Synergistic composition comprising non-racemic proportions of (R) -dimiracetam (1) and (S) -dimiracetam (2)

文档序号:1559399 发布日期:2020-01-21 浏览:27次 中文

阅读说明:本技术 包含非外消旋比例的(r)-地来西坦(1)和(s)-地来西坦(2)的协同组合物 (Synergistic composition comprising non-racemic proportions of (R) -dimiracetam (1) and (S) -dimiracetam (2) ) 是由 M·舍茨 C·法里纳 于 2018-05-30 设计创作,主要内容包括:本发明涉及一种按一定比例存在的3,6,7,7a-四氢-1H-吡咯并[1,5-a]咪唑-2,5-二酮的对映体及其药学上可接受的溶剂化物或共晶体的组合物,一种包含所述组合物的药物组合物,其作为药物的用途以及本发明的组合物或药物组合物在治疗和/或预防疾病或病症中的用途,所述疾病或病症通常且优选地选自周围感觉神经病变、优选周围神经病理性疼痛、癫痫发作、抑郁症或认知障碍。(The present invention relates to a composition of enantiomers of 3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione and pharmaceutically acceptable solvates or co-crystals thereof present in a certain ratio, a pharmaceutical composition comprising said composition, its use as a medicament and the use of the composition or pharmaceutical composition of the invention in the treatment and/or prevention of a disease or condition, which is typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain, seizures, depression or cognitive disorders.)

1. A composition comprising (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)),

Figure FDA0002285200270000011

and/or pharmaceutically acceptable solvates or co-crystals thereof,

wherein the enantiomeric excess (ee) of the (R) -dimiracetam (1) is equal to or higher than 30% and lower than or equal to 60%.

2. The composition according to claim 1, wherein the ratio of (R) -dimiracetam (1) to (S) -dimiracetam (2) is from 2:1 to 3.3:1.

3. The composition according to claim 1 or 2, wherein (R) -dimiracetam (1) and/or a pharmaceutically acceptable solvate or co-crystal thereof, and (S) -dimiracetam (2) and/or a pharmaceutically acceptable solvate or co-crystal thereof, are packaged separately.

4. The composition according to claim 1, wherein the composition is a non-racemic mixture of 3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione (dimiracetam), wherein the non-racemic mixture comprises (R) -dimiracetam (1) and (S) -dimiracetam (2) with an enantiomeric excess (ee) of (R) -dimiracetam (1) equal to or greater than 33% and less than or equal to 54%, and pharmaceutically acceptable solvates or co-crystals thereof.

5. A pharmaceutical composition comprising the composition of any one of the preceding claims and a pharmaceutically acceptable carrier.

6. A kit of parts comprising (R) -dimiracetam (1) and (S) -dimiracetam (2) and instructions to combine (R) -dimiracetam (1) and (S) -dimiracetam (2) to obtain an enantiomeric excess (ee) of (R) -dimiracetam (1) equal to or higher than 33% and lower than or equal to 54%.

7. Use of the composition according to any one of claims 1-4 or the pharmaceutical composition of claim 5 or the kit of claim 6 as a medicament.

8. Use of the composition or the pharmaceutical composition or the kit according to claim 7 for the treatment and/or prevention of a disease or condition, wherein the disease, injury or condition is selected from peripheral sensory neuropathy, seizures, depression or cognitive disorders.

9. Use of the composition or the pharmaceutical composition or the kit according to claim 7 for the treatment and/or prevention of a disease, injury or condition, wherein the disease, injury or condition is selected from a peripheral sensory neuropathy, a neuropsychiatric disease, a motor neuron disease, or a movement disorder.

10. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to claim 8 or 9, wherein the disease, injury or condition is peripheral sensory neuropathy, wherein preferably the peripheral sensory neuropathy is peripheral neuropathic pain.

11. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to any one of claims 8 or 10, wherein the peripheral sensory neuropathy is diabetic neuropathy, post herpetic neuropathy, low back pain, sacral pain, surgical pain, crush injury, spinal cord injury, complex regional pain syndrome, phantom limb sensation, peripheral sensory neuropathy associated with osteoarthritis, peripheral sensory neuropathy associated with rheumatoid arthritis, peripheral sensory neuropathy associated with autoimmune osteoarthritis, headache, fibromyalgia, peripheral sensory neuropathy induced by treatment with a proliferation inhibitor, peripheral sensory neuropathy induced by a chemotherapeutic agent, peripheral sensory neuropathy associated with visceral injury, peripheral sensory neuropathy associated with osteonecrosis, peripheral sensory neuropathy associated with infection by human immunodeficiency virus, peripheral sensory neuropathy induced by treatment with a proliferation inhibitor, peripheral sensory neuropathy induced by a chemotherapeutic agent, peripheral sensory neuropathy associated with visceral injury, peripheral sensory neuropathy induced by infection by human immunodeficiency virus, or a method of the invention, Peripheral neuropathic pain, or peripheral sensory neuropathy caused by antiviral agents.

12. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to any one of claims 8 to 11, wherein the peripheral sensory neuropathy is selected from peripheral sensory neuropathy caused by a chemotherapeutic agent or peripheral sensory neuropathy caused by an antiviral agent.

13. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to any one of claims 8 to 12, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy caused by a chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of: kinase inhibitors, proteasome inhibitors, taxanes, vinca alkaloids and platinum salts, and wherein the chemotherapeutic agent is preferably selected from the group consisting of sorafenib, sunitinib, afatinib, axitinib, vandetanib, vemurafenib, ixazom, bortezomib, paclitaxel, docetaxel, cabazitaxel, vincristine, vinblastine, vindesine, vinorelbine, nedaplatin, lobaplatin, picoplatin, satraplatin, cisplatin, carboplatin and oxaliplatin.

14. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to any one of claims 8 to 13, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy caused by an antiviral agent, wherein the antiviral agent is a nucleoside reverse transcriptase inhibitor, wherein the antiviral agent is preferably selected from the group consisting of zalcitabine, didanosine, stavudine and zidovudine.

15. Use of the composition or use of the pharmaceutical composition or use of the kit according to any one of claims 8 to 14, wherein the composition or the pharmaceutical composition is used together with at least one antineoplastic agent, wherein the antineoplastic agent is preferably selected from the group consisting of kinase inhibitors, proteasome inhibitors, taxanes, vinca alkaloids and platinum salts; and wherein the antineoplastic agent is more preferably selected from the group consisting of sorafenib, sunitinib, afatinib, axitinib, vandetanib, vemurafenib, ixazomi, bortezomib, paclitaxel, docetaxel, cabazitaxel, vincristine, vinblastine, vindesine, vinorelbine, nedaplatin, lobaplatin, picoplatin, satraplatin, cisplatin, carboplatin, and oxaliplatin.

16. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to any one of claims 8 to 15, wherein the composition or the pharmaceutical composition is used together with at least one antiviral agent, wherein the antiviral agent is preferably selected from nucleosides or nucleotides, and wherein the antiviral agent is further preferably selected from zalcitabine, didanosine, stavudine and zidovudine.

17. The use of the composition or the use of the pharmaceutical composition or the use of the kit according to any one of claims 8 to 16, wherein the composition or the pharmaceutical composition is administered orally twice daily in a dose of 10mg to 3000mg per administration, more preferably 20mg to 2000mg per administration, still more preferably 50mg to 1000mg per administration.

18. Use of the composition according to any one of claims 1-4 or the pharmaceutical composition according to claim 5 or the kit according to claim 6 in a method of enhancing learning and memory.

19. A method of preparing the composition according to any one of claims 1 to 4 or the pharmaceutical composition according to claim 5, comprising combining (R) -dimiracetam (1) and (S) -dimiracetam (2), or the racemate of (R) -dimiracetam (1) and dimiracetam.

20. Use of (R) -dimiracetam (1) and/or (S) -dimiracetam (2) and/or a racemate of dimiracetam for the preparation of a composition according to any one of claims 1 to 4 or of a pharmaceutical composition according to claim 5 or of a kit according to claim 6.

21. Non-therapeutic use of the composition according to any one of claims 1-4 for enhancing learning and memory in healthy subjects.

Technical Field

The present invention relates to compositions and kits comprising (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)) and pharmaceutically acceptable solvates or co-crystals thereof, present in a certain ratio, pharmaceutical compositions comprising said compositions, their use as medicaments and the use of the compositions or pharmaceutical compositions or kits of the invention in the treatment and/or prevention of a disease or condition, typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disease states, such as seizures; depression; or a cognitive disorder; and motor neuron diseases such as amyotrophic lateral sclerosis.

Background

Glutamate is an excitatory neurotransmitter that is widely present in the brain. Its role as an excitatory messenger was first discovered in the 1950 s, when some observed convulsions from intravenous glutamic acid. However, the detection of the entire glutamatergic neurotransmitter system (including biosynthetic and catabolic enzymes, cellular uptake mechanisms, intracellular storage and release systems and their cell surface ion channels and G-protein coupled receptors) was not performed until suitable pharmacological tools were first identified in the 1970 s and 1980 s. It was in the 1990's that new molecular biology tools have provided a means for molecular identification and classification of glutamatergic ion channels, receptors, transporters, etc.

Membrane-bound ion channels, gated by the excitatory amino acids glutamate and glycine and also responsive to the exogenous compound N-methyl-D-aspartate (NMDA), control the entry of divalent and monovalent cation fluxes into presynaptic and postsynaptic neurons (see Foster et al, Nature 1987, 329: 395-396; Mayer et al, Trends in Pharmacol. Sci.1990, 11: 254-260). They are molecularly, electrophysiologically and pharmacologically distinct from glutamate-gated cation-conducting ion channels, which are responsive to the exogenous drugs kainic acid or alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA); and they are also distinct from the family of glutamate-gated G protein-coupled receptors (so-called metabotropic glutamate receptors).

NMDA-preferred glutamate-gated ion channels have a heterotetrameric structural basis: two mandatory GluN1 units and two variable GluN2 receptor subunits encoded by GRIN1 gene and one of four GRIN2 genes, respectively. One or both of the GluN2 subunits may potentially be substituted with the GluN3A or GluN3B subunit. The GRIN1 gene product has 8 splice variants, and there are 4 different GRIN2 genes (GRIN2A-D) encoding four different GluN2 subunits. The glycine binding site is present on the GluN1 subunit, while the glutamate binding site is present on the GluN2 subunit (Paoletti P et al, NatRev neurosci.2013; 14 (6): 383-.

Classes of positive or negative allosteric modulators of glutamate-gated ion channels have been described; they bind glutamate-gated ion channels at the inter-subunit interface of the Ligand Binding Domains (LBDs) of the respective ion channels, a site that is different from the glutamate or glycine binding sites present in LBDs (Sun et al, 2002; Jin et al, 2005; Hackos et al, 2016). Allosteric modulators (Wang et al, 2017) have also been described that bind to the transmembrane domain of NMDA-type glutamate-gated ion channels, wherein a highly conserved structural motif (the so-called "Lurcher domain") limits the ion flow through the pore when the ion channel is in a closed or inactivated state (Karakas and Furukawa, 2014; Lee et al, 2014; Ogden and Traynelis, 2013).

