) propyl, 3,3,3-trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2-oxoethyl and 3-(dimethylamino) propyl.
  • cycloalkyl refers to a monovalent group of 3 to 18 carbon atoms, preferably 4-8 carbon atoms, derived from a saturated cyclic or polycyclic hydrocarbon which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • Preferred cycloalkyl group is cycloheptyl.
  • alkylene represents a divalent alkyl group, having straight or branched moieties, containing 1-12 carbon atoms, preferably 1-6 carbon atoms, and being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • Preferred alkylene groups are methylene, ethylene, hydroxyethylene, trimethylene or propylene.
  • cycloalkenyl is defined as a cyclic unsaturated hydrocarbon radical having at least one double bond, containing 4-20 carbon atoms, preferably 5-8 carbon atoms, and being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • Preferred cycloalkenyl group is 6-(hydroxymethyl)cyclohex-3-en-1-yl.
  • aryl is defined as including an organic radical derived from an aromatic hydrocarbon consisting of 1-3 rings and containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, nitro, C1-6-alkyl, C1-6-alkoxy, C1-6-alkylsulfonyl, trifluoromethylthio or pyridinylalkyl.
  • Aryl radicals are preferably phenyl radicals.
  • Preferred aryl groups are phenyl, 3-hydroxyphenyl, 3-fluorophenyl, 3-methylphenyl.
  • halogen includes an atom of Cl, Br, F, I.
  • nitro represents a group of the formula —NO 2 .
  • hydroxy represents a group of the formula —OH.
  • alkoxy represents a group of formula —OR b wherein R b is an alkyl group, as defined above.
  • esters represents a group of formula —COOR c wherein R c is an alkyl group or an aryl group, as defined above.
  • alkoxycarbonyl represents a group of formula —COOR d wherein R d is an alkyl group, as defined above.
  • amino represents a group of the formula —NH 2 .
  • alkylamino represents a group of formula —NHR e or —NR e R f wherein R e and R f are alkyl group as defined above.
  • alkylsulfonyl as used herein is defined as representing a group of formula —SO 2 —R g , wherein R f is C1-4-alkyl.
  • heterocycle as used herein is defined as including an aromatic or non aromatic cycloalkyl or cycloalkenyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl.
  • Non-limiting examples of aromatic heterocycles are pyrazolyl, furyl, imidazolyl, triazolyl, oxazolyl, pyridinyl, pyrrolyl, thienyl, isothiazolyl, benzimidazolyl, tetrazolyl, isooxazolyl, oxazolyl, thiazolyl, 1,2,4-thiadiazolyl, oxadiazole, pyridazinyl, pyrimidinyl, pyrazinyl, isoindolyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, quinazolinyl, quinolizinyl, naphthyridinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, indolyl, indolizinyl, puriny
  • Non-limiting examples of non aromatic heterocycles are tetrahydrofuranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholinyl, thiomorpholinyl, pyrrolidinyl, thiazolidinyl, indolinyl, tetrahydrobenzazocinyl, dihydroisochromenyl, tetrahydropyranyl, oxooctahydroquinolinyl, dioxolanyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, 8-thiabicyclo [3.2.1]cyclooctanyl, 1,4-dithiepanyl, tetrahydro-2H-thiopyranyl, azepanyl and azocanyl, optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy,
  • More preferred non aromatic heterocycles are tetrahydrofuranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholinyl, thiomorpholinyl, pyrrolidinyl, thiazolidinyl, indolinyl, tetrahydro-1-benzazocin-1 (2H)-yl, 3,4-dihydro-1H-isochromen-1-yl, tetrahydropyranyl, oxooctahydroquinolinyl and dioxolanyl.
  • heterocycle also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cycloalkyl ring, a cycloalkenyl ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1)heptanyl, 7-oxabicyclo (2.2.1)heptanyl and 8-azabicyclo (3.2.1)octanyl.
  • pyridinylalkyl represents a group of formula —R h -pyridinyl in which R h is C1-4-alkylene.
  • azido represents a group of the formula —N 3 .
  • cyano as used herein, represents a group of the formula —CN.
  • R 2 is hydrogen or C1-4-alkyl.
  • R 2 is hydrogen, methyl or ethyl. More preferably, R 2 is hydrogen or methyl.
  • R 3 is hydrogen; C1-6-alkyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, hydroxy, alkoxy, alkoxycarbonyl or alkylamino; C5-7-cycloalkyl; (hydroxymethyl)cyclohexenyl; phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl, trifluoromethylthio or pyridinylalkyl; pyridinyl unsubstituted or substituted by methoxy; triazolyl; C1-4-alkoxy; or a group of formula —W—R 8 wherein:
  • W is C1-4-alkylene unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl or alkoxy; —NH—; or —NHC( ⁇ O)—; and
  • R 8 is phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl or trifluoromethylthio; furyl unsubstituted or substituted by methyl; pyrazolyl; pyridinyl; morpholinyl; tetrahydrobenzazocinyl; piperidinyl unsubstituted or substituted by methyl; dihydroisochromenyl or dihydroimidazolyl.
  • R 3 is hydrogen, n-butyl, cycloheptyl, 2-fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2,2-dimethylpropyl, 1-(hydroxymethyl) propyl, 3,3,3-trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2-oxoethyl.
  • R 3a is hydrogen, C1-4-alkyl or a group of formula
  • R 3 is hydrogen, methyl or tetrahydrofuran-2-ylmethyl. More preferably. R 3a is hydrogen.
  • NR 3 R 3a is piperidinyl unsubstituted or substituted by hydroxy; thiomorpholinyl; thiazolidinyl unsubstituted or substituted by C1-4-alkoxycarbonyl; 2,5-dihydro-1H-pyrrol-1-yl; 1,4-dioxa-8-azaspiro[4.5]dec-8-yl; 4-oxooctahydro-1(2H)-quinolinyl; or a group of formula
  • R 14 is pyridinyl; phenyl unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl; or a group of formula —V—R 15 wherein V is unsubstituted C1-4-alkylene and R 15 is phenyl or morpholinyl.
  • NR 3 R 3a is 4-pyridin-2-ylpiperazin-1-yl, 4-(3-methylphenyl)piperazin-1-yl, 4-(4-hydroxyphenyl)piperazin-1-yl, 4-(2-phenylethyl)piperazin-1-yl, 4-(2-morpholin-4-ylethyl)piperazin-1-yl, 3-hydroxypiperidin-1-yl, thiomorpholin-4-yl, 4-methoxycarbonyl-1,3-thiazolidin-3-yl, 2,5-dihydro-1H-pyrrol-1-yl, 1,4-dioxa-8-azaspiro[4.5]dec-8-yl or 4-oxooctahydro-1(2H)-quinolinyl.
  • R 5 is hydrogen, nitro, halogen, C1-4-alkyl, unsubstituted or substituted by halogen, or C1-4-alkoxy unsubstituted or substituted by halogen.
  • R 5 is hydrogen, methyl, ethyl, trifluoromethyl, trifluoromethoxy, n-propyl, isopropyl, nitro, or halogen. More preferably, R 5 is halogen or trifluoromethyl.
  • R 6 is hydrogen, C1-6-alkyl or halogen.
  • R 6 is hydrogen, methyl or Cl. More preferably. R 6 is hydrogen.
  • R 7 is hydrogen, methyl or halogen.
  • R 7 is hydrogen, methyl, Br, F or Cl. More preferably, R 7 is hydrogen, Br or F.
  • Combinations of one or more of these preferred compound groups are especially preferred.
  • the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • R 1 is hydrogen
  • R 2 is hydrogen or C1-4-alkyl
  • R 3 is hydrogen; C1-6-alkyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, hydroxy, alkoxy, alkoxycarbonyl or alkylamino; C5-7-cycloalkyl; (hydroxymethyl)cyclohexenyl; phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl, trifluoromethylthio or pyridinylalkyl; pyridinyl unsubstituted or substituted by methoxy; triazolyl; C1-4-alkoxy; or a group of formula —W—R 8 ,
  • R 3a is hydrogen, C1-4-alkyl or a group of formula
  • R 3a is piperidinyl unsubstituted or substituted by hydroxy; thiomorpholinyl; thiazolidinyl unsubstituted or substituted by C1-4-alkoxycarbonyl: 2,5-dihydro-1H-pyrrol-1-yl; 1,4-dioxa-8-azaspiro[4.5]dec-8-yl; 4-oxooctahydro-1(2H)-quinolinyl; or a group of formula
  • R 4 is hydrogen
  • R 5 is hydrogen; nitro; halogen; C1-4-alkyl, unsubstituted or substituted by halogen; or C1-4-alkoxy unsubstituted or substituted by halogen,
  • R 6 is hydrogen, C1-6-allyl or halogen
  • R 7 is hydrogen, methyl or halogen
  • W is C1-4-alkylene unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl or alkoxy; —NH—; or —NHC( ⁇ O)—,
  • R 8 is phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl or trifluoromethylthio; furyl unsubstituted or substituted by methyl; pyrazolyl; pyridinyl; morpholinyl; tetrahydrobenzazocinyl; piperidinyl unsubstituted or substituted by methyl; dihydroisochromenyl or dihydroimidazolyl,
  • R 14 is pyridinyl; phenyl unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl; or a group of formula —V—R 15
  • V is unsubstituted C1-4-alkylene
  • R 15 is phenyl or morpholinyl
  • n 1 to 4
  • R 1 is H or 2,6-diisopropylphenyl
  • R 3a is H.
  • the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • R 1 is hydrogen
  • R 2 is hydrogen, methyl or ethyl
  • R 3 is hydrogen, n-butyl, cycloheptyl, 2-fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2,2-dimethylpropyl, 1-(hydroxymethyl) propyl, 3,3,3-trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2-oxoethyl, 3-(dimethylamino) propyl, 6-(hydroxymethyl)cyclohex-3-en-1-yl, 3-hydroxyphenyl, 3-fluorophenyl, 3-(2-pyridin-2-ylethyl)phenyl, 3,4-dimethylphenyl, 4-tert-butylphenyl, benzyl, 4-hydroxy-3-methoxybenzyl, 4-methylsulfonylbenzyl, 2-nitrobenzyl, 2-chloro-6-fluorobenzyl, 2-[(trifluoromethyl)thio]benzyl, 2-hydroxy-2-phenylethyl, 2-(3,4
  • R 3a is hydrogen, methyl or tetrahydrofuran-2-ylmethyl, or NR 3 R 3a 4-pyridin-2-ylpiperazin-1-yl.
  • R 4 is hydrogen
  • R 5 is hydrogen, methyl, ethyl, trifluoromethyl, trifluoromethoxy, n-propyl, isopropyl, nitro or halogen.
  • R 6 is hydrogen, methyl or Cl
  • R 7 is hydrogen, methyl, Br, F or Cl
  • R 5 , R 6 or R 7 is different from hydrogen when R 2 is hydrogen, R 3 is H or 2,6-diisopropylphenyl, and R 3a is H.
  • R 2 is hydrogen or methyl.
  • R 3 is hydrogen, R 3a is hydrogen, R 5 is halogen or trifluoromethyl, R 6 is hydrogen and R 7 is hydrogen, Br or F.
  • R 2 when R 2 is C1-20-alkyl, the carbon atom to which R 2 is attached is preferably in the “S”-configuration.
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-iodo-2-oxo-2,3-dihydro-H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5,7-dibromo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-nitro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-methyl-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide, (2R)-2-(5-chloro-2-oxo-2,
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-iodo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5,7-dibromo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; (2S)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; 2-[2-oxo-5-(trifluoromethyl)-2,3-dihydro-1H-indol-1-yl]acetamide and 2-(5-chloro-7-fluoro-2-oxo-2,3-dihydro-1H-indol-1-yl)acet
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-chloro-2-oxo-2,3-dihydro-H-indol-1-yl)acetamide and (2S)-2-(5-chloro-2-oxo-2,3-dihydro-H-indol-1-yl) propanamide.
  • R 1 is hydrogen, C 1-20 alkyl, C 3-8 cycloalkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, guanidine, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl, arylsulfinyl, aryl or heterocycle;
  • R 2 is hydrogen, C 1-20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl:
  • R 3 is hydrogen, C 1-20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;
  • R 2 and R 3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R 4 is hydrogen, C 1-20 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, aryl, azido, alkoxycarbonylamino, arylsulfonyloxy or heterocycle;
  • R 4a is hydrogen or C 1-20 alkyl
  • R 4 and R 4a can form together a C 3-8 cycloalkyl
  • R 5 is hydrogen
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 6 is hydrogen or C1-20 alkyl
  • R 7 is hydrogen
  • R 6 and R 7 are linked together to form a C 3-6 cycloalkyl
  • R 8 is hydrogen, halogen, nitro, cyano, C 1-20 alkyl or alkoxy:
  • R 9 is hydrogen, C1-20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
  • R 10 is hydrogen, C1-20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
  • R 11 is hydrogen, halogen, nitro, cyano, C1-20 alkyl or alkoxy;
  • R 12 is hydrogen or halogen
  • R 13 is hydrogen, nitro, halogen, heterocycle, amino, aryl, C1-20 alkyl unsubstituted or substituted by halogen, or alkoxy unsubstituted or substituted by halogen;
  • R 14 is hydrogen, C1-20 alkyl or halogen
  • R 15 is hydrogen, C1-20 alkyl or halogen
  • R 4 is different from hydrogen when represents a group of formula
  • the asterisk * indicates the point of attachment of the substituents.
  • the compounds have the formula I, their tautomers, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • R 1 is hydrogen, C 1-20 alkyl, C 3-8 cycloalkyl, halogen, hydroxy, ester, amido, cyano, nitro, amino, guanidine, alkylthio, alkylsulfonyl, alkylsulfinyl, aryl or heterocycle;
  • R 2 is hydrogen, C1-20 alkyl, halogen, cyano, ester, carbamate or amido;
  • R 3 is hydrogen, cyano, C 1-20 alkyl, halogen or ester; or R 2 and R 3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R 4 is hydrogen, C1-20 alkyl, C 2-12 alkenyl or aryl
  • R 4a is hydrogen
  • R 5 is hydrogen
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 6 is hydrogen or C1-20 alkyl
  • R 7 is hydrogen; or R 6 and R 7 are linked together to form a C 3-6 cycloalkyl:
  • R 8 is hydrogen
  • R 9 is hydrogen, C 1-20 alkyl, halogen or alkoxy
  • R 10 is hydrogen, C1-20 alkyl, halogen or cyano
  • R 11 is hydrogen
  • R 12 is hydrogen or halogen
  • R 13 is hydrogen, halogen, heterocycle or C 1-20 alkyl
  • R 14 is hydrogen:
  • R 15 is hydrogen
  • alkyl represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched or cyclic or combinations thereof and containing 1-20 carbon atoms, preferably 1-10 carbon atoms, more pre preferred alkyl groups have 1-3 carbon atoms.
  • Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, cyano, azido, aryloxy, alkoxy, alkylthio, alkanoylamino, arylcarbonylamino, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl or aryl.
  • alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, n-heptyl, 2,4,4-trimethylpentyl, n-decyl, chloromethyl, trifluoromethyl, 2-bromo-2,2-difluoroethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino)methyl, (propionylamino)methyl.
  • alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, 2,4,4-trimethylpentyl, chloromethyl, trifluoromethyl. 2,2,2-trifluoroethyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino)methyl, (propionylamino)methyl. (benzoylamino)methyl or 2-(methylthio)ethyl.
  • More preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, azidomethyl or trifluoromethyl. Most preferred alkyl groups are methyl or n-propyl.
  • cycloalkyl represents a monovalent group of 3 to 8 carbon atoms, usually 3-6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • Preferred cycloalkyl groups are cyclopropyl and cyclohexyl.
  • alkenyl represents straight, branched or cyclic unsaturated hydrocarbon radicals or combinations thereof having at least one carbon-carbon double bond, containing 2-12 carbon atoms, preferably usually 2-4 carbon atoms.
  • Alkenyl groups are being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • an alkenyl group is ethenyl (vinyl) optionally substituted by 1 to 3 halogens.
  • Preferred alkenyl group in the present case, is 2,2-difluorovinyl.
  • alkynyl represents straight, branched or cyclic hydrocarbon radicals or combinations thereof containing at least one carbon-carbon triple bond, containing 2-12 carbon atoms, preferably 2-6 carbon atoms, and being optionally substituted by any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • an alkynyl group is a halogenoalkynyl group (haloalkynyl group).
  • Groups qualified by prefixes such as “s”, “i”, “t” and the like are branched derivatives.
  • aryl as used herein, is defined as phenyl optionally substituted by 1 to 4 substituents independently selected from halogen, cyano, alkoxy, alkylthio, C 1-3 alkyl or azido, preferably halogen or azido.
  • aryl groups in the present case are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl. 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl.
  • aryl groups are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • Most preferred aryl groups are phenyl.
  • 3-chlorophenyl 3-fluorophenyl, 3,5-difluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • heterocycle is defined as including an aromatic or non aromatic cycloalkyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure.
  • Heterocyclic ring moieties can be optionally substituted by alkyl groups or halogens and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl.
  • heterocycles are 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-tetrahydrofuranyl, 1H-pyrrol-2-yl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrazol-2-yl, 1H-pyrazol-3-yl, 4-chloro-1-methyl-1H-pyrazol-3-yl, 5-chloro-1,3-dimethyl-1H-pyrazol-4-yl, 1,2,3-thiadiazol-4-yl, 3,5-dimethyl-4-isothiazyl, 1H-imidazol-2-yl, 1-methyl-1H-imidazol-2-yl, 4-methyl-1H-imidazol-5-yl, or 2-methyl-1,3-thiazol-4-yl.
  • Preferred heterocycles are 1H-imidazol-2-yl, 1,2,3-thiadiazol-4-yl, 1H-pyrazol-3-yl.
  • halogen includes an atom of chlorine, bromine, fluorine, iodine. Usually halogens are chlorine, bromine and fluorine. Preferred halogens are fluorine, bromine and chlorine.
  • hydroxy represents a group of formula —OH.
  • alkoxy represents a group of formula —OR a
  • R a is an alkyl group, as defined above.
  • Preferred alkoxy group is methoxy.
  • aryloxy represents a group of formula —OR b wherein R b is an aryl group, as defined above. Preferred aryloxy group is phenoxy.
  • ester represents a group of formula —COOR c wherein R c is an alkyl group or aryl group, as defined above. Preferred ester group is methoxycarbonyl.
  • amino represents a group of formula —NH 2 .
  • amino acid represents an alkylamino or an arylamino group, wherein the terms “alkyl” and “aryl” are defined as above.
  • cyano represents a group of formula —CN.
  • nitro represents a group of formula —NO,.
  • azido represents a group of formula —N 3 .
  • guanidine represents a group of formula —NHC( ⁇ NH)NH 2 .
  • alkylthio represents a group of formula —SR d wherein R d is an alkyl group, as defined above.
  • Preferred alkylthio group is methylthio.
  • alkylsulfonyl represents a group of formula —S( ⁇ O) 2 R wherein R e is an alkyl group, as defined above.
  • R e is an alkyl group, as defined above.
  • Preferred alkylsulfonyl group is methylsulfonyl.
  • alkylsulfinyl represents a group of formula —S( ⁇ O)R f wherein R f is an alkyl group, as defined above.
  • R f is an alkyl group, as defined above.
  • Preferred alkylsulfinyl group is methylsulfinyl.
  • arylthio represents a group of formula —SR g wherein R g is an aryl group, as defined above.
  • arylsulfonyl represents a group of the formula —S( ⁇ O) 2 R h wherein R h is an aryl group, as defined above.
  • arylsulfinyl represents a group of the formula —S( ⁇ O)R 1 wherein R 1 is an aryl group, as defined above.
  • carbamate represents a group of formula —N(H)C(O)OR j , wherein R j is an alkyl or an aryl, as defined above.
  • carbamate groups are (propoxycarbonyl)amino or (benzyloaxycarbonyl)amino.
  • Preferred carbamate group is (benzyloaxycarbonyl)amino.
  • alkanoylamino represents a group of the formula —NHC( ⁇ O)R k wherein R k is an alkyl group, as defined above.
  • (arylcarbonyl)amino represents a group of the formula —NHC( ⁇ O)R m wherein R m is an aryl group, as defined above.
  • Preferred (arylcarbonyl)amino is benzoylamino.
  • R 1 is hydrogen; C 1-10 alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; hydroxy; CL cycloalkyl; halogen; ester; amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl groups; or guanidine.
  • R 1 is hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl: n-butyl: i-butyl; t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; hydroxymethyl; chloromethyl; trifluoromethyl; 2,2,2-trifluoroethyl; cyanomethyl; 2-(methylthio)ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl or 1H-imidazol-2-yl.
  • R 1 is hydrogen; methyl; ethyl: i-propyl; n-propyl; n-butyl; methylthio; nitro; cyano; amino: chloro or 1H-pyrrol-2-yl. Most preferably, R 1 is hydrogen; methyl; methylthio; nitro; cyano; amino or chloro.
  • R 2 is hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino: halogen; ester; cyano; alkyl carbamate; [(N-methoxy-N-methyl)amino]carbonyl.
  • R 2 is hydrogen; methyl; hydroxymethyl; (acetylamino)methyl; (propionylamino)methyl; (benzoylamino)methyl: [(benzyloxy)carbonyl]amino; chloro or cyano. More preferably, R 2 is hydrogen; chloro or cyano.
  • R 3 is hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano.
  • R 3 is hydrogen; hydroxymethyl; chloro; cyano. More preferably, R 3 is hydrogen or cyano. Most preferred R 3 is hydrogen.
  • R 4 is hydrogen; C 1-4 alkyl unsubstituted or substituted by halogens; C 2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens.
  • R 4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl: 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl.
  • R 4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • R 4 is n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • R 4a is hydrogen
  • R 5 is hydrogen
  • R 6 is hydrogen or C 1-10 alkyl unsubstituted or substituted by hydroxy or azido.
  • R 6 is hydrogen or azidomethyl. More preferably R 6 is hydrogen.
  • R 7 is hydrogen
  • R 6 and R 7 are linked to form a cyclopropyl.
  • R 2 and R 3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R 8 is hydrogen
  • R 9 is hydrogen; halogen; C 1-3 alkyl or alkoxy.
  • R 9 is hydrogen; methyl; chloro or methoxy. More preferred R 9 is hydrogen.
  • R 10 is hydrogen; halogen; cyano; C 1-3 alkyl unsubstituted or substituted by halogens; or alkoxy.
  • R 10 is methyl; hydrogen; trifluoromethyl; fluoro; cyano or methoxy. More preferred R 10 is hydrogen; trifluoromethyl; fluoro or cyano.
  • R 11 is hydrogen
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 12 is hydrogen or halogen.
  • R 12 is hydrogen; chloro or fluoro. More preferred R 12 is hydrogen.
  • R 13 is hydrogen; C 1-3 alkyl; halogen or thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl.
  • R 13 is hydrogen; chloro; bromo or methyl.
  • Most preferred R 13 is chloro; bromo or methyl.
  • R 14 is hydrogen.
  • R 15 is hydrogen
  • Combinations of one or more of these preferred compound groups are especially preferred.
  • the compounds of formula I, or pharmaceutically acceptable salts thereof are those wherein
  • R 1 is selected from hydrogen; C 1-10 alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; C 3-6 cycloalkyl: halogen; ester; amido; nitro; cyano: amino; phenyl; alkylthio: alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl group; or guanidine;
  • R 2 is selected from hydrogen; C 1 -4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate or [(N-methoxy-N-methyl)amino]carbonyl.
  • R 3 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy: halogen; ester or cyano;
  • R 4 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by halogens; C 2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens;
  • R 4a is hydrogen
  • R 5 is hydrogen
  • R 6 is selected from hydrogen or C 1-10 alkyl unsubstituted or substituted by hydroxy or azido;
  • R 7 is hydrogen
  • R 6 and R 7 can be linked to form a cyclopropyl
  • R 2 and R 3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R 8 is hydrogen
  • R 9 is selected from hydrogen; halogen; C 1-3 alkyl; alkoxy;
  • R 10 is selected from hydrogen; halogen; cyano or C 1-3 alkyl unsubstituted or substituted by halogens; or alkoxy;
  • R 11 is hydrogen
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 12 is selected from hydrogen or halogen
  • R 13 is selected from hydrogen; C1-3 alkyl; halogen; thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl;
  • R 14 is hydrogen
  • R 15 is hydrogen
  • the compounds of formula I, or pharmaceutically acceptable salt thereof are those wherein
  • R 1 is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl: n-butyl: i-butyl: t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; trifluoromethyl; 2,2,2-trifluoroethyl; hydroxymethyl: chloromethyl; cyanomethyl; 2-(methylthio)ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl; or 1H-imidazol-2-yl;
  • R 2 is selected from hydrogen; methyl; hydroxymethyl; (acetylamino)methyl; (propionylamino)methyl; (benzoylamino)methyl; (benzyloxycarbonyl)amino; chloro; or cyano;
  • R 3 is selected from hydrogen; hydroxymethyl; chloro; cyano;
  • R 2 and R 3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R 8 is hydrogen
  • R 9 is selected from hydrogen; methyl; choro; methoxy;
  • R 10 is selected from methyl; hydrogen; trifluoromethyl; fluoro; cyano; or methoxy;
  • R 11 is hydrogen
  • R 4 is selected from hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl: 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl; or 3-azido-2,4,6-trifluorophenyl.
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 12 is selected from hydrogen; chloro; fluoro;
  • R 13 is selected from hydrogen; chloro; bromo; methyl;
  • R 14 is hydrogen
  • R 6 is selected from hydrogen; azidomethyl
  • R 7 is hydrogen
  • R 6 and R 7 are linked to form a cyclopropyl
  • the compounds of formula I, or pharmaceutically acceptable salt thereof are those wherein
  • R 1 is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; n-butyl; methylthio; nitro; cyano; amino; chloro; or 1H-pyrrol-2-yl;
  • R 2 is selected from hydrogen; chloro; cyano;
  • R 3 is selected from hydrogen; cyano;
  • R 2 and R 3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R 8 is hydrogen
  • R 9 is hydrogen
  • R 10 is selected from hydrogen; trifluoromethyl: fluoro; cyano;
  • R 11 is hydrogen
  • R 4 is selected from hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; or 3-azido-2,4-difluorophenyl;
  • R 4a is hydrogen
  • R 5 is hydrogen
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 12 is hydrogen
  • R 13 is selected from methyl; chloro; bromo;
  • R 14 is hydrogen
  • R 6 is hydrogen
  • R 7 is hydrogen
  • the compounds of formula I, or pharmaceutically acceptable salt thereof are those wherein
  • R 1 is selected from hydrogen; methyl; methylthio; nitro; cyano; amino; chloro;
  • R 2 is selected from hydrogen; chloro; cyano;
  • R 3 is hydrogen
  • R 4 is selected from n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl: 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl;
  • R 4a is hydrogen
  • R 5 is hydrogen
  • R 4 , R 4a and R 5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R 12 is hydrogen
  • R 13 is selected from chloro; bromo; methyl
  • R 14 is hydrogen
  • R 6 is hydrogen
  • R 7 is hydrogen
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: 1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 4-(3-azido-2,4,6-trifluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; ( ⁇ )-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-[(2-ethy
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: 1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one, 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; ( ⁇ )-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-[(2-ethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-isopropyl-1H-imidazol-1
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one: 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; ( ⁇ )-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 4-(2,2-difluorovinyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-on-e; 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: ( ⁇ )-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one.
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen or C 1-4 alkyl
  • R 3 is a group of formula —CHR 5 R 6 or a benzyl group
  • R 4 is C 1-8 is alkyl optionally substituted by alkoxycarbonyl, C 3-6 cycloalkyl, aryl or heterocycle;
  • R 5 is C 2-4 alkyl:
  • R 6 is C 2-4 alkyl, amido or —COOR 7 ;
  • R 7 is C 1-4 alkyl
  • R 4 is C 1-8 alkyl optionally substituted by alkoxycarbonyl.
  • R 3 is a group of formula —CHR 5 R 6 then R 4 is C 1-8 alkyl optionally substituted by C 3-6 cycloalkyl, aryl or heterocycle.
  • alkyl is a group which represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched moieties, or combinations thereof, and containing 1-8 carbon atoms, preferably 1-6 carbon atoms; more preferably alkyl groups have 1-4 carbon atoms.
  • Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl, acyl, aryl or heterocycle.
  • Alkyl moieties may be optionally substituted by a cycloalkyl as defined hereafter.
  • Preferred alkyl groups are methyl, cyanomethyl, ethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-butyl, i-butyl, n-pentyl, 3-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl. 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl.
  • More preferred alkyl groups are methyl, ethyl, cyanomethyl, 2-methoxyethyl, n-propyl, 3-hydroxypropyl, 2-propynyl, n-butyl, 3-pentyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro-2-furyl)methyl.
  • Most preferred alkyl groups are methyl, ethyl, 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • cycloalkyl represents a monovalent group of 3 to 8, preferably 3 to 6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • Preferred cycloalkyl group is cyclohexyl.
  • aryl as used herein, is defined as a phenyl group optionally substituted by 1 to 4 substituents independently selected from halogen, amino, nitro, alkoxy or aminosulfonyl.
  • Preferred aryl groups are phenyl, 2-bromophenyl, 3-bromophenyl, 4-bromophenyl, 3-methoxyphenyl, 3-nitrophenyl, 3-aminophenyl or 4-(aminosulfonyl)phenyl.
  • phenyl represents an aromatic hydrocarbon group of formula —C 6 H 5 .
  • benzyl group represents a group of formula —CH 2 -aryl.
  • Preferred benzyl groups are benzyl, 2-bromobenzyl, 3-bromobenzyl. 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl or 4-(aminosulfonyl)benzyl. More preferred benzyl groups are benzyl. 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl or 3-aminobenzyl. Most preferred alkyl groups are 3-methoxybenzyl or 3-nitrobenzyl.
  • halogen represents an atom of fluorine, chlorine, bromine, or iodine. Preferred halogen is bromine.
  • hydroxy represents a group of formula —OH.
  • cyano represents a group of formula —CN.
  • amino represents a group of formula —NH 2 .
  • ethynyl represents a group of formula —C ⁇ CH.
  • alkoxy represents a group of formula —OR a wherein R a is an alkyl group, as defined above. Preferred alkoxy group is methoxy.
  • nitro represents a group of formula —NO 2 .
  • amido represents a group of formula —C( ⁇ O)NH 2 .
  • acyl represents a group of formula —C( ⁇ O)R b wherein R b is an alkyl group, as defined here above.
  • Preferred acyl group is acetyl (—C( ⁇ O)Me).
  • alkoxycarbonyl (or ester) represents a group of formula —COOR c wherein R c is an alkyl group; with the proviso that R c does not represent an alkyl alpha-substituted by hydroxy.
  • Preferred alkoxycarbonyl group is ethoxycarbonyl.
  • heterocycle represents a 5-membered ring containing one or two heteroatoms selected from O or N.
  • the heterocycle may be substituted by one or two C 1-4 alkyl or nitro.
  • Preferred heterocycles are (3,5-dimethylisoxazol-4-yl) or (5-nitro-2-furyl). Most preferred heterocycle is (5-nitro-2-furyl).
  • R 1 is hydrogen or C 1-6 alkyl.
  • R 1 is hydrogen or C 1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl.
  • R 1 is hydrogen, methyl, cyanomethyl.
  • R 1 is hydrogen, methyl, cyanomethyl.
  • R 1 is hydrogen.
  • R 2 is hydrogen or C 1-4 alkyl. Usually R 2 is hydrogen or unsubstituted C 1-4 alkyl. Preferably R 2 is hydrogen, methyl or n-butyl. More preferably. R 2 is methyl.
  • R 3 is a group of formula —CHR S R 6 or a benzyl group.
  • R 3 is 3-pentyl, 1-(aminocarbonyl)propyl.
  • R 3 is 1-(ethoxycarbonyl)propyl.
  • R 4 is C 1-8 alkyl optionally substituted by alkoxycarbonyl, C 3-6 cycloalkyl, aryl or heterocycle.
  • R 4 is C 1-8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl. 5-nitro-2-furyl or ethoxycarbonyl.
  • R 4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl.
  • R 4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl. Most preferably R 4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • R 5 is C 2-4 alkyl. Usually R 5 is unsubstituted C 2-4 4 alkyl. Preferably R 5 is ethyl.
  • R 6 is C 2-4 alkyl, amido or —COOR 7 .
  • R 6 is unsubstituted C 2-4 alkyl, amido or —COOR 7 .
  • R 6 is ethyl, amido or ethoxycarbonyl. Most preferably R 6 is ethoxycarbonyl.
  • R 7 is C 1-4 alkyl. Usually R 7 is unsubstituted C 1-4 alkyl. Preferably, R 7 is ethyl.
  • the compounds are those having formula I, and their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • R 1 is hydrogen, C 1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl;
  • R 2 is hydrogen or unsubstituted C 1-4 alkyl
  • R 3 is a group of formula —CHR 5 R 6 or a benzyl group
  • R 4 is C 1-8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl, 5-nitro-2-furyl or ethoxycarbonyl;
  • R 5 is unsubstituted C 2-4 alkyl
  • R 6 is unsubstituted C 2-4 alkyl, amido or —COOR 7 ;
  • R 7 is unsubstituted C 1-4 alkyl; with the proviso that when R 1 is hydrogen, R 2 is methyl, R 3 is —CHR 5 R 6 , R 6 is ethoxycarbonyl and R 5 is ethyl, then R 4 is different from n-propyl, i-propyl, n-pentyl, n-heptyl, 3-bromobenzyl, 4-chlorobenzyl, 4-methylbenzyl or 2-phenylethyl.
  • R 4 is C 1-8 alkyl optionally substituted by alkoxycarbonyl.
  • R 4 is C 1-8 alkyl optionally substituted by C 3-6 cycloalkyl, aryl or heterocycle.
  • R 1 is hydrogen, methyl, cyanomethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-pentyl or n-hexyl;
  • R 2 is hydrogen, methyl or n-butyl
  • R 3 is 3-pentyl. 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl;
  • R 4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl; with the proviso that when R L is hydrogen, R 2 is methyl and R 3 is 1-(ethoxycarbonyl)propyl, then R 4 is different from n-pentyl, 3-bromobenzyl or 2-phenylethyl.
  • R 4 is C 1-8 alkyl optionally substituted by alkoxycarbonyl.
  • R 4 is different from 1-(ethoxycarbonyl)propyl.
  • R 1 is hydrogen, methyl, cyanomethyl, 2-methoxyethyl, n-propyl, 3-hydroxypropyl or 2-propynyl;
  • R 2 is methyl
  • R 3 is 3-pentyl, 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl;
  • R 4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl;
  • R 4 is different from 3-bromobenzyl.
  • R 4 is 1-(ethoxycarbonyl)propyl
  • R 4 is different from 1-(ethoxycarbonyl)propyl
  • R 1 is hydrogen; R 2 is methyl; R 3 is 1-(ethoxycarbonyl)propyl; and R 4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • a further embodiment consists in compounds wherein R 2 is methyl, R 3 is a group of formula —CHR 5 R 6 with R 5 being C 2-4 alkyl. R 6 being amido or —COOR 7 and R 7 being methyl or ethyl.
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-[(7-benzyl-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2- ⁇ [7-(3-bromobenzyl)-1-(2-ethoxy-2-oxoethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio ⁇ butanoate; ethyl 2- ⁇ [7-(3-bromobenzyl)-1-(2-methoxyethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio ⁇ butanoate; ethyl 2- ⁇ [7-(3-bromobenzyl)-2
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-[(7-benzyl-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2- ⁇ [7-(3-bromobenzyl)-1-(2-methoxyethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio ⁇ ) butanoate; ethyl 2- ⁇ [7-(3-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio ⁇ butanoate; ethyl 2-([7-(3-bromobenzyl)-1-(cyanomethyl)-3-methyl-2,6
  • compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2- ⁇ [7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio ⁇ butanoate; ethyl 2- ⁇ [3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio ⁇ butanoate; and ethyl 2-( ⁇ 3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl) ⁇ thio)butanoate.
  • the compounds are those having formula II, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts:
  • R.sup.1 is hydrogen or C.sub.1-6 alkyl:
  • R.sup.2 is hydrogen or C.sub. 1-4 alkyl;
  • R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group;
  • R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle;
  • R.sup.5 is hydrogen or C.sub. 1-4 alkyl;
  • R.sup.6 is C.sub.1-4 alkyl, amido or —COOR.sup.7;
  • R.sup.7 is C.sub.1-4 alkyl:
  • R.sup.3 is a benzyl group
  • R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl.
  • R.sup.3 is a group of formula —CHR.sup.5R.sup.6, then R.sup.4 is C.sub. 1-8 alkyl optionally substituted by C.sub.3-6 cycloalkyl, aryl or heterocycle.
  • the compounds are those compounds of formula II, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts
  • R.sup.1 is hydrogen or C.sub. 1-6 alkyl
  • R.sup.2 is hydrogen or C.sub. 1-4 alkyl
  • R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group
  • R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle
  • R.sup.5 is hydrogen or C.sub.1-4 alkyl
  • R.sup.6 is C.sub.1-4 alkyl, amido or —COOR.sup.7
  • R.sup.7 is C.sub.1-4 alkyl.
  • the compounds are compounds of formula IT selected from ethyl 2-[(7-heptyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]but-anoate; 7-(3-bromobenzyl)-3-methyl-8-(propylthio)-3,7-dihydro-1H-purine-2,-6-dione; ethyl 2-[(3-methyl-2,6-dioxo-7-pentyl-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]but-anoate; ethyl 2-[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl-]thiol butanoate; ethyl 2-[(3-methyl-2,6-dioxo-7-prop
  • the compounds are compounds of formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts
  • R.sup.1 is hydrogen or C.sub. 1-6 alkyl
  • R.sup.2 is hydrogen or C.sub.1-4 alkyl
  • R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group
  • R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle
  • R.sup.5 is C.sub.2-4 alkyl
  • R.sup.6 is C.sub.2-4 alkyl, amido or —COOR.sup.7
  • R.sup.7 is C.sub.1-4 alkyl
  • the compounds are compounds having formula II, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • R.sup.1 is hydrogen or C.sub. 1-6 alkyl
  • R.sup.2 is hydrogen or C.sub.1-4 alkyl
  • R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group
  • R.sup.4 is C.sub. 1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle
  • R.sup.5 is hydrogen or C.sub. 1-4 alkyl
  • R.sup.6 is C.sub.1-4 alkyl, amido or —COOR.sup.7
  • R.sup.7 is C.sub.1-4 alkyl:
  • a chemical composition that includes a LEV derivative of Formula 1 or Formula 2 is disclosed.
  • n of Formula 2 and L, X, and Y of Formulas 1 and 2 are defined as follows: a) n is an integer with a value of 0 to 8; b) L is one of the group consisting of CH 2 , CO, NHCO, NHCOO, CONH, NH, O, or S, and combinations thereof; c) X is an end group, an aromatic group, an aryl group, or a saturated, unsaturated, substituted, unsubstituted, straight chain, or branched chain aliphatic group having from 1 to 10 carbon and/or hetero chain atoms, the hetero chain atoms being selected from the group consisting of oxygen, nitrogen, sulfur, or phosphorus, and combinations thereof; and d) Y is optional and if present is one of a functional group selected from group consisting of alcohol amine, amide, carboxylic acid, aldehyde, ester, iminoester, isocyanate, isothiocyanate, anhydride, thiol,
  • the operative group of Z is selected from the group consisting of detectable labels, antigenic carriers, coupling agents, end groups, proteins, lipoproteins, glycoproteins, polypeptides, polysaccharides, nucleic acids, polynucleotides, teichoic acids, radioactive isotopes, enzymes, enzyme fragments, enzyme donor fragments, enzyme acceptor fragments, enzyme substrates, enzyme inhibitors, coenzymes, fluorescent moieties, phosphorescent moieties, anti-stokes up-regulating moieties, chemiluminescent moieties, luminescent moieties, dyes, sensitizers, particles, microparticles, magnetic particles, solid supports, liposomes, ligands, receptors, hapten radioactive isotopes, and combinations thereof.
  • the present invention provides a compound of Formula I:
  • each Z is independently selected from hydrogen and deuterium; R1 is an n-propyl group having zero to seven deuterium atoms; R2 is an ethyl group having zero to five deuterium atoms, and when each R has zero deuterium atoms, at least one Z is deuterium.
  • R1 is selected from CD3CH2CH2-, CD3CD2CH2-, CD3CH2CD2-, CH3CH2CD2-, CH3CD2CD2-, CD3CD2CD2- or CH3CH2CH2-.
  • R1 is CD3CD2CD2- or
  • CD3CD2CH2- CD3CD2CH2-.
  • Z1 and Z2 are both hydrogen.
  • Z1 and Z2 are both deuterium.
  • R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In a more specific embodiment, R2 is selected from CH3CH2- or CD3CD2-. In one aspect of these embodiments. Z1 and Z2 are both hydrogen. In another aspect of these embodiments, Z1 I and Z2 are both deuterium.
  • R1 is CD3CH2CH2-, CD3CD2CH2-, CD3CH2CD2-, CH3CH2CD2-, CH3CD2CD2-, CD3CD2CD2- or CH3CH2CH2-; and R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-.
  • R2 is CH3CH2- or CD3CD2-.
  • R1 is CD3CD2CD2- or CD3CD2CH2-; and R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-.
  • R2 is CH3CH2— or CD3CD2-.
  • Examples of specific compounds of this invention include the following:
  • the compounds of the present invention are those covered by formula (I), their diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.
  • R1 is hydrogen, substituted or unsubstituted C1-12 alkyl, substituted or unsubstituted aryl or substituted or unsubstituted 3-8 membered heterocycle.
  • R2 is hydrogen.
  • R1 and R2 may be linked together in such a way to form a C3-6 cycloalkyl.
  • R3 is either
  • R4 in formula (I) is selected from the group comprising or consisting of hydrogen; C1-12 alkyl optionally substituted by halogen, C1-4 alkoxy, C1-4 alkylthio, azido, nitrooxy or an aryl; C2-12 alkenyl optionally substituted by halogen; C2-12 alkynyl optionally substituted by halogen; azido; alkoxycarbonylamino; arylsulfonyloxy; a substituted or unsubstituted aryl; or a 3-8 membered substituted or unsubstituted heterocycle;
  • R4 is hydrogen; or R4 is C1-12 alkyl or a C1-6 alkyl, optionally substituted by halogen, C1-4 alkoxy, C1-4 alkylthio, azido or nitrooxy; or R4 is C2-12 alkenyl or a C1-6 alkenyl optionally substituted by halogen; or R4 is C2-12 alkynyl or a C1-6 alkynyl optionally substituted by halogen; or R4 is alkoxycarbonylamino.
  • R5 is hydrogen
  • R4 may form together with R5 and the 2-oxo-1-pyrrolidine ring a 1,3-dihydro-2H-indol-2-one ring of the following structure:
  • R6 is hydrogen or halogen.
  • R7 in formula (I) is selected from the group comprising or consisting of hydrogen; nitro; halogen; heterocycle; amino; aryl; C1-12 alkyl optionally substituted by at least one halogen; or C1-12 alkoxy optionally substituted by at least one halogen.
  • R8 in formula (I) is selected from the group comprising or consisting of hydrogen. C1-12 alkyl optionally substituted by halogen, or halogen.
  • R9 in formula (I) is selected from the group comprising or consisting of hydrogen, C1-12 alkyl optionally substituted by halogen, or halogen.
  • a further aspect of the present invention consists in compounds of formula (I) wherein
  • R1 and R2 are both hydrogen.
  • R3 is:
  • R3 is:
  • R4 is n-propyl, 2,2,2-trifluoroethyl, 2-chloro-2,2-difluoroethyl, 2 bromo-2,2-difluoroethyl, 2,2-difluorovinyl.
  • R4 is phenyl, 2,3,5-trifluorophenyl or 3-chloro-4-fluorophenyl.
  • R5 is hydrogen
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R4 forms together with R5a 1,3-dihydro-2H-indol-2-one ring
  • R6 is hydrogen; R7 is chlorine; R8 is hydrogen; R9 is hydrogen.
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of:
  • R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of:
  • Said heterocycles are optionally substituted by e.g. a methyl, n-propyl, trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, iodine, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclopropylmethoxy, cyclobutylmethoxy, amino, methylamino, cyclopropylamino, cyclobutylamino, 1-pyrrolidinyl, cyano, phenyl, benzyl or 3-thienyl.
  • R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: 6-chloro-2-cyclopropylimidazo[1,2-b]pyridazin-3-yl, 6-(cyclopropyloxy)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl. 6-propoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl.
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is a substituted or unsubstituted imidazo[1,2-a]pyridin-3-yl.
  • Said imidazo[1,2-a]pyridin-3-yl is optionally substituted by e.g. a methyl, cyclopropyl, bromine, chlorine, fluorine, iodine.
  • R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: imidazo[1,2-a]pyridin-3-yl, 6-methylimidazo[1,2-a]pyridin-3-yl, 2-chloroimidazo[1,2-a]pyridin-3-yl.
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its N atoms and is selected from the group consisting of:
  • a specific further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a heterocycle linked to the rest of the molecule via one of its N atoms and is selected from the group consisting of:
  • Said heterocycles may optionally be substituted by trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, methoxy or cyano.
  • R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of 6-bromo-2-chloro-3H-imidazo[4,5-b]pyridin-3-yl, 6-bromo-2-cyclopropyl-3H-imidazo[4,5-b]pyridin-3-yl, 1H-pyrrolo[3,2-b]pyridin-1-yl, 2,5-dichloro-1H-pyrrol-1-yl, 2-chloro-5-methoxy-1H-benzimidazol-1-yl, 5-bromo-2-chloro-1H-benzimidazol-1-yl or 2,5-dichloro-1H-benzimidazol-1-yl.
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R1, R2 and R5 are hydrogen.
  • R4 is a C1-6 alkyl optionally substituted by halogen, a C2-6 alkenyl optionally substituted by halogen or C2-12 alkynyl optionally substituted by halogen.
  • R3 is selected from the group consisting of;
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R1, R2 and R5 are hydrogen.
  • R4 is a C1-6 alkyl optionally substituted by halogen, a C2-6 alkenyl optionally substituted by halogen or C2-12 alkynyl optionally substituted by halogen.
  • R3 is selected from the group consisting of
  • a further embodiment of the invention consists in compounds of formula (I), their diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.
  • R1, R2 and R5 are hydrogen.
  • R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms, said heterocycle is selected from the group consisting of:
  • imidazo[1,2-a]pyridin-3-yl imidazo[1,2-a]pyrimidin-3-yl; imidazo[1,2-b]pyridazin-3-yl; 1H-imidazol-4-yl; 1H-imidazol-5-yl;
  • R4 is a substituted or unsubstituted phenyl moiety:
  • a further embodiment of the present invention consists in compounds of formula (I) wherein R1 is hydrogen or C1-12 alkyl;
  • R2 is hydrogen;
  • R3 is an aromatic 5-membered heterocycle linked to the rest of the molecule via one of its C atoms;
  • R4 is hydrogen, C1-12 alkyl or aryl;
  • R5 is hydrogen:
  • R4 can form together with R5 and the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R6 is hydrogen or halogen:
  • R4 may not be hydrogen when R3 is substituted 1H-pyrazol-5-yl. Also this embodiment does not comprise 5-(2′-oxo-1′-pyrrolidinyl)methyl-1,3,4-tricarbomethoxy-pyrazole which is disclosed in A. Padwa et al J. Org. Chem. 2000, 65, 5223-5232 without any biological activity though.
  • R3 is an aromatic 5-membered heterocycle linked to the rest of the molecule via one of its C atoms
  • specific moieties R3 may be selected from 1,3-thiazol-5-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl, 2-oxo-2,3-dihydro-1,3-thiazol-5-yl, each of them being optionally substituted by 1 to 3 substituents independently selected from methyl, chlorine, bromine, amino, methylamino, dimethylamino, (2-oxo-4-propyl-pyrrolidin-1-yl)methyl, 1-pyrrolidinyl, amido, cyano, methoxy, phenyl, 4-methylphenyl-sulfonyl, benzyl or 2-(benzylamino)-2-oxoethyl.
  • more specific moieties R3 are selected from 2-(methylamino)-1,3-thiazol-5-yl; 2-pyrrolidin-1-yl-1,3-thiazol-5-yl: 5-bromo-1H-imidazol-4-yl; 5-chloro-1H-imidazol-4-yl; 1H-imidazol-5-yl; 1-methyl-1H-imidazol-5-yl; 4-bromo-1-methyl-1H-imidazol-5-yl; 4-chloro-1H-imidazol-5-yl; 4-chloro-1-methyl-1H-imidazol-5-yl: 4-cyano-1-methyl-1H-imidazol-5-yl; 1H-pyrazol-4-yl; 3,5-dimethyl-1H-pyrazol-4-yl; 3-methyl-1H-pyrazol-4-yl.
  • most specific moieties R3 are selected from 5-bromo-1H-imidazol-4-yl; 5-chloro-1H-imidazol-4-yl; 1H-imidazol-5-yl; 4-bromo-1-methyl-1H-imidazol-5-yl; 4-chloro-1-methyl-1H-imidazol-5-yl: 1H-pyrazol-4-yl.
  • a specific moiety R1 is selected from hydrogen or ethyl.
  • a specific moiety R4 is selected from hydrogen, n-propyl, 2,3,5-trifluorophenyl or phenyl.
  • a further embodiment of the present invention consists in compounds having the specific formula (Ia).
  • R10 is hydrogen; halogen; C1-4 alkyl optionally substituted by at least one halogen; C1-4 alkoxy; methoxycarbonyl; nitro; amino; alkylamino; amido; or alkanoyl-amino.
  • R10 is hydrogen.
  • R11 is hydrogen; halogen: C1-4 alkyl optionally substituted by at least one halogen; C1-4 alkoxy; methoxycarbonyl; nitro: amino; alkylamino; amido; or alkanoylamino.
  • R11 is hydrogen.
  • R4 is C1-4 alkyl optionally substituted by at least one halogen; or C2-4 alkenyl optionally substituted by at least one halogen.
  • R4 is n-propyl.
  • a specific embodiment relates to an embodiment wherein R10 is selected from hydrogen; methyl; fluorine; chlorine; bromine; methoxy; methoxycarbonyl; nitro; or trifluoromethyl, while R11 is selected from hydrogen; methyl; fluorine; chlorine; bromine; methoxy; methoxycarbonyl; nitro; or trifluoromethyl; and R3 is n-propyl.
  • the present invention relates to the novel compound (S)-alpha-ethyl-2-oxo-1-pyrrolidineacetamide.
  • the present invention relates to the novel compound, (R)-alpha-ethyl-2-oxo-1-pyrrolidineacetamide
  • R1, R2, R3 and R4 which may be the same or different independently represent hydrogen, C1-6 alkyl, phenyl or phenyl substituted by one or more halogen, hydroxyl, nitro, amino, C1-6 alkyl or C1-C6 alkoxy groups;
  • R5 and R6 independently represent hydrogen, C1-C6 alkyl or C3-C6 cycloalkyl, or R5 and R6 together with the nitrogen form a C 4-6 N heterocycle:
  • m represents an integer from 1-2; and
  • n represents an integer from 1-3; provided that, two of the substituents R1, R2, R3 and R4 independently represent phenyl or substituted phenyl and the other two independently represent hydrogen or C1-6 alkyl; or a pharmaceutically acceptable acid addition salt thereof.
  • Pharmaceutically acceptable acid addition salts of the compounds of formula I include salts of mineral acids, for example, hydrohalic acids, e.g. hydrochloric or hydrobromic; or organic acids, e.g. formic, acetic or lactic acids.
  • the acid may be polybasic, for example sulphuric, fumaric, maleic or citric acid.
  • This invention also relates to all stereoisomeric forms and optical enantiomeric forms of the compounds of formula I.
  • alkyl groups which R1, R2, R3, R4, R5 and R6 may represent include methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl and s-butyl;
  • cycloalkyl groups which R5 and R6 may represent include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; C1-6 alkoxy groups include methoxy, ethoxy and propoxy; halogen groups include fluorine, chlorine, bromine or iodine;
  • R1 is hydrogen, phenyl or substituted phenyl, preferably phenyl
  • R2 is hydrogen, phenyl or substituted phenyl, preferably phenyl
  • R3 is hydrogen, phenyl or substituted phenyl, preferably hydrogen:
  • R4 is hydrogen, phenyl or substituted phenyl, preferably hydrogen;
  • R5 is hydrogen, C1-3 alkyl or cyclopropyl, preferably hydrogen or methyl;
  • R6 is hydrogen, C1-3 alkyl or cyclopropyl, preferably hydrogen or methyl;
  • m represents an integer from 1-2 preferably 2;
  • n represents an integer from 1-2, preferably 1.
  • the invention therefore provides a compound having the formula I or a pharmaceutically acceptable salt thereof,
  • RI is hydrogen, CI-20 alkyl, C3 23 cycloalkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, guanidine, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl, arylsulfinyl, aryl or heterocycle;
  • R2 is hydrogen, C1 20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;
  • R3 is hydrogen, C1 20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R4 is hydrogen, C1-20 alkyl, C2-12 alkenyl, C2-12 alkynyl, aryl, azido, alkoxycarbonylamino, arylsulfonyloxy or heterocycle;
  • R4a is hydrogen or C1-20 alkyl; or R4 and R4a can form together a C3-8 cycloalkyl;
  • R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R6 is hydrogen or C1 20 alkyl; R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl;
  • R8 is hydrogen, halogen, nitro, cyano, C1 20 alkyl or alkoxy;
  • R9 is hydrogen, C1-20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
  • RIO is hydrogen, C1 20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulf
  • N represents a group of formula
  • the asterisk * indicates the point of attachment of the substituents.
  • the invention concerns a compound having the formula I, their tautomers, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • RI is hydrogen, C1-20 alkyl, C3-8 cycloalkyl, halogen, hydroxy, ester, amido, cyano, nitro, amino, guanidine, alkylthio, alkylsulfonyl, alkylsulfinyl, aryl or heterocycle:
  • R2 is hydrogen, C1 20 alkyl, halogen, cyano, ester, carbamate or amido;
  • R3 is hydrogen, cyano, C1 20 alkyl, halogen or ester, or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R4a is hydrogen
  • R5 is hydrogen: or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R6 is hydrogen or C1 20 alkyl; R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl; R8 is hydrogen; R9 is hydrogen, C 1-20 alkyl, halogen or alkoxy; RIO is hydrogen, C1 20 alkyl, halogen or cyano; R11 is hydrogen; R12 is hydrogen or halogen; R13 is hydrogen, halogen, heterocycle or C1 20 alkyl; R14 is hydrogen; R15 is hydrogen; with the proviso that R4 is different from hydrogen when
  • alkyl represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched or cyclic or combinations thereof and containing 1-20 carbon atoms, preferably 1-10 carbon atoms, more preferably 1-4 carbon atoms; most preferred alkyl groups have 1-3 carbon atoms.
  • Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, cyano, azido, aryloxy, alkoxy, alkylthio, alkanoylamino, arylcarbonylamino, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl or aryl.
  • alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, n-heptyl, 2,4,4-trimethylpentyl, n-decyl, chloromethyl, trifluoromethyl, 2-bromo-2,2-difluoroethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino) methyl, (propionylamino) methyl, (benzoylamino) methyl, (4-chlorophenoxy)methyl, benzyl, 2-phenylethyl or 2-(methylthio) ethyl.
  • Preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, 2,4,4-trimethylpentyl, chloromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino) methyl, (propionylamino) methyl, (benzoylarino) methyl or 2-(methylthio) ethyl.
  • More preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, azidomethyl or trifluoromethyl. Most preferred alkyl groups are methyl or n-propyl.
  • cycloalkyl represents a monovalent group of 3 to 8 carbon atoms, usually 3-6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups.
  • Preferred cycloalkyl groups are cyclopropyl and cyclohexyl.
  • alkenyl represents straight, branched or cyclic unsaturated hydrocarbon radicals or combinations thereof having at least one carbon-carbon double bond, containing 2-12 carbon atoms, preferably usually 2-4 carbon atoms.
  • Alkenyl groups are being optionally substituted with any suitable group, including but not limited to one or more moities selected from groups as described above for the alkyl groups.
  • an alkenyl group is ethenyl (vinyl) optionally substituted by 1 to 3 halogens.
  • Preferred alkenyl group in the present case, is 2,2-difluorovinyl.
  • alkynyl represents straight, branched or cyclic hydrocarbon radicals or combinations thereof containing at least one carbon-carbon triple bond, containing 2-12 carbon atoms, preferably 2-6 carbon atoms, and being optionally substituted by any suitable group, including but not limited to one or more moities selected from groups as described above for the alkyl groups.
  • an alkynyl group is a halogenoalkynyl group (haloalkynyl group).
  • Groups qualified by prefixes such as “s”, “i”, “t” and the like (e.g. “i-propyl”, “s-butyl”) are branched derivatives.
  • aryl as used herein, is defined as phenyl optionally substituted by 1 to 4 substituents independently selected from halogen, cyano, alkoxy, alkylthio, C1-3 alkyl or azido, preferably halogen or azido.
  • aryl groups in the present case are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl, 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl.
  • aryl groups are phenyl, 3-chlorophenyl.
  • 3-fluorophenyl 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • Most preferred aryl groups are phenyl, 3-chlorophenyl.
  • heterocycle is defined as including an aromatic or non aromatic cycloalkyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure.
  • Heterocyclic ring moities can be optionally substituted by alkyl groups or halogens and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl.
  • heterocycles are 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-tetrahydrofuranyl, 1H-pyrrol-2-yl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrazol-2-yl, 1H-pyrazol-3-yl.
  • Preferred heterocycles are 1H-imidazol-2-yl, 1,2,3-thiadiazol-4-yl, 1H-pyrazol-3-yl, 2-furyl, 3-furyl, 2-thienyl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrrol-2-yl.
  • halogen includes an atom of chlorine, bromine, fluorine, iodine. Usually halogens are chlorine, bromine and fluorine. Preferred halogens are fluorine, bromine and chlorine.
  • hydroxy represents a group of formula —OH.
  • alkoxy represents a group of formula —ORa wherein Ra is an alkyl group, as defined above. Preferred alkoxy group is methoxy.
  • aryloxy represents a group of formula —ORb wherein Rb is an aryl group, as defined above. Preferred aryloxy group is phenoxy.
  • ester represents a group of formula —COORC wherein Rc is an alkyl group or aryl group, as defined above. Preferred ester group is methoxycarbonyl.
  • amino represents a group of formula —NH2.
  • amino acid represents an alkylamino or an arylamino group, wherein the terms “alkyl” and “aryl” are defined as above.
  • cyano represents a group of formula —CN.
  • nitro represents a group of formula —NO2.
  • guanidine represents a group of formula —NHC( ⁇ NH) NH2.
  • alkylthio represents a group of formula —SRd wherein Rd is an alkyl group, as defined above.
  • Rd is an alkyl group, as defined above.
  • One alkylthio group is methylthio.
  • alkylsulfonyl represents a group of formula —S( ⁇ O) 2Re wherein Re is an alkyl group, as defined above.
  • Re is an alkyl group, as defined above.
  • One alkylsulfonyl group is methylsulfonyl.
  • alkylsulfinyl represents a group of formula —S( ⁇ O)Rf wherein Rf is an alkyl group, as defined above.
  • Rf is an alkyl group, as defined above.
  • One alkylsulfinyl group is methylsulfinyl.
  • arylthio represents a group of formula —SRg wherein Rg is an aryl group, as defined above.
  • arylsulfonyl represents a group of the formula —S( ⁇ O) 2Rh wherein Rh is an aryl group, as defined above.
  • arylsulfinyl represents a group of the formula —S( ⁇ O) Ri wherein Ri is an aryl group, as defined above.
  • carbamate represents a group of formula —N(H)C(O) OR1, wherein Ri is an alkyl or an aryl, as defined above.
  • carbamate groups are (propoxycarbonyl)amino or (benzyloaxycarbonyl)amino.
  • One carbamate group is (benzyloaxycarbonyl)amino.
  • alkanoylamino represents a group of the formula —NHC ( ⁇ O) Rk wherein Rk is an alkyl group, as defined above.
  • (arylcarbonyl)amino represents a group of the formula —NHC( ⁇ O)Rm wherein Rm is an aryl group, as defined above.
  • One (arylcarbonyl)amino is benzoylamino.
  • RI is hydrogen; C1 lo alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; hydroxy; C3-6 cycloalkyl; halogen; ester, amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl groups; or guanidine.
  • RI is hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl; n-butyl; i-butyl; t-butyl; -ethylpropyl; 2,4,4-trimethylpentyl; hydroxymethyl; chloromethyl; trifluoromethyl; 2,2,2-trifluoroethyl; cyanomethyl; 2-(methylthio) ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl or 1H-imidazol-2-yl.
  • RI is hydrogen; methyl; ethyl; i-propyl; n-propyl; n-butyl; methylthio: nitro; cyano; amino; chloro or 1H-pyrrol-2-yl.
  • RI is hydrogen; methyl; methylthio; nitro; cyano; amino or chloro.
  • R2 is hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate; [(N-methoxy-N-methyl)amino]carbonyl.
  • R2 is hydrogen; methyl; hydroxymethyl; (acetylamino) methyl; (propionylamino) methyl; (benzoylamino) methyl; [(benzyloxy) carbonyl]amino; chloro or cyano.
  • R2 is hydrogen; chloro or cyano.
  • R3 is hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano. In some embodiments, R3 is hydrogen; hydroxymethyl; chloro; cyano.
  • R3 is hydrogen or cyano. In some embodiments R3 is hydrogen.
  • R4 is hydrogen; C1-4 alkyl unsubstituted or substituted by halogens; C2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens.
  • R4 is hydrogen; C1-4 alkyl unsubstituted or substituted by halogens; C2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens.
  • R4 is hydrogen; C1-4 alkyl unsubstituted or substituted by halogens; C2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens.
  • R4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl.
  • R4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • R4 is n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • R4a is hydrogen
  • R5 is hydrogen
  • R6 is hydrogen or Cl-1 ⁇ 0 alkyl unsubstituted or substituted by hydroxy or azido.
  • R6 is hydrogen or azidomethyl. More preferably R6 is hydrogen.
  • R7 is hydrogen
  • R6 and R7 are linked to form a cyclopropyl.
  • R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R8 is hydrogen
  • R9 is hydrogen; halogen; 1-3 alkyl or alkoxy. In some embodiments, R9 is hydrogen; methyl; chloro or methoxy. In some embodiments R9 is hydrogen.
  • RIO is hydrogen; halogen; cyano; CI 3 alkyl unsubstituted or substituted by halogens; or alkoxy.
  • RIO is methyl; hydrogen; trifluoromethyl; fluoro; cyano or methoxy.
  • R10 is hydrogen; trifluoromethyl; fluoro or cyano.
  • RI 1 is hydrogen
  • R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R12 is hydrogen or halogen. In some embodiments R12 is hydrogen; chloro or fluoro. In some embodiments R12 is hydrogen.
  • R13 is hydrogen; C1 3 alkyl; halogen or thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl.
  • R13 is hydrogen; chloro; bromo or methyl.
  • R13 is chloro; bromo or methyl.
  • R14 is hydrogen.
  • R15 is hydrogen
  • the compounds of formula I, or pharmaceutically acceptable salts thereof are those wherein
  • RI is selected from hydrogen; C1 lo alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; C3 6 cycloalkyl; halogen; ester; amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl group; or guanidine; R2 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate or [(N-methoxy-N-methyl)amino]carbonyl.
  • R3 is selected from hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano;
  • R4 is selected from hydrogen; C1 4 alkyl unsubstituted or substituted by halogens; C2 4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens;
  • R4a is hydrogen;
  • R5 is hydrogen;
  • R6 is selected from hydrogen or C 1-10 alkyl unsubstituted or substituted by hydroxy or azido;
  • R7 is hydrogen; or R6 and R7 can be linked to form a cyclopropyl; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • R8 is hydrogen: R9 is selected from hydrogen; halogen: C1-3 alkyl; alkoxy; R10 is selected from hydrogen; halogen; cyano or Cil alkyl unsubstituted or substituted by halogens; or alkoxy; R 1 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R12 is selected from hydrogen or halogen
  • R13 is selected from hydrogen; CI-3 alkyl; halogen; thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl
  • R14 is hydrogen
  • R15 is hydrogen; with the proviso that R4 is different from hydrogen when
  • the compounds of formula I, or pharmaceutically acceptable salt thereof are those wherein
  • RI is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl; n-butyl; i-butyl; t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; trifluoromethyl; 2,2,2-trifluoroethyl; hydroxymethyl; chloromethyl; cyanomethyl; 2-(methylthio) ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl: methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl; or 1H-imidazol-2-yl; R2 is
  • R8 is hydrogen; R9 is selected from hydrogen; methyl; choro; methoxy; R10 is selected from methyl; hydrogen; trifluoromethyl; fluoro; cyano; or methoxy; R is hydrogen; R4 is selected from hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl: 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl; or 3-azido-2,4,6-trifluorophenyl.
  • R4a is hydrogen; R5 is hydrogen; or R4, R
  • the compounds of formula I, or pharmaceutically acceptable salt thereof are those wherein
  • RI is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; n-butyl; methylthio; nitro; cyano; amino; chloro; or 1H-pyrrol-2-yl; R2 is selected from hydrogen; chloro; cyano; R3 is selected from hydrogen; cyano; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle

Landscapes

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

Abstract

The invention relates to methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania) by using a combination of a synaptic vesicle protein 2A (SV2A) inhibitor and an antipsychotic or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs thereof. In some embodiments, the methods and the compositions are for treating one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia or bipolar disorder (in particular, mania).

