Stable polyunsaturated compounds and their use

文档序号:704130 发布日期:2021-04-13 浏览:17次 中文

阅读说明:本技术 稳定的多不饱和化合物及其用途 (Stable polyunsaturated compounds and their use ) 是由 罗伯特·J·莫利纳里 米哈伊尔·谢尔盖维奇·什切皮诺夫 彼得·米尔纳 于 2019-04-18 设计创作,主要内容包括:本发明提供了使用取代的多不饱和脂肪酸、多不饱和脂肪酸酯、多不饱和脂肪酸硫酯、脂肪酸酰胺、多不饱和脂肪酸模拟物、多不饱和脂肪酸前药或其组合来治疗患有或有风险患有溶酶体贮积病(特别是Tay-Sachs病、Gaucher病、Sandhoff病或Niemann-Pick病)、神经元蜡样脂褐质沉积症、或与磷脂酶A2组VI(PLA2G6)活性受损相关的病况(特别是婴儿神经轴索营养不良或PLA2G6相关的神经退行性变(PLAN))或睡眠障碍的受试者的方法,其中所述取代的化合物包含至少一种减少所述化合物氧化的取代。优选地,所述取代的化合物是氘代多不饱和脂肪酸或其乙酯,例如11,11-D2-亚油酸、11,11-D2-亚油酸乙酯、11,11,14,14-D4-亚麻酸或11,11,14,14-D4-亚麻酸乙酯。(The present invention provides methods of using substituted polyunsaturated fatty acids, polyunsaturated fatty acid esters, polyunsaturated fatty acid thioesters, fatty acid amides, polyunsaturated fatty acid mimetics, polyunsaturated fatty acid prodrugs, or combinations thereof, to treat a subject having or at risk of having a lysosomal storage disease, particularly Tay-Sachs disease, Gaucher disease, sanddhoff disease, or Niemann-Pick disease, neuronal ceroid lipofuscinosis, or a condition associated with impaired activity of phospholipase a2 group VI (PLA2G6), particularly infantile axonal dystrophy or PLA2G 6-associated neurodegeneration (PLAN), or a sleep disorder, wherein the substituted compound comprises at least one substitution that reduces oxidation of the compound. Preferably, the substituted compound is a deuterated polyunsaturated fatty acid or an ethyl ester thereof, such as 11, 11-D2-linoleic acid, 11-D2-ethyl linoleate, 11,14, 14-D4-linolenic acid or ethyl 11,11,14, 14-D4-linolenate.)

1. A method of treating a subject having or at risk of having a disease or condition associated with impaired phospholipase a2 group VI (PLA2G6) activity, the method comprising:

selecting a subject having or at risk of having a disease or condition associated with impaired phospholipase a2 group VI (PLA2G6) activity; and

administering to the subject an effective amount of a substituted compound selected from the group consisting of: polyunsaturated fatty acids, polyunsaturated fatty acid esters, polyunsaturated fatty acid thioesters, fatty acid amides, polyunsaturated fatty acid mimetics, polyunsaturated fatty acid prodrugs, and combinations thereof, wherein the substituted compound comprises at least one substituent that reduces oxidation of the substituted compound.

2. The method of claim 1, wherein the subject has infant neuraxial dystrophy (INAD) or PLA2G 6-associated neurodegeneration (PLAN).

3. A method of treating a subject having or at risk of having a disease or condition associated with Lysosomal Storage Disease (LSD) and/or Neuronal Ceroid Lipofuscinosis (NCL), the method comprising:

selecting a subject having or at risk of having a disease or condition associated with a lysosomal storage disease or neuronal ceroid lipofuscinosis; and

administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, wherein the substituted compound comprises at least one substituent that reduces oxidation of the substituted compound.

4. The method of claim 3, wherein the subject has Tay-sach's disease, Gaucher's disease, Sandhoff's disease, or Niemann-Pick's disease.

5. The method of claim 4, wherein the subject has Tay-Sachs disease.

6. A method of treating a subject suffering from or at risk of suffering from a sleep disorder, the method comprising:

selecting a subject suffering from or at risk of suffering from a sleep disorder; and

administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, wherein the substituted compound comprises at least one substituent that reduces oxidation of the substituted compound.

7. The method of claim 6, wherein the subject has acute or chronic sleep disorder or obstructive sleep apnea syndrome.

8. The method of any one of claims 1 to 7, wherein the substituted compound comprises one or more isotopic atoms, and wherein the amount of the isotopic atoms is substantially higher than the natural abundance level of the isotopic atoms.

9. The method of claim 8, wherein the isotopic atom is deuterium,13C or a combination thereof.

10. The method of any one of claims 1 to 9, wherein the administering comprises repeated administration.

11. The method of any one of claims 1 to 10, wherein the subject has or is at risk of having at least one of a neuropathy or neurodegenerative disease, and the amount of the substituted compound is effective to prevent, ameliorate or inhibit progression of the neuropathy or neurodegenerative disease.

12. The method according to any one of claims 1 to 11, wherein the substituted compound is an isotopically modified polyunsaturated fatty acid, or an ester, amide, thioester, or prodrug thereof.

13. The method of claim 12, wherein the substituted compound is an omega-3 fatty acid, an omega-6 fatty acid, an omega-3 fatty acid ester, an omega-6 fatty acid ester, an omega-3 fatty acid amide, an omega-6 fatty acid amide, an omega-3 fatty acid thioester, or an omega-6 fatty acid thioester, or a combination thereof.

14. The method of any one of claims 1 to 13, wherein the polyunsaturated fatty acid ester is selected from a triglyceride, a diglyceride, a monoglyceride, or an alkyl ester.

15. The method of any one of claims 1 to 14, wherein the substituted compound has the structure of formula (1):

wherein R is H or C3H7

R1Is OH, O-alkyl, amine,An S-alkyl or O-cation;

each Y1And Y2Independently is H or D;

each X1And X2Independently is H or D, wherein Y1、Y2、X1And X2Is D;

m is 0,1, 2,3, 4, 5, 6, 7, 8, 9 or 10;

n is 1,2,3, 4 or 5; and is

p is 0,1, 2,3, 4, 5, 6, 7, 8, 9 or 10.

16. The method of any one of claims 1 to 15, wherein the substituted compound is deuterated linoleic acid, deuterated linolenic acid, deuterated arachidonic acid, deuterated eicosapentaenoic acid, deuterated docosahexaenoic acid, or esters, amides, or thioesters thereof.

17. The method of claim 16, wherein the amount of deuterium in the substituted compound is significantly higher than the natural abundance level of deuterium.

18. The method of claim 17, wherein the substituted compound is deuterated at one or more bis-allylic positions.

19. The method of claim 17, wherein the substituted compound is selected from the group consisting of: 11, 11-D2-linolenic acid; 14, 14-D2-linolenic acid; 11,11,14, 14-D4-linolenic acid; 11, 11-D2-linoleic acid; 7, 7-D2-arachidonic acid; 10, 10-D2-arachidonic acid; 13, 13-D2-arachidonic acid; 7,7,10, 10-D4-arachidonic acid; 7,7,13, 13-D4-arachidonic acid; 10,10,13, 13-D4-arachidonic acid; 7,7,10,10,13, 13-D6-arachidonic acid; 7,7,10,10,13,13,16, 16-D8-eicosapentaenoic acid; 6,6,9,9,12,12,15,15,18, 18-D10-docosahexaenoic acid; esters of any of the foregoing; and combinations thereof.

20. The method of any one of claims 1 to 19, wherein the ester is an ethyl ester.

21. The method of claim 20, wherein the substituted compound is ethyl 11, 11-D2-linoleate.

22. The method of claim 20, wherein the substituted compound is ethyl 11,11,14, 14-D4-linolenate.

23. The method of any one of claims 1 to 22, wherein the subject further ingests at least one of an unsubstituted polyunsaturated fatty acid and an unsubstituted polyunsaturated fatty acid ester.

24. The method of claim 23, wherein the amount of the substituted compound is about 5% or more of the total amount of polyunsaturated fatty acids and polyunsaturated fatty acid esters administered or delivered to the subject.

25. The method of claim 23, wherein the amount of the substituted compound is equal to or less than about 1% of the total amount of polyunsaturated fatty acids and polyunsaturated fatty acid esters administered or delivered to the subject.

26. The method of any one of claims 1 to 25, wherein the amount of the substituted compound administered is from about 10mg/kg to about 200 mg/kg.

27. The method of any one of claims 1 to 26, wherein the amount of the substituted compound administered is from about 20mg/kg to about 100 mg/kg.

28. The method of any one of claims 1 to 27, wherein the substituted compound is administered in an amount of about 1g to about 10 g.

29. The method of claim 28, wherein the amount of the substituted compound administered is from about 2g to about 5 g.

30. The method of any one of claims 1 to 29, wherein the substituted compound is administered once daily.

31. The method of any one of claims 1 to 29, wherein the substituted compound is administered two or more times per day.

32. The method of any one of claims 1 to 31, wherein the amount of the substituted compound administered is from about 1g to about 20g per day.

33. The method of claim 32, wherein the amount of the substituted compound administered is from about 2g to about 10g per day.

34. The method of any one of claims 1 to 33, wherein the substituted compound is administered for at least 2,3, or 4 weeks.

35. The method of any one of claims 1 to 34, wherein the substituted compound is co-administered to the subject with at least one antioxidant.

36. The method of claim 35, wherein the antioxidant is selected from the group consisting of: coenzyme Q, idebenone, mitoxantrone, mitoquinol, plastoquinone, resveratrol, vitamin E, and vitamin C, and combinations thereof.

37. The method of any one of claims 1,2, and 8-36, wherein the substituted compound is administered in an amount effective to alleviate one or more symptoms of a disease or condition associated with impaired phospholipase a2 group VI (PLA2G6) activity.

38. The method of claim 37, wherein the symptom associated with impaired PLA2G6 activity is selected from the group consisting of: hypotonia, nystagmus, strabismus, psychomotor degeneration and low spontaneous locomotor activity, and combinations thereof.

39. A method according to any one of claims 3-5 and 8-36, wherein the amount of the substituted compound administered to the subject is effective to alleviate one or more symptoms associated with LSD and/or NCL.

40. The method of claim 39, wherein the symptoms associated with LSD and/or NCL are selected from the group consisting of: physical difficulty moving, joint stiffness and pain, epilepsy, dementia, mental retardation, high mortality, vision problems, hearing problems, and bulbar function problems, and combinations thereof.

41. The method of any one of claims 6 to 36, wherein the amount of the substituted compound administered to the subject is effective to alleviate one or more symptoms or side effects associated with sleep disorders or sleep deficits.

42. The method of any one of claims 1 to 41, wherein the amount of the substituted compound administered is effective to improve muscle function in the subject.

43. The method of claim 42, wherein the muscle function is selected from the group consisting of: eye tracking, control, lift, fine motor skills and muscle strength, and combinations thereof.

44. The method of any one of claims 1 to 43, wherein the amount of the substituted compound administered is effective to improve neurological function in the subject.

45. The method of claim 44, wherein the neural function is selected from responsiveness to verbal commands, bulbar function, and verbal cognition, and combinations thereof.

Technical Field

The present disclosure relates to the fields of biochemistry and chemistry. Some embodiments relate to stabilized polyunsaturated substances, compositions comprising such stabilized polyunsaturated substances, and therapeutic uses thereof.

Background

In biological systems, the formation of potentially physiologically harmful Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS) may be caused by a variety of metabolic and/or environmental processes. By way of non-limiting example, intracellular reactive oxygen species (e.g., hydrogen peroxide H)2O2(ii) a Superoxide anion O2 -(ii) a OH-OH-(ii) a Nitric oxide NO; etc.) can be generated by several mechanisms (i) activation by radiation, excitation (e.g., UV rays) and ionization (e.g., X-rays); (ii) during xenobiotic and drug metabolism; and (iii) under conditions of relative hypoxia, ischemia and catabolism, and exposure to hyperbaric oxygen. Protection against deleterious physiological activity of ROS and RNS species is mediated by a complex network of overlapping mechanisms and metabolic pathways that utilize redoxThe combination of active small molecules and enzymes and the consumption of reducing equivalents.