Allosteric modulators of glutamate-gated ion channels have therapeutic potential and are even useful in a variety of areas in healthy individuals, such as learning, memory management, mood, attention, emotion, motor neuron disease, peripheral sensory neuropathy, and pain perception (Cull Candy S et al, Curr Opin neurobiol. 2001; 11 (3): 327-35).

Compounds that modulate NMDA receptor function may be useful in the treatment of a number of neurological and psychiatric disorders including, but not limited to, bipolar disorder (Martucci L et al, Schizophrania Res, 2006; 84 (2-3): 214-21), major depression (Li N et al, Biol Psychiatry.2011; 69 (8): 754-61), treatment of resistant depression (Preskorn SH et al, J Clinpychophracol.2008; 28 (6): 631-7) and other mood disorders (including Schizophrenia (Grimwood S et al, neuroreport.1999; 10 (3): 461-5), prenatal and postpartum depression (Weichert CS et al, molecular hypothalamus (2013)18, 1185-1192), seasonal affective disorder, etc.; Alzheimer' S disease (Hanrson JE et al, neurol. Disco., 74: 2011: 18362; Stroke J2011J 31, 18331J 31: 29J 31, J31-31, dementia et al, J31: (Stroke 19: (dementia, J31), Parkinson 'S disease (Duty S, CNS drugs.2012; 26 (12): 1017-32; Steece Collier K et al, Exp neurol.2000; 163 (1): 239-43; Leaver Kr et al, Clin Exp Pharmacol physiol.2008; 35 (11): 1388-94), Huntington' S disease (Tang TS et al, ProcNatl Acad Sci USA 2005; 102 (7): 2602-7; Li L et al, J Neurophylliol.2004; 92 (5): 2738-46), multiple sclerosis (Grasselli G et al, Br J Pharmacol.2013; 168 (2): 502-17), cognitive disorders (Wang D et al, 2014, Expert Oprgets r; 18 (10); 1121-30), head injury (Anochen Y403; Ang J.890, Ang J.11: 11; Epilepsy J.1999; Epilepsy J.51: 11; Epilepsy J.1999; 11; Epilepsy J.11; 11: 11; Epilepsy: 11; Sci J.11; Epilepsy; 11; Sci.11; Sci.3; Sage et al, Sci.3; Epilepsy; Ostrin.11, Sci.11; Ostrin J.3; Ostrin. J, Dyskinesia (e.g., dyskinesia) (Morisette M et al, Mov Disord.2006; 21 (1): 9-17), various neurodegenerative diseases (e.g., amyotrophic lateral sclerosis (Fuller PI et al, Neurosci Lett.2006; 399 (1-2): 157-61) or neurodegenerative diseases associated with bacterial or chronic infections, glaucoma (Naskar R et al, SeminOphthalmol.1999 9 months; 14 (3): 152-8), pain (e.g., chronic, cancer, post-operative and neuropathic pain (Wu LJ and Zhuo M, Neurotherapeutics.2009; 6 (4): 693) 702), diabetic neuropathy, migraine (Peeters M et al, J Pharmacol Exp therapy R.2007; 321; 2: 564-72), cerebral ischemia (Yuan H et al, Neuron 6.1305; Nethers M et al, Neuro 6. 2015 85), Nethers M et al, J Pharmacol Exp therapy R.2007; Wolk.1098; Wolk.7, et al, nature.2012; 486(7402): 261-5), memory and learning disorders (Tang, y.p. et al, nature.1999; 401(6748): 63-9), obsessive compulsive disorder (Arnold PD et al, Psychiatry Res.2009; 172(2): 136-9.), Attention Deficit Hyperactivity Disorder (ADHD) (Dorval KM et al, Genes Brain Behav.2007; 6(5): 444-52), post-traumatic stress disorder (PTSD) (Haller J et al, Behav pharmacol.2011; 22(2): 113-21; leadebrand K et al, Neurobiol Learn Mem.2014; 113:35-40), tinnitus (Guitton MJ and Dudai Y, Neural plate.2007; 80904, respectively; hu SS et al 2016; 273(2): 325-; 18(1): 11-4; starck M et al, J neurol.1997, month 1; 244(1): 9-16), anxiety, autoimmune diseases such as neuropsychiatric systemic lupus erythematosus (Kowal C et al, proc.natl.acad.sci.u.s.a.2006; 10319854-; 3(3): 169 to 79; shen H et al, Proc NatlAcad Sci USA.2011; 108(48): 19407-12).

Symptoms of peripheral sensory neuropathy, including one of the most prominent symptoms, peripheral neuropathic pain (zilliox la, 2017), are frequently encountered in clinical situations: the prevalence is estimated between 7% and 10% in the general population (van Hecke O et al, 2014). In the united states, painful diabetic peripheral neuropathy alone is estimated to affect approximately 1000 million people. Peripheral sensory neuropathy is generally resistant to treatment and is associated with poor patient satisfaction with the treatment. Several drugs have been shown to be effective in treating peripheral sensory neuropathy associated with diabetic neuropathy and post-herpetic neuralgia, and these drugs are also commonly used to treat neuropathic pain associated with other conditions. These treatments often have undesirable side effects, and discontinuing treatment can be problematic. It is important to recognize that peripheral sensory neuropathy affects many aspects of daily life and is associated with poor overall health conditions, decreased quality of life, poor sleep, and increased anxiety and depressed mood. In fact, the quality of life assessment in patients with chronic peripheral sensory neuropathy is as low as that in patients with clinical depression, coronary artery disease, recent myocardial infarction, or poor control of diabetes (Smith-BH et al, 2007).

The american academy of neurology publishes guidelines for the treatment of painful diabetic neuropathy (Bril V et al, 2011), postherpetic neuralgia (Dubinsky Rm et al, 2004), and trigeminal neuralgia (gronesth G et al, 2008). Other clinical practice guidelines for treating neuropathic pain have also been published (Attal N et al, 2010; Moulin D et al, 2014).

Sensory neuropathy is generally classified as central or peripheral depending on the site of the lesion causing the symptoms. Diseases associated with peripheral sensory neuropathy include diabetic neuropathy, neuropathy associated with human immunodeficiency virus, chemotherapy-induced peripheral neuropathy, post-herpetic neuralgia, trigeminal neuralgia, complex regional pain syndrome, compressive mononeuropathy, radiculoneuropathy, inflammatory neuropathy (acute and chronic inflammatory demyelinating polyneuropathy), post-traumatic neuropathy, or phantom limb neuropathy.

Usually, peripheral sensory neuropathy has positive and negative symptoms. Positive symptoms include tingling ("tingling"), tingling, lightning-like or laceration-like sensations, pain, cutting, pulling or tightening-like symptoms, burning or shock-like pain. Negative symptoms include paralysis, numbness, or sensation of wearing socks. Some unique aspects of peripheral sensory neuropathy include hyperalgesia (increased response to a stimulus is often painful); allodynia (pain caused by stimuli that do not normally cause pain); hyperesthesia (increased sensitivity to stimuli); paresthesia (abnormal sensation, whether provoked or spontaneous); sensory disturbance (unpleasant abnormal sensation); sensory decline (impaired pain to normal pain stimuli); analgesia (loss of pain); anesthesia (loss of sensation). Positive signs or symptoms are considered to be sensory pathway hyperactivity due to a decrease in threshold or increased excitability. Negative signs or symptoms are manifested by diminished or diminished sensation and are attributed to loss of sensory function.

Although some pharmacological agents have been found to be effective in treating the symptoms of peripheral sensory neuropathy (Finnerup NB et al, 2015), only a few patients with neuropathic pain have a complete response to drug treatment. For most patients, it is reasonable to expect that treatment will result in pain relief. In general, a 30% reduction in pain on an 11 point value rating scale is considered clinically significant and constitutes "moderate relief" or "great improvement". It is also important to recognize and treat complications such as anxiety and depression, and secondary treatment goals may include improving sleep, enhancing function, and improving overall quality of life. These goals are best achieved when the drug therapy is an integral part of a multidisciplinary therapy approach.

Examples of neuropathic pain medications approved by the U.S. food and drug administration are carbamazepine, duloxetine, pregabalin, gabapentin, topical lidocaine, and topical capsaicin. Tramadol and opioid analgesics are effective on different types of neuropathic pain, but are generally not recommended as first-line therapy due to concerns about long-term safety. However, they are recommended as first line treatments for acute neuropathic pain, cancer-induced neuropathic pain, and exacerbations of severe neuropathic pain. The use of strong opioids (codeine, morphine, oxycodone, and fentanyl) for the treatment of various neuropathic pain is controversial and has become a public health problem given the rising number of deaths attributed to the prescribed opioids. The serious risk of overdose, dependence and addiction presented by these drugs may outweigh their potential benefits.

Therefore, there is still an urgent medical need for the development of new, orally effective treatments for peripheral sensory neuropathy and peripheral neuropathic pain, which are toxicologically benign and free of potential dependence and addictive phenomena.

In addition, there is an important medical need for the development of new, orally effective treatments for neuropsychiatric diseases, such as those described in the handbook of mental disease diagnosis and statistics (DSM-5), 5 th edition, and for the treatment of motor neuron diseases, such as amyotrophic lateral sclerosis.

Dimiracetam (2, 5-dioxohexahydro-1H-pyrrolo [1,2-a ] imidazole, IUPAC name (RS) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione) is a bicyclic 2-pyrrolidone derivative and is a nootropic agent in rasracetam class of drugs:

Figure BDA0002285200280000061

AU 2012/201853 discloses the use of dimiracetam or a pharmaceutically acceptable solvate thereof, alone or in combination with other active ingredients, for the manufacture of a medicament useful for the treatment and/or prevention of chronic pain.

WO 93/09120 relates to processes for the preparation of certain fused imidazole derivatives, in particular certain processes for the preparation of chiral fused imidazole derivatives.

Us patent 5,200,406 mentions that dimiracetam may be useful in restoring learning ability and treating memory difficulties. An example of a disease treated with dimiracetam is alzheimer's disease.