Description

  • This application claims priority and benefit from U.S. Provisional Patent Application 61/726,440, filed Nov. 14, 2012, the contents and disclosures of which are incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • The invention relates to methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania). In particular, it relates to the use of a combination of a synaptic vesicle protein 2A (SV2A) inhibitor and an antipsychotic in treating a subject having or at risk for schizophrenia or bipolar disorder (in particular, mania).
  • BACKGROUND OF THE INVENTION
  • Schizophrenia is a chronic psychiatric disorder, characterized by a wide spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g., hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalities in the brain are proposed to underlie the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely ineffective in treating cognitive impairments in schizophrenia patients.
  • Cognitive impairments in schizophrenia involve both frontal and temporal lobe functions that include memory, attention, processing speed, and executive control. Recent observations, drawn from preclinical animal models and human neuroimaging studies, indicate that altered brain activity/excitability in the medial temporal lobe memory system may contribute to cognitive impairment and may also play a role in augmenting psychotic symptoms due to disinhibition of dopaminergic neurons.
  • Cognitive deficits are increasingly recognized as a key clinical feature that can be detected in a prodromal phase and in remission, as well as during full expression of the illness but are not effectively treated by available antipsychotics. Because untreated features of schizophrenia, especially impaired cognition, predict long-term disability in patients (Green et al., Schizophr. Res. 2004, 72, 41-45), it is critical to develop effective therapies for the spectrum of this illness.
  • SUMMARY OF THE INVENTION
  • In accordance with a first aspect of the present invention, there is provided a method for treating a subject suffering from schizophrenia or bipolar disorder (in particular, mania), or at risk thereof, the method comprising the step of administering to said subject a therapeutically effective amount of a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof in combination with a therapeutically effective amount of an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In some embodiments, the methods of the present invention treat one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia. In some embodiments, the methods of the present invention treat one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). In some embodiments of this invention, the methods of this invention prevent or slow the progression of cognitive impairment or bipolar disorder (in particular, mania) of schizophrenia in said subject.
  • The SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this aspect of the invention include those disclosed in, for example, United States (U.S.) patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, and U.S. Patent Application 61/175,536. However, any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of this aspect of the invention. In other embodiments, the SV2A inhibitor is selected from the group of SV2A inhibitors referred to in International Patent Applications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692 or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs. In other embodiments, the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof. In other embodiments, the SV2A inhibitor is levetiracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2A inhibitor is brivaracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2A inhibitor is seletracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.
  • The antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this invention include both typical and atypical antipsychotics.
  • In some embodiments, the antipsychotics suitable for use in the present invention are selected from atypical antipsychotics, including, but not limited to, those disclosed in, for example, U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
  • In some embodiments, atypical antipsychotics suitable for use in the present invention include, but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof. In some embodiments, the antipsychotic of this invention is selected from aripiprazole (Bristol-Myers Squibb), olanzapine (Lilly) and ziprasidone (Pfizer), and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
  • In some embodiments, the antipsychotics suitable for use in the present invention are typical antipsychotics, including, but not limited to, acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
  • In some embodiments of the present invention, the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be selected from compounds that are dopaminergic agents (such as dopamine D1 receptor antagonists or agonists, dopamine D2 receptor antagonists or partial agonists, dopamine D3 receptor antagonists or partial agonists, dopamine D4 receptor antagonists), glutamatergic agents, N-methyl-D-aspartate (NMDA) receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents (such as alpha-2 adrenergic receptor agonists or antagonists and catechol-O-methyl transferase (COMT) inhibitors), serotonin receptor modulators (such as 5-HT2A receptor antagonists, 5-HT1A receptor partial agonists, 5-HT2C agonists, and 5-HT6 antagonists), cholinergic agents (such as alpha-7 nicotinic receptor agonists, alpha-4-beta2 nicotinic receptor agonists, allosteric modulators of nicotinic receptors and acetylcholinesterase inhibitors, muscarinic receptor agonists and antagonists), cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, monoamine oxidase (MAO) B inhibitors, PDE10 inhibitors, neuronal nitric oxide synthase (nNOS) inhibitors, neurosteroids, and neurotrophic factors.
  • In some embodiments, the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this invention include compounds that may be used to treat at least one sign or symptom of schizophrenia or bipolar disorder (in particular, mania).
  • In some embodiments of this aspect of the invention, the antipsychotic is administered at a dose that is subtherapeutic as compared to the dose at which it is therapeutically effective when administered in the absence of the SV2A inhibitor.
  • In some embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof can be administered at doses as disclosed, for example, in U.S. patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, U.S. Patent Application 61/175,536, and U.S. Patent Application 61/441,251. In other embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.001 mg/kg to 5 mg/kg. In other embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 5 mg/kg, or about 1 to 2 mg/kg, or about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.01 to 1 mg/kg, or about 0.001 to 1 mg/kg, or about 0.5 mg/kg to 5 mg/kg, or about 0.05 mg/kg to 0.5 mg/kg. In other embodiments of this aspect of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 2.0 to 4.0 mg/kg, or about 2.0 to 3.0 mg/kg, or about 3.0 to 4.0 mg/kg, or about 0.2 to 0.4 mg/kg, or about 0.2 to 0.3 mg/kg, or about 0.3 to 0.4 mg/kg, or about 0.001-5 mg/kg, or about 0.001-0.5 mg/kg, or about 0.01-0.5 mg/kg. In some embodiments, the SV2A inhibitor may be selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof, said SV2A inhibitor being administered every 12 or 24 hours at a daily dose selected from any of the above.
  • In some embodiments of the invention, the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs are administered simultaneously, or sequentially, or in a single formulation, or in separate formulations packaged together. In other embodiments, the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs are administered via different routes. As used herein, “combination” includes administration by any of these formulations or routes of administration.
  • In some embodiments of the invention, the combined treatment has a longer or improved therapeutic effect in the subject than is attained by administering the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the absence of the SV2A inhibitor or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×.
  • In some embodiments of the invention, the combined treatment has a longer or improved therapeutic effect in the subject than is attained by administering the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the absence of the antipsychotic or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×.
  • In accordance with another aspect of the present invention, there is provided a method of increasing the therapeutic index of an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in a method of treating schizophrenia or bipolar disorder (in particular, mania) in a subject in need or at risk thereof, comprising administering a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in combination with the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof to said subject.
  • In some embodiments, the increase in the therapeutic index of the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is greater than the therapeutic index of the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof when administered in the absence of the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×.
  • In accordance with another aspect of this invention, there is provided a pharmaceutical composition for treating a subject suffering from schizophrenia or bipolar disorder (in particular, mania), or at risk thereof, the composition comprising a SV2A inhibitor and an antipsychotic or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof. In some embodiments, the composition of this invention is for treating one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia. In some embodiments, the composition of this invention is for treating one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). In some embodiments, the composition is in a liquid form. In some embodiments, the composition is in an aqueous solution. In some embodiments, the composition is in a suspension form. In some embodiments, the composition is in a sustained release form, or a controlled release form, or a delayed release form, or an extended release form. In some embodiments, the composition is in a unit dosage form. In other embodiments, the two components of the compositions are in separate delivery forms packaged together.
  • In some embodiments of this invention, the composition comprises levetiracetam, brivaracetam, seletracetam, or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and an antipsychotic or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.
  • In some embodiments of this invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the composition is present in an amount of 0.07-350 mg, or 50-250 mg, or 3-50 mg. In some embodiments, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is present in an amount less than 350 mg, less than 250 mg, less than 200 mg, less than 150 mg, less than 100 mg, less than 50 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0.5 mg, less than 0.1 mg, or less than 0.07 mg. In certain embodiments of this aspect of the invention, the SV2A inhibitor is present in an amount of 0.07-60 mg, 0.07-350 mg, 25-60 mg, 25-125 mg, 50-250 mg, 5-140 mg, 0.7-180 mg, 125-240 mg, 3-50 mg, or 3-60 mg. In other embodiments of this aspect of the invention, the SV2A inhibitor is present in an amount of 0.05-35 mg. In some embodiments of the composition of this invention, the SV2A inhibitor may be selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof, said SV2A inhibitor being present in an amount selected from any of the above.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts increased mRNA expression of the gene encoding SV2A in the dentate gyrus of the hippocampus of aged-impaired rats (AI) as compared to young rats (Y) and aged-unimpaired rats (AU). Normalized Affymetrix GeneChip probe set signal values (Y-axis), as a measure of mRNA expression, are plotted against learning indices of different rats, as a measure of cognitive impairment.
  • FIG. 2 depicts the effects of administering levetiracetam on the spatial memory retention of six aged-impaired rats (AI) in a Morris Water Maze (MWM) test. Three treatment conditions were employed: vehicle control, levetiracetam (5 mg/kg/day) and levetiracetam (10 mg/kg/day). The AI rats were trained for two consecutive days, with a one-time treatment prior to the training trials per day. 24 hours later, the AI rats were tested. The time the AI rats, 24 hours after treatment with the different conditions and two days of training, spent swimming in the target quadrant or the target annulus in a memory retention trial is used as a measure of spatial memory retention. The target quadrant refers to the quadrant of the maze (which is a circular pool) where the escape platform is placed during the training trials. The target annulus refers to the exact location of the escape platform during the training trials.
  • FIG. 3 depicts the effects of administering levetiracetam on the spatial memory retention of ten aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test. Six treatment conditions were employed: vehicle control, levetiracetam (1.25 mg/kg), levetiracetam (2.5 mg/kg), levetiracetam (5 mg/kg), levetiracetam (10 mg/kg) and levetiracetam (20 mg/kg). In the RAM task used, there was a one-hour delay between presentation of a subset of arms (5 arms available and 3 arms blocked) and completion of the eight-arm win-shift task (eight arms available). Rats were pre-treated 30-40 minutes before daily trials with a one-time drug/control treatment. The number of errors made by the rats after the delay was used as a measure of spatial memory retention. Errors were defined as instances when rats entered an arm from which food had already been retrieved in the pre-delay component of the trial or when rats re-visited an arm in the post-delay session that had already been visited. Paired t-tests were used to compare the number of errors between different doses of levetiracetam and vehicle control.
  • FIG. 4 depicts the effects of administering levetiracetam or valproate separately on the spatial memory retention of ten aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test.
  • FIG. 5 depicts the effects of administering levetiracetam or valproate in combination on the spatial memory retention often aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test.
  • FIG. 6 shows an isobologram plotting levetiracetam dose against valproate dose. The diagonal straight line is the line of additivity, anchored on each axis by the lowest effective doses of valproate and levetiracetam when assessed individually.
  • FIG. 7 depicts the experimental design of the human trials for levetiracetam treatment.
  • FIG. 8A depicts the average activity in the left CA3 of aMCI subjects with placebo treatment and age-matched control subjects with placebo treatment during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 8B depicts the average activity in the left CA3 of aMCI subjects with placebo treatment or levetiracetam treatment (125 mg twice a day for two weeks) during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 8C is a table of the data represented in FIGS. 8A and 8B.
  • FIG. 9A depicts the average activity in the left entorhinal cortex of age-matched control subjects with placebo treatment and aMCI subjects with placebo treatment during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 9B depicts the average activity in the left entorhinal cortex of the same aMCI subjects with placebo treatment or levetiracetam treatment (125 mg twice a day for two weeks) during the presentation of lure stimuli that the subject correctly identified as “similar.”
  • FIG. 9C is a table of the data represented in FIGS. 9A and 9B.
  • FIG. 10A depicts an example of the sequence of images shown to subjects in the explicit 3-alternative forced choice task described in Example 2.
  • FIG. 10B shows sample pairs of similar (“lure”) images.
  • FIG. 11 shows the difference between the aMCI (placebo) subjects and age-matched control (placebo) subjects in their performance of the explicit 3-alternative forced choice task described in Example 2. Each bar represents the proportion of the subject responses (old, similar, or new) when presented with a lure image.
  • FIG. 12 shows the difference between the same aMCI subjects with placebo treatment or with levetiracetam treatment (125 mg twice a day for two weeks) in their performance of the explicit 3-alternative forced choice task described in Example 2. Each bar represents the proportion of the subjects responses (old, similar, or new) when presented with a lure image.
  • FIG. 13 is a table of the data represented in FIGS. 11 and 12.
  • FIG. 14A shows the difference between the age-matched control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Buschke Selective reminding Test—Delayed Recall.
  • FIG. 14B is a table of the data represented in FIG. 14A.
  • FIG. 15A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Benton Visual Retention Test.
  • FIG. 15B is a table of the data represented in FIG. 15A.
  • FIG. 16A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Verbal Paired Associates Test—Recognition.
  • FIG. 16B is a table of the data represented in FIG. 16A.
  • FIG. 17A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Verbal Paired Associates Test—Delayed Recall.
  • FIG. 17B is a table of the data represented in FIG. 17A.
  • FIG. 18A is a table showing the subject selection process for the human levetiracetam trial described in Example 2.
  • FIG. 18B is a table showing the characteristics of the subjects selected for the human levetiracetam trial described in Example 2.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurochemistry, virology, immunology, microbiology, pharmacology, genetics and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art.
  • The methods and techniques of the present invention are generally performed, unless otherwise indicated, according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout this specification. See, e.g. “Principles of Neural Science”, McGraw-Hill Medical. New York, N.Y. (2000); Motulsky, “Intuitive Biostatistics”, Oxford University Press, Inc. (1995); Lodish et al., “Molecular Cell Biology, 4th ed.”, W. H. Freeman & Co. New York (2000); Griffiths et al., “Introduction to Genetic Analysis, 7th ed.”, W. H. Freeman & Co., N.Y. (1999): Gilbert et al., “Developmental Biology, 6th ed.”. Sinauer Associates, Inc., Sunderland, Mass. (2000).
  • Chemistry terms used herein are used according to conventional usage in the art, as exemplified by “The McGraw-Hill Dictionary of Chemical Terms”, Parker S., Ed., McGraw-Hill, San Francisco, C.A. (1985).
  • All of the above, and any other publications, patents and published patent applications referred to in this application are specifically incorporated by reference herein. In case of conflict, the present specification, including its specific definitions, will control.
  • Throughout this specification, the word “comprise” or variations such as “comprises” or “comprising” will be understood to imply the inclusion of a stated integer (or components) or group of integers (or components), but not the exclusion of any other integer (or components) or group of integers (or components).
  • The singular forms “a,” “an,” and “the” include the plurals unless the context clearly dictates otherwise.
  • The term “including” is used to mean “including but not limited to”. “Including” and “including but not limited to” are used interchangeably.
  • The term “agent” is used herein to denote a chemical compound (such as an organic or inorganic compound, a mixture of chemical compounds), a biological macromolecule (such as a nucleic acid, an antibody, including parts thereof as well as humanized, chimeric and human antibodies and monoclonal antibodies, a protein or portion thereof, e.g., a peptide, a lipid, a carbohydrate), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues. Agents include, for example, agents which are known with respect to structure, and those which are not known with respect to structure.
  • A “patient”, “subject”, or “individual” are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (including bovines, porcines, etc.), companion animals (e.g., canines, felines, etc.) and rodents (e.g., mice and rats).
  • “Cognitive function” or “cognitive status” refers to any higher order intellectual brain process or brain state, respectively, involved in learning and/or memory including, but not limited to, attention, information acquisition, information processing, working memory, short-term memory, long-term memory, anterograde memory, retrograde memory, memory retrieval, discrimination learning, decision-making, inhibitory response control, attentional set-shifting, delayed reinforcement learning, reversal learning, the temporal integration of voluntary behavior, expressing an interest in one's surroundings and self-care, speed of processing, reasoning and problem solving and social cognition.
  • In humans, cognitive function may be measured, for example and without limitation, by the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Selective Reminding Test (Buschke and Fuld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest; the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997); the Benton Visual Retention Test, or the explicit 3-alternative forced choice task, or MATRICS consensus neuropsychological test battery. See Folstein et al., J Psychiatric Res 12: 189-98, (1975); Robbins et al., Dementia 5: 266-81, (1994); Rey, L'cxamen clinique en psychologie, (1964); Kluger et al., J Geriatr Psychiatry Neurol 12:168-79. (1999): Marquis et al., 2002 and Masur et al., 1994. Also see Buchanan, R. W., Keefe, R. S. E., Umbricht, D., Green, M. F., Laughren, T., and Marder, S. R. (2011), The FDA-NIMH-MATRICS guidelines for clinical trial design of cognitive-enhancing drugs: what do we know 5 years later?Schizophr. Bull. 37, 1209-1217.
  • In animal model systems, cognitive function may be measured in various conventional ways known in the art, including using a Morris Water Maze (MWM), Barnes circular maze, elevated radial arm maze, T maze or any other mazes in which the animals use spatial information. Cognitive function can be assessed by reversal learning, extradimensional set shifting, conditional discrimination learning and assessments of reward expectancy. Other tests known in the art may also be used to assess cognitive function, such as novel object recognition and odor recognition tasks.
  • Cognitive function may also be measured using imaging techniques such as Positron Emission Tomography (PET), functional magnetic resonance imaging (fMRI), Single Photon Emission Computed Tomography (SPECT), or any other imaging technique that allows one to measure brain function. In animals, cognitive function may also be measured with electrophysiological techniques.
  • “Promoting” cognitive function refers to affecting impaired cognitive function so that it more closely resembles the function of a normal, unimpaired subject. Cognitive function may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject.
  • “Preserving” cognitive function refers to affecting normal or impaired cognitive function such that it does not decline or does not fall below that observed in the subject upon first presentation or diagnosis, or delays such decline.
  • “Improving” cognitive function includes promoting cognitive function and/or preserving cognitive function in a subject.
  • “Cognitive impairment” refers to cognitive function in subjects that is not as robust as that expected in a normal, unimpaired subject. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in a normal, unimpaired subject. In other cases, “cognitive impairment” in subjects affected by schizophrenia or bipolar disorder (in particular, mania) refers to cognitive function in subjects that is not as robust as that expected in normal, unimpaired subject.
  • “Schizophrenia” refers to a chronic debilitating disorder, characterized by a spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g. hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalities in the brain are proposed to underlie the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely ineffective in treating cognitive impairments in patients.
  • “Bipolar disorder” or “BP” or “manic depressive disorder” or “manic depressive illness” refers to a chronic psychological/mood disorder which can be characterized by significant mood changes including periods of depression and euphoric manic periods. BP may be diagnosed by a skilled physician based on personal and medical history, interview consultation and physical examinations. The term “mania” or “manic periods” or other variants refers to periods where an individual exhibits some or all of the following characteristics: racing thoughts, rapid speech, elevated levels of activity and agitation as well as an inflated sense of self-esteem, euphoria, poor judgment, insomnia, impaired concentration and aggression.
  • “Treating” a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results. Beneficial or desired clinical results include, but are not limited to, preventing or slowing the progression of the disease or disorder, or alleviation, amelioration, or slowing the progression, of one or more symptoms associated with CNS disorders with cognitive impairment, such as schizophrenia or bipolar disorder (in particular, mania).
  • “Treating cognitive impairment” refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject's performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline. Preferably, that subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. Treatment of cognitive impairment in humans may improve cognitive function to any detectable degree, but is preferably improved sufficiently to allow the impaired subject to carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject. In some cases. “treating cognitive impairment” refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject's performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline. Preferably, that subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. In some cases, “treating cognitive impairment” in a subject affecting by schizophrenia or bipolar disorder (in particular, mania) refers to takings steps to improve cognitive function in the subject so that the subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject.
  • “Administering” or “administration of” a substance, a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art. For example, a compound or an agent can be administered, intravenously, arterially, intradermally, intramuscularly, intraperitonealy, intravenously, subcutaneously, ocularly, sublingually, orally (by ingestion), intranasally (by inhalation), intraspinally, intracerebrally, and transdermally (by absorption, e.g. through a skin duct). A compound or agent can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, or controlled release of the compound or agent. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. In some aspects, the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug. For example, as used herein, a physician who instructs a patient to self-administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is administering the drug to the patient.
  • Appropriate methods of administering a substance, a compound or an agent to a subject will also depend, for example, on the age of the subject, whether the subject is active or inactive at the time of administering, whether the subject is cognitively impaired at the time of administering, the extent of the impairment, and the chemical and biological properties of the compound or agent (e.g. solubility, digestibility, bioavailability, stability and toxicity). In some embodiments, a compound or an agent is administered orally, e.g., to a subject by ingestion, or intravenously. e.g., to a subject by injection. In some embodiments, the orally administered compound or agent is in an extended release or slow release formulation, or administered using a device for such slow or extended release.
  • “SV2A inhibitor” refers to any agent, substance or compound that binds to SV2A and reduces synaptic function by reducing pre-synaptic vesicle release (See, e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006; Custer et al., 2006; Smedt et al., 2007; Yang et al., 2007; Meehan, “Levetiracetam has an activity-dependent effect on inhibitory transmission,” Epilepsia, 2012 Jan. 31; and Example 8 of WO 2001/62726, all of which are specifically incorporated herein by reference.) A substance, or a compound or an agent is an SV2A inhibitor even if it does not itself bind to SV2A, as long as it causes, or affects the ability of, another compound or agent to bind SV2A or reduce synaptic function by reducing pre-synaptic vesicle release. SV2A inhibitors, as used herein, include pharmaceutically acceptable salts of the inhibitors thereof. They also include hydrates, polymorphs, prodrugs, salts, and solvates of these inhibitors.
  • “Antipsychotic”, “antipsychotic agent”, “antipsychotic drug”, or “antipsychotic compound” refers to (1) a typical or an atypical antipsychotic; (2) an agent that is selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CBI antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, nNOS inhibits, neurosteroids, and neurotrophic factors; and/or (3) an agent that is useful in treating one or more signs or symptoms of schizophrenia or bipolar disorder (in particular, mania).
  • “Typical antipsychotics”, as used herein, refer to conventional antipsychotics, which produce antipsychotic effects as well as movement related adverse effects related to disturbances in the nigrostriatal dopamine system. These extrapyramidal side effects (EPS) include Parkinsonism, akathisia, tardive dyskinesia and dystonia. See Baldessarini and Tarazi in Goodman & Gilman's The Pharmacological Basis of Therapeutics 10 Edition, 2001, pp. 485-520.
  • “Atypical antipsychotics”, as used herein, refer to antipsychotic drugs that produce antipsychotic effects with little or no EPS and include, but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone. “Atypical” antipsychotics differ from conventional antipsychotics in their pharmacological profiles. While conventional antipsychotics are characterized principally by D2 dopamine receptor blockade, atypical antipsychotics show antagonist effects on multiple receptors including the 5HTa and 5HTc serotonin receptors and varying degrees of receptor affinities. Atypical antipsychotic drugs are commonly referred to as serotoninidopamine antagonists, reflecting the influential hypothesis that greater affinity for the 5-HTT2 receptor than for the D2 receptor underlies “atypical” antipsychotic drug action or “second generation” antipsychotic drugs. However, the atypical antipsychotics often display side effects, including, but not limited to, weight gain, diabetes (e.g., type II diabetes mellitus), hyperlipidemia, QTc interval prolongation, myocarditis, sexual side effects, extrapyramidal side effects and cataract. Thus, atypical antipsychotics do not represent a homogeneous class, given their differences in the context of both alleviation of clinical symptoms and their potential for inducing side effects such as the ones listed above. Further, the common side effects of the atypical antipsychotics as described above often limit the antipsychotic doses that can be used for these agents.
  • The term “simultaneous administration,” as used herein, means that the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs, are administered with a time separation of no more than about 15 minutes, and in some embodiments no more than about 10 minutes. When the drugs are administered simultaneously, the SV2A inhibitor and the antipsychotic, or their salts, hydrates, solvates, or polymorphs, may be contained in the same dosage (e.g., a unit dosage form comprising both the SV2A inhibitor and the antipsychotic) or in discrete dosages (e.g., the SV2A inhibitor or its salt, hydrate, solvate, or polymorph is contained in one dosage form and the antipsychotic or its salt, hydrate, solvate, or polymorph is contained in another dosage form).
  • The term “sequential administration” as used herein means that the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, are administered with a time separation of more than about 15 minutes, and in some embodiments more than about one hour, or up to 12-24 hours. Either the SV2A inhibitor or the antipsychotic may be administered first. The SV2A inhibitor and the antipsychotic, or their salts, hydrates, solvents, or polymorphs, for sequential administration may be contained in discrete dosage forms, optionally contained in the same container or package.
  • A “therapeutically effective amount” of a drug or agent is an amount of a drug or an agent that, when administered to a subject will have the intended therapeutic effect. e.g. improving cognitive function in a subject in a subject suffering from a disease or disorder (e.g., schizophrenia or bipolar disorder (in particular, mania)), preventing or slowing the progression of a disease or disorder (e.g., schizophrenia or bipolar disorder (in particular, mania)), and/or alleviating, ameliorating, or slowing the progression of one or more symptoms associated with the disease or disorder (e.g., schizophrenia or bipolar disorder (in particular, mania)). The full therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a therapeutically effective amount may be administered in one or more administrations. The precise effective amount needed for a subject will depend upon, for example, the subject's size, health and age, the nature and extent of the cognitive impairment, and the therapeutics or combination of therapeutics selected for administration, and the mode of administration. The skilled worker can readily determine the effective amount for a given situation by routine experimentation.
  • “Subtherapeutic amount” refers to an amount administered of an agent or compound of the invention that is less than the therapeutic amount, that is, less than the amount normally used when said agent or compound is administered alone (i.e., individually and in the absence of other therapeutic agents or compounds) to treat disorders, such as schizophrenia or bipolar disorder (in particular, mania).
  • “Analog” is used herein to refer to a compound which functionally resembles another chemical entity, but does not share the identical chemical structure. For example, an analog is sufficiently similar to a base or parent compound such that it can substitute for the base compound in therapeutic applications, despite minor structural differences.
  • “Derivative” is used herein to refer to the chemical modification of a compound. Chemical modifications of a compound can include, for example, replacement of hydrogen by an alkyl, acyl, or amino group. Many other modifications are also possible.
  • The term “prodrug” is art-recognized and is intended to encompass compounds or agents which, under physiological conditions, are converted into a SV2A inhibitor or an antipsychotic. A common method for making a prodrug is to select moieties which are hydrolyzed or metabolized under physiological conditions to provide the desired compound or agent. In other embodiments, the prodrug is converted by, for example, an enzymatic activity of the host animal to a SV2A inhibitor or an antipsychotic.
  • The term “aliphatic” as used herein means a straight chained or branched alkyl, alkenyl or alkynyl. It is understood that alkenyl or alkynyl embodiments need at least two carbon atoms in the aliphatic chain. Aliphatic groups typically contains from 1 (or 2) to 12 carbons, such as from 1 (or 2) to 4 carbons.
  • The term “aryl” as used herein means a monocyclic or bicyclic carbocyclic aromatic ring system. For example, aryl as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic carbocyclic aromatic ring system. Phenyl is an example of a monocyclic aromatic ring system. Bicyclic aromatic ring systems include systems wherein both rings are aromatic, e.g., naphthyl, and systems wherein only one of the two rings is aromatic, e.g., tetralin.
  • The term “heterocyclic” as used herein means a monocyclic or bicyclic non-aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH, S, SO, or SO2 in a chemically stable arrangement. For example, heterocyclic as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic non-aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH, S, SO, or SO2 in a chemically stable arrangement. In a bicyclic non-aromatic ring system embodiment of “heterocyclyl”, one or both rings may contain said heteroatom or heteroatom groups. In another bicyclic “heterocyclyl” embodiment, one of the two rings may be aromatic. In yet another heterocyclic ring system embodiment, a non-aromatic heterocyclic ring may optionally be fused to an aromatic carbocycle.
  • Examples of heterocyclic rings include 3-1H-benzimidazol-2-one, 3-(1-alkyl)-benzimidazol-2-one, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholino, 3-morpholino. 4-morpholino, 2-thiomorpholino, 3-thiomorpholino, 4-thiomorpholino, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5-pyrazolinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2-thiazolidinyl, 3-thiazolidinyl, 4-thiazolidinyl, 1-imidazolidinyl, 2-imidazolidinyl, 4-imidazolidinyl, 5-imidazolidinyl, indolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, benzothiolane, benzodithiane, and 1,3-dihydro-imidazol-2-one.
  • The term “heteroaryl” as used herein means a monocyclic or bicyclic aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH or S in a chemically stable arrangement. For example, heteroaryl as used herein can be a C5-C10 monocyclic or C8-C12 bicyclic aromatic ring system having 1 to 3 heteroatom or heteroatom groups in each ring selected from O, N, NH or S in a chemically stable arrangement. In such a bicyclic aromatic ring system embodiment of “heteroaryl”:
  • both rings are aromatic; and
  • one or both rings may contain said heteroatom or heteroatom groups.
  • Examples of heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, benzimidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl. 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-indolyl), pyrazolyl (e.g. 2-pyrazolyl), isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, purinyl, pyrazinyl, 1,3,5-triazinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl).
  • The term “cycloalkyl or cycloalkenyl” refers to a monocyclic or fused or bridged bicyclic carbocyclic ring system that is not aromatic. For example, cycloalkyl or cycloalkenyl as used herein can be a C5-C10 monocyclic or fused or bridged C8-C12 bicyclic carbocyclic ring system that is not aromatic. Cycloalkenyl rings have one or more units of unsaturation. Preferred cycloalkyl or cycloalkenyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, norbornyl, adamantyl and decalinyl.
  • As used herein, the carbon atom designations may have the indicated integer and any intervening integer. For example, the number of carbon atoms in a (C1-C4)-alkyl group is 1, 2, 3, or 4. It should be understood that these designation refer to the total number of atoms in the appropriate group. For example, in a (C3-C10)-heterocyclyl the total number of carbon atoms and heteroatoms is 3 (as in aziridine). 4, 5, 6 (as in morpholine), 7, 8, 9, or 10.
  • “Pharmaceutically acceptable salt” is used herein to refer to an agent or a compound according to the invention that is a therapeutically active, non-toxic base and acid salt form of the compounds. The acid addition salt form of a compound that occurs in its free form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclic, salicylic, p-aminosalicylic, pamoic and the like. See, e.g., WO 01/062726.
  • Compounds containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt form, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts. e.g., lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. Conversely, said salt forms can be converted into the free forms by treatment with an appropriate base or acid. Compounds and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like. See, e.g., WO 01/062726.
  • As used herein, the term “hydrate” refers to a combination of water with a compound wherein the water retains its molecular state as water and is either absorbed, adsorbed or contained within a crystal lattice of the substrate compound.
  • As used herein, the term “polymorph” refers to different crystalline forms of the same compound and other solid state molecular forms including pseudo-polymorphs, such as hydrates (e.g., bound water present in the crystalline structure) and solvates (e.g., bound solvents other than water) of the same compound. Different crystalline polymorphs have different crystal structures due to a different packing of the molecules in the lattice. This results in a different crystal symmetry and/or unit cell parameters which directly influences its physical properties such the X-ray diffraction characteristics of crystals or powders. A different polymorph, for example, will in general diffract at a different set of angles and will give different values for the intensities. Therefore X-ray powder diffraction can be used to identify different polymorphs, or a solid form that comprises more than one polymorph, in a reproducible and reliable way. Crystalline polymorphic forms are of interest to the pharmaceutical industry and especially to those involved in the development of suitable dosage forms. If the polymorphic form is not held constant during clinical or stability studies, the exact dosage form used or studied may not be comparable from one lot to another. It is also desirable to have processes for producing a compound with the selected polymorphic form in high purity when the compound is used in clinical studies or commercial products since Impurities present may produce undesired toxicological effects. Certain polymorphic forms may exhibit enhanced thermodynamic stability or may be more readily manufactured in high purity in large quantities, and thus are more suitable for inclusion in pharmaceutical formulations. Certain polymorphs may display other advantageous physical properties such as lack of hygroscopic tendencies, improved solubility, and enhanced rates of dissolution due to different lattice energies.
  • Many of the compounds useful in the methods and compositions of this invention have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem. (1976), 45, 11-30. The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds or mixtures thereof (including all possible mixtures of stereoisomers). See, e.g., WO 01/062726.
  • Furthermore, certain compounds which contain alkenyl groups may exist as Z (zusammen) or E (entgegen) isomers. In each instance, the invention includes both mixture and separate individual isomers. Multiple substituents on a piperidinyl or the azepanyl ring can also stand in either cis or trans relationship to each other with respect to the plane of the piperidinyl or the azepanyl ring. Some of the compounds may also exist in tautomeric forms. Such forms, although not explicitly indicated in the formulae described herein, are intended to be included within the scope of the present invention. With respect to the methods and compositions of the present invention, reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof unless the particular isomeric form is referred to specifically. See, e.g., WO 01/062726.
  • Description of Methods of the Invention
  • The methods of this invention comprise administration of a SV2A inhibitor or a pharmaceutically acceptable salt thereof in combination with administration of an antipsychotic or a pharmaceutically acceptable salt thereof. The agents or compounds of the SV2A inhibitor or the antipsychotic and their pharmaceutically acceptable salts also include hydrates, solvates, polymorphs, and prodrugs of those agents, compounds, and salts.
  • Methods of Assessing Cognitive Impairment
  • Animal models serve as an important resource for developing and evaluating treatments for CNS disorders with cognitive impairment. Features that characterize cognitive impairment in animal models typically extend to cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be predictive of efficacy in humans. The extent of cognitive impairment in an animal model for a CNS disorder, and the efficacy of a method of treatment for said CNS disorder may be tested and confirmed with the use of a variety of cognitive tests.
  • A Radial Arm Maze (RAM) behavioral task is one example of a cognitive test, specifically testing spacial memory (Chappell et al. Neuropharmacology 37: 481-487, 1998). The RAM apparatus consists of, e.g., eight equidistantly spaced arms. A maze arm projects from each facet of a center platform. A food well is located at the distal end of each arm. Food is used as a reward. Blocks can be positioned to prevent entry to any arm. Numerous extra maze cues surrounding the apparatus may also be provided. After habituation and training phases, spatial memory of the subjects may be tested in the RAM under control or test compound-treated conditions. As a part of the test, subjects are pretreated before trials with a vehicle control or one of a range of dosages of the test compound. At the beginning of each trial, a subset of the arms of the eight-arm maze is blocked. Subjects are allowed to obtain food on the unblocked arms to which access is permitted during this initial “information phase” of the trial. Subjects are then removed from the maze for a delay period, e.g., a 60 second delay, a 15 minute delay, a one-hour delay, a two-hour delay, a six hour delay, a 24 hour delay, or longer) between the information phase and the subsequent “retention test,” during which the barriers on the maze are removed, thus allowing access to all eight arms. After the delay period, subjects are placed back onto the center platform (with the barriers to the previously blocked arms removed) and allowed to obtain the remaining food rewards during this retention test phase of the trial. The identity and configuration of the blocked arms vary across trials. The number of “errors” the subjects make during the retention test phase is tracked. An error occurs in the trial if the subjects entered an arm from which food had already been retrieved in the pre-delay component of the trial, or if it re-visits an arm in the post-delay session that had already been visited. A fewer number of errors would indicate better spatial memory. The number of errors made by the test subject, under various test compound treatment regimes, can then be compared for efficacy of the test compound in treating CNS disorders with cognitive impairment.
  • Another cognitive test that may be used to assess the effects of a test compound on the cognitive impairment of a CNS disorder model animal is the Morris water maze. A water maze is a pool surrounded with a novel set of patterns relative to the maze. The training protocol for the water maze may be based on a modified water maze task that has been shown to be hippocampal-dependent (de Hoz et al., Eur. J. Neurosci., 22:745-54, 2005; Steele and Morris, Hippocampus 9:118-36, 1999). The subject is trained to locate a submerged escape platform hidden underneath the surface of the pool. During the training trial, a subject is released in the maze (pool) from random starting positions around the perimeter of the pool. The starting position varies from trial to trial. If the subject does not locate the escape platform within a set time, the experimenter guides and places the subject on the platform to “teach” the location of the platform. After a delay period following the last training trial, a retention test in the absence of the escape platform is given to assess spatial memory. The subject's level of preference for the location of the (now absent) escape platform, as measured by, e.g., the time spent in that location or the number of crossings of that location made by the mouse, indicates better spatial memory, i.e., treatment of cognitive impairment. The preference for the location of the escape platform under different treatment conditions, can then be compared for efficacy of the test compound in treating CNS disorders with cognitive impairment.
  • There are various tests known in the art for assessing cognitive function in humans, for example and without limitation, the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Selective Reminding Test (Buschke and Fuld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest; the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997); the Benton Visual Retention Test, or MATRICS consensus neuropsychological test battery which includes tests of working memory, speed of processing, attention, verbal learning, visual learning, reasoning and problem solving and social cognition. See Folstein et al., J Psychiatric Res 12: 189-98, (1975): Robbins et al., Dementia 5: 266-81, (1994); Rey, L'examen clinique en psychologie, (1964); Kluger et al., J Geriatr Psychiatry Neurol 12:168-79, (1999); Marquis et al., 2002 and Masur et al., 1994. Also see Buchanan, R. W., Keefe, R. S. E., Umbricht, D., Green, M. F., Laughren, T., and Marder, S. R. (2011) The FDA-NIMH-MATRICS guidelines for clinical trial design of cognitive-enhancing drugs: what do we know 5 years later?Schizophr. Bull. 37, 1209-1217. Another example of a cognitive test in humans is the explicit 3-alternative forced choice task. In this test, subjects are presented with color photographs of common objects consisting of a mix of three types of image pairs: similar pairs, identical pairs and unrelated foils. The second of the pair of similar objects is referred to as the “lure”. These image pairs are fully randomized and presented individually as a series of images. Subjects are instructed to make a judgment as to whether the objects seen are new, old or similar. A “similar” response to the presentation of a lure stimulus indicates successful memory retrieval by the subject. By contrast, calling the lure stimulus “old” or “new” indicates that correct memory retrieval did not occur. Schizophrenia
  • This invention provides methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania) using a SV2A inhibitor or a pharmaceutically acceptable salt thereof in combination with an antipsychotic or a pharmaceutically acceptable salt thereof. In certain embodiments, treatment comprises preventing or slowing the progression of schizophrenia or bipolar disorder (in particular, mania). Schizophrenia is characterized by a wide spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g., hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression of one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia. Further, there are a number of other psychiatric diseases such as schizotypical and schizoaffective disorder, other acute- and chronic psychoses and bipolar disorder (in particular, mania), which have an overlapping symptomatology with schizophrenia. In some embodiments, treatment comprises alleviation, amelioration or slowing the progression of one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). The methods and compositions may be used for human patients in clinical applications in treating schizophrenia or bipolar disorder (in particular, mania). The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
  • Cognitive impairments are associated with schizophrenia. They precede the onset of psychosis and are present in non-affected relatives. The cognitive impairments associated with schizophrenia constitute a good predictor for functional outcome and are a core feature of the disorder. Cognitive features in schizophrenia reflect dysfunction in frontal cortical and hippocampal circuits. Patients with schizophrenia also present hippocampal pathologies such as reductions in hippocampal volume, reductions in neuronal size and dysfunctional hyperactivity. An imbalance in excitation and inhibition in these brain regions has also been documented in schizophrenic patients suggesting that drugs targeting inhibitory mechanisms could be therapeutic. See, e.g., Guidotti et al., Psychopharmacology 180: 191-205, 2005; Zierhut. Psych. Res. Neuroimag. 183:187-194, 2010; Wood et al., Neurolmage 52:62-63, 2010; Vinkers et al., Expert Opin. Investig. Drugs 19:1217-1233, 2009; Young et al., Pharmacol. Ther. 122:150-202, 2009.
  • Animal models serve as an important resource for developing and evaluating treatments for schizophrenia. Features that characterize schizophrenia in animal models typically extend to schizophrenia in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of schizophrenia are known in the art.
  • One animal model of schizophrenia is protracted treatment with methionine. Methionine-treated mice exhibit deficient expression of GAD67 in frontal cortex and hippocampus, similar to those reported in the brain of postmortem schizophrenia patients. They also exhibit prepulse inhibition of startle and social interaction deficits (Tremonlizzo et al., PNAS, 99: 17095-17100, 2002). Another animal model of schizophrenia is methylaoxymethanol acetate (MAM)-treatment in rats. Pregnant female rats are administered MAM (20 mg/kg, intraperitoneal) on gestational day 17. MAM-treatment recapitulate a pathodevelopmental process to schizophrenia-like phenotypes in the offspring, including anatomical changes, behavioral deficits and altered neuronal information processing. More specifically, MAM-treated rats display a decreased density of parvalbumin-positive GABAergic interneurons in portions of the prefrontal cortex and hippocampus. In behavioral tests, MAM-treated rats display reduced latent inhibition. Latent inhibition is a behavioral phenomenon where there is reduced learning about a stimulus to which there has been prior exposure with any consequence. This tendency to disregard previously benign stimuli, and reduce the formation of association with such stimuli is believed to prevent sensory overload. Low latent inhibition is indicative of psychosis. Latent inhibition may be tested in rats in the following manner. Rats are divided into two groups. One group is pre-exposed to a tone over multiple trials. The other group has no tone presentation. Both groups are then exposed to an auditory fear conditioning procedure, in which the same tone is presented concurrently with a noxious stimulus, e.g. an electric shock to the foot. Subsequently, both groups are presented with the tone, and the rats' change in locomotor activity during tone presentation is monitored. After the fear conditioning the rats respond to the tone presentation by strongly reducing locomotor activity. However, the group that has been exposed to the tone before the conditioning period displays robust latent inhibition: the suppression of locomotor activity in response to tone presentation is reduced. MAM-treated rats, by contrast show impaired latent inhibition. That is, exposure to the tone previous to the fear conditioning procedure has no significant effect in suppressing the fear conditioning. (see Lodge et al., J. Neurosci., 29:2344-2354, 2009) Such animal models of schizophrenia may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
  • MAM-treated rats display a significantly enhanced locomotor response (or aberrant locomotor activity) to low dose D-amphetamine administration. The MAM-treated rats also display a significantly greater number of spontaneously firing ventral tegmental dopamine (DA) neurons. These results are believed to be a consequence of excessive hippocampal activity because in MAM-treated rats, the ventral hippocampus (vHipp) inactivation (e.g., by intra-vHipp administration of a sodium channel blocker, tetrodotoxin (TTX), to MAM rats) completely reversed the elevated DA neuron population activity and also normalized the augmented amphetamine-induced locomotor behavior. The correlation of hippocampal dysfunction and the hyper-responsivity of the DA system is believed to underlie the augmented response to amphetamine in MAM-treated animals and psychosis in schizophrenia patients. See Lodge D. J. et al. Neurobiology of Disease (2007), 27(42), 11424-11430. The use of MAM-treated rats in the above study may be suitable for use to assay the effectiveness of the methods and compositions of the present invention in treating schizophrenia or bipolar disorder (in particular, mania). For example, the methods and compositions of this invention maybe evaluated, using MAM-treated animals, for their effects on the central hippocampus (vHipp) regulation, on the elevated DA neuron population activity and on the hyperactive locomotor response to amphetamine in the MAM-treated animals.
  • In MAM-treated rats, hippocampal (HPC) dysfunction leads to dopamine system hyperactivity. A benzodiazepine-positive allosteric modulator (PAM), selective for the α5 subunit of the GABAA receptor, SH-053-2′F—R—CH3, is tested for its effects on the output of the hippocampal (HPC). The effect of SH-053-2′F—R—CH3 on the hyperactive locomotor response to amphetamine in MAM-treated animals is also examined. The α5GABAAR PAM reduces the number of spontaneously active DA neurons in the ventral tegmental area (VTA) of MAM rats to levels observed in saline-treated rats (control group), both when administered systemically and when directly infused into the ventral HPC. Moreover, HPC neurons in both saline-treated and MAM-treated animals show diminished cortical-evoked responses following the α5GABAAR PAM treatment. In addition, the increased locomotor response to amphetamine observed in MAM-treated rats is reduced following the α5GABAAR PAM treatment. See Gill K. M et al. Neuropsychopharmacology (2011), 1-9. The use of MAM-treated rats in the above study may be suitable for use in the present invention to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania). For example, the methods and compositions of this invention maybe evaluated, using MAM-treated animals, for their effects on the output of the hippocampal (HPC) and on the hyperactive locomotor response to amphetamine in the MAM-treated animals.
  • Administration of MAM to pregnant rats on embryonic day 15 (E15) severely impairs spatial memory or the ability to learn the spatial location of four items on an eight-arm radial maze in the offspring. In addition, embryonic day 17 (E17) MAM-treated rats are able to reach the level of performance of control rats at the initial stages of training, but are unable to process and retrieve spatial information when a 30-min delay is interposed, indicating a significant impairment in working memory. See Gourevitch R. et al. (2004). Behav. Pharmacol, 15, 287-292. Such animal models of schizophrenia may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
  • Apomorphine-induced climbing (AIC) and stereotype (AIS) in mice is another animal model useful in this invention. Agents are administered to mice at a desired dose level (e.g., via intraperitoneal administration). Subsequently, e.g., thirty minutes later, experimental mice are challenges with apomorphine (e.g., with 1 mg/kg sc). Five minutes after the apomorphine injection, the sniffing-licking-gnawing syndrome (stereotyped behavior) and climbing behavior induced by apomorphine are scored and recorded for each animal. Readings can be repeated every 5 min during a 30-min test session. Scores for each animal are totaled over the 30-min test session for each syndrome (stereotyped behavior and climbing). If an effect reached at least of 50% inhibition, and ID50 value (95% confidence interval) is calculated using a nonlinear least squares calculation with inverse prediction. Mean climbing and stereotype scores can be expressed as a percent of control values observed in vehible treated (e.g., saline-treated) mice that receive apomorphine. See Grauer S. M. et al. Psychopharmacology (2009) 204, 37-48. This mice model may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
  • The efficacy of the methods and compositions of this invention in treating schizophrenia may also be assessed in animal models of schizophrenia or bipolar disorder (in particular, mania), as well as human subjects with schizophrenia, using a variety of cognitive tests known in the art, as discussed above. SV2A inhibitors
  • “Synaptic vesicle protein-2 (SV2)” is a family of synaptic vesicle proteins, which consists of three members, designated SV2A, SV2B, and SV2C. SV2A is the most widely distributed family member, being expressed ubiquitously in the brain. The proteins are integral membrane proteins and have a low-level homology (20-30%) to the twelve transmembrane family of bacterial and fungal transporter proteins that transport sugar, citrate, and xenobiotics (Bajjalieh et al., Science. 257: 1271-1273. (1992)). SV2 family proteins are present in the brain and endocrine cells, and further are present in all synaptic and endocrine vesicles. SV2 proteins are reported to play a role in normal synaptic function, and functions in a maturation step of primed vesicles that converts the vesicles into a Cat(2+)- and synaptotagmin-responsive state (Sudhof et al., 2009). Functionally, SV2 proteins are reported to enhance synaptic currents and increase the probability of transmitter release by maintaining the size of the readily releasable pool of vesicles (Custer et al., 2006).
  • “SV2A inhibitor” refers to any agent, substance or compound that binds to SV2A and reduces synaptic function by reducing pre-synaptic vesicle release (See, e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006; Custer et al., 2006; Smedt et al., 2007; Yang et al., 2007; Meehan, “Levetiracetam has an activity-dependent effect on inhibitory transmission,” Epilepsia, 2012 Jan. 31; and Example 8 of WO 2001/62726, all of which are specifically incorporated herein by reference.) A substance, or a compound or an agent is an SV2A inhibitor even if it does not itself bind to SV2A, as long as it causes, or affects the ability of, another compound or agent to bind SV2A or reduce synaptic function by reducing pre-synaptic vesicle release. SV2A inhibitors, as used herein, include pharmaceutically acceptable salts of the inhibitors thereof. They also include hydrates, polymorphs, prodrugs, salts, and solvates of these inhibitors.
  • Among the SV2A inhibitors or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof that are useful in the methods and compositions of this invention are those disclosed, for example, U.S. patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, and U.S. Patent Application 61/175,536. However, any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of the invention. In some embodiments, the SV2A inhibitor is selected from the group of SV2A inhibitors referred to in International Patent Applications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692 or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs. Other SV2A inhibitors may also be used in this invention. Applicants also refer to methods of preparing these compounds found in the documents cited above. Other synthetic methods may also be used. These methods are well known to those skilled in the art.
  • In some embodiments of this invention, the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, or prodrugs thereof.
  • In some embodiments of this invention, the SV2A inhibitor is levetiracetam or salts, solvates, hydrates, polymorphs or prodrugs thereof. Levetiracetam refers to the International Union of Pure and Applied Chemistry (IUPAC) name of the compound (2S)-2-(2-oxopyrrolidin-1-yl) butanamide). Levetiracetam is a widely used antiepileptic drug. Levetiracetam binds to a specific site in the CNS: the synaptic vesicle protein 2A (SV2A) (See. e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006) and has further been shown to directly inhibit synaptic activity and neurotransmission by inhibiting presynaptic neurotransmitter release (Yang et al. 2007).
  • Among the SV2A inhibitors useful for the methods and compositions of this invention are the following:
  • i) International Patent Application WO 2001/062726:
  • A compound having the formula I or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00001
  • wherein X is —CA1NR5R6 or —CA1OR7 or —CA1-R8 or CN;
  • A1 and A2 are independently oxygen, sulfur or —NR9;
  • R1 is hydrogen, alkyl, aryl or —CH2—R1a wherein R1a is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
  • R2, R3 and R4 are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative:
  • R2a, R3a and R4a are the same or different and each is independently hydrogen, halogen, alkyl, alkenyl, alkynyl or aryl;
      • R5, R6, R7 and R9 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or an oxy derivative; and
  • R8 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative;
  • with the provisos that at least one of as R2, R3, R4, R2, R3a and R4a is other than hydrogen; and that when the compound is a mixture of all possible isomers, X is —CONR5R6, A2 is oxygen and R1 is hydrogen, methyl, ethyl or propyl then substitution on the pyrollidine ring is other than mono-, di-, or tri-methyl or mono-ethyl; and that when R1, R2, R4, R2a, R3a and R4a are each hydrogen, A2 is oxygen and X is CONR5R6 then R3 is different from carboxy, ester, amido, substituted oxo-pyrrolidine, hydroxy, oxy derivative, amino, amino derivatives, methyl, naphthyl, phenyl optionally substituted by oxy derivatives or in the para position by an halogen atom.
  • In the definitions set forth below, unless otherwise stated, R11 and R12 are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfinyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.
  • The term “oxy derivative”, as used herein is defined as including —O—R11 groups wherein R11 is as defined above except for “oxy derivative”. Non-limiting examples are alkoxy, alkenyloxy, alkynyloxy, acyloxy, oxyester, oxyamido, alkylsulfonyloxy, alkylsulfinyloxy, arylsulfonyloxy, arylsulfinyloxy, aryloxy, aralkoxy or heterocyclooxy such as pentyloxy, allyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methylenedioxy, carbonate.
  • The term “thio derivative” as used herein, is defined as including —S—R11 groups wherein R11 is as defined above except for “thio derivative”. Non-limiting examples are alkylthio, alkenylthio, alkynylthio and arylthio.
  • The term “amino derivative” as used herein, is defined as including —NHR11 or —NR11R12 groups wherein R11 and R12 are as defined above. Non-limiting examples are mono- or di-alkyl-, alkenyl-, alkynyl- and arylamino or mixed amino.
  • The term “acyl derivative” as used herein, represents a radical derived from carboxylic acid and thus is defined as including groups of the formula R11—CO—, wherein R11 is as defined above and may also be hydrogen. Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl, heptanedioyl, cyclohexanecarbonyl, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxalyl, ethoxalyl, cysteinyl, oxamoyl.
  • The term “sulfonyl derivative” as used herein, is defined as including a group of the formula —SO2—R11, wherein R11 is as defined above except for “sulfonyl derivative”. Non-limiting examples are alkylsulfonyl, alkenylsulfonyl, alkynylsulfonyl and arylsulfonyl.
  • The term “sulfinyl derivative” as used herein, is defined as including a group of the formula —SO—R11, wherein R11 is as defined above except for “sulfinyl derivative”. Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl and arylsulfinyl.
  • The term “alkyl”, as used herein, is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and containing 1-20 carbon atoms, preferably 1-6 carbon atoms for non-cyclic alkyl and 3-6 carbon atoms for cycloalkyl (in these two preferred cases, unless otherwise specified, “lower alkyl”). Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cycloalkyl, sulfonic acid, sulfonamide, thio derivative, oxyester, oxyamido, heterocycle, vinyl, C1-5-alkoxy, C6-10-aryloxy and C6-10-aryl.
  • Preferred alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, and 2,2,2-trimethylethyl each optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro and cyano, such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • The term “alkenyl” as used herein, is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond such as ethenyl (=vinyl), 1-methyl-1-ethenyl, 2,2-dimethyl-1-ethenyl, 1-propenyl, 2-propenyl (=allyl), 1-butenyl, 2-butenyl, 3-butenyl, 4-pentenyl, 1-methyl-4-pentenyl, 3-methyl-1-pentenyl, 1-hexenyl, 2-hexenyl, and the like and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl and heterocycle such as mono- and di-halo vinyl where halo is fluoro, chloro or bromo.
  • The term “alkynyl” as used herein, is defined as including a monovalent branched or unbranched hydrocarbon radical containing at least one carbon-carbon triple bond, for example ethynyl, 2-propynyl (=propargyl), and the like and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl and heterocycle, such as haloethynyl.
  • When present as bridging groups, alkyl, alkenyl and alkynyl represent straight- or branched chains, C1-12, preferably C1-4-alkylene or C2-12-, preferably C2-4-alkenylene or -alkynylene moieties respectively.
  • Groups where branched derivatives are conventionally qualified by prefixes such as “n”, “sec”, “iso” and the like (e.g., “n-propyl”, “sec-butyl”) are in the n-form unless otherwise stated.
  • The term “aryl” as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of 1-3 rings and containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfinyl, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, alkylthio, oxyester, oxyamido, aryl, C1-6-alkoxy, C6-10-aryloxy, C1-6-alkyl, C1-6-haloalkyl. Aryl radicals are preferably monocyclic containing 6-10 carbon atoms. Preferred aryl groups are phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, nitro, amino, azido, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkyl, C1-6-haloalkyl and phenyl.
  • The term “halogen”, as used herein, includes an atom of Cl, Br, F, I.
  • The term “hydroxy”, as used herein, represents a group of the formula —OH.
  • The term “thiol”, as used herein, represents a group of the formula —SH.
  • The term “cyano”, as used herein, represents a group of the formula —CN.
  • The term “nitro”, as used herein, represents a group of the formula —NO2.
  • The term “nitrooxy”, as used herein, represents a group of the formula —ONO2.
  • The term “amino”, as used herein, represents a group of the formula —NH2.
  • The term “azido”, as used herein, represents a group of the formula —N3.
  • The term “carboxy”, as used herein, represents a group of the formula —COOH.
  • The term “sulfonic acid”, as used herein, represents a group of the formula —SO3H.
  • The term “sulfonamide”, as used herein, represents a group of the formula —SO2NH2.
  • The term “ester”, as used herein is defined as including a group of formula —COO—R11 wherein R11 is as defined above except oxy derivative, thio derivative or amino derivative.
  • The term “ether” is defined as including a group selected from C1-50-straight or branched alkyl, or C2-50-straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.
  • The term “amido” is defined as including a group of formula —CONH2 or —CONHR11 or —CONR11R12 wherein R11 and R12 are as defined above.
  • The term “heterocycle”, as used herein is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl. Non-limiting examples of aromatic heterocycles are pyridyl, furyl, pyrrolyl, thienyl, isothiazolyl, imidazolyl, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, pyrazolyl, indolyl, indolizinyl, purinyl, isoindolyl, carbazolyl, thiazolyl, 1,2,4-thiadiazolyl, thieno (2,3-b) furanyl, furopyranyl, benzofuranyl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl, benzothiazolyl, or benzoxazolyl, cinnolinyl, phthalazinyl, quinoxalinyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenothiazinyl, furazanyl, isochromanyl, indolinyl, xanthenyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolopyrimidinyl optionally substituted by alkyl or as described above for the alkyl groups. Non-limiting examples of non aromatic heterocycles are tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i.e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) or the same which can optionally be substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups. The term “heterocycle” also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1)heptanyl, 7- oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1)octanyl.
  • In the above definitions it is to be understood that when a substituent such as R2, R3, R4, R2a, R3a, R4a, R5, R6, R7, R8 is attached to the rest of the molecule via a heteroatom or a carbonyl, a straight- or branched chain, C1-12-, preferably C1-4-alkylene or C2-12, preferably C2-4-alkenylene or -alkynylene bridge may optionally be interposed between the heteroatom or the carbonyl and the point of attachment to the rest of the molecule.
  • Preferred examples of X are —COO R7 or —CONR5R6, wherein R5, R6 and R1 are preferably hydrogen, C1-4-alkyl, phenyl or alkylphenyl.
  • Preferably X is carboxy or —CONR5R6, wherein R5 and R6 are preferably hydrogen, C1-4-alkyl, phenyl or alkylphenyl, especially —CONH2.
  • Preferably A1 and A2 are each oxygen.
  • Preferably R1 is hydrogen, alkyl, especially C1-12 alkyl, particularly lower alkyl or aryl especially phenyl.
  • Examples of preferred R1 groups are methyl, ethyl, propyl, isopropyl, butyl, iso- or ter-butyl. 2,2,2-trimethylethyl each optionally attached via a methylene bridge or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • R1 as ethyl is especially preferred.
  • Preferably R2 and R2a are independently hydrogen, halogen or alkyl, especially lower alkyl.
  • Examples of preferred R1 and R2a groups are independently hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl. 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • Especially at least one and most preferably both of R2 and R2a are hydrogen.
  • Preferably R1a, R4 and R4a are independently hydrogen, alkyl, especially methyl or ethyl or aryl especially phenyl or aralkyl, especially benzyl.
  • Examples of preferred R3a, R4 and R4a groups are independently hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl. 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • Especially at least one and most preferably both of R4 and R4a are hydrogen.
  • R3a is particularly hydrogen or alkyl, especially lower alkyl and is most preferably hydrogen.
  • Preferably R3 is hydrogen, C1-12-alkyl, especially C1-6-alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato or alkoxy and attached to the ring either directly or via a thio, sulfinyl, sulfonyl, carbonyl or oxycarbonyl group and optionally, a C1-4-alkylene bridge, particularly methylene; C2-6-alkenyl or -alkynyl, especially C2-3-alkenyl or -alkynyl each optionally substituted by one or more halogens; azido; cyano; amido: carboxy; triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl or piperazinyl each optionally substituted by one or more substituents selected from halogen, C1-6-alkyl and phenyl and attached to the ring either directly or via a carbonyl group or a C1-4-alkylene bridge, particularly methylene; naphthyl; or phenyl, phenylalkyl or phenylalkenyl each optionally substituted by one or more substituents selected from halogen, C1-6-alkyl, C1-6 haloalkyl, C1-6-alkoxy, C1-6-alkylthio, amino, azido, phenyl and nitro and each attached to the ring either directly or via an oxy, sulfonyl, sulfonyloxy, carbonyl or carbonyloxy group and optionally additionally a C1-4-alkylene bridge, particularly methylene.
  • Also, preferably, R3 is C1-6-alkyl optionally substituted by one or more substituents selected from halogen, thiocyanato, azido, alkoxy, alkylthio, phenylsulfonyl; nitrooxy; C2-3-alkenyl or -alkynyl each optionally substituted by one or more halogens or by acetyl; tetrazolyl, pyridyl, furyl, pyrrolyl, thiazolyl or thienyl; or phenyl or phenylalkyl each optionally substituted by one or more substituents selected from halogen, C1-6-alkyl, C1-6 haloalkyl, C1-6-alkoxy, amino, azido, phenyl and nitro and each attached to the ring either directly or via a sulfonyloxy and optionally additionally a C1-4-alkylene bridge, particularly methylene.
  • Other examples of preferred R3 groups are hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethyl-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl.
  • R3 is especially C1-4-alkyl optionally substituted by one or more substituents selected from halogen, thiocyanato or azido; C2-5-alkenyl or -alkynyl, each optionally substituted by one or more halogens; thienyl; or phenyl optionally substituted by one or more substituents selected from halogen, C1-6-alkyl, C1-6 haloalkyl or azido.
  • Further examples of preferred R3 groups are C1-6 alkyl and C2-6 haloalkenyl.
  • Preferably R5 and R6 are independently hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl, especially hydrogen or methyl.
  • Especially at least one and most preferably both of R5 and R6 are hydrogen.
  • Preferably R7 is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or tert-butyl, 2,2,2-trimethylethyl, methoxy, ethoxy, phenyl, benzyl or the same substituted by at least one halogen atom such as trifluoromethyl, chlorophenyl.
  • Preferably R7 is hydrogen, methyl or ethyl especially hydrogen.
  • Preferably R8 is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl. 2,2,2-trimethylethyl, phenyl, benzyl or the same substituted by at least one halogen atom such as trifluoromethyl, chlorobenzyl.
  • Preferably RS is hydrogen or methyl.
  • Combinations of one or more of these preferred compound groups are especially preferred.
  • A particular group of compounds of formula I (Compounds 1A) comprises those wherein,
  • A2 is oxygen;
  • X is —CONR5R6 or —COOR7 or —CO—R8 or CN;
  • R1 is hydrogen or alkyl, aryl, halogen, hydroxy, amino, nitro, cyano;
  • R2, R3, R4, are the same or different and each is independently hydrogen or halogen, hydroxy, amino, nitro, cyano, acyl, acyloxy, a sulfonyl derivative, a sulfinyl derivative, an amino derivative, carboxy, ester, ether, amido, sulfonic acid, sulfonamide, alkoxycarbonyl, a thio derivative, alkyl, alkoxy, oxyester, oxyamido, aryl, an oxy derivative, heterocycle, vinyl and R3 may additionally represent C2-5 alkenyl, C2-5 alkynyl or azido each optionally substituted by one or more halogen, cyano, thiocyano, azido, cyclopropyl, acyl and/or phenyl; or phenylsulfonyloxy whereby any phenyl moiety may be substituted by one or more halogen, alkyl, haloalkyl, alkoxy, nitro, amino, and/or phenyl; most preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl.
  • R2a, R3a and R4a are hydrogen;
  • R4, R6, R7 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or oxy derivative; and
  • R8 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle, alkylthio or thio derivative.
  • Within these Compounds 1A, R1 is preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl; most preferably methyl, ethyl or n-propyl.
  • R2 and R4 are preferably independently hydrogen or halogen or methyl, ethyl, propyl, isopropyl, butyl, isobutyl; and, most preferably, are each hydrogen.
  • R3 is preferably C1-5 alkyl, C2-5 alkenyl, C2-C5 alkynyl, cyclopropyl, azido, each optionally substituted by one or more halogen, cyano, thiocyano, azido, alkylthio, cyclopropyl, acyl and/or phenyl; phenyl; phenylsulfonyl; phenylsulfonyloxy, tetrazole, thiazole, thienyl, furyl, pyrrole, pyridine, whereby any phenyl moiety may be substituted by one or more halogen, alkyl, haloalkyl, alkoxy, nitro, amino, and/or phenyl; most preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl.
  • X is preferably —COOH or —COOMe or —COOEt or —CONH2; most preferably —CONH2.
  • A further particular group of compounds of formula I (Compounds 1B) comprises those wherein,
  • X is —CA1NH2,—CA1NHCH3 or —CA1N(CH3)2;
  • R1 is alkyl or phenyl;
  • R3 is alkyl, alkenyl, alkynyl, cyano, isothiocyanato, ether, carboxyl, amido, aryl, heterocycle; or
  • R3 is CH2R10 wherein R10 is hydrogen, cycloalkyl, oxyester, oxyalkylsulfonyl, oxyarylsulfonyl, aminoalkylsulfonyl, aminoarylsulfonyl, nitrooxy, cyano, isothiocyanato, azido, alkylthio, arylthio, alkylsulfinyl, alkylsulfonyl, heterocycle, aryloxy, alkoxy or trifluoroethyl;
  • R3a is hydrogen, alkyl or aryl (especially with the proviso that when R3a is hydrogen, R1 other than methyl);
  • or R3R3a form a cycloalkyl;
  • and R2, R2a, R4 and R4a are each hydrogen.
  • Within the compounds of formula I,
  • R1 is preferably alkyl especially C1-12- more particularly C1-6-alkyl and is most preferably ethyl;
  • R2, R2a, R3a and R4a are preferably hydrogen;
  • R3 is preferably selected from hydrogen; C1-12-alkyl, especially C1-6-alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato or alkoxy and attached to the ring either directly or via a thio, sulfinyl, sulfonyl, carbonyl or oxycarbonyl group and optionally additionally a C1-4-alkylene bridge, particularly methylene; C2-6-alkenyl or -alkynyl, especially C2-3-alkenyl or -alkynyl, each optionally substituted by one or more halogens; azido; cyano; amido; carboxy; triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl or piperazinyl each optionally substituted by one or more substituents selected from halogen, C1-6-alkyl and phenyl and attached to the ring either directly or via a carbonyl group or a C1-4-alkylene bridge, particularly methylene; naphthyl; or phenyl, phenylalkyl or phenylalkenyl each optionally substituted by one or more substituents selected from halogen, C1-6-alkyl, C1-6 haloalkyl, C1-6-alkoxy, C1-6-alkylthio, amino, azido, phenyl and nitro and each attached to the ring either directly or via an oxy, sulfonyl, sulfonyloxy, carbonyl or carbonyloxy group and optionally additionally a C1-4-alkylene bridge, particularly methylene;
  • R3a is preferably hydrogen or C1-4-alkyl;
  • R4 and R4a are preferably, independently hydrogen, C1-4-alkyl, phenyl or benzyl.
  • A further group of compounds of formula I (Compounds 1C) comprises those in racemic form wherein, when X is —CONR5R6 and R1 is hydrogen, methyl, ethyl or propyl, then substitution on the pyrrolidine ring is other than mono-, di-, or tri-methyl or mono-ethyl.
  • A further group of compound of formula I (Compounds 1D) comprises those in racemic form wherein, when X is —CONR5R6 and R1 is hydrogen or C1-6-alkyl, C2-6-alkenyl or -alkynyl or cycloalkyl, each unsubstituted, then substitution in the ring is other than by alkyl, alkenyl or alkynyl, each unsubstituted.
  • A further particular group of compounds of formula I (Compounds 1E) comprises those wherein,
  • X is —CA1NH2;
  • R1 is H;
  • R3 is azidomethyl, iodomethyl, ethyl optionally substituted by 1 to 5 halogen atoms, n-propyl optionally substituted by 1 to 5 halogen atoms, vinyl optionally substituted by one or two methyl, and/or 1 to 3 halogen atoms, acetylene optionally substituted by C1-4-alkyl, phenyl or halogen;
  • R3a is hydrogen or halogen, preferably fluorine;
  • and R2, R2a, R4 and R4a are each hydrogen;
  • as their racemates or in enantiomerically enriched form, preferably the pure enantiomers.
  • A further particular group of compounds of formula I (Compounds 1F) comprises those wherein,
  • X is —CA1NH2;
  • R1 is H;
  • R3 is C1-6-alkyl, C2-6-alkenyl or C2-6-alkynyl optionally substituted by azido, oxynitro, 1 to 6 halogen atoms;
  • R3a is hydrogen or halogen, preferably fluorine;
  • and R1, R2, R4 and R4a are each hydrogen; as their racemates or in enantiomerically enriched form, preferably the pure enantiomers.
  • In all the above mentioned scopes when the carbon atom to which R1 is attached is asymmetric it is preferably in the “S”-configuration.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of:
    • (2S)-2-[4-(bromomethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[(4R)-4-(iodomethyl)-2-oxopyrrolidinyl]butanamide:
    • (2S)-2-(2-oxo-4-phenyl-1-pyrrolidinyl)butanamide;
    • (2S)-2-[4-(iodomethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(chloromethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • {1-[(1S)-1-(aminocarbonyl)propyl]-5-oxo-3-pyrrolidinyl}methyl 4-methylbenzenesulfonate;
    • (2S)-2-[(4R)-4-(azidomethyl)-2-oxopyrrolidinyl]butanamide;
    • 2-[4-(2,2-dibromovinyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • {1-[(1S)-1-(aminocarbonyl)propyl]-5-oxo-3-pyrrolidinyl}methyl nitrate;
    • (2S)-2-[2-oxo-4-(1H-tetraazol-1-ylmethyl)-1-pyrrolidinyl]butanamide;
    • 2-(2-oxo-4-vinyl-1-pyrrolidinyl)butanamide;
    • 2-{2-oxo-4-[(phenylsulfonyl)methyl]-1-pyrrolidinyl}butanamide;
    • (2S)-2-[(4R)-4-(2,2-dibromovinyl)-2-oxopyrrolidinyl]butanamide;
    • (2S)-2-[(4S)-4-(2,2-dibromovinyl)-2-oxopyrrolidinyl]butanamide;
    • (2S)-2-[4-(isothiocyanatomethyl)-2-oxo-1-pyrrolidinyl]butanamide:
    • 2-[2-oxo-4-(1,3-thiazol-2-yl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(2-thienyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(2-methoxyphenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(3-methoxyphenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(4-azidophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(3-thienyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(3-azidophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(3-thienyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[(4S)-2-oxo-4-vinylpyrrolidinyl]butanamide;
    • (2S)-2-[(4R)-2-oxo-4-vinylpyrrolidinyl]butanamide:
    • 2-[4-(2-bromophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[2-oxo-4-(3-pyridinyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-(4-[1,1′-biphenyl]-4-yl-2-oxo-1-pyrrolidinyl)butanamide;
    • (2S)-2-{4-[(methylsulfanyl)methyl]-2-oxo-1-pyrrolidinyl}butanamide:
    • 2-[4-(iodomethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[(4R)-4-(iodomethyl)-2-oxo-1-pyrrolidinyl]pentanamide;
    • (2S)-2-[(4R)-4-(iodomethyl)-2-oxopyrrolidinyl]propanamide;
    • 2-(2-oxo-4-propyl-1-pyrrolidinyl)propanamide,
    • 2-(2-oxo-4-propyl-1-pyrrolidinyl)butanamide;
    • 2-(2-oxo-4-pentyl-1-pyrrolidinyl)butanamide;
    • (2S)-2-[(4R)-4-(iodomethyl)-2-oxopyrrolidinyl]-N-methylbutanamide;
    • (2S)-2-(4-neopentyl-2-oxo-1-pyrrolidinyl)butanamide;
    • (2S)-2-(4-ethyl-2-oxo-1-pyrrolidinyl)butanamide;
    • 2-[4-(2,2-difluorovinyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(2,2-difluoroethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[(4S)-2-oxo-4-propylpyrrolidinyl]butanamide;
    • (2S)-2-[(4R)-2-oxo-4-propylpyrrolidinyl]butanamide;
    • 2-{4-[(Z)-2-fluoroethenyl]-2-oxo-1-pyrrolidinyl}butanamide;
    • 2-[4-(2-methyl-1-propenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-(4-butyl-2-oxo-1-pyrrolidinyl)butanamide;
    • 2-[4-(cyclopropylmethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-(4-isobutyl-2-oxo-1-pyrrolidinyl)butanamide;
    • 2-[4-(4-chlorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(3-chlorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-{2-oxo-4-[2-(trifluoromethyl)phenyl]-1-pyrrolidinyl}butanamide;
    • 2-[4-(2-fluorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(3-methylphenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(2-phenylethyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(3-bromophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-{4-[3,5-bis(trifluoromethyl)phenyl]-2-oxo-1-pyrrolidinyl}butanamide;
    • 2-[4-(3,4-dichlorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(2,4-dichlorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(2-furyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(3-phenylpropyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(3,5-dibromophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(3,4-dichlorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-(2-oxo-4-propyl-1-pyrrolidinyl)butanamide;
    • 2-[4-(3-chlorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-(4-ethynyl-2-oxo-1-pyrrolidinyl)butanamide;
    • 2-[4-(2-fluorophenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(cyclopropylmethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[(4S)-4-(2,2-difluorovinyl)-2-oxopyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(3,3,3-trifluoropropyl)-1-pyrrolidinyl]butanamide;
    • 2-[4-(3-methylphenyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(cyclopropylmethyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-[(4R)-4-(2,2-difluorovinyl)-2-oxopyrrolidinyl]butanamide;
    • (2S)-2-[2-oxo-4-(1H-pyrrol-1-yl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-(4-allyl-2-oxo-1-pyrrolidinyl)butanamide:
    • (2S)-2-[4-(2-iodopropyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-(4-allyl-2-oxo-1-pyrrolidinyl)butanamide;
    • (2S)-2-[2-oxo-4-(2-oxopropyl)-1-pyrrolidinyl]butanamide;
    • (2S)-2-[4-(2-bromo-1H-pyrrol-1-yl)-2-oxo-1-pyrrolidinyl]butanamide;
    • (2S)-2-(4-methyl-2-oxo-4-propyl-1-pyrrolidinyl)butanamide;
    • (2R)-2-[4-(2,2-dichlorovinyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[4-(bromoethynyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-[(4S)-4-(2,2-difluoropropyl)-2-oxopyrrolidinyl]butanamide;
    • (2S)-2-[4-(bromoethynyl)-2-oxo-1-pyrrolidinyl]butanamide;
    • 2-(2-oxo-4-propyl-1-pyrrolidinyl)pentanamide:
    • 3-cyclopropyl-2-(2-oxo-4-propyl-1-pyrrolidinyl)propanamide;
    • 2-(2-oxo-4-propyl-1-pyrrolidinyl)-3-(1,3-thiazol-4-yl)propanamide;
    • 2-(2-oxo-4-propyl-1-pyrrolidinyl)-4-pentenamide;
    • (2S)-2-[(4R)-2-oxo-4-vinylpyrrolidinyl]butanamide; including all isomeric forms and mixtures thereof or a pharmaceutically acceptable salt thereof.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of:
    • (2S)-2-[(4S)-4-(2,2-difluorovinyl)-2-oxopyrrolidinyl]butanamide;
    • (2S)-2-[(4S)-2-oxo-4-propylpyrrolidinyl]butanamide:
    • (2S)-2-[(4R)-2-oxo-4-propylpyrrolidinyl]butanamide.
  • ii) International Patent Application WO 2002/094787:
  • Compounds of the formula I
  • Figure US20140206667A1-20140724-C00002
  • wherein n represents 0 or 1 whereby R1 is not existent when n=0 and R1 is existent when n=1;
  • A1 represents an oxygen or a sulfur atom;
  • X is —CONR17R8, —COOR9, —CO—R10 or CN;
  • R1 when existent, R2, R3, R4 and R5 are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative,
  • provided that at least one of the substituents R chosen from R1 when existent, R2, R3, R4 or R5 is not hydrogen;
  • R6 is hydrogen, alkyl, aryl or —CH2—R6 wherein R is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
  • R7, R8 and R9 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or an oxy derivative; and
  • R10 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative;
  • their pharmaceutically acceptable salts, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers).
  • In the above formula, at least one substituent R1 to R5 is different from hydrogen. Some non-substituted compounds are referred to in U.S. Pat. Nos. 5,468,733 and 5,516,759. U.S. Pat. No. 5,468,733 refers to non-ring substituted 2-oxo-1-pyrrolidinyl and 2-oxo-1-piperidinyl derivatives as inhibitors of the oncogene Ras protein. In particular, these compounds block the ability of Ras to transform normal cells to cancer cells, and therefore can be included in several chemotherapeutic compositions for treating cancer.
  • U.S. Pat. No. 5,516,759 refers to non-ring substituted 2-oxo-1-pyrrolidinyl, 2-oxo-1-piperidinyl and azepanyl derivatives present at the N-terminus of dodecapeptides possessing LHRH (luteinizing hormone-releasing hormone) antagonistic activity. Such LHRH antagonists are useful in the treatment of a variety of conditions in which suppression of sex steroids plays a key role including contraception, delay of puberty, treatment of benign prostatic hyperplasia a. o.
  • In the definitions set forth below, unless otherwise stated, R11 and R12 are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfinyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.
  • The term “oxy derivative”, as used herein, is defined as including —O—R11 groups wherein R11 is as defined above except for “oxy derivative”. Non-limiting examples are alkoxy, alkenyloxy, alkynyloxy, acyloxy, oxyester, oxyamido, alkylsulfonyloxy, alkylsulfinyloxy, arylsulfonyloxy, arylsulfinyloxy, aryloxy, aralkoxy or heterocyclooxy such as pentyloxy, allyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methylenedioxy, carbonate.
  • The term “thio derivative”, as used herein, is defined as including —S—R11 groups wherein R11 is as defined above except for “thio derivative”. Non-limiting examples are alkylthio, alkenylthio, alkynylthio and arylthio.
  • The term “amino derivative”, as used herein, is defined as including —NHR11 or —NR11R12 groups wherein R11 and R12 are as defined above. Non-limiting examples are mono- or di-alkyl-, alkenyl-, alkynyl- and arylamino or mixed amino.
  • The term “acyl derivative”, as used herein, represents a radical derived from carboxylic acid and thus is defined as including groups of the formula R—CO—, wherein R11 is as defined above and may also be hydrogen. Preferred are acyl derivatives of formula —COR11 wherein R11 is selected from hydrogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkenyl, heterocyle and aryl. Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl, heptanedioyl, cyclohexanecarbonyl, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxalyl, ethoxalyl, cysteinyl, oxamoyl.
  • The term “sulfonyl derivative”, as used herein, is defined as including a group of the formula —SO2—R11, wherein R11 is as defined above except for “sulfonyl derivative”. Non-limiting examples are alkylsulfonyl, alkenylsulfonyl, alkynylsulfonyl and arylsulfonyl.
  • The term “sulfinyl derivative”, as used herein, is defined as including a group of the formula —SO—R11, wherein R11 is as defined above except for “sulfinyl derivative”. Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl and arylsulfinyl.
  • The term “alkyl”, as used herein, is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and generally containing 1-20 carbon atoms, most often 1 to 12 carbon atoms, preferably 1-7 carbon atoms for non-cyclic alkyl and 3-7 carbon atoms for cycloalkyl (in these two preferred cases, unless otherwise specified, “lower alkyl”), each optionally substituted by, preferably 1 to 5, substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cycloalkyl, sulfonic acid, sulfonamide, thio derivative, alkylthio, oxyester, oxyamido, heterocycle, vinyl, alkoxy (preferably C1-5), aryloxy (preferably C6-10) and aryl (preferably C6-10).
  • Preferred are alkyl groups containing 1 to 7 carbon atoms, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl. Most preferred are C1-4 alkyl and C3-7 cycloalkyl, each optionally substituted by one or more hydroxy, halogen, lower alkyl or/and azido.
  • Most preferred alkyl groups are hydroxymethyl, propyl, butyl, 2,2,2-trifluoroethyl, 2-bromo-2,2-difluoroethyl, 2-chloro-2,2-difluoroethyl, 3,3,3-trifluoropropyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2,2-difluoropropyl, 2-iodo-2,2-difluoroethyl.
  • The term “lower alkyl”, as used herein, and unless otherwise specified, refers to C1 to C7 saturated straight, branched or cyclic hydrocarbon. Non limiting examples are methyl, ethyl, propyl, isopropyl, butyl, tertiobutyl, pentyl, cyclopropyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methypentyl, 2,2-dimethylbutyl, optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferably, lower alkyl is methyl.
  • The term “alkenyl”, as used herein, is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, thiocyanato, azido, alkylthio, cycloalkyl, acyl, nitro, cyano, aryl and heterocycle.
  • Preferred alkenyl groups are C2-C1-2 alkenyls, especially C2-6 alkenyls, such as ethenyl (=vinyl), 1-methyl-1-ethenyl, 2,2-dimethyl-1-ethenyl, 1-propenyl, 2-propenyl (=allyl), 1-butenyl, 2-butenyl, 3-butenyl, 4-pentenyl, 1-methyl-4-pentenyl, 3-methyl-1-pentenyl, 1-hexenyl, 2-hexenyl and the like, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl. Most preferred is vinyl, optionally substituted by one or more halogen or/and lower alkyl, and especially 2,2-difluorovinyl, 2,2-dibromovinyl and 2,2-dichlorovinyl.
  • The term “alkynyl” as used herein, is defined as including a monovalent branched or unbranched hydrocarbon radical containing at least one carbon-carbon triple bond, for example ethynyl, 2-propynyl (=propargyl), and the like, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl, heterocycle, thiocyanato, azido, alkylthio, alkyl and acyl.
  • Preferred alkynyl groups are C2-12 alkynyl, especially C2-6 alkynyl, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, acyl, aryl such as phenyl and alkyl, preferably cycloalkyl.
  • Most preferred are ethynyl, propynyl and butynyl, optionally substituted by lower alkyl or/and halogen, and especially 1-propynyl, cyclopropylethynyl, 3-methyl-1-butynyl and 3,3,3-trifluoro-1-propynyl.
  • When present as bridging groups, alkyl, alkenyl and alkynyl represent straight- or branched chains, C1-12, preferably C1-4-alkylene or C2-12-, preferably C2-4-alkenylene or -alkynylene moieties respectively.
  • Groups where branched derivatives are conventionally qualified by prefixes such as “n”, “sec”, “iso” and the like (e.g. “n-propyl”, “sec-butyl”) are in the n-form unless otherwise stated.
  • The term “aryl”, as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of at least one ring, most often 1 to 3 rings and generally containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl, each optionally substituted by one or more substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfinyl, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, C1-6-alkylthio, oxyester, oxyamido, aryl, C1-6-alkoxy, C6-10-aryloxy, C1-6-alkyl, C1-6-haloalkyl. Aryl radicals are preferably monocyclic or bicyclic containing 6-10 carbon atoms. Preferred aryl groups are phenyl and naphthyl each optionally substituted by one or more substituents independently selected from halogen, nitro, amino, azido, C1-6-alkoxy, C1-6-alkyl, C1-6-haloalkyl, sulfonyl and phenyl.
  • Preferred aryl is phenyl, optionally substituted by one or more halogen, lower alkyl, azido or nitro, such as 3-chlorophenyl and 3-azidophenyl.
  • The term “halogen”, as used herein, includes an atom of Cl, Br, F, I.
  • The term “hydroxy”, as used herein, represents a group of the formula —OH.
  • The term “thiol”, as used herein, represents a group of the formula —SH.
  • The term “cyano”, as used herein, represents a group of the formula —CN.
  • The term “nitro”, as used herein, represents a group of the formula —NO2.
  • The term “nitrooxy”, as used herein, represents a group of the formula —ONO2.
  • The term “amino”, as used herein, represents a group of the formula —NH2.
  • The term “azido”, as used herein, represents a group of the formula —N3.
  • The term “carboxy”, as used herein, represents a group of the formula —COOH.
  • The term “sulfonic acid”, as used herein, represents a group of the formula —SO3H.
  • The term “sulfonamide”, as used herein, represents a group of the formula —SO2NH2.
  • The term “ester”, as used herein, is defined as including a group of formula —COO—R11 wherein R11 is as defined above except oxy derivative, thio derivative or amino derivative. Preferred are esters of formula —COOR11 wherein R11 is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl and aryl. Most preferred are esters where R11 is a lower alkyl, especially methyl.
  • The term “ether” is defined as including a group selected from C1-50-straight or branched alkyl, or C2-50-straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.
  • The term “amido” is defined as including a group of formula —CONH2 or —CONHR11 or —CONR11R2 wherein R11 and R12 are as defined above.
  • The term “heterocycle”, as used herein, is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl, and optionally being substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups. Non-limiting examples of heterocycles are pyridyl, furyl, pyrrolyl, thienyl, isothiazolyl, triazolyl, imidazolyl, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, pyrazolyl, indolyl, indolizinyl, purinyl, isoindolyl, carbazolyl, thiazolyl, 1,2,4-thiadiazolyl, thiomorpholinyl, thieno (2,3-b) furanyl, furopyranyl, benzofuranyl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl, benzothiazolyl, or benzoxazolyl, cinnolinyl, phthalazinyl, quinoxalinyl, 1-oxidopyridyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenothiazinyl, furazanyl, benzodioxolyl, isochromanyl, indolinyl, xanthenyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl. 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i.e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) optionally substituted by alkyl or as described above for the alkyl groups. The term “heterocycle” also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1) heptanyl, 7-oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1) octanyl.
  • The heterocycle is preferably selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl.
  • More preferably the heterocycle is selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl, and especially from 2- and 3-thienyl, optionally substituted by one or more halogen, acyl such as formyl, cyano and/or lower alkyl, such as methyl.
  • In the above definitions it is to be understood that when a substituent such as R1, R2, R3, R4, R5, R7, R8, R9, R10 is attached to the rest of the molecule via a heteroatom or a carbonyl, a straight- or branched chain, C1-12-, preferably C1-4-alkylene or C2-12, preferably C2-4-alkenylene or -alkynylene bridge may optionally be interposed between the heteroatom or the carbonyl and the point of attachment to the rest of the molecule.
  • The term “R substituent” refers to R1, R2, R3, R4 or R5, independently.
  • According to a preferred embodiment, a compound of formula I is as defined above wherein n represents 0. The compound is a 6-ring structure (2-thioxo- or 2-oxo-piperidinyl derivative) wherein R1 is not existent since n=0, and is depicted by the formula (I-A).
  • Figure US20140206667A1-20140724-C00003
  • According to a following embodiment, the compound of formula I is as defined above wherein n represents 1. The compound is a 7-ring structure (2-thioxo- or 2-oxo-azepanyl derivative) wherein R1 is existent since n=1 and depicted by the formula (I-B).
  • Figure US20140206667A1-20140724-C00004
  • According to a more preferred embodiment, said compound is as defined above wherein n=0, R3 and/or R4 are different from hydrogen and R2 and R5 represent hydrogen.
  • According to another more preferred embodiment, said compound is as defined above wherein n=1, R2, R3 and/or R4 are different from hydrogen and wherein R1 and R5 represent hydrogen.
  • According to a yet more preferred embodiment, said compound is as defined above wherein only one R substituent chosen from R3 or R4 when n=0 or from R2, R3 or R4 when n=1, is different from hydrogen and the remaining R substituent(s) is/are hydrogen. We hereby refer to a mono-substituted 2-thioxo- or 2-oxo-piperidinyl or 2-thioxo- or 2-oxo-azepanyl derivatives.
  • According to another preferred embodiment, compounds of formula I are as defined above wherein A1 represents an oxygen atom. We hereby refer to 2-oxo-piperidinyl or 2-oxo-azepanyl derivatives.
  • According to another preferred embodiment, compounds of formula I are as defined above wherein X is CONR7R8, especially CONH2. We hereby refer to amido derivatives of 2-oxo (or thioxo)-piperidinyl or 2-oxo (or thioxo)-azepanyl.
  • According to another preferred embodiment, compounds of formula I are as defined above wherein R6 represents hydrogen, C1-4 alkyl, or a CH2—R6a group wherein R6a represents a heterocycle. Most preferably R6 is a C1-4 alkyl, especially ethyl. When R6 is ethyl we refer to 2-(2-oxo (or thioxo)-1-piperidinyl) butanamide or 2-(2-oxo (or thioxo)-1-azepanyl) butanamide derivatives.
  • According to another preferred embodiment, compounds of formula I are as defined above wherein the carbon atom to which R6 is attached is of the S configuration. In case where R6 is ethyl, A is oxygen and X is CONR7R5 we refer then to (2S)-2-(2-oxo-1-piperidinyl) butanamide or (2S)-2-(2-oxo-1-azepanyl) butanamide derivatives.
  • According to a preferred embodiment, the compound is as defined above wherein R2 when n=1. R3 and R4 are the same or different and each is independently hydrogen, halogen, nitro, nitrooxy, cyano, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, acyl derivative, sulfonyl derivative or sulfinyl derivative;
  • R1 when existent, R2 when n=0 and R5 are hydrogen;
  • R6 is hydrogen, alkyl, aryl or —CH2—R6a wherein R6a is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
  • According to this preferred embodiment, the compound is generally such that when R6 is benzyl, X is —COOCH3 and n=1, R2 is different from methyl when R3 and R4 are both hydrogen and R4 is different from methyl when R2 and R3 are both hydrogen.
  • According to another preferred embodiment, the compound is as defined above wherein R2 when n=1, R3 and R4 are the same or different and each is independently hydrogen; cyano; carboxy; amido;
  • C1-12 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkyltio, cycloalkyl, acyl, aryl and heterocycle;
  • C2-12 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl;
  • C2-12 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl; acyl derivative of formula —CO—R11, wherein R11 is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, heterocycle and aryl;
  • ester of formula —CO—O—R11 wherein R11 is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl and aryl:
  • heterocycle selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl;
  • aryl, each optionally substituted by one or more substituents selected from C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 alkylthio, amino, azido, sulfonyl, aryl and nitro.
  • According to another preferred embodiment, the compound is as defined above, wherein R2 when n=1, R3 and R4 are the same or different and each is independently hydrogen;
  • C1-7 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl;
  • C2-6 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl;
  • C2-6 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl;
  • heterocycle selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl;
  • phenyl, each optionally substituted by one or more substituents selected from C1-6 alkyl, halogen substituted alkyl, halogen, alkoxy, amino, azido, sulfonyl, phenyl and nitro.
  • According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R2, R3 and R4 when n=1 or from the group R3 and R4 when n=0, represents independently C1-4-alkyl or C3-7-cycloalkyl, optionally substituted by one or more halogen, hydroxy, lower alkyl and/or azido.
  • According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R2, R3 and R4 when n=1 or from the group R3 and R4 when n=0, represents independently vinyl, optionally substituted by one or more halogen or/and lower alkyl.
  • According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R2, R3 and R4 when n=1 or from the group R3 and R4 when n=0, represents independently ethynyl, propynyl or butynyl, optionally substituted by one or more halogen and/or lower alkyl.
  • According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R2, R3 and R4 when n=1 or from the group R3 and R4 when n=0, represents independently phenyl, optionally substituted by one or more halogen, lower alkyl, azido and/or nitro.
  • According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R2, R3 and R4 when n=1 or from the group R3 and R4 when n=0, represents independently 2- or 3-thienyl, optionally substituted by one or more halogen, acyl, cyano or/and lower alkyl.
  • According to a particular preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R3, R4 and R2 when n=1 or from the group R3 and R4 when n=0, is hydroxymethyl, propyl, butyl. 3,3,3-trifluoropropyl, 2,2,2-trifluoroethyl, cyclopropylmethyl, iodomethyl, azidomethyl. 2-thienyl, 3-thienyl, phenyl, 3-chlorophenyl, 3-azidophenyl, 2,2-difluorovinyl, 2,2-dibromovinyl, 2,2-dichlorovinyl, 2-ethynyl, 5-methyl-2-thienyl, 5-formyl-2-ethynyl, 5-cyano-2-thienyl, 3-bromo-2-thienyl, 4-methyl-2-thienyl, 3,3,3-trifluoro-1-propynyl, 1-propynyl, cyclopropylethynyl, 3-methyl-1-butynyl, 1-butynyl, 2,2-difluoropropyl, 2-chloro-2,2-difluoroethyl, 2-bromo-2,2-difluoroethyl and 2-iodo-2,2-difluoroethyl.
  • According to yet another preferred embodiment, the compound is as defined above wherein R1, R2, R4 and R5 are hydrogen.
  • According to even another preferred embodiment, the compound is as defined above wherein R1, R2, R3 and R5 are hydrogen.
  • According to even another preferred embodiment, the compound is as defined above wherein n=1 and R1, R3, R4 and R6 are hydrogen.
  • In all the above-mentioned scopes when the carbon atom to which R6 is attached is asymmetric it is preferably in the “S”-configuration.
  • Representative compounds useful in the methods and compositions of this invention as defined above are selected from the group consisting of
    • 2-[5-(hydroxymethyl)-2-oxo-1-piperidinyl]butanamide.
    • 2-(2-oxo-5-propyl-1-piperidinyl)butanamide,
    • 2-[2-oxo-5-(3,3,3-trifluoropropyl)-1-piperidinyl]butanamide,
    • 2-[5-(cyclopropylmethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(iodomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(azidomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-(2-oxo-5-phenyl-1-piperidinyl)butanamide,
    • 2-[2-oxo-5-(2-thienyl)-1-piperidinyl]butanamide,
    • 2-[2-oxo-5-(3-thienyl)-1-piperidinyl]butanamide,
    • 2-[5-(3-chlorophenyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(3-azidophenyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(2,2-difluorovinyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(2,2-dibromovinyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(2,2-dichlorovinyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-(5-ethynyl-2-oxo-1-piperidinyl)butanamide,
    • 2-[5-(5-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(5-formyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(5-cyano-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(3-bromo-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(4-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[2-oxo-5-(3,3,3-trifluoro-1-propynyl)-1-piperidinyl]butanamide,
    • 2-[2-oxo-5-(1-propynyl)-1-piperidinyl]butanamide.
    • 2-[5-(cyclopropylethynyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(3-methyl-1-butynyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(1-butynyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(2,2-difluoropropyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(2-chloro-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(2-bromo-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(hydroxymethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-(2-oxo-4-propyl-1-piperidinyl)butanamide,
    • 2-[2-oxo-4-(3,3,3-trifluoropropyl)-1-piperidinyl]butanamide,
    • 2-[4-(cyclopropylmethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(iodomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(azidomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-(2-oxo-4-phenyl-1-piperidinyl)butanamide,
    • 2-[2-oxo-4-(2-thienyl)-1-piperidinyl]butanamide,
    • 2-[2-oxo-4-(3-thienyl)-1-piperidinyl]butanamide,
    • 2-[4-(3-chlorophenyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(3-azidophenyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(2,2-difluorovinyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(2,2-dibromovinyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(2,2-dichlorovinyl)-2-oxo-1-piperidinyl]butanamidc,
    • 2-(4-ethynyl-2-oxo-1-piperidinyl)butanamide,
    • 2-[4-(5-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(5-formyl-2-thienyl)-2-oxo-1-piperidinyl]butanamidc,
    • 2-[4-(5-cyano-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(3-bromo-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(4-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[2-oxo-4-(3,3,3-trifluoro-1-propynyl)-1-piperidinyl]butanamide,
    • 2-[2-oxo-4-(1-propynyl)-1-piperidinyl]butanamide,
    • 2-[4-(cyclopropylethynyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(3-methyl-1-butynyl)-2-oxo-1-piperidinyl]butanamidc,
    • 2-[4-(1-butynyl)-2-oxo-1-piperidinyl]butanamidc.
    • 2-[4-(2,2-difluoropropyl)-2-oxo-1-piperidinyl]butanamide.
    • 2-[4-(2-chloro-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(2-bromo-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[4-(2,2,2-trifluoroethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(hydroxymethyl)-2-oxo-1-azepanyl]butanamide.
    • 2-(2-oxo-5-propyl-1-azepanyl)butanamide,
    • 2-[2-oxo-5-(3,3,3-trifluoropropyl)-1-azepanyl]butanamide,
    • 2-[5-(cyclopropylmethyl)-2-oxo-1-azepanyl]butanamide.
    • 2-[5-(iodomethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(azidomethyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-(2-oxo-5-phenyl-1-azepanyl)butanamide,
    • 2-[2-oxo-5-(2-thienyl)-1-azepanyl]butanamide,
    • 2-[2-oxo-5-(3-thienyl)-1-azepanyl]butanamide,
    • 2-[5-(3-chlorophenyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[5-(3-azidophenyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(2,2-difluorovinyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(2,2-dibromovinyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[5-(2,2-dichlorovinyl)-2-oxo-1-azepanyl]butanamide,
    • 2-(5-ethynyl-2-oxo-1-azepanyl)butanamide,
    • 2-[5-(5-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(5-formyl-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(5-cyano-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(3-bromo-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(4-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[2-oxo-5-(3,3,3-trifluoro-1-propynyl)-1-azepanyl]butanamide,
    • 2-[2-oxo-5-(1-propynyl)-1-azepanyl]butanamide,
    • 2-[5-(cyclopropylethynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(3-methyl-1-butynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(1-butynyl)-2-oxo-1-azepanyl]butanamide.
    • 2-[5-(2,2-difluoropropyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(2-chloro-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[5-(2-bromo-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[5-(2,2,2-trifluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(hydroxymethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-(2-oxo-6-propyl-1-azepanyl)butanamide,
    • 2-[2-oxo-6-(3,3,3-trifluoropropyl)-1-azepanyl]butanamidc,
    • 2-[6-(cyclopropylmethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(iodomethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(azidomethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-(2-oxo-6-phenyl-1-azepanyl)butanamide,
    • 2-[2-oxo-6-(2-thienyl)-1-azepanyl]butanamide,
    • 2-[2-oxo-6-(3-thienyl)-1-azepanyl]butanamide,
    • 2-[6-(3-chlorophenyl)-2-oxo-1-azepanyl]butanamidc.
    • 2-[6-(3-azidophenyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(2,2-difluorovinyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(2,2-dibromovinyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(2,2-dichlorovinyl)-2-oxo-1-azepanyl]butanamide.
    • 2-(6-ethynyl-2-oxo-1-azepanyl)butanamide,
    • 2-[6-(5-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[6-(5-formyl-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(5-cyano-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(3-bromo-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(4-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[2-oxo-6-(3,3,3-trifluoro-1-propynyl)-1-azepanyl]butanamide,
    • 2-[2-oxo-6-(1-propynyl)-1-azepanyl]butanamide,
    • 2-[6-(cyclopropylethynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(3-methyl-1-butynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(1-butynyl)-2-oxo-1-azepanyl]butanamide.
    • 2-[6-(2,2-difluoropropyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(2-chloro-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(2-bromo-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[6-(2,2,2-trifluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(hydroxymethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-(2-oxo-4-propyl-1-azepanyl)butanamide,
    • 2-[2-oxo-4-(3,3,3-trifluoropropyl)-1-azepanyl]butanamide,
    • 2-[4-(cyclopropylmethyl)-2-oxo-1-azepanyl]butanamide.
    • 2-[4-(iodomethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(azidomethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-(2-oxo-4-phenyl-1-azepanyl)butanamide.
    • 2-[2-oxo-4-(2-thienyl)-1-azepanyl]butanamide,
    • 2-[2-oxo-4-(3-thienyl)-1-azepanyl]butanamide,
    • 2-[4-(3-chlorophenyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(3-azidophenyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2,2-difluorovinyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2,2-dibromovinyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2,2-dichlorovinyl)-2-oxo-1-azepanyl]butanamide.
    • 2-(4-ethynyl-2-oxo-1-azepanyl)butanamide,
    • 2-[4-(5-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(5-formyl-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(5-cyano-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(3-bromo-2-thienyl)-2-oxo-1-azepanyl]butanamidc,
    • 2-[4-(4-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[2-oxo-4-(3,3,3-trifluoro-1-propynyl)-1-azepanyl]butanamide,
    • 2-[2-oxo-4-(1-propynyl)-1-azepanyl]butanamide,
    • 2-[4-(cyclopropylethynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(3-methyl-1-butynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(1-butynyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2,2-difluoropropyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2-chloro-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2-bromo-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide,
    • 2-[4-(2,2,2-trifluoroethyl)-2-oxo-1-azepanyl]butanamide.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of:
    • (2S)-2-[5-(iodomethyl)-2-oxo-1-piperidinyl]butanamide,
    • (2S)-2-[5-(azidomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-(2-oxo-5-phenyl-1-piperidinyl]butanamide,
    • (2S)-2-[4-(iodomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(iodomethyl)-2-oxo-1-azepanyl]butanamide.
  • iii) International Patent Application WO 2004/087658:
  • A compound having the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • Figure US20140206667A1-20140724-C00005
  • wherein
  • R1 is hydrogen,
  • R2 is hydrogen or C1-20-alkyl,
  • R3 is hydrogen, C1-20-alkyl, C4-8-cycloalkyl, C5-8-cycloalkenyl, aryl, aromatic or non aromatic heterocycle, C1-20-alkoxy, or a group of formula —W—R8, R3a is hydrogen, C1-20-alkyl or a group of formula:
  • Figure US20140206667A1-20140724-C00006
  • or NR3R3a is a group of formula
  • Figure US20140206667A1-20140724-C00007
  • R4 is hydrogen,
  • R5 is hydrogen; nitro; halogen; azido; cyano; —S—C1-4-alkyl; —SO—C1-4-alkyl; —SO2—C1-4-alkyl: —SONH2; C1-20-alkyl unsubstituted or substituted by halogen; or C1-20-alkoxy unsubstituted or substituted by halogen,
  • R6 is hydrogen, C1-20-alkyl or halogen,
  • R7 is hydrogen, C1-20-alkyl or halogen,
  • W is C1-12-alkylene, —NH— or —NHC(═O)—,
  • X is O, S or NH,
  • Y is O, S, —CR12R13—, —NR14— or —C(—O)—,
  • R8 is aryl or heterocycle,
  • R9, R10, R10a and R11 are independently selected from hydrogen, C1-4-alkyl, halogen, hydroxy or methoxycarbonyl,
  • or R10 and R10a together form a C3-6-alkylene,
  • R12 is hydrogen, C1-4-alkyl, halogen or hydroxy,
  • R13 is hydrogen,
  • or CR12R13 is dioxolanyl,
  • R14 is aryl, heterocycle or a group of formula —V—R15,
  • V is C1-12-alkylene,
  • R15 is aryl or heterocycle,
  • m is 1 to 4.
  • n is 0 or 1,
  • and at least one of R5, R6 or R7 is different from hydrogen when R2 is hydrogen, R3 is H or 2,6-diisopropylphenyl, and R3a is H.
  • In another aspect, the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • Figure US20140206667A1-20140724-C00008
  • wherein
  • R1 is hydrogen,
  • R2 is hydrogen or C1-20-alkyl,
  • R3 is hydrogen, C1-20-alkyl, C4-8-cycloalkyl, C5-8-cycloalkenyl, aryl, aromatic or non aromatic heterocycle, C1-20-alkoxy, or a group of formula —W—RS,
  • R3a is hydrogen, C1-20-alkyl or a group of formula:
  • Figure US20140206667A1-20140724-C00009
  • or NR3R3a is a group of formula
  • Figure US20140206667A1-20140724-C00010
  • R4 is hydrogen,
  • R7 is hydrogen; nitro; halogen; C1-20-alkyl unsubstituted or substituted by halogen; or C1-20-alkoxy unsubstituted or substituted by halogen,
  • R6 is hydrogen, C1-20-alkyl or halogen,
  • R7 is hydrogen, C1-20-alkyl or halogen,
  • W is C1-12-alkylene, —NH— or —NHC(═O)—.
  • X is O, S or NH.
  • Y is O, S, —CR12R13—, —NR14— or —C(═O)—,
  • R8 is aryl or heterocycle,
  • R9, R10, R10a and R11 are independently selected from hydrogen, C1-4-alkyl, halogen, hydroxy or methoxycarbonyl.
  • or R10 and R10a together form a C3-6-alkylene,
  • R12 is hydrogen, C1-4-alkyl, halogen or hydroxy,
  • R13 is hydrogen.
  • or CR12R13 is dioxolanyl,
  • R14 is aryl, heterocycle or a group of formula —V—R15,
  • V is C1-12-alkylene,
  • R15 is aryl or heterocycle,
  • m is 1 to 4,
  • n is 0 or 1,
  • and at least one of R5, R6 or R7 is different from hydrogen when R2 is hydrogen, R3 is H or 2,6-diisopropylphenyl, and R3a is H.
  • The term “alkyl”, as used herein, is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and containing 1-20 carbon atoms, preferably 1-6 carbon atoms and more preferably 1-4 carbon atoms for non-cyclic alkyl and 3-8 carbon atoms for cycloalkyl. Alliyl moieties may optionally be substituted by 1 to 5 substituents independently selected from halogen, hydroxy, alkoxy, alkoxycarbonyl, ester or alkylamino. Preferred alkyl groups are methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, n-butyl, 2-fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2,2-dimethylpropyl, 1-(hydroxymethyl) propyl, 3,3,3-trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2-oxoethyl and 3-(dimethylamino) propyl.
  • The term “cycloalkyl”, as used herein, refers to a monovalent group of 3 to 18 carbon atoms, preferably 4-8 carbon atoms, derived from a saturated cyclic or polycyclic hydrocarbon which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl group is cycloheptyl.
  • The term “alkylene”, as used herein, represents a divalent alkyl group, having straight or branched moieties, containing 1-12 carbon atoms, preferably 1-6 carbon atoms, and being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred alkylene groups are methylene, ethylene, hydroxyethylene, trimethylene or propylene.
  • The term “cycloalkenyl”, as used herein, is defined as a cyclic unsaturated hydrocarbon radical having at least one double bond, containing 4-20 carbon atoms, preferably 5-8 carbon atoms, and being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkenyl group is 6-(hydroxymethyl)cyclohex-3-en-1-yl.
  • The term “aryl”, as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of 1-3 rings and containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, nitro, C1-6-alkyl, C1-6-alkoxy, C1-6-alkylsulfonyl, trifluoromethylthio or pyridinylalkyl. Aryl radicals are preferably phenyl radicals. Preferred aryl groups are phenyl, 3-hydroxyphenyl, 3-fluorophenyl, 3-methylphenyl. 4-methylphenyl, 4-hydroxyphenyl, 4-hydroxy-3-methoxyphenyl, 3-(2-pyridin-2-ylethyl)phenyl, 3,4-dimethylphenyl, 4-tert-butylphenyl, 4-methylsulfonylphenyl, 2-nitrophenyl, 2-chloro-6-fluorophenyl, 2-[(trifluoromethyl)thio]phenyl. 2-chlorophenyl or 4-bromophenyl.
  • The term “halogen”, as used herein, includes an atom of Cl, Br, F, I.
  • The term “nitro”, as used herein, represents a group of the formula —NO2.
  • The term “hydroxy”, as used herein, represents a group of the formula —OH.
  • The term “alkoxy”, as used herein, represents a group of formula —ORb wherein Rb is an alkyl group, as defined above.
  • The term “ester”, as used herein, represents a group of formula —COORc wherein Rc is an alkyl group or an aryl group, as defined above.
  • The term “alkoxycarbonyl”, as used herein, represents a group of formula —COORd wherein Rd is an alkyl group, as defined above.
  • The term “amino”, as used herein, represents a group of the formula —NH2.
  • The term “alkylamino”, as used herein, represents a group of formula —NHRe or —NReRf wherein Re and Rf are alkyl group as defined above.
  • The term alkylsulfonyl, as used herein is defined as representing a group of formula —SO2—Rg, wherein Rf is C1-4-alkyl.
  • The term “heterocycle”, as used herein is defined as including an aromatic or non aromatic cycloalkyl or cycloalkenyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl.