Concentrations of ROS and RNS that are not adequately handled by the endogenous antioxidant system can result in damage to lipids, proteins, carbohydrates and nucleic acids. Changes in oxidative metabolism lead to an increase in the oxidative environment and the formation of potentially physiologically harmful ROS and RNS, commonly referred to in the literature as "oxidative stress". There is a need for an effective method to inhibit or reduce this oxidative stress and further treat medical conditions associated with oxidative stress.

Disclosure of Invention

Some embodiments relate to a method of treating a subject having or at risk of having a disease or condition associated with impaired phospholipase a2 group VI (PLA2G6) activity, comprising administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, the substituted compound comprising at least one substitution that reduces oxidation of the substituted compound.

Some embodiments relate to a method of treating a subject having or at risk of having infant neuroaxonal dystrophy (INAD) or PLA2G 6-related neurodegeneration (PLAN) comprising administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, the substituted compound comprising at least one substitution that reduces oxidation of the substituted compound.

Some embodiments relate to a method of treating a subject having or at risk of having a disease or condition associated with a Lysosomal Storage Disease (LSD), comprising administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, said substituted compound comprising at least one substitution that reduces oxidation of said substituted compound.

Some embodiments relate to a method of treating a subject having or at risk of having a Neuronal Ceroid Lipofuscinosis (NCL) -type disease comprising administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, said substituted compound comprising at least one substitution that reduces oxidation of said substituted compound.

Some embodiments relate to a method of treating a subject having or at risk of having a sleep disorder comprising administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, said substituted compound comprising at least one substitution that reduces oxidation of said substituted compound.

Drawings

FIG. 1 summarizes the baseline and one-year treatment status (extent of injury: (0) severely impaired, (+1) moderately impaired, (+2) mildly impaired or not impaired) for the patients described in example 1.

Detailed Description

Lipid Peroxidation (LPO) is a self-propagating, free-radical chain reaction that amplifies toxic triggering effects in various neurodegenerative diseases. The present disclosure relates to methods of treating various diseases and conditions using isotopically modified polyunsaturated fatty acids or derivatives thereof, wherein these isotopically modified compounds have been stabilized by isotopic substitution of at least one position that reduces oxidation of the compound. These substituted compounds can be easily incorporated into cell membranes and can prevent, delay or reverse lipid peroxidation and oxidative damage caused by lipid peroxidation.

Diseases associated with PLA2G6

The PLA2G6 gene encodes a group of VIA calcium-independent phospholipase a2 β enzymes that selectively hydrolyze glycerophospholipids to release free fatty acids. The mutation of PLA2G6 is related to diseases such as infant neuraxial axon malnutrition (INAD), neurodegeneration with cerebral iron deposition syndrome II and Karak syndrome. PLA2G6 may be the causative gene in an autosomal recessive, early-onset dystonia-parkinson's syndrome patient subgroup. Neuropathological examination can reveal extensive Lewy body lesions and the accumulation of hyperphosphorylated tau.

In some studies, the mechanisms of disease onset and progression suggest that the lipid peroxidation pathway is responsible for the disease phenotype. In the INAD model, PLA2G6 mutations trigger accumulation of lipid peroxidation products of linoleic acid and other PUFAs that lead to PLA2G 6-associated neurodegeneration (PLAN). Studies have been carried out using the fly model of PLAN to solve the problem of whether toxic oxidized cardiolipin actually accumulates within the mitochondrial membrane. The knockdown resulted in a significant increase in lipid peroxidation in the brain of flies due to the development of intracellular accumulation of stored lipids. A high concentration of lipid peroxidation products was observed.

INAD is a neurodegenerative disease that occurs in infancy and dies during adolescence or early adulthood. In addition to the hallmark cerebellar atrophy, its neuropathological features are axonal swelling and the presence of spheroids in the central and peripheral nervous systems. Neurodegeneration with cerebral iron deposition disorders comprises a group of clinically and genetically heterogeneous disorders with progressive extrapyramidal syndrome and high basal ganglia iron, and includes pantothenate kinase-associated neurodegeneration caused by PANK2 mutation (neurodegeneration with cerebral iron deposition disorder type I). Necropsy of the brain of patients with neurodegenerative and cerebral iron deposition associated with the homozygous PLA2G6 mutation revealed extensive Lewy body, dystrophic neurites and pathology of cortical neuronal neurofibrillary tangles.

In most individuals with infantile neuroaxonal dystrophy, mutations in the PLA2G6 gene have been identified. The PLA2G6 gene provides instructions for making an enzyme called a2 phospholipase. This enzyme family is involved in phospholipid metabolism. Phospholipid metabolism is important for many bodily processes, including helping to keep cell membranes intact and functioning properly. The phospholipase a2 produced by the PLA2G6 gene, sometimes referred to as PLA2 group VI, helps regulate the level of a compound known as phosphatidylcholine, which is abundant in cell membranes. Mutations in the PLA2G6 gene impair the function of group VI enzymes of PLA 2. This impairment of enzyme function may disrupt cell membrane maintenance and contribute to spheroid development in nerve axons. Phospholipid metabolism problems arise in this disorder and in a related disorder known as pantothenate kinase-related neurodegeneration.

Lysosomal storage diseases

The term "lysosomal storage disease" or "lysosomal storage disorder" (LSD) refers to a group of nearly 50 relatively rare inherited metabolic disorders that result from deficient lysosomal function due to enzyme deficiency, resulting in improper storage of substances in various cells of the body. These defects are associated with defects in the cellular metabolism of various types of lipids, glycoproteins, and/or glycosaminoglycans. As a result of LSD, excess cellular products, which are typically broken down, instead accumulate to an undesirable extent within the cell. Most lysosomal storage diseases are inherited in an autosomal recessive manner. Symptoms of lysosomal storage disorders often become increasingly severe over a period of time. Some exemplary lysosomal storage diseases include: gaucher disease (type I, P and PI), Pompe disease (glycogen storage disease, including infantile and delayed onset), GM2 gangliosidosis (including Tay-Sachs disease and Sandhoff disease), GM1 gangliosidosis and Niemann-Pick disease.

The term "neuronal ceroid lipofuscinosis" (NCL) is a generic term for a family of genetically distinct neurodegenerative lysosomal storage diseases that are caused by excessive accumulation of lipofuscin (lipofuscin) in body tissues. These lipofuscin are composed of fat and protein. These lipofuscin substances accumulate in neuronal cells and in many organs, including the liver, spleen, heart muscle and kidney.

GM1 gangliosidosis is a rare lysosomal storage disorder that is biochemically characterized by a lack of β -galactosidase activity and clinically characterized by a wide variety of neurovisceral, ophthalmological, and anamorphic characteristics.

GM2 gangliosidosis is a group of three related genetic disorders caused by β -hexosaminidase deficiency. This enzyme catalyzes the biodegradation of fatty acid derivatives known as gangliosides. When β -hexosaminidase no longer functions, lipids accumulate in the nervous tissue of the brain and cause problems. GM2 gangliosidosis includes Tay-Sachs disease, Sandhoff disease, and AB variants.

Tay-Sachs disease is a rare genetic disorder that gradually destroys nerve cells (neurons) in the brain and spinal cord. The most common form of Tay-Sachs disease becomes evident in infancy. Other forms of Tay-sach disease are very rare. Symptoms and signs may occur during childhood, adolescence, or adulthood, and are generally less severe than infant-type symptoms and signs. Characteristic features include muscle weakness, loss of muscle coordination (ataxia) and other motor problems, language problems and mental illness. These symptoms and signs vary widely among patients with late-onset Tay-Sachs. Tay-Sachs disease is caused by a genetic mutation in the HEXA gene on chromosome 15. The mutation causes a problem with an enzyme called β -hexosaminidase a, which causes the accumulation of the ganglioside molecule GM2 inside the cell, resulting in toxicity. Diagnosis is by measuring blood hexosaminidase a levels or genetic testing. In some embodiments of the methods described herein, a subject having Tay-Sachs disease is selected for treatment, comprising measuring blood hexosaminidase a levels, or testing for a genetic mutation in the hex a gene.

Sandhoff disease is a rare autosomal recessive metabolic disorder that can lead to progressive destruction of nerve cells in the brain and spinal cord. This disease is caused by a mutation in the HEXB gene on chromosome 5, which is critical for the lysosomal enzymes β -N-acetylhexosaminidase a and B. In some embodiments of the methods described herein, a subject having Sandhoff disease is selected for treatment comprising detecting a genetic mutation in the HEXB gene.

Gaucher's Disease (GD) is a genetic disorder in which glucocerebroside, a sphingolipid, also known as glucosylceramide, accumulates in cells and certain organs. The condition is characterized by bruising, fatigue, anemia, low platelet count and hepatosplenomegaly, and is caused by a genetic defect in glucocerebrosidase (also known as glucosylceramidase), an enzyme that acts on glucocerebrosidase. When enzymes are deficient, glucocerebroside accumulates, particularly in white blood cells, especially in macrophages (monocytes). Glucocerebroside can accumulate in the spleen, liver, kidney, lung, brain and bone marrow. This disease is caused by a recessive mutation of the GBA gene on chromosome 1. Gaucher's disease is the most common lysosomal storage disease. It is a form of sphingolipid disease (a subgroup of lysosomal storage diseases) because it involves dysfunctional metabolism of sphingolipids. In some embodiments of the methods described herein, the subject having Gaucher's disease is selected for treatment comprising testing for a genetic mutation in the GBA gene.

Niemann-Pick disease is a subset of lysosomal storage disorders, a group of inherited severe metabolic disorders in which sphingomyelin accumulates in the lysosomes of cells. Sphingomyelin is a component of cell membranes, including the membrane of organelles, and thus a deficiency in enzymes prevents degradation of lipids, resulting in accumulation of sphingomyelin in lysosomes of the macrophage-monocyte phagocytic lineage. Mutations in the SMPD1 gene result in Niemann-Pick disease types A and B. They produce a deficiency in the activity of the lysosomal enzyme acid sphingomyelinase, which breaks down the lipid sphingomyelin. Mutations in NPC1 or NPC2 lead to Niemann-Pick disease, type C (NPC), which affects a protein for the transport of lipids. For types a and B, sphingomyelinase levels can be measured from blood samples. For diagnosis of type C, skin samples can help determine whether the transporter is affected.

Typical drugs are directed to enzymatic, protein or genetic pathways. However, many biochemical processes are not controlled by enzymes. These processes are not often addressed therapeutically, in part because the discovery of modern drugs is often based on biochemical pathway mapping known from genomic analysis, and this approach may be relatively blind to non-gene-encoded events. Non-enzymatic in vivo processes involve a large set of oxidation reactions. The oxidative damage that results is harmful and cannot be controlled by antioxidants in diseased cells. Antioxidants are usually present in cells at levels close to saturation by enzyme-controlled active transport, and their concentration cannot easily be increased further. In addition, excess antioxidant may interfere with the desired redox process and result in a net detrimental effect. This may explain why clinical trials of antioxidants in humans generally do not produce positive or negative effects, although the cause of the disease is oxidative in nature.

Lipid peroxidation may lead to lysosomal instability and impaired lysosomal function, leading to LSD. As mentioned above, LSDs represent a class of congenital diseases characterized by the accumulation of substances in lysosomes. These conditions may be caused by a deficiency or reduced activity of lysosomal proteins, which leads to lysosomal accumulation of the substance. Typically, this material is stored because enzyme deficiencies impair digestion, but LSD also occurs when transport from the lysosomal compartment is impaired. In some LSDs, the selection and transport of various structurally impaired portions associated with various lipid subclasses (e.g., sphingolipids) to lysosomes for processing may be compromised. In addition, accumulation of substances such as highly sensitive polyunsaturated fatty acids containing lipids can affect the function of lysosomes as well as other downstream organelles, leading to secondary changes such as impaired autophagy, mitochondrial dysfunction and inflammation. LSDs are often involved in the central nervous system, where dysfunction or loss of neurons leads to mental retardation, progressive motor degeneration and premature death.