Delracetam was originally developed as a cognitive enhancer and has been shown to improve learning and memory in rats (Pinza M et al, 1993; EP 335483). In a single dose healthy human volunteer study (Torchio L et al, 1995), it was found that dimiracetam improved certain indicators of transient decline in cognitive function caused by injection of scopolamine compared to placebo. Other medical uses of dimiracetam have been described, including its broad efficacy in rodent models of neuropathic pain, among others. The efficacy of dimiracetam in the treatment of neuropathic pain of different origins has been demonstrated in established models of neuropathic pain caused by nerve injury, chemotherapy or mono-iodoacetate (MIA) induced osteoarthritis (Fariello R et al, 2014); di Cesare mannell et al, 2015 a; di Cesare mannell et al, 2015 b; WO 2008/125674; EP 2857017B1, US 2010/0125096; WO 2012/055057). Regardless of the chemotherapeutic agent used, neurotoxic symptoms caused by chemotherapy are responsive to dimiracetam: models derived from dideoxycytidine (ddC-), oxaliplatin, vincristine, paclitaxel and sorafenib all reacted with dimiracetam; and it has been demonstrated that dimiracetam is not only effective in treating, but also in preventing the symptoms caused by the administration of these chemotherapeutic agents. A single oral administration of dimiracetam can completely but transiently restore hyperalgesia and allodynia to healthy control levels. The oral dosing was repeated twice daily and even if the dose reduction and the dose interval were increased to the level of once daily oral dosing, the maximal effect was sustained with no evidence of a rapid drug resistance response or tolerance. Moreover, the role of dimiracetam varies with the disease: in a unilateral Chronic Compressive Injury (CCI) model, rats exhibited neuropathic pain in the periphery after surgical ligation of the sciatic nerve periphery in one hindlimb, but not in the other limb undergoing sham surgery, a single oral dose of dimiracetam only reduced the pain response in the nerve-ligated limb, without hyperalgesia or allodynia affecting the sham limb; this is clearly different from, for example, the effects of opioids, which affect both limbs of the model (Christensen D et al, 1998).

The pharmacological mechanism of action of dimiracetam has been explored using synaptosomal preparations of the hippocampus and spinal cord. This test is intended to mimic the physiological process of glutamate-triggered glutamate release from a pharmacological perspective; its pH-, Zn2+And ifenprodil-sensitivity indicates the intervention of NMDA receptor isoforms containing pH-sensitive GluN1 and GluN2A subunits (Fariello et al, 2014). Inhibition of glutamate signaling is the basis for the prevention or treatment of neuropathic pain (Latremolier and Woolf, 2009). In the spinal cord, glutamate-induced at the junction where peripheral sensory afferents form the first and only synaptic junction with central nervous system interneurons (Marieb, Wilhelm and Mallat, 2017)Glutamate release is a component of an up-regulated or "sensitizing" signal caused by damaged peripheral nerves (Latremolier and Woolf, 2009).

In the preparation of synaptosome of hippocampus, dimiracetam is a drug inhibiting NMDA plus glycine triggering which is preloaded in the preparation of synapse3H]D-aspartic acid release is a moderately effective inhibitor, IC50About 3. mu.M. However, in synaptosome preparations of the spinal cord, dimiracetam is much more potent, inhibiting NMDA plus glycine triggered [, [ 2 ] ]3H]IC of-D-aspartic acid release50About 20nM (Fariello R et al, 2014).

The ability of dimiracetam to prevent glutamate-triggered release in the spinal cord is the basis for its use in the prevention or treatment of peripheral sensory neuropathy; other mechanisms in the brain may be the basis for its effective treatment of rat depression (Fariello et al, 2011; WO 2015/010217), as well as its effectiveness in rat and human models of scopolamine-induced cognitive impairment (Pinza et al, 1993).

Dimiracetam is a chiral compound with a single stereocenter, but is a racemic mixture of its (R) -and (S) -enantiomers, which has been developed clinically. Even though (R) -dimiracetam is the more active enantiomer (WO 2008/125674), this was done because the racemate of dimiracetam has been found to be more effective than either single enantiomer. For example, in rats previously treated with 2',3' -dideoxycytidine (ddC, zalcitabine), a single oral administration of (R) -dimiracetam resulted in a partially effective response, while the (S) -enantiomer resulted in a response that was less than the corresponding dose of (R) -enantiomer. On the other hand, racemic dimiracetam gives a better response than (R) -or (S) -dimiracetam alone (WO 2008/125674). This rank order of potency of (S) -, (R) -and racemic dimiracetam is also seen in the restorative effect of dimiracetam on MIA-induced hyperalgesia (WO 2008/125674).

Disclosure of Invention

It has now been surprisingly and unexpectedly found that (R) -and (S) -dimiracetam with an enantiomeric excess (ee) of (R) -dimiracetam (1) of greater than or equal to 30% and less than or equal to 60% >The combination of enantiomers shows a pharmacological potency greater than that of the corresponding single enantiomer or even than that of the racemate, thus providing a synergistic effect that cannot be predicted from the potency of the single enantiomer or racemate. These compositions preferably inhibit NMDA plus glycine induced [ alpha ], [ beta ] -cyclodextrin at a concentration of about 10nM3H]-release of D-aspartic acid from rat synaptosomes of at least about 40%, preferably at least about 45%.

Thus, it has been surprisingly and unexpectedly found that preferred compositions of the present invention having a (R) -dimiracetam enantiomeric excess of 33% (corresponding to a ratio of (R) to (S) of the dimiracetam enantiomer of 2:1) inhibit NMDA plus glycine induced at a concentration of 10nM3H]Release of-D-aspartic acid from rat synaptosomes was about 50% (fig. 1A, table 1). Another preferred composition of the invention having an enantiomeric excess of (R) -dimiracetam of 50% (corresponding to the (R) to (S) ratio of the dimiracetam enantiomer of 3:1) inhibits NMDA plus glycine induced [ alpha ], [ beta ]3H]The release of-D-aspartate from rat synaptosomes reached 52% (FIG. 1B, Table 1). In contrast, racemic dimiracetam inhibits NMDA plus glycine induced at a concentration of 10nM3H]36% release of D-aspartic acid from rat spinal synaptosomes, corresponding to the estimated IC5015nM (FIG. 1C; Table 1); (R) -Delaware estimation IC50123nM (FIG. 1D); (S) -Delaware estimation IC50At 418nM (FIG. 1E).

These surprising results were demonstrated in other different rat models of peripheral neuropathic pain, such as the MIA-induced gonarthritis model or the oxaliplatin-induced neuropathic pain model. Thus, the composition of the invention with an enantiomeric excess of (R) -dimiracetam is more effective in reducing peripheral neuropathic pain (fig. 2), or in preventing oxaliplatin-induced peripheral neuropathic pain (fig. 3), than the racemic mixture of dimiracetam in paw pressure tests after administration of sodium Monoiodoacetate (MIA) (fig. 2). The compositions of the invention with an enantiomeric excess of (R) -dimiracetam are also more effective than compositions in which the excess of (S) -dimiracetam enantiomer is present (fig. 2).

Thus, a combination of the (R) -and (S) -enantiomers of dimiracetam of the invention having an enantiomeric excess (ee) of (R) -dimiracetam greater than or equal to 30% and less than or equal to 60%, is more pharmaceutically effective at a given dose than the pure enantiomer or racemic dimiracetam alone. The term "racemic dimiracetam" refers to a mixture of (R) -and (S) -enantiomers in a weight ratio of 1:1, so that its (R) -dimiracetam enantiomeric excess (ee) is 0%. Thus, the effect associated with the present invention is a synergistic effect, which is surprisingly due to the specific range of ratios between (R) -dimiracetam and (S) -dimiracetam.

Since the plasma concentration-time curves of rats after oral administration of (R) -or (S) -dimiracetam are identical and the (R) -and (S) -enantiomers are not interconverted in vivo (see fig. 4A and fig. 4B, respectively), the behavior of the enantiomeric composition of the present invention cannot be explained by the pharmacokinetics or metabolism of the enantiomers. Indeed, both the (R) -and (S) -enantiomers are individually potent (although not of equal potency) and when combined in appropriate proportions, have pharmacological potency greater than that of the racemate, indicating that they share the same pharmacological mechanisms as racemic dimiracetam and are suitable for use in the treatment of the same medical indications. Thus, the compositions of the present invention are beneficial and can be used to treat and/or prevent a wide variety of diseases and conditions as described in the detailed description. The disease or condition is typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases such as amyotrophic lateral sclerosis. More preferably, the disease or condition is typically selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain, seizures, depression or cognitive disorders.

In a first aspect, the present invention provides a composition comprising (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)),

Figure BDA0002285200280000091

and/or pharmaceutically acceptable solvates or co-crystals thereof,

wherein the enantiomeric excess (ee) of the (R) -dimiracetam (1) is equal to or higher than 30% and lower than or equal to 60%.

A specific example of said composition according to the invention is a non-racemic mixture of 3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione (dimiracetam) and pharmaceutically acceptable solvates or co-crystals thereof, wherein said non-racemic mixture comprises an enantiomeric excess (ee) of (R) -dimiracetam (1) of greater than or equal to 30% and less than or equal to 60% of (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)).

Figure BDA0002285200280000101

In another aspect, the invention provides a pharmaceutical composition comprising the composition of the invention, e.g., the non-racemic mixture of the invention, and a pharmaceutically acceptable carrier.

In yet another aspect, the invention provides a kit of parts comprising (R) -dimiracetam (1) and (S) -dimiracetam (2) and instructions for combining (R) -dimiracetam (1) and (S) -dimiracetam (2) to achieve an enantiomeric excess (ee) of said (R) -dimiracetam (1) that is equal to or greater than 30% and less than or equal to 60%. The same preferred ranges given herein for the enantiomeric excess (ee) of (R) -dimiracetam (1) and the enantiomeric ratio of (R) -dimiracetam (1) to (S) -dimiracetam (2) apply in this respect.

In a further aspect, the invention provides a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention for use as a medicament.

In a further aspect, the invention provides the use of a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention for the treatment or prevention of a disease or condition, wherein said disease or condition is typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases such as amyotrophic lateral sclerosis.

In a further aspect, the invention provides a method of treating and/or preventing a disease or condition in an animal (preferably a human), wherein said disease or condition is typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases, such as amyotrophic lateral sclerosis, wherein the method comprises administering a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention to an animal (preferably a human).

In another aspect, the invention provides the use of a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention for the manufacture of a medicament for the treatment and/or prevention of a disease or condition, wherein said disease or condition is typically and preferably selected from the group consisting of peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases such as amyotrophic lateral sclerosis.

In yet another aspect, the invention provides an article of manufacture comprising a composition of the invention or a pharmaceutical composition of the invention, a container or package, and written instructions such as a package insert and instructions for administration.

It is to be understood that the compositions of the present application include non-racemic mixtures of the present invention and thus any reference to a composition of the present invention is also to be understood as a reference to a non-racemic mixture of the present invention.

Other aspects and embodiments of the invention will become apparent as the description of the specification proceeds.

Drawings

FIG. 1A: increasing the molarity of a composition of the invention having an enantiomeric excess of (R) -dimiracetam of 33.3% (corresponding to an enantiomeric (R): S) ratio of 2:1) stimulated NMDA (10. mu.M) plus glycine (1. mu.M)3H]-effect of release of D-Asp from pre-loaded spinal synaptosomes.

FIG. 1B: increasing the molar concentration of a composition of the invention having an enantiomeric excess of (R) -dimiracetam of 50% (corresponding to an enantiomeric (R): S) ratio of 3:1), NMDA (10. mu.M) plus glycine (1. mu.M) stimulated3H]-effect of release of D-Asp from pre-loaded spinal synaptosomes.

FIG. 1C: increasing the molarity of the deslecetam racemate for NMDA (10. mu.M) plus glycine (1. mu.M) stimulation3H]-effect of release of D-Asp from pre-loaded spinal synaptosomes.