  • Non-limiting examples of aromatic heterocycles are pyrazolyl, furyl, imidazolyl, triazolyl, oxazolyl, pyridinyl, pyrrolyl, thienyl, isothiazolyl, benzimidazolyl, tetrazolyl, isooxazolyl, oxazolyl, thiazolyl, 1,2,4-thiadiazolyl, oxadiazole, pyridazinyl, pyrimidinyl, pyrazinyl, isoindolyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, quinazolinyl, quinolizinyl, naphthyridinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, indolyl, indolizinyl, purinyl, carbazolyl, thieno (2,3-b) furanyl, thianthrenyl, benzothiazolyl, benzoxazolyl, cinnolinyl, quinoxalinyl, phenothiazinyl, isochromanyl and xanthenyl, optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, azido, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkyl, C1-6-haloalkyl, formyl or ester. More preferred aromatic heterocycles are pyrazolyl, furyl, imidazolyl, triazolyl, oxazolyl and pyridinyl.
  • Non-limiting examples of non aromatic heterocycles are tetrahydrofuranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholinyl, thiomorpholinyl, pyrrolidinyl, thiazolidinyl, indolinyl, tetrahydrobenzazocinyl, dihydroisochromenyl, tetrahydropyranyl, oxooctahydroquinolinyl, dioxolanyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, 8-thiabicyclo [3.2.1]cyclooctanyl, 1,4-dithiepanyl, tetrahydro-2H-thiopyranyl, azepanyl and azocanyl, optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, azido, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkyl, C1-6-haloalkyl, formyl or ester. More preferred non aromatic heterocycles are tetrahydrofuranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholinyl, thiomorpholinyl, pyrrolidinyl, thiazolidinyl, indolinyl, tetrahydro-1-benzazocin-1 (2H)-yl, 3,4-dihydro-1H-isochromen-1-yl, tetrahydropyranyl, oxooctahydroquinolinyl and dioxolanyl. The term “heterocycle” also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cycloalkyl ring, a cycloalkenyl ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1)heptanyl, 7-oxabicyclo (2.2.1)heptanyl and 8-azabicyclo (3.2.1)octanyl.
  • The term “pyridinylalkyl”, as used herein, represents a group of formula —Rh-pyridinyl in which Rh is C1-4-alkylene.
  • The term “azido” as used herein, represents a group of the formula —N3.
  • The term “cyano” as used herein, represents a group of the formula —CN.
  • Generally, R2 is hydrogen or C1-4-alkyl.
  • Preferably, R2 is hydrogen, methyl or ethyl. More preferably, R2 is hydrogen or methyl.
  • Generally, R3 is hydrogen; C1-6-alkyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, hydroxy, alkoxy, alkoxycarbonyl or alkylamino; C5-7-cycloalkyl; (hydroxymethyl)cyclohexenyl; phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl, trifluoromethylthio or pyridinylalkyl; pyridinyl unsubstituted or substituted by methoxy; triazolyl; C1-4-alkoxy; or a group of formula —W—R8 wherein:
  • Generally, W is C1-4-alkylene unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl or alkoxy; —NH—; or —NHC(═O)—; and
  • R8 is phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl or trifluoromethylthio; furyl unsubstituted or substituted by methyl; pyrazolyl; pyridinyl; morpholinyl; tetrahydrobenzazocinyl; piperidinyl unsubstituted or substituted by methyl; dihydroisochromenyl or dihydroimidazolyl.
  • Preferably, R3 is hydrogen, n-butyl, cycloheptyl, 2-fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2,2-dimethylpropyl, 1-(hydroxymethyl) propyl, 3,3,3-trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2-oxoethyl. 3-(dimethylamino) propyl, 6-(hydroxymethyl)cyclohex-3-en-1-yl, 3-hydroxyphenyl, 3-fluorophenyl, 3-(2-pyridin-2-ylethyl)phenyl, 3,4-dimethylphenyl, 4-tert-butylphenyl, benzyl, 4-hydroxy-3-methoxybenzyl, 4-methylsulfonylbenzyl, 2-nitrobenzyl, 2-chloro-6-fluorobenzyl, 2-[(trifluoromethyl) thio]benzyl, 2-hydroxy-2-phenylethyl, 2-(3,4-dimethoxyphenyl)ethyl. 2-(2-chlorophenyl)ethyl, 2-(4-methylphenyl)ethyl, (4-bromophenyl)amino, pyridin-3-yl, 6-methoxypyridin-3-yl, 4H-1,2,4-triazol-3-yl, pyridin-4-ylmethyl, (5-methyl-2-furyl)methyl, 3-(1H-pyrazol-1-yl)propyl, 2-morpholin-4-ylethyl, 2-((3,4,5,6-tetrahydro-1-benzazocin-1(2H)-yl) propyl, 2-(2-methylpiperidin-1-yl)ethyl, 3,4-dihydro-1H-isochromen-1-ylmethyl, methoxy, (4-pyridinylcarbonyl)amino or 4,5-dihydro-1H-imidazol-2-ylamino. More preferably. R3 is hydrogen.
  • Generally, R3a is hydrogen, C1-4-alkyl or a group of formula
  • Figure US20140206667A1-20140724-C00011
  • wherein m is 1 to 4.
  • Preferably, R3 is hydrogen, methyl or tetrahydrofuran-2-ylmethyl. More preferably. R3a is hydrogen.
  • In another embodiment, NR3R3a is piperidinyl unsubstituted or substituted by hydroxy; thiomorpholinyl; thiazolidinyl unsubstituted or substituted by C1-4-alkoxycarbonyl; 2,5-dihydro-1H-pyrrol-1-yl; 1,4-dioxa-8-azaspiro[4.5]dec-8-yl; 4-oxooctahydro-1(2H)-quinolinyl; or a group of formula
  • Figure US20140206667A1-20140724-C00012
  • wherein R14 is pyridinyl; phenyl unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl; or a group of formula —V—R15 wherein V is unsubstituted C1-4-alkylene and R15 is phenyl or morpholinyl.
  • In a preferred embodiment, NR3R3a is 4-pyridin-2-ylpiperazin-1-yl, 4-(3-methylphenyl)piperazin-1-yl, 4-(4-hydroxyphenyl)piperazin-1-yl, 4-(2-phenylethyl)piperazin-1-yl, 4-(2-morpholin-4-ylethyl)piperazin-1-yl, 3-hydroxypiperidin-1-yl, thiomorpholin-4-yl, 4-methoxycarbonyl-1,3-thiazolidin-3-yl, 2,5-dihydro-1H-pyrrol-1-yl, 1,4-dioxa-8-azaspiro[4.5]dec-8-yl or 4-oxooctahydro-1(2H)-quinolinyl.
  • Generally, R5 is hydrogen, nitro, halogen, C1-4-alkyl, unsubstituted or substituted by halogen, or C1-4-alkoxy unsubstituted or substituted by halogen.
  • Preferably, R5 is hydrogen, methyl, ethyl, trifluoromethyl, trifluoromethoxy, n-propyl, isopropyl, nitro, or halogen. More preferably, R5 is halogen or trifluoromethyl.
  • Generally, R6 is hydrogen, C1-6-alkyl or halogen.
  • Preferably. R6 is hydrogen, methyl or Cl. More preferably. R6 is hydrogen.
  • Generally, R7 is hydrogen, methyl or halogen.
  • Preferably, R7 is hydrogen, methyl, Br, F or Cl. More preferably, R7 is hydrogen, Br or F.
  • Combinations of one or more of these preferred compound groups are especially preferred.
  • In a preferred embodiment, the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • Figure US20140206667A1-20140724-C00013
  • wherein R1 is hydrogen,
  • R2 is hydrogen or C1-4-alkyl,
  • R3 is hydrogen; C1-6-alkyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, hydroxy, alkoxy, alkoxycarbonyl or alkylamino; C5-7-cycloalkyl; (hydroxymethyl)cyclohexenyl; phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl, trifluoromethylthio or pyridinylalkyl; pyridinyl unsubstituted or substituted by methoxy; triazolyl; C1-4-alkoxy; or a group of formula —W—R8,
  • R3a is hydrogen, C1-4-alkyl or a group of formula
  • Figure US20140206667A1-20140724-C00014
  • or NR3R3a is piperidinyl unsubstituted or substituted by hydroxy; thiomorpholinyl; thiazolidinyl unsubstituted or substituted by C1-4-alkoxycarbonyl: 2,5-dihydro-1H-pyrrol-1-yl; 1,4-dioxa-8-azaspiro[4.5]dec-8-yl; 4-oxooctahydro-1(2H)-quinolinyl; or a group of formula
  • Figure US20140206667A1-20140724-C00015
  • R4 is hydrogen,
  • R5 is hydrogen; nitro; halogen; C1-4-alkyl, unsubstituted or substituted by halogen; or C1-4-alkoxy unsubstituted or substituted by halogen,
  • R6 is hydrogen, C1-6-allyl or halogen,
  • R7 is hydrogen, methyl or halogen,
  • W is C1-4-alkylene unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl or alkoxy; —NH—; or —NHC(═O)—,
  • R8 is phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4-alkyl, hydroxy, methoxy, nitro, methylsulfonyl or trifluoromethylthio; furyl unsubstituted or substituted by methyl; pyrazolyl; pyridinyl; morpholinyl; tetrahydrobenzazocinyl; piperidinyl unsubstituted or substituted by methyl; dihydroisochromenyl or dihydroimidazolyl,
  • R14 is pyridinyl; phenyl unsubstituted or substituted by halogen, hydroxy, C1-4-alkyl; or a group of formula —V—R15
  • V is unsubstituted C1-4-alkylene,
  • R15 is phenyl or morpholinyl,
  • m is 1 to 4,
  • and at least one of Rs, R6 or R7 is different from hydrogen when R2 is hydrogen. R1 is H or 2,6-diisopropylphenyl, and R3a is H.
  • In a more preferred embodiment, the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,
  • Figure US20140206667A1-20140724-C00016
  • R1 is hydrogen,
  • R2 is hydrogen, methyl or ethyl,
  • R3 is hydrogen, n-butyl, cycloheptyl, 2-fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2,2-dimethylpropyl, 1-(hydroxymethyl) propyl, 3,3,3-trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2-oxoethyl, 3-(dimethylamino) propyl, 6-(hydroxymethyl)cyclohex-3-en-1-yl, 3-hydroxyphenyl, 3-fluorophenyl, 3-(2-pyridin-2-ylethyl)phenyl, 3,4-dimethylphenyl, 4-tert-butylphenyl, benzyl, 4-hydroxy-3-methoxybenzyl, 4-methylsulfonylbenzyl, 2-nitrobenzyl, 2-chloro-6-fluorobenzyl, 2-[(trifluoromethyl)thio]benzyl, 2-hydroxy-2-phenylethyl, 2-(3,4-dimethoxyphenyl)ethyl, 2-(2-chlorophenyl)ethyl. 2-(4-methylphenyl)ethyl, (4-bromophenyl)amino, pyridin-3-yl, 6-methoxypyridin-3-yl, 4H-1,2,4-triazol-3-yl, pyridin-4-ylmethyl, (5-methyl-2-furyl)methyl, 3-(1H-pyrazol-1-yl) propyl, 2-morpholin-4-ylethyl, 2-((3,4,5,6-tetrahydro-1-benzazocin-1(2H)-yl) propyl, 2-(2-methylpiperidin-1-yl)ethyl, 3,4-dihydro-1H-isochromen-1-ylmethyl, methoxy, (4-pyridinylcarbonyl) amino or 4,5-dihydro-1H-imidazol-2-ylamino,
  • R3a is hydrogen, methyl or tetrahydrofuran-2-ylmethyl, or NR3R3a 4-pyridin-2-ylpiperazin-1-yl. 4-(3-methylphenyl)piperazin-1-yl, 4-(4-hydroxyphenyl)piperazin-1-yl, 4-(2-phenylethyl)piperazin-1-yl, 4-(2-morpholin-4-ylethyl)piperazin-1-yl, 3-hydroxypiperidin-1-yl, thiomorpholin-4-yl, 4-methoxycarbonyl-1,3-thiazolidin-3-yl, 2,5-dihydro-1H-pyrrol-1-yl, 1,4-dioxa-8-azaspiro[4.5]dec-8-yl or 4-oxooctahydro-1(2H)-quinolinyl,
  • R4 is hydrogen,
  • R5 is hydrogen, methyl, ethyl, trifluoromethyl, trifluoromethoxy, n-propyl, isopropyl, nitro or halogen.
  • R6 is hydrogen, methyl or Cl,
  • R7 is hydrogen, methyl, Br, F or Cl,
  • and at least one of R5, R6 or R7 is different from hydrogen when R2 is hydrogen, R3 is H or 2,6-diisopropylphenyl, and R3a is H.
  • More preferably. R2 is hydrogen or methyl. R3 is hydrogen, R3a is hydrogen, R5 is halogen or trifluoromethyl, R6 is hydrogen and R7 is hydrogen, Br or F.
  • In all the above-mentioned scopes, when R2 is C1-20-alkyl, the carbon atom to which R2 is attached is preferably in the “S”-configuration.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-iodo-2-oxo-2,3-dihydro-H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5,7-dibromo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-nitro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-methyl-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide, (2R)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; (2S)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; 2-[2-oxo-5-(trifluoromethoxy)-2,3-dihydro-1H-indol-1-yl]acetamide; 2-(5-isopropyl-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-ethyl-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-fluoro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5,7-dimethyl-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-bromo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(2-oxo-5-propyl-2,3-dihydro-1H-indol-1-yl)acetamide; 2-[2-oxo-5-(trifluoromethyl)-2,3-dihydro-1H-indol-1-yl]acetamide; 2-(5,6-dimethyl-2-oxo-2,3-dihydro-1H-indol-1I-yl)acetamide; 2-(7-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(6-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) butanamide; (+)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) butanamide; (−)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) butanamide; 2-(5-methyl-2-oxo-2,3-dihydro-1H-indol-1-yl)propanamide, (+)-2-(5-methyl-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; (−)-2-(5-methyl-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; 2-(5-bromo-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; (−)-2-(5-bromo-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide (+)-2-(5-bromo-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; 2-(5-chloro-7-fluoro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3-hydroxyphenyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3-fluorophenyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[3-(2-pyridin-2-ylethyl)phenyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[6-(hydroxymethyl)cyclohex-3-en-1-yl]actinide; 5-chloro-1-[2-oxo-2-(4-pyridin-2-ylpiperazin-1-yl)ethyl3-1,3-dihydro-2H-indol-2-one; 5-chloro-1-{2-[4-(3-methylphenyl)piperazin-1-yl]-2-oxoethyl})-1,3-dihydro-2H-indol-2-one; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(4-hydroxy-3-methoxybenzyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(pyridin-4-ylmethyl)-N-(tetrahydrofuran-2-ylmethyl)acetamide; 5-chloro-1-[2-(3-hydroxypiperidin-1-yl)-2-oxoethyl]-1,3-dihydro-2H-indol-2-one; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N′-isonicotinoylacetohydrazide; 5-chloro-1-(2-oxo-2-thiomorpholin-4-ylethyl)-1,3-dihydro-2H-indol-2-one; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(4H-1,2,4-triazol-3-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[4-(methylsulfonyl)benzyl]acetamide; 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetyl]octahydroquinolin-4 (1H)-one; N′-(4-bromophenyl)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) acetohydrazide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(6-methoxypyridin-3-yl)acetamide; N-butyl-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3-hydroxypropyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[3-(dimethylamino) propyl]acetamide; 5-chloro-1-{2-oxo-2[4-(2-phenylethyl)piperazin-1-yl]ethyl}-1,3-dihydro-2H-indol-2-one; ethyl {[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetyl]amino}acetate; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3-ethoxypropyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(2-fluoroethyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-methoxy-N-methylacetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3,4-dimethylphenyl)acetamide; N-(4-tert-butylphenyl)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3-hydroxy-2,2-dimethylpropyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[1-(hydroxymethyl) propyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3,3,3-trifluoro-2-hydroxypropyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(2-hydroxy-2-phenylethyl)acetamide; 5-chloro-1-{2-[4-(4-hydroxyphenyl)piperazin-1-yl]-2-oxoethyl}-1,3-dihydro-2H-indol-2-one; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(pyridin-4-ylmethyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[(5-methyl-2-furyl)methyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[3-(1H-pyrazol-1-yl) propyl]acetamide; methyl 3-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetyl]-1,3-thiazolidine-4-carboxylate; 5-chloro-1-[2-(2,5-dihydro-1H-pyrrol-1-yl)-2-oxoethyl]-1,3-dihydro-2H-indol-2-one; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N′-(4,5-dihydro-1H-imidazol-2-yl)acetohydrazide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[2-(3,4-dimethoxyphenyl)ethyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[2-(2-chlorophenyl) etl-lyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[2-(4-methylphenyl)ethyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(2-morpholin-4-ylethyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[2-(3,4,5,6-tetrahydro-1-benzazocin-1 (2H)-yl) propyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-[2-(2-methylpiperidin-1-yl)ethyl]acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(2-nitrobenzyl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-(3,4-dihydro-1H-isochromen-1-ylmethyl)acetamide; N-(2-chloro-6-fluorobenzyl)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; N-benzyl-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-methylacetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-{2-[(trifluoromethyl) thio]benzyl}acetamide; 5-chloro-1-[2-(1,4-dioxa-8-azaspiro[4.5]dec-8-yl)-2-oxoethyl]-1,3-dihydro-2H-indol-2-one; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-cycloheptylacetamide; 5-chloro-1-(2-[4-(2-morpholin-4-ylethyl)piperazin-1-yl]-2-oxoethyl)-1,3-dihydro-2H-indol-2-one; and 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)-N-pyridin-3-ylacetamide.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-iodo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; 2-(5,7-dibromo-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide; (2S)-2-(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl) propanamide; 2-[2-oxo-5-(trifluoromethyl)-2,3-dihydro-1H-indol-1-yl]acetamide and 2-(5-chloro-7-fluoro-2-oxo-2,3-dihydro-1H-indol-1-yl)acetamide.
  • In another embodiment, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-chloro-2-oxo-2,3-dihydro-H-indol-1-yl)acetamide and (2S)-2-(5-chloro-2-oxo-2,3-dihydro-H-indol-1-yl) propanamide.
  • iv) U.S. Pat. No. 7,244,747:
  • A compound having the formula I or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00017
  • wherein R1 is hydrogen, C1-20 alkyl, C3-8 cycloalkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, guanidine, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl, arylsulfinyl, aryl or heterocycle;
  • R2 is hydrogen, C1-20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl:
  • R3 is hydrogen, C1-20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;
  • or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00018
  • R4 is hydrogen, C1-20 alkyl, C2-12 alkenyl, C2-12 alkynyl, aryl, azido, alkoxycarbonylamino, arylsulfonyloxy or heterocycle;
  • R4a is hydrogen or C1-20 alkyl;
  • or R4 and R4a can form together a C3-8 cycloalkyl;
  • R5 is hydrogen;
  • or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • R6 is hydrogen or C1-20 alkyl;
  • R7 is hydrogen;
  • or R6 and R7 are linked together to form a C3-6 cycloalkyl;
  • R8 is hydrogen, halogen, nitro, cyano, C1-20 alkyl or alkoxy:
  • R9 is hydrogen, C1-20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
  • R10 is hydrogen, C1-20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
  • R11 is hydrogen, halogen, nitro, cyano, C1-20 alkyl or alkoxy;
  • R12 is hydrogen or halogen;
  • R13 is hydrogen, nitro, halogen, heterocycle, amino, aryl, C1-20 alkyl unsubstituted or substituted by halogen, or alkoxy unsubstituted or substituted by halogen;
  • R14 is hydrogen, C1-20 alkyl or halogen;
  • R15 is hydrogen, C1-20 alkyl or halogen;
  • with the proviso that R4 is different from hydrogen when represents a group of formula
  • Figure US20140206667A1-20140724-C00019
  • The asterisk * indicates the point of attachment of the substituents.
  • In a preferred embodiment, the compounds have the formula I, their tautomers, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00020
  • wherein R1 is hydrogen, C1-20 alkyl, C3-8 cycloalkyl, halogen, hydroxy, ester, amido, cyano, nitro, amino, guanidine, alkylthio, alkylsulfonyl, alkylsulfinyl, aryl or heterocycle;
  • R2 is hydrogen, C1-20 alkyl, halogen, cyano, ester, carbamate or amido;
  • R3 is hydrogen, cyano, C1-20 alkyl, halogen or ester; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00021
  • R4 is hydrogen, C1-20 alkyl, C2-12 alkenyl or aryl;
  • R4a is hydrogen;
  • R5 is hydrogen;
  • or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00022
  • R6 is hydrogen or C1-20 alkyl;
  • R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl:
  • R8 is hydrogen;
  • R9 is hydrogen, C1-20 alkyl, halogen or alkoxy;
  • R10 is hydrogen, C1-20 alkyl, halogen or cyano;
  • R11 is hydrogen;
  • R12 is hydrogen or halogen;
  • R13 is hydrogen, halogen, heterocycle or C1-20 alkyl;
  • R14 is hydrogen:
  • R15 is hydrogen;
  • with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00023
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00024
  • The term “alkyl”, as used herein, represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched or cyclic or combinations thereof and containing 1-20 carbon atoms, preferably 1-10 carbon atoms, more pre preferred alkyl groups have 1-3 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, cyano, azido, aryloxy, alkoxy, alkylthio, alkanoylamino, arylcarbonylamino, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl or aryl. Usually alkyl groups, in the present case, are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, n-heptyl, 2,4,4-trimethylpentyl, n-decyl, chloromethyl, trifluoromethyl, 2-bromo-2,2-difluoroethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino)methyl, (propionylamino)methyl. (benzoylamino)methyl, (4-chlorophenoxy)methyl, benzyl, 2-phenylethyl or 2-(methylthio)ethyl. Preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, 2,4,4-trimethylpentyl, chloromethyl, trifluoromethyl. 2,2,2-trifluoroethyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino)methyl, (propionylamino)methyl. (benzoylamino)methyl or 2-(methylthio)ethyl. More preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, azidomethyl or trifluoromethyl. Most preferred alkyl groups are methyl or n-propyl.
  • The term “cycloalkyl”, as used herein, represents a monovalent group of 3 to 8 carbon atoms, usually 3-6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl groups are cyclopropyl and cyclohexyl.
  • The term “alkenyl” as used herein, represents straight, branched or cyclic unsaturated hydrocarbon radicals or combinations thereof having at least one carbon-carbon double bond, containing 2-12 carbon atoms, preferably usually 2-4 carbon atoms. Alkenyl groups are being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Usually an alkenyl group is ethenyl (vinyl) optionally substituted by 1 to 3 halogens. Preferred alkenyl group, in the present case, is 2,2-difluorovinyl.
  • The term a “alkynyl” as used herein, represents straight, branched or cyclic hydrocarbon radicals or combinations thereof containing at least one carbon-carbon triple bond, containing 2-12 carbon atoms, preferably 2-6 carbon atoms, and being optionally substituted by any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferably an alkynyl group is a halogenoalkynyl group (haloalkynyl group).
  • Groups qualified by prefixes such as “s”, “i”, “t” and the like (e.g. “i-propyl”, “s-butyl”) are branched derivatives.
  • The term “aryl” as used herein, is defined as phenyl optionally substituted by 1 to 4 substituents independently selected from halogen, cyano, alkoxy, alkylthio, C1-3 alkyl or azido, preferably halogen or azido. Usually aryl groups, in the present case are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl. 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl. Preferably, aryl groups are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl. Most preferred aryl groups are phenyl. 3-chlorophenyl, 3-fluorophenyl, 3,5-difluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • The term “heterocycle”, as used herein, is defined as including an aromatic or non aromatic cycloalkyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure. Heterocyclic ring moieties can be optionally substituted by alkyl groups or halogens and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl. Usually heterocycles are 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-tetrahydrofuranyl, 1H-pyrrol-2-yl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrazol-2-yl, 1H-pyrazol-3-yl, 4-chloro-1-methyl-1H-pyrazol-3-yl, 5-chloro-1,3-dimethyl-1H-pyrazol-4-yl, 1,2,3-thiadiazol-4-yl, 3,5-dimethyl-4-isothiazyl, 1H-imidazol-2-yl, 1-methyl-1H-imidazol-2-yl, 4-methyl-1H-imidazol-5-yl, or 2-methyl-1,3-thiazol-4-yl. Preferred heterocycles are 1H-imidazol-2-yl, 1,2,3-thiadiazol-4-yl, 1H-pyrazol-3-yl. 2-furyl, 3-furyl, 2-thienyl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrrol-2-yl.
  • The term “halogen”, as used herein, includes an atom of chlorine, bromine, fluorine, iodine. Usually halogens are chlorine, bromine and fluorine. Preferred halogens are fluorine, bromine and chlorine.
  • The term “hydroxy”, as used herein, represents a group of formula —OH.
  • The term “alkoxy”, as used herein, represents a group of formula —ORa
  • wherein Ra is an alkyl group, as defined above. Preferred alkoxy group is methoxy.
  • The term “aryloxy”, as used herein, represents a group of formula —ORb wherein Rb is an aryl group, as defined above. Preferred aryloxy group is phenoxy.
  • The term “ester”, as used herein, represents a group of formula —COORc wherein Rc is an alkyl group or aryl group, as defined above. Preferred ester group is methoxycarbonyl.
  • The term “amido”, as used herein, represents a group of formula —CONH2.
  • The term “amino”, as used herein, represents a group of formula —NH2.
  • The term “aminoderivative”, as used herein, represents an alkylamino or an arylamino group, wherein the terms “alkyl” and “aryl” are defined as above.
  • The term “cyano”, as used herein, represents a group of formula —CN.
  • The term “nitro”, as used herein, represents a group of formula —NO,.
  • The term “azido”, as used herein, represents a group of formula —N3.
  • The term “guanidine”, as used herein, represents a group of formula —NHC(═NH)NH2.
  • The term “alkylthio”, as used herein, represents a group of formula —SRd wherein Rd is an alkyl group, as defined above. Preferred alkylthio group is methylthio.
  • The term “alkylsulfonyl”, as used herein, represents a group of formula —S(═O)2R wherein Re is an alkyl group, as defined above. Preferred alkylsulfonyl group is methylsulfonyl.
  • The term “alkylsulfinyl”, as used herein, represents a group of formula —S(═O)Rf wherein Rf is an alkyl group, as defined above. Preferred alkylsulfinyl group is methylsulfinyl.
  • The term “arylthio”, as used herein, represents a group of formula —SRg wherein Rg is an aryl group, as defined above.
  • The term “arylsulfonyl”, as used herein, represents a group of the formula —S(═O)2Rh wherein Rh is an aryl group, as defined above.
  • The term “arylsulfinyl”, as used herein, represents a group of the formula —S(═O)R1 wherein R1 is an aryl group, as defined above.
  • The term “carbamate” as used herein, represents a group of formula —N(H)C(O)ORj, wherein Rj is an alkyl or an aryl, as defined above. Usually carbamate groups are (propoxycarbonyl)amino or (benzyloaxycarbonyl)amino. Preferred carbamate group is (benzyloaxycarbonyl)amino.
  • The term “alkanoylamino” as used herein, represents a group of the formula —NHC(═O)Rk wherein Rk is an alkyl group, as defined above.
  • The term “(arylcarbonyl)amino” as used herein, represents a group of the formula —NHC(═O)Rm wherein Rm is an aryl group, as defined above.
  • Preferred (arylcarbonyl)amino is benzoylamino.
  • Usually, R1 is hydrogen; C1-10 alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; hydroxy; CL cycloalkyl; halogen; ester; amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl groups; or guanidine. Preferably, R1 is hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl: n-butyl: i-butyl; t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; hydroxymethyl; chloromethyl; trifluoromethyl; 2,2,2-trifluoroethyl; cyanomethyl; 2-(methylthio)ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl or 1H-imidazol-2-yl. More preferably, R1 is hydrogen; methyl; ethyl: i-propyl; n-propyl; n-butyl; methylthio; nitro; cyano; amino: chloro or 1H-pyrrol-2-yl. Most preferably, R1 is hydrogen; methyl; methylthio; nitro; cyano; amino or chloro.
  • Usually, R2 is hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino: halogen; ester; cyano; alkyl carbamate; [(N-methoxy-N-methyl)amino]carbonyl. Preferably, R2 is hydrogen; methyl; hydroxymethyl; (acetylamino)methyl; (propionylamino)methyl; (benzoylamino)methyl: [(benzyloxy)carbonyl]amino; chloro or cyano. More preferably, R2 is hydrogen; chloro or cyano.
  • Usually, R3 is hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano. Preferably, R3 is hydrogen; hydroxymethyl; chloro; cyano. More preferably, R3 is hydrogen or cyano. Most preferred R3 is hydrogen.
  • Usually, R4 is hydrogen; C1-4 alkyl unsubstituted or substituted by halogens; C2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens. Preferably, R4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl: 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl. More preferably, R4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl. Most preferably, R4 is n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • Usually, R4a is hydrogen.
  • Usually, R5 is hydrogen.
  • Usually, R6 is hydrogen or C1-10 alkyl unsubstituted or substituted by hydroxy or azido. Preferably, R6 is hydrogen or azidomethyl. More preferably R6 is hydrogen.
  • Usually R7 is hydrogen.
  • In other preferred embodiments, R6 and R7 are linked to form a cyclopropyl.
  • In other preferred embodiments, R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00025
  • Usually, R8 is hydrogen.
  • Usually, R9 is hydrogen; halogen; C1-3 alkyl or alkoxy. Preferably, R9 is hydrogen; methyl; chloro or methoxy. More preferred R9 is hydrogen.
  • Usually, R10 is hydrogen; halogen; cyano; C1-3 alkyl unsubstituted or substituted by halogens; or alkoxy. Preferably. R10 is methyl; hydrogen; trifluoromethyl; fluoro; cyano or methoxy. More preferred R10 is hydrogen; trifluoromethyl; fluoro or cyano.
  • Usually, R11 is hydrogen.
  • In other preferred embodiments, R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00026
  • Usually, R12 is hydrogen or halogen. Preferably R12 is hydrogen; chloro or fluoro. More preferred R12 is hydrogen.
  • Usually, R13 is hydrogen; C1-3 alkyl; halogen or thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl. Preferably R13 is hydrogen; chloro; bromo or methyl. Most preferred R13 is chloro; bromo or methyl.
  • Usually R14 is hydrogen.
  • Usually, R15 is hydrogen.
  • Combinations of one or more of these preferred compound groups are especially preferred.
  • Generally, among the embodiments, the compounds of formula I, or pharmaceutically acceptable salts thereof, are those wherein
  • R1 is selected from hydrogen; C1-10alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; C3-6 cycloalkyl: halogen; ester; amido; nitro; cyano: amino; phenyl; alkylthio: alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl group; or guanidine;
  • R2 is selected from hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate or [(N-methoxy-N-methyl)amino]carbonyl.
  • R3 is selected from hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy: halogen; ester or cyano;
  • R4 is selected from hydrogen; C1-4 alkyl unsubstituted or substituted by halogens; C2-4alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens;
  • R4a is hydrogen;
  • R5 is hydrogen;
  • R6 is selected from hydrogen or C1-10 alkyl unsubstituted or substituted by hydroxy or azido;
  • R7 is hydrogen;
  • or R6 and R7 can be linked to form a cyclopropyl;
  • or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00027
  • R8 is hydrogen;
  • R9 is selected from hydrogen; halogen; C1-3 alkyl; alkoxy;
  • R10 is selected from hydrogen; halogen; cyano or C1-3 alkyl unsubstituted or substituted by halogens; or alkoxy;
  • R11 is hydrogen;
  • or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00028
  • R12 is selected from hydrogen or halogen;
  • R13 is selected from hydrogen; C1-3 alkyl; halogen; thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl;
  • R14 is hydrogen;
  • R15 is hydrogen;
  • with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00029
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00030
  • In a preferred embodiment, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein
  • R1 is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl: n-butyl: i-butyl: t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; trifluoromethyl; 2,2,2-trifluoroethyl; hydroxymethyl: chloromethyl; cyanomethyl; 2-(methylthio)ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl; or 1H-imidazol-2-yl;
  • R2 is selected from hydrogen; methyl; hydroxymethyl; (acetylamino)methyl; (propionylamino)methyl; (benzoylamino)methyl; (benzyloxycarbonyl)amino; chloro; or cyano;
  • R3 is selected from hydrogen; hydroxymethyl; chloro; cyano;
  • or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00031
  • R8 is hydrogen;
  • R9 is selected from hydrogen; methyl; choro; methoxy;
  • R10 is selected from methyl; hydrogen; trifluoromethyl; fluoro; cyano; or methoxy;
  • R11 is hydrogen;
  • R4 is selected from hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl: 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl; or 3-azido-2,4,6-trifluorophenyl.
  • R4a is hydrogen; R5 is hydrogen;
  • or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00032
  • R12 is selected from hydrogen; chloro; fluoro;
  • R13 is selected from hydrogen; chloro; bromo; methyl;
  • R14 is hydrogen;
  • R15 hydrogen;
  • R6 is selected from hydrogen; azidomethyl;
  • R7 is hydrogen;
  • or R6 and R7 are linked to form a cyclopropyl;
  • with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00033
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00034
  • In a more preferred embodiment, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein
  • R1 is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; n-butyl; methylthio; nitro; cyano; amino; chloro; or 1H-pyrrol-2-yl;
  • R2 is selected from hydrogen; chloro; cyano;
  • R3 is selected from hydrogen; cyano;
  • or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00035
  • R8 is hydrogen;
  • R9 is hydrogen;
  • R10 is selected from hydrogen; trifluoromethyl: fluoro; cyano;
  • R11 is hydrogen;
  • R4 is selected from hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; or 3-azido-2,4-difluorophenyl;
  • R4a is hydrogen;
  • R5 is hydrogen;
  • or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00036
  • wherein
  • R12 is hydrogen:
  • R13 is selected from methyl; chloro; bromo;
  • R14 is hydrogen;
  • R15 hydrogen;
  • R6 is hydrogen;
  • R7 is hydrogen;
  • with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00037
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00038
  • In a most preferred embodiment, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein
  • R1 is selected from hydrogen; methyl; methylthio; nitro; cyano; amino; chloro;
  • R2 is selected from hydrogen; chloro; cyano;
  • R3 is hydrogen;
  • R4 is selected from n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl: 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl;
  • R4a is hydrogen;
  • R5 is hydrogen;
  • or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00039
  • R12 is hydrogen:
  • R13 is selected from chloro; bromo; methyl;
  • R14 is hydrogen;
  • R15 hydrogen;
  • R6 is hydrogen;
  • R7 is hydrogen.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 4-(3-azido-2,4,6-trifluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-[(2-ethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one: 1-[(2-isopropyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-phenyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-[(2-propyl-1H-imidazol-1-yl)methyl]pyrrolidin-2-one; (+)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; 4-(2,2-difluorovinyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 1-{[2-(methylthio)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[2-(methylsulfinyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(2-tert-butyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[1-(1H-imidazol-1-yl)cyclopropyl]pyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one; 1-{[2-(methylsulfonyl)-1H-imidazol-1-yl]methyl}-propylpyrrolidin-2-one; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-2-carboxamide, 4-(4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,5-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chloro-4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(4-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,4-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-(hydroxymethyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; methyl 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-2-carboxylate; 1-[(2-nitro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-2-carbonitrile; 1-[(2-amino-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2,4-dichloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(5-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; 1-([2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl-1H-imidazole-4-carbonitrile; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (+)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (−)-1-{[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (+)-1-{[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (−)-1-{[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-trifluorophenyl)-1-pyrrolidinyl]methyl}-1H-imidazole-4-carbonitrile; (−)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (+)-1-{[2-oxo-4-(2,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (−)-1-{[2-oxo-4-(2,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (−)-1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (−)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5- -carbonitrile; 1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5- -carbonitrile; 1-[(5-methyl-2-phenyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-phenyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-ethyl-5-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2,5-dimethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one: 1-[2-azido-1-(1H-imidazol-1-yl)ethyl]-4-propylpyrrolidin-2-one; 1-[(4-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; 1-[(2-bromo-4,5-dichloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 1-{[5-(hydroxymethyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[4-(hydroxymethyl)-1H-imidazol-1-yl]methyl})-4-propylpyrrolidin-2-one: benzyl 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazol-5-ylcarbamat-e; N-[(1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazol-5-yl)methyl]acetamide; N-[(1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl)-1H-imidazol-5-yl)methyl}benzamide; N-[(1-[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazol-5-yl)methyl]propanamide; 1-(1H-benzimidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one: 4-propyl-1-[(2-propyl-1H-benzimidazol-1-yl)methyl]pyrrolidin-2-one; 1-[(2-isopropyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}pyrrolidin-2- -one; 1-([2-(methylthio)-1H-benzimidazol-1-yl]methyl)-4-propylpyrrolidin-2- -one; 1-[(2-amino-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[2-(chloromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-on-e; {1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazol-2-yl}acetoni-trile; 1-[(5-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one-; 1-[(5-methyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5,6-dimethyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[2-isopropyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl)-4-propyl-pyrrolidin-2-one; 1-[(6-chloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-propyl-1H-benzimidazole-5-car-bonitrile; 1-[2-ethyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl)-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl})pyrrolidin-2-one; 1-[(5-fluoro-2-propyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin- -2-one; 1-{[6-methyl-2-(1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl}-4-pro-pylpyrrolidin-2-one; 1-[(6-methoxy-2-propyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 2-butyl-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-5- -carbonitrile; 1-{[2-[2-(methylthio)ethyl]-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(5-fluoro-2-isobutyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2- -one; 1-{[5-fluoro-2-(2,4,4-trimethylpentyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 2-cyclopropyl-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-5-carbonitrile; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-(1H-pyrazol-3-yl)-1H-benzimidazole-5-carbonitrile; 1-[(2-cyclopropyl-5-fluoro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-fluoro-2-isopropyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[2-(3-furyl)-6-methoxy-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin- -2-one; 1-[(2-cyclopropyl-6-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylp-yrrolidin-2-one; 1-[(2-isopropyl-6-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin- -2-one; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-(1,2,3-thiadiazol-4-yl-)-1H-benzimidazole-5-carbonitrile; 1-{[2-(1H-imidazol-2-yl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}- -4-propylpyrrolidin-2-one; 1-{[5-fluoro-2-(2,2,2-trifluoroethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[2-(1-ethylpropyl)-6-methoxy-1H-benzimidazol-1-yl]methyl}-4-propylpyrr-olidin-2-one; 1-{[6-methoxy-2-(1-methyl-1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one, 1-{[2-(2-furyl)-5-(trifluoromethyl)-11H-benzimidazol-1-yl]methyl}-4-propyl-pyrrolidin-2-one; 4-propyl-1-{[2-thien-2-yl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl-}pyrrolidin-2-one; 1-{[2-(3-furyl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propyl-pyrrolidin-2-one; 1-[2-cyclopropyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl)-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(1H-pyrrol-2-yl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]-methyl}pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-bromo-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 4-fluoro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 4-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 1-(1H-imidazol-1-ylmethyl)-5-methyl-1,3-dihydro-2H-indol-2-one; 1-[(2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbonitrile; and 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-c-arbonitrile.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one, 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-[(2-ethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-isopropyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-[(2-propyl-1H-imidazol-1-yl)methyl]pyrrolidin-2-one; (+)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; 4-(2,2-difluorovinyl)-1-(H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-{[2-(methylthio)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one; 4-(4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,5-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chloro-4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(4-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,4-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(2-nitro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-2-carbonitrile; 1-[(2-amino-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one: 1-[(5-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; 1-[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; 1-[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl)-1H-imidazole-5- -carbonitrile; (+)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (+); 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4- -carbonitrile; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; 1-[2-azido-1-(1H-imidazol-1-yl)ethyl]-4-propylpyrrolidin-2-one: 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-propyl-1H-benzimidazole-5-car-bonitrile; 1-{[2-ethyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 4-propyl-1-[2-(1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl)pyrrolidin-2-one; 1-[(5-fluoro-2-propyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin- -2-one; 2-butyl-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole- -5-carbonitrile; 1-[(5-fluoro-2-isopropyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-bromo-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 1-(1H-imidazol-1-ylmethyl)-5-methyl-1,3-dihydro-2H-indol-2-one; 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbo-nitrile.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one: 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 4-(2,2-difluorovinyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-on-e; 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-{[2-(methylthio)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,5-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 1-(1H-imidazol-1-ylmethyl)-4-(2,3,4-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(2-nitro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-2-carbonitrile; 1-[(2-amino-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; (+)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 5-bromo-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 1-(1H-imidazol-1-ylmethyl)-5-methyl-1,3-dihydro-2H-indol-2-one; 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbo-nitrile.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one.
  • v) International Patent Application WO 2007/065595:
  • Compounds having formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00040
  • wherein
  • R1 is hydrogen or C1-6 alkyl;
  • R2 is hydrogen or C1-4alkyl;
  • R3 is a group of formula —CHR5R6 or a benzyl group;
  • R4 is C1-8 is alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle;
  • R5 is C2-4 alkyl:
  • R6 is C2-4 alkyl, amido or —COOR7;
  • R7 is C1-4 alkyl;
  • Usually when R3 is a benzyl group, then R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl.
  • Usually when R3 is a group of formula —CHR5R6 then R4 is C1-8 alkyl optionally substituted by C3-6 cycloalkyl, aryl or heterocycle.
  • The term “alkyl”, as used herein, is a group which represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched moieties, or combinations thereof, and containing 1-8 carbon atoms, preferably 1-6 carbon atoms; more preferably alkyl groups have 1-4 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl, acyl, aryl or heterocycle. Alkyl moieties may be optionally substituted by a cycloalkyl as defined hereafter. Preferred alkyl groups are methyl, cyanomethyl, ethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-butyl, i-butyl, n-pentyl, 3-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl. 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl. 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro-2-furyl)methyl. More preferred alkyl groups are methyl, ethyl, cyanomethyl, 2-methoxyethyl, n-propyl, 3-hydroxypropyl, 2-propynyl, n-butyl, 3-pentyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro-2-furyl)methyl. Most preferred alkyl groups are methyl, ethyl, 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • The term “cycloalkyl”, as used herein, represents a monovalent group of 3 to 8, preferably 3 to 6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl group is cyclohexyl.
  • The term “aryl” as used herein, is defined as a phenyl group optionally substituted by 1 to 4 substituents independently selected from halogen, amino, nitro, alkoxy or aminosulfonyl. Preferred aryl groups are phenyl, 2-bromophenyl, 3-bromophenyl, 4-bromophenyl, 3-methoxyphenyl, 3-nitrophenyl, 3-aminophenyl or 4-(aminosulfonyl)phenyl.
  • The term “phenyl”, as used herein, represents an aromatic hydrocarbon group of formula —C6H5.
  • The term “benzyl group”, as used herein, represents a group of formula —CH2-aryl. Preferred benzyl groups are benzyl, 2-bromobenzyl, 3-bromobenzyl. 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl or 4-(aminosulfonyl)benzyl. More preferred benzyl groups are benzyl. 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl or 3-aminobenzyl. Most preferred alkyl groups are 3-methoxybenzyl or 3-nitrobenzyl.
  • The term “halogen”, as used herein, represents an atom of fluorine, chlorine, bromine, or iodine. Preferred halogen is bromine.
  • The term “hydroxy”, as used herein, represents a group of formula —OH.
  • The term “cyano”, as used herein, represents a group of formula —CN.
  • The term “amino”, as used herein, represents a group of formula —NH2.
  • The term “ethynyl”, as used herein, represents a group of formula —C≡CH.
  • The term “alkoxy”, as used herein, represents a group of formula —ORa wherein Ra is an alkyl group, as defined above. Preferred alkoxy group is methoxy.
  • The term “nitro”, as used herein, represents a group of formula —NO2.
  • The term “amido”, as used herein, represents a group of formula —C(═O)NH2.
  • The term “acyl”, as used herein, represents a group of formula —C(═O)Rb wherein Rb is an alkyl group, as defined here above. Preferred acyl group is acetyl (—C(═O)Me).
  • The term “alkoxycarbonyl (or ester)”, as used herein, represents a group of formula —COORc wherein Rc is an alkyl group; with the proviso that Rc does not represent an alkyl alpha-substituted by hydroxy. Preferred alkoxycarbonyl group is ethoxycarbonyl.
  • The term “heterocycle”, as used herein, represents a 5-membered ring containing one or two heteroatoms selected from O or N. The heterocycle may be substituted by one or two C1-4 alkyl or nitro. Preferred heterocycles are (3,5-dimethylisoxazol-4-yl) or (5-nitro-2-furyl). Most preferred heterocycle is (5-nitro-2-furyl).
  • Generally R1 is hydrogen or C1-6 alkyl. Usually R1 is hydrogen or C1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl. Preferably R1 is hydrogen, methyl, cyanomethyl. 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-pentyl or n-hexyl. More preferably R1 is hydrogen, methyl, cyanomethyl. 2-methoxyethyl, n-propyl, 3-hydroxypropyl or 2-propynyl. Most preferably R1 is hydrogen.
  • Generally R2 is hydrogen or C1-4 alkyl. Usually R2 is hydrogen or unsubstituted C1-4 alkyl. Preferably R2 is hydrogen, methyl or n-butyl. More preferably. R2 is methyl.
  • Generally R3 is a group of formula —CHRSR6 or a benzyl group. Preferably R3 is 3-pentyl, 1-(aminocarbonyl)propyl. 1-(ethoxycarbonyl)propyl or 3-bromobenzyl. Most preferably R3 is 1-(ethoxycarbonyl)propyl.
  • Generally R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle. Usually R4 is C1-8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl. 5-nitro-2-furyl or ethoxycarbonyl. Preferably R4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl. More preferably R4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl. Most preferably R4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • Generally R5 is C2-4alkyl. Usually R5 is unsubstituted C2-44 alkyl. Preferably R5 is ethyl.
  • Generally R6 is C2-4alkyl, amido or —COOR7. Usually R6 is unsubstituted C2-4alkyl, amido or —COOR7. Preferably R6 is ethyl, amido or ethoxycarbonyl. Most preferably R6 is ethoxycarbonyl.
  • Generally R7 is C1-4 alkyl. Usually R7 is unsubstituted C1-4 alkyl. Preferably, R7 is ethyl.
  • In some embodiments, the compounds are those having formula I, and their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00041
  • wherein
  • R1 is hydrogen, C1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl;
  • R2 is hydrogen or unsubstituted C1-4 alkyl;
  • R3 is a group of formula —CHR5R6 or a benzyl group;
  • R4 is C1-8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl, 5-nitro-2-furyl or ethoxycarbonyl;
  • R5 is unsubstituted C2-4 alkyl;
  • R6 is unsubstituted C2-4 alkyl, amido or —COOR7;
  • R7 is unsubstituted C1-4 alkyl; with the proviso that when R1 is hydrogen, R2 is methyl, R3 is —CHR5R6, R6 is ethoxycarbonyl and R5 is ethyl, then R4 is different from n-propyl, i-propyl, n-pentyl, n-heptyl, 3-bromobenzyl, 4-chlorobenzyl, 4-methylbenzyl or 2-phenylethyl.
  • In the above embodiment, preferably, when R3 is a benzyl group, then R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl.
  • In the above embodiment, preferably, when R3 is a group of formula —CHR5R6, then R4 is C1-8 alkyl optionally substituted by C3-6 cycloalkyl, aryl or heterocycle.
  • In a preferred embodiment,
  • R1 is hydrogen, methyl, cyanomethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-pentyl or n-hexyl;
  • R2 is hydrogen, methyl or n-butyl;
  • R3 is 3-pentyl. 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl;
  • R4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl; with the proviso that when RL is hydrogen, R2 is methyl and R3 is 1-(ethoxycarbonyl)propyl, then R4 is different from n-pentyl, 3-bromobenzyl or 2-phenylethyl.
  • In the above embodiment, preferably, when R3 is 3-bromobenzyl, then R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl.
  • In the above embodiment, preferably, when R1 is 3-pentyl, 1-(aminocarbonyl)propyl or 1-(ethoxycarbonyl)propyl, then R4 is different from 1-(ethoxycarbonyl)propyl.
  • In a more preferred embodiment,
  • R1 is hydrogen, methyl, cyanomethyl, 2-methoxyethyl, n-propyl, 3-hydroxypropyl or 2-propynyl;
  • R2 is methyl;
  • R3 is 3-pentyl, 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl;
  • R4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl;
  • with the proviso that when R1 is hydrogen, R2 is methyl and R3 is 1-(ethoxycarbonyl)propyl, then R4 is different from 3-bromobenzyl.
  • In the above embodiment, preferably, when R3 is 3-bromobenzyl, then R4 is 1-(ethoxycarbonyl)propyl;
  • In the above embodiment, preferably, when R3 is 3-pentyl, 1-(aminocarbonyl)propyl or 1-(ethoxycarbonyl)propyl, then R4 is different from 1-(ethoxycarbonyl)propyl;
  • In a most preferred embodiment, R1 is hydrogen; R2 is methyl; R3 is 1-(ethoxycarbonyl)propyl; and R4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • A further embodiment consists in compounds wherein R2 is methyl, R3 is a group of formula —CHR5R6 with R5 being C2-4 alkyl. R6 being amido or —COOR7 and R7 being methyl or ethyl.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-[(7-benzyl-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(2-ethoxy-2-oxoethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(2-methoxyethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(2-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(cyanomethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-propyl-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-(2-oxopropyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(3-hydroxypropyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-(2-propynyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio)butanoate; ethyl 2-{[7-(3-aminobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-({7-[4-(aminosulfonyl)benzyl]-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-{[7-(4-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(cyclohexylmethyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[1,3-dimethyl-2,6-dioxo-7-(1-phenylethyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[1,3-dimethyl-2,6-dioxo-7-(2-phenylethyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-({7-[(3,5-dimethylisoxazol-4-yl)methyl]-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate: ethyl 2-[(1,7-dihexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate: ethyl 2-[(7-hexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-[(3-methyl-2,6-dioxo-1,7-dipentyl-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate: 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanamide; 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanamide; 7-(3-bromobenzyl)-8-[(1-ethylpropyl)thio]-3-methyl-3,7-dihydro-1H-purine-2,6-dione; ethyl 2-{8-[(3-bromobenzyl)thio]-1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl}butanoate; and ethyl 2-[(7-isobutyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-[(7-benzyl-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(2-methoxyethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}) butanoate; ethyl 2-{[7-(3-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-([7-(3-bromobenzyl)-1-(cyanomethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-propyl-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(3-hydroxypropyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-(2-propynyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio)butanoate; ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-[7-(3-aminobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio) butanoate; ethyl 2-({7-[(3,5-dimethylisoxazol-4-yl)methyl]-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-[(7-hexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanamide: 7-(3-bromobenzyl)-8-[(1-ethylpropyl)thio]-3-methyl-3,7-dihydro-1H-purine-2,6-dione; and ethyl 2-{8-[(3-bromobenzyl)thio]-1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl}butanoate.
  • In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; and ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)}thio)butanoate.
  • In some embodiments, the compounds are those having formula II, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts:
  • Figure US20140206667A1-20140724-C00042
  • wherein R.sup.1 is hydrogen or C.sub.1-6 alkyl:
    R.sup.2 is hydrogen or C.sub. 1-4 alkyl;
    R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group;
    R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle;
    R.sup.5 is hydrogen or C.sub. 1-4 alkyl;
    R.sup.6 is C.sub.1-4 alkyl, amido or —COOR.sup.7;
    R.sup.7 is C.sub.1-4 alkyl:
  • In the above embodiment, in some cases, when R.sup.3 is a benzyl group, then R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl.
  • In the above embodiment, in some cases, when R.sup.3 is a group of formula —CHR.sup.5R.sup.6, then R.sup.4 is C.sub. 1-8 alkyl optionally substituted by C.sub.3-6 cycloalkyl, aryl or heterocycle.
  • In some embodiments, the compounds are those compounds of formula II, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts
  • Figure US20140206667A1-20140724-C00043
  • wherein
    R.sup.1 is hydrogen or C.sub. 1-6 alkyl;
    R.sup.2 is hydrogen or C.sub. 1-4 alkyl;
    R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group;
    R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle;
    R.sup.5 is hydrogen or C.sub.1-4 alkyl; R.sup.6 is C.sub.1-4 alkyl, amido or —COOR.sup.7;
    R.sup.7 is C.sub.1-4 alkyl.
  • In some embodiments, the compounds are compounds of formula IT selected from ethyl 2-[(7-heptyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]but-anoate; 7-(3-bromobenzyl)-3-methyl-8-(propylthio)-3,7-dihydro-1H-purine-2,-6-dione; ethyl 2-[(3-methyl-2,6-dioxo-7-pentyl-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]but-anoate; ethyl 2-[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl-]thiol butanoate; ethyl 2-[(3-methyl-2,6-dioxo-7-propyl-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]but-anoate; 7-(3-bromobenzyl)-8-[(3-chloro-2-hydroxypropyl)thio]-3-methyl-3,7-dihydro-1H-purine-2,6-dione; and ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl-]thio}propanoate.
  • In some embodiments, the compounds are compounds of formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts
  • Figure US20140206667A1-20140724-C00044
  • wherein
    R.sup.1 is hydrogen or C.sub. 1-6 alkyl;
    R.sup.2 is hydrogen or C.sub.1-4 alkyl;
    R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group;
    R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle;
    R.sup.5 is C.sub.2-4 alkyl:
    R.sup.6 is C.sub.2-4 alkyl, amido or —COOR.sup.7;
    R.sup.7 is C.sub.1-4 alkyl;
  • In another embodiment, the compounds are compounds having formula II, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00045
  • wherein
    R.sup.1 is hydrogen or C.sub. 1-6 alkyl;
    R.sup.2 is hydrogen or C.sub.1-4 alkyl;
    R.sup.3 is a group of formula —CHR.sup.5R.sup.6 or a benzyl group;
    R.sup.4 is C.sub. 1-8 alkyl optionally substituted by alkoxycarbonyl, C.sub.3-6 cycloalkyl, aryl or heterocycle;
    R.sup.5 is hydrogen or C.sub. 1-4 alkyl;
    R.sup.6 is C.sub.1-4 alkyl, amido or —COOR.sup.7;
    R.sup.7 is C.sub.1-4 alkyl:
  • vi) International Patent Application Publication No. WO2010/144712
  • In one embodiment, a chemical composition that includes a LEV derivative of Formula 1 or Formula 2 is disclosed.
  • Figure US20140206667A1-20140724-C00046
  • n of Formula 2 and L, X, and Y of Formulas 1 and 2 are defined as follows: a) n is an integer with a value of 0 to 8; b) L is one of the group consisting of CH2, CO, NHCO, NHCOO, CONH, NH, O, or S, and combinations thereof; c) X is an end group, an aromatic group, an aryl group, or a saturated, unsaturated, substituted, unsubstituted, straight chain, or branched chain aliphatic group having from 1 to 10 carbon and/or hetero chain atoms, the hetero chain atoms being selected from the group consisting of oxygen, nitrogen, sulfur, or phosphorus, and combinations thereof; and d) Y is optional and if present is one of a functional group selected from group consisting of alcohol amine, amide, carboxylic acid, aldehyde, ester, iminoester, isocyanate, isothiocyanate, anhydride, thiol, thiolacetone, diazonium, NHS, CO—NHS, O—NHS, maleimido; or e) Y is a Yi-Z where Yi is selected from the group consisting of COO, CO, O, CONH, NHCO, or NH and Z is an operative group.
  • In one embodiment of the method, the operative group of Z is selected from the group consisting of detectable labels, antigenic carriers, coupling agents, end groups, proteins, lipoproteins, glycoproteins, polypeptides, polysaccharides, nucleic acids, polynucleotides, teichoic acids, radioactive isotopes, enzymes, enzyme fragments, enzyme donor fragments, enzyme acceptor fragments, enzyme substrates, enzyme inhibitors, coenzymes, fluorescent moieties, phosphorescent moieties, anti-stokes up-regulating moieties, chemiluminescent moieties, luminescent moieties, dyes, sensitizers, particles, microparticles, magnetic particles, solid supports, liposomes, ligands, receptors, hapten radioactive isotopes, and combinations thereof.
  • vii) International Patent Application Publication No. WO2010/002869
  • The present invention provides a compound of Formula I:
  • Figure US20140206667A1-20140724-C00047
  • or a pharmaceutically acceptable salt thereof, wherein: each Z is independently selected from hydrogen and deuterium; R1 is an n-propyl group having zero to seven deuterium atoms; R2 is an ethyl group having zero to five deuterium atoms, and when each R has zero deuterium atoms, at least one Z is deuterium.
  • One embodiment of this invention provides compounds of Formula I wherein R1 is selected from CD3CH2CH2-, CD3CD2CH2-, CD3CH2CD2-, CH3CH2CD2-, CH3CD2CD2-, CD3CD2CD2- or CH3CH2CH2-. In a more specific embodiment, R1 is CD3CD2CD2- or
  • CD3CD2CH2-. In one aspect of these embodiments, Z1 and Z2 are both hydrogen.
  • In another aspect of these embodiments, Z1 and Z2 are both deuterium.
  • In another embodiment, R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In a more specific embodiment, R2 is selected from CH3CH2- or CD3CD2-. In one aspect of these embodiments. Z1 and Z2 are both hydrogen. In another aspect of these embodiments, Z1 I and Z2 are both deuterium.
  • The R and Z variables as described above may be selected and taken together to provide more specific embodiments of this invention. For example, in one embodiment, R1 is CD3CH2CH2-, CD3CD2CH2-, CD3CH2CD2-, CH3CH2CD2-, CH3CD2CD2-, CD3CD2CD2- or CH3CH2CH2-; and R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In one aspect of this embodiment. R2 is CH3CH2- or CD3CD2-. [0039]. In another embodiment, R1 is CD3CD2CD2- or CD3CD2CH2-; and R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In one aspect of this embodiment, R2 is CH3CH2— or CD3CD2-.
  • Examples of specific compounds of this invention include the following:
  • Figure US20140206667A1-20140724-C00048
  • viii) 20090312333
  • The compounds of the present invention are those covered by formula (I), their diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.
  • Figure US20140206667A1-20140724-C00049
  • R1 is hydrogen, substituted or unsubstituted C1-12 alkyl, substituted or unsubstituted aryl or substituted or unsubstituted 3-8 membered heterocycle.
  • R2 is hydrogen. Alternatively, R1 and R2 may be linked together in such a way to form a C3-6 cycloalkyl.
  • R3 is either
  • (a) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms, said heterocycle is selected from the group consisting of:
    • 1H-benzimidazol-6-yl;
    • 1H-benzimidazol-7-yl;
    • imidazo[1,2-a]pyridin-3-yl;
    • imidazo[1,2-a]pyrimidin-3-yl;
    • imidazo[1,2-b][1,2,4]triazin-7-yl;
    • imidazo[1,2-b]pyridazin-3-yl;
    • 5,6,7,8-tetrahydroimidazo[1,2-b]pyridazin-3-yl;
    • imidazo[2,1-b][1,3,4]thiadiazol-5-yl;
    • imidazo[2,1-b][1,3]thiazol-5-yl;
    • 3H-imidazo[4,5-b]pyridin-7-yl;
    • 1H-imidazol-4-yl;
    • 1H-imidazol-5-yl;
    • 1H-indol-2-yl;
    • 1H-indol-3-yl;
    • 1H-indol-4-yl;
    • 1H-indol-7-yl;
    • isoxazol-4-yl;
    • 1H-pyrazol-4-yl;
    • 1H-pyrazol-5-yl;
    • 1H-pyrazolo[1,5-a]pyrimidin-3-yl;
    • 1H-pyrazolo[3,4-b]pyridin-3-yl;
    • pyridazin-4-yl;
    • pyridin-2-yl;
    • pyridin-3-yl;
    • pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-5-yl;
    • 1H-pyrrolo[2,3-c]pyridin-2-yl;
    • 1H-pyrrolo[2,3-c]pyridin-3-yl;
    • 1H-pyrrolo[3,2-b]pyridin-3-yl;
    • 1H-pyrrolo[3,2-c]pyridin-2-yl;
    • 1H-pyrrolo[3,2-c]pyridin-3-yl;
    • 1,3,4-thiadiazol-2-yl;
    • 1,3-thiazol-5-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-7-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-8-yl;
    • indolizin-3-yl:
    • or R3 is
      (b) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its N atoms, said heterocycle is selected from the group consisting of: 1H-1,2,3-benzotriazol-1-yl;
    • 1H-imidazo[4,5-b]pyridin-1-yl;
    • 3H-imidazo[4,5-b]pyridin-3-yl;
    • 7H-imidazo[4,5-c]pyridazin-7-yl;
    • 1H-indol-1-yl;
    • 2,3-dihydro-1H-indol-1-yl;
    • 9H-purin-9-yl:
    • 1H-pyrazolo[3,4-b]pyridin-1-yl;
    • 2H-pyrazolo[3,4-b]pyridin-2-yl;
    • 1H-pyrrolo[2,3-b]pyridin-1-yl;
    • 1H-pyrrolo[3,2-b]pyridin-1-yl;
    • 3,4-dihydroquinolin-1(2H)-yl;
    • 8H-isothiazolo[5,4-b]indol-8-yl;
    • 1H-1,2,4-triazol-1-yl;
    • 1H-pyrrol-1-yl;
    • 2-chloro-1H-benzimidazol-1-yl.
  • R4 in formula (I) is selected from the group comprising or consisting of hydrogen; C1-12 alkyl optionally substituted by halogen, C1-4 alkoxy, C1-4 alkylthio, azido, nitrooxy or an aryl; C2-12 alkenyl optionally substituted by halogen; C2-12 alkynyl optionally substituted by halogen; azido; alkoxycarbonylamino; arylsulfonyloxy; a substituted or unsubstituted aryl; or a 3-8 membered substituted or unsubstituted heterocycle;
  • In a specific embodiment R4 is hydrogen; or R4 is C1-12 alkyl or a C1-6 alkyl, optionally substituted by halogen, C1-4 alkoxy, C1-4 alkylthio, azido or nitrooxy; or R4 is C2-12 alkenyl or a C1-6 alkenyl optionally substituted by halogen; or R4 is C2-12 alkynyl or a C1-6 alkynyl optionally substituted by halogen; or R4 is alkoxycarbonylamino.
  • R5 is hydrogen:
  • Alternatively R4 may form together with R5 and the 2-oxo-1-pyrrolidine ring a 1,3-dihydro-2H-indol-2-one ring of the following structure:
  • Figure US20140206667A1-20140724-C00050
  • The asterisk * indicates the point of attachment of the substituents;
  • R6 is hydrogen or halogen.
    R7 in formula (I) is selected from the group comprising or consisting of hydrogen; nitro; halogen; heterocycle; amino; aryl; C1-12 alkyl optionally substituted by at least one halogen; or C1-12 alkoxy optionally substituted by at least one halogen.
    R8 in formula (I) is selected from the group comprising or consisting of hydrogen. C1-12 alkyl optionally substituted by halogen, or halogen.
    R9 in formula (I) is selected from the group comprising or consisting of hydrogen, C1-12 alkyl optionally substituted by halogen, or halogen.
  • A further aspect of the present invention consists in compounds of formula (I) wherein
  • R1 and R2 are both hydrogen.
  • R3 is:
  • (a) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms selected from the group consisting of:
    • 1H-benzimidazol-6-yl;
    • 1H-benzimidazol-7-yl;
    • imidazo[1,2-a]pyridin-3-yl;
    • imidazo[1,2-a]pyrimidin-3-yl;
    • imidazo[1,2-b][1,2,4]triazin-7-yl;
    • imidazo[1,2-b]pyridazin-3-yl;
    • 5,6,7,8-tetrahydroimidazo[1,2-b]pyridazin-3-yl;
    • imidazo[2,1-b][1,3,4]thiadiazol-5-yl;
    • imidazo[2,1-b][1,3]thiazol-5-yl;
    • 3H-imidazo[4,5-b]pyridin-7-yl;
    • 1H-imidazol-4-yl;
    • 1H-imidazol-5-yl;
    • 1H-indol-2-yl;
    • 1H-indol-3-yl;
    • 1H-indol-4-yl;
    • 1H-indol-7-yl;
    • isoxazol-4-yl;
    • 1H-pyrazol-4-yl;
    • 1H-pyrazol-5-yl;
    • 1H-pyrazolo[1,5-a]pyrimidin-3-yl;
    • 1H-pyrazolo[3,4-b]pyridin-3-yl;
    • pyridazin-4-yl;
    • pyridin-2-yl;
    • pyridin-3-yl;
    • pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-5-yl;
    • 1H-pyrrolo[2,3-c]pyridin-2-yl;
    • 1H-pyrrolo[2,3-c]pyridin-3-yl;
    • 1H-pyrrolo[3,2-b]pyridin-3-yl;
    • 1H-pyrrolo[3,2-c]pyridin-2-yl;
    • 1H-pyrrolo[3,2-c]pyridin-3-yl;
    • 1,3,4-thiadiazol-2-yl;
    • 1,3-thiazol-5-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-7-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-8-yl;
    • indolizin-3-yl.
  • Alternatively R3 is:
  • (b) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its N atoms selected from the group consisting of:
    • 1H-1,2,3-benzotriazol-1-yl;
    • 1H-imidazo[4,5-b]pyridin-1-yl;
    • 3H-imidazo[4,5-b]pyridin-3-yl;
    • 7H-imidazo[4,5-c]pyridazin-7-yl;
    • 1H-indol-1-yl;
    • 2,3-dihydro-1H-indol-1-yl;
    • 9H-purin-9-yl;
    • 1H-pyrazolo[3,4-b]pyridin-1-yl;
    • 2H-pyrazolo[3,4-b]pyridin-2-yl;
    • 1H-pyrrolo[2,3-b]pyridin-1-yl;
    • 1H-pyrrolo[3,2-b]pyridin-1-yl;
    • 3,4-dihydroquinolin-1(2H)-yl;
    • 8H-isothiazolo[5,4-b]indol-8-yl;
    • 1H-1,2,4-triazol-1-yl;
    • 1H-pyrrol-1-yl;
    • 2-chloro-1H-benzimidazol-1-yl.
      R4 in formula (I) is selected from the group comprising or consisting of hydrogen; C1-12 alkyl optionally substituted by halogen or C1-4 alkoxy; C2-12 alkenyl optionally substituted by halogen; C2-12 alkynyl optionally substituted by halogen.
  • In a further specific embodiment R4 is n-propyl, 2,2,2-trifluoroethyl, 2-chloro-2,2-difluoroethyl, 2 bromo-2,2-difluoroethyl, 2,2-difluorovinyl.
  • In another specific embodiment R4 is phenyl, 2,3,5-trifluorophenyl or 3-chloro-4-fluorophenyl.
  • R5 is hydrogen;
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R4 forms together with R5a 1,3-dihydro-2H-indol-2-one ring
  • Figure US20140206667A1-20140724-C00051
  • The asterisk * indicates the point of attachment of the heteroaryl alkylene substituent, and wherein
  • R6 is hydrogen;
    R7 is chlorine;
    R8 is hydrogen;
    R9 is hydrogen.
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of:
    • imidazo[1,2-a]pyrimidin-3-yl:
    • imidazo[1,2-b][1,2,4]triazin-7-yl;
    • imidazo[1,2-b]pyridazin-3-yl;
    • 5,6,7,8-tetrahydroimidazo[1,2-b]pyridazin-3-yl
    • imidazo[2,1-b][1,3,4]thiadiazol-5-yl;
    • imidazo[2,1-b][1,3]thiazol-5-yl;
    • 3H-imidazo[4,5-b]pyridin-7-yl;
    • 1H-imidazol-4-yl;
    • 1H-imidazol-5-yl;
    • isoxazol-4-yl;
    • 1H-pyrazol-4-yl;
    • 1H-pyrazol-5-yl;
    • 1H-pyrazolo[1,5-a]pyrimidin-3-yl;
    • 1H-pyrazolo[3,4-b]pyridin-3-yl;
    • pyridin-3-yl:
    • 1H-pyrrolo[2,3-b]pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-5-yl
    • 1H-pyrrolo[2,3-c]pyridin-2-yl;
    • 1H-pyrrolo[2,3-c]pyridin-3-yl;
    • 1,3-thiazol-5-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-8-yl;
    • indolizin-3-yl.
  • In a further specific embodiment R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of:
    • imidazo[1,2-b]pyridazin-3-yl;
    • imidazo[2,1-b][1,3,4]thiadiazol-5-yl;
    • imidazo[2,1-b][1,3]thiazol-5-yl;
    • 3H-imidazo[4,5-b]pyridin-7-yl;
    • 1H-imidazol-4-yl;
    • 1H-imidazol-5-yl;
    • 1H-pyrazol-4-yl;
    • 1H-pyrazolo[1,5-a]pyrimidin-3-yl;
    • pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-4-yl:
    • 1,3-thiazol-5-yl;
  • Said heterocycles are optionally substituted by e.g. a methyl, n-propyl, trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, iodine, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclopropylmethoxy, cyclobutylmethoxy, amino, methylamino, cyclopropylamino, cyclobutylamino, 1-pyrrolidinyl, cyano, phenyl, benzyl or 3-thienyl.
  • In a further specific embodiment R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: 6-chloro-2-cyclopropylimidazo[1,2-b]pyridazin-3-yl, 6-(cyclopropyloxy)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl. 6-propoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl. 6-chloroimidazo[2,1-b][1,3]thiazol-5-yl, 2,6-dichloroimidazo[2,1-b][1,3]thiazol-5-yl, 5-chloro-1H-imidazol-4-yl, 5-bromo-1H-imidazol-4-yl, 4-bromo-1H-imidazol-5-yl, 4-chloro-1H-imidazol-5-yl, 1H-imidazol-5-yl. 1-methyl-1H-imidazol-5-yl, 4-chloro-1-methyl-1H-imidazol-5-yl, 1H-pyrazol-4-yl, 1H-pyrrolo[2,3-b]pyridin-3-yl.
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is a substituted or unsubstituted imidazo[1,2-a]pyridin-3-yl.
  • Said imidazo[1,2-a]pyridin-3-yl is optionally substituted by e.g. a methyl, cyclopropyl, bromine, chlorine, fluorine, iodine.
  • In a further specific embodiment R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: imidazo[1,2-a]pyridin-3-yl, 6-methylimidazo[1,2-a]pyridin-3-yl, 2-chloroimidazo[1,2-a]pyridin-3-yl.
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its N atoms and is selected from the group consisting of:
    • 3H-imidazo[4,5-b]pyridin-3-yl;
    • 1H-indol-1-yl;
    • 1H-pyrrolo[2,3-b]pyridin-1-yl;
    • 1H-pyrrolo[3,2-b]pyridin-1-yl:
    • 1H-pyrrol-1-yl:
    • 2-chloro-1H-benzimidazol-1-yl.
  • A specific further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a heterocycle linked to the rest of the molecule via one of its N atoms and is selected from the group consisting of:
    • 3H-imidazo[4,5-b]pyridin-3-yl;
    • 1H-pyrrolo[3,2-b]pyridin-1-yl:
    • 1H-pyrrol-1-yl:
    • 2-chloro-1H-benzimidazol-1-yl;
  • Said heterocycles may optionally be substituted by trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, methoxy or cyano.
  • In a further specific embodiment R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of 6-bromo-2-chloro-3H-imidazo[4,5-b]pyridin-3-yl, 6-bromo-2-cyclopropyl-3H-imidazo[4,5-b]pyridin-3-yl, 1H-pyrrolo[3,2-b]pyridin-1-yl, 2,5-dichloro-1H-pyrrol-1-yl, 2-chloro-5-methoxy-1H-benzimidazol-1-yl, 5-bromo-2-chloro-1H-benzimidazol-1-yl or 2,5-dichloro-1H-benzimidazol-1-yl.
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R1, R2 and R5 are hydrogen.
  • R4 is a C1-6 alkyl optionally substituted by halogen, a C2-6 alkenyl optionally substituted by halogen or C2-12 alkynyl optionally substituted by halogen.
    R3 is selected from the group consisting of;
    • imidazo[1,2-b]pyridazin-3-yl;
    • imidazo[2,1-b][1,3,4]thiadiazol-5-yl;
    • imidazo[2,1-b][1,3]thiazol-5-yl;
    • 3H-imidazo[4,5-b]pyridin-7-yl;
    • 1H-imidazol-4-yl;
    • 1H-imidazol-5-yl;
    • 1H-pyrazol-4-yl;
    • 1H-pyrazolo[1,5-a]pyrimidin-3-yl;
    • pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-3-yl:
    • 1H-pyrrolo[2,3-b]pyridin-4-yl;
    • 1,3-thiazol-5-yl;
      and optionally substituted by methyl, n-propyl, trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, iodine, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclopropylmethoxy, cyclobutylmethoxy, amino, methylamino, cyclopropylamino, cyclobutylamino, 1-pyrrolidinyl, cyano, phenyl, benzyl or 3-thienyl.
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R1, R2 and R5 are hydrogen.
  • R4 is a C1-6 alkyl optionally substituted by halogen, a C2-6 alkenyl optionally substituted by halogen or C2-12 alkynyl optionally substituted by halogen.
    R3 is selected from the group consisting of
    • 3H-imidazo[4,5-b]pyridin-3-yl;
    • 1H pyrrolo[3,2-b]pyridin-1-yl;
    • 1H-pyrrol-1-yl:
    • 2-chloro-1H-benzimidazol-1-yl;
      optionally substituted by trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, methoxy or cyano.
  • A further embodiment of the invention consists in compounds of formula (I), their diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.
  • Figure US20140206667A1-20140724-C00052
  • R1, R2 and R5 are hydrogen.
    R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms, said heterocycle is selected from the group consisting of:
    • 1H-benzimidazol-6-yl;
    • 1H-benzimidazol-7-yl;
    • imidazo[1,2-a]pyridin-3-yl;
    • imidazo[1,2-a]pyrimidin-3-yl;
    • imidazo[1,2-b][1,2,4]triazin-7-yl;
    • imidazo[1,2-b]pyridazin-3-yl;
    • 5,6,7,8-tetrahydroimidazo[1,2-b]pyridazin-3-yl;
    • imidazo[2,1-b][1,3,4]thiadiazol-5-yl;
    • imidazo[2,1-b][1,3]thiazol-5-yl;
    • 3H-imidazo[4,5-b]pyridin-7-yl;
    • 1H-imidazol-4-yl;
    • 1H-imidazol-5-yl;
    • 1H-indol-2-yl;
    • 1H-indol-3-yl;
    • 1H-indol-4-yl;
    • H-indol-7-yl;
    • isoxazol-4-yl;
    • 1H-pyrazol-4-yl;
    • 1H-pyrazol-5-yl;
    • 1H-pyrazolo[1,5-a]pyrimidin-3-yl;
    • 1H-pyrazolo[3,4-b]pyridin-3-yl;
    • pyridazin-4-yl;
    • pyridin-2-yl;
    • pyridin-3-yl:
    • pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-3-yl;
    • 1H-pyrrolo[2,3-b]pyridin-4-yl;
    • 1H-pyrrolo[2,3-b]pyridin-5-yl;
    • 1H-pyrrolo[2,3-c]pyridin-2-yl;
    • 1H-pyrrolo[2,3-c]pyridin-3-yl;
    • 1H-pyrrolo[3,2-b]pyridin-3-yl;
    • 1H-pyrrolo[3,2-c]pyridin-2-yl;
    • 1H-pyrrolo[3,2-c]pyridin-3-yl;
    • 1,3,4-thiadiazol-2-yl;
    • 1,3-thiazol-5-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-7-yl;
    • [1,2,4]triazolo[4,3-b]pyridazin-8-yl:
    • indolizin-3-yl:
  • Particularly preferred are imidazo[1,2-a]pyridin-3-yl; imidazo[1,2-a]pyrimidin-3-yl; imidazo[1,2-b]pyridazin-3-yl; 1H-imidazol-4-yl; 1H-imidazol-5-yl;
  • R4 is a substituted or unsubstituted phenyl moiety:
  • A further embodiment of the present invention consists in compounds of formula (I) wherein R1 is hydrogen or C1-12 alkyl;
  • R2 is hydrogen;
    R3 is an aromatic 5-membered heterocycle linked to the rest of the molecule via one of its C atoms;
    R4 is hydrogen, C1-12 alkyl or aryl;
    R5 is hydrogen:
  • Alternatively, R4 can form together with R5 and the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00053
  • wherein the asterisk * indicates the point of attachment of the substituents;
    R6 is hydrogen or halogen:
  • In this embodiment R4 may not be hydrogen when R3 is substituted 1H-pyrazol-5-yl. Also this embodiment does not comprise 5-(2′-oxo-1′-pyrrolidinyl)methyl-1,3,4-tricarbomethoxy-pyrazole which is disclosed in A. Padwa et al J. Org. Chem. 2000, 65, 5223-5232 without any biological activity though.
  • In this embodiment wherein R3 is an aromatic 5-membered heterocycle linked to the rest of the molecule via one of its C atoms, specific moieties R3 may be selected from 1,3-thiazol-5-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl, 2-oxo-2,3-dihydro-1,3-thiazol-5-yl, each of them being optionally substituted by 1 to 3 substituents independently selected from methyl, chlorine, bromine, amino, methylamino, dimethylamino, (2-oxo-4-propyl-pyrrolidin-1-yl)methyl, 1-pyrrolidinyl, amido, cyano, methoxy, phenyl, 4-methylphenyl-sulfonyl, benzyl or 2-(benzylamino)-2-oxoethyl.
  • In this embodiment, more specific moieties R3 are selected from 2-(methylamino)-1,3-thiazol-5-yl; 2-pyrrolidin-1-yl-1,3-thiazol-5-yl: 5-bromo-1H-imidazol-4-yl; 5-chloro-1H-imidazol-4-yl; 1H-imidazol-5-yl; 1-methyl-1H-imidazol-5-yl; 4-bromo-1-methyl-1H-imidazol-5-yl; 4-chloro-1H-imidazol-5-yl; 4-chloro-1-methyl-1H-imidazol-5-yl: 4-cyano-1-methyl-1H-imidazol-5-yl; 1H-pyrazol-4-yl; 3,5-dimethyl-1H-pyrazol-4-yl; 3-methyl-1H-pyrazol-4-yl.
  • In this embodiment, most specific moieties R3 are selected from 5-bromo-1H-imidazol-4-yl; 5-chloro-1H-imidazol-4-yl; 1H-imidazol-5-yl; 4-bromo-1-methyl-1H-imidazol-5-yl; 4-chloro-1-methyl-1H-imidazol-5-yl: 1H-pyrazol-4-yl.
  • Still in this embodiment, a specific moiety R1 is selected from hydrogen or ethyl.
  • Still in this embodiment, a specific moiety R4 is selected from hydrogen, n-propyl, 2,3,5-trifluorophenyl or phenyl.
  • A further embodiment of the present invention consists in compounds having the specific formula (Ia).
  • Figure US20140206667A1-20140724-C00054
  • In formula (Ia) the substituent R10 is hydrogen; halogen; C1-4 alkyl optionally substituted by at least one halogen; C1-4 alkoxy; methoxycarbonyl; nitro; amino; alkylamino; amido; or alkanoyl-amino. Preferably R10 is hydrogen.
  • R11 is hydrogen; halogen: C1-4 alkyl optionally substituted by at least one halogen; C1-4 alkoxy; methoxycarbonyl; nitro: amino; alkylamino; amido; or alkanoylamino. Preferably R11 is hydrogen.
    R4 is C1-4 alkyl optionally substituted by at least one halogen; or C2-4 alkenyl optionally substituted by at least one halogen. Preferably R4 is n-propyl.
  • Still in this aspect of the invention a specific embodiment relates to an embodiment wherein R10 is selected from hydrogen; methyl; fluorine; chlorine; bromine; methoxy; methoxycarbonyl; nitro; or trifluoromethyl, while R11 is selected from hydrogen; methyl; fluorine; chlorine; bromine; methoxy; methoxycarbonyl; nitro; or trifluoromethyl; and R3 is n-propyl.
  • Specific compounds of the present invention are those selected from the group consisting of:
    • 1-[(1-methyl-1H-benzimidazol-6-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-(1H-benzimidazol-7-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-propylpyrrolidin-2-one:
    • 1-{[6-chloro-2-(4-methylphenyl)imidazo[1,2-a]pyridin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-([2-(4-chlorophenyl)-6-methylimidazo[1,2-a]pyridin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(5-methylimidazo[1,2-a]pyridin-3-yl)methyl]-4-phenylpyrrolidin-2-one;
    • 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-phenylpyrrolidin-2-one;
    • 1-[(6-methylimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromoimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(8-methylimiidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-iodoimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[8-chloro-6-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl)}-4-propylpyrrolidin-2-one;
    • 1-[(7-methylimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6,8-dibromoimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6,8-dichloroimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-chloroimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloroimidazo[1,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-cyclopropyl-6-fluoroimidazo[1,2-a]pyridin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-chloro-2-cyclopropylimidazo[1,2-a]pyridin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-(imidazo[1,2-a]pyrimidin-3-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-{[2-(4-chlorophenyl)imidazo[1,2-a]pyrimidin-3-yl]methyl}-4-propyl pyrrolidin-2-one;
    • 1-(imidazo[1,2-a]pyrimidin-3-ylmethyl)-4-phenylpyrrolidin-2-one;
    • 1-[(6-chloroimidazo[1,2-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyrimidin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(6-phenylimidazo[1,2-b][1,2,4]triain-7-yl)methyl]-4-propylpyolidin-2-one;
    • 1-{[6-chloro-2-(4-methylphenyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-propylpyrrolidin-2-one;
    • 1-{[6-chloro-2-(4-chlorophenyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(6-chloroimiidazo[1,2-b]pyridazin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-chloroimidazo[1,2-b]pyridazin-3-yl)methyl]-4-phenylpyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-phenylpyrrolidin-2-one;
    • 5-chloro-1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-1,3-dihydro-2H-indol-2-one;
    • 1-{[6-methoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(6-chloro-2-cyclopropylimidazo[1,2-b]pyridazin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[6-isopropoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-(benzyloxy)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-cyclopropyl-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-(dimethylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-methoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2-chloro-2,2-difluoroethyl)-1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl)}pyrrolidin-2-one;
    • 1-{[6-(methylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-hydroxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-(methylthio)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 4-(2-bromo-2,2-difluoroethyl)-1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 1-{[6-(methylsulfonyl)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-(methylsulfinyl)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4(2,2,2-trifluoroethyl)pyrrolidin-2-one;
    • 1-[(6-chloro-2-cyclobutylimidazo[1,2-b]pyridazin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[6-chloro-2-(4-methylphenyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-22 difluorovinyl)pyrrolidin-2-one;
    • 1-{[6-amino-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-(ethylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 4-propyl-1-{[6-(propylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2-bromo-2,2-difluoroethyl)-1-{[6-(propylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-(propylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-methoxy-2-(4-methylphenyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-propyl-1-{[6-pyrrolidin-1-yl-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2-bromo-2,2-difluoroethyl)-1-{[6-methoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 1-{[6-(cyclopropylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-chloro-2-cyclopropylimidazo[1,2-b]pyridazin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-(isopropylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 1-{[2-cyclopropyl-6-(propylamino)imidazo[1,2-b]pyridazin-3-yl]methyl}-22 difluorovinyl)pyrrolidin-2-one;
    • 1-({2-cyclopropyl-6-[(2-fluoroethyl)amino]imidazo[1,2-b]pyridazin-3-yl}methyl)-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-({2-cyclopropyl-6-[(2,2-difluoroethyl)amino]imidazo[1,2-b]pyridazin-3-yl}methyl)-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-({2-cyclopropyl-6-[(2,2,2-trifluoroethyl)amino]imidazo[1,2-b]pyridazin-3-yl}methyl)-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 4-(2,2-difluoroethyl)-1-([2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl)pyrrolidin-2-one;
    • 1-{[2-cyclopropyl-6-(cyclopropylamino)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-chloro-2-cyclobutylimidazo[1,2-b]pyridazin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-chloro-2-cyclopropylimidazo[1,2-b]pyridazin-3-yl)methyl]-4-(3-chloro-4-fluorophenyl)pyrrolidin-2-one;
    • 1-{[6-(butylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-{[6-(cyclobutylamino)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(2-cyclopropyl-6-methoxyimidazo[1,2-b]pyridazin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-[6-ethoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl)pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-isopropoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one,
    • 1-{[6-(cyclopropylmethoxy)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-{[6-(cyclobutylmethoxy)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-{[6-(cyclopropyloxy)-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-propoxy-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 3-{[4-(2,2-difluorovinyl)-2-oxopyrrolidin-1-yl]methyl}-2-(trifluoromethyl)imidazo[1,2-b]pyridazine-6-carbonitrile;
    • 4-(2,2-difluorovinyl)-1-{[6-thien-3-yl-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-phenyl-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-methyl-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-{[6-pyridin-3-yl-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 4-propyl-1-{[2-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[1,2-b]pyridazin-3-yl]methyl}pyrrolidin-2-one;
    • 1-[(6-methylimidazo[2,1-b][1,3,4]thiadiazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[6-(4-methylphenyl)imidazo[2,1-b][1,3,4]thiadiazol-5-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(2-cyclopropyl-6-phenylimidazo[2,1-b][1,3,4]thiadiazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-methylimidazo[2,1-b][1,3]thiazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-chloroimidazo[2,1-b][1,3]thiazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2,6-dichloroimidazo[2,1-b][1,3]thiazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-(3H-imidazo[4,5-b]pyridin-7-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(3H-imidazo[4,5-b]pyridine-7-ylmethyl)-4-phenylpyrrolidin-2-on;
    • 4-phenyl-1-[(5-phenyl-3H-imidazo[4,5-b]pyridin-7-yl)methyl]pyrrolidin-2-one;
    • 4-phenyl-1-{[5-(trifluoromethyl)-3H-imidazo[4,5-b]pyridin-7-yl]methyl}pyrrolidin-2-one;
    • 1-[(6-bromo-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-phenyl-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-methyl-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-methyl-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-{[5-(trifluoromethyl)-3H-imidazo[4,5-b]pyridin-7-yl]methyl}pyrrolidin-2-one;
    • 1-[(6-methyl-3H-imidazo[4,5-b]pyrindin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-phenyl-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[1-(1H-imidazol-4-yl)propyl]pyrrolidin-2-one;
    • 1-[(5-methyl-1H-imidazol-4-yl)methyl]pyrrolidin-2-one;
    • 1-[(2-methyl-1H-imidazol-4-yl)methyl]pyrrolidin-2-one;
    • 1-(1H-imidazol-4-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-({1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazol-4-yl}methyl)-4-propylpyrrolidin-2-one;
    • 1-[(5-chloro-1H-imidazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(5-bromo-1H-imidazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(5-bromo-1H-imidazol-4-yl)methyl]-5-chloro-1,3-dihydro-2H-indol-2-one;
    • 1-(1H-imidazol-5-ylmethyl)pyrrolidin-2-one;
    • 1-[(1-methyl-1H-imidazol-5-yl)methyl]pyrrolidin-2-one;
    • 1-methyl-5-[(2-oxopyrrolidin-1-yl)methyl]-1H-imidazole-4-carbonitrile;
    • 1-(1H-imidazol-5-ylmethyl)-4-phenylpyrrolidin-2-one;
    • 1-[(1-methyl-1H-imidazol-5-yl)methyl]-4-phenylpyrrolidin-2-one;
    • 1-[(4-methoxy-1-methyl-1H-imidazol-5-yl)methyl]pyrrolidin-2-one;
    • 1-[(1-methyl-1H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-methyl-5-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-4-carbonitrile;
    • 1-methyl-5-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-4-carboxamide;
    • N-benzyl-2-{5-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazol-1-yl}acetamide;
    • 1-methyl-5-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-2-carbonitrile;
    • 1-[(4-chloro-1H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-methyl-5-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile;
    • 1-[(4-bromo-1-methyl-1H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2,4-dichloro-1-methyl-1H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • benzyl 1-methyl-5-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazol-2-ylcarbamate;
    • 1-[(4-chloro-1-methyl-1H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1-methyl-1H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;
    • 5-chloro-1-(1H-imidazol-5-ylmethyl)-1,3-dihydro-2H-indol-2-one;
    • 1-[(2,4-dichloro-1H-imidazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(2,4-dichloro-1-methyl-1H-imidazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(2-chloro-1-methyl-1H-imidazol-5-ylmethyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(4-bromo-1-methyl-1H-imidazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 5-chloro-1-[(1-methyl-1H-imidazol-5-yl)methyl]-1,3-dihydro-2H-indol-2-one;
    • 1-[(4-chloro-1-methyl-1H-imidazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-(1H-indol-2-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(1H-indol-3-ylmethyl)-4-propylpyrrolidin-2-one;
    • 3-[(2-oxo-propylpyrrolidin-1-yl)methyl]-1H-indole-5-carbonitrile;
    • 1-[(2-methyl-1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(7-methoxy-1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-nitro-1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-{[6-(trifluoromethyl)-1H-indol-3-yl]methyl}pyrrolidin-2-one;
    • 1-[(5-nitro-1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(7-fluoro-1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-chloro-2-methyl-1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[1H-indol-3-yl(phenyl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[1-(1H-indol-3-yl)propyl]-4-propylpyrrolidin-2-one;
    • 1-[2-furyl(1H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 3-[(2-oxo-4-propylpyrrolidin-1-yl)(phenyl)methyl]-1H-indole-5-carbonitrile;
    • 1-(1H-indol-4-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(1H-indol-7-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(isoxazol-4-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-[(1-phenyl-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(1-methyl-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(1-benzyl-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 4-(2,3,5-trifluorophenyl)-1-[(1,3,5-trimethyl-1H-pyrazol-4-yl)methyl]pyrrolidin-2-one;
    • 4-phenyl-1-(1H-pyrazol-4-ylmethyl)pyrrolidin-2-one;
    • 1-({1-[(4-methylphenyl)sulfonyl]-1H-pyrazol-4-yl}methyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-(1H-pyrazol-4-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(5-chloro-1,3-dimethyl-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl pyrrolidin-2-one;
    • 1-[(1-chloro-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(3,5-dimethyl-1H-pyrazol-4-yl)methyl]-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(3-methyl-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(5-amino-1,3-dimethyl-1H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(5-amino-1-methyl-1H-pyrazol-4-yl)methyl]-4-propylpyrrolidin-2-one;
    • (−)-1-(1H-pyrazol-4-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • (+)-1-(1H-pyrazol-4-ylmethyl)—(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-(1H-pyrazol-4-ylmethyl)-1,3-dihydro-2H-indol-2-one;
    • 5-chloro-1-(1H-pyrazol-4-ylmethyl)-1,3-dihydro-2H-indol-2-one;
    • 5-chloro-1-((1-[(4-methylphenyl)sulfonyl]-1H-pyrazol-4-yl)methyl)-1,3-dihydro-2H-indol-2-one;
    • 1-{[5-chloro-1-methyl-3-(trifluoromethyl)-11H-pyrazol-4-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(5-amino-11H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(1-benzyl-5-chloro-1H-pyrazol-4-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(1,3-dimethyl-1H-pyrazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-(1H-pyrazol-5-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(4-bromo-1-methyl-1H-pyrazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(1-methyl-1H-pyrazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(6-bromo-2-methylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-methylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-thien-2-ylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-[(2-thien-2-ylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 1-[(6-bromo-2-cyclopropylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-tert-butylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-tert-butyl-6-cyclopropylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[2-(2-furyl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(2-methyl-6-thien-2-ylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-methyl-6-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[2-methyl-6-(1H-pyrrol-2-yl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-({6-[(1E)-hex-1-enyl]-2-methylpyrazolo[1,5-a]pyrimidin-3-yl}methyl)-4-propylpyrrolidin-2-one;
    • 1-[(6-chloro-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[2-methyl-6-(phenylethynyl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-hydroxy-2-methylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-methyl-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-[(2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 1-[(6-methoxy-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-chloropyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one:
    • 4-(2,2-difluorovinyl)-1-[(5,6-dimethyl-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-[(6-fluoro-5-methyl-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 1-[(5-methoxypyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[2-(4-bromophenyl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-{[2-(4-fluorophenyl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-[(6-methyl-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-[(5-methyl-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 4-(2,2-difluorovinyl)-1-[(2-thien-2-ylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]pyrrolidin-2-one;
    • 1-{[2-(4-chlorophenyl)-6-methylpyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[2-(4-chlorophenyl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-chloro-2-phenylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-{[6-chloro-2-(4-chlorophenyl)pyrazolo[1,5-a]pyrimidin-3-yl]methyl}-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(2-cyclopropyl-5-methylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-(2.2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(5-chloro-2-cyclopropylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(5-chloro-2,6-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(5-bromo-1H-pyrazolo[3,4-b]pyridin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one,
    • 4-propyl-1-(pyridin-3-ylmethyl)pyrrolidin-2-one;
    • (−)-1-(1-pyridin-3-ylpropyl)pyrrolidin-2-one;
    • 5-chloro-1-[(2-fluoropyridin-3-yl)methyl]-1,3-dihydro-2H-indol-2-one;
    • 1-[(6-chloropyridin-3-yl)methyl]-4-propylpyrrolidin-2-one,
    • 1-{[6-(benzylamino)pyridin-3-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(2-aminopyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[2,3-b]pyridin-3-ylmethyl)pyrrolidin-2-one;
    • 1-[(2-isopropyl-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-phenyl-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-[(2-propyl-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]pyrrolidin-2-one;
    • 1-[(6-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one,
    • 1-[(1-benzoyl-6-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-phenyl-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(7-oxido-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[2,3-b]pyridin-4-ylmethyl)pyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[2,3-b]pyridin-5-ylmethyl)pyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[2,3-c]pyridin-2-ylmethyl)pyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[2,3-c]pyridin-3-ylmethyl)pyrrolidin-2-one:
    • 4-propyl-1-(1H-pyrrolo[3,2-b]pyridin-3-ylmethyl)pyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[3,2-c]pyridin-2-ylmethyl)pyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[3,2-c]pyridin-3-ylmethyl)pyrrolidin-2-one;
    • 4-propyl-1-(1,3,4-thiadiazol-2-ylmethyl)pyrrolidin-2-one;
    • 1-[(2-amino-1,3-thiazol-5-yl)methyl]pyrrolidin-2-one;
    • 1-(1,3-thiazol-5-ylmethyl)pyrrolidin-2-one;
    • 1-[(2-chloro-1,3-thiazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-{[2-(dimethylamino)-1,3-thiazol-5-yl]methyl}-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-{[2-(methylamino)-1,3-thiazol-5-yl]methyl}-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 1-[(2-pyrrolidin-1-yl-1,3-thiazol-5-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one;
    • 5-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1,3-thiazol-2(3H)-one;
    • 4-phenyl-1-{[3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazin-7-yl]methyl}pyrrolidin-2-one;
    • 4-phenyl-1-[(3-phenyl[1,2,4]triazolo[4,3-b]pyridazin-7-yl)methyl]pyrrolidin-2-one;
    • 4-phenyl-1-{[3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazin-8-yl]methyl}pyrrolidin-2-one;
    • 4-propyl-1-{[3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazin-8-yl]methyl}pyrrolidin-2-one;
    • 4-phenyl-1-[(3-phenyl[1,2,4]triazolo[4,3-b]pyridazin-8-yl)methyl]pyrrolidin-2-one;
    • 1-[(6-chloro-3-phenyl[1,2,4]triazolo[4,3-b]pyridazin-8-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-chloro[1,2,4]triazolo[4,3-b]pyridazin-8-yl)methyl]-4-phenylpyrrolidin-2-one;
    • 1-{[6-chloro-3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazin-8-yl]methyl}-4-phenylpyrrolidin-2-one;
    • 1-[(6-chloro-3-phenyl[1,2,4]triazolo[4,3-b]pyridazin-8-yl)methyl]-4-phenylpyrrolidin-2-one;
    • 1-[(2-fluoroindolizin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-(1H-1,2,3-benzotriazol-1-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-chloro-1H-imidazo[4,5-b]pyridin-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-phenyl-1H-imidazo[4,5-b]pyridin-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-(3H-imidazo[4,5-b]pyridin-3-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-3H-imidazo[4,5-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-chloro-3H-imidazo[4,5-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-bromo-2-cyclopropyl-3H-imidazo[4,5-b]pyridin-3-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(3-chloro-7H-imidazo[4,5-c]pyridazin-7-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-methyl-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-methyl-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-phenyl-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-fluoro-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-bromo-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-chloro-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-(2,3-dihydro-1H-indol-1-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-[(5-fluoro-2-phenyl-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-indole-2-carbonitrile;
    • 1-[(2-bromo-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2,5-dichloro-1H-indol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(6-amino-9H-purin-9-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-(9H-purin-9-ylmethyl)pyrrolidin-2-one;
    • 1-{[6-(cyclopropylamino)-9H-purin-9-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[6-(benzylamino)-9H-purin-9-yl]methyl}-4-propylpyrrolidin-2-one;
    • 4-propyl-1-{[6-(propylamino)-9H-purin-9-yl]methyl}pyrrolidin-2-one;
    • 1-({6-[(cyclopropylmethyl)amino]-9H-purin-9-yl}methyl)-4-propylpyrrolidin-2-one;
    • 4-propyl-1-[(6-pyrrolidin-1-yl-9H-purin-9-yl)methyl]pyrrolidin-2-one;
    • 1-[(5-bromo-3-phenyl-1H-pyrazolo[3,4-b]pyridin-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-bromo-2H-pyrazolo[3,4-b]pyridin-2-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-bromo-3-phenyl-2H-pyrazolo[3,4-b]pyridin-2-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 4-propyl-1-(1H-pyrrolo[3,2-b]pyridin-1-ylmethyl)pyrrolidin-2-one;
    • 1-(3,4-dihydroquinolin-1(2H)-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(8H-isothiazolo[5,4-b]indol-8-ylmethyl)-4-propylpyrrolidin-2-one;
    • 1-(1H-1,2,4-triazol-1-ylmethyl)pyrrolidin-2-one;
    • 1-[(2,5-dichloro-1H-pyrrol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-pyrrol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-benzimidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one;
    • 2-chloro-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-5-carbonitrile;
    • 2-chloro-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-6-carbonitrile;
    • 4-propyl-1-[(2,5,6-trichloro-1H-benzimidazol-1-yl)methyl]pyrrolidin-2-one;
    • 1-[(2-chloro-6-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-5-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-6-nitro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-5-nitro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-6-methyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-benzimidazol-1-yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one;
    • 1-[(6-bromo-2-chloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(5-bromo-2-chloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-6-fluoro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-5-fluoro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2,6-dichloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-[(2,5-dichloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-{[2-chloro-6-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-{[2-chloro-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one;
    • 1-[(2-chloro-1H-benzimidazol-1-yl)methyl]pyrrolidin-2-one;
    • 1-[(2-chloro-6-hydroxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one;
    • 1-(pyridin-4-ylmethyl)pyrrolidin-2-one, and
    • 1-[(2-chloro-5-hydroxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one.
  • viii) U.S. Pat. No. 4,696,943
  • The present invention relates to the novel compound (S)-alpha-ethyl-2-oxo-1-pyrrolidineacetamide.
  • ix) U.S. Pat. No. 4,696,942
  • The present invention relates to the novel compound, (R)-alpha-ethyl-2-oxo-1-pyrrolidineacetamide
  • x) U.S. Pat. No. 5,334,720
  • According to this invention we provide novel compounds of the formula I,
  • Figure US20140206667A1-20140724-C00055
  • wherein, R1, R2, R3 and R4, which may be the same or different independently represent hydrogen, C1-6 alkyl, phenyl or phenyl substituted by one or more halogen, hydroxyl, nitro, amino, C1-6 alkyl or C1-C6 alkoxy groups;
    R5 and R6 independently represent hydrogen, C1-C6 alkyl or C3-C6 cycloalkyl, or R5 and R6 together with the nitrogen form a C4-6 N heterocycle:
    m represents an integer from 1-2; and
    n represents an integer from 1-3;
    provided that,
    two of the substituents R1, R2, R3 and R4 independently represent phenyl or substituted phenyl and the other two independently represent hydrogen or C1-6 alkyl;
    or a pharmaceutically acceptable acid addition salt thereof.
  • Pharmaceutically acceptable acid addition salts of the compounds of formula I include salts of mineral acids, for example, hydrohalic acids, e.g. hydrochloric or hydrobromic; or organic acids, e.g. formic, acetic or lactic acids. The acid may be polybasic, for example sulphuric, fumaric, maleic or citric acid.
  • This invention also relates to all stereoisomeric forms and optical enantiomeric forms of the compounds of formula I.
  • In the compounds of formula I: alkyl groups which R1, R2, R3, R4, R5 and R6 may represent include methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl and s-butyl;
  • cycloalkyl groups which R5 and R6 may represent include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl;
    C1-6 alkoxy groups include methoxy, ethoxy and propoxy; halogen groups include fluorine, chlorine, bromine or iodine;
  • We prefer compounds of formula I or a pharmaceutically acceptable acid addition salt thereof, in which;
  • R1 is hydrogen, phenyl or substituted phenyl, preferably phenyl;
    R2 is hydrogen, phenyl or substituted phenyl, preferably phenyl;
    R3 is hydrogen, phenyl or substituted phenyl, preferably hydrogen:
    R4 is hydrogen, phenyl or substituted phenyl, preferably hydrogen;
    R5 is hydrogen, C1-3 alkyl or cyclopropyl, preferably hydrogen or methyl;
    R6 is hydrogen, C1-3 alkyl or cyclopropyl, preferably hydrogen or methyl;
    m represents an integer from 1-2 preferably 2;
    n represents an integer from 1-2, preferably 1.
  • We especially prefer compounds of formula I in which R1 and R2 are both phenyl.
  • We especially prefer compounds of formula I in which one of R5 and R6 is hydrogen and the other is hydrogen or methyl.
  • xi) International Patent Application Publication No. WO2005/054188
  • In one aspect the invention therefore provides a compound having the formula I or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00056
  • wherein
    RI is hydrogen, CI-20 alkyl, C3 23 cycloalkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, guanidine, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl, arylsulfinyl, aryl or heterocycle; R2 is hydrogen, C1 20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;
    R3 is hydrogen, C1 20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;
    or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00057
  • R4 is hydrogen, C1-20 alkyl, C2-12 alkenyl, C2-12 alkynyl, aryl, azido, alkoxycarbonylamino, arylsulfonyloxy or heterocycle; R4a is hydrogen or C1-20 alkyl; or R4 and R4a can form together a C3-8 cycloalkyl; R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00058
  • R6 is hydrogen or C1 20 alkyl; R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl; R8 is hydrogen, halogen, nitro, cyano, C1 20 alkyl or alkoxy; R9 is hydrogen, C1-20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
    RIO is hydrogen, C1 20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonyl, arylsulfonyl, alkylsulfinyl or arylsulfinyl;
    RI1 is hydrogen, halogen, nitro, cyano, C1 20 alkyl or alkoxy; R12 is hydrogen or halogen;
    R13 is hydrogen, nitro, halogen, heterocycle, amino, aryl, C1-20 alkyl unsubstituted or substituted by halogen, or alkoxy unsubstituted or substituted by halogen; R14 is hydrogen, C1-20 alkyl or halogen:
    R15 is hydrogen, C1 20 alkyl or halogen; with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00059
  • N represents a group of formula
  • Figure US20140206667A1-20140724-C00060
  • The asterisk * indicates the point of attachment of the substituents.
  • In a preferred embodiment, the invention concerns a compound having the formula I, their tautomers, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00061
  • wherein
    RI is hydrogen, C1-20 alkyl, C3-8 cycloalkyl, halogen, hydroxy, ester, amido, cyano, nitro, amino, guanidine, alkylthio, alkylsulfonyl, alkylsulfinyl, aryl or heterocycle: R2 is hydrogen, C1 20 alkyl, halogen, cyano, ester, carbamate or amido; R3 is hydrogen, cyano, C1 20 alkyl, halogen or ester, or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00062
  • hydrogen, C1 20 alkyl, C2 12 alkenyl or aryl; R4a is hydrogen;
    R5 is hydrogen: or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00063
  • R6 is hydrogen or C1 20 alkyl; R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl; R8 is hydrogen; R9 is hydrogen, C 1-20 alkyl, halogen or alkoxy; RIO is hydrogen, C1 20 alkyl, halogen or cyano; R11 is hydrogen; R12 is hydrogen or halogen; R13 is hydrogen, halogen, heterocycle or C1 20 alkyl; R14 is hydrogen; R15 is hydrogen; with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00064
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00065
  • The term “alkyl”, as used herein, represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched or cyclic or combinations thereof and containing 1-20 carbon atoms, preferably 1-10 carbon atoms, more preferably 1-4 carbon atoms; most preferred alkyl groups have 1-3 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, cyano, azido, aryloxy, alkoxy, alkylthio, alkanoylamino, arylcarbonylamino, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl or aryl. Usually alkyl groups, in the present case, are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, n-heptyl, 2,4,4-trimethylpentyl, n-decyl, chloromethyl, trifluoromethyl, 2-bromo-2,2-difluoroethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino) methyl, (propionylamino) methyl, (benzoylamino) methyl, (4-chlorophenoxy)methyl, benzyl, 2-phenylethyl or 2-(methylthio) ethyl. Preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, 2,4,4-trimethylpentyl, chloromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino) methyl, (propionylamino) methyl, (benzoylarino) methyl or 2-(methylthio) ethyl. More preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, azidomethyl or trifluoromethyl. Most preferred alkyl groups are methyl or n-propyl.
  • The term “cycloalkyl”, as used herein, represents a monovalent group of 3 to 8 carbon atoms, usually 3-6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl groups are cyclopropyl and cyclohexyl.
  • The term “alkenyl” as used herein, represents straight, branched or cyclic unsaturated hydrocarbon radicals or combinations thereof having at least one carbon-carbon double bond, containing 2-12 carbon atoms, preferably usually 2-4 carbon atoms. Alkenyl groups are being optionally substituted with any suitable group, including but not limited to one or more moities selected from groups as described above for the alkyl groups. Usually an alkenyl group is ethenyl (vinyl) optionally substituted by 1 to 3 halogens. Preferred alkenyl group, in the present case, is 2,2-difluorovinyl.
  • The term “alkynyl” as used herein, represents straight, branched or cyclic hydrocarbon radicals or combinations thereof containing at least one carbon-carbon triple bond, containing 2-12 carbon atoms, preferably 2-6 carbon atoms, and being optionally substituted by any suitable group, including but not limited to one or more moities selected from groups as described above for the alkyl groups. Preferably an alkynyl group is a halogenoalkynyl group (haloalkynyl group). Groups qualified by prefixes such as “s”, “i”, “t” and the like (e.g. “i-propyl”, “s-butyl”) are branched derivatives.
  • The term “aryl” as used herein, is defined as phenyl optionally substituted by 1 to 4 substituents independently selected from halogen, cyano, alkoxy, alkylthio, C1-3 alkyl or azido, preferably halogen or azido. Usually aryl groups, in the present case are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl, 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl. Preferably, aryl groups are phenyl, 3-chlorophenyl. 3-fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3-chloro-4-fluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl. Most preferred aryl groups are phenyl, 3-chlorophenyl. 3-fluorophenyl, 3,5-difluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • The term “heterocycle”, as used herein, is defined as including an aromatic or non aromatic cycloalkyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure. Heterocyclic ring moities can be optionally substituted by alkyl groups or halogens and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl. Usually heterocycles are 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-tetrahydrofuranyl, 1H-pyrrol-2-yl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrazol-2-yl, 1H-pyrazol-3-yl. 4-chloro-1-methyl-1H-pyrazol-3-yl, 5-chloro-1,3-dimethyl-1H-pyrazol-4-yl, 1,2,3-thiadiazol-4-yl, 3,5-dimethyl-4-isothiazyl, 1H-imidazol-2-yl, 1-methyl-1H-imidazol-2-yl, 4-methyl-1H-imidazol-5-yl, or 2-methyl-1,3-thiazol-4-yl. Preferred heterocycles are 1H-imidazol-2-yl, 1,2,3-thiadiazol-4-yl, 1H-pyrazol-3-yl, 2-furyl, 3-furyl, 2-thienyl, 1-methyl-1H-pyrrol-2-yl, 1H-pyrrol-2-yl.
  • The term “halogen”, as used herein, includes an atom of chlorine, bromine, fluorine, iodine. Usually halogens are chlorine, bromine and fluorine. Preferred halogens are fluorine, bromine and chlorine.
  • The term “hydroxy”, as used herein, represents a group of formula —OH.
  • The term “alkoxy”, as used herein, represents a group of formula —ORa wherein Ra is an alkyl group, as defined above. Preferred alkoxy group is methoxy.
  • The term “aryloxy”, as used herein, represents a group of formula —ORb wherein Rb is an aryl group, as defined above. Preferred aryloxy group is phenoxy.
  • The term “ester”, as used herein, represents a group of formula —COORC wherein Rc is an alkyl group or aryl group, as defined above. Preferred ester group is methoxycarbonyl.
  • The term “amido”, as used herein, represents a group of formula —CONH2.
  • The term “amino”, as used herein, represents a group of formula —NH2.
  • The term “aminoderivative”, as used herein, represents an alkylamino or an arylamino group, wherein the terms “alkyl” and “aryl” are defined as above.
  • The term “cyano”, as used herein, represents a group of formula —CN.
  • The term “nitro”, as used herein, represents a group of formula —NO2.
  • The term “azido”, as used herein, represents a group of formula —N3.
  • The term “guanidine”, as used herein, represents a group of formula —NHC(═NH) NH2.
  • The term “alkylthio”, as used herein, represents a group of formula —SRd wherein Rd is an alkyl group, as defined above. One alkylthio group is methylthio.
  • The term “alkylsulfonyl”, as used herein, represents a group of formula —S(═O) 2Re wherein Re is an alkyl group, as defined above. One alkylsulfonyl group is methylsulfonyl.
  • The term “alkylsulfinyl”, as used herein, represents a group of formula —S(═O)Rf wherein Rf is an alkyl group, as defined above. One alkylsulfinyl group is methylsulfinyl.
  • The term “arylthio”, as used herein, represents a group of formula —SRg wherein Rg is an aryl group, as defined above.
  • The term “arylsulfonyl”, as used herein, represents a group of the formula —S(═O) 2Rh wherein Rh is an aryl group, as defined above.
  • The term “arylsulfinyl”, as used herein, represents a group of the formula —S(═O) Ri wherein Ri is an aryl group, as defined above.
  • The term “carbamate”, as used herein, represents a group of formula —N(H)C(O) OR1, wherein Ri is an alkyl or an aryl, as defined above. Usually carbamate groups are (propoxycarbonyl)amino or (benzyloaxycarbonyl)amino. One carbamate group is (benzyloaxycarbonyl)amino.
  • The term “alkanoylamino”, as used herein, represents a group of the formula —NHC (═O) Rk wherein Rk is an alkyl group, as defined above.
  • The term “(arylcarbonyl)amino”, as used herein, represents a group of the formula —NHC(═O)Rm wherein Rm is an aryl group, as defined above. One (arylcarbonyl)amino is benzoylamino.
  • Usually, RI is hydrogen; C1 lo alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; hydroxy; C3-6 cycloalkyl; halogen; ester, amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl groups; or guanidine.
  • In some embodiments, RI is hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl; n-butyl; i-butyl; t-butyl; -ethylpropyl; 2,4,4-trimethylpentyl; hydroxymethyl; chloromethyl; trifluoromethyl; 2,2,2-trifluoroethyl; cyanomethyl; 2-(methylthio) ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl or 1H-imidazol-2-yl. More preferably, RI is hydrogen; methyl; ethyl; i-propyl; n-propyl; n-butyl; methylthio: nitro; cyano; amino; chloro or 1H-pyrrol-2-yl. Most preferably, RI is hydrogen; methyl; methylthio; nitro; cyano; amino or chloro.
  • Usually, R2 is hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate; [(N-methoxy-N-methyl)amino]carbonyl. Preferably, R2 is hydrogen; methyl; hydroxymethyl; (acetylamino) methyl; (propionylamino) methyl; (benzoylamino) methyl; [(benzyloxy) carbonyl]amino; chloro or cyano. In some embodiments, R2 is hydrogen; chloro or cyano.
  • Usually, R3 is hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano. In some embodiments, R3 is hydrogen; hydroxymethyl; chloro; cyano.
  • In some embodiments, R3 is hydrogen or cyano. In some embodiments R3 is hydrogen.
  • Usually, R4 is hydrogen; C1-4 alkyl unsubstituted or substituted by halogens; C2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens. Preferably. R4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl or 3-azido-2,4,6-trifluorophenyl. More preferably, R4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl. Most preferably, R4 is n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl or 3-azido-2,4-difluorophenyl.
  • Usually, R4a is hydrogen.
  • Usually, R5 is hydrogen.
  • Usually, R6 is hydrogen or Cl-1˜0 alkyl unsubstituted or substituted by hydroxy or azido. Preferably, R6 is hydrogen or azidomethyl. More preferably R6 is hydrogen.
  • Usually R7 is hydrogen.
  • In other embodiments, R6 and R7 are linked to form a cyclopropyl.
  • In other embodiments, R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00066
  • Usually, R8 is hydrogen.
  • Usually, R9 is hydrogen; halogen; 1-3 alkyl or alkoxy. In some embodiments, R9 is hydrogen; methyl; chloro or methoxy. In some embodiments R9 is hydrogen.
  • Usually, RIO is hydrogen; halogen; cyano; CI 3 alkyl unsubstituted or substituted by halogens; or alkoxy. In some embodiments. RIO is methyl; hydrogen; trifluoromethyl; fluoro; cyano or methoxy. In some embodiments R10 is hydrogen; trifluoromethyl; fluoro or cyano.
  • Usually, RI 1 is hydrogen.
  • In other embodiments, R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00067
  • Usually, R12 is hydrogen or halogen. In some embodiments R12 is hydrogen; chloro or fluoro. In some embodiments R12 is hydrogen.
  • Usually, R13 is hydrogen; C1 3 alkyl; halogen or thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl. In some embodiments R13 is hydrogen; chloro; bromo or methyl. In some embodiments R13 is chloro; bromo or methyl.
  • Usually R14 is hydrogen.
  • Usually, R15 is hydrogen.
  • In a general embodiment of the invention, the compounds of formula I, or pharmaceutically acceptable salts thereof, are those wherein
  • RI is selected from hydrogen; C1 lo alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; C3 6 cycloalkyl; halogen; ester; amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl group; or guanidine; R2 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate or [(N-methoxy-N-methyl)amino]carbonyl.
  • R3 is selected from hydrogen; C1-4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano; R4 is selected from hydrogen; C1 4 alkyl unsubstituted or substituted by halogens; C2 4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens;
    R4a is hydrogen; R5 is hydrogen; R6 is selected from hydrogen or C 1-10 alkyl unsubstituted or substituted by hydroxy or azido;
    R7 is hydrogen; or R6 and R7 can be linked to form a cyclopropyl; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00068
  • R8 is hydrogen: R9 is selected from hydrogen; halogen: C1-3 alkyl; alkoxy;
    R10 is selected from hydrogen; halogen; cyano or Cil alkyl unsubstituted or substituted by halogens; or alkoxy; R 1 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00069
  • R12 is selected from hydrogen or halogen; R13 is selected from hydrogen; CI-3 alkyl; halogen; thiazolyl unsubstituted or substituted by alkyl groups, such as methylthiazolyl; R14 is hydrogen; R15 is hydrogen; with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00070
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00071
  • In an embodiment of the invention, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein
  • RI is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; cyclopropyl; n-butyl; i-butyl; t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; trifluoromethyl; 2,2,2-trifluoroethyl; hydroxymethyl; chloromethyl; cyanomethyl; 2-(methylthio) ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl: methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1H-pyrrol-2-yl; 1-methyl-1H-pyrrol-2-yl; 2-thienyl; 1H-pyrazol-3-yl; 1,2,3-thiadiazol-4-yl; or 1H-imidazol-2-yl; R2 is selected from hydrogen; methyl; hydroxymethyl; (acetylamino) methyl; (propionylamino) methyl; (benzoylamino) methyl; (benzyloxycarbonyl)amino; chloro; or cyano; R3 is selected from hydrogen; hydroxymethyl: chloro; cyano; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00072
  • R8 is hydrogen; R9 is selected from hydrogen; methyl; choro; methoxy; R10 is selected from methyl; hydrogen; trifluoromethyl; fluoro; cyano; or methoxy; R is hydrogen; R4 is selected from hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl: 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl; or 3-azido-2,4,6-trifluorophenyl. R4a is hydrogen; R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00073
  • R12 is selected from hydrogen; chloro; fluoro; R13 is selected from hydrogen; chloro: bromo; methyl: R14 is hydrogen: R15 hydrogen; R6 is selected from hydrogen; azidomethyl; R7 is hydrogen; or R6 and R7 are linked to form a cyclopropyl; with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00074
  • represents a group of formula
  • Figure US20140206667A1-20140724-C00075
  • In one embodiment of the invention, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein
  • RI is selected from hydrogen; methyl; ethyl; i-propyl; n-propyl; n-butyl; methylthio; nitro; cyano; amino; chloro; or 1H-pyrrol-2-yl; R2 is selected from hydrogen; chloro; cyano; R3 is selected from hydrogen; cyano; or R2 and R3 can form together with the imidazole ring the following 1H-benzimidazole cycle
  • Figure US20140206667A1-20140724-C00076
  • R8 is hydrogen; R9 is hydrogen;
    R10 is selected from hydrogen; trifluoromethyl; fluoro; cyano;
    R11 is hydrogen; R4 is selected from hydrogen: n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fluorophenyl; 3,5-difluorophenyl; 3,4-difluorophenyl; 3-chloro-4-fluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; or 3-azido-2,4-difluorophenyl; R4a is hydrogen; R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00077
  • wherein R12 is hydrogen; R13 is selected from methyl; chloro; bromo; R14 is hydrogen; R15 hydrogen; R6 is hydrogen; R7 is hydrogen; with the proviso that R4 is different from hydrogen when
  • Figure US20140206667A1-20140724-C00078
  • R11 represents a group of formula
  • Figure US20140206667A1-20140724-C00079
  • In one embodiment of the invention, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein
  • RI is selected from hydrogen; methyl; methylthio; nitro; cyano; amino; chloro; R2 is selected from hydrogen; chloro; cyano; R3 is hydrogen; R4 is selected from n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5-trifluorophenyl; 3-azido-2,4-difluorophenyl; R4a is hydrogen;
    R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-1-pyrrolidine ring the following 1,3-dihydro-2H-indol-2-one cycle
  • Figure US20140206667A1-20140724-C00080