Reactive Oxygen Species (ROS) play a critical role and may be a common mediator of cell death in many LSDs. Thus, up-regulation of purine-free endonuclease 1(APE1), a protein that repairs oxidative DNA damage, was observed in Gaucher fibroblasts, but not in Gaucher mesenchymal stromal cells. Inducible nitric oxide synthase and nitrotyrosine are elevated in activated microglia/macrophages in GM1 and GM2 gangliosidoses, and ROS are elevated in the Fabry disease model. Genetic microarray analysis of Niemann-Pick disease type C1 (NPC1) fibroblasts is consistent with increased oxidative stress, and elevated ROS and lipid peroxidation makes fibroblasts more susceptible to death following acute oxidative injury. In mucopolysaccharidosis type IIIB (MPSIIIB), increased oxidative stress leads to oxidation of proteins, lipids and DNA, and an oxidative imbalance is found in mucopolysaccharidosis type I (MPSI). In Neuronal Ceroid Lipofuscinosis (NCL), elevated levels of ROS and superoxide dismutase are considered downstream of ER stress, a significant increase in manganese-dependent superoxide dismutase activity can be detected in fibroblasts and brain extracts of CLN6 sheep, and an increase in the expression of 4-hydroxynonenal can be detected in late infant and juvenile forms of NCL.

The central role played by oxidative stress in integrating other cellular pathways and stresses suggests that it is most likely activated in LSD as a secondary biochemical pathway rather than as a direct result of the accumulation of primary substrates. Furthermore, the possible effects of oxidative stress may have real implications in describing LSD pathology, particularly since oxidative stress plays a central role in other, more well-studied neurodegenerative conditions.

Sleep disorders

A large proportion of adults (about 40%) are affected variously by sleep abnormalities (dyssomnia), either chronic or acute. Sleep plays an important multifunctional role in physiological homeostasis. Grade n.sleep Med Clin 2011; 6:171. This includes a clean up (clearance) function that ensures that unwanted inter-and intra-neuronal metabolites that accumulate during the day are broken down and removed. A large part of this fragment consists of substances produced by oxidative stress, such as various LPO derivatives per se or conjugates with other biomolecules (e.g. DNA, phospholipid headgroups, proteins or peptides). Mathangi DC et al, Ann Neurosci 2012; 19: 161; thamaiselivi K et al, int.j.biol.med.res.2012; 3:1754. It is well known that this derivative is elevated in the event of insufficient sleep and indeed can be used as a marker of insufficient sleep. Weljie AM et al, PNAS USA 2015; 112:2569.

External (lifestyle choices such as shortened sleep time or jet lag) or internal (various sleep disorders) factors can adversely affect this elimination process, leading to incomplete removal or gradual accumulation of LPO products, resulting in metabolic disorders, various neurological conditions including, but not limited to, psychosis and bipolar disorder, and accelerated aging. Schmidt SM et al, Lancet Diabetes endocrinol.2014; 3:52. A related problem is interference of circadian rhythmicity and fluctuations, which affect a variety of metabolic pathways. Moeller-Levet CS et al, PNAS USA 2013; and 110: E1132. This is particularly relevant to lipid processing, which is largely controlled by the diurnal cycle. Chua EC-P et al, PNAS USA 2013; 110:14468.

Elevated oxidative stress markers are associated with obstructive sleep apnea syndrome and are often associated with many other sub-categories of sleep abnormalities. Passali d.et al, Acta Otorhinolaryngoh Itah 2015; 35: 420; hachul DE et al, clemeric 2006; 9: 312; gulec M et al, Prog Neuropsychypharmacol Biol Psychiatry 2012; 37: 247; liang B et al, Eur Rev Med Pharmacol Sci 2013; 17: 2517; semenova NV et al, Neuropsychiatry 2018; 8:1452.

In some embodiments, substituted compounds (e.g., D-PUFA) may be used alone or in combination with other therapeutic approaches (including but not limited to antioxidants, melatonin, glycine, sleep medications, antidepressants, etc.) to reduce the side effects of sleep deficits and sleep disorders caused by various background conditions, including but not limited to lifestyle-related sleep deficits; alcohol-related sleep deficits; idiopathic hypersomnia; hypersomnia, various sleep apneas; various abnormal sleep states; restless leg syndrome; illusion of sleep state; chronic Fatigue Syndrome (CFS) (also known as Myalgic Encephalomyelitis (ME)); mood disorders such as depression; anxiety disorders; panic; psychosis such as schizophrenia; and circadian rhythm-related sleep disorders, including jet lag-related disorders and night shift-related conditions. In some further embodiments, substituted compounds may also help to reduce the amount of sleep required and to alleviate lethargy. In some further embodiments, substituted compounds may also be used to ameliorate, reduce or alleviate other physiological effects, side effects or symptoms of sleep disorders, such as muscle pain; confusion; memory decline or loss; depression; the development of error memory; before falling asleep and before waking up hallucinations appear; hand tremor; headache; cachexia and lassitude; hordeolum; periorbital edema; an increase in blood pressure; increased levels of stress hormones; increased risk of diabetes; decreased immunity, increased susceptibility to disease; increased risk of fibromyalgia; irritability; rapid involuntary rhythmic eye movements; obesity; epilepsy; children develop spleen qi; and symptoms similar to attention deficit hyperactivity disorder and confusion.

Unlike mono-unsaturated or saturated fats, polyunsaturated lipids (e.g., polyunsaturated fats) contain one or more bisallyl positions-CH-in the long carbon chain of the fatty acid2Groups, which are non-conjugated moieties between two unsaturated double bonds. These positions are characteristic of PUFAs and are particularly susceptible to the formation of free radicals by oxidative stress through hydrogen abstraction. Once formed, the free radicals are more reactive than the PUFA itself and immediately react further, usually with oxygen, to form peroxy radicals, and these trigger even better than the original disease the propagation of more hydrogen extraction from the PUFA (see scheme 1).

The chain reaction of PUFA autoxidation is described in scheme 1 as the linoleic acid chain reaction. More PUFA enters the circulation during the propagation step. The extraction of the bisallyl hydride is the rate limiting step. The peroxide is further degraded to cytotoxic aldehydes, which further destroys proteins and DNA, creating more peroxidative stress.

Scheme one chemical chain reaction for linoleic acid lipid peroxidation

Unlike the classical mechanisms of abnormal proteins, different expression levels or chemical toxicity can directly lead to disease damage of cells. Factors such as differential expression levels, chemical toxicity and/or lipid peroxidation may be triggers that do not necessarily lead to clinical disease until the free radical mechanisms of chemical oxidation of sensitive fatty acids amplify it. Since the mechanisms of free radical lipid oxidation are well known and are known to involve accelerated cycling of autocatalytic damage, this common mechanism, independent of the trigger used to trigger it, may be responsible for a large number of cellular injuries in many indications.

Downregulating free radical initiation of lipid peroxidation (scheme 1) can prevent and treat disease-causing cell damage, even reversing the in vivo phenotype. Thus, stabilizing lipids against this injury becomes a new treatment modality.

The mechanisms of disease development and progression suggest that lipid peroxidation pathways are associated with disease phenotypes. The method comprises the following steps: 1) high concentrations of ROS produced by cellular energy production; 2) the concentrated accumulation of highly sensitive polyunsaturated fats in the lipid membrane; and 3) inadequate protection of the antioxidant for various reasons including hydrophobicity of the film, which limits the solubility and diffusion of the antioxidant to sensitive areas.

The final metabolites of the impaired polyunsaturated fatty acids, molecules such as 4-hydroxy-2-nonenal (4-HNE), 4-hydroxyhexenal (HHE), Malondialdehyde (MDA) and many other active carbonyl compounds, are candidate markers of neurodegeneration and loss of mitochondrial function, and are observed in almost all diseases involving lipid peroxidation.

Scheme 2 PUFA degradation products

There is a large body of literature supporting the role of lipid peroxidation in neurodegeneration with brain iron deposition (NBIA) and related neurodegenerative diseases. See Reed et al, "Lipid oxidation and neuro-integrative disease," Free radial Biology & Medicine 51(2011): 1302-. Recent literature also describes the role of lipid peroxidation in INAD, PKAN and PLA2G6 disorders. See Kinghom et al, "Loss of PLA2G6 leads to elongated myocardial lipid oxidation and mitochondral dysfunction," Brain 2015; 138: 1801-1816; kinghom et al, "Mitochondrial dysfunction and defects in lipid homeostatic targets in neurological targets with broad immunity," Rare Diseases 2016, VOL.4, NO.1, el 128616. PLA2G6 is particularly relevant in diseases associated with brain iron accumulation, such as Friedreich's ataxia, NBIA and alzheimer's disease, to name a few, are more susceptible because iron is a catalyst for the Fenton reaction of membrane lipid peroxidation pathway initiation events.

It is clear that lipid peroxidation plays an important role in LSD and/or NCL and related neurodegenerative diseases. Failure during normal lipid or oxidized lipid processing can excite LPO and exacerbate LPO product toxicity, thereby producing systemic toxic effects on any lipid membrane-containing structures, particularly PUFA-rich membranes. Examples of LSD diseases include, but are not limited to, sphingolipid deposits, ceramidase, Farber's disease, Krabbe's disease (infancy and late onset), galactosialiosis, gangliosides: gangliosidosis, alpha-galactosidases (including Fabry disease (alpha-galactosidase A), Schindler disease (alpha-galactosidase B)), beta-galactosidase/GM 1 gangliosidosis (infantile, juvenile and adult/chronic), GM2 gangliosidosis (AB variant, activator deficiency), Sandhoff disease (infantile, juvenile and adult/chronic), Tay-Sachs (juvenile hexosaminidase A deficiency, chronic hexosaminidase A deficiency), glucocerebrosidase (Gaucher disease, type I, P, type IP), sphingomyelinase (lysosomal acid lipase deficiency, early or late), MPS Ninn-Pick disease (type A or B), brain sulfation (sulfosulfatosis), metachromatic brain dystrophy, SapoB deficiency, multiple enzyme sulfate, mucopolysaccharidosis (type I: I Hurler syndrome), MPS I S Scheie syndrome, MPS I H-S Hurler-Scheie syndrome; type II (Hunter syndrome); type III (Sanfilippo syndrome) MPS III A (type A), MPS III B (type B), MPS III C (type C), MPS III D (type D); type IV (Morquio): MPS IV a (type a), MPS IV B (type B); type VI (maroteeaux-Lamy syndrome); type VII (Sly syndrome); type IX (hyaluronidase deficiency)), mucolipidosis (type I (sialidase deficiency), type II (I cell disease), type III (pseudohurler multiple dystrophy/phosphotransferase deficiency), type IV (mucoipidin 1 deficiency), lipidosis (Niemann-Pick disease, type C or D), Wolman disease, oligosaccharides (α -mannosidosis, β -mannosidosis, aspartylglucosaminuria, fucosidosis), lysosomal transport diseases (cystinosis, compact osteogenesis imperfecta (pycnodysostososis), sala disease/sialoprosidosis, free sialyl storage disease in infants), glycogen storage disease (Pompe disease type II, Danon disease type IIb), cholesteryl ester storage disease, and lysosome disease. Examples of NCL-type diseases include, but are not limited to, santavori-halita disease type 1/infant NCL (CLN1 PPT1), Jansky-Bielschowsky disease type 2/advanced infant NCL (CLN2/LINCL TPP1), Batten-Spielmeyer-Vogt disease type 3/juvenile NCL (CLN3), Kufs disease type 4/adult NCL (CLN4), Finnish variant/advanced infant type 5 (CLN5), advanced infant type 6 variant (CLN6), CLN7 type 7, Northern epilepsy type 8 (CLN8), Turkish advanced infant type 8 (CLN8), German/serban advanced infant type 9 (unknown), congenital cathepsin D deficiency type 10 (CTSD) and barton disease.

Lipid peroxidation chain reactions are targets for substituted compounds described herein. This chain reaction leads to cell damage, death and disease. To stop this damaging process, the substituted compounds described herein can be directed to the underlying cause of the disease, i.e., the amplification of the original disease caused by lipid peroxidation. Since PUFAs are also transformed (turn over) in diseased and normal cells, the substituted compounds described herein are capable of both maintaining and restoring their health and function.

The initial event of lipid peroxidation chain reaction is caused by the extraction of hydrogen from the bisallyl (between double bonds) methylene carbon in lipids by ROS-a rate determining step of lipid peroxidation chain reaction. If the start rate can be slowed, it will have a dramatic impact on down-regulated PUFA oxidation by eliminating all downstream multiplicative "cycling" damage for each extraction.

By replacing the hydrogen atom at the bisallylmethylene position with a deuterium atom, the initial extraction rate can be reduced. Deuterium is naturally occurring and is considered by living systems as a normal variant of hydrogen (hydrogen in all natural substances usually consists of about 1 deuterium/7000 hydrogen). Deuterium is also a well-known cause of "isotopic effects" (IE): when H is substituted by D, the reaction involving C-H bond cleavage is greatly slowed. This substitution reduces the ability of the C-H bond to break.