FIG. 1D: increasing the molarity of (R) -dimiracetam on NMDA (10. mu.M) plus glycine (1. mu.M) stimulation3H]-effect of release of D-Asp from pre-loaded spinal synaptosomes.

FIG. 1E: increasing the molarity of (S) -dimiracetam on NMDA (10. mu.M) plus glycine (1. mu.M) stimulation3H]-effect of release of D-Asp from pre-loaded spinal synaptosomes.

In fig. 1A-E, results are expressed as% increase in basal release, data are mean ± s.e.m of 6 experiments performed in triplicate.

FIG. 2: in the MIA-induced rat osteoarthritis model, the anti-hyperalgesic effect of a composition of the invention with a single dose of (R) -dimiracetam enantiomeric excess of 50% (corresponding to an enantiomeric (R): (S) ratio of 3:1) compared to the dimiracetam racemate and a dimiracetam composition with an (S) -enantiomeric excess ((R): (S) ratio of 1: 3). Pain threshold is determined by Randall&Selitto analgesic apparatus evaluation. Results are expressed in grams, each value representing the mean ± s.e.m. of 20 rats. With respect to the vehicle-MIA,**:P<0.01。

FIG. 3: repeated oral administration of a composition of the invention with 50% enantiomeric excess of (R) -dimiracetam (corresponding to a 3:1 ratio of enantiomer (R): S) and the effect of dimiracetam racemate on mechanical hyperalgesia caused by oxaliplatin. In the morningBefore morning administration, Randall is administered&The Selitto analgesic apparatus assessed pain thresholds, each value representing the mean ± s.e.m. of 6 rats. Relative to the animals treated with oxaliplatin + vehicle,*P<0.05 and**P<0.01. relative to oxaliplatin + delacetam 15mg/kg, ° P<0.05 and DEG P<0.01; relative to oxaliplatin + dimiracetam 50mg/kg,#P<0.05 and##P<0.01。

FIG. 4A: comparison of plasma concentration-time curves of the (R) -enantiomer after oral administration of (R) -dimiracetam (75mg/kg) and after oral administration of racemic dimiracetam (150 mg/kg).

FIG. 4B: comparison of plasma concentration-time curves of the (S) -enantiomer after oral administration of (S) -dimiracetam (2) (75mg/kg) and after oral administration of racemic dimiracetam (150 mg/kg).

Figures 4A and 4B demonstrate that the in vivo pharmacological superiority of the compositions of the invention is not due to pharmacokinetic differences.

FIG. 5: racemic dilaceptan (circles) and five different enantiomeric mixtures (circles with R to S ratios from 2:1 to 4: 1) inhibit NMDA + glycine induced3H]D-aspartic acid release (squares). This data was obtained in example 5.

FIG. 6: passive avoidance testing. Three mixtures of the enantiomers of dimiracetam with different R: S ratios (1:1, 2:1 and 3:1) were administered orally 30 minutes before the training session on day 1, at doses of 3, 10 or 30 mg/kg. Scopolamine (1.5mg/kg i.p.) was injected immediately after the 1 st day penalty. The latencies recorded in the training sessions were comparable in all groups (about 15 seconds). On day 2, a reservation session is performed and the latency is reported in a histogram. Data are presented as mean ± s.e.m of 12 mice in2 different experimental groups. Relative to vehicle + vehicle (vehicle),##P<0.01; relative to scopolamine + vehicle (scopolamine),*P<0.05 and**P<0.01。

FIG. 7: forced swimming test. Mixtures of three different R: S ratios (1:1, 2:1 and 3:1) of the enantiomers of dimiracetam were administered orally at doses of 10, 30 or 100mg/kg 25 minutes before the test. The mice were placed in a glass cylinder filled with water for 6 minutes and the last 4 minutes were recordedThe duration of the movement. Data are presented as mean ± s.e.m. of 12 mice analyzed in2 different experimental groups. Relative to vehicle + vehicle (vehicle) (indicating treatment of animals with vehicle instead of scopolamine, followed by treatment again with vehicle instead of test compound),*P<0.05 and**P<0.01。

Detailed Description

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

The term "about" as used to indicate enantiomeric excess refers to ± 4% of a given value, if not otherwise indicated. In each embodiment of the invention, "about" may be deleted.

The term "preferably" is used to describe features or embodiments that are not required in the present invention but which may lead to an improved technical effect and is therefore desirable but not necessary.

For the numerical values mentioned herein, the last decimal place of the numerical value preferably indicates the degree of precision thereof, unless explicitly stated otherwise. Therefore, unless other error margins are given, the maximum margin is preferably determined by applying a rounding rule to the last decimal point. Therefore, a value of 2.5 preferably has a margin of error of 2.45 to 2.54.

The present invention relates to a composition comprising (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)) present in a certain ratio. It is to be understood that the term "composition" does not require that (R) -dimiracetam (1) and (S) -dimiracetam (2) must be mixed. They may be formulated in combination or separately and may be administered simultaneously or sequentially, as long as the ratio of (R) -dimiracetam (1) and (S) -dimiracetam (2) achieved in the subject to be treated meets the requirements of the invention. Preferably, the composition of the invention is a mixture of (R) -dimiracetam (1) and (S) -dimiracetam (2), but the composition of the invention can also be a combination comprising one or more articles containing (R) -dimiracetam (1) and one or more articles containing (S) -dimiracetam (2), or a combination of one or more articles containing (R) -dimiracetam (1) and one or more articles containing dimiracetam racemate.

Furthermore, the dimiracetam contained in the composition according to the invention must be present in the range of the total ratio of (R) -dimiracetam (1) and (S) -dimiracetam (2), or in the range of the (R) -dimiracetam (1) enantiomeric excess indicated as required for the invention. In other words, it is against the gist of the present invention to theoretically separate a composition comprising equal amounts of (R) -dimiracetam (1) and (S) -dimiracetam (2) into one component containing an excess of (R) -dimiracetam (1) and another component containing an excess of (S) -dimiracetam (2). Thus, whatever the physical form in which the composition of the invention is present, the composition as a whole must satisfy the requirements as regards the range of proportions of (R) -dimiracetam (1) and (S) -dimiracetam (2), or expressed as enantiomeric excess of (R) -dimiracetam (1) of the invention. It will be appreciated that the ratio of (R) -dimiracetam (1) and (S) -dimiracetam (2), or the enantiomeric excess expressed as (R) -dimiracetam (1), is based on the number of statistically significant dimiracetam molecules, typically exceeding 1000 dimiracetam molecules. In the present invention, the relative amounts of (R) -dimiracetam (1) and (S) -dimiracetam (2) are expressed as the ratio of (R) -dimiracetam (1) to (S) -dimiracetam (2) or as the enantiomeric excess of (R) -dimiracetam (1).

It is to be understood that as used herein, unless otherwise specifically indicated, "ratio" of (R) -dimiracetam (1) to (S) -dimiracetam (2) refers to the weight ratio of (R) -dimiracetam to (S) -dimiracetam (2). If solvates of (R) -dimiracetam (1) and/or (S) -dimiracetam (2) are used, the solvent is not taken into account in the present calculation. In other words, the ratio of "(R) -dimiracetam (1) and (S) -dimiracetam (2) is calculated as follows:

Figure BDA0002285200280000141

as known to those skilled in the art, the proportion of compounds that differ only chirally, for example in the case of (R) -dimiracetam (1) and (S) -dimiracetam (2), can be determined in a number of ways known in the art, including but not limited to chromatography using chiral supports, polarized light measurement with polarized light rotation, nuclear magnetic resonance spectroscopy using chiral shift reagents, or derivatization of compounds using chiral compounds such as moservic acid (Mosher acid) followed by analysis by chromatography or nuclear magnetic resonance spectroscopy. Enantiomers can also be separated from mixtures by methods known to those skilled in the art, including chiral High Pressure Liquid Chromatography (HPLC) and direct fractional crystallization of the racemate (i.e., dimiracetam) by chiral co-crystallization techniques, which exploit the formation of specific hydrogen bonding interactions present in co-crystals (see Springuel GR et al, 2012; and U.S. Pat. No. 6,570,036). Useful co-crystallization partners include enantiomers of mandelic acid, malic acid, tartaric acid and derivatives thereof; or enantiomers which can be prepared by asymmetric synthesis. See, e.g., Eliel and Wilen, 1994.

The ratio of (R) -dimiracetam (1) and (S) -dimiracetam (2) (which may also be referred to as chiral purity) of the compositions of the invention (e.g. non-racemic mixtures) can also be expressed in terms of their enantiomeric excess (ee), as determined generally and preferably by chiral HPLC (see examples for details), and is calculated by the following formula:

ee=(AR–AS)/(AR+AS)×100%,

wherein A isRIs (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] in an HPLC chromatogram of a sample solution]Peak area of imidazole-2, 5-dione (i.e., (R) -enantiomer (1) of dimiracetam); a. theSIs (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] in an HPLC chromatogram of a sample solution]Peak area of imidazole-2 (i.e. (S) -enantiomer (2) of dimiracetam).

In this respect, it should be noted that, although chiral "purity" is mentioned above, the gist of the present invention is not to obtain high chiral purity of (R) -dimiracetam (1) or (S) -dimiracetam (2). In contrast, the gist of the present invention is that a certain range of the ratio of (R) -dimiracetam (1) to (S) -dimiracetam (2) results in a particular synergistic effect. The basis of the present invention is the previously unknown ratio of the two compounds, i.e. (R) -dimiracetam (1) and (S) -dimiracetam (2), as opposed to merely improving the purity of the compounds, i.e. the target is known and one particular compound is to be obtained in the desired 100% purity.

The term "pharmaceutically acceptable" means that the compound or composition (typically and preferably, solvate, co-crystal or carrier) must be chemically or toxicologically compatible with the other ingredients (typically and preferably, the composition of the invention) when typically and preferably used in a formulation or typically and preferably used in the treatment of an animal (preferably a human) therewith. Preferably, the term "pharmaceutically acceptable" means that the compound or composition (typically and preferably, solvate, co-crystal or carrier) must be chemically and toxicologically compatible with the other ingredients (typically and preferably, the composition of the invention) when typically and preferably used in a formulation or typically and preferably used in the treatment of an animal (preferably a human) therewith. It is to be noted that this can be done by techniques known to those skilled in The art (for example, in "Remington: The Science and Practice of Pharmacy", Pharmaceutical Press, 22ndedition, the techniques disclosed in (e) et al) to formulate a pharmaceutical composition.

"solvate" refers to an association or complex of one or more solvent molecules with the (R) -enantiomer of dimiracetam (1) or the (S) -enantiomer of dimiracetam (2). Examples of solvate-forming solvents include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide (DMSO), ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" refers to a complex in which the solvent molecule is water.