  • R12 is hydrogen; R13 is selected from chloro; bromo; methyl; R14 is hydrogen; R15 hydrogen; R6 is hydrogen; R7 is hydrogen.
  • In some embodiments, compounds are: 1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 4-(3-azido-2,4,6-trifluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-[(2-ethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-isopropyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-phenyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-[(2-propyl-1H-imidazol-1-yl)methyl]pyrrolidin-2-one; (+)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; 4-(2,2-difluorovinyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-([2-(methylthio)-1H-imidazol-1-yl]methyl-4-propylpyrrolidin-2-one; 1-([2-(methylsulfinyl)-1H-imidazol-1-yl]methyl)-4-propylpyrrolidin-2-one; 1-[(2-tert-butyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[1-(1H-imidazol-1-yl)cyclopropyl]pyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one; 1-{[2-(methylsulfonyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-2-carboxamide; 4-(4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,5-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chloro-4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl) pyrrolidin-2-one; 4-(4-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,4-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-(hydroxymethyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; methyl 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazole-2-carboxylate; 1-[(2-nitro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one: 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-2-carbonitrile; 1-[(2-amino-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2,4-dichloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(5-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (+)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (−)-1-{[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (+)-1-[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-1-yl]methyl)-1H-imidazole-5-carbonitrile; (−)-1-{[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (+)-{[2-oxo-4-(2,3,4-trifluorophenyl)-1-pyrrolidinyl]methyl}-1H-imidazole-4-carbonitrile; (−)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl)}-H-imidazole-4-carbonitrile; (+)-1-{[2-oxo-4-(2,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile: (−)-1-[2-oxo-4-(2,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl)-1H-imidazole-4-carbonitrile; (−)-1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-ylmethyl}-1H-imidazole-4-carbonitrile; (−)-1-[2-oxo-4-(3, 4, 5=trifluorophenyl)pyrrolidin-1-yl]methyl)-1H-imidazole-5-carbonitrile; 1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 1-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-methyl}-1H-imidazole-5-carbonitrile; 1-[(5-methyl-2-phenyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-phenyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-ethyl-5-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2,5-dimethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one: 1-[2-azido-1-(1H-imidazol-1-yl)ethyl]-4-propylpyrrolidin-2-one; 1-[(4-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(2-bromo-4,5-dichloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one: 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl})-1H-imidazole-5-carbonitrile; 1-{[5-(hydroxymethyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[4-(hydroxymethyl)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; benzyl 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-imidazol-5-ylcarbamate; N-[(1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazol-5-yl)methyl]acetamide; N-[(1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl)-1H-imidazol-5-yl)methyl]benzamide; N-1 (1-1 [2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazol-5-yl)methyl]propanamide; 1-(1H-benzimidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-[(2-propyl-1H-benzimidazol-1-yl)methyl]pyrrolidin-2-one; 1-[(2-isopropyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}pyrrolidin-2-one; 1-[2-(methylthio)-1H-benzimidazol-1-yl]methyl)-4-propylpyrrolidin-2-one; 1-[(2-amino-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[2-(chloromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; (1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazol-2-yl)acetonitrile; 1-[(5-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-methyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5,6-dimethyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[2-isopropyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(6-chloro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-propyl-1H-benzimidazole-5-carbonitrile; 1-[2-ethyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl)-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl}pyrrolidin-2-one; 1-[(5-fluoro-2-propyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[6-methyl-2-(1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(6-methoxy-2-propyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 2-butyl-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-5-carbonitrile; 1-{[2-[2-(methylthio)ethyl]-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(5-fluoro-2-isobutyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[5-fluoro-2-(2,4,4-trimethylpentyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 2-cyclopropyl-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-5-carbonitrile; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-(1H-pyrazol-3-yl)-1H-benzimidazole-5-carbonitrile; 1-[(2-cyclopropyl-5-fluoro-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-fluoro-2-isopropyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-{[2-(3-furyl)-6-methoxy-1H-benzimidazol-1-ylmethyl}-4-propylpyrrolidin-2-one; 1-[(2-cyclopropyl-6-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-isopropyl-6-methoxy-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-(1,2,3-thiadiazol-4-yl)-1H-benzimidazole-5-carbonitrile; 1-{[2-(1H-imidazol-2-yl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[5-fluoro-2-(2,2,2-trifluoroethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[2-(1-ethylpropyl)-6-methoxy-IH-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[6-methoxy-2-(1-methyl-1H-pyrrol-2-yl)-IH-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-([2-(2-furyl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl)-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-thien-2-yl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]melthyl}pyrrolidin-2-one; 1-{[2-(3-furyl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-{[2-cyclopropyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(1H-pyrrol-2-yl)-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-bromo-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one: 5-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 4-fluoro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 4-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 1-(1H-imidazol-1-ylmethyl)-5-methyl-1,3-dihydro-2H-indol-2-one; 1-[(2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbonitrile; and 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbonitrile.
  • In some embodiments, compounds are: 1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one, 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-[(2-ethyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-isopropyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl-1-[(2-propyl-1H-imidazol-1-yl)methyl]pyrrolidin-2-one; (+)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; 4-(2,2-difluorovinyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-{[2-(methylthio)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one; 4-(4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,5-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-chloro-4-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 4-(4-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,4-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(2-nitro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-2-carbonitrile; 1-[(2-amino-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-1 (5-chloro-IH-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one: 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-H-imidazole-4-carbonitrile; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; (+)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (−)-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (+); 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-4-carbonitrile; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[2-azido-1-(1H-imidazol-1-yl)ethyl]-4-propylpyrrolidin-2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-2-propyl-1H-benzimidazole-5-carbonitrile; 1-{[2-ethyl-5-(trifluoromethyl)-1H-benzimidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 4-propyl-1-{[2-(1H-pyrrol-2-yl)-1H-benzimidazol-1-yl]methyl}pyrrolidin-2-one; 1-[(5-fluoro-2-propyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 2-butyl-1-[(2-oxo-4-propylpyrrolidin-1-yl)methyl]-1H-benzimidazole-5-carbonitrile; 1-[(5-fluoro-2-isopropyl-1H-benzimidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-bromo-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 5-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 1-(1H-imidazol-1-ylmethyl)-5-methyl-1,3-dihydro-2H-indol-2-one; 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbonitrile.
  • In some embodiments, compounds are: 1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpyrrolidin-2-one; (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 4-(2,2-difluorovinyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one: 4-(3-chlorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-{[2-(methylthio)-1H-imidazol-1-yl]methyl}-4-propylpyrrolidin-2-one; 1-[(2-methyl-1H-imidazol-1-yl)methyl]-4-phenylpyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3,5-difluoromethyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,4-trifluorophenyl)pyrrolidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrrolidin-2-one; 1-H-imidazol-1-ylmethyl)-4-(2,4,5-trifluorophenyl)pyrrolidin-2-one; 1-[(2-nitro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; 1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-H-imidazole-2-carbonitrile; 1-[(2-amino-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; 1-[(5-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one; (+)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; (−)-1-(1H-imidazol-1-ylmethyl)-4-phenylpyrrolidin-2-one; 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 1-[(2-chloro-1H-imidazol-1-yl)methyl]-4-propylpyrrolidin-2-one; (+)-1-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-1-yl]methyl}-1H-imidazole-5-carbonitrile; 5-bromo-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one: 5-chloro-1-(1H-imidazol-1-ylmethyl)-1,3-dihydro-2H-indol-2-one; 1-(1H-imidazol-1-ylmethyl)-5-methyl-1,3-dihydro-2H-indol-2-one; 1-[(5-chloro-2-oxo-2,3-dihydro-1H-indol-1-yl)methyl]-1H-imidazole-5-carbonitrile.
  • Some compounds are: (−)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; (+)-4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one; 4-(3-azido-2,4-difluorophenyl)-1-(1H-imidazol-1-ylmethyl)pyrrolidin-2-one.
  • The acid addition salt form of a compound of formula I that occurs in its free form as a base can be obtained by treating the free base with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, trifluoroacetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclic, salicylic, p-aminosalicylic, pamoic and the like.
  • The compounds of formula I containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt forms, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts. e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.
  • Compounds of the formula I and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like.
  • Many of the compounds of formula I and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem., 45 (1976) 11-30.
  • The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula I or mixtures thereof (including all possible mixtures of stereoisomers).
  • Some of the compounds of formula I may also exist in tautomeric forms. Such forms although not explicitly indicated in the above formula are intended to be included within the scope of the present invention.
  • In another preferred embodiment, the present invention concerns also compounds of formula IA and their tautomeric form IB
  • Figure US20140206667A1-20140724-C00081
  • With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof, unless the particular isomeric form is referred to specifically. Compounds according to the present invention may exist in different polymorphic forms. Although not explicitly indicated in the above formula, such forms are intended to be included within the scope of the present invention.
  • The invention also includes within its scope pro-drug forms of the compounds of formula I and its various sub-scopes and sub-groups.
  • xii) U.S. Patent Application Publication No. 20090018148
  • In one aspect the invention provides compounds having formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00082
  • wherein
    R1 is hydrogen or C1-6 alkyl;
    R2 is hydrogen or C1-4 alkyl;
    R3 is a group of formula —CHR5R6 or a benzyl group;
    R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle;
    R5 is C2-4 alkyl;
    R6 is C2-4 alkyl, amido or —COOR7;
    R7 is C1-4 alkyl;
  • In one aspect, the invention provides compounds:
  • When R1 is hydrogen, R2 is methyl, R3 is —CHR5R6, R6 is ethoxycarbonyl and R5 is ethyl, then R4 is different from methyl, n-propyl, i-propyl, n-pentyl, n-heptyl, 3-bromobenzyl, 4-chlorobenzyl, 4-methylbenzyl or 2-phenylethyl;
  • When R1 is hydrogen, R2 is methyl, R3 is benzyl, then R4 is different from i-propyl, n-butyl, 3-methylbutyl, benzyl, phenylethyl-, or 3-phenylpropyl;
  • When R1 and R2 are methyl, R3 is benzyl, R4 is different from methyl, 3-methylbutyl, benzyl, 3-phenylpropyl or 4-chlorophenylmethyl; Finally 8-(2-chloro-benzylsulfanyl)-3-methyl-7-octyl-3,7-dihydro-purine-2,6-dione is considered.
  • Usually when R3 is a benzyl group, then R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl.
  • Usually when R3 is a group of formula —CHR5R6, then R4 is C1-8 alkyl optionally substituted by C3-6 cycloalkyl, aryl or heterocycle.
  • The term “alkyl”, as used herein, is a group which represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched moieties, or combinations thereof, and containing 1-8 carbon atoms, preferably 1-6 carbon atoms; more preferably alkyl groups have 1-4 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl, acyl, aryl or heterocycle. Alkyl moieties may be optionally substituted by a cycloalkyl as defined hereafter. Preferred alkyl groups according to the present invention are methyl, cyanomethyl, ethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl. 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-butyl, i-butyl, n-pentyl, 3-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro-2-furyl)methyl. More preferred alkyl groups are methyl, ethyl, cyanomethyl, 2-methoxyethyl, n-propyl, 3-hydroxypropyl, 2-propynyl, n-butyl, 3-pentyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro-2-furyl)methyl. Most preferred alkyl groups are methyl, ethyl, 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • The term “cycloalkyl”, as used herein, represents a monovalent group of 3 to 8, preferably 3 to 6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl group according to the present invention is cyclohexyl.
  • The term “aryl” as used herein, is defined as a phenyl group optionally substituted by 1 to 4 substituents independently selected from halogen, amino, nitro, alkoxy or aminosulfonyl. Preferred aryl groups are phenyl, 2-bromophenyl, 3-bromophenyl, 4-bromophenyl, 3-methoxyphenyl, 3-nitrophenyl, 3-aminophenyl or 4-(aminosulfonyl)phenyl.
  • The term “phenyl”, as used herein, represents an aromatic hydrocarbon group of formula —C6H5.
  • The term “benzyl group”, as used herein, represents a group of formula —CH2-aryl. Preferred benzyl groups are benzyl, 2-bromobenzyl, 3-bromobenzyl. 4-bromobenzyl. 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl or 4-(aminosulfonyl)benzyl. More preferred benzyl groups are benzyl, 3-bromobenzyl. 3-methoxybenzyl, 3-nitrobenzyl or 3-aminobenzyl. In some embodiments alkyl groups are 3-methoxybenzyl or 3-nitrobenzyl.
  • The term “halogen”, as used herein, represents an atom of fluorine, chlorine, bromine, or iodine. In some embodiments the halogen is bromine.
  • The term “hydroxy”, as used herein, represents a group of formula —OH.
  • The term “cyano”, as used herein, represents a group of formula —CN.
  • The term “amino”, as used herein, represents a group of formula —NH2.
  • The term “ethynyl”, as used herein, represents a group of formula —C≡CH.
  • The term “alkoxy”, as used herein, represents a group of formula —ORa wherein
  • Ra is an alkyl group, as defined above. In some embodiments the alkoxy group is methoxy.
  • The term “nitro”, as used herein, represents a group of formula —NO2.
  • The term “amido”, as used herein, represents a group of formula —C(═O)NH2.
  • The term “acyl”, as used herein, represents a group of formula —C(═O)Rb wherein Rb is an alkyl group, as defined here above. In some embodiments the acyl group is acetyl (—C(═O)Me).
  • The term “alkoxycarbonyl (or ester)”, as used herein, represents a group of formula —COORc wherein Rc is an alkyl group; with the proviso that Rc does not represent an alkyl alpha-substituted by hydroxy. In some embodiments the alkoxycarbonyl group is ethoxycarbonyl.
  • The term “heterocycle”, as used herein, represents a 5-membered ring containing one or two heteroatoms selected from O or N. The heterocycle may be substituted by one or two C1-4 alkyl or nitro. In some embodiments the heterocycles are (3,5-dimethylisoxazol-4-yl) or (5-nitro-2-furyl). Most preferred heterocycle is (5-nitro-2-furyl).
  • Generally R1 is hydrogen or C1-6 alkyl. Usually R1 is hydrogen or C1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl. In some embodiments R1 is hydrogen, methyl, cyanomethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl. 2-propynyl, n-pentyl or n-hexyl. In some embodiments R1 is hydrogen, methyl, cyanomethyl, 2-methoxyethyl, n-propyl. 3-hydroxypropyl or 2-propynyl. In some embodiments R1 is hydrogen.
  • Generally R2 is hydrogen or C1-4 alkyl. Usually R2 is hydrogen or unsubstituted C1-4 alkyl. In some embodiments R2 is hydrogen, methyl or n-butyl. In some embodiments, R2 is methyl.
  • Generally R3 is a group of formula —CHR5R6 or a benzyl group. In some embodiments R3 is 3-pentyl, 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl. In some embodiments R3 is 1-(ethoxycarbonyl)propyl.
  • Generally R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle. Usually R4 is C1-8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl. 3,5-dimethylisoxazol-4-yl, 5-nitro-2-furyl or ethoxycarbonyl. In some embodiments R4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl. In some embodiments R4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl. In some embodiments R4 is 3-methoxybenzyl. 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • Generally R5 is C2-4 alkyl. Usually R5 is unsubstituted C2-4 alkyl. In some embodiments R5 is ethyl.
  • Generally R6 is C2-4 alkyl, amido or —COOR7. Usually R6 is unsubstituted C2-4 alkyl, amido or —COOR7. In some embodiments R6 is ethyl, amido or ethoxycarbonyl. In some embodiments R6 is ethoxycarbonyl.
  • Generally R7 is C1-4 alkyl. Usually R7 is unsubstituted C1-4 alkyl. In some embodiments, R7 is ethyl.
  • Usually the invention provides compounds having formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,
  • Figure US20140206667A1-20140724-C00083
  • wherein
    R1 is hydrogen, C1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl;
    R2 is hydrogen or unsubstituted C1-4 alkyl;
    R3 is a group of formula —CHR5R6 or a benzyl group;
    R4 is C1-8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl, 5-nitro-2-furyl or ethoxycarbonyl;
    R5 is unsubstituted C2-4 alkyl;
    R6 is unsubstituted C2-4 alkyl, amido or —COOR7;
    R7 is unsubstituted C1-4 alkyl;
    with the proviso that when R1 is hydrogen, R2 is methyl, R3 is —CHR5R6, R6 is ethoxycarbonyl and R5 is ethyl, then R4 is different from n-propyl, i-propyl, n-pentyl, n-heptyl, 3-bromobenzyl, 4-chlorobenzyl, 4-methylbenzyl or 2-phenylethyl. In the above embodiment, sometimes, when R3 is a benzyl group, then R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl.
  • In the above embodiment, sometimes, when R3 is a group of formula —CHR5R6, then R4 is C1-8 alkyl optionally substituted by C3-6 cycloalkyl, aryl or heterocycle.
  • In one embodiment,
  • R1 is hydrogen, methyl, cyanomethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-pentyl or n-hexyl;
    R2 is hydrogen, methyl or n-butyl;
    R3 is 3-pentyl, 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl;
    R4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl. 3-bromobenzyl, 4-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1-phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl;
    with the proviso that when R1 is hydrogen, R2 is methyl and R3 is 1-(ethoxycarbonyl)propyl, then R4 is different from n-pentyl, 3-bromobenzyl or 2-phenylethyl.
  • In the above embodiment, sometimes, when R3 is 3-bromobenzyl, then R4 is C1-8 alkyl optionally substituted by alkoxycarbonyl.
  • In the above embodiment, sometimes, when R3 is 3-pentyl, 1-(aminocarbonyl)propyl or 1-(ethoxycarbonyl)propyl, then R4 is different from 1-(ethoxycarbonyl)propyl.
  • In a more preferred embodiment, R1 is hydrogen, methyl, cyanomethyl, 2-methoxyethyl, n-propyl, 3-hydroxypropyl or 2-propynyl;
  • R2 is methyl;
    R3 is 3-pentyl, 1-(aminocarbonyl)propyl, 1-(ethoxycarbonyl)propyl or 3-bromobenzyl;
    R4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2-furyl)methyl or 1-(ethoxycarbonyl)propyl;
    with the proviso that when R1 is hydrogen. R2 is methyl and R3 is 1-(ethoxycarbonyl)propyl, then R4 is different from 3-bromobenzyl.
  • In the above embodiment, sometimes, when R3 is 3-bromobenzyl, then R4 is 1-(ethoxycarbonyl)propyl;
  • In the above embodiment, sometimes, when R3 is 3-pentyl, 1-(aminocarbonyl)propyl or 1-(ethoxycarbonyl)propyl, then R4 is different from 1-(ethoxycarbonyl)propyl;
  • In one embodiment, R1 is hydrogen; R2 is methyl; R3 is 1-(ethoxycarbonyl)propyl; and R4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.
  • A further embodiment consists in compounds wherein R2 is methyl, R3 is a group of formula —CHR5R6 with R5 being C2-4 alkyl, R6 being amido or —COOR7 and R7 being methyl or ethyl.
  • In some embodiments, compounds are ethyl 2-[(7-benzyl-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(2-ethoxy-2-oxoethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(2-methoxyethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(2-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(cyanomethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-propyl-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-(2-oxopropyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(3-hydroxypropyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-(2-propynyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-aminobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-({7-[4-(aminosulfonyl)benzyl]-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-{[7-(4-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(cyclohexylmethyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[1,3-dimethyl-2,6-dioxo-7-(1-phenylethyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[1,3-dimethyl-2,6-dioxo-7-(2-phenylethyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-({7-[(3,5-dimethylisoxazol-4-yl)methyl]-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-[(1,7-dihexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-[(7-hexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-[(3-methyl-2,6-dioxo-1,7-dipentyl-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanamide; 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanamide; 7-(3-bromobenzyl)-8-[(1-ethylpropyl)thio]-3-methyl-3,7-dihydro-1H-purine-2,6-dione; ethyl 2-{8-[(3-bromobenzyl)thio]-1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl}butanoate; and ethyl 2-[(7-isobutyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate.
  • In some embodiments compounds are: ethyl 2-[(7-benzyl-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(2-methoxyethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1,3-dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(cyanomethyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-propyl-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-1-(3-hydroxypropyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thiol}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-1-(2-propynyl)-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-aminobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-({7[(3,5-dimethylisoxazol-4-yl)methyl]-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate: ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate; ethyl 2-[(7-hexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio]butanoate: 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanamide; 7-(3-bromobenzyl)-8-[(1-ethylpropyl)thio]-3-methyl-3,7-dihydro-1H-purine-2,6-dione; and ethyl 2-{8-[(3-bromobenzyl)thio]-1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl}butanoate.
  • In some embodiments compounds are: ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl]thio}butanoate; and ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl}thio)butanoate.
  • The acid addition salt form of a compound of formula I that occurs in its free form as a base can be obtained by treating the free base with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, trifluoroacetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like.
  • The compounds of formula I containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt forms, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts. e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.
  • Compounds of the formula I and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like.
  • Many of the compounds of formula I and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem., 45 (1976) 11-30.
  • The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula I or mixtures thereof (including all possible mixtures of stereoisomers).
  • With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof, unless the particular isomeric form is referred to specifically. Compounds according to the present invention may exist in different polymorphic forms. Although not explicitly indicated in the above formula, such forms are intended to be included within the scope of the present invention.
  • xiii) U.S. Pat. No. 7,465,549
  • In some embodiments, the compound includes optionally substituted N-alkylated 2-oxo-pyrrolidine derivatives. In some embodiments, those compounds are alkyl amides derivatives substituted on the positions 4 and/or 5 of the pyrrolidone ring. Examples of optionally substituted N-alkylated 2-oxo-pyrrolidine derivatives include, but are not limited to, compounds such as (2S)-2-[(4S)-4-(2,2-difluorovinyl)-2-oxopyrrolidinyl]butanamide, (2S)-2-[(4R)-2-oxo-4-propylpyrrolidinyl]butanamide, (2S)-2-[(4S)-2-oxo-4-propylpyrrolidinyl]butanamide, and (2S)-2-[4-(3-azidophenyl)-2-oxopyrrolidin-1-yl]butanamide.
  • In some embodiments, the compounds further include optionally substituted N-alkylated 2-oxo-piperidinyl derivatives. In some embodiments, those compounds are alkyl amides derivatives substituted on the position 4 and/or 5 and/or 6 of the 2-oxo-piperidinyl ring. Examples of optionally substituted N-alkylated 2-oxo-pyrrolidine derivatives include, but are not limited to, compounds such as those referred to in international patent application PCT/EP02/05503 such as (2S)-2-[5-(iodomethyl)-2-oxo-1-piperidinyl]butanamide, (2S)-2-[5-(azidomethyl)-2-oxo-1-piperidinyl]butanamide, 2-(2-oxo-5-phenyl-1-piperidinyl)butanamide, (2S)-2-[4-(iodomethyl)-2-oxo-1-piperidinyl]butanamide, and (2S)-2-[4-(2-fluoro-2-methylpropyl)-2-oxo-1-pyrrolidinyl]butanamide.
  • In some embodiments, the compounds include any acetam compound of formula I, in racemic or isomeric form, or a pharmaceutically acceptable salt thereof.
  • Figure US20140206667A1-20140724-C00084
  • wherein
    R represents hydrogen or hydroxy;
    R1 and R2 represent independently hydrogen or an alkyl group of 1-4 carbon atoms; and
    R3 and R4 represent independently hydrogen, an alkyl group of 1-4 carbon atoms or —(CH2)n-NR5R6 wherein n is 1, 2 or 3 and R5 and R6 represent independently hydrogen or an alkyl group of 1-4 carbon atoms.
  • An example of such an acetam compound includes, but is not limited to, a compound of formula I wherein R, R1, R2, R3 and R4 are hydrogen, 2-oxo-pyrrolidineacetamide, known by the generic name piracetam as described in UK Patents Nos. 1,039,113 and 1,309.692.
  • In some embodiments, the compounds also include optionally substituted N-alkylated 2-oxo-azepanyl derivatives. Preferably, those compounds are alkyl amides derivatives substituted on the positions 4 and/or 5 and/or 6 and/or 7 of the 2-oxo-azepanyl ring. Examples of optionally substituted N-alkylated 2-oxo-azepanyl derivatives include, but are not limited to, compounds such as those referred to in international patent application PCT/EP02/05503 such as 2-[5-(iodomethyl)-2-oxo-1-azepanyl]butanamide.
  • xiv) U.S. Patent Application Publication No. 2006258704
  • This invention provides novel compounds of the formula I
  • Figure US20140206667A1-20140724-C00085
  • wherein
    n represents 0 or 1 whereby R<1> is not existent when n=0 and R<1> is existent when n=1;
    A<1> represents an oxygen or a sulfur atom;
  • X is —CONR<7>R<8>, —COOR<9>, —CO—R<10> or CN;
  • R<1> when existent, R<2>, R<3>, R<4> and R<5> are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative, provided that at least one of the substituents R chosen from R<1> when existent, R<2>, R<3>, R<4> or R<5> is not hydrogen;
    R<6> is hydrogen, alkyl, aryl or —CH2-R<6a> wherein R<6a> is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
    R<7>, R<8> and R<9> are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or an oxy derivative; and
    R<10> is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative;
    their pharmaceutically acceptable salts, geometrical Isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers).
  • In the above formula, at least one substituent R<1> to R<5> is different from hydrogen. Some non-substituted compounds are referred to in U.S. Pat. Nos. 5,468,733 and 5,516,759. U.S. Pat. No. 5,468,733 discloses non-ring substituted 2-oxo-1-pyrrolidinyl and 2-oxo-1-piperidinyl derivatives as inhibitors of the oncogene Ras protein. In particular, these compounds block the ability of Ras to transform normal cells to cancer cells, and therefore can be included in several chemotherapeutic compositions for treating cancer.
  • U.S. Pat. No. 5,516,759 discloses non-ring substituted 2-oxo-1-pyrrolidinyl. 2-oxo-1-piperidinyl and azepanyl derivatives present at the N-terminus of dodecapeptides possessing LHRH (luteinizing hormone-releasing hormone) antagonistic activity. Such LHRH antagonists are useful in the treatment of a variety of conditions in which suppression of sex steroids plays a key role including contraception, delay of puberty, treatment of benign prostatic hyperplasia a.o.
  • In the definitions set forth below, unless otherwise stated. R<11> and R<12> are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfinyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.
  • The term “oxy derivative”, as used herein, is defined as including —O—R<11> groups wherein R<11> is as defined above except for “oxy derivative”. Non-limiting examples are alkoxy, alkenyloxy, alkynyloxy, acyloxy, oxyester, oxyamido, alkylsulfonyloxy, alkylsulfinyloxy, arylsulfonyloxy, arylsulfinyloxy, aryloxy, aralkoxy or heterocyclooxy such as pentyloxy, allyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methylenedioxy, carbonate.
  • The term “thio derivative”, as used herein, is defined as including —S—R<11> groups wherein R<11> is as defined above except for “thio derivative”. Non-limiting examples are alkylthio, alkenylthio, alkynylthio and arylthio.
  • The term “amino derivative”, as used herein, is defined as including —NHR<11> or —NR<11>R<12> groups wherein R<11> and R<12> are as defined above. Non-limiting examples are mono- or di-alkyl-, alkenyl-, alkynyl- and arylamino or mixed amino.
  • The term “acyl derivative”, as used herein, represents a radical derived from carboxylic acid and thus is defined as including groups of the formula R<11>-CO—, wherein R<11> is as defined above and may also be hydrogen. Preferred are acyl derivatives of formula —COR<11> wherein R<11> is selected from hydrogen, C1-12 alkyl, C2-12 alkenyl, C2-12 alkenyl, heterocyle and aryl. Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl, heptanedioyl, cyclohexanecarbonyl, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxalyl, ethoxalyl, cysteinyl, oxamoyl.
  • The term “sulfonyl derivative”, as used herein, is defined as including a group of the formula —SO—R<11>, wherein R<11> is as defined above except for “sulfonyl derivative”. Non-limiting examples are alkylsulfonyl, alkenylsulfonyl, alkynylsulfonyl and arylsulfonyl.
  • The term “sulfinyl derivative”, as used herein, is defined as including a group of the formula —SO—R<1>, wherein R<11> is as defined above except for “sulfinyl derivative”. Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, alkynylsulfinyl and arylsulfinyl.
  • The term “alkyl”, as used herein, is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and generally containing 1-20 carbon atoms, most often 1 to 12 carbon atoms, preferably 1-7 carbon atoms for non-cyclic alkyl and 3-7 carbon atoms for cycloalkyl (in these two preferred cases, unless otherwise specified, “lower alkyl”), each optionally substituted by, preferably 1 to 5, substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cycloalkyl, sulfonic acid, sulfonamide, thio derivative, alkylthio, oxyester, oxyamido, heterocycle, vinyl, alkoxy (preferably C1-5), aryloxy (preferably C6-10) and aryl(preferably C6-10).
  • In some embodiments are alkyl groups containing 1 to 7 carbon atoms, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl. Most preferred are C1-4 alkyl and C3-7 cycloalkyl, each optionally substituted by one or more hydroxy, halogen, lower alkyl or/and azido.
  • In some embodiments are alkyl groups are hydroxymethyl, propyl, butyl, 2,2,2-trifluoroethyl, 2-bromo-2,2-difluoroethyl, 2-chloro-2,2-difluoroethyl, 3,3,3-trifluoropropyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2,2-difluoropropyl. 2-iodo-2,2-difluoroethyl.
  • The term “lower alkyl”, as used herein, and unless otherwise specified, refers to C1 to C7 saturated straight, branched or cyclic hydrocarbon. Non limiting examples are methyl, ethyl, propyl, isopropyl, butyl, tertiobutyl, pentyl, cyclopropyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methypentyl, 2,2-dimethylbutyl, optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferably, lower alkyl is methyl.
  • The term “alkenyl”, as used herein, is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, thiocyanato, azido, alkylthio, cycloalkyl, acyl, nitro, cyano, aryl and heterocycle.
  • In some embodiments are alkenyl groups are C2-C1-2 alkenyls, especially C2-6 alkenyls, such as ethenyl (=vinyl), 1-methyl-1-ethenyl, 2,2-dimethyl-1-ethenyl, 1-propenyl, 2-propenyl (=allyl), 1-butenyl, 2-butenyl, 3-butenyl, 4-pentenyl, 1-methyl-4-pentenyl, 3-methyl-1-pentenyl, 1-hexenyl. 2-hexenyl and the like, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl. Most preferred is vinyl, optionally substituted by one or more halogen or/and lower alkyl, and especially 2,2-difluorovinyl, 2,2-dibromovinyl and 2,2-dichlorovinyl.
  • The term “alkynyl” as used herein, is defined as including a monovalent branched or unbranched hydrocarbon radical containing at least one carbon-carbon triple bond, for example ethynyl. 2-propynyl (=propargyl), and the like, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl, heterocycle, thiocyanato, azido, alkylthio, alkyl and acyl.
  • In some embodiments are alkynyl groups are C2-12 alkynyl, especially C2-6 alkynyl, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, acyl, aryl such as phenyl and alkyl, preferably cycloalkyl.
  • In some embodiments are ethynyl, propynyl and butynyl, optionally substituted by lower alkyl or/and halogen, and especially 1-propynyl, cyclopropylethynyl, 3-methyl-1-butynyl and 3,3,3-trifluoro-1-propynyl.
  • When present as bridging groups, alkyl, alkenyl and alkynyl represent straight- or branched chains, C1-12, preferably C1-4-alkylene or C2-12-, preferably C2-4-alkenylene or -alkynylene moieties respectively.
  • Groups where branched derivatives are conventionally qualified by prefixes such as “n”, “sec”, “iso” and the like (e.g. “n-propyl”, “sec-butyl”) are in the n-form unless otherwise stated.
  • The term “aryl”, as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of at least one ring, most often 1 to 3 rings and generally containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl, each optionally substituted by one or more substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfinyl, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, C1-6-alkylthio, oxyester, oxyamido, aryl, C1-6-alkoxy, C6-10-aryloxy, C1-6-alkyl, C1-6-haloalkyl. Aryl radicals are preferably monocyclic or bicyclic containing 6-10 carbon atoms. Preferred aryl groups are phenyl and naphthyl each optionally substituted by one or more substituents independently selected from halogen, nitro, amino, azido, C1-6-alkoxy, C1-6-alkyl, C1-6-haloalkyl, sulfonyl and phenyl.
  • In some embodiments the aryl is phenyl, optionally substituted by one or more halogen, lower alkyl, azido or nitro, such as 3-chlorophenyl and 3-azidophenyl.
  • The term “halogen”, as used herein, includes an atom of Cl, Br, F, 1.
  • The term “hydroxy”, as used herein, represents a group of the formula —OH.
  • The term “thiol”, as used herein, represents a group of the formula —SH.
  • The term “cyano”, as used herein, represents a group of the formula —CN.
  • The term “nitro”, as used herein, represents a group of the formula —NO2.
  • The term “nitrooxy”, as used herein, represents a group of the formula —ONO2.
  • The term “amino”, as used herein, represents a group of the formula —NH2.
  • The term “azido”, as used herein, represents a group of the formula —N3.
  • The term “carboxy”, as used herein, represents a group of the formula —COOH.
  • The term “sulfonic acid”, as used herein, represents a group of the formula —SO3H.
  • The term “sulfonamide”, as used herein, represents a group of the formula —SO2NH2.
  • The term “ester”, as used herein, is defined as including a group of formula —COO—R<11> wherein R<11> is as defined above except oxy derivative, thio derivative or amino derivative. Preferred are esters of formula —COOR<11> wherein R<11> is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl and aryl. Most preferred are esters where R<11> is a lower alkyl, especially methyl.
  • The term “ether” is defined as including a group selected from C1-50-straight or branched alkyl, or C2-50-straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.
  • The term “amido” is defined as including a group of formula —CONH2 or —CONHR<11> or —CONR<11> R<12> wherein R<11> and R<12> are as defined above.
  • The term “heterocycle”, as used herein, is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl, and optionally being substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups. Non-limiting examples of heterocycles are pyridyl, furyl, pyrrolyl, thienyl, isothiazolyl, triazolyl, imidazolyl, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolyl, isoquinolyl, isobenzofuranyl, benzothienyl, pyrazolyl, indolyl, indolizinyl, purinyl, isoindolyl, carbazolyl, thiazolyl, 1,2,4-thiadiazolyl, thiomorpholinyl, thieno(2,3-b)furanyl, furopyranyl, benzofuranyl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl, benzothiazolyl, or benzoxazolyl, cinnolinyl, phthalazinyl, quinoxalinyl, 1-oxidopyridyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenothiazinyl, furazanyl, benzodioxolyl, isochromanyl, indolinyl, xanthenyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro(4.5)dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i.e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) optionally substituted by alkyl or as described above for the alkyl groups. The term “heterocycle” also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo(2.2.1)heptanyl, 7-oxabicyclo(2.2.1)heptanyl, 8-azabicyclo(3.2.1)octanyl.
  • The heterocycle may be selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl. In some embodiments the heterocycle is selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl, and especially from 2- and 3-thienyl, optionally substituted by one or more halogen, acyl such as formyl, cyano and/or lower alkyl, such as methyl.
  • In the above definitions it is to be understood that when a substituent such as R<1>, R<2>, R<3>, R<4>, R<5>. R<7>, R<8>, R<9>, R<10> is attached to the rest of the molecule via a heteroatom or a carbonyl, a straight- or branched chain, C1-12-, preferably C1-4-alkylene or C2-12, preferably C2-4-alkenylene or -alkynylene bridge may optionally be interposed between the heteroatom or the carbonyl and the point of attachment to the rest of the molecule.
  • The acid addition salt form of a compound of formula (I) that occurs in its free form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like.
  • The compounds of formula (I) containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt form, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.
  • Compounds of the formula I and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like.
  • Many of the compounds of formula I and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem. (1976), 45, 11-30.
  • The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula I or mixtures thereof (including all possible mixtures of stereoisomers).
  • Furthermore, certain compounds of formula I which contain alkenyl groups may exist as Z (zusammen) or E (entgegen) isomers. In each instance, the invention includes both mixture and separate individual isomers.
  • Multiple substituents on the piperidinyl or the azepanyl ring can also stand in either cis or trans relationship to each other with respect to the plane of the piperidinyl or the azepanyl ring.
  • Some of the compounds of formula I may also exist in tautomeric forms. Such forms although not explicitly indicated in the above formula are intended to be included within the scope of the present invention.
  • With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof unless the particular isomeric form is referred to specifically.
  • The invention also includes within its scope prodrug forms of the compounds of formula I and Its various sub-scopes and sub-groups.
  • The term “prodrug” as used herein includes compound forms which are rapidly transformed in vivo to the parent compound according to the invention, for example, by hydrolysis in blood. Prodrugs are compounds bearing groups which are modified by biotransformation prior to exhibiting their pharmacological action. Such groups include moieties which are readily oxidised, cyclised or cleaved, which compound after biotransformation remains or becomes pharmacologically active. For example, metabolically cleavable groups form a class of groups well known to practitioners of the art. They include, but are not limited to such groups as alkanoyl (i.e. acetyl, propionyl, butyryl, and the like), unsubstituted and substituted carbocyclic aroyl (such as benzoyl, substituted benzoyl and 1- and 2-naphthoyl), alkoxycarbonyl (such as ethoxycarbonyl), trialkylsilyl (such as trimethyl- and triethylsilyl), monoesters formed with dicarboxylic acids (such as succinyl), phosphate, sulfate, sulfonate, sulfonyl, sulfinyl and the like. The compounds bearing the biotransformable groups have the advantage that they may exhibit improved bioavailability as a result of enhanced solubility and/or rate of absorption conferred upon the parent compound by virtue of the presence of the biotransformable group. T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery System”, Vol. 14 of the A.C.S. Symposium Series; “Bioreversible Carriers in Drug Design”, ed. Edward B. Roche. American Pharmaceutical Association and Pergamon Press, 1987.
  • The term “R substituent” refers to R<1>, R<2>, R<3>, R<4> or R<5>, independently.
  • According to one embodiment, the present invention relates to a compound of formula I as defined above wherein n represents 0. The compound is a 6-ring structure (2-thioxo- or 2-oxo-piperidinyl derivative) wherein R<1> is not existent since n=0, and is depicted by the formula (I-A).
  • Figure US20140206667A1-20140724-C00086
  • According to a following embodiment, the present invention relates to a compound of formula I according to the invention as defined above wherein n represents 1. The compound is a 7-ring structure (2-thioxo- or 2-oxo-azepanyl derivative) wherein R<1> is existent since n=1 and depicted by the formula (I-B).
  • Figure US20140206667A1-20140724-C00087
  • According to one embodiment, the invention relates to said compound as defined above wherein n=0, R<3> and/or R<4> are different from hydrogen and R<2> and R<5> represent hydrogen.
  • According to another embodiment, the invention relates to said compound as defined above wherein n=1, R<2>, R<3> and/or R<4> are different from hydrogen and wherein R<1> and R<5> represent hydrogen.
  • According to another embodiment, the invention relates to said compound as defined above wherein only one R substituent chosen from R<3> or R<4> when n=0 or from R<2>, R<3> or R<4> when n=1, is different from hydrogen and the remaining R substituent(s) is/are hydrogen. We hereby refer to a mono-substituted 2-thioxo- or 2-oxo-piperidinyl or 2-thioxo- or 2-oxo-azepanyl derivatives.
  • According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein A<1> represents an oxygen atom. We hereby refer to 2-oxo-piperidinyl or 2-oxo-azepanyl derivatives.
  • According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein X is CONR<7> R<8>, especially CONH2. We hereby refer to amido derivatives of 2-oxo(or thioxo)-piperidinyl or 2-oxo(or thioxo)-azepanyl.
  • According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein R<6> represents hydrogen, C1-4 alkyl, or a CH2-R<6a> group wherein R<6a> represents a heterocycle. Most preferably R<6> is a C1-4 alkyl, especially ethyl. When R<6> is ethyl we refer to 2-(2-oxo(or thioxo)-1-piperidinyl)butanamide or 2-(2-oxo(or thioxo)-1-azepanyl)butanamide derivatives.
  • According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein the carbon atom to which R<6> is attached is of the S configuration. In case where R<6> is ethyl, A is oxygen and X is CON R<7>R<8>, we refer then to (2S)-2-(2-oxo-1-piperidinyl)butanamide or (2S)-2-(2-oxo-1-azepanyl)butanamide derivatives.
  • According to one embodiment, the present invention relates to a compound as defined above wherein R<2> when n=1, R<3> and R<4> are the same or different and each is independently hydrogen, halogen, nitro, nitrooxy, cyano, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, acyl derivative, sulfonyl derivative or sulfinyl derivative:
  • R<1> when existent, R<2> when n=0 and R<5> are hydrogen;
    R<6> is hydrogen, alkyl, aryl or —CH2-R<6a> wherein R<6a> is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;
    provided that, when R<6> is hydrogen, X is —CONR<7>R<8> and that the compound is
    neither methyl (2R)-2-[(6R)-6-methyl-2-oxoazepanyl]-3-phenylpropanoate nor methyl (2S)-2-[(4R)-4-methyl-2-oxoazepanyl]-3-phenylpropanoate.
  • According to this embodiment, the compound is generally such that when R<6> is benzyl, X is —COOCH3 and n=1, R<2> is different from methyl when R<3> and R<4> are both hydrogen and R<4> is different from methyl when R<2> and R<3> are both hydrogen.
  • According to another embodiment, the present invention relates to a compound as defined above wherein R<2> when n=1, R<3> and R<4> are the same or different and each is independently hydrogen; cyano; carboxy; amido;
  • C1-12 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cycloalkyl, acyl, aryl and heterocycle;
    C2-12 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl;
    C2-12 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl: acyl derivative of formula —CO—R<11>, wherein R<11> is selected from C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, heterocycle and aryl; ester of formula —CO—O—R<11> wherein R<1> is selected from C1-12 alkyl. C2-12 alkenyl, C2-12 alkynyl and aryl:
    heterocycle selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1-oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl;
    aryl, each optionally substituted by one or more substituents selected from C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 alkylthio, amino, azido, sulfonyl, aryl and nitro.
  • According to another embodiment, the present invention relates to a compound as defined above, wherein R<2> when n=1, R<3> and R<4> are the same or different and each is independently hydrogen;
  • C1-7 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl:
    C2-6 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl:
    C2-6 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl: heterocycle selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl: phenyl, each optionally substituted by one or more substituents selected from C1-6 alkyl, halogen substituted alkyl, halogen, alkoxy, amino, azido, sulfonyl, phenyl and nitro.
  • According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2>, R<3> and R<4> when n=1 or from the group R<3> and R<4> when n=0, represents independently C1-4-alkyl or C3-7-cycloalkyl, optionally substituted by one or more halogen, hydroxy, lower alkyl and/or azido.
  • According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2>, R<3> and R<4> when n=1 or from the group R<3> and R<4> when n=0, represents independently vinyl, optionally substituted by one or more halogen or/and lower alkyl.
  • According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2>, R<3> and R<4> when n=1 or from the group R3 and R<4> when n=0, represents independently ethynyl, propynyl or butynyl, optionally substituted by one or more halogen and/or lower alkyl.
  • According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2>, R<3> and R<4> when n=1 or from the group R<3> and R<4> when n=0, represents independently phenyl, optionally substituted by one or more halogen, lower alkyl, azido and/or nitro.
  • According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2>, R<3> and R<4> when n=1 or from the group R<3> and R<4> when n=0, represents independently 2- or 3-thienyl, optionally substituted by one or more halogen, acyl, cyano or/and lower alkyl.
  • According to a particular embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<3>, R<4> and R<2> when n=1 or from the group R<3> and R<4> when n=0, is hydroxymethyl, propyl, butyl, 3,3,3-trifluoropropyl, 2,2,2-trifluoroethyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2-thienyl, 3-thienyl, phenyl. 3-chlorophenyl, 3-azidophenyl, 2,2-difluorovinyl, 2,2-dibromovinyl, 2,2-dichlorovinyl, 2-ethynyl, 5-methyl-2-thienyl, 5-formyl-2-ethynyl, 5-cyano-2-thienyl, 3-bromo-2-thienyl, 4-methyl-2-thienyl. 3,3,3-trifluoro-1-propynyl, 1-propynyl, cyclopropylethynyl, 3-methyl-1-butynyl, 1-butynyl, 2,2-difluoropropyl, 2-chloro-2,2-difluoroethyl, 2-bromo-2,2-difluoroethyl and 2-iodo-2,2-difluoroethyl.
  • According to yet another embodiment, the present invention relates to a compound as defined above wherein R<1>, R<2>, R<4> and R<5> are hydrogen.
  • According to another embodiment, the present invention relates to a compound as defined above wherein R<1>. R<2>, R<3> and R<5> are hydrogen.
  • According to another embodiment, the present invention relates to a compound as defined above wherein n=1 and R<1>, R<3>, R<4> and R<5> are hydrogen.
  • In all the above-mentioned scopes when the carbon atom to which R<6> is attached is asymmetric it may be in the “S”-configuration.
  • Representative compounds of this invention as defined above are selected from the group consisting of 2-[5-(hydroxymethyl)-2-oxo-1-piperidinyl]butanamide, 2-(2-oxo-5-propyl-1-piperidinyl)butanamide. 2-[2-oxo-5-(3,3,3-trifluoropropyl)-1-piperidinyl]butanamide, 2-[5-(cyclopropylmethyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(iodomethyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(azidomethyl)-2-oxo-1-piperidinyl]butanamide, 2-(2-oxo-5-phenyl-1-piperidinyl)butanamide, 2-[2-oxo-5-(2-thienyl)-1-piperidinyl]butanamide, 2-[2-oxo-5-(3-thienyl)-1-piperidinyl]butanamide, 2-[5-(3-chlorophenyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(3-azidophenyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(2,2-difluorovinyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(2,2-dibromovinyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(2,2-dichlorovinyl)-2-oxo-1-piperidinyl]butanamide, 2-(5-ethynyl-2-oxo-1-piperidinyl)butanamide, 2-[5-(5-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(5-formyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(5-cyano-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(3-bromo-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(4-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[2-oxo-5-(3,3,3-trifluoro-1-propynyl)-1-piperidinyl]butanamide, 2-[2-oxo-5-(1-propynyl)-1-piperidinyl]butanamide, 2-[5-(cyclopropylethynyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(3-methyl-1-butynyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(1-butynyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(2,2-difluoropropyl)-2-oxo 1-piperidinyl]butanamide, 2-[5-(2-chloro-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(2-bromo-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(hydroxymethyl)-2-oxo-1-piperidinyl]butanamide, 2-(2-oxo-4-propyl-1-piperidinyl)butanamide, 2-[2-oxo-4-(3,3,3-trifluoropropyl)-1-piperidinyl]butanamide, 2-14-(cyclopropylmethyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(iodomethyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(azidomethyl)-2-oxo-1-piperidinyl]butanamide, 2-(2-oxo-4-phenyl-1-piperidinyl)butanamide, 2-12-oxo-4-(2-thienyl)-1-piperidinyl]butanamide, 2-[2-oxo-4-(3-thienyl)-1-piperidinyl]butanamide, 2-[4-(3-chlorophenyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(3-azidophenyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2,2-difluorovinyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2,2-dibromovinyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2,2-dichlorovinyl)-2-oxo-1-piperidinyl]butanamide, 2-(4-ethynyl-2-oxo-1-piperidinyl)butanamide, 2-[4-(5-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(5-formyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(5-cyano-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(3-bromo-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(4-methyl-2-thienyl)-2-oxo-1-piperidinyl]butanamide, 2-[2-oxo-4-(3,3,3-trifluoro-1-propynyl)-1-piperidinyl]butanamide, 2-[2-oxo-4-(1-propynyl)-1-piperidinyl]butanamide, 2-[4-(cyclopropylethynyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(3-methyl-1-butynyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(1-butynyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2,2-difluoropropyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2-chloro-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2-bromo-2,2-difluoroethyl)-2-oxo-1-piperidinyl]butanamide, 2-[4-(2,2,2-trifluoroethyl)-2-oxo-1-piperidinyl]butanamide, 2-[5-(hydroxymethyl)-2-oxo-1-azepanyl]butanamide. 2-(2-oxo-5-propyl-1-azepanyl)butanamide, 2-[2-oxo-5-(3,3,3-trifluoropropyl)-1-azepanyl]butanamide, 2-(5-(cyclopropylmethyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(iodomethyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(azidomethyl)-2-oxo-1-azepanyl]butanamide, 2-(2-oxo-5-phenyl-1-azepanyl)butanamide, 2-[2-oxo-5-(2-thienyl)-1-azepanyl]butanamide, 2-[2-oxo-5-(3-thienyl)-1-azepanyl]butanamide, 2-[5-(3-chlorophenyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(3-azidophenyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(2,2-difluorovinyl)-2-oxo-1-azepanyl]butanamide. 2-[5-(2,2-dibromovinyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(2,2-dichlorovinyl)-2-oxo-1-azepanyl]butanamide, 2-(5-ethynyl-2-oxo-1-azepanyl)butanamide, 2-[5-(5-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(5-formyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(5-cyano-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(3-bromo-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(4-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[2-oxo-5-(3,3,3-trifluoro-1-propynyl)-1-azepanyl]butanamide, 2-[2-oxo-5-(1-propynyl)-1-azepanyl]butanamide, 2-[5-(cyclopropylethynyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(3-methyl-1-butynyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(1-butynyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(2,2-difluoropropyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(2-chloro-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(2-bromo-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[5-(2,2,2-trifluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(hydroxymethyl)-2-oxo-1-azepanyl]butanamide, 2-(2-oxo-6-propyl-1-azepanyl)butanamide, 2-[2-oxo-6-(3,3,3-trifluoropropyl)-1-azepanyl]butanamide, 2-[6-(cyclopropylmethyl)-2-oxo-1-azepanyl]butanamide. 2-[6-(iodomethyl)-2-oxo-1-azepanyl]butanamide, 2-16-(azidomethyl)-2-oxo-1-azepanyl]butaxiamide, 2-(2-oxo-6-phenyl-1-azepanyl)butanamide, 2-[2-oxo-6-(2-thienyl)-1-azepanyl]butanamide, 2-[2-oxo-6-(3-thienyl)-1-azepanyl]butanamide, 2-[6-(3-chlorophenyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(3-azidophenyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(2,2-difluorovinyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(2,2-dibromovinyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(2,2-dichlorovinyl)-2-oxo-1-azepanyl]butanamide, 2-(6-ethynyl-2-oxo-1-azepanyl)butanamide. 2-[6-(5-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(5-formyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(5-cyano-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(3-bromo-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(4-methyl-2-thienyl]-2-oxo-1-azepanyl]butanamide, 2-[2-oxo-6-(3,3,3-trifluoro-1-propynyl)-1-azepanyl]butanamide, 2-[2-oxo-6-(1-propynyl)-1-azepanyl]butanamide, 2-[6-(cyclopropylethynyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(3-methyl-1-butynyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(1-butynyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(2,2-difluoropropyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(2-chloro-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[6-(2-bromo-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide. 2-[6-(2,2,2-trifluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(hydroxymethyl)-2-oxo-1-azepanyl]butanamide, 2-(2-oxo-4-propyl-1-azepanyl)butanamide, 2-[2-oxo-4-(3,3,3-trifluoropropyl)-1-azepanyl]butanamide, 2-14-(cyclopropylmethyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(iodomethyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(azidomethyl)-2-oxo-1-azepanyl]butanamide, 2-(2-oxo-4-phenyl-1-azepanyl)butanamide, 2-[2-oxo-4-(2-thienyl)-1-azepanyl]butanamide, 2-[2-oxo-4-(3-thienyl)-1-azepanyl]butanamide, 2-[4-(3-chlorophenyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(3-azidophenyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(2,2-difluorovinyl)-2-oxo-1-azepanyl]butanamide. 2-[4-(2,2-dibromovinyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(2,2-dichlorovinyl)-2-oxo-1-azepanyl]butanamide, 2-(4-ethynyl-2-oxo-1-azepanyl)butanamide, 2-[4-(5-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(5-formyl-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[<4>-(5-cyano-<2>-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(3-bromo-2-thienyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(4-methyl-2-thienyl)-2-oxo-1-azepanyl]butanamide. 2-[2-oxo-4-(3,3,3-trifluoro-1-propynyl)-1-azepanyl]butanamide, 2-[2-oxo-4-(1-propynyl)-1-azepanyl]butanamide, 2-[4-(cyclopropylethynyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(3-methyl-1-butynyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(1-butynyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(2,2-difluoropropyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(2-chloro-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(2-bromo-2,2-difluoroethyl)-2-oxo-1-azepanyl]butanamide, 2-[4-(2,2,2-trifluoroethyl)-2-oxo-1-azepanyl]butanamide.
  • Results have been obtained with the following compounds:
    • (2S)-2-[5-(iodomethyl)-2-oxo-1-piperidinyl]butanamide,
    • (2S)-2-[5-(azidomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-(2-oxo-5-phenyl-1-piperidinyl]butanamide.
    • (2S)-2-[4-(iodomethyl)-2-oxo-1-piperidinyl]butanamide,
    • 2-[5-(iodomethyl)-2-oxo-1-azepanyl]butanamide.
  • xv) International Patent Application Publication No. WO2008/132139
  • In some embodiments, the compounds are of formula (I) as follows:
  • Figure US20140206667A1-20140724-C00088
  • wherein
    Y is O or S. In some embodiments Y is O. R1 is hydrogen or C-|.g alkyl:
    R2 is hydrogen;
    R3 is —CONR5R6, —COR7, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; R5, R6 are the same or different and are independently selected from hydrogen and C-16 alkyl;
    R7 is C<;|6 alkyl;
    A is a monocyclic or bicyclic heterocyclic moiety selected from the group consisting of imidazolidin-1-yl, 1,3-oxazolidin-3-yl, 2,5-dihydro-1H-pyrrol-1-yl, 1,3-thiazol-3(2H)-yl, 1,3-thiazolidin-3-yl, piperidin-1-yl, azepan-1-yl, 5,6-dihydro-4H-thieno[3,2-b]pyrrol-4-yl, hexahydro-4H-thieno[3,2-b]pyrrol-4-yl, 2,3-dihydro-1H-thieno[3,4-b]pyrrol-1-yl, 1,3-benzothiazol-3(2H)-yl, 1,3-benzoxazol-3(2H)-yl, pyrazolo[1,5-a]pyridin-1(2H)-yl, 3,4-dihydroisoquinolin-2(1H)-yl, 3,4-dihydroquinolin-1(2H)-yl, 1,3,4,5-tetrahydro-2H-2-benzazepin-2-yl, 1,2,4,5-tetrahydro-3H-3-benzazepin-3-yl; R4 is either R̂a or R̂ depending on whether A being is a monocyclic or a bicyclic heterocycle:
    where A is a monocyclic heterocyclic moiety. R̂ is R̂a which is selected from the group consisting of hydrogen; C-|.g alkyl optionally substituted by a substituent selected from halogen, C-1.4 alkoxy, C-1.4 alkylthio, azido, nitrooxy or an aryl;
    C2-6 alkenyl optionally substituted by halogen; C2-6 alkynyl optionally substituted by halogen; azido; alkoxycarbonylamino; arylsulfonyloxy; a substituted or unsubstituted aryl; or a 3-8 membered substituted or unsubstituted heterocycle;
    where A is a bicyclic heterocyclic moiety R̂ is R̂ which is selected from the group comprising or consisting of hydrogen; nitro; cyano; halogen: heterocycle; amino; aryl; C-|.g alkyl optionally substituted by at least one halogen; or C-|.g alkoxy optionally substituted by at least one halogen;
  • In some embodiments the compounds are as follows:
  • For compounds where A=Y is selected from a 2-oxo-piperidin-1-yl, a 2-oxo-azepan-1-yl, a 2-oxo-1,3-benzothiazol-3(2H)-yl or a 2-oxo-1,3-benzoxazol-3(2H)-yl, R3 must be selected from an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl.
  • For compounds where A=Y is a 5-oxoimidazolidin-1-yl, R̂ and R̂ are hydrogen, R3 is —CONR5R6, R5 and| R6 are as above defined, then R̂a may not be an alkyl, aralkyl or substituted aralkyl.
  • Where A=Y is either of a 2-oxo-piperidin-1-yl and a 2-oxo-azepan-1-yl, R̂, R̂ and R̂a are all hydrogen, then R̂ could not be a 2-phenylimidazo[1,2-a]pyridin-3-yl.
  • In a specific embodiment A=Y is selected from the list consisting of:
  • Figure US20140206667A1-20140724-C00089
  • wherein X is O or S, in a more specific embodiment O; in another embodiment, X is S.
  • The asterisks in the above illustration indicate the attachment sites of the substituent R̂a.
  • In a specific embodiment, when R̂ is —CONR5R6 and R̂ is C-μg alkyl, the carbon atom to which R-I and R̂ are attached is preferably in the “S”-configuration.
  • In a specific embodiment R̂ is hydrogen, methyl, ethyl and R̂ is hydrogen. In a specific embodiment R3 is —CONH2.
  • In a further specific embodiment R̂ is 1H-imidazol-1-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, imidazo[1,2-a]pyridin-3-yl or imidazo[1,2-b]pyridazin-3-yl. In a specific embodiment R̂a is a C-|.g alkyl which may optionally be substituted by a halogen; or a phenyl.
  • In another specific embodiment R̂b is hydrogen, halogen, nitro, cyano or a C-μg alkyl optionally substituted by a halogen.
  • In still a further embodiment compounds may be used in the treatment of the above mentioned disorders, in particular of epilepsy, having the formula (I-E), as wells as its geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00090
  • wherein
  • X is O or S;
  • R-I is hydrogen or C-|.g alkyl, in a more specific embodiment hydrogen:
    R3 is an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; R̂b is hydrogen: nitro; cyano; halogen; C-|.g alkyl optionally substituted by halogen; C-|.g alkoxy optionally substituted by halogen.
  • A further aspect of the present invention consists in novel compounds having the formula (I-A), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00091
  • wherein
    R1 is hydrogen or C-|.g alkyl, preferably hydrogen, methyl or ethyl; in a more specific embodiment R̂ is ethyl.
    R3 is —CONH2, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl, preferably R̂ is —CONH2.
    R̂a is either hydrogen or an aryl; with the proviso that 2-(5-oxoimidazolidin-1-yl)acetamide is excluded. Preferably R̂a is an aryl, e.g. a phenyl which may be substituted preferably by halogen, nitro, alkoxy, in particular by nitro.
  • In a particular embodiment, when R̂ is —CONH2 and R̂ is C-|.g alkyl, the carbon atom to which R1 and R̂ are attached is preferably in the “S”-configuration. A further aspect of the present invention consists in novel compounds having the formula (I-B1 or I-B2), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00092
  • wherein X in formula (I-B2) is either S or O, in a more specific embodiment S;
    R1 is hydrogen or C-|.g alkyl, preferably hydrogen, methyl or ethyl; in a more specific embodiment R̂ is ethyl.
    R3 is —CONH2, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; preferably R̂ is —CONH2
    R̂a is hydrogen; C-|.g alkyl optionally substituted by halogen or C-1.4 alkoxy: an aryl; or C2.g alkenyl optionally substituted by halogen. Preferably, R̂a is C-|.g alkyl optionally substituted by halogen or C2-6 alkenyl optionally substituted by halogen or an aryl. In a more specific embodiment R̂a is C-|.g alkyl optionally substituted by halogen or aryl.
  • In a particular embodiment, when R̂ is —CONH2 and R̂ is C-|.g alkyl, the carbon atom to which R-I and R̂ are attached is preferably in the “S”-configuration.
  • A further aspect of the present invention consists in novel compounds having the formula (I-B3), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00093
  • wherein
    R1 is either hydrogen or C-μg alkyl, preferably hydrogen, methyl or ethyl; more preferably R1 is ethyl.
    R3 is —CONH2, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; preferably R̂ is —CONH2R̂a is C-|5 alkyl optionally substituted by halogen or C-1.4 alkoxy; an aryl; or C2_g alkenyl optionally substituted by halogen.
  • Preferably, R̂a is C-|.g alkyl optionally substituted by halogen or C2_g alkenyl optionally substituted by halogen.
  • In a particular embodiment, when R̂ is —CONH2 and R̂ is C-|.g alkyl, the carbon atom to which R-I and R̂ are attached is preferably in the “S”-configuration.
  • A further aspect of the present invention consists in novel compounds having the formula (I-C), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.
  • Figure US20140206667A1-20140724-C00094
  • wherein
    R1 is hydrogen or C-|.g alkyl, in particular hydrogen, methyl or ethyl.
    R3 is —CONH2, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; in particular R̂ is —CONH2
    R̂a is methyl, ethyl, butyl optionally substituted by halogen or C-1.4 alkoxy, an unsubstituted phenyl or a phenyl substituted by halogen, a C-|.g alkyl optionally substituted by halogen or a C-1.4 alkoxy; or R̂a is a C2-6 alkenyl optionally substituted by halogen. Preferably, R̂a is methyl, optionally substituted by halogen, an unsubstituted phenyl or a phenyl substituted by halogen.
  • In a particular embodiment, when R̂ is —CONH2 and R̂ is C-|.g alkyl, the carbon atom to which R1 and Rβ are attached is preferably in the “S”-configuration.
  • A further aspect of the present invention consists in compounds having the formula (I-D1 or I-D2), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00095
  • wherein
    R-I is hydrogen or C-|.g alkyl, in particular hydrogen; R3 is an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl. In one embodiment, R̂ is 1H-imidazol-1-yl, 1H-imidazol-4-yl. 1H-imidazol-5-yl, imidazo[1,2-a]pyridin-3-yl or imidazo[1,2-b]pyridazin-3-yl. In a more specific embodiment, R̂ is 1H-imidazol-1-yl. 1H-imidazol-4-yl. 1H-imidazol-5-yl, imidazo[1,2-a]pyridin-3-yl; R̂a is hydrogen, C-|.g alkyl optionally substituted by halogen or C-1.4 alkoxy; aryl; or C2-g alkenyl optionally substituted by halogen. In a specific embodiment, R̂a is C-|.g alkyl optionally substituted by halogen; aryl; or C2-6 alkenyl optionally substituted by halogen. In a more specific embodiment R̂a is C-|.g alkyl optionally substituted by halogen; or aryl; e.g., propyl or phenyl;
    with the proviso that when R̂ and R̂a are hydrogen, R̂ is not 2-phenylimidazo[1,2-a]pyridin-3-yl.
  • A further aspect of the present invention consists in compounds having the formula (I-F1, I-F2 or I-F3), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00096
  • wherein
    R-I is hydrogen or C-|.g alkyl, preferably hydrogen, methyl or ethyl; more preferably, R̂ is hydrogen.
    R3 is —CONH2, an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl; in a more specific embodiment R3 is —CONH2, 1H-imidazol-1-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, imidazo[1,2-a]pyridin-3-yl or imidazo[1,2-b]pyridazin-3-yl. R̂b is hydrogen; halogen; nitro; cyano; C1.4 alkyl optionally substituted by halogen;
    C-1.4 alkoxy optionally substituted by halogen. In a more specific embodiment R̂ is hydrogen, halogen or cyano, more specifically halogen.
  • In a particular embodiment, when R̂ is —CONH2 and R̂ is C-|.g alkyl, the carbon atom to which R1 and Rβ are attached is preferably in the “S”-configuration.
  • A further aspect of the present invention consists in compounds having the formula (I-F4), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00097
  • wherein
    R-I is hydrogen or C-|.g alkyl, preferably hydrogen;
    R3 is an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl; more specifically R̂ is 1H-imidazol-1-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, imidazo[1,2-a]pyridin-3-yl or imidazo[1,2-b]pyridazin-3-yl. More specifically R̂ is 1H-imidazol-4-yl or imidazo[1,2-a]pyridin-3-yl.
    R̂b is hydrogen; halogen; nitro; cyano; C-1.4 alkyl optionally substituted by halogen; C-1.4 alkoxy optionally substituted by halogen; specifically R̂ is hydrogen, halogen or cyano.
  • In a particular embodiment, when R̂ is —CONH2 and R̂ is C-|.g alkyl, the carbon atom to which R-I and R̂ are attached is preferably in the “S”-configuration.
  • A further aspect of the present invention consists in compounds having either of the formula (I-G1, I-G2 or I-G3), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,
  • Figure US20140206667A1-20140724-C00098
  • wherein
    R-I is hydrogen or C-|.g alkyl; preferably hydrogen;
    R3 is —CONH2, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; in a more specific embodiment R̂ is —CONH2, 1H-imidazol-1-yl, 1H-imidazol-4-yl, 1H-imidazol-5-yl, imidazo[1,2-a]pyridin-3-yl or imidazo[1,2-b]pyridazin-3-yl. In a even more specific embodiment R3 is an 1H-imidazol-4-yl or imidazo[1,2-a]pyridin-3-yl;
    R4D is hydrogen; halogen; C-1.4 alkyl optionally substituted by halogen; C-1.4 alkoxy optionally substituted by halogen.
  • Specific compounds of the present invention are those selected from the group consisting of: (2S)-2-[3-(4-nitrophenyl)-5-oxoimidazolidin-1-yl]butanamide; (2S)-2-[3-(2,4-dinitrophenyl)-5-oxoimidazolidin-1-yl]butanamide: (2S)-2-(5-oxo-3-phenylimidazolidin-1-yl)butanamide; 2-[5-(iodomethyl)-2-oxo-1,3-oxazolidin-3-yl]butanamide; 2-(2-oxo-2,5-dihydro-1H-pyrrol-1-yl)butanamide; 2-(2-oxo-4-phenyl-2,5-dihydro-1H-pyrrol-1-yl)butanamide; 2-(4-methyl-2-oxo-2,5-dihydro-1H-pyrrol-1-yl)butanamide; (2S)-2-(2-oxo-5-propyl-1,3-thiazol-3(2H)-yl)butanamide; 2-(2-oxo-5-propyl-1,3-thiazol-3(2H)-yl)propanamide; 2-(5-butyl-2-oxo-1,3-thiazolidin-3-yl)butanamide; 2-(5-butyl-2-oxo-1,3-thiazolidin-3-yl)propanamide; 2-(2-oxo-5-phenyl-1,3-thiazolidin-3-yl)propanamide; 2-(2-oxo-5-propyl-1,3-thiazolidin-3-yl)butanamide; 2-(2-oxo-5-phenyl-1,3-thiazolidin-3-yl)butanamide; 2-(2-oxo-5-propyl-1,3-thiazolidin-3-yl)propanamide; (2S)-2-[2-oxo-5-(2,2,2-trifluoroethyl)-1,3-thiazolidin-3-yl]butanamide; 1-([6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl)piperidin-2-one; 1-(1H-imidazol-4-ylmethyl)-5-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-5-propylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-5-propylpiperidin-2-one; 141H-imidazol-1-ylmethyl)-5-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-5-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-phenylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-5-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpiperidin-2-one; 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}azepan-2-one; 1-(1H-imidazol-5-ylmethyl)-5-propylazepan-2-one; 5-propyl-1-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}azepan-2-one; 5-phenyl-1-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}azepan-2-one; 1-(1H-imidazol-5-ylmethyl)-6-propylazepan-2-one; 1-(1H-imidazol-4-ylmethyl)-4-propylazepan-2-one; 4-(1H-imidazol-4-ylmethyl)-4,6-dihydro-5H-thieno[3,2-b]pyrrol-5-one; 2-(5-oxo-5,6-dihydro-4H-thieno[3,2-b]pyrrol-4-yl)acetamide; 4-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}-4,6-dihydro-5H-thieno[3,2-b]pyrrol-5-one; 4-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}hexahydro-5H-thieno[3,2-b]pyrrol-5-one; 14(1H-imidazol-4-ylmethyl)-1H-thieno[3,4-b]pyrrol-2(3H)-one; 2-(6-chloro-2-OXO-1,3-benzothiazol-3(2H)-yl)acetamide; 6-bromo-3-(1H-imidazol-1-ylmethyl)-1,3-benzothiazol-2(3H)-one; 2-(6-bromo-2-oxo-1,3-benzothiazol-3(2H)-yl)propanamide; 2-(6-bromo-2-oxo-1,3-benzothiazol-3(2H)-yl)propanamide; 2-(6-fluoro-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide; 2-(6-methyl-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide; 6-fluoro-3-(1H-imidazol-1-ylmethyl)-1,3-benzoxazol-2(3H)-one; 1-(1H-imidazol-4-ylmethyl)pyrazolo[1,5-a]pyridin-2(1H)-one; 2-(6-chloro-3-oxo-3,4-dihydroisoquinolin-2(1H)-yl)propanamide; 5-chloro-2-(1H-imidazol-4-ylmethyl)-1,4-dihydroisoquinolin-3(2H)— one; 2-(6-chloro-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide; 2-(6-bromo-2-oxo-3,4-dihydroquinolin-I (2H)-yl)acetamide; 1-(1H-imidazol-4-ylmethyl)-3,4-dihydroquinolin-2(1H)— one; 2-(6-iodo-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide; 2-(6-cyano-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide; 7-chloro-2-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}-1,2,4,5-tetrahydro-3H-2-benzazepin-3-one; 7-chloro-2-(1H-imidazol-4-ylmethyl)-1,2,4,5-tetrahydro-3H-2-benzazepin-3-one; 7-chloro-3-(1H-imidazol-4-ylmethyl)-1,3,4,5-tetrahydro-2H-3-benzazepin-2-one; and 7-chloro-3-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}-1,3,4,5-tetrahydro-2H-3-benzazepin-2-one.
  • In some embodiments, compounds of the present invention are those selected from the group consisting of: 1-(1H-imidazol-4-ylmethyl)-5-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-5-propylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-5-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-5-phenylpiperidin-2-one: 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-phenylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-5-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-4-ylmethyl)-1H-thieno[3,4-b]pyrrol-2(3H)-one; 6-bromo-3-(1H-imidazol-1-ylmethyl)-1,3-benzothiazol-2(3H)-one; 2-(6-bromo-2-oxo-1,3-benzothiazol-3(2H)-yl)propanamide; and 5-chloro-2-(1H-imidazol-4-ylmethyl)-1,4-dihydroisoquinolin-3(2H)-one.
  • The following paragraphs provide definitions of the various chemical moieties that make up the compounds according to the invention and are intended to apply uniformly through-out the specification and embodiments unless an otherwise expressly set out definition provides a broader definition.
  • “C-|3 alkyl” refers to alkyl groups having 1 to 6, or 1 to 4 carbon atoms. This term is exemplified by groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, n-hexyl, trifluoromethyl and the like. “Aryl” refers to an unsaturated aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl). Preferred aryl include phenyl, naphthyl, phenantrenyl and the like.
  • “Heterocycle” refers to a saturated or unsaturated ring system containing, in addition to carbon atoms, at least one hetero atom, such as nitrogen, oxygen and/or sulfur. “Heterocycle” includes both “heteroaryl” and “heterocycloalkyl”.
  • “Heteroaryl” refers to a monocyclic heteroaromatic, or a bicyclic or a tricyclic fused-ring heteroaromatic group. Particular examples of heteroaromatic groups include optionally substituted pyridyl, pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrazolyl, 1,2,3-triazolyl. 1,2,4-triazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadia-zolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, 1,3,4-triazinyl, 1,2,3-triazinyl, benzofuryl, [2,3-dihydro]benzofuryl, isobenzofuryl, benzothienyl, benzotriazolyl, isobenzothienyl, indolyl, isoindolyl, 3H-indolyl, benzimidazolyl, imidazopyridinyl, benzothiazolyl, benzoxazolyl, quinolizinyl, quinazolinyl, pthalazinyl, quinoxalinyl, cinnolinyl, napthyridinyl, pyrido[3,4-b]pyridyl, pyrido[3,2-b]pyridyl, pyrido[4,3-b]pyridyl, quinolyl, isoquinolyl, tetrazolyl, 5,6,7,8-tetrahydroquinolyl. 5,6,7,8-tetrahydroisoquinolyl, purinyl, pteridinyl, carbazolyl, xanthenyl, benzoquinolyl, imidazopyrimidinyl, imidazopyridazinyl, imidazothiazolyl or imidazothiadiazolyl.
  • “C2-6 alkenyl” refers to alkenyl groups preferably having from 2 to 6 carbon atoms and having at least 1 or 2 sites of alkenyl unsaturation. Preferable alkenyl groups include ethenyl (vinyl, —CH═CH2), n-2-propenyl (allyl, —CH2CH═CH2) and the like.
  • “C2-6 alkynyl” refers to alkynyl groups preferably having from 2 to 6 carbon atoms and having at least 1-2 sites of alkynyl unsaturation, preferred alkynyl groups include ethynyl (—C—CH), propargyl (—CH2C—CH), and the like.
  • “C3.8 cycloalkyl” refers to a saturated carbocyclic group of from 3 to 8 carbon atoms having a single ring (e.g. cyclohexyl) or multiple condensed rings (e.g., norbornyl). Preferred cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl and the like.
  • “Heterocycloalkyl” refers to a C3.8 cycloalkyl group according to the definition above, in which 1 to 3 carbon atoms are replaced by hetero atoms chosen from the group consisting of O, S, NR, R being defined as hydrogen or C-|.g alkyl.
  • “Alkoxy” refers to the group —O—R where R includes “C-μg alkyl”, “C2-6 alkenyl”, “C2-6 alkynyl”, “C3.8 cycloalkyl”, “heterocycloalkyl”, “aryl”, “heteroaryl”.
  • “Amino” refers to the group —NRR′ where each R, R′ is independently hydrogen, “C-|.g alkyl”, “C2-6 alkenyl”, “C2-6 alkynyl”, “C3-8 cycloalkyl”, “heterocycloalkyl”, “aryl”, “heteroaryl”, and where R and R′, together with the nitrogen atom to which they are attached, can optionally form a 3-8-membered heterocycloalkyl ring.
  • “Amido” refers to the group —C(═O)NRR′ where each R, R′ is independently hydrogen, “C-|5 alkyl”, “C2-6 alkenyl”, “C2-6 alkynyl”, “C3.8 cycloalkyl”, “heterocycloalkyl”, “aryl”,
  • “heteroaryl”, and where R and R′, together with the nitrogen atom to which they are attached, can optionally form a 3-8-membered heterocycloalkyl ring.
  • “Acylamino” refers to the group —NRC(O)R′ wherein R and R′ are as defined hereabove for the amino group.
  • “Ureido” refers to the group —NR″C(O)NRR′ wherein R and R′ are as defined hereabove for the amino group, and R″ is as defined hereabove. “Sulfanyl” refers to the group —SR where R is “C-|.g alkyl”. “C2-6 alkenyl”, “C2-6 alkynyl”, “C3.8 cycloalkyl”, “heterocycloalkyl”, “aryl” or “heteroaryl”.
  • “Sulfinyl” refers to the group —S(═O)R where R is “C-|.g alkyl”, “C2-6 alkenyl”, “C2-6 alkynyl”, “C3.8 cycloalkyl”, “heterocycloalkyl”, “aryl” or “heteroaryl”.
  • “Sulfonyl” refers to the group —S(═O)2R where R is “C-|.g alkyl”, “C2-6 alkenyl”, “C2-6 alkynyl”, “C3.8 cycloalkyl”, “heterocycloalkyl”, “aryl” or “heteroaryl”.
  • “Halogen” refers to fluoro, chloro, bromo and iodo atoms.
  • “Substituted or unsubstituted”: Unless otherwise constrained by the definition of the individual substituent, the above set out groups, like “alkyl”, “alkenyl”, “alkynyl”, “aryl” and “heteroaryl” etc. groups can optionally be substituted with from 1 to 5 substituents selected from the group consisting of “C-|.g alkyl”, “C2-6 alkenyl”, “C2-6 alkynyl”.
  • “cycloalkyl”, “heterocycloalkyl”, “amino”, “amido”, “acylamino”, “ureido”, “aryl”, “heteroaryl”, “alkoxy”, “halogen”, cyano, hydroxy, mercapto, nitro, “amido”, “sulfanyl”, “sulfinyl”, “sulfonyl” and the like.
  • The acid addition salt form of a compound of formula (I) that occurs in its free form as a base can be obtained by treating the free base with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, trifluoroacetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like.
  • The compounds of formula (I) containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt forms, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.
  • Compounds of the formula (I) and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like.
  • Many of the compounds of formula (I) and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem., 45 (1976) 11-30.
  • The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula (I) or mixtures thereof (including all possible mixtures of stereoisomers). With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof, unless the particular isomeric form is referred to specifically.
  • Compounds according to the present invention may exist in different polymorphic forms. Although not explicitly indicated in the above formula, such forms are intended to be included within the scope of the present invention.
  • Some of the compounds of formula (I) may also exist in tautomeric forms. Such forms although not explicitly indicated in the above formula are intended to be included within the scope of the present invention.
  • The invention also includes within its scope pro-drug forms of the compounds of formula (I) and its various sub-scopes and sub-groups.
  • In a specific embodiment, the present invention concerns a compound selected from the group consisting of: (2S)-2-[3-(4-nitrophenyl)-5-oxoimidazolidin-1-yl]butanamide; (2S)-2-[3-(2,4-dinitrophenyl)-5-oxoimidazolidin-1-yl]butanamide; (2S)-2-(5-oxo-3-phenylimidazolidin-1-yl)butanamide: 2-[5-(iodomethyl)-2-oxo-1,3-oxazolidin-3-yl]butanamide; 2-(2-oxo-2,5-dihydro-1H-pyrrol-1-yl)butanamide; 2-(2-oxo-4-phenyl-2,5-dihydro-1H-pyrrol-1-yl)butanamide; 2-(4-methyl-2-oxo-2,5-dihydro-1H-pyrrol-1-yl)butanamide; (+)-(2S)-2-(2-oxo-4-propyl-2,5-dihydro-1H-pyrrol-1-yl)butanamide; (2S)-2-(2-oxo-5-propyl-1,3-thiazol-3(2H)-yl)butanamide; 2-(2-oxo-5-propyl-1,3-thiazol-3(2H)-yl)propanamide; 2-(5-butyl-2-oxo-1,3-thiazolidin-3-yl)butanamide; 2-(5-butyl-2-oxo-1,3-thiazolidin-3-yl)propanamide; 2-(2-oxo-5-phenyl-1,3-thiazolidin-3-yl)propanamide; 2-(2-oxo-5-propyl-1,3-thiazolidin-3-yl)butanamide; 2-(2-oxo-5-phenyl-1,3-thiazolidin-3-yl)butanamide; 2-(2-oxo-5-propyl-1,3-thiazolidin-3-yl)propanamide; (2S)-2-[2-oxo-5-(2,2,2-trifluoroethyl)-1,3-thiazolidin-3-yl]butanamide; 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}piperidin-2-one; 1-(1H-imidazol-4-ylmethyl)-5-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-5-propylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-5-propylpiperidin-2-one; 14(1H-imidazol-1-ylmethyl)-5-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-5-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-phenylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-phenylpiperidin-2-one; 1-(imidazo[1,2-a]pyridin-3-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-5-ylmethyl)-4-propylpiperidin-2-one; 1-(1H-imidazol-1-ylmethyl)-4-propylpiperidin-2-one; 1-{[6-chloro-2-(trifluoromethyl)imidazo[1,2-b]pyridazin-3-yl]methyl}azepan-2-one; 1-(1H-imidazol-5-ylmethyl)-5-propylazepan-2-one; 5-propyl-1-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}azepan-2-one; 141H-imidazol-5-ylmethyl)-5-phenylazepan-2-one; 5-phenyl-1-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}azepan-2-one; 1-(1H-imidazol-5-ylmethyl)-6-propylazepan-2-one; 1-(1H-imidazol-4-ylmethyl)-4-propylazepan-2-one; 4-(1H-imidazol-4-ylmethyl)-4,6-dihydro-5H-thieno[3,2-b]pyrrol-5-one; 2-(5-oxo-5,6-dihydro-4H-thieno[3,2-b]pyrrol-4-yl)acetamide; 4-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl})-4,6-dihydro-5H-thieno[3,2-b]pyrrol-5-one, 4-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}hexahydro-5H-thieno[3,2-b]pyrrol-5-one; 1-(1H-imidazol-4-ylmethyl)-1H-thieno[3,4-b]pyrrol-2(3H)-one; 2-(6-bromo-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide; 2-(2-OXO-1,3-benzothiazol-3(2H)-yl)acetamide; 2-(6-chloro-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide; 6-bromo-3-(1H-imidazol-1-ylmethyl)-1,3-benzothiazol-2(3H)-one; 6-bromo-3-(2-oxopropyl)-1,3-benzothiazol-2(3H)-one; 2-(6-nitro-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide; 2-(6-bromo-2-oxo-1,3-benzothiazol-3(2H)-yl)propanamide; 2-(6-bromo-2-oxo-1,3-benzothiazol-3(2H)-yl)propanamide; 2-(6-fluoro-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide; 2-(6-methyl-2-oxo-1,3-benzothiazol-3(2H)-yl)acetamide: 6-fluoro-3-(1H-imidazol-1-ylmethyl)-1,3-benzoxazol-2(3H)-one; 1-(1H-imidazol-4-ylmethyl)pyrazolo[1,5-a]pyridin-2(1H)-one; 2-(6-chloro-3-oxo-3,4-dihydroisoquinolin-2(1H)-yl)propanamide: 5-chloro-2-(1H-imidazol-4-ylmethyl)-1,4-dihydroisoquinolin-3(2H)-one; 2-(6-chloro-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide; 2-(6-bromo-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide: 1-(1H-imidazol-4-ylmethyl)-3,4-dihydroquinolin-2(1H)-one: 2-(6-iodo-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide; 2-(6-cyano-2-oxo-3,4-dihydroquinolin-1(2H)-yl)acetamide; 7-chloro-2-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}-1,2,4,5-tetrahydro-3H-2-benzazepin-3-one: 7-chloro-2-(1H-imidazol-4-ylmethyl)-1,2,4,5-tetrahydro-3H-2-benzazepin-3-one; 7-chloro-3-(1H-imidazol-4-ylmethyl)-1,3,4,5-tetrahydro-2H-3-benzazepin-2-one; and 7-chloro-3-{[2-(trifluoromethyl)imidazo[1,2-a]pyridin-3-yl]methyl}-1,3,4,5-tetrahydro-2H-3-benzazepin-2-one.
  • xvi) UK Patent 1,039,113
  • The new compounds according to the present invention are N-substituted lactams of the general formula:
  • Figure US20140206667A1-20140724-C00099
  • wherein N is a whole number of from 3 to 5 and R represents a
  • Figure US20140206667A1-20140724-C00100
  • radical in which m is 0.1 or 2 and R′ is a hydrogen atom or an alkyl, cycloalkyl, alkenyl or alkynyl radical, which may contain 3 to 6 carbon atoms, or an aryl radical, and R″ is a hydrogen atom or an alkyl radical, or both R′ and R″, together with the nitrogen atom to which they are attached, form a heterocyclic ring, such as 5a pyrrolidine ring.
  • xvii) UK Patent 1,309,692
  • According to the present invention, there are provided new N-substituted lactams of the general formula:
  • Figure US20140206667A1-20140724-C00101
  • wherein X is a hydrogen atom or an alkyl, alkenyl or alkynyl radical containing 1 to 6 carbon atoms, p is a whole number of from 1 to 6, Y is a hydrogen atom or an alkyl, alkenyl or alkynyl radical containing 1 to 6 carbon atoms or a cycloalkyl radical and R′ and R″, which may be the same or different, are hydrogen atoms or alkyl, alkenyl, alkynyl, cycloalkyl or aryl radicals or R′ and R″, together with the nitrogen atom to which they are attached, form a heterocyclic radical which may contain further heteroatoms, with the proviso that at least one of the symbols X and Y is other than a hydrogen atom.
  • Antipsychotics
  • The antipsychotics suitable for use in the present invention may be any antipsychotic drugs or agents or pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof.
  • (1) “Typical” and “Atypical” Antipsychotics
  • Among the antipsychotics or pharmaceutically acceptable salts, hydrates, solvates, polymorphs and prodrugs thereof that are useful in the methods and compositions of this invention are atypical and typical antipsychotics.
  • In some embodiments, the antipsychotic is an atypical antipsychotic or pharmaceutically acceptable salts, hydrates, solvates, prodrugs and polymorphs thereof. Atypical antipsychotics offer several clinical benefits including, for example, superior side effect profiles, particularly with regard to extrapyramidal side effects (EPS). Atypical antipsychotics typically differ from typical antipsychotics in their “limbic-specific” dopamine type 2 (D2)-receptor binding. Atypical antipsychotics, also display a high ratio of serotonin type 2 (5-HT2)-receptor binding to D2 binding. Atypical antipsychotics have high affinity for the 5-HT2-receptor and function as antagonists of serotonin for the 5-HT2-receptor.
  • Examples of atypical antipsychotics include, but are not limited to: Aripiprazole, 7-[4-[4-(2,3-dichloropheny 1)-1-piperazinyl]butoxy]-3,4-di-hydrocarbostyril (commercially available from Bristol-Meyers Squibb Co., Princeton, N.J. under the trade name Ability®) is disclosed in U.S. Pat. Nos. 4,734,416 and 5,006,528, which are incorporated herein by reference. Exemplary formulations and dosages of aripiprazole suitable for use in treating schizophrenia and bipolar disorder are described in U.S. Pat. Nos. 6,977,257; 7,115,587; and 7,550,445, which are herein incorporated by reference in their entirety. Asenapine, trans-5-chloro-2-methyl-2,3,3a,12b-tetrahydro-1H-dibenz[2,3:6,7]oxepint[4,5-c]pyrrole (under trade name Saphris® or Sycrest®) is disclosed in U.S. Pat. Nos. 4,145,434 and 5,763,476, which are herein incorporated by reference in their entirety. An orthorhombic crystal form of asenapine is described in U.S. Pat. No. 7,741,358, which is also incorporated herein by reference. Clozapine, 8-chloro-11-(4-methyl-1-piperaziny 1)-5H-dibenzo[b,e][1,4]-diazepine (commercially available from Mylan Pharmaceuticals, Morgantown. W.Va. under the trade name Mylan®) is disclosed in U.S. Pat. No. 3,539,573, which is herein incorporated by reference. Clinical efficacy of Clozapine in the treatment of schizophrenia has previously been disclosed. Hanes, et al., Psychopharmacol. Bull., 24, 62 (1988).
  • Iloperidone, 1-[4-[3-[4-(6-Fluoro-1,2-benzisoxazol-3-yl-1-piperidinyl]propoxy]-3-methoxyphenyl]ethanone (under trade name Fanapt®) is disclosed in EP Patent EP402644, which is incorporated herein by reference. The use of iloperidone in treating psychotic symptom and exemplary dosages of iloperidone suitable for such treatment are disclosed in U.S. Patent U.S. RE39198, which is incorporated herein by reference.
  • Olanzapine, 2-methyl-4-(4-methyl-1-piperaziny I)-10H-thieno[2,3-b][1,5]benzodiazepine, disclosed in U.S. Pat. No. 5,229,382 (commercially available from Eli Lilly, Indianapolis, Ind. under the trade name Zyprexa®) which is hereby incorporated by reference, as being useful for the treatment of schizophrenia, schizophreniform disorder, acute mania, mild anxiety states, and psychosis. The use of olanzapine in treating schizophrenia and exemplary dosages of olanzapine for such use are disclosed in U.S. Pat. Nos. 5,625,897, 5,627,178, 5,817,655, 5,919,485 and 6,960,577. Olanzapine polymorphs are disclosed in U.S. Pat. No. 5,736,541, incorporated herein by reference. Olanzapine hydrate forms are disclosed in U.S. Pat. No. 6,251,895, incorporated herein by reference. Lurasidone, (3aR,4S,7R,7aS)-2-; (1R,2R)-2-[4-(1,2-benzisothiazol 3-yl)-piperazin-1-yl-methyl]cyclohexylmethyl hexahydro-4,7-methano-2H-isoindole-1,3-dione (developed by Dainippon Sumitomo Pharmna Co., Ltd. under trade name Latuda®) is disclosed in U.S. Pat. No. 5,532,372, incorporated herein by reference. Paliperidone, 3-[2-[4-(6-fluoro-1,2-benzisoxazol-3-yl)-1-piperidinyl]ethyl]-6,7,8,9-tetrahydro-9-hydroxy-2-methyl-4Hpyrido[1,2-a]pyrimidin-4-one (developed by Janssen Pharmaceutica under the trade name Invega® or Invega sustenna®), is disclosed in EP Patent 368388. The use of paliperidone in treating psychosis and exemplary formulations for such use are disclosed in U.S. Pat. Nos. 5,158,952. 5,254,556, 5,352,459, 6,077,843 and 6,555,544, all of which are incorporated herein by reference.
  • Quetiapine, 5-[2-(4-dibenzo[b,f][1,4]thiazepin-11-yl-1-piperaziny 1)-eth-oxy]ethanol (commercially available from Astra Zeneca, Wilmington, Del. under the tradename Seroquel(Y) its activity in assays which demonstrate utility in the treatment of schizophrenia are disclosed in U.S. Pat. No. 4,879,288, which is herein incorporated by reference. Exemplary formulations of quetiapine for use in treating schizophrenia and bipolar disorder are disclosed in U.S. Pat. No. 5,948,437, incorporated herein by reference.
  • Risperidone, 3-[2-[4-(6-fluoro-1,2-benzisoxazol-3-yl)piperidino]ethyl]-2-methyl-6,7,8,9-tetrahydro-4H-pyrido-[1,2-a]pyrimidin-4-one (commercially available from Janssen under the trade name Risperdal®) and its use in the treatment of psychotic diseases are disclosed in U.S. Pat. No. 4,804,663, which is herein incorporated by reference.
  • Sertindole, 1-[2-[4-[5-chloro-1-(4-fluoropheny1)-1H-indol-3-yl]-1-piperidinyl]ethyl]imidazolidin-2-one, is described in U.S. Pat. No. 4,710,500. Its use in the treatment of schizophrenia is described in U.S. Pat. Nos. 5,112,838 and 5,238.945. U.S. Pat. Nos. 4,710,500; 5,112,838; and 5,238,945 are herein incorporated by reference in their entirety.
  • Ziprasidone, 5-[2-[4-(1,2-benzoisothiazol-3-yl)-1-piperazinyl]ethyl]-6-chloro-1,3-dihydro-2H-indol-2-one, (commercially available from Pfizer Inc., New York, N.Y. under the trade name Geodoni®) is disclosed in U.S. Pat. Nos. 4,831,031 and 5,312,925 and its activity in assays which demonstrate utility in the treatment of schizophrenia are described in U.S. Pat. No. 4,831,031, all of which are herein incorporated by reference.
  • Surmontil (trimipramine maleate), 5-(3-dimethylamino-2-methylpropyl)-10,11-dihydro-5H-dibcnz(b,f) azepine acid malcate (Commercially available from Odyssey Pharmaceuticals, Inc., North Hanover, N.J. under the trade name Surmotil®).
  • In some embodiment, the antipsychotic for the methods and compositions of this invention is selected from aripiprazole, olanzapine and ziprasidone, or pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof.
  • In some embodiments of the invention, the antipsychotic is a typical antipsychotic. Such typical antipsychotics include, but are not limited to, acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupcntixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, pcrphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and pharmaceutically acceptable salts, hydrates, solvates, prodrugs and polymorphs thereof.
  • (2) Antipsychotics Displaying Various Pharmacology/Mechanisms
  • Suitable antipsychotics or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof for the present invention may be selected from compounds/agents that are dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CBI antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, nNOS inhibits, neurosteroids, and neurotrophic factors.
  • In some embodiments, the antipsychotic is a dopaminergic agent selected from dopamine D1 receptor antagonists or agonists (for example, dihydrexidine, A77636 and SKF81297), dopamine D2 receptor antagonists or partial agonists (e.g., some typical and atypical antipsychotics), dopamine D3 receptor antagonists or agonists (for example, S33084, SB-277011-A, AVE5997 and (±)-PD128907), dopamine D4 receptor antagonists (for examples, clozapine and sonepiprazole (U-101387 or PNU-101387G)).
  • In some embodiments, the antipsychotic is a glutamatergic agent selected from NMDA receptor positive allosteric modulators (e.g., glycine, D-cycloserine and D-serine), glycine reuptake inhibitors (e.g. N-(3-(4′-fluorophenyl)-3-(4′-phenylphenoxy)propyl) sarcosine and glycyldodecylamide), glutamate reuptake inhibitor (e.g., excitatory amino-acid transporters EAAT3 antagonists), metabotropic glutamate receptors agonists (e.g., LY-354740), AMPA/kainate receptor antagonists (e.g., LY-293558, GYKI52466 and LY-326325), ampakines (CX-516), and glutathione prodrugs.
  • In some embodiments, the antipsychotic is a noradrenergic agent selected from alpha-2 adrenergic receptor agonists or antagonists (e.g., guanfacine, clozapine and risperidone) and COMT inhibitors (e.g., tolcapone).
  • In some embodiments, the antipsychotic is a serotonin receptor modulator selected from 5-HT2A receptor antagonists, 5-HT1A receptor partial agonists, 5-HT2C agonists, and 5-HT6 antagonists (e.g., some atypical antipsychotics).
  • In some embodiments, the antipsychotic is a cholinergic agent selected from alpha-7 nicotinic receptor agonists (e.g., 3-2,4-dimethoxybenzylidene anabaseine (DMXB-A or GTS-21)), alpha-4-beta2 nicotinic receptor agonists (e.g. SIB-1553A), allosteric modulators of nicotinic receptors and acetylcholinesterase inhibitors, muscarinic receptor agonists and antagonists (e.g., N-desmethylclozapine, xanomeline, PTAC, and BuTAC).
  • In some embodiments, the antipsychotic is selected from cannabinoid CB1 antagonists (e.g., SR141716), neurokinin 3 antagonists (e.g., osanetant (SR-142801) and talnetant), neurotensin agonists (e.g., SR-48692), MAO B inhibitors (e.g., Selegiline (deprenyl) and rasagiline), PDE10 inhibitors (e.g., Papaverine), NNOS inhibits (e.g., methylene blue, LNOARG, L-NAME, and 7-nitroindazole), neurosteroids (e.g., dehydroepiandrosterone (DHEA) and its sulfate derivative (DHEA-S), pregnenolone (PREG) and pregnenolone sulfate (PREGS)), and neurotrophic factors (e.g., nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and neurotrophin (NT)-3/4/5)).
  • (3) Antipsychotics Useful for Treating Symptoms of Schizophrenia or Bipolar Disorder (in Particular, Mania)
  • In some embodiments, the antipsychotics or pharmaceutically acceptable salts, hydrates, solvates, polymorphs and prodrugs thereof that are useful in the methods and compositions of this invention include those that are useful in treating one or more signs or symptoms of schizophrenia or bipolar disorder (in particular, mania).
  • Schizophrenia is characterized by psychological symptoms such as perception (hallucinations), ideation, reality testing (delusions), thought processes (loose associations), feeling (flatness, inappropriate effect), behavior (catatonia, disorganization), attention, concentration, motivation (avolition, impaired intentions and planning) and judgment (see for example Diagnostic and Statistical Manual of Mental Disorders IV, American Psychiatric Association). In general, the symptoms of schizophrenia are divided into positive and negative symptoms with hallucinations and delusions being positive features, and features such as flatness, poverty of speech and impaired executive functions representing negative symptoms. Clinical rating scales such as Positive and Negative Syndrome Scale and Scale for the Assessment of Negative Symptoms provide criteria to differentiate between, and rate, positive and negative symptoms. Frequently included in the description of negative symptoms are the cognitive deficits schizophrenic and schizotypical patients suffer from. These include impairment in attention, verbal fluency, executive functions such as planning, working memory and visual and verbal learning and memory. These types of cognitive dysfunction can be measured with a variety of tests, such as Visual Search, Verbal Fluency, Wisconsin Card Sorting, Trail Making—Part B, Symbol Digit, Hopkins Verbal Learning, Digit Span, Stroop-Color-Word and Attentional Capacity. MATRICS consensus neuropsychological test battery which includes tests of working memory, speed of processing, attention, verbal learning, visual learning, reasoning and problem solving and social cognition. Moreover, it has been found that cognitive measures predict work function and overall outcome as assessed by the Global Assessment Scale and Quality of Life Scale. Several studies have now demonstrated that neuropsychological functions, reflecting several negative and cognitive symptoms of the disease, may be more impaired in male schizophrenic patients when compared to female patients. Further, there are a number of other psychiatric diseases such as schizotypical and schizoaffective disorder, other acute- and chronic psychoses and bipolar disorder which have an overlapping symptomatology with schizophrenia. Any compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs and prodrugs thereof that are useful in treating at least one of the signs or symptoms of schizophrenia or bipolar disorder (in particular, mania), including, for example, those recited above, are useful in the methods and compositions of this invention.
  • Among the antipsychotics or pharmaceutically acceptable salts, hydrates, solvates, polymorphs and prodrugs thereof that are useful in the methods and compositions of this invention are those disclosed, for example, in U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, polymorphs and prodrugs thereof.
  • In some embodiments, the antipsychotics useful in this invention include those compounds/agents disclosed, for example, in U.S. Patents or Patent Publications US20020052401A1; US20020091118A1; US20020091119A1; US20020094986A1; US200201123490A1; US20020147194A1; US20020156089A1; US20020165217A1; US20030008806A1; US20030008892A1; US20030013887A1; US20030018047A1; US20030027812A 1: US20030032579A1; US20030130303A1; US200301158208A1; US200301162766A1; US20030181458A1; US20030191176A1; US20030208081A1; US20030232841A1; US20030235631A1; US20040001895A1; US20040006135A1; US20040029876A1; US20040039022A1; US20040048869A1; US20040049039A1; US20040082597A1; US20040106603A1; US20040110817A1; US20040116443A1; US20040132713A1; US20040138232A1; US20040162293A1; US20040162294A1; US200401167200A1; US200402044114A1; US20040204415A1; US20040204445A1; US20040204453A1; US20040209887A1; US20040220184A1; US20040220274A1; US20040229874A1; US20040229911A1; US20040242587A1; US200402541193A1; US20040259859A1; US20040266781A1; US2004026681A1; US20050004106A1; US20050004137A1; US20050009927A1; US20050014764A 1: US20050014848A1; US20050026937A1; US20050026946A1; US20050032837A1; US20050038036A1; US20050043292A1; US20050070577A1; US20050080099A1; US20050080100A1; US20050101613A1; US20050107425A1; US20050113437A1; US20050143403A1; US20050171086A1; US20050171095A1; US200501182049A1; US20050203296A1; US20050209250A1; US20050215571A1; US20050227980A1; US20050227981A1; US20050234065A1; US20050245521A1; US20050245543A1; US20050250803A1; US200502561112A1; US20050256162A1; US20050282811A1; US20050282816A1; US20050288304A1; US20060014733A1; US20060019998A1; US20060025421A1; US20060047114A1; US20060052373A1; US20060058361A1; US20060069087A1; US20060084692A1; US20060094719A1; US20060148807A1; US20060166974A1; US20060166998A1; US20060173179A1; US20060183763A1; US20060217398A1; US20060229455A1; US20060270656A1; US20070015763A1; US20070027178A1; US20070049613A1; US20070054913A1; US20070093509A1; US20070155779A1; US20070185097A 1: US20070213337A1; US20070224636A1; US20080045512A1; US20080096955A1; US20080139567A1; US20080167319A1; US20080176925A1; US20080207897A1; US20080269110A1; US20080269246A1; US20080305161A1; US20080318926A1; US20090011994A1; US20090023756A1; US200900935112A1; US20090131433A1; US20090176829A1; US20090197859A1; US20090215857A1; US2009029881A1; US20100004259A1; US20100029684A1; US20100113465A1; US20100130501A1; US20100222353A1; US20100249128A1; US20100324043A1; US20110003759A1; US20110028520A1; US20110034484A1; US20110144060A1; US20110144159A 1; US20110152334A1; US20110160220A1; US20110166160A1; US20110230493A1; US20110269745A1; US20110319439A1; US20110319449A1; US20120028961A1; US20120108588A1; US20120142729A1; US20120202809A1; US20120214784A1; US20120214791A1; US20120220568A1; U.S. Pat. No. 3,953,603A; U.S. Pat. No. 4,018,895A; U.S. Pat. No. 4,337,250A; U.S. Pat. No. 4,352,807A; U.S. Pat. No. 4,427,679A; U.S. Pat. No. 4,431,649A; U.S. Pat. No. 4,495,187A; U.S. Pat. No. 4,547,501A; U.S. Pat. No. 4,593,037A; U.S. Pat. No. 4,599,339A; U.S. Pat. No. 4,605,655A; U.S. Pat. No. 4,619,930A; U.S. Pat. No. 4,656,173A; U.S. Pat. No. 4,677,104A; U.S. Pat. No. 4,757,073A; U.S. Pat. No. 4,771,053A; U.S. Pat. No. 4,784,998A; U.S. Pat. No. 4,879,391A; U.S. Pat. No. 4,883,795A; U.S. Pat. No. 4,891,375A; U.S. Pat. No. 4,931,447A; U.S. Pat. No. 4,933,343A; U.S. Pat. No. 4,956,368A; U.S. Pat. No. 4,977,178A; U.S. Pat. No. 4,981,870A; U.S. Pat. No. 5,006,525A; U.S. Pat. No. 5,011,841A; U.S. Pat. No. 5,043,341A; U.S. Pat. No. 5,051,412A; U.S. Pat. No. 5,077,295A; U.S. Pat. No. 5,157,034A; U.S. Pat. No. 5,162,339A; U.S. Pat. No. 5,232,929A; U.S. Pat. No. 5,238,959A; U.S. Pat. No. 5,242,911A; U.S. Pat. No. 5,256,664A; U.S. Pat. No. 5,276,040A; U.S. Pat. No. 5,294,619A; U.S. Pat. No. 5,312,925A; U.S. Pat. No. 5,350,747A; U.S. Pat. No. 5,364,863A; U.S. Pat. No. 5,373,003A; U.S. Pat. No. 5,385,916A; U.S. Pat. No. 5,399,565A; U.S. Pat. No. 5,422,354A; U.S. Pat. No. 5,451,586A; U.S. Pat. No. 5,498,610A; U.S. Pat. No. 5,498,614A; U.S. Pat. No. 5,498,626A; U.S. Pat. No. 5,521,220A; U.S. Pat. No. 5,527,808A; U.S. Pat. No. 5,559,129A; U.S. Pat. No. 5,563,148A; U.S. Pat. No. 5,569,662A; U.S. Pat. No. 5,576,321A; U.S. Pat. No. 5,578,612A; U.S. Pat. No. 5,594,014A; U.S. Pat. No. 5,597,826A; U.S. Pat. No. 5,604,241A; U.S. Pat. No. 5,604,252A; U.S. Pat. No. 5,627,200A; U.S. Pat. No. 5,639,752A; U.S. Pat. No. 5,658,590A; U.S. Pat. No. 5,688,804A; U.S. Pat. No. 5,696,168A; U.S. Pat. No. 5,698,568A; U.S. Pat. No. 5,703,065A; U.S. Pat. No. 5,710,168A; U.S. Pat. No. 5,716,965A; U.S. Pat. No. 5,721,255A; U.S. Pat. No. 5,731,307A; U.S. Pat. No. 5,736,541A; U.S. Pat. No. 5,741,797A; U.S. Pat. No. 5,744,480A; U.S. Pat. No. 5,747,501A; U.S. Pat. No. 5,789,423A; U.S. Pat. No. 5,817,656A; U.S. Pat. No. 5,821,248A; U.S. Pat. No. 5,837,711A; U.S. Pat. No. 5,849,739A; U.S. Pat. No. 5,854,232A; U.S. Pat. No. 5,854,239A; U.S. Pat. No. 5,854,256A; U.S. Pat. No. 5,886,008A; U.S. Pat. No. 5,889,010A; U.S. Pat. No. 5,912,256A; U.S. Pat. No. 5,939,433A; U.S. Pat. No. 5,942,524A; U.S. Pat. No. 5,958,921A; U.S. Pat. No. 5,985,322A; U.S. Pat. No. 5,994,352A; U.S. Pat. No. 6,020,335A; U.S. Pat. No. 6,043,258A; U.S. Pat. No. 6,046,193A; U.S. Pat. No. 6,046,213A; U.S. Pat. No. 6,060,479A; U.S. Pat. No. 6,083,943A; U.S. Pat. No. 6,087,392A; U.S. Pat. No. 6,110,918A; U.S. Pat. No. 6,110,919A; U.S. Pat. No. 6,117,890A; U.S. Pat. No. 6,127,373A; U.S. Pat. No. 6,143,767A; U.S. Pat. No. 6,147,072A; U.S. Pat. No. 6,150,366A; U.S. Pat. No. 6,150,388A; U.S. Pat. No. 6,166,020A; U.S. Pat. No. 6,166,064A; U.S. Pat. No. 6,172,073B1; U.S. Pat. No. 6,174,895B1; U.S. Pat. No. 6,194,454B1; U.S. Pat. No. 6,197,773B1; U.S. Pat. No. 6,235,734B1; U.S. Pat. No. 6,235,747B1; U.S. Pat. No. 6,245,765B1; U.S. Pat. No. 6,245,766B1; U.S. Pat. No. 6,284,771B1; U.S. Pat. No. 6,312,717B1: U.S. Pat. No. 6,323,208B1: U.S. Pat. No. 6,326,398B1; U.S. Pat. No. 6,329,396B1; U.S. Pat. No. 6,358,950B1; U.S. Pat. No. 6,369,074B1: U.S. Pat. No. 6,380,186B1; U.S. Pat. No. 6,380,233B1; U.S. Pat. No. 6,395,735B2; U.S. Pat. No. 6,395,784B1; U.S. Pat. No. 6,399,609B1; U.S. Pat. No. 6,410,739B1; U.S. Pat. No. 6,429,317B1; U.S. Pat. No. 6,433,009B1; U.S. Pat. No. 6,436,914B1; U.S. Pat. No. 6,436,938B1; U.S. Pat. No. 6,441,015B2; U.S. Pat. No. 6,444,665B1; U.S. Pat. No. 6,448,261B1; U.S. Pat. No. 6,462,048B2; U.S. Pat. No. 6,465,491B2; U.S. Pat. No. 6,476,051B2; U.S. Pat. No. 6,506,775B1; U.S. Pat. No. 6,515,005B2; U.S. Pat. No. 6,518,271B1; U.S. Pat. No. 6,525,048B1; U.S. Pat. No. 6,525,196B1; U.S. Pat. No. 6,545,018B2; U.S. Pat. No. 6,545,022B1; U.S. Pat. No. 6,548,493B1; U.S. Pat. No. 6,548,502B2; U.S. Pat. No. 6,552,017B1; U.S. Pat. No. 6,596,900B2; U.S. Pat. No. 6,620,830B2; U.S. Pat. No. 6,627,771B1; U.S. Pat. No. 6,630,469B2: U.S. Pat. No. 6,630,476B2: U.S. Pat. No. 6,632,831B2; U.S. Pat. No. 6,635,270B2; U.S. Pat. No. 6,638,934B2; U.S. Pat. No. 6,673,811B1; U.S. Pat. No. 6,686,361B2; U.S. Pat. No. 6,710,040B1; U.S. Pat. No. 6,710,071B2; U.S. Pat. No. 6,713,490B2; U.S. Pat. No. 6,734,185B2; U.S. Pat. No. 6,777,406B2; U.S. Pat. No. 6,777,437B2; U.S. Pat. No. 6,784,180B2; U.S. Pat. No. 6,818,648B2; U.S. Pat. No. 6,821,976B2; U.S. Pat. No. 6,835,733B2; U.S. Pat. No. 6,844,344B2; U.S. Pat. No. 6,849,619B2; U.S. Pat. No. 6,875,771B2; U.S. Pat. No. 6,888,004B2; U.S. Pat. No. 6,894,045B2; U.S. Pat. No. 6,900,210B2; U.S. Pat. No. 6,924,310B2; U.S. Pat. No. 6,936,601B2; U.S. Pat. No. 6,958,351B2; U.S. Pat. No. 6,960,577B2; U.S. Pat. No. 6,992,087B2; U.S. Pat. No. 7,015,229B2; U.S. Pat. No. 7,030,145B2; U.S. Pat. No. 7,041,672B2; U.S. Pat. No. 7,045,529B2; U.S. Pat. No. 7,045,551B2; U.S. Pat. No. 7,049,314B2; U.S. Pat. No. 7,053,122B2; U.S. Pat. No. 7,067,658B2: U.S. Pat. No. 7,087,609B2: U.S. Pat. No. 7,098,217B2; U.S. Pat. No. 7,101,881B2; U.S. Pat. No. 7,101,885B2; U.S. Pat. No. 7,105,516B2: U.S. Pat. No. 7,109,164B2; U.S. Pat. No. 7,112,585B2; U.S. Pat. No. 7,115,587B2; U.S. Pat. No. 7,115,600B2; U.S. Pat. No. 7,135,472B2; U.S. Pat. No. 7,144,881B2; U.S. Pat. No. 7,144,898B2; U.S. Pat. No. 7,157,488B2; U.S. Pat. No. 7,238,699B2; U.S. Pat. No. 7,276,526B2; U.S. Pat. No. 7,319,100B2; U.S. Pat. No. 7,345,038B2; U.S. Pat. No. 7,384,934B2; U.S. Pat. No. 7,439,236B2; U.S. Pat. No. 7,485,636B2; U.S. Pat. No. 7,553,836B2; U.S. Pat. No. 7,662,817B2; U.S. Pat. No. 7,671,072B2; U.S. Pat. No. 7,678,793B2; U.S. Pat. No. 7,709,522B2; U.S. Pat. No. 7,851,622B2; U.S. Pat. No. 7,932,249B2; U.S. Pat. No. 7,956,049B2; U.S. Pat. No. 7,973,159B2; U.S. Pat. No. 8,022,062B2; U.S. Pat. No. 8,124,639B2; U.S. RE039679E; in EP Patents or Patent Publications EP1033364B1; EP104860B1; EP106486A2: EP106487A2: EP1070058B1; EP1082960A2; EP1088819B1; EP1099446B1; EP104420B1; EP11113015B1; EP1114817B1; EP1140929B1; EP1140931B1; EP1157001B1; EP177792A2; EP177798A2; EP1186318A2; EP11189904B1; EP1189905B1; EP1192165B1; EP1192952A2; EP1199068B1; EP1209157A1; EP1211247A1; EP1213031A2; EP1224930A1; EP1230921A1; EP1238676A1; EP1242411B1; EP1250336B1; EP1254662A2; EP1254668A2; EP1257526B1; EP1260221A2; EP1268396B1; EP1268404B1; EP1272484B1; EP1280781B1; EP1284257A2; EP1292568B1; EP1294677B1; EP1297833B1; EP1344779B1; EP1345942B1; EP1347760B1; EP1368094B1; EP1379239B1, EP1380298A2; EP1399445B1; EP1458368B1; EP1468686A2: EP1492794B1; EP1499606B1; EP1542668B1; EP1546134B1; EP1556378B1; EP1641454B1; EP1641455B1; EP1666886A2; EP16897211B1; EP171550B1, EP1727794B1; EP1805165B1; EP1824852B1: EP1838716B1; EP1846410B1; EP1899296B1; EP1908764A1; EP1924560B1; EP2044029B1; EP2094684B1; EP2124933B1; EP2231630B1; EP2252581B1; EP2280961B1; EP2298776A1; EP24791168A1; EP25603A1; EP25985A1; EP279598A2; EP281309A1; EP307172A2; EP318933A2; EP3291168A2; EP397364B1; EP397365A1; EP409435B1; EP436334A2; EP497314A1; EP532527B1; EP533487A1; EP537993A 1: EP545421A1; EP589924B1; EP591333B1; EP592438B1; EP594636B1; EP607164B1; EP613458B31; EP635015B1; EP641328B1; EP668863B I; EP687268B1; EP689536B I; EP7087711B1; EP716649B1; EP722941A2, EP737194B1; EP738513A1; EP747353A2; EP756869A2; EP773023A1; EP806423A1; EP810220B31; EP8211955B1; EP830864B1; EP868892A1; EP874625B1; EP884310A1; EP884316A1; EP891332B1; EP894085B1; EP901374B1; EP901789A1; EP904273B1; EP909561A2; EP915880B1; EP918772B1; EP929528B1; EP9311547B1; EP937077B1; EP9521154A2; EP958824A2; EP964849B1; EP965343A2; EP966967A2; and in PCT Patent Publications WO0016777A1; WO0041684A1; WO0071107A2; WO0074784A1; WO0146177A1; WO01146179A1; WO0146181A1; WO01146186A1; WO0146187A1; WO0160784A1; WO0168592A1; WO0172692A1; WO0177100A2; WO0185145A2; WO0189530A2; WO0192526A1; WO0194293A2; WO0203684A2; WO02058704A1; WO02059124A2; WO02059127A2; WO02059129A2; WO020721101A1; WO02072202A1; WO02079152A1; WO02092090A1; WO0219998A2; WO02461167A1; WO0247685A2; WO03000646A1; WO03006015A1; WO03009851A1; WO03010161A1; WO03022820A1; WO03032974A2; WO03043637A1; WO03049724A1; WO03082877A1; WO030846110A1; WO03093499A2; WO03105815A1; WO03105902A1; WO2004000355A1; WO2004014895A1; WO2004016583A1; WO2004016593A 1: WO2004017897A2; WO2004031189A1; WO2004039367A1; WO2004085439A1; WO2004096773A1; WO2004100954A1; WO2004100956A1; WO2004100957A1; WO20041100992A2; WO2005002578A1; WO2005013961A1; WO2005019180A1; WO2005023265A1; WO2005035523A1; WO2005040110A1; WO2005051488A1; WO2005051919A1; WO2005060949A2; WO2005060963A1; WO2005061491A2; WO2005063296A2; WO20050661126A1; WO2005067973A2; WO20050709116A1; WO2005079807A1; WO2005080361A1; WO2005082370A1; WO2005090300A1; WO2005092318A1; WO2005102272A2; WO2006016278A1; WO2006019886A2: WO2006019940A2; WO2006027691A2; WO2006044176A1; WO2006044454A1; WO2006048727A1; WO2006061711A1; WO2006073886A1; WO2006086464A2; WO2006088716A1; WO2006090272A1; WO2006090273A2; WO2006103559A1; WO2006106416A1; WO2006116401A1; WO2006136924A1; WO20070117750A1; WO20070262119A2; WO2007028082A1; WO2007028083A2; WO2007028131A1; WO2007028132A2; WO20070311828A2; WO2007050723A1; WO2007057742A2; WO2007063385A2; WO2007069053A1; WO2007085954A2; WO2007088450A2; WO2007088462A1; WO2007096743A1; WO2007099423A1; WO2007105053A2; WO2007129183A2; WO2007138431A2; WO2008001182A1; WO2008010073A1; WO2008015516A1; WO2008020302A2; WO2008020306A2; WO2008026046A1; WO2008032164A2; WO2008065500A2; WO2008070306A2; WO2008096260A1; WO2008125945A2; WO20081134480A1; WO2009009501A2; WO20090711988A1; WO2009094260A1; WO2009098576A1; WO2009127944A1; WO2009131814A2: WO2009149258A2; WO2010009062A1; WO2010014280A1; WO2010049841A1; WO2010058318A1; WO2010104818A1; WO2010104830A1; WO2010111080A2; WO2010146488A1; WO2011060035A1; WO2012004698A1; WO2012038850A1; WO2012056402A2; WO2012073143A1; WO2012073146A1; WO2012114222A1; WO8803024A1; WO9002552A1; WO9005525A 1; WO9006303A1; WO9007926A1; WO9100863A1; WO9109844A1; WO9206079A1; WO9306101A1; WO9320073A1; WO9403445A1; WO9410171A1; WO9413676A1; WO9413677A1; WO9534563A1; WO9603400A1; WO9606081A1; WO9610570A 1: WO9624353A1; WO9703066A1; WO9703665A1; WO9723220A1; WO9725983A1; WO9733577A1; WO9735584A1; WO9736867A1; WO9742190A1; WO9742191A1; WO9814433A1; WO9818798A1; WO9845268A1; WO9845287A1; WO9846225A1; WO9846226A1; WO9904778A1; WO9939725A1; WO9952889A1; WO9952907A1; WO9959593A1; WO9961441A1, and pharmaceutically acceptable salts, hydrates, solvates polymorphs or prodrugs thereof.
  • Method of Treating Schizophrenia or Bipolar Disorder (in Particular, Mania) with the Administration of a SV2a Inhibitor and an Antipsychotic or Pharmaceutically Acceptable Salts Thereof
  • In one aspect, the invention provides methods for treating a subject suffering from schizophrenia or bipolar disorder (in particular, mania), or at risk thereof, by administering a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in combination with an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. In some embodiments, the methods of this invention treat one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia In some embodiments, the methods of this invention treat one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania).
  • The SV2A inhibitor and the antipsychotic suitable for the method of this invention may be selected from any of those as described above. In some embodiments, the SV2A inhibitor is selected from any of those described above; and the antipsychotic is selected from (1) atypical and typical antipsychotics (such as those described above); (2) agents that are dopaminergic agents (such as dopamine D1 receptor antagonists or agonists, dopamine D2 receptor antagonists or partial agonists, dopamine D3 receptor antagonists or partial agonists, dopamine D4 receptor antagonists), glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA, kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents (such as alpha-2 adrenergic receptor agonists or antagonists and COMT inhibitors), serotonin receptor modulators (such as 5-HT2A receptor antagonists, 5-HT1A receptor partial agonists, 5-HT2C agonists, and 5-HT6 antagonists), cholinergic agents (such as alpha-7 nicotinic receptor agonists, alpha-4-beta2 nicotinic receptor agonists, allosteric modulators of nicotinic receptors and acetylcholinesterase inhibitors, muscarinic receptor agonists and antagonists), cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, nNOS inhibits, neurosteroids, and neurotrophic factors, including, e.g., those specific such agents as described above, and (3) any compounds that are useful in treating one or more sign or symptoms of schizophrenia or bipolar disorder (in particular, mania) (including, e.g., the agents disclosed in any of the above-listed patents or patent application publications), and pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof. In some embodiments, the SV2A inhibitor is selected from the group consisting of levetiracetam, seletracetam, and brivaracetam or derivatives or analogs or pharmaceutically acceptable salts, or solvates, or hydrates, or polymorphs, or prodrugs thereof; and the antipsychotic is an atyptical antipsychotic selected from, e.g., aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof. In some embodiments, the SV2A inhibitor is selected from levetiracetam or derivatives or analogs or pharmaceutically acceptable salts, or solvates, or hydrates, or polymorphs, or prodrugs thereof; and the antipsychotic is an atyptical antipsychotic selected from, e.g., aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof.
  • In some embodiments, the subject that suffers schizophrenia or bipolar disorder (in particular, mania) is a human patient. The subject may be a human or other mammal such as a non-human primate, or rodent (e.g., rat). In some embodiments, the subject is a human patient.
  • In some embodiments, the use of the SV2A inhibitors and pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof in combination with antipsychotics and their pharmaceutically acceptable salts, hydrates, solvates and polymorphs may reduce the amount of antipsychotics necessary for the treatment of schizophrenia or bipolar disorder (in particular, mania). In some embodiments, the subject that suffers schizophrenia or bipolar disorder (in particular, mania) is a human patient, and thus the use of the SV2A inhibitors reduce the side effects caused by antipsychotics without diminishing efficacy. Further, in some embodiments, the efficacy of a combination of the SV2A inhibitors and antipsychotics and pharmaceutically acceptable salts, solvates, hydrates, and polymorphs thereof exceeds the efficacy of either drug administered alone at its optimal dose and thus, is an improved treatment for schizophrenia or bipolar disorder (in particular, mania).
  • It will be appreciated that compounds and agents used in the compositions and methods of this invention preferably should readily penetrate the blood-brain barrier when peripherally administered. Compounds which cannot penetrate the blood-brain barrier, however, can still be effectively administered directly into the central nervous system, e.g., by an intraventricular or other neuro-compatible routes.
  • As used herein, administration of SV2A inhibitor and an antipsychotic or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof “in combination” includes simultaneous administration and/or administration at different times, such as sequential administration. Simultaneous administration of the SV2A inhibitor and the antipsychotic or their pharmaceutically acceptable salts, hydrates, solvates and polymorphs can optionally be combined with supplemental doses of the SV2A inhibitor and/or the antipsychotic and their salts, hydrates, solvates and polymorphs. Simultaneous administration of drugs encompasses administration as co-formulation or, alternatively, as separate compositions.
  • In accordance with this invention, the SV2A inhibitor and the psychotic, and pharmaceutically acceptable salts, solvates, hydrates, polymorphs thereof, can be administered to a subject via any suitable route or routes. In some embodiments, the drugs are administered orally; however, administration intravenously, subcutaneously, intra-arterially, intramuscularly, intraspinally, rectally, intrathoracically, intraperitoneally, intracentricularly, or transdermally, topically, or by inhalation is also contemplated. The agents can be administered orally, for example, in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, or the like, prepared by art recognized procedures. In certain embodiments, the SV2A inhibitor and the antipsychotic, and pharmaceutically acceptable salts, solvates, hydrates, polymorphs thereof, can be administered to a subject via different routes. For example, the SV2A inhibitor or its salt, solvate, hydrate, or polymorph is administered intravenously and the antipsychotic or its salt, solvate, hydrate, or polymorph is administered orally.
  • In some embodiments, the administration is a slow or extended release. The term “extended release” is widely recognized in the art of pharmaceutical sciences and is used herein to refer to a controlled release of an active compound or agent from a dosage form to an environment over (throughout or during) an extended period of time, e.g. greater than or equal to one hour. An extended release dosage form will release drug at substantially constant rate over an extended period of time or a substantially constant amount of drug will be released incrementally over an extended period of time. The term “extended release” used herein includes the terms “controlled release,” “prolonged release,” “sustained release.” “delayed release,” or “slow release” as these terms are used in the pharmaceutical sciences. In some embodiments, the extended release dosage is administered in the form of a patch or a pump.
  • When a solid carrier is used for administration, the preparation may be in a tablet, placed in a hard gelatin capsule in powder or pellet form, or it may be in the form of a troche or lozenge. If a liquid carrier is used, the preparation may be in the forms of a syrup, emulsion, soft gelatin capsule, or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution.
  • Dosage schedules of the agents and compositions according to the methods of the invention will vary according to the particular compound or compositions selected, the route of administration, the nature of the condition being treated, the age, and condition of the patient, the course, or stage of treatment, and will ultimately be at the discretion of the attending physician. It will be understood that the amount of the SV2A inhibitor and the antipsychotic and their pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof administered will be amounts effective to produce a desired biological effect, such as beneficial results, including clinical results. It will be understood that an effective amount can be administered in more than one dose and over a course of treatment.
  • Desired duration of administration of the SV2A inhibitor and the antipsychotic and their pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof can be determined by routine experimentation by one skilled in the art. For example, the SV2A inhibitor and the antipsychotic and their pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof may be administered for a period of 1-4 weeks, 1-3 months, 3-6 months, 6-12 months, 1-2 years, or more, up to the lifetime of the patient.
  • It is known in the art that normalization to body surface area is an appropriate method for extrapolating doses between species. The human equivalent dose (HED) for this dosage can be estimated using the following formula that accounts for differences in body surface area (see Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, December 2002, Center for Biologics Evaluation and Research):