In some embodiments, substituted compounds described herein (e.g., PUFAs) that are specifically substituted with deuterium at the bisallyl position can be prepared in large quantities using well-optimized pharmaceutical synthetic methods. This modification is both "natural" (deuterium is present in nature) and "gaming-rule-altering" (game-playing): although the lipid peroxidation process is autocatalytic, the stabilization of the initial step is "reverse" catalyzed, resulting in a > 10-fold reduction in isotope multiplicities at each step, essentially shutting down the chain process rapidly. Thus, the vulnerable target bonds of the chain reaction are "flameproof" and not damaged by ROS. Importantly, enzymatic processes involving PUFAs, such as beta-oxidation, conversion involving other enzymes (all stoichiometric 1:1 enzyme: substrate reactions) are largely unaffected. Furthermore, this "fire-fighting" process requires only a small fraction of the total deuterated PUFA to effectively shut down the chain reaction of lipid peroxidation.

The substituted compounds described herein are deuterated (e.g., deuterated PUFAs), and thus represent a novel class of sensitive and specific drugs that are structurally similar to the corresponding compounds with only native deuterated levels, but that prevent destructive, non-enzymatic oxidation processes without substantially interfering with biologically essential enzymatic transformations. For example, because the PUFA in the membrane will rapidly convert-even if the cell is free-deuterium PUFA will rapidly replace the original hydrogen-containing molecule in all compartments of all tissues. All active transport for transferring normal PUFAs from oral foods works the same for deuterated PUFAs and transports them to the place where they are needed. As a result, D-PUFA is rapidly incorporated into the brain, retina and other tissues that are difficult to treat.

The substituted compounds described herein (e.g., deuterated PUFA, ethyl 11, 11-D2-linoleate) are unique in drug discovery and therapy. Some PUFAs, such as linoleic acid, are part of the human diet and have no pharmacological effect, but in deuterated form they may be useful as sensitive and specific drugs. These types of substituted compounds also do not have any observable side effects.

Linoleic Acid (LA) is an essential fatty acid in the human diet, called GRAS, with no known upper toxicity limit for nutritional use. LA was identified in the 20 th century, and more than 1300 studies have been published on LA in humans. There have been over 23,000 published human studies on omega-3 PUFAs, of which about 2,500 are randomized controlled clinical trials comparing omega-3 PUFAs and LA. No safety issues associated with LA were found in these studies.

The substituted compounds described herein are effective in treating a disease or condition associated with a lysosomal storage disease or a condition associated with impaired PLA2G6 activity. The role of the substituted compounds (e.g., deuterated LAs and esters thereof) in promoting cell survival, reducing and/or preventing damage caused by free radical chain reactions has been demonstrated. Coq-deficient coq mutant yeast strains are highly sensitive to oxidative damage by exogenous PUFAs because they lack antioxidant control and the critical hydrophobic intracellular mitochondrial membrane domain is not accessible to other hydrophilic antioxidants. In the coq mutant yeast model, healthy cells do not grow well on PUFAs (e.g., LA) when grown on MUFA (monounsaturated fatty acids) and SFA (saturated fatty acids). However, substituted compounds (e.g., deuterated LAs and esters thereof) are effective in increasing and/or maintaining cell viability.

In addition, the substituted compounds described herein are effective in reducing and preventing oxidative stress and damage associated with iron accumulation. Yeast, murine and human in vitro models of Friedreich's ataxia (FRDA) demonstrate that the substituted compounds described herein (e.g., D-PUFAs and esters thereof) are effective in controlling oxidative stress associated with increased iron. In FRDA, destructive iron accumulation was observed in brain tissue, similar to that in INAD. Treatment with substituted compounds described herein (e.g., D-PUFAs and esters thereof) and D4-ALA (deuterated linolenic acids, such as 11,11,14, 14-D4-linolenic acid) can result in reduced lipid peroxidation in an FRDA yeast model. In a murine FRDA cell model, a substituted compound described herein (e.g., 11, 11-D2-linoleic acid, 11,11,14, 14-D4-linolenic acid, or esters thereof) can protect cells from loss of viability. In the human fibroblast FRDA model, substituted compounds described herein (e.g., 11, 11-D2-linoleic acid, 11,11,14, 14-D4-linolenic acid, or esters thereof) can rescue cells from losing viability. See Cotticelli et al, Redox biology.2013(l): 398-. In another FRDA murine model, treatment with D4-ALA prevented lipid peroxidation. See Abeti et al, Cell Death dis.2016 May 26; 7: e 2237.

Andreyev reports the effect of substituted compounds described herein (e.g., D-PUFAs and esters thereof) on mitochondrial function under conditions of oxidative stress. See Andreyev et al, Free Radio Biol med.2015 Jan 8.80891-5849(15) 00003-9. In deuterated cells, the levels of F2-isoprostane oxidation product can be significantly reduced, indicating that treatment with the substituted compounds described herein can reduce the free radical chain processes in the cells as a whole. In H9C2 myoblasts, treatment with t-ButOOH resulted in increased respiratory depression and membrane leakage. Substituted compounds described herein (e.g., D-PUFAs and esters thereof) can protect mitochondrial function from t-ButOOH induced stress; maintaining maximum breathing and/or reducing the increase in membrane leakage. Other oxidative stress paradigms may work as well, D4-ALA and its esters may protect against oxidative stress, confirming that a combination of non-deuterated and deuterated PUFAs may effectively protect against oxidative stress. In the presence of unprotected polyunsaturated fatty acids, small amounts of D-PUFA can provide significant protection from Fe2+The effects of very severe oxidative stress induced.

In vivo models of mitochondrial dysfunction show that substituted compounds described herein (e.g., D-PUFAs and esters thereof) reduce oxidative stress-related damage. In the Parkinson's disease model, C57BL/6 mice were treated with neurotoxin 1-methyl-4-phenyl-1, 2,3, 6-tetrahydropyridine (MPTP). See shchenopov et al, Toxicol lett.2011 Nov 30; 207(2):97-103. MPP active metabolite MPP+Complex I, which inhibits the mitochondrial electron transport chain. Measurement of brain deuterium concentration 12-13 days after dietary administration of the substituted compounds described herein (e.g., D-PUFAs and esters thereof) indicates deuterium incorporation. Comparing to control with the above-described extractionAnimals fed secondary compounds (e.g., D-PUFAs and esters thereof) exhibit less MPTP effects. Treatment with substituted compounds described herein (e.g., D-PUFAs and esters thereof) can also rescue the dopaminergic phenotype.

Single and repeated dose studies for up to 26 weeks were performed in mice and rats using oral gavage and diet administration of substituted compounds described herein (e.g., D-PUFAs and esters thereof). The substituted compounds studied (e.g., 11,11-D2 linoleic acid) were well tolerated in all studies conducted. NOAEL determined in the study at 8 and 26 weeks corresponds to the average consumption of the substituted compound studied, at amounts of about 362 and about 452mg/kg, respectively. The high dose diets in these studies did not contain native LA. In this study, no signs of essential fatty acid (linoleic acid) deficiency were observed, which were characterized by changes, especially skin changes, including hair loss and flaky tails, which occurred within several months of continued feeding of the LA-deficient diet. This is consistent with the substituted compounds described herein (e.g., D-PUFAs and esters thereof) (dideuterolinoleic acid), which for dietary purposes are equivalent to and biologically interchangeable with normal dietary LA as the only source of LA in rats in this study. Analysis of tissue uptake and distribution indicates that administration of the substituted compounds described herein (e.g., D-PUFAs and esters thereof) does not appear to alter the enzymatic process or availability of PUFAs as measured in the heart, brain, lung, kidney, and liver. The substituted compounds described herein (e.g., D-PUFAs and esters thereof) and derivatives thereof are incorporated into tissues and do not result in any significant morphological or functional change in the treated animal as compared to their control. The data show that the enzymatic process of the substituted compounds described herein (e.g., D-PUFAs and their esters), and the subsequent mode of selective PUFA species incorporation for each tissue tested, are the same for the low dose, high dose, and control groups of the 8-and 26-week study. Body weight, organ weight, percentage of PUFA composition per organ, distribution of PUFA species in each tissue, and PUFA composition of red blood cells were unchanged compared to the control group.

These data indicate that the substituted compounds described herein (e.g., D-PUFAs and esters thereof) can effectively inhibit free radical degradation of lipids, but do not affect metabolic enzymatic processing of lipids. Thus, the substituted compounds described herein (e.g., D-PUFAs and esters thereof) appear to be treated the same as the essential fatty acid LA of dietary origin, and thus have well-known LA characteristics and safety.

The substituted compounds described herein (e.g., D-PUFAs and esters thereof) have been demonstrated to alleviate cell death and disease symptoms in a number of preclinical models of neurodegenerative diseases. > 90% of the INAD disease-associated gene defects, PLA2G6, result in increased cell death due to failure to clear lipid peroxidation. These effects can be reversed in the INAD stem cell and fibroblast studies dosed with D-PUFA compared to the control group, and the climbing performance of the drosophila model improved. Since D-PUFA has been shown to be safe in preclinical toxicity studies and phase 1/2 clinical studies of Friedreich's ataxia, and to show benefit in various models of INAD, it may also be effective to treat diseases or conditions associated with impaired activity of phospholipase a2 group VI (PLA2G6), such as INAD and PLAN, or diseases or conditions associated with lysosomal storage diseases and/or NCL diseases.

Definition of

The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.

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. The use of the term "including" is not limiting. The use of the term "having" is not limiting. As used in this specification, the term "comprising" shall be interpreted as having an open-ended meaning, whether in the transitional phrase or in the subject of the claims. That is, the above terms are to be construed as synonymous with the phrases "having at least" or "including at least". For example, when used in the context of a process, the term "comprising" means that the process includes at least the recited steps, but may include additional steps. The term "comprising" when used in the context of a compound, composition, formulation or device means that the compound, composition, formulation or device includes at least the recited features or ingredients, but may also include additional features or ingredients.

As used herein, common abbreviations are defined as follows:

ANOVA analysis of variance

BID twice daily

Neural axonal dystrophy in INAD infants

Late-onset Tay-Sachs disease of LOTS

LPO lipid peroxidation

LSD lysosomal storage disease or disorder

RBC red blood cells

PLA2G6 phospholipase A2 group VI

PLAN PLA2G 6-related neurodegeneration

PK pharmacokinetics

PUFA polyunsaturated fatty acids

T25FW timed 25 foot walk

The term "about" as used herein means that a quantity, value, number, percentage, amount, or weight differs from a reference quantity, value, number, percentage, amount, or weight by a variance deemed acceptable by one of ordinary skill in the art to be of that type of quantity, value, number, percentage, amount, or weight. In various embodiments, the term "about" refers to a change of 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% relative to a reference parameter, value, quantity, number, percentage, amount, or weight.

As used herein, the term "oral dosage form" has the ordinary meaning as understood by those skilled in the art, and thus includes, by way of non-limiting example, pharmaceutical formulations or forms for administration to humans, including pills, tablets, drug cores (cores), capsules, caplets, loose powders (los powder), solutions and suspensions.

As used herein, the term "ester" refers to the structure-C (═ O) OR, where R may include unsubstituted moietiesSubstituted or substituted C1-30Alkyl (branched or straight chain), unsubstituted or substituted C6-10Aryl, unsubstituted or substituted 5-to 10-membered heteroaryl, unsubstituted or substituted C3-10Carbocyclyl, or unsubstituted or substituted 3-10 membered heterocyclyl.

As used herein, the term "thioester" refers to the structure-C (═ O) SR, where R may include unsubstituted or substituted C1-30Alkyl (branched or straight chain), unsubstituted or substituted C6-10Aryl, unsubstituted or substituted 5-to 10-membered heteroaryl, unsubstituted or substituted C3-10Carbocyclyl, or unsubstituted or substituted 3-10 membered heterocyclyl.

As used herein, the term "amide" refers to the structure-C (O) NR1R2or-S (O) NR1R2Wherein R is1And R2C which may be independently unsubstituted or substituted1-30Alkyl (branched or straight chain), unsubstituted or substituted C6-10Aryl, unsubstituted or substituted 5-to 10-membered heteroaryl, unsubstituted or substituted C3-10Carbocyclyl, or unsubstituted or substituted 3-10 membered heterocyclyl.