"cocrystal" refers to a crystal structure comprising at least two different compounds that are solid in their pure form at ambient conditions. The at least two different compounds may comprise (R) -dimiracetam (1) and/or (S) -dimiracetam (2) and/or any other component of the composition or excipient of the pharmaceutical composition. The co-crystal consists of neutral molecular species, all of which remain neutral after crystallization; furthermore, typically and preferably, they are crystalline homogeneous substances in which two or more building compounds are present in a defined stoichiometric ratio. See Wang Y and Chen a, 2013; SpringuelGR et al, 2012; and us patent 6,570,036. It is to be understood that (R) -dimiracetam (1) and/or (S) -dimiracetam (2) may be in the form of any polymorph. Various Co-crystals and techniques for preparing such Co-crystals are described in "RSC Drug Discovery, Pharmaceutical Salts and Co-crystals" (in particular chapters 15 and 16 thereof), edited by Johan Wouters and Luc Qu é, published by the royal chemical society in 2012. Preferred examples of co-crystal formers are those disclosed in table 16.1 of this reference. Further preferred co-crystals include co-crystals of alpha-hydroxy acids, alpha-keto acids and/or alpha-keto amides with the enantiomer of dimiracetam having a (R)/(S) ratio as disclosed herein. Examples of alpha-hydroxy acids include atrolactic acid, benzoic acid, 4-chloromandelic acid, citric acid, 3, 4-dihydroxymandelic acid, ethyl pyruvate, galacturonic acid, gluconolactone, glucuronic acid, glucuronolactone, glycolic acid, 2-hydroxybutyric acid, 2-hydroxyvaleric acid, 2-hydroxyhexanoic acid, 2-hydroxyheptanoic acid, 2-hydroxyoctanoic acid, 2-hydroxynonanoic acid, 2-hydroxydecanoic acid, 2-hydroxyundecanoic acid, 4-hydroxymandelic acid, 3-hydroxy-4-methoxymandelic acid, 4-hydroxy-3-methoxymandelic acid, alpha-hydroxyarachidonic acid, alpha-hydroxybutyric acid, alpha-hydroxyisobutyric acid, alpha-hydroxylauric acid, alpha-hydroxymyristic acid, alpha-hydroxymandelic acid, alpha-hydroxymande, Alpha-hydroxypalmitic acid, alpha-hydroxystearic acid, 3- (2 '-hydroxyphenyl) lactic acid, 3- (4' -hydroxyphenyl) lactic acid, malic acid, mandelic acid, methyl lactic acid, methyl pyruvate, mucic acid, alpha-phenylacetic acid, alpha-phenylpyruvic acid, pyruvic acid, saccharic acid, tartaric acid and tartronic acid. Examples of alpha-keto acids include 2-keto acetic acid (glyoxylic acid), 2-keto methyl acetate, 2-keto propionic acid (pyruvic acid), 2-keto methyl propionate (pyruvic acid methyl ester), 2-keto ethyl propionate (pyruvic acid ethyl ester), 2-keto propyl propionate (pyruvic acid propyl ester), 2-phenyl-2-keto acetic acid (benzoylcarboxylic acid), 2-phenyl-2-keto methyl acetate (benzoylcarboxylic acid methyl ester), 2-phenyl-2-keto ethyl acetate (benzoylcarboxylic acid ethyl ester), 3-phenyl-2-keto propionic acid (phenylpyruvic acid), 3-phenyl-2-keto methyl propionate (phenylpyruvic acid methyl ester), 3-phenyl-2-keto ethyl propionate (phenylpyruvic acid ethyl ester), 2-ketobutyric acid, 2-ketovaleric acid, 2-ketocaproic acid, 2-ketoheptanoic acid, 2-ketocaprylic acid, 2-ketocapric acid and methyl 2-ketocaprylate. Examples of α -ketoamides include any compounds obtainable by reacting any of the examples of α -ketoacids described above with a primary or secondary amine.

In a first aspect, the present invention provides a composition comprising (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)),

Figure BDA0002285200280000171

and/or pharmaceutically acceptable solvates or co-crystals thereof,

wherein the enantiomeric excess (ee) of the (R) -dimiracetam (1) is equal to or higher than 30% and lower than or equal to 60%. This composition, as well as any other compositions and pharmaceutical compositions according to the present invention, preferably inhibits NMDA plus glycine induced release of [3H ] -D-aspartic acid from rat spinal synaptosomes at a concentration of about 10nM, preferably at least about 36%, preferably at least about 40%, preferably at least about 45%, even more preferably about 50%. Example 5 shows the measurement method for measuring this parameter.

An example of such a composition is a non-racemic mixture of 3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione and a pharmaceutically acceptable solvate or co-crystal thereof, wherein the non-racemic mixture comprises (R) -dimiracetam (1) with an enantiomeric excess (ee) of greater than or equal to 30% and less than or equal to 60% (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)).

Figure BDA0002285200280000181

Generally, non-solvated or non-co-crystallized compositions are preferred. Further preferred are non-solvated and non-co-crystallized compositions.

Thus, in another aspect, the present invention provides a composition of 3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione, wherein the composition comprises (R) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((R) -dimiracetam (1)) and (S) -3,6,7,7 a-tetrahydro-1H-pyrrolo [1,5-a ] imidazole-2, 5-dione ((S) -dimiracetam (2)), the enantiomeric excess (ee) of (R) -dimiracetam (1) being greater than or equal to 30% and less than or equal to 60%.

More preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 30% and less than or equal to about 54%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 30% and less than or equal to 54%.

More preferably, the enantiomeric excess (ee) of the (R) -dimiracetam (1) is greater than or equal to about 33% and less than or equal to about 54%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 33% and less than or equal to 54%.

More preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 30% and less than or equal to about 53%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 30% and less than or equal to 53%.

More preferably, the enantiomeric excess (ee) of the (R) -dimiracetam (1) is greater than or equal to about 33% and less than or equal to about 53%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 33% and less than or equal to 53%.

More preferably, the enantiomeric excess (ee) of the (R) -dimiracetam (1) is greater than or equal to about 33% and less than or equal to about 50%. Still more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 33% and less than or equal to 50%.

More preferably, the enantiomeric excess (ee) of the (R) -dimiracetam (1) is greater than or equal to about 30% and less than or equal to about 50%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 30% and less than or equal to 50%.

More preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to about 35% and less than or equal to about 54%, and preferably less than or equal to about 53%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 35% and less than or equal to 54%, and preferably less than or equal to 53%.

More preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to about 40% and less than or equal to about 54%, and preferably less than or equal to about 53%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 40% and less than or equal to 54%, and preferably less than or equal to 53%.

More preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to about 45% and less than or equal to about 54%, and preferably less than or equal to about 53%. Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is greater than or equal to 45% and less than or equal to 54%, and preferably less than or equal to 53%.

Still more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is selected from the group consisting of about 33%, about 35%, about 37%, about 39%, about 41%, about 43%, about 45%, about 47%, about 50% and about 53%. Still more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is selected from the group consisting of 33%, 35%, 37%, 39%, 41%, 43%, 45%, 47%, 50% and 53%.

Even more preferably, said enantiomeric excess (ee) of said (R) -dimiracetam (1) is selected from about 50%.

Even more preferably, the enantiomeric excess (ee) of the (R) -dimiracetam (1) is 50%.

As known to those skilled in the art, the enantiomeric excess can be replaced by the ratio of (R) -dimiracetam (1) to (S) -dimiracetam (2). The preferred range of the ratio of (R) -dimiracetam (1) to (S) -dimiracetam (2) is 2:1 to 3.5:1, preferably 2:1 to 3.3:1, more preferably 2.0:1.0 to 3.3:1.0, even more preferably 2.00:1.00 to 3.30: 1.00. Further preferred ranges are 2.0:1.0 to 3.25:1.0, 2.00:1.00 to 3.25:1.00, 2:1 to 3:1, 2.0:1.0 to 3.0:1.0 and 2.00:1.00 to 3.00: 1.00. Further preferred ranges include 2.3:1 to 3.3:1, 2.6:1 to 3.3:1, 2.7:1 to 3.2:1, 2.8:1 to 3.2:1, 2.9:1 to 3.1:1, and 3:1 and 3.0: 1.0. Other preferred ranges include 2.1:1 to 2.9:1, 2.2:1 to 2.8:1, 2.3:1 to 2.7:1, 2.4:1 to 2.6:1, and 2.5:1 and 2.5: 1.0.

In another aspect, the invention provides a pharmaceutical composition comprising a composition of the invention and a pharmaceutically acceptable carrier.

In another aspect, the invention provides a kit of parts comprising (R) -dimiracetam (1) and (S) -dimiracetam (2) and instructions for combining (R) -dimiracetam (1) and (S) -dimiracetam (2) to achieve an enantiomeric excess (ee) of said (R) -dimiracetam (1) of equal to or greater than 30% and less than or equal to 60%. Hereinafter, it is to be understood that whenever the use of the composition of the invention is described, the kit according to the invention may alternatively be used. It will be understood by those skilled in the art that the components of the kit may be combined prior to administration (which is preferred), or the components of the kit may be administered separately. In the latter case, the components of the kit are typically administered over a time frame of up to 30 minutes to achieve the effect of the invention.

In a further aspect, the invention provides a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention for use as a medicament.

In another aspect, the invention provides the use of a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention in the treatment or prevention of various diseases and conditions, such as those described below:

a) for preventing or treating positive symptoms of peripheral neuropathy, including cold sensitivity, tingling, burning or painful sensations, such as those associated with chemotherapy, proliferation-inhibiting therapy, viral infections and treatments, post-herpetic neuralgia, osteonecrosis, trigeminal neuralgia or diabetic peripheral neuropathy, including primary allodynia, secondary allodynia or other pain or discomfort associated with sensitization of the spinal cord or higher brain structures or neural pathways;

b) for the prevention or treatment of pain, including skeletal and joint pain, osteonecrosis pain, repetitive motion pain, dental pain, dysmenorrhea pain, cancer pain, myofascial pain, surgical pain, perioperative pain, and post-operative pain syndromes, such as post-mastectomy syndrome, post-thoracotomy syndrome, or stump pain, as well as pain associated with angina, neuroma pain, complex regional pain syndrome, chronic pelvic pain, chronic lower back pain;

c) for the prevention or treatment of inflammatory pain, such as osteoarthritis, rheumatoid arthritis, rheumatism, chronic arthritis pain and associated neuralgia, tenosynovitis and gout;

d) for the prevention or treatment of neuropathic pain, such as chemotherapy-induced pain, post-traumatic pain, crush pain, painful traumatic mononeuropathy, painful polyneuropathy, pain caused by spinal injury, low back pain, nerve compression or entrapment, sacral pain, trigeminal neuralgia, migraine and migraine headaches, post-herpetic neuralgia, phantom limb pain, post-herpetic pain, diabetic neuropathy, and pathologies of all levels of the peripheral nervous system resulting from central pain syndrome;

e) for the prevention or treatment of neuropsychiatric diseases, examples of which include schizophrenia, psychosis including schizophrenia, schizophreniform symptoms, schizoaffective disorders, delusional disorders, brief psychotic disorders, substance-related disorders, paranoid schizophrenia, disorganized schizophrenia, catatonic schizophrenia, undifferentiated schizophrenia, substance-induced psychotic disorders, substance-related disorders and addictive behaviors;

f) focal and generalized epilepsy and other seizures;