  • HED=animal dose×(Km animal/Km human)
  • where the Km factor is body weight divided by body surface area (Km rat has been determined as 6, and Km human is 37; see Reagan-Saw, Nihal, Ahmad, 2007). Thus, a dosage of 10 mg/kg in rats is equivalent to 1.6 mg/kg in humans (10 mg/kg X (6/37)=1.6 mg/kg). For human subjects, to calculate a dose in mg from the dose in mg/kg, the dose in mg/kg is multiplied by a typical adult weight of 70 kg.
  • In certain embodiments of the invention, the dose of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is 0.1 to 5 mg/kg/day (which, given a typical human subject of 70 kg, is 7 to 350 mg/day).
  • In certain embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate and polymorph thereof can be administered at doses according to, for example, U.S. patent application Ser. No. 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, and U.S. Patent Application 61/175,536 and U.S. Patent Application 61/441,251. In certain embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate and polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.001 mg/kg to 5 mg/kg. In some embodiments, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate and polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 5 mg/kg, or about 1 to 2 mg/kg, or about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.01 to 1 mg/kg, or about 0.001 to 1 mg/kg, or about 0.5 to 5 mg/kg, or about 0.05 to 0.5 mg/kg. In certain embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.001 to 5 mg/kg, about 0.001 to 0.5 mg/kg, about 0.01 to 0.5 mg/kg, about 0.1 to 5 mg/kg, or about 1 to 2 mg/kg, or about 2 to 4 mg/kg, or about 2 to 3 mg/kg, or about 3 to 4 mg/kg, or about 0.2 to 0.4 mg/kg, or about 0.2 to 0.3 mg/kg, or about 0.3 to 0.4 mg/kg, or about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg-kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.5 to 2 mg/kg, or about 0.8 to 1.6, or about 0.8 to 3.6, or about 0.5 to 4 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.05 to 3 mg/kg or about 0.08 to about 1.6 mg/kg, or about 0.08 to 3.6 or about 0.05 to 2 mg/kg, or about 0.01 to 1 mg/kg, or about 0.001 to 1 mg/kg, or about 0.5 to 5 mg/kg, or about 0.05 to 0.5 mg/kg, or about 0.8 mg/kg, or about 1.6 mg/kg, or about 3.6 mg/kg, or about 0.08 mg/kg, or about 0.16 mg/kg, or about 0.36 mg/kg. Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art following the methods of this invention. For repeated administrations over several days or weeks or longer, depending on the condition, the treatment is sustained until a sufficient level of cognitive function is achieved.
  • In certain embodiments of the invention, the dose of the SV2A inhibitor is 0.001-5 mg/kg/day (which, given a typical human subject of 70 kg, is about 0.07-350 mg/day). Doses that may be used include, but are not limited to 0.001 mg/kg/day, 0.0015 mg/kg/day, 0.002 mg/kg/day, 0.005 mg/kg/day, 0.0075 mg/kg/day, 0.01 mg/kg/day, 0.015 mg/kg/day, 0.02 mg/kg/day, 0.03 mg/kg/day, 0.04 mg/kg/day, 0.05 mg/kg/day, 0.1 mg/kg/day, 0.2 mg/kg/day, 0.3 mg/kg/day, 0.4 mg/kg/day, 0.5 mg/kg/day, 0.75 mg/kg/day, 1.0 mg/kg/day, 1.5 mg/kg/day, 2.0 mg/kg/day, 2.5 mg/kg/day, 3.0 mg/kg/day, 4.0 mg/kg/day, or 5.0 mg/kg/day. In some embodiments, the dose of the SV2A inhibitor is 0.001-0.5 mg/kg/day (which, given a typical human subject of 70 kg, is about 0.07-35 mg/day), or 0.01-0.5 mg/kg/day (which is about 0.7-35 mg/day). Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art following the methods of this invention.
  • In certain embodiments of the invention, the dose of the SV2A inhibitor is 0.1 to 5 mg/kg/day (which, given a typical human subject of 70 kg, is 7 to 350 mg/day). Doses that may be used include, but are not limited to 0.1 mg/kg/day, 0.5 mg/kg/day, 1 mg/kg/day, 1.5 mg/kg/day, 2 mg/kg/day. 2.5 mg/kg/day. 3 mg/kg/day, 4 mg/kg/day, or 5 mg/kg/day. In certain embodiments, the dose is 1-2 mg/kg/day (which, given a typical human subject of 70 kg, is 70-140 mg/day). In other embodiments of the invention, the dose of the SV2A inhibitor is 0.1 to 0.2 mg/kg/day. Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art following the methods of this invention.
  • In certain embodiments of the invention, the dose of the SV2A inhibitor is 0.01 to 2.5 mg/kg/day (which, given a typical human subject of 70 kg, is about 0.7-180 mg/day). Doses that may be used include, but are not limited to 0.01 mg/kg/day, 0.02 mg/kg/day, 0.03 mg/kg/day, 0.04 mg/kg/day, 0.06 mg/kg/day. 0.08 mg/kg/day, 0.12 mg/kg/day, 0.14 mg/kg/day, 0.16 mg/kg/day, 0.18 mg/kg/day. 0.2 mg/kg/day, 0.4 mg/kg/day, 0.6 mg/kg/day, 0.8 mg/kg/day, 1.0 mg/kg/day, 1.2 mg/kg/day, 1.4 mg/kg/day, 1.6 mg/kg/day. 1.8 mg/kg/day. 2.0 mg/kg/day, 2.2 mg/kg/day, 2.4 mg/kg/day, or 2.5 mg/kg/day. In some embodiments, the dose of the SV2A inhibitor is 0.1-2.5 mg/kg/day (which, given a typical human subject of 70 kg, is about 7-180 mg/day), 0.1-0.2 mg/kg/day (which is about 7-15 mg/day), 0.2-0.4 mg/kg/day (about 14-30 mg/day), 0.4-2.5 mg/kg/day (about 25-180 mg/day), 0.6-1.8 mg/kg/day (about 40-130 mg/day), 0.04-2.5 mg/kg/day (about 2.5-180 mg/day) or 0.06-1.8 mg/kg/day (about 4-130 mg/day). In some embodiments of the invention, the dose of the SV2A inhibitor is 40 to 130 mg, 140 to 300 mg, 200 to 300 mg or 140 to 200 mg. Other doses higher than, intermediate to or less than these doses may also be used and may be determined by one skilled in the art following the methods of this invention.
  • In certain embodiments of the invention, the interval of administration is 12 or 24 hours. Administration at less frequent intervals, such as once every 6 hours, may also be used. In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a total daily dose of 0.1 to 5 mg/kg (e.g., in the case of administration every 12 hours of a daily dose of 2 mg/kg, each administration is 1 mg/kg). In some embodiments, the SV2A inhibitor is administered every 24 hours at a daily dose of 1 to 2 mg/kg. In another embodiment, the SV2A inhibitor is administered every 24 hours at a daily dose of 0.1-0.2 mg/kg. In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of 0.01 to 2.5 mg/kg (e.g., in the case of administration every 12 hours of a daily dose of 0.8 mg/kg, each administration is 0.4 mg/kg). In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of 0.1 to 2.5 mg/kg. In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of 0.4 to 2.5 mg/kg. In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of 0.6 to 1.8 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.04-2.5 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.06-1.8 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.001-5 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.001-0.5 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.01-0.5 mg/kg.
  • In certain embodiments of the invention, the SV2A inhibitor is levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. The levetiracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours at a daily dose of about 1 to 2 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 2.0 to 3.0 mg/kg, or about 3.0 to 4.0 mg/kg, or about 2.0 to 4.0 mg/kg, or about 0.1 to 5 mg/kg, or about 70 to 140 mg, or about 7 to 180 mg, or about 25 to 180 mg, or about 40 to 130 mg, or about 140 to 300 mg, or about 200 to 300 mg, or about 140 to 200 mg, or about 7 to 350 mg.
  • In other embodiments, the levetiracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours according to
  • TABLE 1
    Daily Doses of Levetiracetam
    Lower range
    0.1 mg/ 0.4 mg/
    Upper range kg kg 0.6 mg/kg 1 mg/kg 2 mg/kg 3 mg/kg
    1.8 mg/kg   + + + +
    2 mg/kg + + + +
    2.5 mg/kg   + + + + +
    3 mg/kg + + + + +
    4 mg/kg + + + + + +
    5 mg/kg + + + + + +
  • TABLE 2
    Daily Doses of Levetiracetam in a Human Subject of 70 KG
    Lower range
    Upper range
    7 mg 25 mg 40 mg 70 mg 140 mg 200 mg
    130 mg + + + +
    140 mg + + + +
    180 mg + + + + +
    200 mg + + + + +
    300 mg + + + + + +
    350 mg + + + + + +
  • In certain embodiments of the invention, the SV2A inhibitor is brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. The brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.2 to 0.4 mg/kg, or about 7 to 15 mg, or about 0.7 to 180 mg, or about 2.5 to 180 mg, or about 4.0 to 130 mg, or about 14 to 30 mg.
  • In other embodiments, the brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours at a daily dose of at least 0.1 mg, 0.5 mg, 0.75 mg, 1.0 mg, 1.5 mg, or 2.0 mg, but no more than a daily dose of 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg. 25 mg, 30 mg, or 35 mg. In other embodiments, the brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 mg/kg. 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/kg, but no more than a daily dose of 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.15 mg/kg, 0.1 mg/kg, 0.05 mg/kg, or 0.04 mg/kg.
  • In other embodiments, the brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours according to one of the daily dose ranges indicated as “+” listed in Table 3 or Table 4. For example, the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be administered every 12 or 24 hours at a daily dose of 0.1-35 mg, 0.5-35 mg, 0.75-35 mg, 1.0-35 mg, 1.5-35 mg, 2.0-35 mg, 0.1-30 mg, 0.1-25 mg, 0.1-20 mg, 0.1-15 mg, 0.1-10 mg, 0.1-5 mg, 0.1-2.5 mg, 0.0015-0.5 mg/kg, 0.0075-0.5 mg/kg, 0.01-0.5 mg/kg, 0.015-0.5 mg/kg, 0.02-0.5 mg/kg, 0.03-0.5 mg/kg, 0.0015-0.4 mg/kg, 0.0015-0.3 mg/kg, 0.0015-0.2 mg/kg. 0.0015-0.15 mg/kg, 0.0015-0.1 mg/kg, 0.0015-0.05 mg/kg, or 0.0015-0.04 mg/kg.
  • TABLE 3
    Daily Doses of Brivaracetam
    Lower range
    Upper range 0.0015 mg/kg 0.0075 mg/kg 0.01 mg/kg 0.015 mg/kg 0.02 mg/kg 0.03 mg/kg
    0.04 mg/kg  + + + + + +
    0.05 mg/kg  + + + + + +
    0.1 mg/kg + + + + + +
    0.15 mg/kg  + + + + + +
    0.2 mg/kg + + + + + +
    0.3 mg/kg + + + + + +
    0.4 mg/kg + + + + + +
    0.5 mg/kg + + + + + +
  • TABLE 4
    Daily Doses of Brivaracetam in a Human Subject of 70 KG
    Lower range
    Upper range 0.1 mg 0.5 mg 0.75 mg 1.0 mg 1.5 mg 2.0 mg
    2.5 mg  + + + + + +
     5 mg + + + + + +
    10 mg + + + + + +
    15 mg + + + + + +
    20 mg + + + + + +
    25 mg + + + + + +
    30 mg + + + + + +
    35 mg + + + + + +
  • In certain embodiments of the invention, the SV2A inhibitor is seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. In some embodiments, the seletracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.1 mg, 0.5 mg. 0.75 mg, 1.0 mg, 1.5 mg, or 2.0 mg, but no more than a daily dose of 2.5 mg, 5 mg, 10 mg. 15 mg, 20 mg, 25 mg, 30 mg, or 35 mg. In other embodiments, the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 mg/kg, 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/kg, but no more than a daily dose of 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.15 mg/kg. 0.1 mg/kg, 0.05 mg/kg, or 0.04 mg/kg.
  • In certain embodiments of the invention, the seletracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered according to one of the daily dose ranges indicated as “+” listed in Table 5 or Table 6. For example, the seletracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph may be administered every 12 or 24 hours at a daily dose of 0.1-35 mg, 0.5-35 mg, 0.75-35 mg, 1.0-35 mg, 1.5-35 mg, 2.0-35 mg. 0.1-30 mg. 0.1-25 mg. 0.1-20 mg, 0.1-15 mg, 0.1-10 mg. 0.1-5 mg, 0.1-2.5 mg, 0.0015-0.5 mg/kg, 0.0075-0.5 mg/kg, 0.01-0.5 mg/kg, 0.015-0.5 mg/kg, 0.02-0.5 mg/kg, 0.03-0.5 mg/kg, 0.0015-0.4 mg/kg, 0.0015-0.3 mg/kg, 0.0015-0.2 mg/kg, 0.0015-0.15 mg/kg, 0.0015-0.1 mg/kg, 0.0015-0.05 mg/kg, or 0.0015-0.04 mg/kg.
  • TABLE 5
    Daily Doses of Seletracetam
    Lower range
    Upper range 0.0015 mg/kg 0.0075 mg/kg 0.01 mg/kg 0.015 mg/kg 0.02 mg/kg 0.03 mg/kg
    0.04 mg/kg  + + + + + +
    0.05 mg/kg  + + + + + +
    0.1 mg/kg + + + + + +
    0.15 mg/kg  + + + + + +
    0.2 mg/kg + + + + + +
    0.3 mg/kg + + + + + +
    0.4 mg/kg + + + + + +
    0.5 mg/kg + + + + + +
  • TABLE 6
    Daily Doses of Seletracetam in a Human Subject of 70 KG
    Lower range
    Upper range 0.1 mg 0.5 mg 0.75 mg 1.0 mg 1.5 mg 2.0 mg
    2.5 mg  + + + + + +
     5 mg + + + + + +
    10 mg + + + + + +
    15 mg + + + + + +
    20 mg + + + + + +
    25 mg + + + + + +
    30 mg + + + + + +
    35 mg + + + + + +
  • In certain embodiments of the invention, the seletracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered according to one of the daily dose ranges indicated above for levetiracetam.
  • In some embodiments, the antipsychotic useful in the present invention is an atypical antipsychotic. Generally the amount of an atypical antipsychotic administered to a patient is an amount sufficient to have a therapeutic effect. In a preferred embodiment the amount of an atypical antipsychotic administered to a patient is an amount sufficient to treat at least one symptom or sign of schizophrenia or bipolar disorder (in particular, mania), wherein the one sign or symptom may include, but are not limited to any of those described above, including, for example, delusions, hallucinations, disorganized speech (e.g., frequent derailment or incoherence), grossly disorganized or catatonic behavior and negative symptoms (e.g., affective flattening, alogia, avolition). One skilled in the art will recognize that the amount of atypical antipsychotic will vary with many factors including the potency of the atypical antipsychotic, the age and weight of the patient, and the severity of the condition or disorder to be treated. The dosages of the drugs used in the present invention can, in the final analysis, be set by the physician in charge of the case, using knowledge of the drugs, the properties of the drugs in combination as determined in clinical trials, and the characteristics of the patient, including diseases other than that for which the physician is treating the patient.
  • Non-limiting daily dosage amounts for several atypical antipsychotics are provided herein:
  • Aripiprazole, about 0.1-150 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 1-80 mg/day, about 1-50 mg/day, or about 5-50 mg/day, and in some embodiments, up to about 30 mg/day or about 10-15 mg/day;
    Asenapine, about 0.1-150 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 1-80 mg/day, about 1-50 mg/day, or about 5-50 mg/day, and in some embodiments, about 10 mg/day;
    Clozapine, about 0.1-1000 mg/day, about 1-900 mg/day, about 5-900 mg/day, about 10-900 mg/day, about 100-900 mg/day, about 100-800 mg/day or about 100-750 mg/day, and in some embodiments, about 150-450 mg/day or about 300-450 mg/day;
    Iloperidone, about 0.1-150 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 1-80 mg/day, about 1-50 mg/day, or about 5-50 mg/day, and in some embodiments, about 12-24 mg/day;
    Olanzapine, about 0.1-150 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 1-80 mg/day, about 1-50 mg/day, or about 5-50 mg/day, and in some embodiments, about 10-15 mg/day;
    Lurasidone, about 0.1-500 mg/day, about 1-500 mg/day, about 1-250 mg/day, about 10-250 mg/day, about 10-100 mg/day, or about 20-100 mg/day, and in some embodiments, about 40-80 mg/day:
    Paliperidone, about 0.1-150 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 1-80 mg/day, about 1-50 mg/day, or about 5-50 mg/day, and in some embodiments, about 6 mg/day;
    Quetiapine, about 0.1-1000 mg/day, about 1-900 mg/day, about 1-800 mg/day, about 50-800, about 100-800, or about 200-800 mg/day, and in some embodiments, about 150-750 mg/day, about 300 mg/day or about 400-800 mg/day;
    Risperidone, about 0.1-150 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 1-80 mg/day, about 1-50 mg/day, or about 5-50 mg/day, and in some embodiments, about 4-8 mg/day or 1-6 mg/day;
    Ziprasidone, about 0.1-250 mg/day, about 1-150 mg/day, about 1-100 mg/day, about 20-100, or about 20-80 mg/day, and in some embodiments, up to about 40 mg/day, or up to about 80 mg/day or about 40-80 mg/day.
  • For repeated administrations over several days or weeks or longer, depending on the condition, the treatment is sustained until a sufficient level of cognitive function is achieved.
  • The antipsychotic or a salt, hydrate, solvate or polymorph thereof may be administered at dosage levels distinct from conventional levels (e.g., at subtherapeutic doses) when provided in combination with SV2A inhibitor, due to a SV2A inhibitor-dependent increase in the antipsychotic's therapeutic index. In some embodiments, the increase in the antipsychotic's therapeutic index due to the combination with SV2A inhibitor is greater than the therapeutic index of the antipsychotic administered in the absence of a SV2A inhibitor by at least about 1.5× or 2.0× or 2.5× or 3.0× or 3.5× or 4.0× or 4.5× or 5.0× or 5.5× or 6.0× or 6.5× or 7.0× or 7.5× or 8.0× or 8.5× or 9.0× or 9.5× or 10×, or greater than about 10×. In some embodiments, combination of an antipsychotic with the SV2A inhibitor reduces the dosage of the antipsychotic required for its therapeutic effect. In some embodiments, the antipsychotic is an atypical antipsychotic. When used in combination with SV2A inhibitor, such atypical antipsychotic is administered at a dose lower than required for its therapeutic effect when administered in the absence of SV2A inhibitor.
  • The frequency of administration of the composition of this invention may be adjusted over the course of the treatment, based on the judgment of the administering physician. It will be clear that the SV2A inhibitor and the antipsychotic and their salts, hydrates, solvates and polymorphs can be administered at different dosing frequencies or intervals. For example, SV2A inhibitor can be administered daily (including multiple doses per day) or less frequently. An antipsychotic can be administered daily (including multiple doses per day) or less frequently. In some embodiments, sustained continuous release formulations of a SV2A inhibitor and an antipsychotic may be desired. Various formulations and devices for achieving sustained release are known in the art.
  • The use of a combination of an SV2A inhibitor and an antipsychotic may reduce the amount of the antipsychotic necessary for treatment of schizophrenia or bipolar disorder (in particular, mania), and may thus reduce the side effects caused by the antipsychotics. In particular, the combination of an SV2A inhibitor with a reduced amount of antipsychotic may reduce the side effects without negatively impacting efficacy. Accordingly, in some embodiments, a subtherapeutic amount of antipsychotic is administered.
  • In some embodiments, a suitable amount of the SV2A inhibitor is administered so as to reduce the dose of the antipsychotic by at least about 20%, at least about 30%, at least about 40%, or at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or more from to the dose of the antipsychotic normally used when administered alone (i.e., individually and not in combination with other therapeutic agents or compounds). The reduction may be reflected in terms of amount administered at a given administration and/or amount administered over a given period of time (reduced frequency).
  • In certain embodiments of the invention, the combined administration of SV2A inhibitor or a salt, hydrate, solvate and polymorph thereof and an antipsychotic or a salt, hydrate, solvate and polymorph thereof can attain a longer or improved therapeutic effect in the subject than that attained by administering only the SV2A inhibitor or only the antipsychotic, by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×.
  • Compositions of this Invention
  • In one aspect, the invention provides compositions comprising a SV2A inhibitor and at least one antipsychotic and their salts, hydrates, solvates and polymorphs. In some embodiments, the SV2A inhibitor and the antipsychotic may be present in a single dosage unit (e.g., combined together in one capsule, tablet, powder, or liquid, etc.).
  • The SV2A inhibitor and the antipsychotic suitable for the compositions of this invention may be selected from any of those as described above. In some embodiments, the SV2A inhibitor is selected from any of those described above; and the antipsychotic is selected from (1) atypical and typical antipsychotics (such as those described above); (2) agents that are dopaminergic agents (such as dopamine D1 receptor antagonists or agonists, dopamine D2 receptor antagonists or partial agonists, dopamine D3 receptor antagonists or partial agonists, dopamine D4 receptor antagonists), glutamatergic agents. NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs)agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents (such as alpha-2 adrenergic receptor agonists or antagonists and COMT inhibitors), serotonin receptor modulators (such as 5-HT2A receptor antagonists, 5-HT1A receptor partial agonists, 5-HT2C agonists, and 5-HT6 antagonists), cholinergic agents (such as alpha-7 nicotinic receptor agonists, alpha-4-beta2 nicotinic receptor agonists, allosteric modulators of nicotinic receptors and acetylcholinesterase inhibitors, muscarinic receptor agonists and antagonists), cannabinoid CBI antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, nNOS inhibits, neurosteroids, and neurotrophic factors, including, e.g. those specific such agents described above, and (3) any compounds that are useful in treating one or more sign or symptoms of schizophrenia or bipolar disorder (in particular, mania) (including, e.g., the agents disclosed in any of the above-listed patents or patent application publications), and pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof. In some embodiments, the SV2A inhibitor is selected from the group consisting of levetiracetam, seletracetam, and brivaracetam or derivatives or analogs or pharmaceutically acceptable salts, or solvates, or hydrates, or polymorphs, or prodrugs thereof; and the antipsychotic is an atyptical antipsychotic selected from, e.g., aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof. In some embodiments, the SV2A inhibitor is selected from levetiracetam or derivatives or analogs or pharmaceutically acceptable salts, or solvates, or hydrates, or polymorphs, or prodrugs thereof; and the antipsychotic is an atyptical antipsychotic selected from, e.g., aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, polymorphs or prodrugs thereof.
  • The composition described herein can contain more than one SV2A inhibitor and/or more than one antipsychotic. In some embodiments, the SV2A inhibitor and the antipsychotic are in a single dosage form, in a unit dosage form, in separate dosage forms, or in separate dosage forms packaged together.
  • The compositions described herein can further contain pharmaceutically acceptable excipient(s) and may contain other agents that serve to enhance and/or complement the effectiveness of the SV2A inhibitor and/or the antipsychotic. The compositions may also contain additional agents known to be useful for treating cognitive function disorder.
  • The composition in the present invention may be in solid dosage forms such as capsules, tablets, dragrees, pills, lozenges, powders and granule. Where appropriate, they may be prepared with coatings such as enteric coatings or they may be formulated so as to provide controlled releases of one or more active ingredient such as sustained or prolonged release according to methods well known in the art. In certain embodiments, the composition is in form of a slow, controlled, or extended release. The term “extended release” is widely recognized in the art of pharmaceutical sciences and is used herein to refer to a controlled release of an active compound or agent from a dosage form to an environment over (throughout or during) an extended period of time, e.g. greater than or equal to one hour. An extended release dosage form will release drug at substantially constant rate over an extended period of time or a substantially constant amount of drug will be released incrementally over an extended period of time. The term “extended release” used herein includes the terms “controlled release”, “prolonged release”, “sustained release”, or “slow release”, as these terms are used in the pharmaceutical sciences. In some embodiments, the extended release dosage is administered in the form of a patch or a pump. The composition may also be in liquid dosage forms including solutions, emulsions, suspensions, syrups, and clixirs.
  • The compositions may be specifically formulated for administration by any suitable route as described herein and known in the art. Compositions for parental administration include sterile aqueous and nonaqueous injectable solutions, dispersions, suspensions or emulsions as well as sterile powders to be reconstituted in sterile injectable solutions or dispersions prior to use. Compositions for intraoral and oral delivery (including sublingual and buccal administration, e.g. Danckwerts et al, and oral) include but are not limited to bioadhesive polymers, tablets, patches, liquids and semisolids (see e.g., Smart et al). Compositions for respiratory delivery (pulmonary and nasal delivery) include but are not limited to a variety of pressurized metered dose inhalers, dry powder inhalers, nebulizers, aqueous mist inhalers, drops, solutions, suspensions, sprays, powders, gels, ointments, and specialized systems such as liposomes and microspheres (see e.g. Owens et al, “Alternative Routes of Insulin Delivery” and Martini et al). Compositions for transdermal delivery include but are not limited to colloids, patches, and microemulsions. Other suitable administration forms for the above and other include depot injectable formulations, suppositories, sprays, ointments, cremes, gels, inhalants, dermal patches, implants etc.
  • The compositions may also contain adjuvants, such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and gelatin.
  • Therapeutic formulations can be prepared by methods well known in the art of pharmacy, see. e.g., Goodman et al., 2001; Ansel, et al., 2004; Stoklosa et al., 2001; and Bustamante, et al., 1993.
  • In certain embodiments of the invention, a composition containing an SV2A inhibitor and an antipsychotic and their salts, hydrates, solvates and polymorphs comprises an amount of the SV2A inhibitor between 0.07 and 350 mg, or between 50 and 200 mg, or between 3 and 50 mg. In some embodiments, the amount of the SV2A inhibitor is less than 350 mg, less than 250 mg, less than 200 mg, less than 150 mg, less than 100 mg, less than 50 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0.5 mg, less than 0.1 mg, or less than 0.07 mg. In certain embodiments of the invention, a composition containing an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph comprises the SV2A inhibitor in an amount of 0.07-60 mg. 0.07-350 mg, 25-60 mg, 25-125 mg. 50-250 mg, 5-140 mg, 0.7-180 mg, 125-240 mg, 3-50 mg, or 3-60 mg. In some embodiments, a composition containing an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph comprises the SV2A inhibitor in an amount of 0.05-35 mg. In some embodiments of the composition of the present invention, the SV2A inhibitor may be selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof, said SV2A inhibitor being present in an amount selected from any of the above.
  • It will be understood by one of ordinary skill in the art that the compositions and methods described herein may be adapted and modified as is appropriate for the application being addressed and that the compositions and methods described herein may be employed in other suitable applications, and that such other additions and modifications will not depart from the scope hereof.
  • This invention will be better understood from the Experimental Details which follow. However, one skilled in the art will readily appreciate that the specific methods and results discussed are merely illustrative of the invention as described more fully in the embodiments which follow thereafter.
  • EXAMPLES Introduction and Models of Cognitive Impairment
  • A variety of conditions characterized by cognitive impairment, e.g., Age-Associated Memory Impairment (AAMI), Mild Cognitive Impairment (MCI) and Age-related Cognitive Decline (ARCD) are believed to be related to aging. Others are related to disease, for example, AD. Animal models serve as an important resource for developing and evaluating treatments for such age-related cognitive impairments. Features that characterize age-related cognitive impairment in animal models typically extend to age-related cognitive impairment in humans. Efficacy in such animal models is, thus, predictive of efficacy in humans.
  • Of available models, a Long-Evans rat model of cognitive impairment is particularly well suited for distinguishing the difference between cognitive impairment related to illness and that related to aging. Indeed, extensive behavioral characterization has identified a naturally occurring form of cognitive impairment in an outbred strain of aged Long-Evans rats (Charles River Laboratories; Gallagher et al., Behav. Neurosci. 107:618-626, (1993)). In a behavioral assessment with the Morris Water Maze (MWM), rats learn and remember the location of an escape platform guided by a configuration of spatial cues surrounding the maze. The cognitive basis of performance is tested in probe trials using measures of the animal's spatial bias in searching for the location of the escape platform. Aged rats in the study population have no difficulty swimming to a visible platform, but an age-dependent impairment is detected when the platform is camouflaged, requiring the use of spatial information. Performance for individual aged rats in the outbred Long-Evans strain varies greatly. For example, a proportion of those rats perform on a par with young adults. However, approximately 40-50% fall outside the range of young performance. This variability among aged rats reflects reliable individual differences. Thus, within the aged population some animals are cognitively impaired and designated aged-impaired (AI) and other animals are not impaired and are designated aged-unimpaired (AU). See, e.g., Colombo et al., Proc. Natl. Acad. Sci. 94: 14195-14199, (1997); Gallagher and Burwell, Neurobiol. Aging 10: 691-708, (1989); Rapp and Gallagher, Proc. Natl. Acad. Sci. 93: 9926-9930, (1996); Nicolle et al., Neuroscience 74: 741-756. (1996); and Nicolle et al., J. Neurosci. 19: 9604-9610, (1999).
  • We used the above-described rat model to identify individual AI and AU rats. We then conducted behavioral assessment on AI rats while administering various pharmacological treatments.
  • Example 1 Increased Gene Expression of SV2A in Aged-Impaired Rats Behavioral Characterization of Young, Aged-Impaired and Aged-Unimpaired Rats in Morris Water Maze (MWM)
  • Behavioral tests were performed on young (4 months old) and aged (24 months old) pathogen-free male Long-Evans rats.
  • The MWM apparatus consists of a large, circular pool (diameter 1.83 m; height, 0.58 m) filled with water (27° C.) that is made opaque through the addition of non-toxic pigment or some other substance. In the typical “hidden platform” version of the test, rats are trained to find a camouflaged white escape platform (height, 34.5 cm) that is positioned in the center of one quadrant of the maze about 1.0 cm below the water surface. This platform can be retracted to the bottom of the tank or raised to its normal position from outside the maze during behavioral testing. The location of the platform remains constant from trial to trial. Because there are no local cues that mark the position of the platform, the rat's ability to locate it efficiently from any starting position at the perimeter of the pool depends on using information surrounding the maze. The maze is surrounded by black curtains to which white patterns are affixed to provide a configuration of spatial cues. A second platform (height 37.5 cm), with its surface painted black is elevated 2 cm above the water surface during cue training to control for factors unrelated to cognition. The behavior of a rat in the pool is recorded by a camera that is suspended 2.5 m above the center of the pool. The camera is connected to a video tracking system (HVS Image Advanced Tracker VP200) and a PC computer running HVS software developed by Richard Baker of HVS Image. Hampton, UK.
  • The MWM protocol is optimized for sensitivity to the effects of aging on cognition and for measures of reliable individual differences within the aged population of out-bred Long-Evans rats (Gallagher et al. Behav. Neurosci. 107:618-626. (1993)). Rats receive three trials per day for 8 consecutive days, using a 60 sec inter-trial interval. On each training trial, the rat is released into the maze from one of four equally spaced starting positions around the perimeter of the pool. The starting position varies from trial to trial, thus preventing the use of a response strategy (e.g., always turning left from the start location to locate the escape platform). If a rat does not locate the escape platform within 90 sec on any trial, the experimenter guides the rat to the platform, where it remains for 30 sec. Every sixth trial consists of a probe trial to assess the development of spatial bias in the maze. During these trials, the rat swims with the platform retracted to the bottom of the pool for 30 sec, at which time the platform is raised to its normal position for completion of the escape trial. At the completion of the protocol using the hidden platform, rats are assessed for cue learning using the visible platform. The location of this platform varies from trial to trial in a single session of 6 training trials.
  • The proximity of the animal's position with respect to the goal is used to analyze the training trial and probe trial performance. The proximity measure is obtained by sampling the position of the animal in the maze (10 times/sec) to provide a record of distance from the escape platform in 1 sec averages. For both probe trials and training trials, a correction procedure is implemented so that trial performance is relatively unbiased by differences in distance to the goal from the various start locations at the perimeter of the pool. In making this correction, the average swimming speed is calculated for each trial (path length/latency). Then, the amount of time required to swim to the goal at that speed from the start location used for the trial is removed from the record prior to computing trial performance, i.e., cumulative distance on training trials and average distance from the goal on probe trials. Thus, scores obtained using the proximity measure are designed to reflect search error, representing deviations from an optimal search, i.e. direct path to the goal and search in the immediate vicinity of that location during probe trials.
  • Computer records of video-tracking are compiled to provide data on each rat's performance in the maze. Measures on training trials and probe trials are analyzed by Analysis of Variance (ANOVA).
  • In one set of trials, the performance during training with the hidden, camouflaged platform differs between the groups of young and aged rats [F (1, 23)=12.69, p<0.002]. In this set of trials, no difference between the groups is observed for the cue training trials with a visible platform. In this set of trials, latencies to escape during cue training averaged 9.36 seconds for young and 10.60 seconds for the aged rats.
  • An average proximity measure on interpolated probe trials is used to calculate a spatial learning index for each individual subject as described in detail in Gallagher et al., Behav. Neurosci. 107:618-26, (1993). When a rat rapidly learns to search for the platform close to its position, its spatial learning index is low. Overall, in one set of trials aged rats differed from young rats [F (1, 23)=15.18, p<0.001]. Aged rats are classified as either unimpaired or impaired relative to the learning index profile of the young study population. Aged rats that fall within the normative range of young rats (index scores <241) are designated aged-unimpaired (AU). The remaining aged subjects that have index scores outside the range of young performance are designated aged-impaired (AI).
  • Preparation of RNA from Behaviorally Characterized Rats
  • Twenty-four outbred Long-Evans rats, behaviorally characterized as is described above, are killed by live decapitation to obtain fresh brain tissue. The brain is removed, and the dentate gyrus hippocampal region is microdissected from 500 micron sections taken through the transverse axis of the entire hippocampal formation (both left and right hippocampi) of 24 characterized rats. There are 8 animals in each group (AI, AU, and Y).
  • Total RNA is isolated using Trizol reagent (Invitrogen, Carlsbad, Calif.) according to the standard protocol (homogenization in Trizol reagent followed by chloroform extraction and isopropanol precipitation). Total RNA is further purified using the RNeasy mini kit (Qiagen, Valencia, Calif.), cRNA probes are then generated from the RNA samples at the Johns Hopkins Microarray Core Facility, generally according to Affymetrix specifications.
  • Briefly, 5 μg of total RNA is used to synthesize first strand cDNA using oligonucleotide probes with 24 oligo-dT plus T7 promoter as primer (Proligo LLC, Boulder, Calif.), and the SuperScript Choice System (Invitrogen). Following the double stranded cDNA synthesis, the product is purified by phenol-chloroform extraction, and biotinilated anti-sense cRNA is generated through in vitro transcription using the BioArray RNA High Yield Transcript Labeling kit (ENZO Life Sciences Inc., Farmingdale, N.Y.). 15 μg of the biotinilated cRNA is fragmented at 94° C. for 35 min (100 mM Trix-acetate, pH 8.2, 500 mM KOAC. 150 mM MgOAC). 10 μg of total fragmented cRNA is hybridized to the RAT genome 230-2 Affymetrix GeneChip array for 16 hours at 45° C. with constant rotation (60 rpm).
  • Affymetrix Fluidics Station 450 is then used to wash and stain the chips, removing the non-hybridized target and incubating with a streptavidin-phycoerythrin conjugate to stain the biotinilated cRNA. The staining is then amplified using goat immunoglobulin-G (IgG) as blocking reagent and biotinilated anti-streptavidin antibody (goat), followed by a second staining step with a streptavidin-phycoerythrin conjugate.
  • For quality control of the total RNA from the samples, the Agilent Bioanalyzer, Lab on a Chip technology, is used to confirm that all the samples had optimal rRNA ratios (1:2, for 18S and 28S, respectively) and clean run patterns.
  • For quality control of the hybridization, chip image, and comparison between chips, the following parameters are considered: Scaling factor: related to the overall intensity of the chip, to confirm the similar signal intensity and staining through out the samples; Background: estimation of unspecific or cross-hybridization; Percentage of present calls: percentage of transcripts that are considered significantly hybridized to the chip (present) by the algorithm; Glyseraldehyde-3-phosphate dehydrogenase (GAPDH) (3′/5′): representation of the RNA integrity by measuring the ratio of 3′ to 5′ regions for the housekeeping gene GAPDH, its presence in the chip and a ratio close to 1 advocates for a good integrity of the target (sample); Spikes (BioB/BioC) to confirm the detection level and sensitivity after hybridization.
  • Data Analysis of Microarray
  • Fluorescence is detected using the Affymetrix G3000 GeneArray Scanner and image analysis of each GeneChip is done through the GeneChip Operating System 1.1.1 (GCOS) software from Affymetrix, using the standard default settings. All of the GeneChip arrays use short oligonucleotides for genes in an RNA sample.
  • For comparison between different chips, global scaling is used, scaling all probe sets to target intensity (TGT) of 150. Total number of present calls and scaling factors are similar across all chips. Further analysis for presence/absence and statistical difference is performed on a region by region basis in the following manner. Probe sets are determined to be present in a region if it had a present call in four of eight animals in a single group.
  • Probe sets are annotated using the Affymetrix annotation of Jun. 20, 2005, and all probe sets representing a specific gene are identified.
  • An ANOVA is conducted on the probe set signal values for all present probe sets by combining two groups of animals and comparing them to the third group. An “AI ANOVA” is performed, where AU group are combined with Young group and compared to AT group.
  • Pearsons's correlations comparing probe set signal values to learning indices were calculated for the aged animals (excluding young) across all present probe sets. As shown in FIG. 1, expression of genes encoding SV2A was significantly increased in aged-impaired (AI) individuals relative to young individuals (Y) and aged-unimpaired individuals (AU) in a set of experiments performed as above. These results show that increased SV2A expression was correlated to the development of age-related cognitive impairment.
  • Example 2 Effect of Levetiracetam in Aged-Impaired Rats
  • Morris Water Maze Results
  • Six Age-Impaired (AI) Long-Evans rats (as characterized above) were tested for their memory of new spatial information in the MWM, under different drug/control treatment conditions (vehicle control and two different dosage levels of levetiracetam). The MWM protocol was substantially the same as the one described in Example 1. Specifically for this study, a retention trial was performed after the training trials, as described below.
  • AI rats were given six training trials per training day with a 60-sec inter-trial interval between each training trial for two consecutive days. On each training trial, the rat was released in the maze from one of four equally spaced starting positions around the perimeter of the pool. If the rat did not locate the escape platform within 90 sec on any trial, the experimenter guided the rat to the platform, where it remained for 30 sec. 30 minutes to 1 hour prior to all the training trials on each training day, AI rats were pretreated with one of three drug conditions: 1) vehicle control (0.9% saline solution); 2) levetiracetam (5 m/kg/day); and 3) levetiracetam (10 mg/kg/day); through intraperitoneal (i.p.) injection. The same six AI rats were used for the entire trials so that each treatment condition was tested on all six rats. Therefore, to counterbalance any potential bias, both the location of the escape platform and the spatial cues surrounding the water maze were different in the three treatment conditions. Therefore, using one set of locations and spatial cues, two rats were treated with saline control solution, two with levetiracetam (5 m/kg/day) and two with levetiracetam (10 mg/kg/day). Using the second set of locations and spatial cues, the two rats treated with saline control solution in the first test were treated with either levetiracetam (5 m/kg/day) or levetiracetam (10 mg/kg/day), and the two rats previously treated with levetiracetam (5 m/kg/day) were treated with either saline control solution or levetiracetam (10 mg/kg/day), and the two rats previously treated with levetiracetam (10 mg/kg/day) were treated with either saline control solution or levetiracetam (5 m/kg/day). Using the last set of locations and spatial cues, the rat groupings were again switched so that each group was treated with a different condition than they had been treated previously.
  • After the second training day and completion of the twelve training trials (over the two days), the rat was returned to its home cage and placed in the animal housing room. After a delay of 24 hours from the last training trial, the rat was given one testing trial (the “retention trial”), which was the same MWM task as the training trials, but with the escape platform removed.
  • For the retention trial, the MWM circular pool was divided into 4 quadrants. The particular quadrant where the escape platform was placed in the training trials is referred as “target quadrant”. The particular region where the platform was located in the training trials is referred as “target annulus”. In the retention trial, the time the AI rats spent swimming in the target quadrant is measured and further plotted as a percentage of total swimming time. FIG. 2 displays the results of one such set of retention trials. The time the AI rats spend in the target annulus is also measured. FIG. 2 displays the results of one such set of retention trials. Time data are collected for all three drug treatment conditions.
  • In the retention trial, whose results are depicted in FIG. 2, the time the AI rats spent in the target quadrant was approximately 25%, which is a performance equivalent to them having no memory of the platform location. This performance did not significantly improve in the group treated with levetiractam at 5 mg/kg/day. However, the group treated with levetiractam at 10 mg/kg/day demonstrated significantly improved memory as compared to vehicle-treated controls, as indicated by a significant increase in the time spent in the target quadrant to approximately 35% of total swimming time (see FIG. 2). That level of performance is equivalent to young and age-unimpaired rats, indicating that treatment with 10 mg/kg/day levetiractam resulted in a significant recovery of the AI rats' ability to navigate this MWM. The effectiveness of the 10 mg/kg/day levetiracetam treatment was also seen in the time spent in the target annulus (see FIG. 2).
  • Radial Arm Maze Results
  • The effects of levetiracetam on the spatial memory retention of aged-impaired (AI) rats were assessed in a Radial Arm Maze (RAM) behavioral task using vehicle control and five different dosage levels of levetiracetam (1.25 mg/kg/day, 2.5 mg/kg/day, 5 mg/kg/day, 10 mg/kg/day and 20 mg/kg/day). RAM behavioral tasks were preformed on ten AI rats. All six treatment conditions were tested on all ten rats, as described above for the MWM test.
  • The RAM apparatus used consisted of eight equidistantly-spaced arms. An elevated maze arm (7 cm width×75 cm length) projected from each facet of an octagonal center platform (30 cm diameter, 51.5 cm height). Clear side walls on the arms were 10 cm high and were angled at 65° to form a trough. A food well (4 cm diameter, 2 cm deep) was located at the distal end of each arm. Froot Loops™ (Kellogg Company) were used as rewards. Blocks constructed of Plexiglas™ (30 cm height×12 cm width) could be positioned to prevent entry to any arm. Numerous extra maze cues surrounding the apparatus were also provided.
  • The AI rats were initially subjected to a pre-training test (Chappell et al. Neuropharmacology 37: 481-487, 1998). The pre-training test consisted of a habituation phase (4 days), a training phase on the standard win-shift task (18 days) and another training phase (14 days) in which a brief delay was imposed between presentation of a subset of arms designated by the experimenter (e.g., 5 arms available and 3 arms blocked) and completion of the eight-arm win-shift task (i.e. with all eight arms available).
  • In the habituation phase, rats were familiarized to the maze for an 8-minute session on four consecutive days. In each of these sessions food rewards were scattered on the RAM, initially on the center platform and arms and then progressively confined to the arms. After this habituation phase, a standard training protocol was used, in which a food pellet was located at the end of each arm. Rats received one trial each day for 18 days. Each daily trial terminated when all eight food pellets had been obtained or when either 16 choices were made or 15 minutes had elapsed. After completion of this training phase, a second training phase was carried out in which the memory demand was increased by imposing a brief delay during the trial. At the beginning of each trial, three arms of the eight-arm maze were blocked. Rats were allowed to obtain food on the five arms to which access was permitted during this initial ‘information phase’ of the trial. Rats were then removed from the maze for 60 seconds, during which time the barriers on the maze were removed, thus allowing access to all eight arms. Rats were then placed back onto the center platform and allowed to obtain the remaining food rewards during this ‘retention test’ phase of the trial. The identity and configuration of the blocked arms varied across trials.
  • The number of “errors” the AI rats made during the retention test phase was tracked. An error occurred in the trial if the rats entered an arm from which food had already been retrieved in the pre-delay component of the trial, or if it re-visited an arm in the post-delay session that had already been visited.
  • After completion of the pre-training test, rats were subjected to trials with more extended delay intervals, i.e., a one-hour delay, between the information phase (presentation with some blocked arms) and the retention test (presentation of all arms). During the delay interval, rats remained off to the side of the maze in the testing room, on carts in their individual home cages. AI rats were pretreated 30-40 minutes before daily trials with a one-time shot of the following six conditions: 1) vehicle control (0.9% saline solution); 2) levetiracetam (1.25 mg/kg/day); 3) levetiracetam (2.5 mg/kg/day); 4) levetiracetam (5 mg/kg/day); 5) levetiracetam (10 mg/kg/day); 6) levetiracetam (20 mg/kg/day); through intraperitoneal (i.p.) injection. Injections were given every other day with intervening washout days. Each AI rat was treated with all six conditions within 23 days of testing. To counterbalance any potential bias, drug effect was assessed using ascending-descending dose series, i.e., the dose series was given first in an ascending order and then repeated in a descending order. Therefore, each dose had two determinations.
  • Parametric statistics (paired t-tests) was used to compare the retention test performance of the AI rats in the one-hour delay version of the RAM task in the context of different doses of levetiracetam and vehicle control (see FIG. 3). The average numbers of errors that occurred in the trials were also significantly fewer with levetiracetam treatment of 5 mg/kg/day (average no. of errors±standard error of the mean (SEM)=0.75±0.32) and 10 mg/kg/day (average no. of errors±SEM=0.80±0.27) than using vehicle control (average no. of errors±SEM=2.00±0.42). Relative to vehicle control treatment, levetiracetam significantly improved memory performance at 5 mg/kg/day (t(9)=2.18, p=0.057) and 10 mg/kg/day (t(9)=2.37, p=0.042).
  • The radial arm maze task was also used to evaluate the effect of a combination therapy with Levetiracetam (i.p. administration) and valproate (subcutaneous administration). Levetiracetam, on its own, was effective in reducing the number of errors by AI rats in the radial arm maze at 5-10 mg/kg doses, but not at 1.25 mg/kg or 2.5 mg/kg. Valproate, on its own, was effective at 100 mg/kg but not at 25 mg/kg or 50 mg/kg. See FIG. 4. Combining the two drugs, however, had a synergistic effect. A combined administration of 50 mg/kg valproate with 2.5 mg/kg levetiracetam, neither being an effective dose when administered individually, resulted in a reduced number of errors in the radial arm maze task. This result was also obtained at an even lower dose of 1.25 mg/kg levetiracetam combined with 50 mg/kg valproate. See FIG. 5. An isobologram of levetiracetam and valproate dosages confirmed that the effect of the combined 50 mg/kg valproate and 1.25 mg/kg levetivacetam (VPA 50+LEV 1.25; empty circle) had a synergistic (super-additive) effect. The combined 50 mg/kg valproate and 2.5 mg/kg levetivacetam (VPA 50+LEV 2.5; dark circle), on the other hand, had a simple additive effect, as indicated by its placement on the line. See FIG. 6.
  • To calculate the human equivalent dose (HED) for levetiracetam dosage for treatment of age-dependent cognitive impairment in humans, we employed the formula HED (mg/kg)=rat dose (mg/kg)×0.16 (see Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, December 2002, Center for Biologics Evaluation and Research). Therefore, the dosage of 5 mg/kg/day in rats is equivalent to 0.8 mg/kg/day in humans and the dosage of 10 mg/kg/day in rats is equivalent to 1.6 mg/kg/day in humans.
  • Example 3 Effect of Levetiracetam in Human Subjects with aMCI
  • A within-subjects trial of 8 weeks duration, involving 17 amnestic MCI (aMCI) subjects and 17 age-matched controls with a low dose treatment of levetiracetam is conducted. During the course of the study, each aMCI subject receives both drug and placebo treatments separately in two periods of two weeks each, with the order of treatments among different aMCI subjects counterbalanced (see FIG. 7). Age-matched control subjects treated with placebo serve as a further control. Cognitive testing and fMRI imaging data are obtained from the subjects after each two week period of drug/placebo treatment.
  • Participants and Clinical Characterization
  • 17 right-handed aMCI patients are recruited from the Alzheimer's Disease Research Center (ADRC) at the Johns Hopkins Hospital and other referrals. An additional 17 right-handed healthy volunteers are recruited from the pool of control participants in the ADRC and other referrals. All participants are administered the Telephone Interview of Cognitive Status to determine if they are likely to pass the entry criteria of the study (including criteria for MRI scanning). All participants further undergo neurological, psychiatric, and neuropsychological examination using standardized instruments and methods. The psychiatric evaluation includes administration of the Structured Clinical Interview for DSM-IV Axis I Disorders and the Clinical Dementia Rating (CDR) scale. All aMCI patients have CDR scores of 0.5. Diagnosis of aMCI is based on the criteria proposed by Petersen et al. (e.g., “Mild cognitive impairment: Aging to Alzheimer's Disease,” Oxford University Press. N.Y. (2003), which include a memory complaint (corroborated by an informant), impaired memory function on testing (1.5 standard deviations below norm), otherwise preserved cognitive functioning (within 1 standard deviation of norm), no decline in functional ability, and no dementia. Final aMCI diagnoses are reached by clinical consensus. Exclusion criteria include major neurological or psychiatric disorders, head trauma with loss of consciousness, history of drug abuse or dependency, and general contraindications to an MRI examination (e.g. cardiac pacemaker, aneurysm coils, claustrophobia). Each aMCI subject is required to have a study partner (i.e., an informant) who can provide information about the subject's daily function and assure that medications are taken appropriately. See FIGS. 18A and 18B.
  • Study Visits:
  • The study consists of 4 visits over the course of 8 weeks (see FIG. 7). The Baseline Visit is for the purpose of performing medical, neurological, psychiatric, and neurocognitive assessments. Visits 1 and 2 are identical to the Baseline Visit but include a fMRI session. The Washout Visit, at the end of a 4 week washout period, is for the purpose of a brief clinical assessment and initiation of the second drug/placebo phase.
  • Baseline Visit:
  • At the screening visit, informed consent is obtained from the subject (and an informant in the case of MCI subjects). The subject and the informant participate in a standardized clinical interview that is used to determine the degree of the subject's functional impairment in daily life, based on the Clinical Dementia Rating (CDR) scale. The subject's medical, neurological, and psychiatric history is obtained (including a review of current medications), as well as the family history of dementia. Brief medical, neurological and psychiatric exams are conducted (including vital signs). Blood is drawn in order to perform standard laboratory tests needed to determine if the subject meets the entry criteria. The subject is re-screened for contraindications to MRI scanning, using the standard form employed at the Kirby Imaging Center. Brief cognitive testing is performed (described in section on neuropsychological assessment below). These assessments are used to determine if the subject meets the entry criteria. All of the foregoing are completed using standardized forms. If the subject meets entry criteria for the study, the subject is given the study medication (drug or placebo, randomly selected), and instructions about how it should be taken. The subject is advised about the potential for having suicidal thoughts and advised to stop taking the medication and immediately contact the study physician if this occurs.
  • Visit 1:
  • At the end of the first drug/placebo period 2 weeks after the Baseline Visit, the medical, neurological and psychiatric evaluations and cognitive testing are repeated. The subject is also clinically evaluated for suicidal ideation. Blood is drawn again to repeat the standard tests and to determine whether there are any changes related to drug treatment; the subject's blood levetiracetam level is also obtained. All medication dispensed at the Baseline Visit (drug or placebo) is collected and subject compliance with the medication regimen is assessed. The first fMRI session (with cognitive tests) is conducted on the same day, either immediately before or immediately after the clinical assessment. Subjects discontinue first period treatment at this visit.
  • Washout Visit:
  • At the end of a washout period (4 weeks) following Visit 1, the subject receives a brief medical screening, including a medical and psychiatric evaluation. Blood is drawn to obtain the blood levetiracetam level (to confirm washout). The subject is provided with new medication (drug or placebo, alternated from what was assigned in the previous treatment period) for the final phase of the study with instructions about how it should be taken.
  • Visit 2:
  • At approximately 2 weeks after the Washout Visit (i.e., 2 weeks after starting the second treatment period), the medical, neurological and psychiatric evaluations and the cognitive testing are repeated. The subject is clinically evaluated for suicidal ideation. Blood is drawn again to repeat the standard tests and to determine whether there were any changes related to drug treatment; the subject's blood levetiracetam level is also obtained. All medication dispensed at the Washout Visit is collected and subject compliance with the medication regimen is assessed. The second fMRI session (with cognitive tests) is repeated on the same day, either immediately before or immediately after the clinical assessment.
  • Neuropsychological Assessment
  • All participants undergo neuropsychological evaluation at the time of assessment for treatment efficacy (Visits 1 and 2), as well as at the Baseline Visit. The evaluation occurs outside of the scanner and includes the Buschke Selective Reminding Test (Buschke and Fuld. 1974) and the Verbal Paired Associates subtest, the Logical Memory subtest, the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997), and the Benton Visual Retention Test, as these tasks are particularly sensitive to medial temporal lobe function and early memory problems (Marquis et al., 2002 and Masur et al., 1994). Additionally, subjects are asked to complete tests of more general cognitive function such as tests to assess general mental status, executive function, attention and general naming ability. All neuropsychological tests are administered by a trained research assistant during a 60-minute session. As the three neuropsychological assessments in this study occur within a time period of 8 weeks, different versions of the neuropsychological tests are used to minimize test specific practice effects. Breaks are provided to the subject as needed.
  • Drug Administration
  • As described above, the drug treatment period is the two weeks preceding Visit 1 or 2 (with the two week period preceding the other Visit being the placebo phase). For the subjects receiving the drug treatment, half a scored 250 mg tablet of levetiracetam is used to achieve a dose of 125 mg/kg twice a day, which is approximately 3.6 mg/kg/day (assuming an average adult human weight of 70 kg).
  • All drug and placebo preparations are performed on a 1:1 allocation. The pharmacy randomize patients as they enroll, and keep a list of drug assignment.
  • Levetiracetam is rapidly and almost completely absorbed after oral administration, and its bioavailability is not affected by food. Plasma half-life of levetiracetam is approximately 7±1 hour (expected to be 9-10 hours in elderly due to decreased renal function). Absorption is rapid, with peak plasma concentrations occurring about 1 hour following oral administration. Steady state can be achieved after 2 days of multiple twice-daily dosing.
  • A typical starting dose of levetiracetam in treating epilepsy in humans is 500 mg twice a day, which is approximately 14.3 mg/kg/day. The dosage is then is increased until optimal efficacy, up to 50 mg/kg/day. Thus, the dose used in this experiment is a quarter of the lowest human dose used for treating epilepsy.
  • Even lower dosages, e.g., of 25-60 mg twice a day, are contemplated, based on the results of previous animal studies that indicated low-dose efficacy. The highest effective doses of levetiracetam used in the animal model are 5-10 mg/kg (given acutely). The human equivalent dose (HED), calculated as described above, of this dosage for treatment of age-dependent cognitive impairment in humans is equivalent to 0.8-1.6 mg/kg/day (or 28-56 mg twice a day).
  • MRI Data Acquisition
  • Imaging data are obtained through high-resolution methods developed in the Stark laboratory. Data are collected on a Phillips 3 Tesla scanner (Eindhoven, The Netherlands) equipped with an 8-channel SENSE (Sensitivity Encoding) head coil, located at the F. M. Kirby Research Center for Functional Brain Imaging at the Kennedy Krieger Institute (Baltimore, Md.). High-resolution echo-planar images are collected using an acquisition matrix of 64×64, a repetition time of 1500 milliseconds, an echo time of 30 milliseconds, a flip angle of 70 degrees, a SENSE factor of 2, and an isotropic resolution of 1.5 mm×1.5 mm×1.5 mm with no gap. Nineteen oblique slices are acquired parallel to the principal longitudinal axis of the hippocampus and covered the entire medial temporal lobe region bilaterally. In addition to the functional runs, a whole-brain MPRAGE structural scan (parameters: 150 oblique slices, 1 mm isotropic resolution) is acquired.
  • Image Analysis
  • Data analysis is carried out using the Analysis for Functional Neuroimages (AFNI, release 200807181710) software. Images are first co-registered to correct for within- and across-scan head motion. Acquisitions in which a significant motion event occur (more than 3 degrees of rotation or 2 mm of translation in any direction relative to prior acquisition), plus and minus one time repetition for 1.5 seconds, are excluded from the analyses. Structural anatomical data are registered to standard stereotaxic space (Talairach & Toumoux, 1988), and the same parameters are subsequently applied to the functional data. Behavioral vectors are produced to model different trial types.
  • The ROI-LDDMM (large deformation diffeomorphic metric mapping of the region of interest) method, a technique for cross-subject alignment, increases the power of multisubject regional fMRI studies by focusing the alignment power specifically on the ROIs (regions of interest) and not elsewhere in the brain. First, all subjects' anatomical and functional scans are normalized to the Talairach atlas using AFNI. Sub-regions of the medial temporal lobe and the hippocampus (bilateral entorhinal cortex, perirhinal cortex, parahippocampal cortex, CA3/dentate region, CA1 region, and subiculum) are segmented in three dimensions on the MPRAGE scans. The labels for the CA3 region and dentate gyrus (DG) are combined. The anatomically defined ROIs are then used to calculate the ROI-LDDMM 3D vector field transformation for each subject using a customized template based on the mean of the entire sample tested as the target. The ROI-LDDMM transformations for each individual subject's ROIs are then applied to the fit coefficient maps.
  • Group data are analyzed using a two-way Analysis of Variance (ANOVA) with trial types and group as fixed factors, and subject as a random factor nested within group. A liberal peak threshold of p<0.05, along with a spatial extent threshold of 10 voxels are used to define functional ROIs on the overall F statistic. This approach, rather than using a direct pair-wise contrast, reduces voxel selection biases because any differences amongst the various conditions allowed for a voxel to be selected. This threshold is then combined with the anatomical segmentations to only include voxels inside the regions of interest. This serves to exclude voxels that does not change with any of the model's factors, effectively limiting the analysis to voxels showing any changes with task condition or group. Voxels within each functional ROT are collapsed for further analysis.
  • Cognitive Tests During fMRI Scans at Visits 1 and 2
  • The activity of the subject's medial temporal lobe is measured by functional MRI during the subject's participation in an explicit 3-alternative forced choice task, where participants view novel, repeated and similar (“lure”) stimuli. The Psychophysics Toolbox extensions in Matlab 7.0 (The MathWorks, Natick, Mass.) is used for stimulus presentation and behavioral data collection. Stimuli are color photographs of common objects. Each participant undergoes a series of testing runs during the functional imaging sessions, each run consisting of a mix of three types of image pairs: similar pairs, identical pairs and unrelated foils. These image pairs are fully randomized throughout the run and presented individually as a series of images (see FIG. 10A). Participants are instructed to make a judgment as to whether each object seen is new, old or similar. Of critical interest are the participants' responses when presented with the second of the pair of similar objects (the “lure”; see FIG. 10B). The correct identification by the subject of lure stimuli as “similar,” provides behavioral evidence of pattern separation, i.e., the separation of similar experiences into distinct non-overlapping representations. However, an incorrect identification of lure stimuli as “old” or “new,” indicates a failure of pattern separation. Identification of lure stimuli as “old” indicates that the subject focused on the similarities between the lure stimulus and the earlier-shown partner image. Identification of the lure stimulus as “new” indicates that the subject failed to recall the earlier-shown partner image altogether. Each run also contains a number of baseline trials that use a challenging perceptual discrimination task known to provide a lower and more-stable estimate of baseline activity in the medial temporal lobe (Stark & Squire, 2001 PNAS; Law et al, 2005).
  • A survey of the activity level of various subregions in the medial temporal lobe during the cognitive test, as measured by fMRI, shows that aMCI subjects have hyperactive DG/CA3 regions and a hypoactive entorhinal cortex during the performance of memory tasks, compared to age-matched control subjects.
  • We assess the level of activity in DG/CA3 during successful memory judgments in control and aMCI subjects. The mean activity is calculated from the average activity, as measured by fMRI, during the presentation of lure stimuli correctly identified by subject as “similar” that is calibrated for baseline activity. FIG. 8A shows that aMCI patients exhibit DG/CA3 hyperactivity when making these judgments (p=0.013). FIG. 8B, however, shows that treatment with levetiracetam reduces DG/CA3 hyper-activity in aMCI subjects (p=0.037). The activity level in the aMCI subject treated with the drug, in fact, is normalized to the extent that that it is statistically indistinguishable from the activity of control subjects treated with placebo. See FIG. 8C for the mean activity values shown in FIGS. 8A and 8B.
  • The activity level during successful memory judgments in EC is significantly lower in placebo-treated aMCI subjects compared to controls (p=0.003). See FIG. 9A. However, levetiracetam treatment normalizes activity in aMCI subjects in EC as well. See FIG. 9B. Levetiracetam treatment increases EC activity during memory judgments in aMCI subjects, such that it is statistically indistinguishable from placebo-treated control subjects. See FIG. 9B. See FIG. 9C for the mean activity values shown in FIGS. 9A and 9B.
  • The normalization of DG/CA3 and EC activity during memory judgments by levetiracetam treatment is mirrored in the change seen in the aMCI subjects' performance in the cognitive task. With placebo treatment, aMCI patients perform worse that control subjects, correctly identify lure items as “similar” less often and incorrectly identifying them as “old” more often (p=0.009). See FIG. 11. However, the performance of aMCI subjects improves significantly under levetiracetam treatment. See FIG. 12. The interaction of more correct “similar” identifications with less incorrect “old” identifications under drug treatment results in a significant improvement in the performance of this memory task (p=0.039). See FIG. 13 for a table of the data represented in FIGS. 11 and 12.
  • The performance of control-placebo subjects and aMCI subjects with drug or placebo treatment is also compared in other common cognitive tests, such as the Buschke Selective Reminding Test—Delayed Recall (FIGS. 14A and 14B), the Benton Visual Retention Test (FIGS. 15A and 15B), Verbal Paired Associates Test—Recognition (FIGS. 16A and 16B) and Verbal Paired Associates Test—Delayed Recall (FIGS. 17A and 17B). In all of these tests, aMCI subjects treated with placebo perform worse than placebo-treated control subjects, and levetiracetam treatment fail to rescue performance in aMCI subjects.
  • There are a number of possible reasons why levetiracetam treatment does not help aMCI subjects with performance in these other cognitive tests. The explicit 3-alternative forced choice task done in the fMRI study is a task that is especially sensitive to DG/CA3 function. As such, the performance of the subjects in this task may be particularly attuned to the changes in DG/CA3 activity resulting from levetiracetam treatment. Further, the aMCI subjects were treated with levetiracetam for only two weeks prior to the administration of the cognitive tests. It is contemplated that a treatment duration of longer than two weeks, e.g., 16 weeks or 8 months, for the drug treatment will result in improved efficacy. Finally, comparative animal studies (see Example 1) indicate that an even lower dose would be more effective. The human dosage of 125 mg twice a day is equivalent to a rat dosage of 22.3 mg/kg/day. As is shown in Example 2 and FIG. 3, 20 mg/kg levetiracetam is too high a dose in rats, and it fails to improve the performance of AI rats in the radial maze task. The effective doses of levetiracetam used in the animal model are 5-10 mg/kg. The human equivalent dose (HED) of the optimal rat dose is 0.8-1.6 mg/kg/day. Such a dosage would result in the administration of 28-56 mg twice a day (which is substantially lower than the 125 mg twice a day used in this study). Thus, it is contemplated that aMCI subjects will exhibit a further normalization of DG/CA3 and EC activity, as well as further improved performance in cognitive tests, if they are treated with lower doses equivalent to the effective doses in rat, e.g., 25-60 mg twice a day of levetiracetam.