As used herein, "subject" refers to a human or non-human mammal (e.g., dog, cat, mouse, rat, cow, sheep, pig, goat), a non-human primate or bird (e.g., chicken), as well as any other vertebrate or invertebrate animal.

The term "pediatric patient" as used herein refers to a human patient 17 years of age or less. In certain non-limiting embodiments, the patient is 16 years or less, or 15 years or less, or 14 years or less, or 13 years or less, or 12 years or less, or 11 years or less, or 10 years or less, or 9 years or less, or 8 years or less, or 7 years or less, or 6 years or less, or 5 years or less, or 4 years or less, or 3 years or less, or 2 years or less, or 1 year or less, or 6 months or less, or 4 months or less, or 2 months or less, or 1 month or less. In particular embodiments, the pediatric patient is between about 12 years of age to about 17 years of age. In one embodiment, the pediatric patient is selected from the group consisting of about 12 to about 17 years of age and about 2 years of age or less.

As used herein, the act of "providing" includes supplying, obtaining, or administering (including self-administration) a composition described herein.

As used herein, the term "administering" a drug includes the individual obtaining and taking the drug himself. For example, in some embodiments, an individual obtains a drug from a pharmacy and self-administers the drug according to the methods provided herein.

The term "therapeutically effective amount" as used herein refers to an amount of a substituted compound described herein that is sufficient to treat, ameliorate, or exhibit a detectable therapeutic effect on a disease or condition described herein. The effect may be detected by any means known in the art. In some embodiments, the precise effective amount of a subject may depend on the weight, size, and health of the subject; the nature and extent of the condition; and selecting the therapeutic agent or combination of therapeutic agents for administration. A therapeutically effective amount in a given situation may be determined by routine experimentation within the skill and judgment of the clinician. In some embodiments, the substituted compound is a polyunsaturated acid (PUFA) or an ester, thioester, amide or other prodrug thereof, or a combination thereof, for use in treating or ameliorating the diseases or conditions described herein. In some further embodiments, the substituted compound is 11, 11-D2-linoleic acid or an ester thereof.

As used herein, "treatment" refers to the administration of a compound or pharmaceutical composition to a subject for prophylactic and/or therapeutic purposes. The term "prophylactic treatment" refers to treating a subject who has not yet exhibited symptoms of a disease or condition, but who is predisposed to, or at risk of, a particular disease or condition, whereby the treatment reduces the likelihood that the patient will develop the disease or condition. The term "therapeutic treatment" refers to the treatment of a subject already suffering from a disease or condition.

The pharmaceutical compositions described herein are preferably provided in unit dosage form. As used herein, a "unit dosage form" is a composition/formulation that contains an amount of a compound suitable for administration to an animal (preferably a mammalian subject) in a single administration, in accordance with good medical practice. However, the preparation of a single or unit dosage form does not imply that the dosage form is administered once per day or once per course of treatment, or that the unit dosage form contains all the doses administered once. Such dosage forms are intended to be administered once, twice, three times or more daily, and may be administered more than once during the course of treatment, although single administrations are not specifically excluded. In addition, multiple unit dosage forms can be administered substantially simultaneously to achieve a desired overall dose (e.g., a patient can swallow two or more tablets to obtain a complete dose). One skilled in the art will recognize that the formulation does not specifically consider the entire course of therapy, and such a decision is left to those skilled in the therapeutic arts rather than the formulation arts.

In any of the embodiments described herein, the method of treatment may optionally encompass a use claim, such as a swiss-type use claim. For example, a method of treating a subject with impaired PLA2G6 activity may optionally involve the use of a compound in the manufacture of a medicament for treating a disease or condition described herein, or a compound for use in treating a disease or condition described herein.

Method of treatment

Some embodiments relate to a method of treating a subject having or at risk of having a disease or condition associated with impaired activity of phospholipase a2 group VI, the method comprising: selecting a subject having or at risk of having a disease or condition associated with impaired activity of phospholipase a2 group VI; and administering to the subject an effective amount of a substituted compound selected from the group consisting of a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, and combinations thereof, wherein the substituted compound comprises at least one substituent that reduces oxidation of the substituted compound. In some embodiments, the subject has an infant neuraxial dystrophy or a PLA2G 6-associated neurodegenerative disease. In one embodiment, the subject has an infant neuraxial dystrophy. In some such embodiments, the infant neuraxial dystrophy is caused by a PLA2G6 mutation.

Some embodiments relate to a method of treating a subject having or at risk of having a disease or condition associated with a lysosomal storage disease and/or neuronal ceroid lipofuscinosis, comprising: selecting a subject having or at risk of having a disease or condition associated with a lysosomal storage disease or neuronal ceroid lipofuscinosis; and administering to the subject an effective amount of a substituted compound selected from a polyunsaturated fatty acid, a polyunsaturated fatty acid ester, a polyunsaturated fatty acid thioester, a fatty acid amide, a polyunsaturated fatty acid mimetic, a polyunsaturated fatty acid prodrug, or a combination thereof, wherein the substituted compound comprises at least one substituent that reduces oxidation of the substituted compound. In some embodiments, the subject has Tay-sach's disease, Gaucher's disease, Sandhoff's disease, or Niemann-Pick's disease. In one embodiment, the subject has Tay-Sachs disease, e.g., late-onset Tay-Sachs disease. In some such embodiments, the Tay-Sachs disease is caused by a genetic mutation in the HEXA gene. In some embodiments, the LSD is GM1 gangliosidosis. In some embodiments, the LSD is GM2 gangliosidosis. In some embodiments, the LSD is a sphingolipid deposition disease.

Some embodiments relate to a method of treating a subject having or at risk of having a sleep disorder, the method comprising: selecting a subject having or at risk of having a sleep disorder, and administering to the subject an effective amount of a substituted compound selected from the group consisting of polyunsaturated fatty acids, polyunsaturated fatty acid esters, polyunsaturated fatty acid thioesters, fatty acid amides, polyunsaturated fatty acid mimetics, polyunsaturated fatty acid prodrugs, and combinations thereof, wherein the substituted compound comprises at least one substituent that reduces oxidation of the substituted compound. In some embodiments, the subject has acute or chronic sleep abnormalities. In some embodiments, the subject has obstructive sleep apnea syndrome.

In some embodiments of the methods described herein, the administering step comprises repeating the administering. In some embodiments, the subject has or is at risk of having a neuropathy or neurodegenerative disease, and the amount of the substituted compound is effective to prevent, ameliorate or inhibit progression of the neuropathy or neurodegenerative disease.

In some embodiments of the methods described herein, the substituted compound comprises one or more isotopes, and the amount of the isotope is substantially above the natural abundance level of the isotope. For example, in some embodiments, the amount of the isotope is two or more times greater than the natural abundance level of the isotope. In some embodiments, the isotope is selected from deuterium,13C and combinations thereof. In some embodiments, the isotopic atom is deuterium. The substituted compounds (e.g., isotopically modified PUFAs (e.g., deuterated PUFAs)) can reduce oxidation by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%.

In some embodiments, the methods described herein comprise identifying or selecting a subject with impaired PLA2G6 activity for treatment. In some such embodiments, identifying or selecting a subject with impaired PLA2G6 activity may comprise sequencing the DNA of the subject or using genetic testing to identify and screen patients with a mutation in the PLA2G6 gene. In some embodiments, patients with impaired PLA2G6 activity are identified in probands (probands) by identifying a biallelic pathogenic variant of PLA2G6 in a molecular genetic test. In some embodiments, patients with impaired PLA2G6 activity can be identified in probands where no pathogenic variant of PLA2G6 has been identified by electron microscopy of nerve biopsies of dystrophic axons (axon spheroids).

In some embodiments, the methods described herein comprise identifying or selecting a subject with LSD and/or NCL for treatment. In some such embodiments, identifying or selecting a subject with LSD and/or NCL may comprise sequencing the DNA of the subject or using genetic testing to identify and screen patients with genetic mutations associated with LSD and/or NCL-type diseases. In some such embodiments, identifying or selecting a subject with LSD and/or NCL comprises sequencing the DNA of the subject or using a genetic test to identify and determine the expression or activity of PGRN, or detecting one or more mutations in the gene or genomic DNA encoding PGRN. In some embodiments, identifying or selecting a subject with LSD and/or NCL comprises sequencing the DNA of the subject or using a genetic test to identify and determine the expression or activity of PGRN, or detecting one or more mutations in a gene or genomic DNA encoding HEX (e.g., HEX a, HEX b, or HEX s). In some further embodiments, identifying or selecting a subject with LSD and/or NCL comprises sequencing the DNA of the subject and detecting one or more mutations in a gene or genomic DNA encoding HEX (e.g., HEX a, HEX b, or HEX s). In some embodiments, the LSD and/or NCL-associated gene mutation may be a GBA mutation. In some embodiments, the LSD and/or NCL-associated gene mutation may be a PGRN mutation. In some embodiments, identifying a subject with LSD and/or NCL comprises sequencing the DNA of the subject or using a genetic test to identify and determine the expression or activity of SMPD1, NPC1, or NPC2, or detecting one or more mutations in a gene or genomic DNA encoding SMPD1, NPC1, or NPC 2.

In some embodiments of the methods described herein, the subject has or is at risk of having at least one of a neuropathy or neurodegenerative disease associated with impaired PLA2G6 activity. In some embodiments, the subject has infant neuraxial dystrophy (INAD) or PLA2G 6-associated neurodegenerative disease (PLAN). In some embodiments, the neuropathy or neurodegenerative disease associated with impaired PLA2G6 activity does not include alzheimer's disease. In some embodiments, the neuropathy or neurodegenerative disease associated with impaired PLA2G6 activity does not include parkinson's disease.

In some embodiments, the amount of substituted compound administered to the subject is effective to alleviate one or more symptoms of a disease or condition associated with impaired phospholipase a2 group VI (PLA2G6) activity. In some such embodiments, the symptom of the disease or condition is selected from the group consisting of hypotonia, nystagmus, strabismus, psychomotor degeneration (regression), and low spontaneous motor activity.

In some embodiments, the amount of substituted compound administered to the subject is effective to alleviate one or more symptoms associated with LSD and/or NCL. The symptoms of LSD and/or NCL may vary from patient to patient. In some embodiments, the symptom of the disease or condition is selected from the group consisting of: physical movement difficulties (e.g., joint stiffness and pain), epilepsy, dementia, mental retardation, high mortality, vision problems (e.g., blindness) or hearing problems (deafness), and bulbar function problems.

In some embodiments, the amount of substituted compound administered to the subject is effective to alleviate one or more symptoms or side effects of a sleep disorder or a lack of sleep. In some such embodiments, the side effects or symptoms of sleep disorders or hyposomnia are selected from the group consisting of: muscle pain; confusion; memory decline or loss; depression; the development of error memory; before falling asleep and before waking up hallucinations appear; tremor of the hands; headache; cachexia and lassitude; hordeolum; periorbital edema; an increase in blood pressure; increased levels of stress hormones; increased risk of diabetes; the immunity is reduced; increased susceptibility to disease; increased risk of fibromyalgia; irritability; rapid involuntary rhythmic eye movements; obesity; epilepsy; children develop spleen qi; and symptoms like attention deficit hyperactivity disorder and confusion.

In some further embodiments, the amount of the substituted compound administered to the subject is effective to increase muscle function in the subject. In some embodiments, the muscle function is selected from the group consisting of: eye tracking, control, lift, fine motor skills and muscle strength.

In some further embodiments, the amount of the substituted compound administered to the subject is effective to increase neurological function in the subject. In some embodiments, the neural function is selected from the group consisting of: responsiveness to verbal commands, bulbar function, and verbal cognition. In some embodiments, the neural function is bulbar function.