g) obesity or other eating disorders associated with excessive food intake, bulimia nervosa;

h) cerebral damage following stroke, cerebral edema, cerebral ischemia, cerebral hemorrhage, neurodegenerative disease, cardiac bypass surgery and transplantation, perinatal hypoxia, cardiac arrest, and hypoglycemic brain injury;

i) sleep disorders, such as insomnia, narcolepsy, or restless legs;

j) anxiety disorders such as affective disorders, panic attacks, panic disorders, acute stress disorders, agoraphobia, generalized anxiety disorder, obsessive compulsive disorder, post-traumatic stress disorder, separation anxiety disorder, social phobia, specific subject phobia, substance-induced anxiety disorder;

k) mood disorders such as depression, anhedonia, unipolar depression, bipolar disorder, psychotic depression;

l) substance addiction, drug dependence, tolerance, dependence on or withdrawal from substances including alcohol, amphetamines, cannabis, ***e, hallucinogens, inhalants, nicotine, opioids, phencyclidine, sedatives, hypnotics, or anxiolytics;

m) impaired cognitive function, such as age-related cognitive decline, or cognitive disorders, such as dementia associated with alzheimer's disease, ischemia, trauma, vascular problems or stroke, HIV disease, parkinson's disease, huntington's disease, pick's disease, creutzfeldt-jakob disease, chemotherapy, perinatal hypoxia, other general medical conditions, or drug abuse;

n) Parkinson's disease, including drug-induced Parkinson's disease or post-encephalitic Parkinson's disease;

o) attention deficit disorders, such as Attention Deficit Hyperactivity Disorder (ADHD), obsessive-compulsive disorder, phobias, post-traumatic stress disorder syndrome, autism and autism spectrum disorders, impulse control disorders;

p) tinnitus, presbycusis;

q) enhancing learning and memory;

r) for the prevention or treatment of genetic or sporadic motor neuron diseases, examples of which include amyotrophic lateral sclerosis, primary lateral sclerosis, progressive muscle atrophy, progressive bulbar palsy, friedrich's ataxia, fragile-X syndrome;

s) for the prevention or treatment of movement disorders, examples of which include dystonia, chorea, including Huntington's chorea, Parkinson's-related dystonia, Creutzfeld-Jakob disease, progressive supranuclear palsy, multiple system atrophy, corticobasal degeneration, basal ganglia calcification;

t) for akinesia, such as rigidity-of-motility syndrome,

u) for movement disorders such as drug-induced Parkinson's disease, e.g. antipsychotic-induced Parkinson's disease, antipsychotic-induced malignant syndrome, antipsychotic-induced acute dystonia, antipsychotic-induced acute akathisia, antipsychotic-induced tardive dyskinesia and drug-induced postural tremor, including resting tremor, postural tremor and intention tremor, chorea (e.g. Sydney's chorea, Huntington's chorea, benign hereditary chorea, neuroerythrocytosis, symptomatic chorea, drug-induced chorea and hemichorea), systemic or focal myoclonus, tics (including simple, complex and symptomatic dystonia) and dystonia (including iodine-induced dystonia, drug-induced dystonia, etc.) Symptomatic and paroxysmal dystonia, and local dystonia such as blepharospasm, oromaxillofacial dystonia, spasmodic vocalization, spasmodic torticollis, axonal dystonia, dystonic writer's spasm, and hemiplegic dystonia), muscle spasms, and diseases associated with muscle spasms or weakness;

v) and for urinary incontinence, multiple system atrophy, tuberous sclerosis, olivopontocerebellar atrophy, cerebral palsy, drug optic neuritis, ischemic retinopathy, diabetic retinopathy, glaucoma, spasms, myoclonus, and dyskinesia associated with tourette's syndrome.

It should be understood that the above list of diseases is given only as a specific example and should not be construed as limiting the invention. Among them, preferred is one or more selected from a), e), q), r) and s).

The disease or condition is typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases such as amyotrophic lateral sclerosis.

In addition, the compositions of the present invention may also be used in non-therapeutically useful forms to enhance learning and memory in healthy subjects (e.g., healthy humans).

In a further aspect, the invention provides a method for the treatment and/or prevention of a disease or condition, wherein the disease or condition is typically and preferably selected from peripheral sensory neuropathy, preferably selected from peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases, such as amyotrophic lateral sclerosis, wherein the method comprises administering a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention.

It is also part of the present invention to provide a method of treating a disease or condition wherein a therapeutically effective amount of a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention is administered to an animal, preferably a human, in need thereof. The term "therapeutically effective amount" as used herein refers to an amount sufficient to modulate one or more symptoms of the condition or disease being treated, preferably from 10mg to 3000mg per oral administration, once daily or twice daily or three times daily. It is yet another part of the present invention to provide a method for the prevention of a disease or disorder, wherein a therapeutically effective amount of a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention is administered to an animal, preferably a human, reasonably expected to be in need thereof. The term "therapeutically effective amount" as used herein refers to an amount sufficient to modulate one or more of the desired symptoms of the condition or disease to be avoided, preferably 10mg to 3000mg per oral administration, once or twice or three times daily.

In a further aspect, the invention provides the use of a composition of the invention or a pharmaceutical composition of the invention or a kit of the invention in the manufacture of a medicament for the treatment and/or prevention of a disease or condition, wherein the disease or condition is typically and preferably selected from peripheral sensory neuropathy, preferably peripheral neuropathic pain and other symptoms of peripheral sensory neuropathy; and neuropsychiatric disorders, such as seizures; depression; or a cognitive disorder; and motor neuron diseases such as amyotrophic lateral sclerosis.

It is also part of the present invention to administer the composition of the invention or the pharmaceutical composition of the invention, respectively, in combination with an active ingredient and an active agent (especially an anti-tumour and an anti-viral drug) which cause the onset of peripheral neuropathic pain and other symptoms of peripheral neuropathy which occur as side effects. The composition or pharmaceutical composition or kit is preferably used alone or together with at least one antineoplastic agent or at least one antiviral agent. More preferably, the composition or pharmaceutical composition or kit is used alone. More preferably, the composition or pharmaceutical composition or kit is used together with at least one antineoplastic agent. Alternatively, preferably, the composition or pharmaceutical composition or kit is used together with at least one antiviral agent.

Further preferably, said composition or pharmaceutical composition or kit is administered in combination with at least one antineoplastic agent or at least one antiviral agent, wherein said combined administration of said composition or said pharmaceutical composition with said at least one antineoplastic agent or with said at least one antiviral agent is concurrent, simultaneous, sequential or separate administration.

Non-limiting examples of such antineoplastic agents are selected from the group consisting of kinase inhibitors, proteasome inhibitors, taxanes, vinca alkaloids, and platinum salts. Non-limiting examples of such antiviral drugs are selected from nucleoside analogs or nucleotide analogs. Further preferably, the antineoplastic agent is selected from the group consisting of kinase inhibitors, proteasome inhibitors, taxanes, vinca alkaloids, and platinum salts. The antineoplastic agent is preferably selected from sorafenib, sunitinib, afatinib, axitinib, vandetanib, vemurafenib, ixazomi, bortezomib, paclitaxel, docetaxel, cabazitaxel, vincristine, vinblastine, vindesine, vinorelbine, nedaplatin, lobaplatin, picoplatin, lobaplatin, satraplatin, cisplatin, carboplatin and oxaliplatin. The antiviral agent is preferably selected from zalcitabine, didanosine, stavudine and zidovudine.

The composition or pharmaceutical composition or kit is preferably for use with at least one antiviral drug, wherein preferably the antiviral drug is selected from a nucleoside or a nucleotide, and wherein further preferably the antiviral drug is selected from zalcitabine, didanosine, stavudine or zidovudine.

Preferably, the disease or condition is a seizure. Alternatively, the disease or condition is preferably depression. Further preferably, the disease or condition is a cognitive disorder. Even more preferably, the disease or disorder is peripheral sensory neuropathy. More preferably, the disease or condition is peripheral neuropathic pain.

More preferably, the disease or condition is a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is selected from the group consisting of (i) diabetic neuropathy, (ii) post-herpetic neuropathy, (iii) low back pain, (iv) sacral pain, (v) surgical pain, (vi) crush injury, (vii) spinal injury, (viii) complex regional pain syndrome, (ix) phantom limb sensation, (x) peripheral sensory neuropathy associated with osteoarthritis, (xi) peripheral sensory neuropathy associated with rheumatoid arthritis, (xii) peripheral sensory neuropathy associated with autoimmune osteoarthritis, (xiii) headache, (xiv) fibromyalgia, (xv) peripheral sensory neuropathy caused by treatment with a proliferation-increasing inhibitor, (xvi) peripheral sensory neuropathy caused by a chemotherapeutic agent, (xvii) peripheral sensory neuropathy associated with visceral injury, (xviii) Peripheral sensory neuropathy associated with osteonecrosis, (xix) peripheral sensory neuropathy associated with human immunodeficiency virus infection, and (xx) peripheral sensory neuropathy caused by antiviral agents.

Preferably, the disease or condition is a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is selected from the group consisting of (i) diabetic neuropathy, (ii) post-herpetic neuropathy, (iii) low back pain, (iv) sacral pain, (v) surgical pain, (vi) crush injury, (vii) spinal injury, (viii) complex regional pain syndrome, (ix) phantom limb sensation, (x) peripheral sensory neuropathy associated with osteoarthritis, (xi) peripheral sensory neuropathy associated with rheumatoid arthritis, (xii) peripheral sensory neuropathy associated with autoimmune osteoarthritis, (xiii) headache, (xiv) fibromyalgia, (xv) peripheral sensory neuropathy caused by treatment with a proliferation inhibitor, (xvi) peripheral sensory neuropathy caused by a chemotherapeutic agent, (xvii) peripheral sensory neuropathy associated with visceral injury, (xviii) Peripheral sensory neuropathy associated with osteonecrosis, (xix) peripheral sensory neuropathy associated with human immunodeficiency virus infection, (xx) peripheral sensory neuropathy caused by antiviral agents, (xxi) peripheral neuropathic pain.

The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes diabetic neuropathy. Even more preferably, the disease or disorder is a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is post-herpetic neuropathy. The disease or condition is preferably peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is lumbago. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes sacral pain. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes surgical pain. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes a crush injury. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes a spinal injury. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes a complex regional pain syndrome. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes phantom limb sensation. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy associated with osteoarthritis. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy associated with rheumatoid arthritis. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy associated with autoimmune osteoarthritis. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes headache. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes fibromyalgia. Still more preferably, the disease or disorder is peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy caused by treatment with a proliferation inhibitor. More preferably, the disease or disorder is peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy induced by a chemotherapeutic agent. Still more preferably, the disease or disorder is peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes peripheral sensory neuropathy associated with visceral injury. Still more preferably, the disease or disorder is peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes peripheral sensory neuropathy associated with osteonecrosis. Still more preferably, the disease or disorder is a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes a peripheral sensory neuropathy associated with infection by a human immunodeficiency virus. Still more preferably, the disease or disorder is peripheral sensory neuropathy, wherein the peripheral sensory neuropathy is peripheral sensory neuropathy caused by an antiviral agent. The disease or condition is further preferably a peripheral sensory neuropathy, wherein the peripheral sensory neuropathy becomes peripheral neuropathic pain.