Claims (41)

1. A method for treating schizophrenia or bipolar disorder in a patient in need or at risk thereof, the method comprising the step of administering to said subject a therapeutically effective amount of a synaptic vesicle protein 2A (SV2A) inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, polymorph thereof and a therapeutically effective amount of an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate, polymorph thereof.
2. The method of claim 1, wherein the antipsychotic is administered at a dose that is subtherapeutic as compared to the dose at which it is therapeutically effective when administered in the absence of the SV2A inhibitor.
3. The method of claim 1, wherein the SV2A inhibitor is:
a) selected from the group of SV2A inhibitors referred to in International Patent Application PCT/US2009/005647; International Patent Application Publications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. patent application Ser. Nos. 12/580,464; 61/105,847; 61/152,631; and 61/175,536; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692; or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof; or
b) selected from the group consisting of levetiracetam, seletracetam, and brivaracetam and derivatives, analogs, pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof.
4. (canceled)
5. The method of claim 1, wherein the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of:
a) 0.001 mg/kg to 5 mg/kg;
b) 0.5 mg/kg to 5 mg/kg; or
c) 0.05 mg/kg to 0.5 mg/kg.
6. (canceled)
7. (canceled)
8. The method of claim 1, wherein the antipsychotic is selected from atypical and typical antipsychotics.
9. The method of claim 8, wherein the antipsychotic is:
a) an atypical antipsychotic;
b) selected from aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, polymorphs thereof;
c) selected from aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof;
d) a typical antipsychotic;
e) selected from acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof;
f) a compound selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, NNOS inhibits, neurosteroids, and neurotrophic factors;
g) selected from the group of compounds referred to in U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof; or
h) selected from the group of compounds referred to in U.S. Patents or Patent Publications US20020052401A1; US20020091118A1; US20020091119A1; US20020094986A1; US20020123490A1; US20020147194A1; US20020156089A1; US20020165217A1; US20030008806A1; US20030008892A1; US20030013887A1; US20030018047A1; US20030027812A1; US20030032579A1; US20030130303A1; US20030158208A1; US20030162766A1; US20030181458A1; US20030191176A1; US20030208081A1; US20030232841A1; US20030235631A1; US20040001895A1; US20040006135A1; US20040029876A1; US20040039022A1; US20040048869A1; US20040049039A1; US20040082597A1; US20040106603A1; US20040110817A1; US20040116443A1; US20040132713A1; US20040138232A1; US20040162293A1; US20040162294A1; US20040167200A1; US20040204414A1; US20040204415A1; US20040204445A1; US20040204453A1; US20040209887A1; US20040220184A1; US20040220274A1; US20040229874A1; US20040229911A1; US20040242587A1; US20040254193A1; US20040259859A1; US20040266781A1; US20040266815A1; US20050004106A1; US20050004137A1; US20050009927A1; US20050014764A1; US20050014848A1; US20050026937A1; US20050026946A1; US20050032837A1; US20050038036A1; US20050043292A1; US20050070577A1; US20050080099A1; US20050080100A1; US20050101613A1; US20050107425A1; US20050113437A1; US20050143403A1; US20050171086A1; US20050171095A1; US20050182049A1; US20050203296A1; US20050209250A1; US20050215571A1; US20050227980A1; US20050227981A1; US20050234065A1; US20050245521A1; US20050245543A1; US20050250803A1; US20050256112A1; US20050256162A1; US20050282811A1; US20050282816A1; US20050288304A1; US20060014733A1; US20060019998A1; US200600254211A1; US20060047114A1; US20060052373A1; US20060058361A1; US20060069087A1; US20060084692A1; US20060094719A1; US20060148807A1; US20060166974A1; US20060166998A1; US20060173179A1; US20060183763A1; US20060217398A1; US20060229455A1; US20060270656A1; US20070015763A1; US20070027178A1; US20070049613A1; US20070054913A1; US20070093509A1; US20070155779A1; US20070185097A1; US20070213337A1; US20070224636A1; US20080045512A1; US20080096955A1; US20080139567A1; US20080167319A1; US20080176925A1; US20080207897A1; US20080269110A1; US20080269246A1; US20080305161A1; US20080318926A1; US20090011994A1; US20090023756A1; US20090093512A1; US20090131433A1; US20090176829A1; US20090197859A1; US20090215857A1; US20090298811A1; US20100004259A1; US20100029684A1; US20100113465A1; US20100130501A1; US20100222353A1; US20100249128A1; US20100324043A1; US20110003759A1; US20110028520A1; US20110034484A1; US20110144060A1; US20110144159A1; US20110152334A1; US20110160220A1; US20110166160A1; US20110230493A1; US20110269745A1; US20110319439A1; US20110319449A1; US20120028961A1; US20120108588A1; US20120142729A1; US20120202809A1; US20120214784A1; US20120214791A1; US20120220568A1; U.S. Pat. No. 3,953,603A; U.S. Pat. No. 4,018,895A; U.S. Pat. No. 4,337,250A; U.S. Pat. No. 4,352,807A; U.S. Pat. No. 4,427,679A; U.S. Pat. No. 4,431,649A; U.S. Pat. No. 4,495,187A; U.S. Pat. No. 4,547,501A; U.S. Pat. No. 4,593,037A; U.S. Pat. No. 4,599,339A; U.S. Pat. No. 4,605,655A; U.S. Pat. No. 4,619,930A; U.S. Pat. No. 4,656,173A; U.S. Pat. No. 4,677,104A; U.S. Pat. No. 4,757,073A; U.S. Pat. No. 4,771,053A; U.S. Pat. No. 4,784,998A; U.S. Pat. No. 4,879,391A; U.S. Pat. No. 4,883,795A; U.S. Pat. No. 4,891,375A; U.S. Pat. No. 4,931,447A; U.S. Pat. No. 4,933,343A; U.S. Pat. No. 4,956,368A; U.S. Pat. No. 4,977,178A; U.S. Pat. No. 4,981,870A; U.S. Pat. No. 5,006,525A; U.S. Pat. No. 5,011,841A; U.S. Pat. No. 5,043,341A; U.S. Pat. No. 5,051,412A; U.S. Pat. No. 5,077,295A; U.S. Pat. No. 5,157,034A; U.S. Pat. No. 5,162,339A; U.S. Pat. No. 5,232,929A; U.S. Pat. No. 5,238,959A; U.S. Pat. No. 5,242,911A; U.S. Pat. No. 5,256,664A; U.S. Pat. No. 5,276,040A; U.S. Pat. No. 5,294,619A; U.S. Pat. No. 5,312,925A; U.S. Pat. No. 5,350,747A; U.S. Pat. No. 5,364,863A; U.S. Pat. No. 5,373,003A; U.S. Pat. No. 5,385,916A; U.S. Pat. No. 5,399,565A; U.S. Pat. No. 5,422,354A; U.S. Pat. No. 5,451,586A; U.S. Pat. No. 5,498,610A; U.S. Pat. No. 5,498,614A; U.S. Pat. No. 5,498,626A; U.S. Pat. No. 5,521,220A; U.S. Pat. No. 5,527,808A; U.S. Pat. No. 5,559,129A; U.S. Pat. No. 5,563,148A; U.S. Pat. No. 5,569,662A; U.S. Pat. No. 5,576,321A; U.S. Pat. No. 5,578,612A; U.S. Pat. No. 5,594,014A; U.S. Pat. No. 5,597,826A; U.S. Pat. No. 5,604,241A; U.S. Pat. No. 5,604,252A; U.S. Pat. No. 5,627,200A; U.S. Pat. No. 5,639,752A; U.S. Pat. No. 5,658,590A; U.S. Pat. No. 5,688,804A; U.S. Pat. No. 5,696,168A; U.S. Pat. No. 5,698,568A; U.S. Pat. No. 5,703,065A; U.S. Pat. No. 5,710,168A; U.S. Pat. No. 5,716,965A; U.S. Pat. No. 5,721,255A; U.S. Pat. No. 5,731,307A; U.S. Pat. No. 5,736,541A; U.S. Pat. No. 5,741,797A; U.S. Pat. No. 5,744,480A; U.S. Pat. No. 5,747,501A; U.S. Pat. No. 5,789,423A; U.S. Pat. No. 5,817,656A; U.S. Pat. No. 5,821,248A; U.S. Pat. No. 5,837,711A; U.S. Pat. No. 5,849,739A; U.S. Pat. No. 5,854,232A; U.S. Pat. No. 5,854,239A; U.S. Pat. No. 5,854,256A; U.S. Pat. No. 5,886,008A; U.S. Pat. No. 5,889,010A; U.S. Pat. No. 5,912,256A; U.S. Pat. No. 5,939,433A; U.S. Pat. No. 5,942,524A; U.S. Pat. No. 5,958,921A; U.S. Pat. No. 5,985,322A; U.S. Pat. No. 5,994,352A; U.S. Pat. No. 6,020,335A; U.S. Pat. No. 6,043,258A; U.S. Pat. No. 6,046,193A; U.S. Pat. No. 6,046,213A; U.S. Pat. No. 6,060,479A; U.S. Pat. No. 6,083,943A; U.S. Pat. No. 6,087,392A; U.S. Pat. No. 6,110,918A; U.S. Pat. No. 6,110,919A; U.S. Pat. No. 6,117,890A; U.S. Pat. No. 6,127,373A; U.S. Pat. No. 6,143,767A; U.S. Pat. No. 6,147,072A; U.S. Pat. No. 6,150,366A; U.S. Pat. No. 6,150,388A; U.S. Pat. No. 6,166,020A; U.S. Pat. No. 6,166,064A; U.S. Pat. No. 6,172,073B1; U.S. Pat. No. 6,174,895B1; U.S. Pat. No. 6,194,454B1; U.S. Pat. No. 6,197,773B1; U.S. Pat. No. 6,235,734B1; U.S. Pat. No. 6,235,747B1; U.S. Pat. No. 6,245,765B1; U.S. Pat. No. 6,245,766B1; U.S. Pat. No. 6,284,771B1; U.S. Pat. No. 6,312,717B1; U.S. Pat. No. 6,323,208B1; U.S. Pat. No. 6,326,398B1; U.S. Pat. No. 6,329,396B1; U.S. Pat. No. 6,358,950B1; U.S. Pat. No. 6,369,074B1; U.S. Pat. No. 6,380,186B1; U.S. Pat. No. 6,380,233B1; U.S. Pat. No. 6,395,735B2; U.S. Pat. No. 6,395,784B1; U.S. Pat. No. 6,399,609B1; U.S. Pat. No. 6,410,739B1; U.S. Pat. No. 6,429,317B1; U.S. Pat. No. 6,433,009B1; U.S. Pat. No. 6,436,914B1; U.S. Pat. No. 6,436,938B1; U.S. Pat. No. 6,441,015B2; U.S. Pat. No. 6,444,665B1; U.S. Pat. No. 6,448,261B1; U.S. Pat. No. 6,462,048B2; U.S. Pat. No. 6,465,491B2; U.S. Pat. No. 6,476,051B2; U.S. Pat. No. 6,506,775B1; U.S. Pat. No. 6,515,005B2; U.S. Pat. No. 6,518,271B1; U.S. Pat. No. 6,525,048B1; U.S. Pat. No. 6,525,196B1; U.S. Pat. No. 6,545,018B2; U.S. Pat. No. 6,545,022B1; U.S. Pat. No. 6,548,493B1; U.S. Pat. No. 6,548,502B2; U.S. Pat. No. 6,552,017B1; U.S. Pat. No. 6,596,900B2; U.S. Pat. No. 6,620,830B2; U.S. Pat. No. 6,627,771B1; U.S. Pat. No. 6,630,469B2; U.S. Pat. No. 6,630,476B2; U.S. Pat. No. 6,632,831B2; U.S. Pat. No. 6,635,270B2; U.S. Pat. No. 6,638,934B2; U.S. Pat. No. 6,673,811B1; U.S. Pat. No. 6,686,361B2; U.S. Pat. No. 6,710,040B1; U.S. Pat. No. 6,710,071B2; U.S. Pat. No. 6,713,490B2; U.S. Pat. No. 6,734,185B2; U.S. Pat. No. 6,777,406B2; U.S. Pat. No. 6,777,437B2; U.S. Pat. No. 6,784,180B2; U.S. Pat. No. 6,818,648B2; U.S. Pat. No. 6,821,976B2; U.S. Pat. No. 6,835,733B2; U.S. Pat. No. 6,844,344B2; U.S. Pat. No. 6,849,619B2; U.S. Pat. No. 6,875,771B2; U.S. Pat. No. 6,888,004B2; U.S. Pat. No. 6,894,045B2; U.S. Pat. No. 6,900,210B2; U.S. Pat. No. 6,924,310B2; U.S. Pat. No. 6,936,601B2; U.S. Pat. No. 6,958,351B2; U.S. Pat. No. 6,960,577B2; U.S. Pat. No. 6,992,087B2; U.S. Pat. No. 7,015,229B2; U.S. Pat. No. 7,030,145B2; U.S. Pat. No. 7,041,672B2; U.S. Pat. No. 7,045,529B2; U.S. Pat. No. 7,045,551B2; U.S. Pat. No. 7,049,314B2; U.S. Pat. No. 7,053,122B2; U.S. Pat. No. 7,067,658B2; U.S. Pat. No. 7,087,609B2; U.S. Pat. No. 7,098,217B2; U.S. Pat. No. 7,101,881B2; U.S. Pat. No. 7,101,885B2; U.S. Pat. No. 7,105,516B2; U.S. Pat. No. 7,109,164B2; U.S. Pat. No. 7,112,585B2; U.S. Pat. No. 7,115,587B2; U.S. Pat. No. 7,115,600B2; U.S. Pat. No. 7,135,472B2; U.S. Pat. No. 7,144,881B2; U.S. Pat. No. 7,144,898B2; U.S. Pat. No. 7,157,488B2; U.S. Pat. No. 7,238,699B2; U.S. Pat. No. 7,276,526B2; U.S. Pat. No. 7,319,100B2; U.S. Pat. No. 7,345,038B2; U.S. Pat. No. 7,384,934B2; U.S. Pat. No. 7,439,236B2; U.S. Pat. No. 7,485,636B2; U.S. Pat. No. 7,553,836B2; U.S. Pat. No. 7,662,817B2; U.S. Pat. No. 7,671,072B2; U.S. Pat. No. 7,678,793B2; U.S. Pat. No. 7,709,522B2; U.S. Pat. No. 7,851,622B2; U.S. Pat. No. 7,932,249B2; U.S. Pat. No. 7,956,049B2; U.S. Pat. No. 7,973,159B2; U.S. Pat. No. 8,022,062B2; U.S. Pat. No. 8,124,639B2; U.S. RE039679E; in EP Patents or Patent Publications EP1033364B1; EP104860B1; EP106486A2; EP106487A2; EP1070058B1 EP1082960A2; EP1088819B1; EP1099446B1; EP1104420B1; EP1113015B1; EP1114817B1; EP1140929B1; EP1140931B1; EP1157001B1; EP1177792A2; EP1177798A2; EP1186318A2; EP1189904B1; EP1189905B1; EP1192165B1; EP1192952A2; EP1199068B1; EP1209157A1; EP1211247A1; EP1213031A2; EP1224930A1; EP1230921A1; EP1238676A1; EP1242411B1; EP1250336B1; EP1254662A2; EP1254668A2; EP1257526B1; EP1260221A2; EP1268396B1; EP1268404B1; EP1272484B1; EP1280781B1; EP1284257A2; EP1292568B1; EP1294677B1; EP1297833B1; EP1344779B1; EP1345942B1; EP1347760B1; EP1368094B1; EP1379239B1; EP1380298A2; EP1399445B1; EP1458368B1; EP1468686A2; EP1492794B1; EP1499606B1; EP1542668B1; EP1546134B1; EP1556378B1; EP1641454B1; EP1641455B1; EP1666886A2; EP1689721B1; EP171550B1; EP1727794B1; EP1805165B1; EP1824852B1; EP1838716B1; EP1846410B1; EP1899296B1; EP1908764A1; EP1924560B1; EP2044029B1; EP2094684B1; EP2124933B1; EP2231630B1; EP2252581B1; EP2280961B1; EP2298776A1; EP2479168A1; EP25603A1; EP25985A1; EP279598A2; EP281309A1; EP307172A2; EP318933A2; EP329168A2; EP397364B1; EP397365A1; EP409435B1; EP436334A2; EP497314A1; EP532527B1; EP533487A1; EP537993A1; EP545421A1; EP589924B1; EP591333B1; EP592438B1; EP594636B1; EP607164B1; EP613458B1; EP635015B1; EP641328B1; EP668863B1; EP687268B1; EP689536B1; EP708771B1; EP716649B1; EP722941A2; EP737194B1; EP738513A1; EP747353A2; EP756869A2; EP773023A1; EP806423A1; EP810220B1; EP821955B1; EP830864B1; EP868892A1; EP874625B1; EP884310A1; EP884316A1; EP891332B1; EP894085B1; EP901374B1; EP901789A1; EP904273B1; EP909561A2; EP915880B1; EP918772B1; EP929528B1; EP931547B1; EP937077B1; EP952154A2; EP958824A2; EP964849B1; EP965343A2; EP966967A2; and in PCT Patent Publications WO0016777A1; WO0041684A1; WO0071107A2; WO0074784A1; WO146177A1; WO146179A1; WO146181A1; WO146186A1; WO146187A1; WO0160784A1; WO0168592A1; WO0172692A1; WO0177100A2; WO0185145A2; WO0189530A2; WO0192526A1; WO0194293A2; WO0203684A2; WO02058704A1; WO02059124A2; WO02059127A2; WO02059129A2; WO02072101A1; WO02072202A1; WO02079152A1; WO02092090A1; WO0219998A2; WO0246167A1; WO0247685A2; WO03000646A1; WO03006015A1; WO03009851A1; WO3010161A1; WO3022820A1; WO03032974A2; WO03043637A1; WO03049724A1; WO03082877A1; WO03084610A1; WO03093499A2; WO03105815A1; WO03105902A1; WO2004000355A1; WO2004014895A1; WO2004016583A1; WO2004016593A1; WO2004017897A2; WO2004031189A1; WO2004039367A1; WO2004085439A1; WO2004096773A1; WO2004100954A1; WO2004100956A1; WO2004100957A1; WO2004100992A2; WO2005002578A1; WO20050139611A1; WO2005019180A1; WO2005023265A1; WO2005035523A1; WO2005040110A1; WO2005051488A1; WO2005051919A1; WO2005060949A2; WO2005060963A1; WO2005061491A2; WO2005063296A2; WO2005066126A1; WO2005067973A2; WO2005070916A1; WO2005079807A1; WO2005080361A1; WO2005082370A1; WO2005090300A1; WO2005092318A1; WO2005102272A2; WO2006016278A1; WO2006019886A2; WO2006019940A2; WO2006027691A2; WO2006044176A1; WO2006044454A1; WO2006048727A1; WO2006061711A1; WO2006073886A1; WO2006086464A2; WO2006088716A1; WO2006090272A1; WO2006090273A2; WO2006103559A1; WO2006106416A1; WO2006116401A1; WO2006136924A1; WO2007017750A1; WO2007026219A2; WO2007028082A1; WO2007028083A2; WO2007028131A1; WO2007028132A2; WO2007031828A2; WO2007050723A1; WO2007057742A2; WO2007063385A2; WO2007069053A1; WO2007085954A2; WO2007088450A2; WO2007088462A1; WO2007096743A1; WO2007099423A1; WO2007105053A2; WO2007129183A2; WO2007138431A2; WO2008001182A1; WO2008010073A1; WO2008015516A1; WO2008020302A2; WO2008020306A2; WO2008026046A1; WO2008032164A2; WO2008065500A2; WO2008070306A2; WO2008096260A1; WO2008125945A2; WO2008134480A1; WO2009009501A2; WO2009071988A1; WO2009094260A1; WO2009098576A1; WO2009127944A1; WO2009131814A2; WO2009149258A2; WO2010009062A1; WO2010014280A1; WO2010049841A1; WO2010058318A1; WO2010104818A1; WO2010104830A1; WO2010111080A2; WO2010146488A1; WO2011060035A1; WO2012004698A1; WO2012038850A1; WO2012056402A2; WO2012073143A1; WO2012073146A1; WO2012114222A1; WO8803024A1; WO9002552A1; WO9005525A1; WO9006303A1; WO9007926A1; WO9100863A1; WO9109844A1; WO9206079A1; WO9306101A1; WO9320073A1; WO9403445A1; WO9410171A1; WO9413676A1; WO9413677A1; WO9534563A1; WO9603400A1; WO9606081A1; WO9610570A 1; WO9624353A1; WO9703066A1; WO9703665A1; WO9723220A1; WO9725983A1; WO9733577A1; WO9735584A1; WO9736867A1; WO9742190A1; WO9742191A1; WO9814433A1; WO9818798A1; WO9845268A1; WO9845287A1; WO9846225A1; WO9846226A1; WO9904778A1; WO9939725A1; WO9952889A1; WO9952907A1; WO9959593A1; WO9961441A1, or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof.
10-14. (canceled)
15. The method of claim 1, wherein the antipsychotic is useful in treating at least one sign or symptom of schizophrenia or bipolar disorder.
16. (canceled)
17. (canceled)
18. The method of claim 1, wherein the SV2A inhibitor and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs are administered simultaneously, or sequentially, or in a single formulation, or in separate formulations.
19-21. (canceled)
22. The method of claim 1, wherein the SV2A inhibitor is levetiracetam or a derivative, analog, pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and the antipsychotic is selected from aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
23. The method of claim 1,
a) wherein the treatment has a longer therapeutic effect in the subject than is attained by administering the antipsychotic in the absence of the SV2A inhibitor by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×; or
b) wherein the treatment has a longer therapeutic effect in the subject than is attained by administering the SV2A inhibitor in the absence of the antipsychotic by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×.
24. (canceled)
25. A method of increasing the therapeutic index of an antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in a method of treating schizophrenia or bipolar disorder in a subject in need or at risk thereof, comprising administering an SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in combination with the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof to said subject.
26. The method of claim 25, wherein the increase in the therapeutic index of the antipsychotic is greater than the therapeutic index of the antipsychotic when administered in the absence of the SV2A inhibitor by at least about 1.5×, or 2.0×, or 2.5×, or 3.0×, or 3.5×, or 4.0×, or 4.5×, or 5.0×, or 5.5×, or 6.0×, or 6.5×, or 7.0×, or 7.5×, or 8.0×, or 8.5×, or 9.0×, or 9.5×, or 10×, or greater than about 10×.
27. The method of claim 25, wherein the SV2A inhibitor is:
a) selected from the group of SV2A inhibitors referred to in International Patent Application PCT/US2009/005647; International Patent Application Publications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. patent application Ser. Nos. 12/580,464; 61/105,847; 61/152,631; and 61/175,536; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692; or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof; or
b) selected from the group consisting of levetiracetam, seletracetam, and brivaracetam, and derivatives, analogs, pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof.
28. (canceled)
29. The method of claim 25, wherein the antipsychotic is selected from atypical and typical antipsychotics.
30. The method of claim 29, wherein the antipsychotic is:
a) an atypical antipsychotic;
b) selected from aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, polymorphs thereof;
c) selected from aripiprazole, olanzapine and ziprasidone, and pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof;
d) a typical antipsyhotic;
e) selected from acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof;
f) a compound selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, NNOS inhibits, neurosteroids, and neurotrophic factors;
g) selected from the group of compounds referred to in U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof; or
h) selected from the group of compounds referred to in U.S. Patents or Patent Publications US20020052401A1; US20020091118A1; US20020091119A1; US20020094986A1; US20020123490A1; US20020147194A1; US20020156089A1; US20020165217A1; US20030008806A1; US20030008892A1; US20030013887A1; US20030018047A1; US20030027812A1; US20030032579A1; US20030130303A1; US20030158208A1; US20030162766A1; US20030181458A1; US20030191176A1; US20030208081A1; US20030232841A1; US20030235631A1; US20040001895A1; US20040006135A1; US20040029876A1; US20040039022A1; US20040048869A1; US20040049039A1; US20040082597A1; US20040106603A1; US20040110817A1; US20040116443A1; US20040132713A1; US20040138232A1; US20040162293A1; US20040162294A1; US20040167200A1; US20040204414A1; US20040204415A1; US20040204445A1; US20040204453A1; US20040209887A1; US20040220184A1; US20040220274A1; US20040229874A1; US20040229911A1; US20040242587A1; US20040254193A1; US20040259859A1; US20040266781A1; US20040266815A1; US20050004106A1; US20050004137A1; US20050009927A1; US20050014764A1; US20050014848A1; US20050026937A1; US20050026946A1; US20050032837A1; US20050038036A1; US20050043292A1; US20050070577A1; US20050080099A1; US20050080100A1; US20050101613A1; US20050107425A1; US20050113437A1; US20050143403A1; US20050171086A1; US20050171095A1; US20050182049A1; US20050203296A1; US20050209250A1; US20050215571A1; US20050227980A1; US20050227981A1; US20050234065A1; US20050245521A1; US20050245543A1; US20050250803A1; US20050256112A1; US20050256162A1; US20050282811A1; US20050282816A1; US20050288304A1; US20060014733A1; US20060019998A1; US200600254211A1; US20060047114A1; US20060052373A1; US20060058361A1; US20060069087A1; US20060084692A1; US20060094719A1; US20060148807A1; US20060166974A1; US20060166998A1; US20060173179A1; US20060183763A1; US20060217398A1; US20060229455A1; US20060270656A1; US20070015763A1; US20070027178A1; US20070049613A1; US20070054913A1; US20070093509A1; US20070155779A1; US20070185097A1; US20070213337A1; US20070224636A1; US20080045512A1; US20080096955A1; US20080139567A1; US20080167319A1; US20080176925A1; US20080207897A1; US20080269110A1; US20080269246A1; US20080305161A1; US20080318926A1; US20090011994A1; US20090023756A1; US20090093512A1; US20090131433A1; US20090176829A1; US20090197859A1; US20090215857A1; US20090298811A1; US20100004259A1; US20100029684A1; US20100113465A1; US20100130501A1; US20100222353A1; US20100249128A1; US20100324043A1; US20110003759A1; US20110028520A1; US20110034484A1; US20110144060A1; US20110144159A1; US20110152334A1; US20110160220A1; US20110166160A1; US20110230493A1; US20110269745A1; US20110319439A1; US20110319449A1; US20120028961A1; US20120108588A1; US20120142729A1; US20120202809A1; US20120214784A1; US20120214791A1; US20120220568A1; U.S. Pat. No. 3,953,603A; U.S. Pat. No. 4,018,895A; U.S. Pat. No. 4,337,250A; U.S. Pat. No. 4,352,807A; U.S. Pat. No. 4,427,679A; U.S. Pat. No. 4,431,649A; U.S. Pat. No. 4,495,187A; U.S. Pat. No. 4,547,501A; U.S. Pat. No. 4,593,037A; U.S. Pat. No. 4,599,339A; U.S. Pat. No. 4,605,655A; U.S. Pat. No. 4,619,930A; U.S. Pat. No. 4,656,173A; U.S. Pat. No. 4,677,104A; U.S. Pat. No. 4,757,073A; U.S. Pat. No. 4,771,053A; U.S. Pat. No. 4,784,998A; U.S. Pat. No. 4,879,391A; U.S. Pat. No. 4,883,795A; U.S. Pat. No. 4,891,375A; U.S. Pat. No. 4,931,447A; U.S. Pat. No. 4,933,343A; U.S. Pat. No. 4,956,368A; U.S. Pat. No. 4,977,178A; U.S. Pat. No. 4,981,870A; U.S. Pat. No. 5,006,525A; U.S. Pat. No. 5,011,841A; U.S. Pat. No. 5,043,341A; U.S. Pat. No. 5,051,412A; U.S. Pat. No. 5,077,295A; U.S. Pat. No. 5,157,034A; U.S. Pat. No. 5,162,339A; U.S. Pat. No. 5,232,929A; U.S. Pat. No. 5,238,959A; U.S. Pat. No. 5,242,911A; U.S. Pat. No. 5,256,664A; U.S. Pat. No. 5,276,040A; U.S. Pat. No. 5,294,619A; U.S. Pat. No. 5,312,925A; U.S. Pat. No. 5,350,747A; U.S. Pat. No. 5,364,863A; U.S. Pat. No. 5,373,003A; U.S. Pat. No. 5,385,916A; U.S. Pat. No. 5,399,565A; U.S. Pat. No. 5,422,354A; U.S. Pat. No. 5,451,586A; U.S. Pat. No. 5,498,610A; U.S. Pat. No. 5,498,614A; U.S. Pat. No. 5,498,626A; U.S. Pat. No. 5,521,220A; U.S. Pat. No. 5,527,808A; U.S. Pat. No. 5,559,129A; U.S. Pat. No. 5,563,148A; U.S. Pat. No. 5,569,662A; U.S. Pat. No. 5,576,321A; U.S. Pat. No. 5,578,612A; U.S. Pat. No. 5,594,014A; U.S. Pat. No. 5,597,826A; U.S. Pat. No. 5,604,241A; U.S. Pat. No. 5,604,252A; U.S. Pat. No. 5,627,200A; U.S. Pat. No. 5,639,752A; U.S. Pat. No. 5,658,590A; U.S. Pat. No. 5,688,804A; U.S. Pat. No. 5,696,168A; U.S. Pat. No. 5,698,568A; U.S. Pat. No. 5,703,065A; U.S. Pat. No. 5,710,168A; U.S. Pat. No. 5,716,965A; U.S. Pat. No. 5,721,255A; U.S. Pat. No. 5,731,307A; U.S. Pat. No. 5,736,541A; U.S. Pat. No. 5,741,797A; U.S. Pat. No. 5,744,480A; U.S. Pat. No. 5,747,501A; U.S. Pat. No. 5,789,423A; U.S. Pat. No. 5,817,656A; U.S. Pat. No. 5,821,248A; U.S. Pat. No. 5,837,711A; U.S. Pat. No. 5,849,739A; U.S. Pat. No. 5,854,232A; U.S. Pat. No. 5,854,239A; U.S. Pat. No. 5,854,256A; U.S. Pat. No. 5,886,008A; U.S. Pat. No. 5,889,010A; U.S. Pat. No. 5,912,256A; U.S. Pat. No. 5,939,433A; U.S. Pat. No. 5,942,524A; U.S. Pat. No. 5,958,921A; U.S. Pat. No. 5,985,322A; U.S. Pat. No. 5,994,352A; U.S. Pat. No. 6,020,335A; U.S. Pat. No. 6,043,258A; U.S. Pat. No. 6,046,193A; U.S. Pat. No. 6,046,213A; U.S. Pat. No. 6,060,479A; U.S. Pat. No. 6,083,943A; U.S. Pat. No. 6,087,392A; U.S. Pat. No. 6,110,918A; U.S. Pat. No. 6,110,919A; U.S. Pat. No. 6,117,890A; U.S. Pat. No. 6,127,373A; U.S. Pat. No. 6,143,767A; U.S. Pat. No. 6,147,072A; U.S. Pat. No. 6,150,366A; U.S. Pat. No. 6,150,388A; U.S. Pat. No. 6,166,020A; U.S. Pat. No. 6,166,064A; U.S. Pat. No. 6,172,073B1; U.S. Pat. No. 6,174,895B1; U.S. Pat. No. 6,194,454B1; U.S. Pat. No. 6,197,773B1; U.S. Pat. No. 6,235,734B1; U.S. Pat. No. 6,235,747B1; U.S. Pat. No. 6,245,765B1; U.S. Pat. No. 6,245,766B1; U.S. Pat. No. 6,284,771B1; U.S. Pat. No. 6,312,717B1; U.S. Pat. No. 6,323,208B1; U.S. Pat. No. 6,326,398B1; U.S. Pat. No. 6,329,396B1; U.S. Pat. No. 6,358,950B1; U.S. Pat. No. 6,369,074B1; U.S. Pat. No. 6,380,186B1; U.S. Pat. No. 6,380,233B1; U.S. Pat. No. 6,395,735B2; U.S. Pat. No. 6,395,784B1; U.S. Pat. No. 6,399,609B1; U.S. Pat. No. 6,410,739B1; U.S. Pat. No. 6,429,317B1; U.S. Pat. No. 6,433,009B1; U.S. Pat. No. 6,436,914B1; U.S. Pat. No. 6,436,938B1; U.S. Pat. No. 6,441,015B2; U.S. Pat. No. 6,444,665B1; U.S. Pat. No. 6,448,261B1; U.S. Pat. No. 6,462,048B2; U.S. Pat. No. 6,465,491B2; U.S. Pat. No. 6,476,051B2; U.S. Pat. No. 6,506,775B1; U.S. Pat. No. 6,515,005B2; U.S. Pat. No. 6,518,271B1; U.S. Pat. No. 6,525,048B1; U.S. Pat. No. 6,525,196B1; U.S. Pat. No. 6,545,018B2; U.S. Pat. No. 6,545,022B1; U.S. Pat. No. 6,548,493B1; U.S. Pat. No. 6,548,502B2; U.S. Pat. No. 6,552,017B1; U.S. Pat. No. 6,596,900B2; U.S. Pat. No. 6,620,830B2; U.S. Pat. No. 6,627,771B1; U.S. Pat. No. 6,630,469B2; U.S. Pat. No. 6,630,476B2; U.S. Pat. No. 6,632,831B2; U.S. Pat. No. 6,635,270B2; U.S. Pat. No. 6,638,934B2; U.S. Pat. No. 6,673,811B1; U.S. Pat. No. 6,686,361B2; U.S. Pat. No. 6,710,040B1; U.S. Pat. No. 6,710,071B2; U.S. Pat. No. 6,713,490B2; U.S. Pat. No. 6,734,185B2; U.S. Pat. No. 6,777,406B2; U.S. Pat. No. 6,777,437B2; U.S. Pat. No. 6,784,180B2; U.S. Pat. No. 6,818,648B2; U.S. Pat. No. 6,821,976B2; U.S. Pat. No. 6,835,733B2; U.S. Pat. No. 6,844,344B2; U.S. Pat. No. 6,849,619B2; U.S. Pat. No. 6,875,771B2; U.S. Pat. No. 6,888,004B2; U.S. Pat. No. 6,894,045B2; U.S. Pat. No. 6,900,210B2; U.S. Pat. No. 6,924,310B2; U.S. Pat. No. 6,936,601B2; U.S. Pat. No. 6,958,351B2; U.S. Pat. No. 6,960,577B2; U.S. Pat. No. 6,992,087B2; U.S. Pat. No. 7,015,229B2; U.S. Pat. No. 7,030,145B2; U.S. Pat. No. 7,041,672B2; U.S. Pat. No. 7,045,529B2; U.S. Pat. No. 7,045,551B2; U.S. Pat. No. 7,049,314B2; U.S. Pat. No. 7,053,122B2; U.S. Pat. No. 7,067,658B2; U.S. Pat. No. 7,087,609B2; U.S. Pat. No. 7,098,217B2; U.S. Pat. No. 7,101,881B2; U.S. Pat. No. 7,101,885B2; U.S. Pat. No. 7,105,516B2; U.S. Pat. No. 7,109,164B2; U.S. Pat. No. 7,112,585B2; U.S. Pat. No. 7,115,587B2; U.S. Pat. No. 7,115,600B2; U.S. Pat. No. 7,135,472B2; U.S. Pat. No. 7,144,881B2; U.S. Pat. No. 7,144,898B2; U.S. Pat. No. 7,157,488B2; U.S. Pat. No. 7,238,699B2; U.S. Pat. No. 7,276,526B2; U.S. Pat. No. 7,319,100B2; U.S. Pat. No. 7,345,038B2; U.S. Pat. No. 7,384,934B2; U.S. Pat. No. 7,439,236B2; U.S. Pat. No. 7,485,636B2; U.S. Pat. No. 7,553,836B2; U.S. Pat. No. 7,662,817B2; U.S. Pat. No. 7,671,072B2; U.S. Pat. No. 7,678,793B2; U.S. Pat. No. 7,709,522B2; U.S. Pat. No. 7,851,622B2; U.S. Pat. No. 7,932,249B2; U.S. Pat. No. 7,956,049B2; U.S. Pat. No. 7,973,159B2; U.S. Pat. No. 8,022,062B2; U.S. Pat. No. 8,124,639B2; U.S. RE039679E; in EP Patents or Patent Publications EP1033364B1; EP104860B1; EP106486A2; EP106487A2; EP1070058B1 EP1082960A2; EP1088819B1; EP1099446B1; EP1104420B1; EP1113015B1; EP1114817B1; EP1140929B1; EP1140931B1; EP1157001B1; EP1177792A2; EP1177798A2; EP1186318A2; EP1189904B1; EP1189905B1; EP1192165B1; EP1192952A2; EP1199068B1; EP1209157A1; EP1211247A1; EP1213031A2; EP1224930A1; EP1230921A1; EP1238676A1; EP1242411B1; EP1250336B1; EP1254662A2; EP1254668A2; EP1257526B1; EP1260221A2; EP1268396B1; EP1268404B1; EP1272484B1; EP1280781B1; EP1284257A2; EP1292568B1; EP1294677B1; EP1297833B1; EP1344779B1; EP1345942B1; EP1347760B1; EP1368094B1; EP1379239B1; EP1380298A2; EP1399445B1; EP1458368B1; EP1468686A2; EP1492794B1; EP1499606B1; EP1542668B1; EP1546134B1; EP1556378B1; EP1641454B1; EP1641455B1; EP1666886A2; EP1689721B1; EP171550B1; EP1727794B1; EP1805165B1; EP1824852B1; EP1838716B1; EP1846410B1; EP1899296B1; EP1908764A1; EP1924560B1; EP2044029B1; EP2094684B1; EP2124933B1; EP2231630B1; EP2252581B1; EP2280961B1; EP2298776A1; EP2479168A1; EP25603A1; EP25985A1; EP279598A2; EP281309A1; EP307172A2; EP318933A2; EP329168A2; EP397364B1; EP397365A1; EP409435B1; EP436334A2; EP497314A1; EP532527B1; EP533487A1; EP537993A1; EP545421A1; EP589924B1; EP591333B1; EP592438B1; EP594636B1; EP607164B1; EP613458B1; EP635015B1; EP641328B1; EP668863B1; EP687268B1; EP689536B1; EP708771B1; EP716649B1; EP722941A2; EP737194B1; EP738513A1; EP747353A2; EP756869A2; EP773023A1; EP806423A1; EP810220B1; EP821955B1; EP830864B1; EP868892A1; EP874625B1; EP884310A1; EP884316A1; EP891332B1; EP894085B1; EP901374B1; EP901789A1; EP904273B1; EP909561A2; EP915880B1; EP918772B1; EP929528B1; EP931547B1; EP937077B1; EP952154A2; EP958824A2; EP964849B1; EP965343A2; EP966967A2; and in PCT Patent Publications WO0016777A1; WO0041684A1; WO0071107A2; WO0074784A1; WO146177A1; WO146179A1; WO146181A1; WO146186A1; WO146187A1; WO0160784A1; WO0168592A1; WO0172692A1; WO0177100A2; WO0185145A2; WO0189530A2; WO0192526A1; WO0194293A2; WO0203684A2; WO02058704A1; WO02059124A2; WO02059127A2; WO02059129A2; WO02072101A1; WO02072202A1; WO02079152A1; WO02092090A1; WO0219998A2; WO0246167A1; WO247685A2; WO3000646A1; WO3006015A1; WO03009851A1; WO3010161A1; WO3022820A1; WO03032974A2; WO03043637A1; WO03049724A1; WO03082877A1; WO03084610A1; WO3093499A2; WO03105815A1; WO03105902A1; WO2004000355A1; WO2004014895A1; WO2004016583A1; WO2004016593A1; WO2004017897A2; WO2004031189A1; WO2004039367A1; WO2004085439A1; WO2004096773A1; WO2004100954A1; WO2004100956A1; WO2004100957A1; WO2004100992A2; WO2005002578A1; WO20050139611A1; WO2005019180A1; WO2005023265A1; WO2005035523A1; WO200504011A1; WO2005051488A1; WO2005051919A1; WO2005060949A2; WO2005060963A1; WO2005061491A2; WO2005063296A2; WO2005066126A1; WO2005067973A2; WO2005070916A1; WO2005079807A1; WO2005080361A1; WO2005082370A1; WO2005090300A1; WO2005092318A1; WO2005102272A2; WO2006016278A1; WO2006019886A2; WO2006019940A2; WO2006027691A2; WO2006044176A1; WO2006044454A1; WO2006048727A1; WO2006061711A1; WO2006073886A1; WO2006086464A2; WO2006088716A1; WO2006090272A1; WO2006090273A2; WO2006103559A1; WO2006106416A1; WO2006116401A1; WO2006136924A1; WO2007017750A1; WO2007026219A2; WO2007028082A1; WO2007028083A2; WO2007028131A1; WO2007028132A2; WO2007031828A2; WO2007050723A1; WO2007057742A2; WO2007063385A2; WO2007069053A1; WO2007085954A2; WO2007088450A2; WO2007088462A1; WO2007096743A1; WO2007099423A1; WO2007105053A2; WO2007129183A2; WO2007138431A2; WO2008001182A1; WO2008010073A1; WO2008015516A1; WO2008020302A2; WO2008020306A2; WO2008026046A1; WO2008032164A2; WO2008065500A2; WO2008070306A2; WO2008096260A1; WO2008125945A2; WO2008134480A1; WO2009009501A2; WO2009071988A1; WO2009094260A1; WO2009098576A1; WO2009127944A1; WO2009131814A2; WO2009149258A2; WO2010009062A1; WO2010014280A1; WO2010049841A1; WO2010058318A1; WO2010104818A1; WO2010104830A1; WO2010111080A2; WO2010146488A1; WO2011060035A1; WO2012004698A1; WO2012038850A1; WO2012056402A2; WO2012073143A1; WO2012073146A1; WO2012114222A1; WO8803024A1; WO9002552A1; WO9005525A1; WO9006303A1; WO9007926A1; WO9100863A1; WO9109844A1; WO9206079A1; WO9306101A1; WO9320073A1; WO9403445A1; WO9410171A1; WO9413676A1; WO9413677A1; WO9534563A1; WO9603400A1; WO9606081A1; WO9610570A1; WO9624353A1; WO9703066A1; WO9703665A1; WO9723220A1; WO9725983A1; WO9733577A1; WO9735584A1; WO9736867A1; WO9742190A1; WO9742191A1; WO9814433A1; WO9818798A1; WO9845268A1; WO9845287A1; WO9846225A 1; WO9846226A1; WO9904778A1; WO9939725A1; WO9952889A1; WO9952907A1; WO9959593A1; WO9961441A1, or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof.
31-35. (canceled)
36. The method of claim 25, wherein the antipsychotic is useful in treating at least one sign or symptom of schizophrenia or bipolar disorder.
37. (canceled)
38. (canceled)
39. A pharmaceutical composition comprising a SV2A inhibitor and an antipsychotic or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs.
40. The pharmaceutical composition of claim 39, wherein the SV2A inhibitor and the antipsychotic or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs are in separate dosage forms or in a unit dosage form.
41. The composition of claim 39, wherein the composition is in a solid form, a liquid form, a suspension form, a sustained release form, a delayed release form, or an extended release form.
42. The composition of claim 39, wherein the SV2A inhibitor is:
a) selected from the group of SV2A inhibitors referred to in International Patent Application PCT/US2009/005647; International Patent Application Publications WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787; WO2001/062726; U.S. Pat. Nos. 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. patent application Ser. Nos. 12/580,464; 61/105,847; 61/152,631; and 61/175,536; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692; or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof; or
b) selected from the group consisting of levetiracetam, seletracetam, and brivaracetam and derivatives, analogs, pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof.
43. (canceled)
44. The composition of claim 39, wherein the SV2A inhibitor in the composition is present in an amount of:
a) 0.07-350 mg;
b) 50-250 mg;
c) 3-50 mg; or
d) less than 350 mg, less than 250 mg, less than 200 mg, less than 150 mg, less than 100 mg, less than 50 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0.5 mg, less than 0.1 mg, or less than 0.07 mg.
45-47. (canceled)
48. The composition of claim 39, wherein the antipsychotic is selected from atypical and typical antipsychotics.
49. The composition of claim 48, wherein the antipsychotic is:
a) an atypical antipsychotic;
b) selected from aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvates, polymorphs thereof;
c) selected from aripiprazole, olanzapine and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof;
d) a typical antipsychotic;
e) selected from acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof;
f) a compound selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, NNOS inhibits, neurosteroids, and neurotrophic factors;
g) selected from the group of compounds referred to in U.S. Pat. Nos. 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof; or
h) selected from the group of compounds referred to in U.S. Patents or Patent Publications US20020052401A1; US20020091118A1; US20020091119A1; US20020094986A1; US20020123490A1; US20020147194A1; US20020156089A1; US20020165217A1; US20030008806A1; US20030008892A1; US20030013887A1; US20030018047A1; US20030027812A1; US20030032579A1; US20030130303A1; US20030158208A1; US20030162766A1; US20030181458A1; US20030191176A1; US20030208081A1; US20030232841A1; US20030235631A1; US20040001895A1; US20040006135A1; US20040029876A1; US20040039022A1; US20040048869A1; US20040049039A1; US20040082597A1; US20040106603A1; US20040110817A1; US20040116443A1; US20040132713A1; US20040138232A1; US20040162293A1; US20040162294A1; US20040167200A1; US20040204414A1; US20040204415A1; US20040204445A1; US20040204453A1; US20040209887A1; US20040220184A1; US20040220274A1; US20040229874A1; US20040229911A1; US20040242587A1; US20040254193A1; US20040259859A1; US20040266781A1; US20040266815A1; US20050004106A1; US20050004137A1; US20050009927A1; US20050014764A1; US20050014848A1; US20050026937A1; US20050026946A1; US20050032837A1; US20050038036A1; US20050043292A1; US20050070577A1; US20050080099A1; US20050080100A1; US20050101613A1; US20050107425A1; US20050113437A1; US20050143403A1; US20050171086A1; US20050171095A1; US20050182049A1; US20050203296A1; US20050209250A1; US20050215571A1; US20050227980A1; US20050227981A1; US20050234065A1; US20050245521A1; US20050245543A1; US20050250803A1; US20050256112A1; US20050256162A1; US20050282811A1; US20050282816A1; US20050288304A1; US20060014733A1; US20060019998A1; US200600254211A1; US20060047114A1; US20060052373A1; US20060058361A1; US20060069087A1; US20060084692A1; US20060094719A1; US20060148807A1; US20060166974A1; US20060166998A1; US20060173179A1; US20060183763A1; US20060217398A1; US20060229455A1; US20060270656A1; US20070015763A1; US20070027178A1; US20070049613A1; US20070054913A1; US20070093509A1; US20070155779A1; US20070185097A1; US20070213337A1; US20070224636A1; US20080045512A1; US20080096955A1; US20080139567A1; US20080167319A1; US20080176925A1; US20080207897A1; US20080269110A1; US20080269246A1; US20080305161A1; US20080318926A1; US20090011994A1; US20090023756A1; US20090093512A1; US20090131433A1; US20090176829A1; US20090197859A1; US20090215857A1; US20090298811A1; US20100004259A1; US20100029684A1; US20100113465A1; US20100130501A1; US20100222353A1; US20100249128A1; US20100324043A1; US20110003759A1; US20110028520A1; US20110034484A1; US20110144060A1; US20110144159A1; US20110152334A1; US20110160220A1; US20110166160A1; US20110230493A1; US20110269745A1; US20110319439A1; US20110319449A1; US20120028961A1; US20120108588A1; US20120142729A1; US20120202809A1; US20120214784A1; US20120214791A1; US20120220568A1; U.S. Pat. No. 3,953,603A; U.S. Pat. No. 4,018,895A; U.S. Pat. No. 4,337,250A; U.S. Pat. No. 4,352,807A; U.S. Pat. No. 4,427,679A; U.S. Pat. No. 4,431,649A; U.S. Pat. No. 4,495,187A; U.S. Pat. No. 4,547,501A; U.S. Pat. No. 4,593,037A; U.S. Pat. No. 4,599,339A; U.S. Pat. No. 4,605,655A; U.S. Pat. No. 4,619,930A; U.S. Pat. No. 4,656,173A; U.S. Pat. No. 4,677,104A; U.S. Pat. No. 4,757,073A; U.S. Pat. No. 4,771,053A; U.S. Pat. No. 4,784,998A; U.S. Pat. No. 4,879,391A; U.S. Pat. No. 4,883,795A; U.S. Pat. No. 4,891,375A; U.S. Pat. No. 4,931,447A; U.S. Pat. No. 4,933,343A; U.S. Pat. No. 4,956,368A; U.S. Pat. No. 4,977,178A; U.S. Pat. No. 4,981,870A; U.S. Pat. No. 5,006,525A; U.S. Pat. No. 5,011,841A; U.S. Pat. No. 5,043,341A; U.S. Pat. No. 5,051,412A; U.S. Pat. No. 5,077,295A; U.S. Pat. No. 5,157,034A; U.S. Pat. No. 5,162,339A; U.S. Pat. No. 5,232,929A; U.S. Pat. No. 5,238,959A; U.S. Pat. No. 5,242,911A; U.S. Pat. No. 5,256,664A; U.S. Pat. No. 5,276,040A; U.S. Pat. No. 5,294,619A; U.S. Pat. No. 5,312,925A; U.S. Pat. No. 5,350,747A; U.S. Pat. No. 5,364,863A; U.S. Pat. No. 5,373,003A; U.S. Pat. No. 5,385,916A; U.S. Pat. No. 5,399,565A; U.S. Pat. No. 5,422,354A; U.S. Pat. No. 5,451,586A; U.S. Pat. No. 5,498,610A; U.S. Pat. No. 5,498,614A; U.S. Pat. No. 5,498,626A; U.S. Pat. No. 5,521,220A; U.S. Pat. No. 5,527,808A; U.S. Pat. No. 5,559,129A; U.S. Pat. No. 5,563,148A; U.S. Pat. No. 5,569,662A; U.S. Pat. No. 5,576,321A; U.S. Pat. No. 5,578,612A; U.S. Pat. No. 5,594,014A; U.S. Pat. No. 5,597,826A; U.S. Pat. No. 5,604,241A; U.S. Pat. No. 5,604,252A; U.S. Pat. No. 5,627,200A; U.S. Pat. No. 5,639,752A; U.S. Pat. No. 5,658,590A; U.S. Pat. No. 5,688,804A; U.S. Pat. No. 5,696,168A; U.S. Pat. No. 5,698,568A; U.S. Pat. No. 5,703,065A; U.S. Pat. No. 5,710,168A; U.S. Pat. No. 5,716,965A; U.S. Pat. No. 5,721,255A; U.S. Pat. No. 5,731,307A; U.S. Pat. No. 5,736,541A; U.S. Pat. No. 5,741,797A; U.S. Pat. No. 5,744,480A; U.S. Pat. No. 5,747,501A; U.S. Pat. No. 5,789,423A; U.S. Pat. No. 5,817,656A; U.S. Pat. No. 5,821,248A; U.S. Pat. No. 5,837,711A; U.S. Pat. No. 5,849,739A; U.S. Pat. No. 5,854,232A; U.S. Pat. No. 5,854,239A; U.S. Pat. No. 5,854,256A; U.S. Pat. No. 5,886,008A; U.S. Pat. No. 5,889,010A; U.S. Pat. No. 5,912,256A; U.S. Pat. No. 5,939,433A; U.S. Pat. No. 5,942,524A; U.S. Pat. No. 5,958,921A; U.S. Pat. No. 5,985,322A; U.S. Pat. No. 5,994,352A; U.S. Pat. No. 6,020,335A; U.S. Pat. No. 6,043,258A; U.S. Pat. No. 6,046,193A; U.S. Pat. No. 6,046,213A; U.S. Pat. No. 6,060,479A; U.S. Pat. No. 6,083,943A; U.S. Pat. No. 6,087,392A; U.S. Pat. No. 6,110,918A; U.S. Pat. No. 6,110,919A; U.S. Pat. No. 6,117,890A; U.S. Pat. No. 6,127,373A; U.S. Pat. No. 6,143,767A; U.S. Pat. No. 6,147,072A; U.S. Pat. No. 6,150,366A; U.S. Pat. No. 6,150,388A; U.S. Pat. No. 6,166,020A; U.S. Pat. No. 6,166,064A; U.S. Pat. No. 6,172,073B1; U.S. Pat. No. 6,174,895B1; U.S. Pat. No. 6,194,454B1; U.S. Pat. No. 6,197,773B1; U.S. Pat. No. 6,235,734B1; U.S. Pat. No. 6,235,747B1; U.S. Pat. No. 6,245,765B1; U.S. Pat. No. 6,245,766B1; U.S. Pat. No. 6,284,771B1; U.S. Pat. No. 6,312,717B1; U.S. Pat. No. 6,323,208B1; U.S. Pat. No. 6,326,398B1; U.S. Pat. No. 6,329,396B1; U.S. Pat. No. 6,358,950B1; U.S. Pat. No. 6,369,074B1; U.S. Pat. No. 6,380,186B1; U.S. Pat. No. 6,380,233B1; U.S. Pat. No. 6,395,735B2; U.S. Pat. No. 6,395,784B1; U.S. Pat. No. 6,399,609B1; U.S. Pat. No. 6,410,739B1; U.S. Pat. No. 6,429,317B1; U.S. Pat. No. 6,433,009B1; U.S. Pat. No. 6,436,914B1; U.S. Pat. No. 6,436,938B1; U.S. Pat. No. 6,441,015B2; U.S. Pat. No. 6,444,665B1; U.S. Pat. No. 6,448,261B1; U.S. Pat. No. 6,462,048B2; U.S. Pat. No. 6,465,491B2; U.S. Pat. No. 6,476,051B2; U.S. Pat. No. 6,506,775B1; U.S. Pat. No. 6,515,005B2; U.S. Pat. No. 6,518,271B1; U.S. Pat. No. 6,525,048B1; U.S. Pat. No. 6,525,196B1; U.S. Pat. No. 6,545,018B2; U.S. Pat. No. 6,545,022B1; U.S. Pat. No. 6,548,493B1; U.S. Pat. No. 6,548,502B2; U.S. Pat. No. 6,552,017B1; U.S. Pat. No. 6,596,900B2; U.S. Pat. No. 6,620,830B2; U.S. Pat. No. 6,627,771B1; U.S. Pat. No. 6,630,469B2; U.S. Pat. No. 6,630,476B2; U.S. Pat. No. 6,632,831B2; U.S. Pat. No. 6,635,270B2; U.S. Pat. No. 6,638,934B2; U.S. Pat. No. 6,673,811B1; U.S. Pat. No. 6,686,361B2; U.S. Pat. No. 6,710,040B1; U.S. Pat. No. 6,710,071B2; U.S. Pat. No. 6,713,490B2; U.S. Pat. No. 6,734,185B2; U.S. Pat. No. 6,777,406B2; U.S. Pat. No. 6,777,437B2; U.S. Pat. No. 6,784,180B2; U.S. Pat. No. 6,818,648B2; U.S. Pat. No. 6,821,976B2; U.S. Pat. No. 6,835,733B2; U.S. Pat. No. 6,844,344B2; U.S. Pat. No. 6,849,619B2; U.S. Pat. No. 6,875,771B2; U.S. Pat. No. 6,888,004B2; U.S. Pat. No. 6,894,045B2; U.S. Pat. No. 6,900,210B2; U.S. Pat. No. 6,924,310B2; U.S. Pat. No. 6,936,601B2; U.S. Pat. No. 6,958,351B2; U.S. Pat. No. 6,960,577B2; U.S. Pat. No. 6,992,087B2; U.S. Pat. No. 7,015,229B2; U.S. Pat. No. 7,030,145B2; U.S. Pat. No. 7,041,672B2; U.S. Pat. No. 7,045,529B2; U.S. Pat. No. 7,045,551B2; U.S. Pat. No. 7,049,314B2; U.S. Pat. No. 7,053,122B2; U.S. Pat. No. 7,067,658B2; U.S. Pat. No. 7,087,609B2; U.S. Pat. No. 7,098,217B2; U.S. Pat. No. 7,101,881B2; U.S. Pat. No. 7,101,885B2; U.S. Pat. No. 7,105,516B2; U.S. Pat. No. 7,109,164B2; U.S. Pat. No. 7,112,585B2; U.S. Pat. No. 7,115,587B2; U.S. Pat. No. 7,115,600B2; U.S. Pat. No. 7,135,472B2; U.S. Pat. No. 7,144,881B2; U.S. Pat. No. 7,144,898B2; U.S. Pat. No. 7,157,488B2; U.S. Pat. No. 7,238,699B2; U.S. Pat. No. 7,276,526B2; U.S. Pat. No. 7,319,100B2; U.S. Pat. No. 7,345,038B2; U.S. Pat. No. 7,384,934B2; U.S. Pat. No. 7,439,236B2; U.S. Pat. No. 7,485,636B2; U.S. Pat. No. 7,553,836B2; U.S. Pat. No. 7,662,817B2; U.S. Pat. No. 7,671,072B2; U.S. Pat. No. 7,678,793B2; U.S. Pat. No. 7,709,522B2; U.S. Pat. No. 7,851,622B2; U.S. Pat. No. 7,932,249B2; U.S. Pat. No. 7,956,049B2; U.S. Pat. No. 7,973,159B2; U.S. Pat. No. 8,022,062B2; U.S. Pat. No. 8,124,639B2; U.S. RE039679E; in EP Patents or Patent Publications EP1033364B1; EP104860B1; EP106486A2; EP106487A2; EP1070058B1 EP1082960A2; EP1088819B1; EP1099446B1; EP1104420B1; EP1113015B1; EP1114817B1; EP1140929B1; EP1140931B1; EP1157001B1; EP1177792A2; EP1177798A2; EP1186318A2; EP1189904B1; EP1189905B1; EP1192165B1; EP1192952A2; EP1199068B1; EP1209157A1; EP1211247A1; EP1213031A2; EP1224930A1; EP1230921A1; EP1238676A1; EP1242411B1; EP1250336B1; EP1254662A2; EP1254668A2; EP1257526B1; EP1260221A2; EP1268396B1; EP1268404B1; EP1272484B1; EP1280781B1; EP1284257A2; EP1292568B1; EP1294677B1; EP1297833B1; EP1344779B1; EP1345942B1; EP1347760B1; EP1368094B1; EP1379239B1; EP1380298A2; EP1399445B1; EP1458368B1; EP1468686A2; EP1492794B1; EP1499606B1; EP1542668B1; EP1546134B1; EP1556378B1; EP1641454B1; EP1641455B1; EP1666886A2; EP1689721B1; EP171550B1; EP1727794B1; EP1805165B1; EP1824852B1; EP1838716B1; EP1846410B1; EP1899296B1; EP1908764A1; EP1924560B1; EP2044029B1; EP2094684B1; EP2124933B1; EP2231630B1; EP2252581B1; EP2280961B1; EP2298776A1; EP2479168A1; EP25603A1; EP25985A1; EP279598A2; EP281309A1; EP307172A2; EP318933A2; EP329168A2; EP397364B1; EP397365A1; EP409435B1; EP436334A2; EP497314A1; EP532527B1; EP533487A1; EP537993A1; EP545421A1; EP589924B1; EP591333B1; EP592438B1; EP594636B1; EP607164B1; EP613458B1; EP635015B1; EP641328B1; EP668863B1; EP687268B1; EP689536B1; EP708771B1; EP716649B1; EP722941A2; EP737194B1; EP738513A1; EP747353A2; EP756869A2; EP773023A1; EP806423A1; EP810220B1; EP821955B1; EP830864B1; EP868892A1; EP874625B1; EP884310A1; EP884316A1; EP891332B1; EP894085B1; EP901374B1; EP901789A1; EP904273B1; EP909561A2; EP915880B1; EP918772B1; EP929528B1; EP931547B1; EP937077B1; EP952154A2; EP958824A2; EP964849B1; EP965343A2; EP966967A2; and in PCT Patent Publications WO0016777A1; WO0041684A1; WO0071107A2; WO0074784A1; WO0146177A1; WO0146179A1; WO0146181A1; WO0146186A1; WO0146187A1; WO0160784A1; WO0168592A1; WO0172692A1; WO0177100A2; WO0185145A2; WO0189530A2; WO0192526A1; WO0194293A2; WO0203684A2; WO02058704A1; WO02059124A2; WO02059127A2; WO02059129A2; WO02072101A1; WO02072202A1; WO02079152A1; WO02092090A1; WO0219998A2; WO0246167A1; WO0247685A2; WO03000646A1; WO03006015A1; WO03009851A1; WO03010161A1; WO03022820A1; WO03032974A2; WO03043637A1; WO03049724A1; WO03082877A1; WO03084610A1; WO03093499A2; WO03105815A1; WO03105902A1; WO2004000355A1; WO2004014895A1; WO2004016583A1; WO2004016593A1; WO2004017897A2; WO2004031189A1; WO2004039367A1; WO2004085439A1; WO2004096773A1; WO2004100954A1; WO2004100956A1; WO2004100957A1; WO2004100992A2; WO2005002578A1; WO20050139611A1; WO2005019180A1; WO2005023265A1; WO2005035523A1; WO200504011A1; WO2005051488A1; WO2005051919A1; WO2005060949A2; WO2005060963A1; WO2005061491A2; WO2005063296A2; WO2005066126A1; WO2005067973A2; WO2005070916A1; WO2005079807A1; WO2005080361A1; WO2005082370A1; WO2005090300A1; WO2005092318A1; WO2005102272A2; WO2006016278A1; WO2006019886A2; WO2006019940A2; WO2006027691A2; WO2006044176A1; WO2006044454A1; WO2006048727A1; WO2006061711A1; WO2006073886A1; WO2006086464A2; WO2006088716A1; WO2006090272A1; WO2006090273A2; WO2006103559A1; WO2006106416A1; WO2006116401A1; WO2006136924A1; WO2007017750A1; WO2007026219A2; WO2007028082A1; WO2007028083A2; WO2007028131A1; WO2007028132A2; WO2007031828A2; WO2007050723A1; WO2007057742A2; WO2007063385A2; WO2007069053A1; WO2007085954A2; WO2007088450A2; WO2007088462A1; WO2007096743A1; WO2007099423A1; WO2007105053A2; WO2007129183A2; WO2007138431A2; WO2008001182A1; WO2008010073A1; WO2008015516A1; WO2008020302A2; WO2008020306A2; WO2008026046A1; WO2008032164A2; WO2008065500A2; WO2008070306A2; WO2008096260A1; WO2008125945A2; WO2008134480A1; WO2009009501A2; WO2009071988A1; WO2009094260A1; WO2009098576A1; WO2009127944A1; WO2009131814A2; WO2009149258A2; WO2010009062A1; WO2010014280A1; WO2010049841A1; WO2010058318A1; WO2010104818A1; WO2010104830A1; WO2010111080A2; WO2010146488A1; WO2011060035A1; WO2012004698A1; WO2012038850A1; WO2012056402A2; WO2012073143A1; WO2012073146A1; WO2012114222A1; WO8803024A1; WO9002552A1; WO9005525A1; WO9006303A1; WO9007926A1; WO9100863A1; WO9109844A1; WO9206079A1; WO9306101A1; WO9320073A1; WO9403445A1; WO9410171A1; WO9413676A1; WO9413677A1; WO9534563A1; WO9603400A1; WO9606081A1; WO9610570A1; WO9624353A1; WO9703066A1; WO9703665A1; WO9723220A1; WO9725983A1; WO9733577A1; WO9735584A1; WO9736867A1; WO9742190A1; WO9742191A1; WO9814433A1; WO9818798A1; WO9845268A1; WO9845287A1; WO9846225A1; WO9846226A1; WO9904778A1; WO9939725A1; WO9952889A1; WO9952907A1; WO9959593A1; WO9961441A1, or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof.
50-54. (canceled)
55. The composition of claim 39, wherein the antipsychotic is useful in treating at least one sign or symptom of schizophrenia or bipolar disorder.
56. (canceled)
57. (canceled)
US14/080,531 2012-11-14 2013-11-14 Methods and compositions for treating schizophrenia Abandoned US20140206667A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/080,531 US20140206667A1 (en) 2012-11-14 2013-11-14 Methods and compositions for treating schizophrenia
US15/629,314 US10624875B2 (en) 2012-11-14 2017-06-21 Methods and compositions for treating schizophrenia