In some embodiments, administration of a substituted compound described herein may be used in combination with one or more additional therapies for the treatment of INAD selected from the group consisting of pharmacological treatment of spasticity and seizures; trials of oral or intrathecal injection of baclofen for the treatment of atypical INAD-related dystonia; the treatment of those patients with tardive neuropsychiatric symptoms by psychiatrists; fiber supplement and/or stool softener treatment for constipation; controlling secretion with scopolamine transdermal patch as required; prevention of aspiration pneumonia and feed adjustments needed to obtain adequate nutrition, and combinations thereof. In some embodiments, administration of a substituted compound described herein may be used in combination with one or more additional therapies for treating PLAN selected from the group consisting of: treatment with dopaminergic agents; the treatment of neuropsychiatric symptoms by a psychiatrist; assessment of postural instability and gait difficulty managed by physical therapy; occupational therapy to assist with activities of daily living; prevention of aspiration pneumonia and feed adjustments needed to obtain adequate nutrition, and combinations thereof.

In some embodiments, administration of a substituted compound described herein may be used in combination with one or more additional therapies for treating LSD and/or NCL disease. For example, the administration may be to a subject suffering from a sleep disorder described herein, along with antioxidants, melatonin, glycine, sleep medications, antidepressants, to improve or modulate the sleep-wake cycle and/or reduce side effects associated with the sleep disorder or the lack of sleep.

Dosage form

In some embodiments of the methods described herein, the therapeutically effective amount of the substituted compound administered to the subject is about 0.1g, 0.2g, 0.5g, 1.0g, 1.5g, 2.0g, 2.5g, 3.0g, 3.5g, 4.0g, 4.5g, 5.0g, 5.5g, 6.0g, 6.5g, 7.0g, 7.5g, 8.0g, 8.5g, 9.0g, 9.5g, lOg, 10.5g, 11g, 11.5g, 12g, 12.5g, 13g, 13.5g, 14g, 14.5g, 15g, 15.5g, l6g, 16.5g, 17g, 17.5g, 18g, 18.5g, 19g, 19.5g, or 20g, or a value defined by any two of the values above. In some embodiments, the amount of the substituted compound administered is from about 0.1g to about 20g, from about 1g to about 10g, from about 2g to about 5 g. In some further embodiments, the amount of the substituted compound administered is from about 1.8g to about 4.5 g. In some embodiments, the substituted compound is administered in a single unit dosage form. In some other embodiments, the substituted compound is administered in two or more unit dosage forms (i.e., divided doses). For example, when the dose is about 5g, it may be provided in the form of 4 or 5 tablets, each containing about 1.25g or 1g of the substituted compound. In some such embodiments, a dose of lg to 10g comprises administering 1,2,3, 4, or 5 unit dosage forms each comprising about lg to about 2g of the substituted compound, or about 2,3, or 4 unit dosage forms each comprising about 0.5g to about 2.5g of the substituted compound. In another embodiment, a dose of 2g to 5g comprises administering 1,2,3, 4, or 5 unit dosage forms, each dosage form comprising about 1g to about 2g of the substituted compound. In some embodiments, the unit dosage form is a tablet, capsule, pill, or pellet. In some further embodiments, the unit dosage form for oral administration, i.e., an oral dosage form.

In some embodiments of the methods described herein, the substituted compound may be administered once daily. In some other embodiments, the substituted compound may be administered two or more times per day, for example, two or three times per day. In some embodiments, the therapeutically effective amount of the substituted compound administered per day is about 1.0g, 2.0g, 3.0g, 3.5g, 4.0g, 4.5g, 5.0g, 5.5g, 6.0g, 6.5g, 7.0g, 7.5g, 8.0g, 8.5g, 9.0g, 9.5g, 10g, 10.5g, 11g, 11.5g, 12g, 12.5g, 13g, 13.5g, 14g, 14.5g, 15g, 15.5g, 16g, 16.5g, 17g, 17.5g, 18g, 18.5g, 19g, 19.5g, 20g, 25g, 30g, 35g, 40g, 45g, or 50g, or a range defined by any two of the values above. In some such embodiments, the amount of the substituted compound administered per day is from about 1g to about 20g, from about 2g to about 10g, from about 3g to about 8g, from about 4g to about 7g, or from about 5g to about 6 g. In one embodiment, the amount of 11, 11-D2-linoleic acid or ester thereof administered per day is from about 2g to about 10 g. In another embodiment, the amount of 11, 11-D2-linoleic acid or ester thereof administered per day is from about 1.8g to about 9 g.

In some embodiments of the methods described herein, the substituted compound may be administered for at least 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks. In some embodiments, the method further comprises detecting a steady state plasma level of the substituted compound, or a level of the substituted compound within the erythrocyte membrane, to determine the level of incorporation of the substituted compound. In some such embodiments, the plasma levels of the substituted compound reach a steady state after 1,2,3, or 4 weeks. In one embodiment, the plasma levels of the substituted compound may reach a steady state within 15 days, 20 days, 30 days, 40 days, 50 days, or 60 days.

In some embodiments, the dose of the substituted compound ranges from about 10mg/kg to about 200mg/kg, or about 20mg/kg to about 100 mg/kg. In some embodiments, the dose of the substituted compound ranges from about 30mg/kg to about 80 mg/kg. In some embodiments, the daily dose of the substituted compound ranges from about 1g to about 10 g. In some embodiments, the daily dose of the substituted compound is about 1.8g or about 9 g. In some embodiments, the daily dose of the substituted compound is about 1.8 g. In one embodiment, the substituted compound is administered in a daily dose of about 4.5g twice daily. In another embodiment, the substituted compound is administered in a daily dose of about 2.7g twice daily.

In some embodiments, the substituted compound is co-administered to the subject with at least one antioxidant. In some embodiments, the antioxidant is selected from the group consisting of coenzyme Q, idebenone (idebenone), mitoquinone (mitoquinone), mitoquinol (mitoquinol), plastoquinone, resveratrol, vitamin E, and vitamin C, and combinations thereof. In some such embodiments, the antioxidant may be administered simultaneously with, prior to, or after administration of the substituted compound. In some embodiments, the antioxidant and the substituted compound may be in a single dosage form. In some embodiments, the single dosage form is selected from the group consisting of: pills, tablets and capsules.

Substituted compounds

In some embodiments, the substituted compound comprises at least one isotope, and the amount of the isotope is substantially greater than the natural abundance level of the isotope. For example, in some embodiments, the amount of the isotope is two or more times greater than the natural abundance level of the isotope. In some such embodiments, the substituted compound comprises deuterium in an amount significantly above the natural abundance level of deuterium. For example, in some embodiments, the amount of deuterium in the substituted compound is two or more times greater than the natural abundance level of deuterium.

In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid ester. In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid. In some embodiments, the polyunsaturated fatty acid ester is a triglyceride, diglyceride, monoglyceride, or alkyl ester. In some embodiments, the polyunsaturated fatty acid ester is an ethyl ester.

The term "substituted compound" as used herein refers to a compound that is modified by substitution at one or more positions, thereby reducing the rate of oxidation of the compound. The modification may be isotopically substituted or non-isotopically chemical modification. The isotopic substitution may refer to substitution with an isotope such as deuterium or13One or more substitutions of isotopes of C. The non-isotopic modification may refer to substitution with another chemical group on the allylic hydrogen, or changing the position of the unsaturated bond to eliminate the allylic hydrogen position, thereby reducing oxidation of the substituted compound.

The term "polyunsaturated lipid" as used herein refers to a lipid that contains one or more unsaturated bonds (e.g., double or triple bonds) in its hydrophobic tail. The polyunsaturated lipid may be a polyunsaturated fatty acid (PUFA) or an ester thereof.

The term "bisallyl position" as used herein refers to the position of a polyunsaturated lipid (such as a polyunsaturated fatty acid or ester thereof) which corresponds to the methylene group of the 1, 4-diene system. Examples of polyunsaturated lipids with deuterium at one or more of the bisallyl positions include, but are not limited to, 11-dideutero-cis, cis-9, 12-octadecadienoic acid (11, 11-dideutero- (9Z,12Z) -9, 12-octadecadienoic acid; D2-LA); and 11,11,14, 14-tetradeutero-cis, cis-9, 12, 15-octadecatrienoic acid (11,11,14, 14-tetradeutero- (9Z,12Z,15Z) -9,12, 15-octadecatrienoic acid; D4-ALA).

The term "pro-bis-allylic position" as used herein refers to a methylene group in a compound that becomes a diallylic position upon desaturation. For example, some of the non-bisallyl sites in the precursor PUFA become bisallyl groups upon biochemical conversion. The pro-bisallyl position may be further substituted, in addition to being deuterated, with carbon-13, each position having isotopic abundance levels above its natural abundance level. For example, in addition to the existing bisallyl position, the pro-bisallyl position may be enhanced by isotopic substitution as shown in formula (1) below, wherein R is1is-OH, -O-alkyl, -amine, -S-alkyl or-O-cation (for example, the cation is Na)+Or K+) (ii) a m is 0 to 10; n is 1 to 5; and p is 0 to 10. In formula (1), the position of the X atom represents the pre-bisallyl position, and the position of the Y atom represents the bisallyl position, X1、X2、Y1And Y2Each of the atoms may independently be a hydrogen or deuterium atom, and X1、X2、Y1Or Y2At least one of the atoms is a deuterium atom. Each Y of each n unit1And Y2May independently be a hydrogen or deuterium atom, each X of each m unit1And X2May independently be a hydrogen or deuterium atom.

R=H,C3H7;R1OH, alkyl, S-alkyl, O-amino or O-cation

Each Y1And Y2(for each n unit) is independently H or D

Each X1And X2(for each n unit) is independently H or D

Another example of a substituted compound having bisallyl and propbisallyl positions is shown in formula (2), wherein Y is1-YnAnd/or X1-XmEach pair independently represents a di-allylic and a pre-di-allylic position of the PUFA, respectively, and these positions may contain deuterium atoms. X1、X2、...Xn,Y1、Y2、...YnThe atoms may independently be hydrogen or deuterium atoms, and X1、X2、...Xn,Y1、Y2、...YnAt least one or more of the atoms is deuterium. In some embodiments, Y is1、Y2、...YnAt least one of the atoms is deuterium. In some embodiments, p is 0,1 or 2. In some embodiments, m is 0,1, 2, or 3. In some embodiments, n is 1,2,3, or 4. In some embodiments, n is greater than 1. In some embodiments, n is less than 4.

R=H,C3H7;R1OH, O-alkyl, amine, SH, S-alkyl or O-cation;

Y1to YnH or D; x1To XmH or D; m is 1-10; n is 1-6; and p is 1-10

The substituted compounds described herein can be polyunsaturated lipids having at least one substitution that reduces oxidation of the substituted compound. In some embodiments, the substituted compound is isotopically modified to reduce oxidation. In some embodiments, the substituted compound is non-isotopically modified at one or more positions to reduce oxidation. The substituted compounds, e.g., isotopically modified PUFAs (e.g., deuterated PUFAs), can reduce oxidation by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%.

In some embodiments, the substituted compounds described herein comprise isotopically modified polyunsaturated fatty acids, isotopically modified polyunsaturated fatty acid esters, isotopically modified polyunsaturated fatty acid thioesters, isotopically modified polyunsaturated fatty acid amides, isotopically modified polyunsaturated fatty acid mimetics, or isotopically modified polyunsaturated fatty acid prodrugs. In some embodiments, the substituted compounds described herein may be isotopically modified polyunsaturated fatty acids or fatty acid esters. In some embodiments, the substituted compound may be an isotopically modified naturally occurring PUFA. In some embodiments, the substituted compound may have conjugated double bonds. In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid. In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid thioester. In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid amide. In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid mimetic. In some embodiments, the substituted compound is an isotopically modified polyunsaturated fatty acid prodrug.

In some embodiments, the substituted compound may be a deuterated polyunsaturated lipid. In some embodiments, the substituted compound can be a deuterated polyunsaturated fatty acid, a deuterated polyunsaturated fatty acid ester, a deuterated polyunsaturated fatty acid thioester, a deuterated fatty acid amide, a deuterated polyunsaturated fatty acid mimetic, a deuterated polyunsaturated fatty acid prodrug, or a combination thereof.

In some embodiments, the substituted compound is deuterated at one or more bis-allylic positions. In some embodiments, the substituted compound is further deuterated at one or more of the pre-bisallyl positions.