The peripheral sensory neuropathy is preferably selected from the group consisting of peripheral sensory neuropathy caused by a chemotherapeutic agent or peripheral sensory neuropathy caused by an antiviral agent.

Still more preferably, the disease or disorder is peripheral sensory neuropathy caused by a chemotherapeutic agent, wherein typically and preferably the chemotherapeutic agent is selected from the group consisting of a kinase inhibitor, a proteasome inhibitor, a taxane, a vinca alkaloid and a platinum salt. More preferably, the disease or disorder is a peripheral sensory neuropathy induced by a chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of a kinase inhibitor, a proteasome inhibitor, a taxane, a vinca alkaloid, and a platinum salt. Still more preferably, the disease or condition is a peripheral sensory neuropathy induced by a chemotherapeutic agent, wherein the chemotherapeutic agent is selected from sorafenib, sunitinib, afatinib, axitinib, vandetanib, vemurafenib, ixazomide, bortezomib, paclitaxel, docetaxel, cabazitaxel, vincristine, vinblastine, vindesine, vinorelbine, nedaplatin, lobaplatin, picoplatin, lobaplatin, satraplatin, cisplatin, carboplatin, and oxaliplatin. Still more preferably, the disease or disorder is peripheral sensory neuropathy induced by a chemotherapeutic agent, wherein the chemotherapeutic agent is selected from sorafenib, vincristine, paclitaxel, or oxaliplatin. Very preferably, the peripheral sensory neuropathy is induced by a chemotherapeutic agent, wherein the chemotherapeutic agent is sorafenib, paclitaxel, vincristine, cisplatin, carboplatin, or oxaliplatin.

Further preferably, the disease or disorder is peripheral sensory neuropathy caused by an antiviral agent, wherein preferably the antiviral agent is a nucleoside reverse transcriptase inhibitor. More preferably, the disease or disorder is peripheral sensory neuropathy induced by an antiviral agent, wherein the antiviral agent is selected from zalcitabine, didanosine, stavudine or zidovudine. More preferably, the disease or disorder is zalcitabine-induced peripheral sensory neuropathy.

Preferably, the chemotherapy-induced peripheral sensory neuropathy causes symptoms of allodynia or sensory disorder, more preferably, symptoms of allodynia or sensory disorder of the hand or foot, and further preferably, allodynia or sensory disorder of the hand or foot caused by sorafenib, vincristine, paclitaxel, or carboplatin, cisplatin, or oxaliplatin.

More preferably, the peripheral sensory neuropathy is associated with pain, paresthesia, dysesthesia or allodynia.

More preferably, the composition of the present invention or the pharmaceutical composition of the present invention may be administered prophylactically before the anti-tumor chemotherapeutic agent causes peripheral sensory neuropathy and its accompanying symptoms.

Further preferably, the composition of the invention or the pharmaceutical composition of the invention may be administered intermittently. Furthermore, it is preferred in the present invention that the composition of the present invention or the pharmaceutical composition of the present invention may be administered in synchronization with the repeated cycle of the antitumor chemotherapeutic agent.

When determining the individual regimen and dosage level for a particular patient or subject, the dosage will depend upon the route of administration, the severity of the disease, the age and weight of the patient or subject, and other factors normally considered by the attending physician.

The composition or pharmaceutical composition of the invention may be administered by any route, including oral, intramuscular, subcutaneous, topical, transdermal, intranasal, intravenous, sublingual or intrarectal administration. Typically and preferably, the pharmaceutical compositions of the present invention are administered by the oral route in a single dosage unit once daily, twice daily or three times daily, and most preferably once daily or twice daily. In a most preferred embodiment, the composition or pharmaceutical composition of the invention is administered twice daily.

Typically and preferably, the oral dose of the composition of the invention or the pharmaceutical composition of the invention is from 10mg to 3000mg per administration, more preferably from 20mg to 2000mg per administration, and still more preferably from 50mg to 1000mg per administration. Typically and preferably, the composition or the pharmaceutical composition is administered orally twice daily in a dose of 10mg to 3000mg per administration, more preferably 20mg to 2000mg per administration, still more preferably 50mg to 1000mg per administration.

The pharmaceutical compositions of the present invention may be prepared by mixing with appropriately selected and pharmaceutically acceptable excipients, vehicles, adjuvants, additives, surfactants, desiccants or diluents, which are known to those skilled in the art and may be suitable for oral, parenteral or topical administration. Typically and preferably, the pharmaceutical composition of the invention is administered in the form of tablets, capsules, sachets, powders, granules, pellets, oral or parenteral solutions, suspensions, suppositories, ointments, creams, lotions, gels, pastes and/or containing liposomes, micelles and/or microspheres.

The pharmaceutically acceptable carrier of the pharmaceutical composition of the present invention is, but not limited to, any pharmaceutically acceptable excipient, vehicle, adjuvant, additive, surfactant, desiccant or diluent. Suitable pharmaceutically acceptable carriers are magnesium carbonate, magnesium stearate, talc, lactose, sucrose, pectin, dextrin, starch, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter. The pharmaceutically acceptable carrier of the present invention may be solid, semi-solid or liquid.

Tablets, capsules or sachets for oral administration are generally provided in dosage units and may contain conventional excipients such as binding agents, fillers, diluents, tabletting agents, lubricants, detergents, disintegrating agents, coloring agents, flavoring agents and wetting agents. The tablets may be coated according to methods well known in the art. Suitable fillers include or preferably are cellulose, mannitol, lactose and similar agents. Suitable disintegrants include or are preferably starch, polyvinylpyrrolidone and starch derivatives, such as sodium starch glycolate. Suitable lubricants include or preferably are, for example, magnesium stearate. Suitable wetting agents include or are preferably sodium lauryl sulfate. These solid oral compositions may be prepared by conventional mixing, filling or tableting methods. The mixing operation can be repeated to disperse the active agent in a composition containing a large amount of filler. These operations are conventional.

Oral liquid compositions may be provided in the form of, for example, aqueous solutions, emulsions, syrups or elixirs, or as a dry product for reconstitution with water or a suitable liquid carrier at the time of use. The liquid composition may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminium stearate colloid or hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate or acacia; non-aqueous carriers (which may include edible oils), such as almond oil, fractionated coconut oil, oily esters, such as glycerides, propylene glycol or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and, if desired, conventional perfumes or colorants. Oral formulations may also include or may be formulated into conventional formulations, such as tablets or granules. For parenteral administration, liquid dosage units comprising the compositions of the present invention and a sterile carrier may be prepared.

The oral formulation may optionally further comprise a taste masking ingredient to optimize the taste of the oral formulation. Examples of such taste masking ingredients may be citrus, licorice, mint, grape, blackcurrant or eucalyptus based flavours known to the person skilled in the art.

Parenteral solutions are generally prepared by dissolving the compound in the carrier and sterilizing by filtration, followed by filling in suitable vials or ampoules and sealing.

Adjuvants such as local anesthetics, preservatives and buffering agents can be added to the pharmaceutical composition. To increase stability, the composition can be frozen after filling the vial and the water removed under vacuum. To facilitate uniform distribution of the compositions of the present invention, surfactants or humectants may advantageously be included in the pharmaceutical composition.

Topical formulations include or are preferably ointments, creams, lotions, gels, glues, solutions, pastes or may contain liposomes, micelles or microspheres.

The subjects treated with the composition or pharmaceutical composition of the present invention are humans and animals. Preferred animals are livestock and farm animals including, but not limited to, guinea pigs, rabbits, horses, donkeys, camels, cows, sheep, goats, pigs, cats, dogs, and parrots. More preferably the subject is a mammal, more preferably a human.

In yet another aspect, the invention provides an article of manufacture comprising a composition of the invention or a pharmaceutical composition of the invention or a kit, container or package of the invention, and written instructions such as a package insert and instructions for administration.

It is further contemplated that the ratio of (R) -dimiracetam or (S) -dimiracetam to other rasitapids (e.g., aniracetam, brivaracetam, sibutramine, coraracetam, doxetaracetam, etiracetam/levetiracetam, fasoracetam, oxiracetam, methylphenidate, neritaracetam, nefiracetam, N- (1- (phenylacetyl) -L-prolyl) glycine ethyl ester (omberacetam) (Noopept), oxiracetam, phenylpiracetam hydrazide, piracetam, pramipentam, rolipramipentam, luritaracetam and/or seletracetam), particularly if the ratio of (R) -dimiracetam or (S) -dimiracetam to other rasitapids (or other enantiomers of rasitapids) is disclosed herein for a mixture of (R) -dimiracetam and (S) -dimiracetam When selected within the range of compounds, may also be used to prepare synergistic mixtures and compositions.

It is further envisaged that compositions of (R) -dimiracetam or (S) -dimiracetam with other dimiracetam derivatives (such as those disclosed in US7544705 or US 8334286), in particular if the ratio of (R) -dimiracetam to dimiracetam-like compound (or enantiomer of dimiracetam-like compound) is selected within the range disclosed herein for mixtures of (R) -dimiracetam and (S) -dimiracetam, may also be used for the preparation of synergistic mixtures and compositions.

The present invention also relates to methods of treating and/or preventing a disease, injury, or condition, comprising: administering to a subject the composition of claim 1, wherein the disease, injury, or disorder is peripheral sensory neuropathy, seizures, depression, or cognitive disorders. In the method, the disease, injury or disorder is preferably a peripheral sensory neuropathy, a neuropsychiatric disorder, a motor neuron disorder or a movement disorder. More preferably, the disease, injury or disorder is peripheral sensory neuropathy. The peripheral sensory neuropathy is preferably peripheral neuropathic pain. The peripheral sensory neuropathy is preferably selected from diabetic neuropathy, post-herpetic neuropathy, low back pain, sacral pain, surgical pain, crush injury, spinal injury, complex regional pain syndrome, phantom limb sensation, peripheral sensory neuropathy associated with osteoarthritis, peripheral sensory neuropathy associated with rheumatoid arthritis, peripheral sensory neuropathy associated with autoimmune osteoarthritis, headache, fibromyalgia, peripheral sensory neuropathy induced by treatment with a proliferation inhibitor, peripheral sensory neuropathy induced by a chemotherapeutic agent, peripheral sensory neuropathy associated with visceral injury, peripheral sensory neuropathy associated with osteonecrosis, peripheral sensory neuropathy associated with human immunodeficiency virus infection, peripheral neuropathic pain, or peripheral sensory neuropathy induced by an antiviral agent. In some cases, the peripheral sensory neuropathy is peripheral sensory neuropathy induced by a chemotherapeutic agent or peripheral sensory neuropathy induced by an antiviral agent. In certain instances, the peripheral sensory neuropathy is a peripheral sensory neuropathy induced by a chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of kinase inhibitors, proteasome inhibitors, taxanes, vinca alkaloids, and platinum salts, and wherein preferred chemotherapeutic agents are selected from the group consisting of sorafenib, sunitinib, afatinib, axitinib, vandetanib, vemurafenib, ixazomide, bortezomib, paclitaxel, docetaxel, cabazitaxel, vincristine, vinblastine, vindesine, vinorelbine, nedaplatin, lobaplatin, picoplatin, lobaplatin, satraplatin, cisplatin, carboplatin, and oxaliplatin. In some cases, the peripheral sensory neuropathy is peripheral sensory neuropathy induced by an antiviral agent, wherein the antiviral agent is a nucleoside reverse transcriptase inhibitor. In certain instances, the nucleoside reverse transcriptase inhibitor is zalcitabine, didanosine, stavudine, or zidovudine.