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261726440P 2012-11-14 2012-11-14
US14/080,531 US20140206667A1 (en) 2012-11-14 2013-11-14 Methods and compositions for treating schizophrenia

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/629,314 Continuation US10624875B2 (en) 2012-11-14 2017-06-21 Methods and compositions for treating schizophrenia

Publications (1)

Publication Number Publication Date
US20140206667A1 true US20140206667A1 (en) 2014-07-24

Family

ID=50731694

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/442,996 Active US10154988B2 (en) 2012-11-14 2013-11-14 Methods and compositions for treating schizophrenia
US14/080,531 Abandoned US20140206667A1 (en) 2012-11-14 2013-11-14 Methods and compositions for treating schizophrenia
US15/629,314 Active US10624875B2 (en) 2012-11-14 2017-06-21 Methods and compositions for treating schizophrenia

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/442,996 Active US10154988B2 (en) 2012-11-14 2013-11-14 Methods and compositions for treating schizophrenia

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/629,314 Active US10624875B2 (en) 2012-11-14 2017-06-21 Methods and compositions for treating schizophrenia

Country Status (6)

Country Link
US (3) US10154988B2 (en)
EP (2) EP2919788A4 (en)
JP (2) JP6440625B2 (en)
CA (1) CA2891122C (en)
HK (1) HK1215170A1 (en)
WO (1) WO2014078568A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10154988B2 (en) 2012-11-14 2018-12-18 The Johns Hopkins University Methods and compositions for treating schizophrenia
US10159648B2 (en) 2015-05-22 2018-12-25 Agenebio, Inc. Extended release pharmaceutical compositions of levetiracetam
US10806717B2 (en) 2013-03-15 2020-10-20 The Johns Hopkins University Methods and compositions for improving cognitive function
US10898449B2 (en) 2016-12-20 2021-01-26 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
US11033512B2 (en) 2017-06-26 2021-06-15 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and silicone acrylic hybrid polymer
US11160785B2 (en) 2013-03-15 2021-11-02 Agenebio Inc. Methods and compositions for improving cognitive function
US11253173B1 (en) * 2017-05-30 2022-02-22 Verily Life Sciences Llc Digital characterization of movement to detect and monitor disorders
US11337932B2 (en) 2016-12-20 2022-05-24 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and polysiloxane or polyisobutylene
US11648213B2 (en) 2018-06-20 2023-05-16 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
LT3096790T (en) 2014-01-21 2019-10-10 Janssen Pharmaceutica, N.V. Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
WO2015110435A1 (en) * 2014-01-21 2015-07-30 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
KR101634440B1 (en) 2014-08-21 2016-06-30 서울대학교산학협력단 Pharmaceutical Composition Comprising Inhibitors of Phosphorylation of GluA1 Subunit of AMPA receptor for preventing or treating mental disorders
WO2017195144A1 (en) * 2016-05-12 2017-11-16 Jubilant Generics Limited Pharmaceutical compositions comprising brivaracetam
CN106109402A (en) * 2016-07-20 2016-11-16 南通雅本化学有限公司 A kind of levetiracetam injection
EP4178582A4 (en) * 2020-07-10 2024-07-10 Agenebio Inc Combinations of gabaa alpha 5 agonists and sv2a inhibitors and methods of using in the treatment of cognitive impairment

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070212428A1 (en) * 2004-06-04 2007-09-13 Mood Management Sciences, Inc. Methods and compositions for treating mood disorder
US20070244143A1 (en) * 2006-03-08 2007-10-18 Braincells, Inc Modulation of neurogenesis by nootropic agents
US20070264358A1 (en) * 2004-06-04 2007-11-15 Wittlin William A Methods and Compositions for Treating Mood Disorder