In some embodiments, the substituted compound is an omega-3 fatty acid, an omega-6 fatty acid, an omega-3 fatty acid ester, an omega-6 fatty acid ester, an omega-3 fatty acid amide, an omega-6 fatty acid amide, an omega-3 fatty acid thioester, or an omega-6 fatty acid thioester, or a combination thereof. In some embodiments, the substituted compound is an omega-3 fatty acid, an omega-3 fatty acid ester, an omega-3 fatty acid amide, an omega-3 fatty acid thioester, a prodrug thereof, or a combination thereof. In some embodiments, the substituted compound is an omega-6 fatty acid, an omega-6 fatty acid ester, an omega-6 fatty acid amide, an omega-6 fatty acid thioester, a prodrug thereof, or a combination thereof. In some embodiments, the substituted compound is linoleic acid, linolenic acid, arachidonic acid, eicosapentaenoic acid, docosahexaenoic acid, or esters, amides, thioesters, or prodrugs thereof, or a combination thereof.

In some embodiments, the subject also ingests at least one of an unsubstituted polyunsaturated fatty acid and an unsubstituted polyunsaturated fatty acid ester.

In some embodiments, the amount of the substituted compound is about 5% or more greater than the total amount of polyunsaturated fatty acids and polyunsaturated fatty acid esters administered or delivered to the subject. In some embodiments, the amount of the substituted compound is about 10% or more greater than the total amount of polyunsaturated fatty acids and polyunsaturated fatty acid esters administered to the patient. In some embodiments, the amount of the substituted compound is about 15% or more greater than the total amount of polyunsaturated fatty acids and polyunsaturated fatty acid esters administered to the subject. In some other embodiments, the amount of the substituted compound is equal to or less than about 1% of the total amount of polyunsaturated fatty acids and polyunsaturated fatty acid esters administered or delivered to the subject.

In some embodiments, the polyunsaturated fatty acid, polyunsaturated fatty acid ester, polyunsaturated fatty acid thioester, polyunsaturated fatty acid amide, polyunsaturated fatty acid mimetic, or polyunsaturated fatty acid prodrug can be a naturally occurring PUFA. In some embodiments, the polyunsaturated fatty acid, polyunsaturated fatty acid ester, polyunsaturated fatty acid thioester, polyunsaturated fatty acid amide, polyunsaturated fatty acid mimetic, or polyunsaturated fatty acid prodrug can have a conjugated double bond.

In some embodiments, the substituted compound is deuterated at one or more positions. In some embodiments, the substituted compound is deuterated at one or more bis-allylic positions. In some embodiments, the polyunsaturated fatty acid, polyunsaturated fatty acid ester, polyunsaturated fatty acid thioester, polyunsaturated fatty acid amide, polyunsaturated fatty acid mimetic, or polyunsaturated fatty acid prodrug is deuterated at one or more positions. In some embodiments, the polyunsaturated fatty acid, polyunsaturated fatty acid ester, polyunsaturated fatty acid thioester, polyunsaturated fatty acid amide, polyunsaturated fatty acid mimetic, or polyunsaturated fatty acid prodrug is deuterated at one or more diallyl positions.

In some embodiments, the substituted compound is a fatty acid or fatty acid ester. In some such embodiments, the ester may be a triglyceride, a diglyceride, a monoglyceride, or an alkyl ester. In some further embodiments, the polyunsaturated fatty acid ester is a methyl ester or an ethyl ester.

In some embodiments, the deuterated fatty acid or fatty acid ester is co-administered to the patient with a non-deuterated fatty acid or fatty acid ester.

In some embodiments, the substituted compound comprises about 1 wt% to about 100 wt%, about 5 wt% to about 90 wt%, about 10 wt% to about 50 wt%, about 20 wt% to about 40 wt% of the total amount of polyunsaturated fatty acids, polyunsaturated fatty acid esters, polyunsaturated fatty acid thioesters, polyunsaturated fatty acid amides, polyunsaturated fatty acid mimetics, and polyunsaturated fatty acid prodrugs administered or delivered to the patient. In some embodiments, the substituted compound comprises about 10 wt% to about 40 wt% of the total amount of polyunsaturated fatty acids, polyunsaturated fatty acid esters, polyunsaturated fatty acid thioesters, polyunsaturated fatty acid amides, polyunsaturated fatty acid mimetics, and polyunsaturated fatty acid prodrugs administered to the patient. In some embodiments, the substituted compound comprises about 1 wt%, 2 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 8 wt%, 9 wt%, 10 wt%, 15 wt%, 20 wt% or more of the total amount of polyunsaturated fatty acids, polyunsaturated fatty acid esters, polyunsaturated fatty acid thioesters, polyunsaturated fatty acid amides, polyunsaturated fatty acid mimetics, and polyunsaturated fatty acid prodrugs administered or delivered to the patient. In some further embodiments, the substituted compound is a deuterated fatty acid or fatty acid ester.

In some embodiments, the deuterated fatty acid or fatty acid ester comprises from about 1 wt% to about 100 wt%, from about 5 wt% to about 90 wt%, from about 10 wt% to about 50 wt%, from about 20 wt% to about 40 wt% of the total amount of fatty acid or fatty acid ester administered or delivered to the subject. In some embodiments, the deuterated fatty acid or fatty acid ester comprises about 1 wt%, 2 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 8 wt%, 9 wt%, 10 wt%, 15 wt%, 20 wt%, or more of the total amount of fatty acid or fatty acid ester administered or delivered to the subject.

In some embodiments, the cells or tissues of the patient maintain a sufficient concentration of the deuterated fatty acid or fatty acid ester to prevent or reduce autoxidation of naturally-occurring non-deuterated fatty acids or fatty acid esters.

In some embodiments, the deuterated substituted compound has an isotopic purity in a range from about 20% to about 99%.

In some embodiments, the fatty acid or fatty acid ester is one or more selected from the group consisting of: 11, 11-D2-linolenic acid, 14, 14-D2-linolenic acid, 11,14, 14-D4-linolenic acid, 11-D2-linoleic acid, 14, 14-D2-linoleic acid and 11,11,14, 14-D4-linoleic acid. In some embodiments, the substituted compound is an omega-3 fatty acid or an omega-3 fatty acid ester. In some embodiments, the substituted compound is an omega-6 fatty acid or an omega-6 fatty acid ester.

In some embodiments, the substituted compound is linoleic acid, linolenic acid, arachidonic acid (ARA), docosahexaenoic acid (DHA), or eicosapentaenoic acid (EPA), or a combination thereof. In some embodiments, the substituted compound is arachidonic acid, docosahexaenoic acid, eicosapentaenoic acid, which contains one or more deuterium. In some embodiments, the substituted compound is arachidonic acid, docosahexaenoic acid, eicosapentaenoic acid, each of which contains one or more deuterium at one or more bisallyl positions.

In some embodiments, the substituted compound is selected from the group consisting of: 11, 11-D2-linolenic acid, 14, 14-D2-linolenic acid, 11,14, 14-D4-linolenic acid, 11-D2-linoleic acid, esters thereof, and combinations thereof. In some embodiments, the substituted compound is selected from the group consisting of: 7, 7-D2-arachidonic acid; 10, 10-D2-arachidonic acid; 13, 13-D2-arachidonic acid; 7,7,10, 10-D4-arachidonic acid; 7,7,13,13-D4 arachidonic acid; 10,10,13, 13-D4-arachidonic acid; 7,7,10,10,13, 13-D6-arachidonic acid; 7,7,10,10,13,13,16, 16-D8-eicosapentaenoic acid; 6,6,9,9,12,12,15,15,18, 18-D10-docosahexaenoic acid; esters thereof, and combinations thereof. In some embodiments, the substituted compound is ethyl 11, 11-D2-linoleate. In some embodiments, the substituted compound is ethyl 11,11,14, 14-D4-linolenate. In some embodiments, the substituted compound is 7,7,10,10,13, 13-D6-arachidonic acid; 7,7,10,10,13,13,16, 16-D8-eicosapentaenoic acid; 6,6,9,9,12,12,15,15,18, 18-D10-docosahexaenoic acid; or an ester thereof. In some embodiments, the substituted compound is 7,7,10,10,13, 13-D6-arachidonic acid; 7,7,10,10,13,13,16, 16-D8-eicosapentaenoic acid, or an ester thereof. In some embodiments, the substituted compound is 7,7,10,10,13,13,16, 16-D8-eicosapentaenoic acid, or an ester thereof. In some embodiments, the substituted compound is 6,6,9,9,12,12,15,15,18, 18-D10-docosahexaenoic acid; or an ester thereof.

In some embodiments, the fatty acid or fatty acid ester is an omega-3 fatty acid. In some embodiments, the omega-3 fatty acid is alpha linolenic acid. In some embodiments, the omega-3 fatty acid is ARA. In some embodiments, the omega-3 fatty acid is EPA. In some embodiments, the omega-3 fatty acid is DHA.

In some embodiments, the fatty acid or fatty acid ester is an omega-6 fatty acid. In some embodiments, the omega-6 fatty acid is linoleic acid. In some embodiments, the omega-6 fatty acid is gamma linolenic acid, dihomogamma linolenic acid, arachidonic acid, or docosatetraenoic acid. In some embodiments, the fatty acid or fatty acid ester is an omega-6 ARA. In some embodiments, the fatty acid or fatty acid ester is omega-6 DHA. In some embodiments, the fatty acid or fatty acid ester is omega-6 EPA.

The substituted compounds isotopically enhanced at the oxidation-sensitive sites by the above structures are heavily isotopically enriched at the sites as compared to the natural abundance of the appropriate isotope. In some embodiments, the substituted compound has a deuterium atom present at a level greater than its natural abundance level. Deuterium is naturally present in an abundance of about 0.0156%. Thus, greater than 0.0156% of the hydrogen atoms of a substituted compound having a natural abundance of deuterium relative to one or more hydrogen atoms in each molecule of the substituted compound are replaced or "enhanced" by deuterium, e.g., 0.02% deuterium, but preferably about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% deuterium. In other embodiments, the percentage of total hydrogen atoms fortified with deuterium in the substituted compound is at least 0.02%, 0.03% (about twice the natural abundance), 0.05%, 0.1%, 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.

In some aspects, the compositions of substituted compounds comprise an isotopically modified polyunsaturated lipid and an isotopically unmodified polyunsaturated lipid. In some embodiments, isotopic purity refers to the percentage of molecules of the isotopically modified polyunsaturated lipid in the composition relative to the total number of molecules of the isotopically modified polyunsaturated lipid plus the polyunsaturated lipid containing no heavy atoms. For example, the isotopic purity can be about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the molecules of the isotopically modified polyunsaturated lipid relative to the total number of molecules of both the isotopically modified polyunsaturated lipid and the polyunsaturated lipid that do not contain heavy atoms. In other embodiments, the isotopic purity is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%. In some embodiments, the isotopic purity of the polyunsaturated lipid can be about 10% -100%, 10% -95%, 10% -90%, 10% -85%, 10% -80%, 10% -75%, 10% -70%, 10% -65%, 10% -60%, 10% -55%, 10% -50%, 10% -45%, 10% -40%, 10% -35%, 10% -30%, 10% -25%, or 10% -20% of the total number of molecules of the polyunsaturated lipid in the composition. In other embodiments, the isotopic purity of the polyunsaturated lipid can be about 15% -100%, 15% -95%, 15% -90%, 15% -85%, 15% -80%, 15% -75%, 15% -70%, 15% -65%, 15% -60%, 15% -55%, 15% -50%, 15% -45%, 15% -40%, 15% -35%, 15% -30%, 15% -25%, or 15% -20% of the total number of molecules of the polyunsaturated lipid in the composition. In some embodiments, the isotopic purity of the polyunsaturated lipid can be about 20% -100%, 20% -95%, 20% -90%, 20% -85%, 20% -80%, 20% -75%, 20% -70%, 20% -65%, 20% -60%, 20% -55%, 20% -50%, 20% -45%, 20% -40%, 20% -35%, 20% -30%, or 20% -25% of the total number of molecules of the polyunsaturated lipid in the composition. Two isotopically modified polyunsaturated lipid molecules out of the total number of molecules of 100 total of isotopically modified polyunsaturated lipids and polyunsaturated lipids without heavy atoms may have an isotopic purity of 2% regardless of the number of heavy atoms contained in the two isotopically modified molecules.