In some cases, the method further comprises administering an anti-neoplastic agent, wherein the anti-neoplastic agent is selected from the group consisting of a kinase inhibitor, a proteasome inhibitor, a taxane, a vinca alkaloid, and a platinum salt. In some cases, the antineoplastic agent is selected from sorafenib, sunitinib, afatinib, axitinib, vandetanib, vemofinib, ixazomide, bortezomib, paclitaxel, docetaxel, cabazitaxel, vincristine, vinblastine, vindesine, vinorelbine, nedaplatin, lobaplatin, picoplatin, lobaplatin, satraplatin, cisplatin, carboplatin, and oxaliplatin. In some cases, the method further comprises administering an antiviral agent, wherein the antiviral agent is a nucleoside or nucleotide. In certain instances, the antiviral agent is zalcitabine, didanosine, stavudine, or zidovudine. In some cases, the composition is administered orally twice daily at a dose of 10mg to 3000mg per administration, 20mg to 2000mg per administration, or 50mg to 1000mg per administration.

The present invention also relates to a method of enhancing learning and memory comprising administering to a subject a composition of the invention as described herein. In certain instances of the method, the subject is a healthy subject.

The non-patent references cited herein are abbreviated as first author and year of publication. The complete citations are listed below.

Attal N,Cruccu G,Baron R,

Figure BDA0002285200280000311

M,Hansson P,Jensen TS,Nurmikko T;European Federation of Neurological Societies.EFNS guidelines on thepharmacological treatment of neuropathic pain:2010 revision.Eur J Neurol.2010Sep;17(9):1113-e88.PMID:20402746

Bradford MM.A rapid and sensitive method for the quantitation ofmicrogram quantities of protein utilizing the principle of protein-dyebinding.Anal Biochem.1976May 7;72:248-54.PMID:942051

Bril V,England J,Franklin GM,Backonja M,Cohen J,Del Toro D,Feldman E,Iverson DJ,Perkins B,Russell JW,Zochodne D;American Academy of Neurology;American Association of Neuromuscular and Electrodiagnostic Medicine;AmericanAcademy of Physical Medicine and Rehabilitation.Evidence-based guideline:Treatment of painful diabetic neuropathy:report of the American Academy ofNeurology,the American Association of Neuromuscular and ElectrodiagnosticMedicine,and the American Academy of Physical Medicine andRehabilitation.Neurology.2011 May 17;76(20):1758-65.Erratum in:Neurology.2011Aug 9;77(6):603.PMID:21482920

Camilleri P,Eggleston D,Farina C,Murphy JA,Pfeiffer U,Pinza M,SeniorLA.Chiral high-performance liquid chromatography of some related bicycliclactams.Journal of Chromatography A,654(1993)207-213.

Cavaletti G,Tredici G,Petruccioli MG,DondèE,Tredici P,Marmiroli P,Minoia C,Ronchi A,Bayssas M,Etienne GG.Effects of different schedules ofoxaliplatin treatment on the peripheral nervous system of the rat.Eur JCancer.2001 Dec;37(18):2457-63.PMID:11720843

Christensen D,

Figure BDA0002285200280000321

JJ,Guilbaud G,Kayser V.Theantinociceptive effect of combined systemic administration of morphine andthe glycine/NMDA receptor antagonist,(+)-HA966 in a rat model of peripheralneuropathy.Br J Pharmacol.1998 Dec;125(8):1641-50.Erratum in:Br J Pharmacol1999 Apr;126(8):1881.PMID:9886755

Dubinsky RM,Kabbani H,El-Chami Z,Boutwell C,Ali H;Quality StandardsSubcommittee of the American Academy of Neurology.Practice parameter:treatment of postherpetic neuralgia:an evidence-based report of the QualityStandards Subcommittee of the American Academy of Neurology.Neurology.2004Sep28;63(6):959-65.PMID:15452284

Di Cesare Mannelli L,Maresca M,Farina C,Scherz MW,Ghelardini C.Amodel of neuropathic pain induced by sorafenib in the rat:Effect of dimiracetam.Neurotoxicology.2015a Sep;50:101-7.PMID:26254739

Di Cesare Mannelli L,Micheli L,Farina C,Scherz M,and GhelardiniC.Effects of dimiracetam on oxaliplatin-induced hyperalgesia and allodynia inthe rat.Journal of Clinical Oncology 2015b 33:15_suppl,e20650-e20650.

Eliel EL,Wilen SH.Stereochemistry of Organic Compounds,Wiley-Interscience,New York(1994).ISBN:978-0-471-01670-0

Fariello RG,Ghelardini C,Di Cesare Mannelli L,Bonanno G,Pittaluga A,Milanese M,Misiano P,Farina C.Broad spectrum and prolonged efficacy ofdimiracetam in models of neuropathic pain.Neuropharmacology.2014 Jun;81:85-94.PMID:24486381

Fariello RG,Ghelardini C,Di Cesare Manneli L,Zanardelli M,FarinaC.Antidepressant-like activity of dimiracetam(NT-11624)in the rat forcedswimming test.Program No.789.08/EE25.2011 Neuroscience MeetingPlanner.Washington,DC:Society for Neuroscience,2011.Online.

Fernihough J,Gentry C,Malcangio M,Fox A,Rediske J,Pellas T,Kidd B,Bevan S,Winter J.Pain related behaviour in two models of osteoarthritis inthe rat knee.Pain.2004;Nov;112(1-2):83-93.PMID:15494188

Finnerup NB,Attal N,Haroutounian S,McNicol E,Baron R,Dworkin RH,Gilron I,

Figure BDA0002285200280000341

M,Hansson P,Jensen TS,Kamerman PR,Lund K,Moore A,Raja SN,Rice AS,Rowbotham M,Sena E,Siddall P,Smith BH,Wallace M.Pharmacotherapy forneuropathic pain in adults:a systematic review and meta-analysis.LancetNeurol.2015 Feb;14(2):162-73.PMID:25575710

Gronseth G,Cruccu G,Alksne J,Argoff C,Brainin M,Burchiel K,NurmikkoT,Zakrzewska JM.Practice parameter:the diagnostic evaluation and treatment oftrigeminal neuralgia(an evidence-based review):report of the QualityStandards Subcommittee of the American Academy of Neurology and the EuropeanFederation of Neurological Societies.Neurology.2008 Oct 7;71(15):1183-90.PMID:18716236

Guingamp C,Gegout-Pottie P,Philippe L,Terlain B,Netter P,GilletP.Mono-iodoacetate-induced experimental osteoarthritis:a dose-response studyof loss of mobility,morphology,and biochemistry.Arthritis Rheum.1997 Sep;40(9):1670-9.PMID:9324022

Guzman RE,Evans MG,Bove S,MorenkoB,Kilgore K.Mono-iodoacetate-induced histologic changes in subchondral bone and articular cartilage of ratfemorotibial joints:an animal model of osteoarthritis.Toxicol Pathol.2003;31(6):619-24.PMID:14585729

Latremoliere A,Woolf CJ.Central sensitization:a generator of painhypersensitivity by central neural plasticity.J Pain.2009 Sep;10(9):895-926.PMID:19712899.

Marieb EN,Wilhelm PB&Mallat JB,eds.Human Anatomy,8thedition.Pearson,London(2017).

Moulin D,Boulanger A,Clark AJ,Clarke H,Dao T,Finley GA,Furlan A,Gilron I,Gordon A,Morley-Forster PK,Sessle BJ,Squire P,Stinson J,Taenzer P,Velly A,Ware MA,Weinberg EL,Williamson OD;Canadian PainSociety.Pharmacological management of chronic neuropathic pain:revisedconsensus statement from the Canadian Pain Society.Pain Res Manag.2014 Nov-Dec;19(6):328-35.PMID:25479151

Nakamura Y,Iga K,Shibata T,Shudo M,Kataoka K.Glial plasmalemmalvesicles:a subcellular fraction from rat hippocampal homogenate distinct fromsynaptosomes.Glia.1993 Sep;9(1):48-56.PMID:7902337

Paluzzi S,Alloisio S,Zappettini S,Milanese M,Raiteri L,Nobile M,Bonanno G.Adult astroglia is competent for Na+/Ca2+exchanger-operatedexocytotic glutamate release triggered by mild depolarization.JNeurochem.2007 Nov;103(3):1196-207.PMID:17935604

Pinza M,Farina C,Cerri A,Pfeiffer U,Riccaboni MT,Banfi S,Biagetti R,Pozzi O,Magnani M,Dorigotti L.Synthesis and pharmacological activity of aseries of dihydro-1H-pyrrolo[1,2-a]imidazole-2,5(3H,6H)-diones,a novel classof potent cognition enhancers.J Med Chem.1993 Dec 24;36(26):4214-20.PMID:8277504

Randall LO,Selito JJ.A method for measurement of analgesic activityon inflamed tissue.Arch Int Pharmacodyn Ther.1957 Sep 1;111(4):409-19.PMID:13471093

Smith BH,Torrance N,Bennett MI,Lee AJ.Health and quality of lifeassociated with chronic pain of predominantly neuropathic origin in thecommunity.Clin J Pain.2007 Feb;23(2):143-9.PMID:17237663

Springuel GR,Leyssens T.Innovative chiral resolution usingenantiospecific co-crystallization in solution.Cryst Growth Des.2012;12(7):3374–3378.DOI:10.1021/cg300307z.

Torchio L,Lombardi F,Visconti M,Doyle E.Determination of the polardrug dimiracetam in human plasma and serum by column-switching high-performance liquid chromatography.J Chromatogr B Biomed Appl.1995 Apr 7;666(1):169-77.PMID:7655615

van Hecke O,Austin SK,Khan RA,Smith BH,Torrance N.Neuropathic pain inthe general population:a systematic review of epidemiologicalstudies.Pain.2014 Apr;155(4):654-62.Erratum in:Pain.2014 Sep;155(9):1907.PMID:24291734

Wang Y,Chen A.Crystallization-based separation of enantiomers,inStereoselective Synthesis of Drugs and Natural Products,2ndvolume;AndrushkoV,Andrushko N,Eds.Wiley-Interscience,New York(2013).ISBN:978-1-118-03217-6

Zilliox LA.Neuropathic Pain.Continuum(Minneap Minn).2017 Apr;23(2,Selected Topics in Outpatient Neurology):512-532.PMID:28375916.

40页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于治疗NASH和用于预防NASH诱导的HCC的方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!