Family Cites Families (728)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5516A (en) 1848-04-18 Combining lantebns and lamps
US759A (en) 1838-05-30 Reacting watek-wheels
US1015498A (en) 1911-03-01 1912-01-23 Edward L Kraus Folding-desk blackboard.
NL181407B (en) 1952-09-20 Merck & Co Inc IMPROVEMENT OF A PROCESS FOR PREPARING A PHARMACEUTICAL PREPARATION WITH ANTIBACTERIAL ACTION CONTAINING A COMBINATION OF A 3-FLUOR-D-ALANINE COMPOUND AND A CYCLOSERINE COMPOUND.
GB1039113A (en) 1964-08-06 1966-08-17 Ucb Sa New n-substituted lactams
GB1309692A (en) 1970-02-13 1973-03-14 Ucb Sa N-substituted lactams
US3539573A (en) 1967-03-22 1970-11-10 Jean Schmutz 11-basic substituted dibenzodiazepines and dibenzothiazepines
JP2000319257A (en) 1969-01-06 2000-11-21 Eisai Co Ltd 1-benzyl-4-[(5,6-dimethoxy-2-fluoro-1-indanon)-2-yl methyl piperidine
US3953603A (en) 1973-11-05 1976-04-27 Eli Lilly And Company Hexahydro-dibenzo[b,d,]pyran-9-ones as psychotropic, particularly anti-depressant drugs
US4018895A (en) 1974-01-10 1977-04-19 Eli Lilly And Company Aryloxyphenylpropylamines in treating depression
IT1045043B (en) 1975-08-13 1980-04-21 Isf Spa PYROLIDINE DERIVATIVES
NL7605526A (en) 1976-05-24 1977-11-28 Akzo Nv NEW TETRACYCLICAL DERIVATIVES.
IT1075280B (en) 1977-02-11 1985-04-22 Isf Spa PROCEDURE FOR THE PREPARATION OF PYROLIDINE DERIVATIVES
BE864269A (en) 1977-03-03 1978-06-16 Parke Davis & Co NEW N- (AMINOALKYL SUBSTITUTE) -2-OXO-1-PYRROLIDINE-ACETAMIDES AND METHODS FOR PRODUCING THEM
JPS54130587A (en) 1978-03-30 1979-10-09 Otsuka Pharmaceut Co Ltd Carbostyryl derivative
ATE27T1 (en) 1978-05-08 1981-04-15 Ucb Sa LACTAM-N-ACETIC ACIDS AND THEIR AMIDES, PROCESS FOR THEIR PREPARATION AND MEDICINES CONTAINING THEM
US4654370A (en) 1979-03-12 1987-03-31 Abbott Laboratories Glyceryl valproates
US4431649A (en) 1979-07-30 1984-02-14 Pfizer Inc. Hexahydro-trans- and tetrahydropyridoindole neuroleptic agents
US4352807A (en) 1979-07-30 1982-10-05 Pfizer Inc. Hexahydro-trans-pyridoindole neuroleptic agents
US4337250A (en) 1979-07-30 1982-06-29 Pfizer Inc. Hexahydro-trans- and tetrahydropyridoindole neuroleptic agents
US4465683A (en) 1979-09-14 1984-08-14 Mead Johnson & Company Anti-psychotic agents
US4254124A (en) 1979-09-24 1981-03-03 Mead Johnson & Company Antidepressant agent
US4372960A (en) 1980-12-12 1983-02-08 Warner-Lambert Company Quaternary derivatives of N-(substituted-aminoalkyl)-2-oxo-1-pyrrolidine-acetamides as cognition activators
US4427679A (en) 1981-01-16 1984-01-24 Pfizer Inc. Hexahydro-trans- and tetrahydropyridoindole neuroleptic agents
FR2515179A1 (en) 1981-07-24 1983-04-29 Hoffmann La Roche PYRROLIDINE DERIVATIVES, PROCESS FOR THEIR PREPARATION, INTERMEDIATES FOR THEIR SYNTHESIS AND THEIR THERAPEUTIC APPLICATION
US4415085A (en) 1981-12-21 1983-11-15 Eli Lilly And Company Dry pharmaceutical system
PT77267B (en) 1982-09-07 1986-03-21 Lilly Co Eli Improved process for preparing novel octahydrobenz <f> isoquinolines or compounds relating thereto
GR81301B (en) 1982-09-07 1984-12-11 Lilly Co Eli
US4977178A (en) 1982-09-20 1990-12-11 Pfizer Inc. Method of treating anxiety and depression with 1-phenyl-2(1H,3H)-indolone psycho-therapeutic agents
US4879391A (en) 1982-09-20 1989-11-07 Pfizer Inc. 1-Phenyl-2(1H,3H)-indolone psychotherapeutic agents
US4476307A (en) 1982-09-20 1984-10-09 Pfizer Inc. Heteroylidene indolone compounds
US4495187A (en) 1982-10-18 1985-01-22 Pfizer Inc. Method of using [1,2,4]triazolo[4,3-a]quinoxaline-4-amine derivatives as antidepressant and antifatigue agents
IL67623A (en) 1983-01-05 1984-09-30 Teva Pharma 1'-ethoxycarbonyloxyethyl ester of valproic acid,its preparation and pharmaceutical compositions containing it
IL72381A (en) 1983-07-20 1988-03-31 Sanofi Sa Pharmaceutical composition based on valproic acid
US4547501A (en) 1983-09-02 1985-10-15 Pfizer Inc. Method of using [1,2,4]triazolo[4,3-a]quinoxaline-4-amine derivatives as antidepressant and antifatigue agents
US4599339A (en) 1983-09-26 1986-07-08 Eli Lilly And Company Use of pyrimido[4,5-g]quinolines in treating parkinsonism
US4558070A (en) 1983-10-26 1985-12-10 Abbott Laboratories Acid salts of valproic acid
US4605655A (en) 1984-03-06 1986-08-12 Bristol-Myers Company Antipsychotic 1-fluorophenylbutyl-4-(2-pyrimidinyl)piperazine derivatives
GB8412357D0 (en) 1984-05-15 1984-06-20 Ucb Sa Pharmaceutical composition
GB8412358D0 (en) 1984-05-15 1984-06-20 Ucb Sa Pharmaceutical composition
US4668687A (en) 1984-07-23 1987-05-26 Bristol-Myers Company Psychogeriatric 1-(2-pyrimidinyl)piperazinyl derivatives of 1-pyrrolidin-2-ones
US4593037A (en) 1984-07-26 1986-06-03 Pfizer Inc. 1,3-disubstituted piperidine compounds as neuroleptic agents
US4619930A (en) 1985-01-16 1986-10-28 Bristol-Myers Company Antipsychotic cyclic imide derivatives of 2-(4-butylpiperazin-1-yl)pyridines, compositions and use
US4913906B1 (en) 1985-02-28 2000-06-06 Yissum Res Dev Co Controlled release dosage form of valproic acid
IL74497A (en) 1985-03-05 1990-02-09 Proterra Ag Pharmaceutical compositions containing phenyl carbamate derivatives and certain phenyl carbamate derivatives
US4804663A (en) 1985-03-27 1989-02-14 Janssen Pharmaceutica N.V. 3-piperidinyl-substituted 1,2-benzisoxazoles and 1,2-benzisothiazoles
IE58370B1 (en) 1985-04-10 1993-09-08 Lundbeck & Co As H Indole derivatives
US4656173A (en) 1985-04-24 1987-04-07 Bristol-Myers Company Antipsychotic benzisothiazole S-oxide compound
US4677104A (en) 1985-05-06 1987-06-30 Bristol-Myers Company Antipsychotic fused-ring pyridinylpiperazine derivatives
JPS6222785A (en) 1985-07-23 1987-01-30 Sanwa Kagaku Kenkyusho:Kk Novel 2-oxopyrrolidine compound and salt thereof, and preventing and treating agent for cerebral dysfunction containing said compound and salt as active constituent
US4663318A (en) 1986-01-15 1987-05-05 Bonnie Davis Method of treating Alzheimer's disease
GB8607684D0 (en) 1986-03-27 1986-04-30 Ici America Inc Thiazepine compounds
IT1190133B (en) 1986-06-19 1988-02-10 Chiesi Farma Spa VALPROIC ACID AND (E) -2-VALPROENOIC ACID DERIVATIVES, PROCEDURE FOR THEIR PREPARATION AND RELATED PHARMACEUTICAL COMPOSITIONS
US4757073A (en) 1986-09-30 1988-07-12 Bristol-Myers Company Antipsychotic cyclic imide derivatives of 2-(4-butylipiperazin-1-yl) pyridines, compositions and use
US4816456A (en) 1986-10-01 1989-03-28 Summers William K Administration of monoamine acridines in cholinergic neuronal deficit states
DD263531A5 (en) 1986-10-21 1989-01-04 ������@���Kk�� PROCESS FOR PREPARING BROKEN BICYCLIC IMIDENCES
MX174210B (en) 1987-02-17 1994-04-28 Pfizer PROCEDURE FOR THE PREPARATION OF ARILPIPERAZINYL-ALKYLENPHENYL-P-HETEROCICLICOS COMPOUNDS
MX173362B (en) 1987-03-02 1994-02-23 Pfizer PIPERAZINIL HETERO-CYCLIC COMPOUNDS AND PROCEDURE FOR THE PREPARATION
US4771053A (en) 1987-03-02 1988-09-13 Bristol-Myers Company Method for alleviation of primary depressive disorders
HU201906B (en) 1987-03-04 1991-01-28 Sandoz Ag Process for producing phenyl-carbamate derivative and acid additional salts and pharmaceutical compositions containing them
FI91401C (en) 1987-03-17 1994-06-27 Hoechst Roussel Pharma Process for the preparation of therapeutically active substituted 9-aminotetrahydroacrines and related compounds
DE3709230A1 (en) 1987-03-20 1988-10-06 Desitin Arzneimittel Gmbh NEW VALPROINIC ACID CALCIUM
IT1225462B (en) 1987-04-03 1990-11-14 Mediolanum Farmaceutici Srl ORGANIC SALTS OF PHYSOSTIGMIN DERIVATIVES
US4784998A (en) 1987-04-06 1988-11-15 Bristol-Myers Company 1,3,4-oxadiazole pyschotropic compounds
US5002955A (en) 1987-04-23 1991-03-26 Hoechst-Roussel Pharmaceuticals Inc. Fused heteroalkylene quinolinamines and use as cholinergic agents
KR100195399B1 (en) 1987-05-04 1999-06-15 보니데이비스 Compounds for the treatment of alzheimer's disease
US5187165A (en) 1987-05-15 1993-02-16 Hoechst-Roussel Pharmaceuticals Inc. Memory enhancing and analgesic 1,2,3a,8,8a-hexahydro-3a,8(and 1,3a,8)-di(and tri)methylpyrrolo[2,3-b]indoles
US4931447A (en) 1987-06-15 1990-06-05 Eli Lilly And Company Cycloalkylamides of (8β)-1-alkyl-6-(substituted) ergolines
FI95572C (en) 1987-06-22 1996-02-26 Eisai Co Ltd Process for the preparation of a medicament useful as a piperidine derivative or its pharmaceutical salt
JPS6422883A (en) 1987-07-17 1989-01-25 Sanwa Kagaku Kenkyusho Co 1-pyrrolidine acetamide derivative, its salt, production thereof and prophylactic and remedial agent for cereral dysfunction containing said derivative and salt as active ingredient
ATE68346T1 (en) 1987-07-22 1991-11-15 Farvalsa Ag MOISTURE-STABLE SOLID VALPROIC ACID PREPARATION AND PROCESS FOR THEIR PRODUCTION.
ZA886585B (en) 1987-09-08 1990-05-30 Lilly Co Eli Specific 5-ht,antagonists
US5364863A (en) 1987-09-08 1994-11-15 Eli Lilly And Company Specific 5-HT3 antagonists
US4880930A (en) 1987-11-30 1989-11-14 New James S Psychotropic acyclic amide derivatives
US4891375A (en) 1988-01-13 1990-01-02 Pfizer Inc. Arylpiperazinyl-alkylene-phenyl-heterocyclic compounds
US4883795A (en) 1988-01-22 1989-11-28 Pfizer Inc. Piperazinyl-heterocyclic compounds
US4831031A (en) 1988-01-22 1989-05-16 Pfizer Inc. Aryl piperazinyl-(C2 or C4) alkylene heterocyclic compounds having neuroleptic activity
US5001130A (en) 1988-02-18 1991-03-19 Bristol-Myers Company Psychotropic heterobicycloalkylpiperazine derivatives
US5238959A (en) 1988-04-08 1993-08-24 Eli Lilly And Company 3-phenyloxy-3-phenyl propanamines
US5242911A (en) 1988-06-17 1993-09-07 Pfizer Inc. Bridged bicyclic imides as anxiolytics and antidepressants
US5077295A (en) 1988-09-16 1991-12-31 Pfizer Inc. Antipsychoic 4-(4-(3-benzisothiazolyl)-1-piperazinyl)buytl bridged bicycle imides
US5006528A (en) 1988-10-31 1991-04-09 Otsuka Pharmaceutical Co., Ltd. Carbostyril derivatives
CA2000786C (en) 1988-11-07 1999-01-26 Cornelus G. M. Janssen 3-piperidinyl-1,2-benzisoxazoles
US5158952A (en) 1988-11-07 1992-10-27 Janssen Pharmaceutica N.V. 3-[2-[4-(6-fluoro-1,2-benzisoxozol-3-yl)-1-piperidinyl]ethyl]-6,7,8,9 tetrahydro-9-hydroxy-2-methyl-4H-pyrido [1,2-a]pyrimidin-4-one, compositions and method of use
US5254556A (en) 1988-11-07 1993-10-19 Janssen Pharmaceutica N.V. 3-piperidinyl-1,2-benzisoxazoles
MX18467A (en) 1988-11-23 1993-07-01 Pfizer THERAPEUTIC AGENTS OF QUINUCLIDINES
WO1990006303A1 (en) 1988-12-02 1990-06-14 Pfizer Inc. Arylpiperidine derivatives
US4950658A (en) 1988-12-06 1990-08-21 Board Of Trustees Of Southern Illinois Univ. Method of medical treatment of Alzheimer's disease
HUT53607A (en) 1989-01-17 1990-11-28 Hoffmann La Roche Process for production of derivatives of cyclohexan-acetamid
WO1990007926A1 (en) 1989-01-20 1990-07-26 Pfizer Inc. 3-(1,2,5,6-tetrahydropyridyl)-pyrrolopyridines
FR2643556B1 (en) 1989-02-27 1993-03-05 Sanofi Sa PHARMACEUTICAL COMPOSITION WITH SUSTAINED RELEASE OF VALPROIC ACID
US4981870A (en) 1989-03-07 1991-01-01 Pfizer Inc. Use of 4-phenyl-1,2,3,4-tetrahydro-1-naphthalenamine derivatives in the treatment of psychosis, inflammation and as immunosuppressants
US5238945A (en) 1989-04-11 1993-08-24 H. Lundbeck A/S Method of treating psychoses
GB8908085D0 (en) 1989-04-11 1989-05-24 Lundbeck & Co As H New therapeutic use
US4914207A (en) 1989-05-09 1990-04-03 Pfizer Inc. Arylthiazolylimidazoles
US4963689A (en) 1989-05-19 1990-10-16 Pfizer Inc. Heterocyclicguanidines as 5HT3 antagonists
US5364866A (en) 1989-05-19 1994-11-15 Hoechst-Roussel Pharmaceuticals, Inc. Heteroarylpiperidines, pyrrolidines and piperazines and their use as antipsychotics and analetics
ES2076253T3 (en) 1989-05-19 1995-11-01 Hoechst Roussel Pharma N- (ARYLOXYALKYL) -HETEROARILPIPERIDINES AND -HETEROARILPIPERAZINAS, A PROCEDURE FOR ITS PREPARATION AND USE AS MEDICINES.
US5693668A (en) 1989-06-22 1997-12-02 Merrell Pharmaceuticals Inc. Acetylcholinesterase inhibitors
EP0403713A1 (en) 1989-06-22 1990-12-27 Merrell Dow Pharmaceuticals Inc. Novel acetylcholinesterase inhibitors
US5350747A (en) 1989-07-07 1994-09-27 Pfizer Inc Heteroaryl piperazine antipsychotic agents
WO1991000863A1 (en) 1989-07-07 1991-01-24 Pfizer Inc. Heteroaryl piperazine antipsychotic agents
IT1231477B (en) 1989-07-12 1991-12-07 Sigma Tau Ind Farmaceuti (PIRROLIDIN-2-ONE-1-IL) ACETAMIDES AS ACTIVATORS OF LEARNING PROCESSES AND MEMORY AND PHARMACEUTICAL COMPOSITIONS INCLUDING SUCH COMPOUNDS
US4956368A (en) 1989-07-24 1990-09-11 Bristol-Myers Company Metabolites and prodrug formulations of 8-[4-[4-(1,2-benzisothiazol-3-yl)-1-piperazinyl]butyl]-8-azaspiro[4.5]decane-7,9-dione
US5006525A (en) 1989-07-24 1991-04-09 Eli Lilly And Company Dopamine agonists method
US4999430A (en) 1989-07-31 1991-03-12 Warner-Lambert Company Derivatives of 1,2,3,4-tetrahydro-9-acrisinamine
DE3927049A1 (en) 1989-08-16 1991-02-21 Sandoz Ag HALOGENALKYL-PHENYL-KETONE AND THEIR HYDRATES, THEIR PRODUCTION AND USE
WO1991003467A1 (en) 1989-08-30 1991-03-21 Pfizer Inc. Benzazabicyclic carbamates as novel cholinesterase inhibitors
US4914102A (en) 1989-09-28 1990-04-03 Hoechst Roussel Pharmaceuticals, Inc. N-aminocarbamates related to physostigmine, pharmacentical compositions and use
US5011841B1 (en) 1989-11-14 1994-09-06 Pfizer Treatment of depression
WO1991009844A1 (en) 1990-01-04 1991-07-11 Pfizer Inc. Substance p antagonists
US5232929A (en) 1990-11-28 1993-08-03 Pfizer Inc. 3-aminopiperidine derivatives and related nitrogen containing heterocycles and pharmaceutical compositions and use
NZ237241A (en) 1990-03-02 1993-11-25 Pharmetrix Corp Method for increasing the storage stability of physostigmine
US5043341A (en) 1990-04-11 1991-08-27 Eli Lilly And Company N-(2-hydroxycyclopentyl)-1-isopropyl-6-methylergoline-8-carboxamides
US5229382A (en) 1990-04-25 1993-07-20 Lilly Industries Limited 2-methyl-thieno-benzodiazepine
DK0532527T3 (en) 1990-06-01 1995-01-02 Pfizer 3-amino-2-arylquinuclidines, process for their preparation and pharmaceutical compositions containing them
JP2807577B2 (en) 1990-06-15 1998-10-08 エーザイ株式会社 Cyclic amide derivative
JP2800953B2 (en) 1990-07-06 1998-09-21 住友製薬株式会社 New imide derivatives
US5399565A (en) 1990-07-17 1995-03-21 Eli Lilly And Company Pyrazolidinone CCK and gastrin antagonists and pharmaceutical formulations therof
EP0540526B1 (en) 1990-07-23 1994-12-28 Pfizer Inc. Quinuclidine derivatives
US5102891A (en) 1990-07-23 1992-04-07 Hoechst-Roussel Pharmaceuticals Inc. 1-(substituted pyridinylamino)-1H-indol-5-yl substituted carbamates
US5264442A (en) 1990-08-13 1993-11-23 Hoechst-Roussel Pharmaceuticals Incorporated Carbamoyl-1-(pyridinylalkyl)-1H-indoles, indolines and related analogs
TW200462B (en) 1990-09-27 1993-02-21 Hoechst Roussel Pharma
JPH0772175B2 (en) 1990-09-28 1995-08-02 フアイザー・インコーポレイテツド Fused ring analogs of nitrogen-containing non-aromatic heterocycles
US5559129A (en) 1990-10-15 1996-09-24 Pfizer Inc Indole derivatives
EP0592438B1 (en) 1990-10-15 1997-08-27 Pfizer Inc. Indole derivatives
US5578612A (en) 1990-10-15 1996-11-26 Pfizer Inc. Indole derivatives
US5190951A (en) 1990-10-19 1993-03-02 Ss Pharmaceutical Co., Ltd. Quinoline derivatives
US5147881A (en) 1990-11-14 1992-09-15 Pfizer Inc 4-(1,2-benzisoxazolyl)piperidine antipsychotic agents
JP2965675B2 (en) 1990-11-21 1999-10-18 エーザイ株式会社 Method for producing (-)-1-benzyl-4-[(5,6-dimethoxy-1-indanone) -2-yl] methylpiperidine
TW197435B (en) 1990-11-22 1993-01-01 Takeda Pharm Industry Co Ltd
JPH04327532A (en) 1991-01-31 1992-11-17 Bristol Myers Squibb Co Medicinal preparation used for treating attensiveness defficiency disease accompanied by hyperactivity
US5668117A (en) 1991-02-22 1997-09-16 Shapiro; Howard K. Methods of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with previously known medicaments
US5157034A (en) 1991-02-27 1992-10-20 Pfizer Inc. Neuroleptic perhydro-1H-pyrido[1,2-a]pyrazines
DK0573522T3 (en) 1991-03-01 1995-03-06 Pfizer 1-azabicyclo (3.2.2) nonan-3-amine derivatives
US5334720A (en) 1991-03-07 1994-08-02 Fisons Corporation Diphenyl-1-(aminoalkyl)-2-piperidinone and -2-pyrrolidinone derivatives having anticonvulsant properties
ES2188584T3 (en) 1991-03-28 2003-07-01 Eisai Co Ltd AMINA HETEROCICLICAL DERIVATIVES.
US5750542A (en) 1993-09-28 1998-05-12 Pfizer Benzisoxazole and benzisothizole derivatives as cholinesterase inhibitors
US5627178A (en) 1991-04-23 1997-05-06 Lilly Industries Limited 2-methyl-thieno-benzodiazepine
US5817655A (en) 1991-04-23 1998-10-06 Eli Lilly And Company Methods of treatment using a thieno-benzodiazepine
US5817656A (en) 1991-04-23 1998-10-06 Eli Lilly And Company Mental disorders
US5104880A (en) 1991-05-01 1992-04-14 Mayo Foundation For Medical Education And Research Huperzine a analogs as acetylcholinesterase inhibitors
CA2067614C (en) 1991-05-02 2002-07-30 Eiichi Otomo Agent for improving dementia
US5716965A (en) 1991-05-22 1998-02-10 Pfizer Inc. Substituted 3-aminoquinuclidines
FR2677019B1 (en) 1991-05-27 1994-11-25 Pf Medicament NOVEL PIPERIDINES DISUBSTITUEES-1,4, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION.
DE122006000066I2 (en) 1991-05-31 2007-12-06 Pfizer quinuclidine derivatives
US5106856A (en) 1991-06-07 1992-04-21 Hoechst-Roussel Pharmaceuticals Inc. [(Arylalkylpiperidin-4-yl)methyl]-2a,3,4,5-tetrahydro-1(2H)-acenaphthylen-1-ones and related compounds
RU2114848C1 (en) 1991-06-20 1998-07-10 Пфайзер Инк. Fluoroalkoxybenzylamine derivatives of nitrogen-containing heterocycles, pharmaceutical composition, method of inhibition of substance p effect in mammalian
TW202432B (en) 1991-06-21 1993-03-21 Pfizer
FR2679555B1 (en) 1991-07-25 1993-11-19 Fabre Medicament Pierre NEW DERIVATIVES OF UREAE, THEIR PREPARATION AND THEIR APPLICATION IN THERAPEUTICS.
AU665207B2 (en) 1991-07-29 1995-12-21 Warner-Lambert Company Quinazoline derivatives as acetylcholinesterase inhibitors
IT1251166B (en) 1991-08-09 1995-05-04 Chiesi Farma Spa GENESERINE DERIVATIVES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US5161533A (en) 1991-09-19 1992-11-10 Xomed-Treace Inc. Break-apart needle electrode system for monitoring facial EMG
US5246947A (en) 1991-09-23 1993-09-21 Hoechst-Roussel Pharmaceuticals Incorporated Substituted pyridinylamino-1,2-benzisothiazoles and their use for treating depression
CA2118936A1 (en) 1991-09-25 1993-04-01 Gene M. Bright Neuroleptic 2-substituted perhydro-1-h-pyrido[1,2-a]pyrazines
DK0607164T3 (en) 1991-09-26 2002-06-17 Pfizer Condensed tricyclic nitrogen-containing heterocyclic compounds as substance P receptor antagonists
US5231093A (en) 1991-10-01 1993-07-27 Hoechst-Roussel Pharmaceuticals Incorporated Carbamate derivatives of 4-amino-3-isoxazolidinones, 3-amino-1-hydroxypyrrolidin-2-ones and 1-amino-1-cyclopropanecarboxylic acid analogs
TW263504B (en) 1991-10-03 1995-11-21 Pfizer
EP0537993A1 (en) 1991-10-17 1993-04-21 Eli Lilly And Company Nicotinic activity of a series of arecolones and isoarecolones
US5710168A (en) 1991-10-23 1998-01-20 Pfizer Inc. 2-piperidino-1-alkanol derivatives as neuroprotective agents
AU2681392A (en) 1991-11-12 1993-06-15 Pfizer Inc. Acyclic ethylenediamine derivatives as substance p receptor antagonists
DK0619805T3 (en) 1991-11-25 2000-06-05 Pfizer 5- (Hetero- or carbocyclylamino) -indole derivatives, their preparation and use as 5-HT1 agonists
US5137894A (en) 1991-12-05 1992-08-11 New James S 4-(4-Piperidinyl-thieno[3,2-c]pyridine derivatives of n-alkylglutarimides
ES2042384B1 (en) 1991-12-26 1994-06-01 Boehringer Ingelheim Espana PROCEDURE FOR OBTAINING BIS-PYRIDINE DERIVATIVES.
EP0619814A1 (en) 1991-12-31 1994-10-19 Fujisawa Pharmaceutical Co., Ltd. Oxadiazole derivatives having acetylcholinesterase-inhibitory and muscarinic agonist activity
WO1993016690A1 (en) 1992-02-25 1993-09-02 Warner-Lambert Company Cytoprotective compositions containing pyruvate and antioxidants
TW288010B (en) 1992-03-05 1996-10-11 Pfizer
US5569662A (en) 1992-03-23 1996-10-29 Pfizer Inc. Quinuclidine derivatives as substance P antagonists
RU2101283C1 (en) 1992-04-07 1998-01-10 Пфайзер Инк. Indole derivatives or their pharmaceutically acceptable salts
US6380233B1 (en) 1992-04-07 2002-04-30 Pfizer Inc Indole derivatives as 5-HT1 agonists
ATE148465T1 (en) 1992-04-10 1997-02-15 Pfizer ACYLAMINOINDOL DERIVATIVES AS 5-HT1 AGONISTS
US5439930A (en) 1992-04-14 1995-08-08 Russian-American Institute For New Drug Development Biologically active n-acylprolydipeptides having antiamnestic, antihypoxic and anorexigenic effects
US5256664A (en) 1992-04-28 1993-10-26 Bristol-Myers Squibb Company Antidepressant 3-halophenylpiperazinylpropyl derivatives of substituted triazolones and triazoldiones
US20050256162A1 (en) 1992-05-05 2005-11-17 Pfizer Inc Fluoroalkoxybenzylamino derivatives of nitrogen containing heterocycles
CA2134964C (en) 1992-05-18 1997-12-30 Manoj C. Desai Bridged aza-bicyclic derivatives as substance p antagonists
IT1254996B (en) 1992-06-25 1995-10-11 Mediolanum Farmaceutici Srl AMINO ALKYLCARBAMMIC ESTERS OF ESEROLINE ACTS FOR USE AS ANTICOLINESTERASICS AND RELATED PREPARATION PROCEDURE
US5688804A (en) 1992-08-04 1997-11-18 Pfizer Inc. 3-Benzylamino-2-phenyl-piperidine derivatives as substance P receptor antagonists
EP0655996B1 (en) 1992-08-19 2001-11-07 Pfizer Inc. Substituted benzylamino nitrogen containing non-aromatic heterocycles
US5312925A (en) 1992-09-01 1994-05-17 Pfizer Inc. Monohydrate of 5-(2-(4-(1,2-benzisothiazol-3-yl)-1-piperazinyl)-ethyl)-6-chloro-1,3-dihydro-2H-indol-2-one-hydrochloride
US5440023A (en) 1992-09-18 1995-08-08 Beckman Instruments, Inc. Method for making valproic acid derivatives
US5604241A (en) 1992-10-21 1997-02-18 Pfizer Inc. Substituted benzylaminoquinuclidines as substance P antagonists
US5837711A (en) 1992-10-28 1998-11-17 Pfizer Inc. Substituted quinuclidines as substance P antagonists
TW251284B (en) 1992-11-02 1995-07-11 Pfizer
US5498610A (en) 1992-11-06 1996-03-12 Pfizer Inc. Neuroprotective indolone and related derivatives
AU676489B2 (en) 1992-11-12 1997-03-13 Pfizer Inc. Quinuclidine derivative as substance P antagonist
US6369074B1 (en) 1992-12-10 2002-04-09 Pfizer Inc. Aminomethylene substituted non-aromatic heterocycles and use as substance P antagonists
ATE194340T1 (en) 1992-12-10 2000-07-15 Pfizer AMINOMETHYLENE SUBSTITUTED HETEROCYCLIC COMPOUNDS AND THEIR USE AS SUBSTANCE PANTAGONISTS
US5352459A (en) 1992-12-16 1994-10-04 Sterling Winthrop Inc. Use of purified surface modifiers to prevent particle aggregation during sterilization
RU2124015C1 (en) 1992-12-17 1998-12-27 Пфайзер Инк. Derivatives of pyrrolo[2,3-d]pyrimidines, pharmaceutical composition, pyrrolo[2,3-d]pyrimidines, derivatives of pyrrole
TW444018B (en) 1992-12-17 2001-07-01 Pfizer Pyrazolopyrimidines
EP0611769A1 (en) 1993-02-16 1994-08-24 Merrell Dow Pharmaceuticals Inc. Silylated acetylcholinesterase inhibitors
AU4982293A (en) 1993-03-02 1994-09-26 Fujisawa Pharmaceutical Co., Ltd. Novel heterocyclic compound
ATE166650T1 (en) 1993-03-04 1998-06-15 Pfizer SPIROAZACYCLIC DERIVATIVES AS SUBSTANCE P ANTAGONISTS
AU6391894A (en) 1993-03-16 1994-10-11 Pfizer Inc. Naphthalene derivatives
IL109646A0 (en) 1993-05-19 1994-08-26 Pfizer Heteroatom substituted alkyl benzylamino-quinuclidines
EP0627400A1 (en) 1993-06-04 1994-12-07 Merrell Dow Pharmaceuticals Inc. Aromatic acetylcholinesterase inhibitors
SE9302080D0 (en) 1993-06-16 1993-06-16 Ab Astra NEW COMPOUNDS
US5869499A (en) 1993-07-15 1999-02-09 Pfizer Inc Benzyloxyquinuclidines as substance P antagonists
JP2860603B2 (en) 1993-08-31 1999-02-24 ファイザー・インク. 5-arylindole derivatives
US6083943A (en) 1993-09-17 2000-07-04 Pfizer Inc Substituted azaheterocyclecarboxylic acid
EP1209157A1 (en) 1993-09-17 2002-05-29 Pfizer Inc. Heteroarylamino and heteroarylsulfonamido substituted 3-benzylaminomethyl piperidines and related compounds
GB9319732D0 (en) 1993-09-24 1993-11-10 Ucb Sa Use of (s)-alpha-ethyl-2-oxo-l-pyrrolidineacetamide for the treatment of anxiety
US5468733A (en) 1993-09-30 1995-11-21 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
KR100197454B1 (en) 1993-12-29 1999-06-15 디. 제이. 우드, 스피겔 알렌 제이 Diazabicyclic neurokinin antagonists
DE122010000050I2 (en) 1994-03-02 2011-07-21 Organon Nv Sublingual or buccal medicine.
DE69533284T2 (en) 1994-04-29 2005-07-21 Pfizer Inc. NEW CYCLIC AND ACYCLIC AMIDS FOR INCREASING NEUROTRANSMITTER DISTRIBUTION
PL181895B1 (en) 1994-06-16 2001-10-31 Pfizer Pyrazole and pyrrole pyridines
US5744476A (en) 1994-06-27 1998-04-28 Interneuron Pharmaceuticals, Inc. Dopamine D1 agonists for the treatment of dementia
EP0767782B1 (en) 1994-06-29 2001-10-31 Pfizer Inc. Aryl and heteroaryl alkoxynaphthalene derivatives
CA2194984C (en) 1994-07-26 2002-07-02 John Eugene Macor 4-indole derivatives as serotonin agonists and antagonists
ES2201106T3 (en) 1994-08-18 2004-03-16 Pfizer Inc. NEUROPROTECTORS DERIVATIVES OF 3- (PIPERIDINIL-1) -CROMAN-4,7-DIOL AND 1- (4-HYDROPHENYL) -2- (PIPERIDINIL-1) -ALCANOL.
US5597826A (en) 1994-09-14 1997-01-28 Pfizer Inc. Compositions containing sertraline and a 5-HT1D receptor agonist or antagonist
US5627200A (en) 1994-09-26 1997-05-06 Pfizer Inc β3 -Adrenoceptor agonists and antagonists for the treatment of intestinal motility disorders, depression, prostate disease and dyslipidemia
WO1996010570A1 (en) 1994-09-30 1996-04-11 Pfizer Inc. NEUROLEPTIC 2,7-DISUBSTITUTED PERHYDRO-1H-PYRIDO[1,2-a]PYRAZINES
US5516759A (en) 1994-12-08 1996-05-14 Tap Holdings Inc. LHRH antagonists having lactam groups at the N-terminus
US5658590A (en) 1995-01-11 1997-08-19 Eli Lilly And Company Treatment of attention-deficit/hyperactivity disorder
US5741789A (en) 1995-01-17 1998-04-21 Eli Lilly And Company Compounds having effects on serotonin-related systems
US5576321A (en) 1995-01-17 1996-11-19 Eli Lilly And Company Compounds having effects on serotonin-related systems
WO1996024353A1 (en) 1995-02-10 1996-08-15 Eli Lilly And Company Methods of treating or preventing psychiatric disorders
EG23659A (en) 1995-03-24 2007-03-26 Lilly Co Eli Process and crystal forms of methyl-thieno-benzodiazepine
CR5278A (en) 1995-03-24 1996-07-04 Lilly Co Eli ORAL FORMULATION OF 2-METHYL-THENO-BENZODIACEPINE
JPH11503757A (en) 1995-04-18 1999-03-30 イーライ・リリー・アンド・カンパニー Use of ergoline compounds that produce physiological and pathological functions at 5-HT (7) receptors
US5889010A (en) 1995-05-18 1999-03-30 Pfizer Inc. Benzimidazole derivatives having dopaminergic activity
US20010023248A1 (en) 1995-05-25 2001-09-20 Howard Harry R. Aminomethylene substituted non-aromatic heterocycles and use as substance P antagonists
US6403599B1 (en) 1995-11-08 2002-06-11 Pfizer Inc Corticotropin releasing factor antagonists
IL118279A (en) 1995-06-07 2006-10-05 Abbott Lab 3 - pyridyloxy (or thio) alkyl heterocyclic compounds, pharmaceutical compositions containing them and their uses in the preparation of medicaments for controlling chemical synaptic transmission
ATE199552T1 (en) 1995-07-07 2001-03-15 Pfizer SUBSTITUTED BENZOLACTAM COMPOUNDS AS SUBSTANCE P ANTAGONISTS
US5696168A (en) 1995-07-24 1997-12-09 Eli Lilly And Company Treatment of attention-deficit/hyperactivity disorder
CA2227410A1 (en) 1995-07-24 1997-02-06 Eli Lilly And Company Treatment of attention-deficit/hyperactivity disorder
TW340842B (en) 1995-08-24 1998-09-21 Pfizer Substituted benzylaminopiperidine compounds
IL115113A (en) 1995-08-31 2002-11-10 Israel State 3-carbamoyloxy pyridinium derivatives and pharmaceutical compositions containing them
ES2100129B1 (en) 1995-10-11 1998-02-16 Medichem Sa NEW POLYCLIC AMINOPYRIDINE COMPOUNDS ACETYLCHOLINESTERASE INHIBITORS, PROCEDURE FOR THE PREPARATION AND USE.
WO1997019059A1 (en) 1995-11-17 1997-05-29 Sibia Neurosciences, Inc. Novel substituted aryl compounds useful as modulators of acetylcholine receptors
US5783584A (en) 1995-12-11 1998-07-21 Mayo Foundation For Medical Education And Research THA analogs useful as cholinesterase inhibitors
PT780375E (en) 1995-12-21 2002-12-31 Pfizer 3-¬ (BENZYL SUBSTITUTED IN 5) AMINO | -2-PHENYLPIPERIDINES AS ANTAGONISTS OF THE SUBSTANCE P
IL125031A0 (en) 1995-12-22 1999-01-26 Lilly Co Eli Method for treating depression
US5625897A (en) 1996-01-11 1997-05-06 Park; Kelly R. Upper torso garment
WO1997025983A1 (en) 1996-01-22 1997-07-24 Eli Lilly And Company Indane derivatives for antipsychotic compositions
GB9606736D0 (en) 1996-02-19 1996-06-05 Shire International Licensing Therapeutic method
PL328925A1 (en) 1996-03-11 1999-03-01 Lilly Co Eli Method of treating bipolar disorders
CA2250042A1 (en) 1996-03-25 1997-10-02 Eli Lilly And Company Treating pain using a synergistic combination of an atypical antipsychotic and a drug used in treatment of pain
US6107300A (en) 1996-03-27 2000-08-22 Dupont Pharmaceuticals Arylamino fused pyrimidines
US6326368B1 (en) 1996-03-27 2001-12-04 Dupont Pharmaceuticals Company Aryloxy- and arylthiosubstituted pyrimidines and triazines and derivatives thereof
JP3118467B2 (en) 1996-03-29 2000-12-18 ファイザー インク. Benzyl (idene) -lactam derivatives as selective (ant) agonists of 5-HT1A and / or 5-HT1D receptors, their preparation and use
DE69728138T2 (en) 1996-03-29 2004-09-16 Pfizer Inc. 6-phenylpyridine
DE69707659T2 (en) 1996-04-12 2002-05-08 Aventis Pharma Inc ISATINE DERIVATIVES AS ACETYLCHOLINESTERASE INHIBITORS AND ANALGETICS
TW491847B (en) 1996-05-07 2002-06-21 Pfizer Mesylate dihydrate salts of 5-(2-(4-(1,2-benzisothiazol-3-yl)-1-piperazinyl)-ethyl)-6-chloro-1,3-dihydro-2h-indol-2-one
IL126590A (en) 1996-05-07 2001-11-25 Pfizer Mesylate trihydrates salt of 5-(2-(4-(1, 2-benzisothiazol-3-yl)-1-piperazinyl) ethyl)-6-chloro-1, 3-dihydro-2(1h)-indol-2-one (=ziprasidone) and pharmaceutical compositions comprising it
TW487572B (en) 1996-05-20 2002-05-21 Janssen Pharmaceutica Nv Aqueous suspensions of 9-hydroxyrisperidone fatty acid esters
EP1380298A3 (en) 1996-05-23 2004-04-07 Bristol-Myers Squibb Pharma Company Tetrahydropteridines and pyridylpiperazines for treatment of neurological disorders, anorexia, inflammation
US5948437A (en) 1996-05-23 1999-09-07 Zeneca Limited Pharmaceutical compositions using thiazepine
ZA973884B (en) 1996-05-23 1998-11-06 Du Pont Merck Pharma Tetrahydropteridines and pyridylpiperazines for treatment of neurological disorders
ATE210649T1 (en) 1996-05-28 2001-12-15 Pfizer ARYLACRYLAMIDE DERIVATIVES AS 5HT1 AGONISTS OR ANTAGONISTS
WO1997046526A1 (en) 1996-06-07 1997-12-11 Eisai Co., Ltd. Stable polymorphs of donepezil (1-benzyl-4-[(5,6-dimethoxy-1-indanon)-2-yl]methylpiperidine) hydrochloride and process for production
TW513409B (en) 1996-06-07 2002-12-11 Eisai Co Ltd Polymorphs of donepezil hydrochloride
US6329396B1 (en) 1996-06-10 2001-12-11 Pfizer Inc. Substituted benzylaminopiperidine compounds
US5912256A (en) 1996-06-20 1999-06-15 Eli Lilly And Company Compounds having effects on serotonin-related systems
CA2180703A1 (en) 1996-07-08 1998-01-09 Paul Marie Victor Gilis Fast-dissolving galanthamine hydrobromide tablet
US7094782B1 (en) 1996-07-24 2006-08-22 Bristol-Myers Squibb Company Azolo triazines and pyrimidines
US6191131B1 (en) 1997-07-23 2001-02-20 Dupont Pharmaceuticals Company Azolo triazines and pyrimidines
EA006626B1 (en) 1996-07-24 2006-02-24 Дюпон Фармасьютикалз Компани Azolo pyrimidines, pharmaceutical composition and method for treatment
US6043258A (en) 1996-08-01 2000-03-28 Eli Lilly And Company Method for treating disruptive behavior disorders with xanomeline
PT821955E (en) 1996-08-01 2002-07-31 Lilly Co Eli Use of 3- (4-HEXYLXY-1,2,5-TIADIAZOL-3-YL) -1,2,5,6-TETRAHYDRO-1-METHYLPYRIDINE (XANOMELINE) FOR THE TREATMENT OF BIPOLAR DISEASE
US6117890A (en) 1996-08-01 2000-09-12 Eli Lilly And Company Method for treating bipolar disorder
MX9706196A (en) 1996-08-14 1998-02-28 Pfizer Piperidinylamino tricyclic compounds as substance p antagonists.
AU4078997A (en) 1996-08-22 1998-03-06 New York University Cholinesterase inhibitors for treatment of parkinson's disease
ZA977967B (en) 1996-09-23 1999-03-04 Lilly Co Eli Combination therapy for treatment of psychoses
AU720366B2 (en) 1996-09-23 2000-06-01 Eli Lilly And Company Olanzapine dihydrate D
US6423708B1 (en) 1996-09-30 2002-07-23 Pfizer Inc Aralkyl and aralkylidene heterocyclic lactams and imides
UA56185C2 (en) 1996-09-30 2003-05-15 Пфайзер Інк. Aralkyl- and aralkylidene heterocyclic lactams and imids, a pharmaceutical composition and a treatment method
IT1285801B1 (en) 1996-10-10 1998-06-24 Sigma Tau Ind Farmaceuti IMPROVED PROCEDURE FOR THE PREPARATION OF VALPROIC ACID
US6235734B1 (en) 1996-10-30 2001-05-22 Pfizer Inc Pyridone-fused azabicyclic- or cytisine derivatives, their preparation and their use in addiction therapy
WO1998030243A1 (en) 1997-01-08 1998-07-16 Warner-Lambert Company Acetylcholinesterase inhibitors in combination with muscarinic agonists for the treatment of alzheimer's disease
ES2198598T3 (en) 1997-01-29 2004-02-01 Pfizer Inc. DERIVATIVES OF SULFONILUREA AND ITS USE IN THE CONTROL OF THE ACTIVITY OF INTERLEUQUINA-1.
US6020335A (en) 1997-02-06 2000-02-01 Pfizer Inc (N-(pyridinylmethyl)-heterocyclic)ylideneamine compounds as nicotinic acetylcholine receptor binding agents
ATE241358T1 (en) 1997-03-03 2003-06-15 Eisai Co Ltd USE OF CHOLINESTERASE INHIBITORS TO TREAT CONCENTRATION DISORDERS
GB2323788B (en) 1997-04-02 1999-02-17 Bristol Myers Squibb Co Venting valve for ostomy bag
ID22781A (en) 1997-04-04 1999-12-09 Pfizer Prod Inc Nicotynamide derivatives
ZA982877B (en) 1997-04-09 1999-10-04 Lilly Co Eli Treatment of central nervous system disorders with selective estrogen receptor modulators.
WO1998046225A1 (en) 1997-04-11 1998-10-22 Eli Lilly And Company Method for treating schizophrenia
EP1009403A4 (en) 1997-04-11 2002-08-28 Lilly Co Eli Method for treating schizophrenia
US5910319A (en) 1997-05-29 1999-06-08 Eli Lilly And Company Fluoxetine enteric pellets and methods for their preparation and use
US6060479A (en) 1997-06-09 2000-05-09 Pfizer Inc Quinazoline-4-one AMPA antagonists
ATE303997T1 (en) 1997-06-09 2005-09-15 Pfizer Prod Inc CHINAZOLIN-4-ON AMPA ANTAGONISTS
US6627755B1 (en) 1997-06-09 2003-09-30 Pfizer Inc Quinazolin-4-one AMPA antagonists
JP2001510797A (en) 1997-07-22 2001-08-07 イーライ・リリー・アンド・カンパニー Pharmaceutical compounds
GB9716879D0 (en) 1997-08-08 1997-10-15 Shire Int Licensing Bv Treatment of attention deficit disorders
AU8742198A (en) 1997-08-15 1999-03-08 Shire International Licensing B.V. Use of cholinesterase inhibitor for treating diseases associated with pro teolytic enzyme activity
US6479523B1 (en) 1997-08-26 2002-11-12 Emory University Pharmacologic drug combination in vagal-induced asystole
IL125951A (en) 1997-09-05 2003-09-17 Pfizer Prod Inc A pharmaceutical composition comprising a piperazinyl-heterocyclic compound for treating tourette's syndrome, obsessive compulsive disorder, chronic motor or vocal tic disorder in a mammal
US6323208B1 (en) 1997-09-05 2001-11-27 Pfizer Inc Atropisomers of 2,3-disubstituted-(5.6)-heteroaryl fused-pyrimidin-4-ones
AU9214498A (en) 1997-09-23 1999-04-12 Eli Lilly And Company Treatment of attention-deficit/hyperactivity disorder
UA72189C2 (en) 1997-11-17 2005-02-15 Янссен Фармацевтика Н.В. Aqueous suspensions of 9-hydroxy-risperidone fatty acid esters provided in submicron form
EP1048653B1 (en) 1997-12-05 2004-03-03 Eisai Co., Ltd. Donepezil polycrystals and process for producing the same
US6545022B1 (en) 1997-12-16 2003-04-08 Pfizer Inc. 4(3)substituted-4(3)-aminomethyl-(thio)pyran or piperidine derivatives (=gabapentin analogues), their preparation and their use in the treatment of neurological disorders
IL127497A (en) 1997-12-18 2002-07-25 Pfizer Prod Inc Pharmaceutical compositions containing piperazinyl-heterocyclic compounds for treating psychiatric disorders
CN1137123C (en) 1998-01-28 2004-02-04 杜邦药品公司 Azolo triazizes and pyrimidines
US6131106A (en) 1998-01-30 2000-10-10 Sun Microsystems Inc System and method for floating-point computation for numbers in delimited floating point representation
AU2495399A (en) 1998-02-06 1999-08-23 Eli Lilly And Company Desert hedgehog related nucleic acids and proteins
GB9805561D0 (en) 1998-03-16 1998-05-13 Merck Sharp & Dohme A combination of therapeutic agents
PA8469101A1 (en) 1998-04-09 2000-09-29 Pfizer Prod Inc AZABICICLIC LEAGUES OF RECEIVERS 5HT1
PA8469501A1 (en) 1998-04-10 2000-09-29 Pfizer Prod Inc HYDROXAMIDES OF THE ACID (4-ARILSULFONILAMINO) -TETRAHIDROPIRAN-4-CARBOXILICO
EP0957099B1 (en) 1998-04-15 2002-11-20 Pfizer Products Inc. Heterocyclic carboxamides
DE69919436T2 (en) 1998-04-16 2005-09-15 Pfizer Products Inc., Groton N-acyl and N-aroyl aralkylamides
EP1077704A4 (en) 1998-05-21 2002-01-30 Lilly Co Eli Combination therapy for treatment of depression
US6960577B2 (en) 1998-05-22 2005-11-01 Eli Lilly And Company Combination therapy for treatment of refractory depression
GT199900060A (en) 1998-05-22 2000-10-14 POLYMORPHES OF A CRYSTALLINE PYRAZINE MONOCLORHYDRATE AND ITS PHARMACEUTICAL COMPOSITIONS.
KR20010043730A (en) 1998-05-22 2001-05-25 피터 지. 스트링거 Combination Therapy for Treatment of Refractory Depression
WO1999062522A1 (en) 1998-05-29 1999-12-09 Eli Lilly And Company Combination therapy for treatment of bipolar disorders
US6150366A (en) 1998-06-15 2000-11-21 Pfizer Inc. Ziprasidone formulations
US6436914B1 (en) 1998-06-30 2002-08-20 Bristol-Myers Squibb Company 2-hydroxy-3—(4-hydroxy-3-sulfonamidophenyl)—propylamines useful as beta 3 adrenergic agonists
US6312717B1 (en) 1998-07-07 2001-11-06 Bristol-Myers Squibb Company Method for treatment of anxiety and depression
US6262081B1 (en) 1998-07-10 2001-07-17 Dupont Pharmaceuticals Company Composition for and method of treating neurological disorders
US6218383B1 (en) 1998-08-07 2001-04-17 Targacept, Inc. Pharmaceutical compositions for the prevention and treatment of central nervous system disorders
BR9912902A (en) 1998-08-11 2001-05-08 Pfizer Prod Inc 1,8-naphthyridin-4 (1h) -ones substituted as phosphodiesterase 4 inhibitors
IL125809A (en) 1998-08-17 2005-08-31 Finetech Lab Ltd Process and intermediates for production of donepezil and related compounds
IT1304904B1 (en) 1998-09-11 2001-04-05 Eisai Co Ltd ANTICOLINESTERASIC DERIVATIVES FOR THE TREATMENT OF FUNCTIONAL AND / OR ORGANIC SYNDROME
HN1999000146A (en) 1998-09-21 2000-11-11 Pfizer Prod Inc PHARMACEUTICAL AGENTS FOR THE TREATMENT OF PARKINSON'S DISEASE, ADHD AND MICROADENOMAS.
CA2345767A1 (en) 1998-10-16 2000-04-27 Paul Leonce Irma De Nijs Therapy for improving cognition
US5994352A (en) 1998-11-13 1999-11-30 Pfizer Inc. 5-arylindole derivatives
AU1738800A (en) 1998-11-23 2000-06-13 Bonnie Davis Dosage formulations for acetylcholinesterase inhibitors
WO2000031020A1 (en) 1998-11-25 2000-06-02 Warner-Lambert Company Improved gamma amino butyric acid analogs
ATE306264T1 (en) 1998-12-11 2005-10-15 Bonnie M Davis USE OF ACETYLCHOLINESTERASE INHIBITORS TO MODULATE THE HYPOTHALAMUS-PITOPHYSIS-GONAD AXIS
UA62015C2 (en) 1998-12-28 2003-12-15 Pfizer Prod Inc Benzoizoxazol derivatives, a pharmaceutical composition (variants) based thereon (variants) and a method for treatment (variants)
US6271380B1 (en) 1998-12-30 2001-08-07 Dupont Pharmaceuticals Company 1H-imidazo[4,5-d]pyridazin-7-ones, 3H-imidazo-[4,5-c]pyridin-4-ones and corresponding thiones as corticotropin releasing factor (CRF) receptor ligands
AU2180400A (en) 1999-01-13 2000-08-01 Eli Lilly And Company A pharmaceutical combination for the treatment of depression
DE60018225T2 (en) 1999-03-01 2005-12-29 Pfizer Products Inc., Groton Oxamic acids having a cyano group as ligands for the thyroid receptor
CA2363145C (en) 1999-03-01 2006-02-14 Pfizer Products Inc. Oxamic acids and derivatives as thyroid receptor ligands
US6277866B1 (en) 1999-03-03 2001-08-21 Eisai Co., Ltd. 1-benzyl-4[(5,6-dimethoxy-2-fluoro-1-indanon)-2-yl]methylpiperidine
DE69913138T2 (en) 1999-03-31 2004-08-26 Eisai Co., Ltd. STABILIZED COMPOSITION WITH NOOTROPIC ACTIVE SUBSTANCES
EP1050303A3 (en) 1999-04-27 2003-01-15 Pfizer Products Inc. Methods and compositions for treating age-related behavioral disorders in companion animals
OA11943A (en) 1999-05-21 2006-04-12 Pfizer Prod Inc New pharmacetical combinations for nos inhibitors
AU4973800A (en) 1999-06-09 2000-12-28 Eli Lilly And Company Combination for treating weight gain associated with antipsychotic use comprising an atypical antipsychotic and an h2 antagonist
DE60014370T2 (en) 1999-06-15 2006-02-09 Bristol-Myers Squibb Pharma Co., Wilmington SUBSTITUTED HETEROCYCLYL CONDENSED GAMMA CARBOLINE
US6713471B1 (en) 1999-06-15 2004-03-30 Bristol-Myers Squibb Pharma Company Substituted heterocycle fused gamma-carbolines
US6524616B1 (en) 1999-06-25 2003-02-25 Wake Forest University Health Services Compositions and methods for treating or preventing neurodegeneration and cognitive decline and dysfunction associated with alzheimer's disease, aging, other dementia related disorders and estrogen deficiency related conditions
US6432989B1 (en) 1999-08-27 2002-08-13 Pfizer Inc Use of CRF antagonists to treat circadian rhythm disorders
JP4242048B2 (en) 1999-09-01 2009-03-18 エーザイ・アール・アンド・ディー・マネジメント株式会社 4-substituted piperidine derivatives
DK1209151T3 (en) 1999-09-01 2007-08-20 Eisai R&D Man Co Ltd 4-SUBSTITUTED PIPERIDE INGREDIATES
JP2003509494A (en) 1999-09-22 2003-03-11 シェーリング コーポレイション Muscarinic antagonist
PT1088819E (en) 1999-09-30 2005-09-30 Pfizer Prod Inc 6-AZAURACYL DERIVATIVES AS LIGNED FROM TIROIDE RECEPTORS
CA2324813A1 (en) 1999-11-10 2001-05-10 Susan Beth Sobolov-Jaynes Combination treatment for depression and anxiety
US6620802B1 (en) 1999-11-23 2003-09-16 Corcept Therapeutics, Inc. Methods of treating mild cognitive impairment using a glucocorticoid-specific receptor antagonist
JP4249415B2 (en) * 1999-12-01 2009-04-02 ユセベ,ソシエテ アノニム Pyrrolidineacetamide derivatives alone or in combination for the treatment of CNS diseases
AU3264201A (en) 1999-12-17 2001-06-25 Du Pont Pharmaceuticals Company Imidazopyrimidinyl and imidazopyridinyl derivatives
AU3789901A (en) 1999-12-17 2001-06-25 Bristol-Myers Squibb Company Antipsychotic heterocycle compounds
DE60026581T2 (en) 1999-12-20 2006-12-28 Eli Lilly And Co., Indianapolis INDOLE DERIVATIVES FOR THE TREATMENT OF DEPRESSION AND SCARENESS
AU2049501A (en) 1999-12-20 2001-07-03 Eli Lilly And Company Benzofuran derivatives
US6844344B2 (en) 1999-12-20 2005-01-18 Eli Lilly And Company Benzofuran derivatives
AU1930901A (en) 1999-12-20 2001-07-03 Eli Lilly And Company Azabicyclo(3.2.1)octane derivatives
AU2251801A (en) 1999-12-20 2001-07-03 Eli Lilly And Company Piperidine derivatives and their use as serotonin receptor antagonists
ATE276249T1 (en) 1999-12-29 2004-10-15 Pfizer Prod Inc OPTICALLY ACTIVE 3-((2-PIPERAZINYL-PHENYL)METHYL)-1-(4-(TRIFLUORO METHYL)-PHENYL)-2-PYRROLIDINONE AS SELECTIVE 5-HT1D RECEPTOR ANTAGONISTS
EP1127882A1 (en) 2000-01-25 2001-08-29 Pfizer Products Inc. Tetrazole compounds as thyroid receptor ligands
CZ20022771A3 (en) 2000-02-17 2003-09-17 Bristol-Myers Squibb Company Aniline-derived ligands for the thyroid receptor
GB0004297D0 (en) 2000-02-23 2000-04-12 Ucb Sa 2-oxo-1 pyrrolidine derivatives process for preparing them and their uses
PT1311272E (en) 2000-03-03 2007-02-28 Eisai R&D Man Co Ltd Novel methods using cholinesterase inhibitors
WO2001066096A2 (en) 2000-03-06 2001-09-13 Immune Network Ltd. Compositions for prevention and treatment of dementia
US6911476B2 (en) 2000-03-13 2005-06-28 Eli Lilly And Company Sulfonamide derivatives
GB0007884D0 (en) 2000-03-31 2000-05-17 Pfizer Ltd Diphenyl ether compounds useful in therapy
US6664291B2 (en) 2000-03-31 2003-12-16 Pfizer, Inc. Malonamic acids and derivatives thereof as thyroid receptor ligands
EP1268404B1 (en) 2000-03-31 2008-01-02 Pfizer Products Inc. Malonamic acids and derivatives thereof as thyroid receptor ligands
EP1280798B1 (en) 2000-04-08 2007-01-03 Boehringer Ingelheim Pharma GmbH & Co.KG Bicyclic heterocylces, medicaments containing said compounds, the use thereof and method for producing them
ES2244599T3 (en) 2000-04-10 2005-12-16 Pfizer Products Inc. BENZAMIDO-PIPERIDINE COMPOUNDS AS SUBSTANCE ANTAGONISTS P.
JP4150519B2 (en) 2000-04-13 2008-09-17 エーザイ・アール・アンド・ディー・マネジメント株式会社 Acetylcholinesterase inhibitor comprising 1-benzylpyridinium salt
US6620830B2 (en) 2000-04-21 2003-09-16 Pfizer, Inc. Thyroid receptor ligands
US20010036949A1 (en) 2000-05-09 2001-11-01 Coe Jotham Wadsworth Pharmaceutical composition and method of treatment of diseases of cognitive dysfunction in a mammal
US6630469B2 (en) 2000-05-09 2003-10-07 Bristol-Myers Squibb Company 5-HT7 receptor antagonists
US20040229911A1 (en) 2000-05-17 2004-11-18 Pfizer Inc New pharmaceutical combinations for NOS inhibitors
EP1292311A2 (en) 2000-05-24 2003-03-19 Eli Lilly And Company Combination therapy for the treatment of depression comprising an antidepressant and an ampa receptor potentiator
US6893858B2 (en) 2000-05-26 2005-05-17 Bristol-Myers Squibb Company Human kcnq5 potassium channel, methods and compositions thereof
EP1698338B1 (en) 2000-06-02 2008-04-09 Pfizer Products Inc. S-methyl-dihydro-ziprasidone for the treatment of schizophrenia
US6395784B1 (en) 2000-06-07 2002-05-28 Bristol-Myers Squibb Company Benzamide ligands for the thyroid receptor
DOP2001000189A (en) 2000-06-30 2002-03-30 Pfizer Prod Inc BENZOPHENONS AND SULPHONES AS INHIBITORS OF GLYCINE CAPTURE
CA2411386A1 (en) 2000-06-30 2002-01-10 Eli Lilly And Company Combination therapy for treatment of psychoses
US6630476B2 (en) 2000-07-07 2003-10-07 Bristol-Myers Squibb Pharma Company Pyrrolo [3,4-d] pyrimidines as corticotropin releasing factor (CRF) antagonists
EP1177792A3 (en) 2000-07-27 2002-10-23 Pfizer Products Inc. Dopamine D4 Ligands for the treatment of novelty-seeking disorders
US6756385B2 (en) 2000-07-31 2004-06-29 Pfizer Inc. Imidazole derivatives
US20020016334A1 (en) 2000-07-31 2002-02-07 Coe Jotham Wadsworth Pharmaceutical composition for the treatment of attention deficit hyperactivity disorder (ADHD)
US20020049211A1 (en) 2000-09-06 2002-04-25 Sobolov-Jaynes Susan Beth Combination treatment for depression and anxiety
WO2002019998A2 (en) 2000-09-08 2002-03-14 Eli Lilly And Company A method of treating weight gain associated with atypical antipsychotic use
WO2002024200A1 (en) 2000-09-21 2002-03-28 Bristol-Myers Squibb Company Substituted azole derivatives as inhibitors of corticotropin releasing factor
EP1192952A3 (en) 2000-09-28 2003-03-26 Pfizer Products Inc. Combination, for treating depression and anxiety, containing an NK-3 receptor antagonist and a CNS penetrant NK-1 receptor antagonist
IL145584A0 (en) 2000-10-02 2002-06-30 Pfizer Prod Inc Nmda nr2b antagonists for treatment
US20020151591A1 (en) 2000-10-17 2002-10-17 Anabella Villalobos Combination use of acetylcholinesterase inhibitors and GABAa inverse agonists for the treatment of cognitive disorders
US20020103198A1 (en) 2000-12-04 2002-08-01 Fliri Anton F.J Acylamino cyclopropane derivatives
CA2364211A1 (en) 2000-12-05 2002-06-05 Phillip Branch Chappell Combination treatment for depression, anxiety and psychosis
UA73619C2 (en) 2000-12-13 2005-08-15 Pfizer Prod Inc Stable pharmaceutical compositions of nmda receptor agonist (variants) and method of treatment
AU2002248216B2 (en) 2000-12-20 2007-03-01 Bristol-Myers Squibb Pharma Company Pyrazinoquinoxaline derivatives as serotonin agonists and antagonists
US6849619B2 (en) 2000-12-20 2005-02-01 Bristol-Myers Squibb Company Substituted pyridoindoles as serotonin agonists and antagonists
IL156261A0 (en) 2000-12-20 2004-01-04 Bristol Myers Squibb Co Pyrroloquinoline and pyridoquinoline derivatives and pharmaceutical compositions containing the same
EP1343791A2 (en) 2000-12-20 2003-09-17 Bristol-Myers Squibb Company Substituted tetracyclic pyridoindoles as serotonin agonists and antagonists
JPWO2002053153A1 (en) 2000-12-28 2004-04-30 第一製薬株式会社 Drugs for the treatment and prevention of neuropathic pain
US20020086871A1 (en) 2000-12-29 2002-07-04 O'neill Brian Thomas Pharmaceutical composition for the treatment of CNS and other disorders
US6436938B1 (en) 2001-01-22 2002-08-20 Pfizer Inc. Combination treatment for depression
EE200300342A (en) 2001-01-26 2003-12-15 Bristol-Myers Squibb Company Imidazolyl Derivatives as Inhibitors of Corticotropin Releasing Factor, Methods of their Preparation and Use
US20020107244A1 (en) 2001-02-02 2002-08-08 Howard Harry R. Combination treatment for depression
US6610326B2 (en) 2001-02-16 2003-08-26 Andrx Corporation Divalproex sodium tablets
JP2004523557A (en) 2001-02-23 2004-08-05 ジョンズ・ホプキンス・ユニバーシティ Treatment of tics, tremors, and related disorders
US20020123490A1 (en) 2001-03-01 2002-09-05 Pfizer Inc. Combination treatment for anxiety, depression, obsessive compulsive disorder and psychosis
MXPA03008185A (en) 2001-03-13 2004-01-29 Bristol Myers Squibb Pharma Co 4-(2-butylamino)-2,7-dimethyl-8-(2-methyl-6-methoxypyrid-3-yl) pyrazolo-[1,5-a]-1,3, 5-triazine, its enantiomers and pharmaceutically acceptable salts as corticotropin releasing factor receptor ligands.
WO2002072101A1 (en) 2001-03-13 2002-09-19 Bristol-Myers Squibb Pharma Company A corticotropin releasing factor receptor ligand, its enantiomer and pharmaceutically acceptable salts
EP1372620A2 (en) 2001-03-15 2004-01-02 Saegis Pharmaceuticals Methods for restoring cognitive function following systemic stress
MXPA03008726A (en) 2001-03-29 2003-12-12 Lilly Co Eli N-(2-arylethyl)benzylamines as antagonists of the 5-ht6.
PL364580A1 (en) 2001-03-29 2004-12-13 Bristol-Myers Squibb Company Cyclopropylindole derivatives as selective serotonin reuptake inhibitors
TNSN03094A1 (en) 2001-04-19 2005-12-23 Warner Lambert Co BICYCLIC OR TRICYCLIC CONDENSED AMINO ACIDS
IL149106A0 (en) 2001-04-20 2002-11-10 Pfizer Prod Inc Therapeutic use of selective pde10 inhibitors
US20030032579A1 (en) 2001-04-20 2003-02-13 Pfizer Inc. Therapeutic use of selective PDE10 inhibitors
EP1666886A3 (en) 2001-04-20 2006-06-21 Pfizer Products Inc. Method of identifying selective PDE10 inhibitor compounds
US20030018047A1 (en) 2001-04-20 2003-01-23 Pfizer Inc. Therapeutic use of selective PDE10 inhibitors
MY129350A (en) 2001-04-25 2007-03-30 Bristol Myers Squibb Co Aripiprazole oral solution
AU2756602A (en) 2001-04-25 2002-10-31 Pfizer Products Inc. Methods and kits for treating depression or preventing deterioration of cognitive function
US20020165217A1 (en) 2001-05-01 2002-11-07 Pfizer Inc. Combination treatment for anxiety and depression
ES2254611T3 (en) 2001-05-11 2006-06-16 Pfizer Products Inc. DERIVATIVES OF TIAZOL.
PL366577A1 (en) 2001-05-14 2005-02-07 Bristol-Myers Squibb Pharma Company Substituted pyrazinones, pyridines and pyrimidines as corticotropin releasing factor ligands
EP1260221A3 (en) 2001-05-23 2002-12-18 Pfizer Products Inc. Combination treatment for depression and anxiety
WO2002094787A1 (en) 2001-05-23 2002-11-28 Ucb, S.A. 2-oxo-piperidinyl- and 2-oxo-azepanyl alkanoic acid derivativ es for the treatment of epilepsy and other neurological disorders
US6566550B2 (en) 2001-06-21 2003-05-20 Pfizer Inc Substituted aromatic ethers as inhibitors of glycine transport
US20030008892A1 (en) 2001-07-09 2003-01-09 Pfizer Inc. Pharmaceutical composition and method of modulating cholinergic function in a mammal
AU2002354575A1 (en) 2001-07-12 2003-01-29 Bristol-Myers Squibb Pharma Company Tetrahydropurinones as corticotropin releasing factor
WO2003006015A1 (en) 2001-07-13 2003-01-23 Bristol-Myers Squibb Pharma Company Substituted thiazoles and oxazoles as corticotropin releasing hormone ligands
WO2003010161A1 (en) 2001-07-24 2003-02-06 Bristol-Myers Squibb Company S-6-hydroxy-buspirone
EP1408979A4 (en) 2001-07-24 2005-03-23 Bristol Myers Squibb Co R-6-hydroxy-buspirone
US8354438B2 (en) 2001-08-08 2013-01-15 Michael Chez Neurological functions
ATE305917T1 (en) 2001-08-16 2005-10-15 Pfizer Prod Inc DIFLUOROMETHYLENE AROMATIC ETHERS AND THEIR USE AS GLYCINE TYPE 1 TRANSPORTER INHIBITORS
ATE404192T1 (en) 2001-08-22 2008-08-15 Hamilton Pharmaceuticals Inc USE OF NEFIRACETAM TO TREAT POSTISCHEMIC NEURODEGENERATION
WO2003020289A1 (en) 2001-08-30 2003-03-13 Ortho-Mcneil Pharmaceutical, Inc. Treatment of dementia and memory disorders with anticonvulsants and acetylcholinesterase inhibitors
WO2003022820A1 (en) 2001-09-06 2003-03-20 Bristol-Myers Squibb Pharma Company. Heteroaromatic substituted cyclopropane as corticotropin releasing hormone
ATE313325T1 (en) 2001-09-26 2006-01-15 Pfizer Prod Inc INDOLCAROBOXYLIC ACID AS A THYROID RECEPTOR LIGAND
EP1438036A2 (en) 2001-10-12 2004-07-21 Eli Lilly And Company Use of sulfonamide derivatives as pharmaceuticals compounds
DE60233884D1 (en) 2001-10-16 2009-11-12 Memory Pharmaceutical Corp 4 (4-ALKOXY-3-HYDROXYPHENYL) -2-PYRROLIDONE DERIVATIVES AS PDE-4 INHIBITORS FOR THE TREATMENT OF NEUROLOGICAL SYNDROMES
KR20010113584A (en) 2001-11-08 2001-12-28 (주)시스튜디오 a method for providing comics-animation by computers and a computer-readable medium storing data of comics-animation
US6673811B1 (en) 2001-11-19 2004-01-06 Neurogen Corporation 1H-pyrrolo [3,2-b] pyridine-3-carboxylic acid amines as GABAA receptor ligands
US20030119831A1 (en) 2001-11-20 2003-06-26 Hartz Richard A. 3,7-dihydro-purine-2,6-dione derivatives as CRF receptor ligands
US6992087B2 (en) 2001-11-21 2006-01-31 Pfizer Inc Substituted aryl 1,4-pyrazine derivatives
MXPA02010430A (en) 2001-11-27 2003-06-02 Warner Lambert Co Branched chain amino acid-dependent aminotransferase inhibitors and their use in the treatment of neurodegenerative diseases.
HUP0402619A3 (en) 2001-12-11 2008-04-28 Lilly Co Eli Use of norepinephrine reuptake inhibitors for the preparation of a medicament for treatment of cognitive failure
US6495700B1 (en) 2002-01-09 2002-12-17 Axonyx, Inc. Process for producing phenserine and its analog
US20050014848A1 (en) 2002-01-23 2005-01-20 Pfizer Inc. Combination of serotonin reuptake inhibitors and norephinephrine reuptake inhibitors
US20050009927A1 (en) 2002-01-23 2005-01-13 Pfizer Inc Combination of serotonin reuptake inhibitors and norepinephrine reuptake inhibitors
EP1476165A1 (en) 2002-02-22 2004-11-17 PHARMACIA &amp; UPJOHN COMPANY Substituted pyrimidinones and pyrimidinthiones
US20040167200A1 (en) 2002-03-25 2004-08-26 Pfizer Inc. Pharmaceutical composition and method of modulating cholinergic function in a mammal
GB0207104D0 (en) 2002-03-26 2002-05-08 Pfizer Ltd Stable hydrate of a muscarinic receptor antagonist
US6759552B2 (en) 2002-03-28 2004-07-06 Council Of Scientific And Industrial Research Compound as cholinesterase inhibitor and its isolation from fungus sporotrichum species
US7214673B2 (en) 2002-03-28 2007-05-08 Eli Lilly And Company Piperazine substituted aryl benzodiazepines and their use as dopamine receptor antagonists for the treatment of psychotic disorders
JP4405811B2 (en) 2002-03-29 2010-01-27 エーザイ・アール・アンド・ディー・マネジメント株式会社 (1-Indanone)-(1,2,3,6-tetrahydropyridine) derivative
AU2003218063A1 (en) 2002-04-03 2003-10-20 Eli Lilly And Company Therapy for psychoses combining an atypical antipsychotic and an mglu2/3 receptor agonist
US6713490B2 (en) 2002-04-26 2004-03-30 Pfizer, Inc. 3,4-dihydroquinolin-2(1H)-one compounds as NR2B receptor antagonists
CN1649845A (en) 2002-04-26 2005-08-03 先灵公司 Muscarinic antagonists
AU2003298514A1 (en) 2002-05-17 2004-05-04 Eisai Co., Ltd. Methods and compositions using cholinesterase inhibitors
WO2003101458A1 (en) 2002-05-31 2003-12-11 H. Lundbeck A/S A combination of an nmda-antagonist and acetylcholine esterase inhibitors for the treatment of alzheimer's disease
US6710040B1 (en) 2002-06-04 2004-03-23 Pfizer Inc. Fluorinated cyclic amides as dipeptidyl peptidase IV inhibitors
US20040001895A1 (en) 2002-06-17 2004-01-01 Pfizer Inc. Combination treatment for depression and anxiety
US20030235631A1 (en) 2002-06-17 2003-12-25 Pfizer Inc. Combination treatment for depression and anxiety
US20040006135A1 (en) 2002-06-19 2004-01-08 Pfizer Inc. Combination treatment for depression and anxiety
US20040116506A1 (en) 2002-06-20 2004-06-17 Krusz John Claude Use of levetiracetam for treating or preventing acute headaches
US6875771B2 (en) 2002-07-26 2005-04-05 Bristol-Myers Squibb Company Pyridopyrimidine derivatives as 5-HT6 antagonists
DE60313634T2 (en) 2002-08-05 2008-01-31 Eli Lilly And Co., Indianapolis PIPERAZINE-SUBSTITUTED ARYLBENZODIAZEPINE
US7557137B2 (en) 2002-08-05 2009-07-07 Bristol-Myers Squibb Company Gamma-lactams as beta-secretase inhibitors
US7053122B2 (en) 2002-08-09 2006-05-30 Pfizer Inc Therapeutic use of aryl amino acid derivatives
GB0219154D0 (en) 2002-08-16 2002-09-25 Pfizer Ltd Diaryl compounds
GB0219153D0 (en) 2002-08-16 2002-09-25 Pfizer Ltd Substituted glycine derivatives for use as medicaments
DE60327225D1 (en) 2002-08-20 2009-05-28 Bristol Myers Squibb Co ARIPIPRAZOL COMPLEX FORMULATION AND METHOD
JP2006504690A (en) 2002-09-10 2006-02-09 ファイザー・プロダクツ・インク Diazabicyclic compounds useful for the treatment of CNS and other disorders
AU2003270669A1 (en) 2002-09-18 2004-04-08 Bristol-Myers Squibb Company Cyclopentyl indole derivatives
TW200418838A (en) 2002-09-18 2004-10-01 Bristol Myers Squibb Co Compounds for the treatment of premature ejaculation
US7635709B2 (en) 2002-09-26 2009-12-22 The United States Of America As Represented By The Department Of Veterans Affairs Compositions and methods for bowel care in individuals with chronic intestinal pseudo-obstruction
US7067658B2 (en) 2002-09-30 2006-06-27 Bristol-Myers Squibb Company Pyridino and pyrimidino pyrazinones
GB0223494D0 (en) 2002-10-09 2002-11-13 Neuropharma Sa Dual binding site acetylcholinesterase inhibitors for the treatment of alzheimer's disease
KR100852834B1 (en) 2002-10-24 2008-08-18 메르츠 파마 게엠베하 운트 코. 카가아 A pharmaceutical product comprising 1-aminocyclohexane derivatives and acetylcholinesterase inhibitors and combination therapy using these compounds
ES2297200T3 (en) 2002-10-24 2008-05-01 Pfizer Products Inc. ACILO DERIVATIVES OF 5- (2- (4- (1,2 BENCISOTIAZOL-3-IL) -1-PIPERAZINIL) ETIL) -6-CHLORINE-1,3-DIHYDRO-2H-INDOL-2-ONA PRESENTING ACTIVITY NEUROLEPTICS
GB0225379D0 (en) 2002-10-31 2002-12-11 Pfizer Ltd Therapeutic proline derivatives
CA2502068A1 (en) 2002-11-18 2004-06-03 Pfizer Products Inc. Dipeptidyl peptidase iv inhibiting fluorinated cyclic amides
EP1581498A2 (en) 2002-11-22 2005-10-05 Bristol-Myers Squibb Company Pyridinyl, pyrimidinyl and pyrazinyl amides as potassium channel openers
WO2004047744A2 (en) 2002-11-22 2004-06-10 Bristol-Myers Squibb Company 3-heterocyclic benzylamide derivatives as potassium channel openers
WO2004047743A2 (en) 2002-11-22 2004-06-10 Bristol-Myers Squibb Company 1-aryl-2-hydroxyethyl amides as potassium channel openers
US7087609B2 (en) 2002-11-22 2006-08-08 Bristol-Myers Squibb Company 3-(pyridinyl-piperazin-1-yl)-phenylethyl amides as potassium channel openers
US7144881B2 (en) 2002-11-22 2006-12-05 Bristol-Myers Squibb Company Arylcyclopropylcarboxylic amides as potassium channel openers
ATE540129T1 (en) 2002-11-22 2012-01-15 Univ Johns Hopkins TARGET FOR THE THERAPY OF COGNITIVE DISABILITIES
WO2004051222A2 (en) 2002-12-03 2004-06-17 Ucb, S.A. Methods for the identification of agents for the treatment of seizures, neurological diseases, endocrinopathies and hormonal diseases
US7090985B2 (en) 2002-12-03 2006-08-15 Ucb, S.A. Methods for the identification of agents for the treatment of seizures, neurological diseases, endocrinopathies and hormonal diseases
WO2004052348A2 (en) 2002-12-11 2004-06-24 Pharmacia & Upjohn Company Llc Treatment of diseases with combinations of alpha 7 nicotinic acetylcholine receptor agonists and other compounds
OA12969A (en) 2002-12-11 2006-10-13 Pharmacia & Upjohn Co Llc Combination for the treatment of ADHD.
BRPI0406704A (en) 2003-01-31 2005-12-20 Pfizer Prod Inc 5ht ~ 7 ~ Antagonists and Inverse Agonists
KR100831545B1 (en) 2003-03-06 2008-05-21 셀진 코포레이션 Methods of using and compositions comprising selective cytokine inhibitory drugs for the treatment and management of disorders of the central nervous system
MXPA05009660A (en) 2003-03-12 2005-10-20 Pfizer Prod Inc Pyridyloxymethyl and benzisoxazole azabicyclic derivatives.
US20040191334A1 (en) 2003-03-24 2004-09-30 Pang-Chui Shaw Use of transhinone derivates as cholinesterase inhibitors in treating related diseases
ITMI20030573A1 (en) 2003-03-24 2004-09-25 Nikem Research Srl NOOTROPIC ACTION COMPOUNDS, THEIR PREPARATION,
MXPA05010293A (en) 2003-03-27 2005-11-17 Pfizer Prod Inc Substituted 4-amino[1,2,4]triazolo[4,3-a]quinoxalines.
TW200508197A (en) 2003-03-31 2005-03-01 Ucb Sa Indolone-acetamide derivatives, processes for preparing them and their uses
US7041671B2 (en) 2003-04-02 2006-05-09 Pfizer Inc Pyrrolo[1,2-b]pyridazine compounds and their uses
US7056920B2 (en) 2003-04-04 2006-06-06 Pfizer Inc Pyrrolo[1,2-B]pyridazine compounds and their uses
US7034023B2 (en) 2003-04-04 2006-04-25 Pfizer Inc Pyrrolo[1,2-B]pyridazine compounds and their uses
EP1615894A2 (en) 2003-04-14 2006-01-18 Pfizer Products Inc. 3-azabicyclo 3.2.1 octane derivatives
US20040204453A1 (en) 2003-04-14 2004-10-14 Pfizer Inc 4-Phenyl-piperidine compounds and their use as modulators of opioid receptors
US7056930B2 (en) 2003-04-14 2006-06-06 Pfizer Inc. 2-Azabicyclo[3.3.1]nonane derivatives
US7030145B2 (en) 2003-04-18 2006-04-18 Bristol-Myers Squibb Company Pyridinyl derivatives for the treatment of depression
US7112585B2 (en) 2003-04-18 2006-09-26 Bristol-Myers Squibb Company Pyrimidine derivatives as corticotropin releasing factor inhibitors
EP1620428A1 (en) 2003-04-23 2006-02-01 Pharmacia & Upjohn Company LLC Substituted pyrimidinones and pyrimidinthiones as crf antagonists
GB0309440D0 (en) 2003-04-25 2003-06-04 Lilly Co Eli Quinolone derivatives
US7732162B2 (en) 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
MXPA05011993A (en) 2003-05-07 2006-02-02 Pharmacia & Upjohn Co Llc Pyrrolo (1,2-b) pyridazine compounds and their use as crf-1 receptor antagonists.
WO2004099148A1 (en) 2003-05-09 2004-11-18 Pharmacia & Upjohn Company Llc Substituted pyrimidine derivatives
WO2004100954A1 (en) 2003-05-16 2004-11-25 Pfizer Products Inc. Treatment of psychotic and depressive disorders
MXPA05012320A (en) 2003-05-16 2006-01-30 Pfizer Prod Inc Treatment of bipolar disorders and associated symptoms.
MXPA05012325A (en) 2003-05-16 2006-01-30 Pfizer Prod Inc Method for enhancing cognition using ziprasidone.
CA2525366A1 (en) 2003-05-16 2004-11-25 Pfizer Products Inc. Therapeutic combinations of atypical antipsychotics with gaba modulators, anticonvulsants or benzodiazapines
CN1794994A (en) 2003-05-23 2006-06-28 大塚制药株式会社 Carbostyril derivatives and mood stabilizers for treating mood disorders
US7101881B2 (en) 2003-06-11 2006-09-05 Pfizer Inc Tetrahydroquinolines
EP1638932A1 (en) 2003-06-18 2006-03-29 Pfizer Products Inc. Novel piperazinyl-aryloxy and piperazinyl-heteroaryloxy-n-aryl lactams
ATE409480T1 (en) 2003-06-27 2008-10-15 Pfizer Prod Inc PYRAZOLOA3,4-BUPYRIDINE-6-ONE AS GSK-3 INHIBITORS
EP1641455B1 (en) 2003-06-27 2008-08-06 Pfizer Products Inc. Pyrazolo[3,4-b]pyridin-6-ones as gsk-3 inhibitors
US20050070577A1 (en) 2003-07-03 2005-03-31 Pfizer Inc. Compositions containing a serotonin selective reuptake inhibitor and a 5-HT2A receptor antagonist
WO2005013961A1 (en) 2003-07-17 2005-02-17 Eli Lilly And Company Combination therapy for treatment of cognitive disorders or psychoses
TW200510324A (en) 2003-08-11 2005-03-16 Lilly Co Eli 6-(2,2,2-trifluoroethylamino)-7-chiloro-2, 3, 4, 5-tetrahydro-1h-benzo[d]azepine as a 5-ht2c receptor agonist
WO2005019168A2 (en) 2003-08-20 2005-03-03 Pfizer Products Inc. Fluorinated lysine derivatives as dipeptidyl peptidase iv inhibitors
US20060270656A1 (en) 2003-09-09 2006-11-30 Eli Lilly And Company Substituted piperazines of azepines, oxazepines and thiazepines
CA2542339A1 (en) 2003-09-09 2005-03-17 Pfizer Products Inc. Combination of serotonin reuptake inhibitors and norepinephrine reuptake inhibitors
GB0322140D0 (en) 2003-09-22 2003-10-22 Pfizer Ltd Combinations
US7244843B2 (en) 2003-10-07 2007-07-17 Bristol-Myers Squibb Company Modulators of serotonin receptors
EP1673367A1 (en) 2003-10-08 2006-06-28 Pfizer, Inc. Fused lactam compounds
ES2324044T3 (en) 2003-10-08 2009-07-29 Eli Lilly And Company PIRROL AND PIRAZOL DERIVATIVES AS POTENTIALS OF GLUTAMATE RECEIVERS.
US20050080100A1 (en) 2003-10-09 2005-04-14 Pfizer Inc Pyridylamino compounds and methods of use thereof
WO2005039580A1 (en) 2003-10-16 2005-05-06 Boehringer Ingelheim International Gmbh Pharmaceutical composition comprising a monoamine neurotransmitter re-uptake inhibitor and an acetylcholinesterase inhibitor
WO2005037790A1 (en) 2003-10-16 2005-04-28 Pfizer Products Inc. Preparation of 3-azabicyclo [3.1.0] hexane derivatives
US7897639B2 (en) 2003-10-21 2011-03-01 Colucid Pharmaceuticals, Inc. Carbamoyl esters that inhibit cholinesterase and release pharmacologically active agents
WO2005053701A1 (en) 2003-11-26 2005-06-16 Pfizer Products Inc. Combination of dopamine agonists and aralkyl and aralkylidene heterocyclic lactams and imides
DE602004028150D1 (en) 2003-11-26 2010-08-26 Pfizer Prod Inc AMINOPYRAZOL DERIVATIVES AS GSK-3 INHIBITORS
WO2005051488A1 (en) 2003-11-26 2005-06-09 Pfizer Products Inc. Combination of dopamine agonists and monoamine reuptake inhibitors
ES2323313T3 (en) 2003-12-02 2009-07-13 Ucb Pharma, S.A. IMIDAZOL DERIVATIVES, PROCEDURES TO PREPARE THEM AND THEIR USES.
CA2548304A1 (en) 2003-12-12 2005-07-07 Eli Lilly And Company Selective norepinephrine reuptake inhibitors for the treatment of hot flashes, impulse control disorders and personality change due to a general medical condition
US20050227980A1 (en) 2003-12-15 2005-10-13 Pfizer Inc. Aralkyl and aralkylidene heterocyclic lactam and imides
BRPI0417709A (en) 2003-12-15 2007-03-20 Pfizer Prod Inc heterocyclic aralkylidem and aralkyl lactams and imides
JP2007514731A (en) 2003-12-19 2007-06-07 ファイザー インコーポレイテッド Benzenesulfonylamino-pyridin-2-yl derivatives and related compounds as inhibitors of 11-beta-hydroxysteroid dehydrogenase type 1 (11-β-hsd-1) for the treatment of diabetes and obesity
CA2549638A1 (en) 2003-12-23 2005-07-14 Pfizer Products Inc. Therapeutic combination for cognition enhancement and psychotic disorders
TW200522944A (en) 2003-12-23 2005-07-16 Lilly Co Eli CB1 modulator compounds
US20050171095A1 (en) 2004-01-06 2005-08-04 Pfizer Inc Combination of CRF antagonists and 5-HT1B receptor antagonists
ATE552251T1 (en) 2004-01-09 2012-04-15 Lilly Co Eli THIOPHENE AND FURAN COMPOUNDS
US20050222123A1 (en) 2004-01-27 2005-10-06 North Shore-Long Island Jewish Research Institute Cholinesterase inhibitors for treating inflammation
EP1737466A1 (en) 2004-01-29 2007-01-03 Pfizer Products Inc. COMBINATION OF y-AMINOBUTYRIC ACID MODULATORS AND 5-HT-1b RECEPTOR ANTAGONISTS
WO2005082370A1 (en) 2004-01-29 2005-09-09 Pfizer Products Inc. Combinations of an atypical antipsychotic and an aminomethylpyridyloxymethyl/benzisoxazole azabicyclic derivatives for treating cns disorders
WO2005074535A2 (en) 2004-01-30 2005-08-18 Eisai Co., Ltd. Cholinesterase inhibitors for spinal cord disorders
WO2005080361A1 (en) 2004-02-02 2005-09-01 Pfizer Products Inc. Histamine-3 receptor modulators
CA2556160A1 (en) 2004-02-13 2005-09-01 Pfizer Products Inc. Therapeutic combinations of atypical antipsychotics with corticotropin releasing factor antagonists
JP2007522248A (en) 2004-02-13 2007-08-09 ニューロモレキュラー・インコーポレイテッド Combination of NMDA receptor antagonists and antiepileptic drugs for the treatment of epilepsy and other CNS disorders
US20050182044A1 (en) 2004-02-17 2005-08-18 Bruinsma Gosse B. Combinatorial therapy with an acetylcholinesterase inhibitor and (3aR)-1,3a,8-trimethyl-1,2,3,3a,8,8a-hexahydropyrrolo[2,3,-b]indol-5-yl phenylcarbamate
CN1934088A (en) 2004-02-25 2007-03-21 伊莱利利公司 6-substituted 2,3,4,5-tetrahydro-1h-benzo[d]azepines as 5-ht2c receptor agonists
GB0405200D0 (en) 2004-03-08 2004-04-21 Pfizer Ltd Combinations comprising alpha-2-delta ligands
US20060148807A1 (en) 2004-03-12 2006-07-06 Pfizer Inc Pyrrolo[1,2b]pyridazine compounds and their uses
US20060166998A1 (en) 2004-03-12 2006-07-27 Pfizer Inc Pyrrolo[1,2-B]pyridazine compounds and their uses
ATE533745T1 (en) 2004-03-17 2011-12-15 Pfizer Prod Inc NEW BENZYL(IDENE) LACTAM DERIVATIVES
EP1740172A4 (en) 2004-03-19 2007-10-10 Axonyx Inc Acetylcholinesterase inhibitors and n-methyl-d-aspartate antagonists useful in the treatment of cognitive disorders
WO2005100334A1 (en) 2004-04-14 2005-10-27 Pfizer Products Inc. Dipeptidyl peptidase-iv inhibitors
WO2005102272A2 (en) 2004-04-21 2005-11-03 Pfizer Products Inc. Sustained-release dosage forms for cabergoline
JP2007533741A (en) 2004-04-22 2007-11-22 イーライ リリー アンド カンパニー BACE inhibitor
US20050245543A1 (en) 2004-04-30 2005-11-03 Pfizer Inc Histamine-3 receptor antagonists
JP2007537232A (en) 2004-05-11 2007-12-20 ファイザー・プロダクツ・インク Combination of atypical antipsychotic and 5-HT1B receptor antagonist
WO2005113527A1 (en) 2004-05-21 2005-12-01 Pfizer Products Inc. Tetrahydronaphthyl- piperazines as 5-ht1b antagonists, inverse agonists and partial agonists
EP1753745A2 (en) 2004-05-21 2007-02-21 Pfizer Products Inc. Pyrazinylmethyl lactam derivatives
WO2005121082A1 (en) 2004-06-11 2005-12-22 Ucb, S.A. Process for preparing 2-oxo-1-pyrrolidine derivatives by intramolecular allylation
JP2008503560A (en) 2004-06-22 2008-02-07 ファイザー・プロダクツ・インク Diazabicyclo histamine-3 receptor antagonist
JP2008503559A (en) 2004-06-25 2008-02-07 ファイザー・プロダクツ・インク Pyridylpiperazine for treating CNS disorders
US7572805B2 (en) 2004-07-14 2009-08-11 Bristol-Myers Squibb Company Pyrrolo(oxo)isoquinolines as 5HT ligands
US7618980B2 (en) 2004-07-14 2009-11-17 Bristol-Myers Squibb Company Pyrrolo(oxo)quinolines as 5HT ligands
US20060014733A1 (en) 2004-07-19 2006-01-19 Pfizer Inc Histamine-3 agonists and antagonists
BRPI0513444A (en) 2004-07-21 2008-05-06 Pfizer Prod Inc histamine 3 receptor antagonists
PA8640801A1 (en) 2004-08-09 2006-08-03 Warner Lambert Co NEW INHIBITORS OF THE QUINOXALINONE NOREPINEFRINE RECOVERY FOR THE TREATMENT OF CENTRAL NERVOUS SYSTEMS
MX2007002136A (en) 2004-09-01 2007-04-02 Pfizer Prod Inc Azabicyclic histamine-3 receptor antagonists.
JP2008512438A (en) 2004-09-10 2008-04-24 ファイザー・プロダクツ・インク Diphenyl ether ligands for therapy
MX2007002460A (en) 2004-09-10 2007-05-04 Pfizer Prod Inc Methods of treating mood disorders using benzisoxazole containing diazabicyclic compounds.
WO2006034196A1 (en) 2004-09-17 2006-03-30 Lifelike Biomatic Inc. Compositions for enhancing memory and methods therefor
US20060069087A1 (en) 2004-09-27 2006-03-30 Pfizer Inc Histamine-3 receptor antagonists
WO2006040688A2 (en) 2004-10-12 2006-04-20 Ernir Snorrason Inhibitors of acetylcholinesterase for treating skin diseases
US20060084692A1 (en) 2004-10-15 2006-04-20 Pfizer Inc. Treatment of bipolar disorders and associated symptoms
EP1805165B1 (en) 2004-10-18 2009-12-16 Eli Lilly And Company 1-(hetero)aryl-3-amino-pyrrolidine derivatives for use as mglur3 receptor antagonists
US7354951B2 (en) 2004-10-18 2008-04-08 Eli Lilly And Company Substituted benzopyrans as selective estrogen receptor-beta agonists
WO2006060082A1 (en) 2004-10-22 2006-06-08 THE GOVERNMENT OF THE U.S.A. as represented by THE SEC., DEPT. OF HEALTH & HUMAN SERVICES, NATIONAL INSTITUTES OF HEALTH Tricyclic compounds, preparation thereof and use thereof as cholinesterase activity inhibitors
WO2006046131A1 (en) 2004-10-29 2006-05-04 Pfizer Products Inc. Tetralin histamine-3 receptor antagonists
WO2006048727A1 (en) 2004-11-02 2006-05-11 Pfizer Products Inc. Piperazinylphenalkyl lactam/amine ligands for the 5ht1b receptor
JP2008519818A (en) 2004-11-15 2008-06-12 ファイザー・プロダクツ・インク Azabenzoxazole for the treatment of CNS disorders
WO2006051407A1 (en) 2004-11-15 2006-05-18 Pfizer Products Inc. Azabenzoxazoles for the treatment of cns disorders
WO2006056873A2 (en) 2004-11-29 2006-06-01 Warner-Lambert Company Llc Therapeutic pyrazolo[3,4-b] pyridines and indazoles
EP1824852B1 (en) 2004-12-07 2008-05-14 Pfizer Products Incorporated 1,2,3,3a,8,8a-hexahydro-2,7a-diaza-cyclopenta[a]inden-7-one derivatives which bind to neuronal nicotinic acetylcholine specific receptor sites and are useful in modulating cholinergic function and in the treatment of addictive disorders
WO2006071274A2 (en) 2004-12-23 2006-07-06 Voyager Pharmaceutical Corporation Leuprolide acetate and acetylcholinesterase inhibitors or nmda receptor antagonists for the treatment of alzheimer’s disease
EP1831172B1 (en) 2004-12-28 2009-02-18 Council of Scientific and Industrial Research Substituted carbamic acid quinolin-6-yl esters useful as acetylcholinesterase inhibitors
US20060183763A1 (en) 2004-12-31 2006-08-17 Pfizer Inc Novel pyrrolidyl derivatives of heteroaromatic compounds
SI1838716T1 (en) 2005-01-05 2011-10-28 Lilly Co Eli Olanzapine pamoate dihydrate
CA2595988A1 (en) 2005-01-27 2006-08-03 Alembic Limited Extended release formulation of levetiracetam
WO2006086464A2 (en) 2005-02-10 2006-08-17 Bristol-Myers Squibb Company Dihydroquinazolinones as 5ht modulators
FR2881618B1 (en) 2005-02-10 2007-04-13 Centre Nat Rech Scient USE OF PYRIMIDINETRIONE-DERIVED COMPOUNDS AS ACETYLCHOLINESTERASE INHIBITORS, COMPOSITIONS CONTAINING THESE DERIVATIVES, AND USES THEREOF AS INSECTICIDES
EP1853578A1 (en) 2005-02-15 2007-11-14 Eli Lilly And Company Substituted tetralins as selective estrogen receptor-beta agonists
US20070298098A1 (en) 2005-02-16 2007-12-27 Elan Pharma International Limited Controlled Release Compositions Comprising Levetiracetam
WO2006090272A1 (en) 2005-02-22 2006-08-31 Warner-Lambert Company Llc Isoquinoline [1,8]naphthyridin-2-ones and related compounds for treatment of schizophrenia
WO2006090273A2 (en) 2005-02-22 2006-08-31 Warner-Lambert Company Llc [1,8]naphthyridin-2-ones and related compounds with keto or hydroxyl linkers for the treatment of schizophrenia
US8431156B2 (en) 2005-02-22 2013-04-30 Sun Pharma Advanced Research Company Ltd. Pharmaceutical composition
US8216611B2 (en) 2005-03-30 2012-07-10 Mylan Pharmaceuticals Ulc Combined-step process for pharmaceutical compositions
JP4109709B1 (en) 2005-04-01 2008-07-02 ワーナー−ランバート カンパニー リミテッド ライアビリティー カンパニー Tetrahydro-pyridazepin-8-one and related compounds for the treatment of schizophrenia
PL1874282T3 (en) 2005-04-06 2011-04-29 Adamas Pharmaceuticals Inc Methods and compositions for treatment of cns disorders
CA2603939C (en) 2005-04-08 2013-08-27 Pfizer Products Inc. Bicyclic [3.1.0] heteroaryl amides as type i glycine transport inhibitors
WO2006106416A1 (en) 2005-04-08 2006-10-12 Pfizer Products Inc. PYRIDIL-LACTAMS AND THEIR USE 5 -HTl RECEPTORS LIGAN
US20080305161A1 (en) 2005-04-13 2008-12-11 Pfizer Inc Injectable depot formulations and methods for providing sustained release of nanoparticle compositions
US7741358B2 (en) 2005-04-14 2010-06-22 N.V. Organon Crystal form of asenapine maleate
US20060241144A1 (en) 2005-04-20 2006-10-26 Albert Cha Method for treating apathy syndrome
WO2006116401A1 (en) 2005-04-28 2006-11-02 Bristol-Myers Squibb Company C-5 substituted quinazolinone derivatives as selective estrogen receptor beta modulators
JP2008542378A (en) * 2005-05-31 2008-11-27 オレキシジェン・セラピューティクス・インコーポレーテッド Methods and compositions for managing psychotic disorders
EP2308870A3 (en) 2005-06-01 2011-10-19 UCB Pharma S.A. 2-oxo-1-pyrrolidine deriatives, processes for preparing them and their uses
EP1731149A1 (en) 2005-06-08 2006-12-13 Ucb S.A. Use of 2-oxo-1-pyrrolidone derivatives for the treatment of diseases characterized by progressive myoclonic epilepsy
US7812040B2 (en) 2005-06-22 2010-10-12 Pfizer Inc. Histamine-3 receptor antagonists
US20070015763A1 (en) 2005-07-12 2007-01-18 Pfizer Inc Treatment of psychosis associated with parkinson's disease and subcortical dementias using a combination of an atypical antipsychotic with a dopamine agonist
JP2009502959A (en) 2005-07-28 2009-01-29 ブリストル−マイヤーズ スクイブ カンパニー Substituted tetrahydro-1H-pyrido [4,3, B] indoles as serotonin receptor agonists and antagonists
WO2007019312A2 (en) 2005-08-03 2007-02-15 The Johns Hopkins University Methods for characterizing and treating cognitive impairment in aging and disease
TW200738734A (en) 2005-08-08 2007-10-16 Pfizer Prod Inc Benzoate salt of 4-(5-methyl-oxazolo[4,5-b]pyridin-2-yl)-1,4-diaza-bicyclo[3.2.2]nonane
US7795436B2 (en) 2005-08-24 2010-09-14 Bristol-Myers Squibb Company Substituted tricyclic heterocycles as serotonin receptor agonists and antagonists
GB0517740D0 (en) 2005-08-31 2005-10-12 Novartis Ag Organic compounds
WO2007026219A2 (en) 2005-08-31 2007-03-08 Pfizer Products Inc. Combinations of a 5-ht1b antagonist with a noradrenalin re-uptake inhibitor or a serotonin noradrenalin reutake inhibitor for treating cns conditions
EP1924578B1 (en) 2005-09-01 2013-11-06 Eli Lilly And Company 6-N-LINKED HETEROCYCLE-SUBSTITUTED 2,3,4,5-TETRAHYDRO-1H-BENZO[d]AZEPINES AS 5-HT2C RECEPTOR AGONISTS
BRPI0615048A2 (en) 2005-09-01 2010-03-30 Lilly Co Eli compound, pharmaceutical composition, and use of a compound
CA2619566C (en) 2005-09-01 2013-08-13 Eli Lilly And Company 6-substituted-2,3,4,5-tetrahydro-1h-benzo[d]azepines as 5-ht2c receptor agonists
ATE437859T1 (en) 2005-09-01 2009-08-15 Lilly Co Eli 6-SUBSTITUTED-2,3,4,5-TETRAHYDRO-1H-BENZOEDUEAZEPINES AS 5-HT2C RECEPTOR AGONISTS
US20090215857A1 (en) 2005-09-13 2009-08-27 Pfizer Products Inc. Therapeutic Pyrrolidines
US8618115B2 (en) 2005-10-26 2013-12-31 Bristol-Myers Squibb Company Substituted thieno[3,2-d]pyrimidinones as MCHR1 antagonists and methods for using them
US7745447B2 (en) 2005-10-26 2010-06-29 Bristol-Myers Squibb Company Substituted thieno[3,2-D]pyrimidines as non-basic melanin concentrating hormone receptor-1 antagonists
US8158673B2 (en) 2005-10-27 2012-04-17 Pfizer Inc. Histamine-3 receptor antagonists
WO2007057742A2 (en) 2005-11-18 2007-05-24 Pfizer Products Inc. Novel piperazinone derivatives
WO2007063385A2 (en) 2005-12-01 2007-06-07 Pfizer Products Inc. Spirocyclic amine histamine-3 receptor antagonists
AU2006322349A1 (en) 2005-12-07 2007-06-14 Ucb Pharma, S.A. Xanthine derivatives, processes for preparing them and their uses
WO2007069053A1 (en) 2005-12-14 2007-06-21 Pfizer Products Inc. Benzimidazole antagonists of the h-3 receptor
NL2000397C2 (en) 2006-01-05 2007-10-30 Pfizer Prod Inc Bicyclic heteroaryl compounds as PDE10 inhibitors.
BRPI0707223A2 (en) 2006-01-27 2011-04-26 Pfizer Prod Inc aminophthalazine derivative compounds
WO2007088450A2 (en) 2006-02-01 2007-08-09 Pfizer Products Inc. Chromane antagonist of the h-3 receptor
WO2007088462A1 (en) 2006-02-01 2007-08-09 Pfizer Products Inc. Spirochromane antagonists of the h-3 receptor
US7553836B2 (en) 2006-02-06 2009-06-30 Bristol-Myers Squibb Company Melanin concentrating hormone receptor-1 antagonists
JP2009527542A (en) 2006-02-23 2009-07-30 ファイザー・プロダクツ・インク Substituted quinazolines as PDE10 inhibitors
WO2007099423A1 (en) 2006-03-02 2007-09-07 Pfizer Products Inc. 1-pyrrolidine indane derivatives as histamine-3 receptor antagonists
US20100216734A1 (en) 2006-03-08 2010-08-26 Braincells, Inc. Modulation of neurogenesis by nootropic agents
EP2007749A2 (en) 2006-03-13 2008-12-31 Pfizer Products Inc. Tetralines antagonists of the h-3 receptor
EP2001463B1 (en) 2006-03-20 2013-07-03 Council of Scientific and Industrial Research A pharmaceutical composition useful as acetyl cholinesterase inhibitors
US20070224636A1 (en) 2006-03-24 2007-09-27 Pfizer Inc Pyrrolo[1,2b]pyridazine compounds and their uses
CA2650711A1 (en) 2006-04-28 2007-11-08 Northwestern University Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
US20090176829A1 (en) 2006-05-02 2009-07-09 Pfizer Inc Bicyclic heteroaryl compounds as pde10 inhibitors
US7858611B2 (en) 2006-05-09 2010-12-28 Braincells Inc. Neurogenesis by modulating angiotensin
JP2009536667A (en) 2006-05-09 2009-10-15 ブレインセルス,インコーポレイティド 5HT receptor-mediated neurogenesis
WO2007138431A2 (en) 2006-05-30 2007-12-06 Pfizer Products Inc. Azabicyclic ether histamine-3 antagonists
JP5214597B2 (en) 2006-06-08 2013-06-19 ユセベ ファルマ ソシエテ アノニム Co-crystal of pyrrolidinone
WO2008001182A1 (en) 2006-06-26 2008-01-03 Pfizer Products Inc. Tricyclic heteroaryl compounds as pde10 inhibitors
US20080014271A1 (en) 2006-07-13 2008-01-17 Ucb, S.A. Novel pharmaceutical compositions comprising levetiracetam
KR20090029299A (en) 2006-07-14 2009-03-20 화이자 프로덕츠 인코포레이티드 Tartrate salt of (7s)-7-[(5-fluoro-2-methyl-benzyl)oxy]-2-[(2r)-2-methylpiperazin-1-yl]-6,7-dihydro-5h-cyclopenta[b]pyridine
WO2008015516A1 (en) 2006-07-28 2008-02-07 Pfizer Products Inc. Fused tricyclic heterocycles for the treatment of schizophrenia
US20080045512A1 (en) 2006-08-09 2008-02-21 Pfizer Inc. Benzoate salt of 4-(5-methyl-oxazolo[4,5-b]-pyridin-2-yl)-1,4-diazabicyclo[3.2.2]nonane
WO2008020302A2 (en) 2006-08-17 2008-02-21 Pfizer Products Inc. Heteroaromatic quinoline-based compounds as phosphodiesterase (pde) inhibitors
EP2051696A2 (en) 2006-08-18 2009-04-29 Morton Grove Pharmaceuticals, Inc. Stable liquid levetiracetam compositions and methods
WO2008020306A2 (en) 2006-08-18 2008-02-21 Pfizer Products Inc. Isoindole derivatives
WO2008026046A1 (en) 2006-08-30 2008-03-06 Pfizer Products Inc. Morpholine d3 dopamine antagonists
JP2010503654A (en) 2006-09-12 2010-02-04 ファイザー・プロダクツ・インク Benzimidazolone derivatives
WO2008062446A2 (en) 2006-09-14 2008-05-29 Alembic Limited An extended release composition of levetiracetam, which exhibits no adverse food effect
WO2008036846A2 (en) 2006-09-22 2008-03-27 Braincells, Inc. Combination comprising an hmg-coa reductase inhibitor and a second neurogenic agent for treating a nervous system disorder and increasing neurogenesis
PE20080926A1 (en) 2006-10-23 2008-07-19 Lilly Co Eli COMPOUNDS CB1
KR20090087009A (en) 2006-10-27 2009-08-14 메디베이션 뉴롤로지 인코퍼레이티드 Methods and combination therapies for treating alzheimer's disease
WO2008065500A2 (en) 2006-11-30 2008-06-05 Pfizer Products Inc. Heteroaryl amides as type i glycine transport inhibitors
CN101626688A (en) 2006-12-11 2010-01-13 雷维瓦药品公司 Compositions, synthesis, and methods of using indanone based cholinesterase inhibitors
WO2008074896A1 (en) 2006-12-21 2008-06-26 Prendergast Patrick T Compositions and methods for treatment of chronic neurological disorders
WO2008084324A1 (en) 2007-01-04 2008-07-17 Pfizer Products Inc. Naphthyridinone compound
CA2676456C (en) 2007-01-22 2012-03-27 Pfizer Products Inc. Tosylate salt of a therapeutic compound and pharmaceutical compositions thereof
EP2125709A2 (en) 2007-02-02 2009-12-02 Colucid Pharmaceuticals, Inc. Compounds that inhibit cholinesterase
FR2912056B1 (en) 2007-02-05 2009-12-18 Rd Pharmagal EXTENDED RELEASE COMPOSITION OF LEVETIRACETAM AND PREPARATION METHOD
AU2007346591A1 (en) 2007-02-07 2008-08-14 Gosforth Centre (Holdings) Pty Ltd Treatment of ADHD
WO2008096260A1 (en) 2007-02-07 2008-08-14 Pfizer Inc. 3-amino-pyrrolo[3,4-c] pyrazole- 5 (1h, 4h, 6h) carbaldehyde derivatives as pkc inhibitors
MX2009010951A (en) 2007-04-12 2009-10-29 Pfizer 3-amido-pyrrolo [3, 4-c] pyrazole-5 (1h, 4h, 6h) carbaldehyde derivatives as inhibitors of protein kinase c.
US7851622B2 (en) 2007-04-25 2010-12-14 Bristol-Myers Squibb Company Non-basic melanin concentrating hormone receptor-1 antagonists
WO2008132139A2 (en) 2007-04-27 2008-11-06 Ucb Pharma S.A. New heterocyclic derivatives useful for the treatment of cns disorders
WO2008144305A1 (en) 2007-05-18 2008-11-27 Janssen Pharmaceutica N.V. Diaryl-substituted tetrahydroisoquinolines as histamine h3 receptor and serotonin transporter modulators
US20090011994A1 (en) 2007-07-06 2009-01-08 Bristol-Myers Squibb Company Non-basic melanin concentrating hormone receptor-1 antagonists and methods
RU2339620C1 (en) 2007-07-09 2008-11-27 Общество С Ограниченной Ответственностью "Фармвинг" Oxalate n,n-dimethyl-2-n,n-dimethylaminomethylpyridyl-3-carbamate, possessing anti-cholinesterase activity and ability to improve cognitive functions
CA2690110A1 (en) 2007-07-13 2009-01-22 Eisai R&D Management Co., Ltd. Ampa receptor antagonists for neuropathic pain
US20090030403A1 (en) 2007-07-27 2009-01-29 Leyde Kent W Methods and Systems for Attenuating the Tolerance Response to a Drug
KR20090031333A (en) 2007-09-21 2009-03-25 주식회사 엘지생명과학 Beta-secretase inhibiting compounds
US20090176740A1 (en) 2007-11-30 2009-07-09 Phillips Ii Dauglas James Treatment of neurological conditions by the co-administration of aniracetam and l-alpha glycerylphosphorylcholine
AU2008332880C1 (en) 2007-12-07 2012-07-19 Pfizer Inc. Tosylate salt of trans-N-isobutyl-3-fluoro-3- [3-fluoro-4- (pyrrolidin-1-yl-methyl) -phenyl] cyclobut anecarboxamide
US20100297181A1 (en) * 2007-12-26 2010-11-25 Eisai R&D Management Co., Ltd. AMPA Receptor Antagonists for Epilepsy, Mental Disorders or Deficits in Sensory Organ
CA2713025C (en) 2008-01-22 2012-12-04 Eli Lilly And Company Kappa selective opioid receptor antagonist
CA2714232A1 (en) 2008-02-05 2009-08-13 Pfizer Inc. Pyridinyl amides for the treatment of cns and metabolic disorders
RS53418B (en) 2008-03-03 2014-12-31 Ucb Pharma S.A. Pharmaceutical solutions, process of preparation and therapeutic uses
EP2276735A1 (en) 2008-04-17 2011-01-26 Pfizer Inc. Ether benzylidene piperidine aryl carboxamide compounds useful as faah inhibitors
US8426448B2 (en) 2008-04-22 2013-04-23 Eli Lilly And Company 1,5-diphenyl-pyrrolidin-2-one compounds as CB-1 ligands
SI2280961T1 (en) 2008-04-22 2012-11-30 Lilly Co Eli 1,5-diphenyl-pyrrolidin-2-one compounds as cb-1 ligands
AR071997A1 (en) 2008-06-04 2010-07-28 Bristol Myers Squibb Co CRYSTAL FORM OF 6 - ((4S) -2-METHYL-4- (2-NAFTIL) -1,2,3,4-TETRAHYDROISOQUINOLIN-7-IL) PIRIDAZIN-3-AMINA
WO2010002869A1 (en) 2008-07-01 2010-01-07 Concert Pharmaceuticals, Inc. 2-oxo-1-pyrrolidine derivatives
JP2010024156A (en) 2008-07-16 2010-02-04 Ucb Pharma Sa Pharmaceutical composition comprising levetiracetam
TW201006801A (en) 2008-07-18 2010-02-16 Lilly Co Eli Imidazole carboxamides
US7932256B2 (en) 2008-07-31 2011-04-26 Bristol-Myers Squibb Company (S)-4-(1-cyclopropyl-2-methoxyethyl)-6-(6-(difluoromethoxy)-2,5-dimethylpyridin-3-ylamino)-5-oxo-4,5-dihydropyrazine-2-carbonitrile: a pyrazinone modulator of corticotropin-releasing factor receptor activity
MX2011001384A (en) 2008-08-06 2011-09-27 Gosforth Ct Holdings Pty Ltd Compositions and methods for treating psychiatric disorders.
CN102227217A (en) 2008-10-16 2011-10-26 约翰斯.霍普金斯大学 Methods and compositions for improving cognitive function
US20100113465A1 (en) 2008-10-30 2010-05-06 Pfizer Inc. 7-azaspiro[3.5]nonane-7-carboxamide compounds
US9125898B2 (en) 2008-11-14 2015-09-08 Neurotune Ag Acetam derivatives for pain relief
WO2010057088A2 (en) 2008-11-17 2010-05-20 Auspex Pharmaceuticals, Inc. Pyrrolidinyl modulators of nicotinic acetylcholine receptors
PT2358360T (en) 2008-11-18 2016-11-15 Ucb Biopharma Sprl Prolonged release formulations comprising an 2-oxo-1-pyrrolidine derivative
CA2741839A1 (en) 2008-11-21 2010-05-27 Pfizer Inc. 1-oxa-8-azaspiro [4,5] decane-8-carboxamide compounds as faah inhibitors
US20100172979A1 (en) 2008-12-24 2010-07-08 Zhongshui Yu Controlled-release formulations
CN104906085A (en) 2009-01-29 2015-09-16 Ucb医药有限公司 Pharmaceutical compositions comprising 2-oxo-1-pyrrolidine derivatives
EP2393483B1 (en) 2009-02-09 2017-06-28 UCB Biopharma SPRL Pharmaceutical compositions comprising brivaracetam
WO2010104830A1 (en) 2009-03-09 2010-09-16 Bristol-Myers Squibb Company Pyridone analogs useful as melanin concentrating hormone receptor-1 antagonists
US8278316B2 (en) 2009-03-09 2012-10-02 Bristol-Myers Squibb Company Aza pyridone analogs useful as melanin concentrating hormone receptor-1 antagonists
WO2010111080A2 (en) 2009-03-27 2010-09-30 Eli Lilly And Company Optimized treatment of schizophrenia
US8846411B2 (en) 2009-06-11 2014-09-30 Microgenics Corporation Derivatives, reagents, and immunoassay for detecting levetiracetam
MA33407B1 (en) 2009-06-18 2012-07-03 Pfizer BICYCLIC AND TRICYCLIC COMPOUNDS AS KAT II INHIBITORS
US20120171125A1 (en) 2009-08-07 2012-07-05 Ucb Pharma, S.A. Methods for Enhancing the Cognitive Function
CN101647789B (en) 2009-09-01 2011-08-17 天津药物研究院药业有限责任公司 Levetiracetam slow release pellet capsule preparation
CN102548544B (en) 2009-10-09 2015-01-21 永进药品工业株式会社 Pharmaceutical composition with both immediate and extended release characteristics
WO2011057199A1 (en) * 2009-11-06 2011-05-12 Adenios, Inc. Compositions for treating cns disorders
KR101368130B1 (en) 2009-11-16 2014-02-27 일라이 릴리 앤드 캄파니 Spiropiperidine compounds as orl-1 receptor antagonists
UA107943C2 (en) 2009-11-16 2015-03-10 Lilly Co Eli Compounds of spiropiperidines as antagonists of the orl-1 receptors
AR079343A1 (en) 2009-12-21 2012-01-18 Lilly Co Eli ACID COMPOUND 2-AMINO-4- (4H-1,2,4-TRIAZOL-3-ILSULFANIL) BICYCLE [3.1.0] HEXANO-2,6-DICARBOXILICO, PHARMACEUTICAL COMPOSITION THAT INCLUDES IT AND ITS USE TO PREPARE A MEDICINAL PRODUCT USEFUL TO TREAT A PSYCHIATRIC DISORDER
EA037187B1 (en) * 2010-02-09 2021-02-17 Дзе Джонс Хопкинс Юниверсити Method and composition for treating a cognitive disorder
WO2011143721A1 (en) 2010-05-21 2011-11-24 Gosforth Centre (Holdings) Pty Ltd Compositions and methods for treating neurodegenerative disorders
MX2013000138A (en) 2010-07-08 2013-03-05 Pfizer Piperidinyl pyrimidine amides as kv7 potassium channel openers.
CA2809217A1 (en) 2010-09-21 2012-03-29 Pfizer Inc. Pyrimidones for treatment of potassium channel related diseases
MX2013003832A (en) 2010-10-26 2013-10-01 Alpharma Pharmaceuticals Llc Formulations and methods for attenuating respiratory depression induced by opioid overdose.
KR20120055313A (en) 2010-11-23 2012-05-31 주식회사 바이오파마티스 Sustained-release pharmaceutical composition comprising levetiracetam or pharmaceutically acceptable salt thereof having improved dissolution stability and method for manufacturing the same
WO2012073143A1 (en) 2010-12-01 2012-06-07 Pfizer Inc. Kat ii inhibitors
CN103228631B (en) 2010-12-01 2015-04-29 辉瑞大药厂 Kat II inhibitors
EP3400934A1 (en) * 2011-02-09 2018-11-14 The Johns Hopkins University Levetiracetam for improving cognitive function
JP5543039B2 (en) 2011-02-23 2014-07-09 ファイザー・インク Imidazo [5,1-f] [1,2,4] triazine for the treatment of neurological disorders
DE102011103270A1 (en) 2011-05-26 2012-11-29 Stada Arzneimittel Ag Powdered mixture for the preparation of levetiracetam-containing tablets
GB201111712D0 (en) 2011-07-08 2011-08-24 Gosforth Ct Holdings Pty Ltd Pharmaceutical compositions
US10154988B2 (en) 2012-11-14 2018-12-18 The Johns Hopkins University Methods and compositions for treating schizophrenia
WO2014144801A1 (en) 2013-03-15 2014-09-18 Agenebio Inc. Methods and compositions for improving cognitive function
WO2014144663A1 (en) 2013-03-15 2014-09-18 The Johns Hopkins University Methods and compositions for improving cognitive function
EP2968257A4 (en) 2013-03-15 2016-08-31 Univ Johns Hopkins Methods and compositons for improving cognitive function
US10925834B2 (en) 2015-05-22 2021-02-23 Agenebio, Inc. Extended release pharmaceutical compositions of levetiracetam

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070212428A1 (en) * 2004-06-04 2007-09-13 Mood Management Sciences, Inc. Methods and compositions for treating mood disorder
US20070264358A1 (en) * 2004-06-04 2007-11-15 Wittlin William A Methods and Compositions for Treating Mood Disorder
US20070244143A1 (en) * 2006-03-08 2007-10-18 Braincells, Inc Modulation of neurogenesis by nootropic agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Bora et al., "Theory of mind impairment: a distinct trait-marker for schizophrenia spectrum disorders and bipolar disorder?", 2009, Acta Psychiatr. Scand., 120(4), pp. 253-264. *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10154988B2 (en) 2012-11-14 2018-12-18 The Johns Hopkins University Methods and compositions for treating schizophrenia
US10806717B2 (en) 2013-03-15 2020-10-20 The Johns Hopkins University Methods and compositions for improving cognitive function
US11160785B2 (en) 2013-03-15 2021-11-02 Agenebio Inc. Methods and compositions for improving cognitive function
US10159648B2 (en) 2015-05-22 2018-12-25 Agenebio, Inc. Extended release pharmaceutical compositions of levetiracetam
US10925834B2 (en) 2015-05-22 2021-02-23 Agenebio, Inc. Extended release pharmaceutical compositions of levetiracetam
US10898449B2 (en) 2016-12-20 2021-01-26 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
US10980753B2 (en) 2016-12-20 2021-04-20 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
US11337932B2 (en) 2016-12-20 2022-05-24 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and polysiloxane or polyisobutylene
US11253173B1 (en) * 2017-05-30 2022-02-22 Verily Life Sciences Llc Digital characterization of movement to detect and monitor disorders
US11998317B1 (en) * 2017-05-30 2024-06-04 Verily Life Sciences Llc Digital characterization of movement to detect and monitor disorders
US11033512B2 (en) 2017-06-26 2021-06-15 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and silicone acrylic hybrid polymer
US11648213B2 (en) 2018-06-20 2023-05-16 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine

Also Published As

Publication number Publication date
US10154988B2 (en) 2018-12-18
EP2919788A1 (en) 2015-09-23
JP6440625B2 (en) 2018-12-19
CA2891122A1 (en) 2014-05-22
US20180125818A1 (en) 2018-05-10
EP2919788A4 (en) 2016-05-25
US20150313876A1 (en) 2015-11-05
HK1215170A1 (en) 2016-08-19
JP2018090609A (en) 2018-06-14
CA2891122C (en) 2021-07-20
WO2014078568A1 (en) 2014-05-22
EP3610890A1 (en) 2020-02-19
US10624875B2 (en) 2020-04-21
JP2015537003A (en) 2015-12-24

Similar Documents

Publication Publication Date Title
US10624875B2 (en) Methods and compositions for treating schizophrenia
US20210244709A1 (en) Methods and compositions for improving cognitive function
US20230033195A1 (en) Methods and compositions for improving cognitive function
US20160030391A1 (en) Methods and compositions for improving cognitive function
CA2789014C (en) Methods and compositions for improving cognitive function
US11160785B2 (en) Methods and compositions for improving cognitive function
AU2018208662B2 (en) Methods and Compositions for Treating Schizophrenia
RU2792010C2 (en) Methods and compositions to improve cognitive functions
AU2013344646A1 (en) Methods and compositions for treating schizophrenia

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: THE JOHNS HOPKINS UNIVERSITY, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GALLAGHER, MICHELA;REEL/FRAME:044755/0572

Effective date: 20131112

AS Assignment

Owner name: THE JOHNS HOPKINS UNIVERSITY, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KOH, MING TENG;REEL/FRAME:059657/0650

Effective date: 20210325