In some aspects, isotopically modified PUFA molecules may contain one deuterium atom, such as when one of the two hydrogens in a methylene group is replaced with deuterium, and thus may be referred to as a "D1" PUFA. Similarly, isotopically modified PUFA molecules may contain two deuterium atoms, for example when both hydrogens in a methylene group are replaced with deuterium, and thus may be referred to as a "D2" PUFA. Similarly, isotopically modified PUFA molecules may contain three deuterium atoms and may be referred to as "D3" PUFA. Similarly, isotopically modified PUFA molecules may contain four deuterium atoms and may be referred to as "D4" PUFAs. In some embodiments, isotopically modified PUFA molecules may contain five deuterium atoms or six deuterium atoms, and may be referred to as "D5" or "D6" PUFAs, respectively.

The number of heavy atoms, or the isotopic content (load), in a molecule may vary. For example, molecules with relatively low isotopic content may contain about 1,2,3, 4, 5, or 6 deuterium atoms. A medium isotope content molecule may contain about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 deuterium atoms. In a very high content molecule, each hydrogen atom may be replaced by a deuterium atom. Thus, isotopic content refers to the percentage of heavy atoms of that type of atom in each substituted compound or polyunsaturated lipid molecule. For example, the isotopic content can be about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% by number of atoms of the same type as compared to a substituted compound or polyunsaturated lipid that does not have heavy atoms of the same type (e.g., hydrogen and deuterium are "of the same type"). In some embodiments, the isotopic content is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%. When the isotopic purity of the substituted compound or polyunsaturated lipid composition is higher, but the isotopic content of a given molecule is lower, unexpected side effects are expected to be reduced. For example, metabolic pathways may be less affected when composed of substituted compounds or polyunsaturated lipids with high isotopic purity but low isotopic content.

It is readily understood that when one of the two hydrogens of the methylene group is replaced with a deuterium atom, the resulting compound may have a stereogenic center. In some embodiments, it may be desirable to use racemic compounds. In other embodiments, it may be desirable to use enantiomerically pure compounds. In other embodiments, it may be desirable to use diastereomerically pure compounds. In some embodiments, it may be desirable to use a mixture of compounds with enantiomeric and/or diastereomeric excess of about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%. In other embodiments, the enantiomeric excess and/or diastereomeric excess is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 65%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%. In some embodiments, it may be preferred to utilize stereochemically pure enantiomers and/or diastereomers of the embodiments, for example when contact with chiral molecules is intended to mitigate oxidative damage. However, in many cases, achiral molecules are used to mitigate oxidative damage. In this case, embodiments may be utilized regardless of their stereochemical purity. Furthermore, in some embodiments, mixtures of enantiomers and diastereomers may be used even when the compound is targeted to chiral molecules to mitigate oxidative damage.

In some aspects, the isotopically modified polyunsaturated lipids impart an amount of heavy atoms in a particular tissue. Thus, in certain aspects, the number of heavy molecules will be a specific percentage of the same type of molecules in the tissue. For example, the number of heavy molecules may be about 1% to 100% of the total amount of molecules of the same type in the tissue. In some aspects, 10-50% of the molecules are substituted with the same type of heavy molecule.

In some embodiments, the polyunsaturated lipid is deuterated at one or more bis-allylic positions. An example of a polyunsaturated lipid is an essential PUFA isotopically modified at the bisallyl position as shown in formula (3) below, wherein R is1is-O-alkyl, -OH, amine-SH, -S-alkyl, or-O-cation (for example, the cation is Na)+Or K+) (ii) a m is 1 to 10; n is 1 to 5; r is H or alkyl (e.g., C)3H7). In addition to deuteration, the bisallyl position may be further enhanced by carbon-13, with each position having isotopic abundance levels above the natural abundance levels. One or two Y in each n unit at each bisallyl position in formula (3)1、Y2The atoms are independently deuterium atoms. In some embodiments, n is 1,2,3, or 4. In some embodiments, m is 1,2,3, or 4.

The exact structures of the above compounds are shown below, they provide isotopically enhanced n-3 (omega-3) and n-6 (omega-6) essential polyunsaturated fatty acids, as well as PUFAs biochemically produced from them by desaturation/elongation. Any of these compounds may be used to slow oxidation. In the following compounds, the PUFAs are isotopically enhanced at oxidation-sensitive sites and/or sites that may become oxidation-sensitive upon biochemical desaturation/elongation. R1May be H, alkyl or cationic (e.g., Na)+Or K+);R2May be H or D;*represents12C or13C。

The deuterated linoleic acid may comprise:

the following fully deuterated linoleic acid can be produced by microbiological methods, for example, by growth in deuterium and/or carbon-13 containing media.

Deuterated arachidonic acid may include:

the following per-deuterated arachidonic acid can be produced microbiologically, for example, by growth in deuterium and/or carbon-13 containing media.

Deuterated linolenic acids can include:

and

the following deuterated linolenic acids can be produced microbiologically, for example, by growth in deuterium and/or carbon-13 containing media.

Deuterated polyunsaturated fatty acids and esters can also include:

and esters thereof.

In another embodiment, the oxidizable bis-allylic sites of the substituted compounds described herein (e.g., PUFAs) can prevent hydrogen extraction by further separating the bis-allylic hydrogen activated double bonds, thereby eliminating the bis-allylic sites as shown below while retaining certain PUFA functionality. These PUFA mimics do not have a diallyl position.

In another embodiment, the oxidizable bisallyl sites of the substituted compounds described herein (e.g., PUFAs) can prevent hydrogen abstraction by incorporating a divalent heteroatom (e.g., S, O), thereby eliminating bisallyl hydrogens as shown below. These PUFA mimetics also lack bisallyl hydrogen.

In another embodiment, PUFA mimetics (i.e., compounds that are structurally similar to natural PUFAs but are more resistant to oxidation due to structural differences) can be used for the purposes described above. The readily oxidizable bisallyl sites of PUFAs can prevent hydrogen extraction by dimethylation or halogenation as shown below. The hydrogen atom on the methyl group may optionally be halogen, such as fluorine, or deuterium. These PUFA mimics are dimethylated at the bisallyl site.

In another embodiment, the oxidizable bis-allyl sites of the PUFA can be protected from hydrogen extraction by alkylation as shown below. These PUFA mimics are dialkylated at the bisallyl site.

In another embodiment, cyclopropyl may be used to replace the double bond, thereby imparting certain functions to the acid while eliminating the bis allyl sites, as shown below. These PUFA mimics have cyclopropyl groups instead of double bonds.

In another embodiment, the double bond may be substituted with a 1, 2-substituted cyclobutyl group in the appropriate conformation to impart certain functions to the acid while eliminating the bis allyl sites, as shown below. These PUFA mimetics have a 1, 2-cyclobutyl substituted double bond.

In a modification of the previous embodiment of the mimetic of double bond substitution with 1, 2-cyclobutyl, the double bond can be substituted with a 1, 3-substituted cyclobutyl of appropriate conformation, thereby imparting certain functions to the acid while eliminating the bis allyl sites. The following PUFA mimetics have 1, 3-cyclobutyl substituted double bonds.

Bioisosteres are substituents or groups with similar physical or chemical properties that result in biological properties that are substantially similar to those of the compound. For example, well-known isosteres of hydrogen and/or bioisosteres include halogens (e.g., fluorine); isosteres of alkenes and/or bioisosteres include alkynes, benzene rings, cyclopropyl rings, cyclobutyl rings, cyclopentyl rings, cyclohexyl rings, thioethers, and the like; isosteres of carbonyl and/or bioisosteres include sulfoxide, sulfone, thiocarbonyl, and the like; isosteres of esters and/or bioisosteres include amides, sulfonates, sulfonamides, sulfinates, sulfenylamides (sulfenylamides), and the like. Thus, PUFA mimetics also include compounds having isosteric and/or bioisosteric functionalities.

In some embodiments, the PUFAs and/or PUFA mimetics are formulated into prodrugs for use in the various methods described herein. A prodrug is a pharmacological substance that may itself be biologically active, but after administration, the prodrug is metabolized to a form that also exerts biological activity. Many different types of prodrugs are known and can be divided into two main types according to their cellular metabolic sites. Type I prodrugs are those that are metabolized intracellularly, while type II are those that are metabolized extracellularly. It is well known that carboxylic acids can be converted to esters and various other functional groups to enhance pharmacokinetics (e.g., absorption, distribution, metabolism, and secretion). Esters are well known prodrug forms of carboxylic acids, formed by the condensation of an alcohol (or chemical equivalent thereof) with a carboxylic acid (or chemical equivalent thereof). In some embodiments, the alcohol (or chemical equivalent thereof) used to incorporate the PUFA prodrug comprises a pharmaceutically acceptable alcohol or chemical (which when metabolized results in a pharmaceutically acceptable alcohol). Such alcohols include, but are not limited to, propylene glycol, ethanol, isopropanol, 2- (2-ethoxyethoxy) ethanol (ii) ((iii))Gattefosse, Westwood, n.j.07675), benzyl alcohol, glycerol, polyethylene glycol 200, polyethylene glycol 300, or polyethylene glycol 400; polyoxyethylene castor oil derivatives (e.g., polyoxyethylene glycerol triricinoleate or polyoxyethylene 35 castor oil: (a)EL, BASF Corp.), polyoxyethylene glyceryl oxystearate (e.g., polyoxyethylene glyceryl oxystearate, and polyoxyethylene sorbitan monostearate (e.g., polyoxyethylene sorbitan monostearate)RH 40 (polyethylene glycol 40 hydrogenated castor oil) orRH 60 (polyethylene glycol 60 hydrogenated castor oil), BASF Corp.); a saturated polyglycolized glyceride (for example,35/10、44/14、46/07、50/13 or53/10, available from Gattefosse, Westwood, n.j.07675); polyoxyethylene alkyl ethers (e.g., cetomacrogol 1000); polyoxyethylene stearate (e.g., PEG-6 stearate, PEG-8 stearate, polyoxyethylene 40 stearate NF, polyoxyethylene 50 stearate NF, PEG-12 stearate, PEG-20 stearate, PEG-100 stearate, PEG-12 distearate, PEG-32 distearate or PEG-150 distearate); ethyl oleate, isopropyl palmitate, isopropyl myristate; dimethyl isosorbide; n-methyl pyrrolidone; paraffin wax; cholesterol; lecithin; a suppository base; a pharmaceutically acceptable wax (e.g., carnauba wax, yellow wax, white wax, microcrystalline wax, or emulsifying wax); a pharmaceutically acceptable silicon fluid; sorbitol fatty acid esters (including sorbitol laurate, sorbitol oleate, sorbitol palmitate, or sorbitol stearate); pharmaceutically acceptable saturated fats or pharmaceutically acceptable saturated oils (e.g. hydrogenated castor oil (glycerol-tri-12-hydroxystearate), cetyl esters wax (mainly C)14-C18C of saturated fatty acids14-C18A mixture of saturated esters having a melting point in the range of about 43-47 ℃), or glyceryl monostearate), and combinations thereof.

In some embodiments, the fatty acid prodrugs are represented by ester P-B, where group P is a PUFA and group B is a biologically acceptable molecule. Thus, cleavage of the ester P-B provides the PUFA and a biologically acceptable molecule. This cleavage can be induced by acids, bases, oxidizing agents and/or reducing agents. Examples of biologically acceptable molecules include, but are not limited to, nutrients, peptides, amino acids, proteins, carbohydrates (including mono-, di-, poly-, glycosaminoglycans, and oligosaccharides), nucleotides, nucleosides, lipids (including mono-, di-, and tri-substituted glycerols, glycerophospholipids, sphingolipids, and steroids), and combinations thereof.

In some embodiments, the alcohol (or chemical equivalent thereof) used to incorporate the PUFA prodrug includes a polyol (e.g., a diol, triol, tetraol, pentaol, etc.). Examples of the polyhydric alcohol include ethylene glycol, propylene glycol, 1, 3-butylene glycol, polyethylene glycol, methyl propylene glycol, ethoxy diethylene glycol, hexylene glycol, dipropylene glycol glycerin and carbohydrates. The esters formed from the polyols and PUFAs may be monoesters, diesters, triesters, and the like. In some embodiments, multiple esterified polyols are esterified with the same PUFA. In other embodiments, multiple esterified polyols are esterified with different PUFAs. In some embodiments, different PUFAs are stabilized in the same manner. In other embodiments, different PUFAs are stabilized in different ways (e.g., substituted with deuterium in one PUFA and substituted with deuterium in another PUFA)13C substitution). In some embodiments, one or more PUFAs are omega-3 fatty acids and one or more PUFAs are omega-6 fatty acids.

It is also contemplated that it may be useful to formulate the PUFA and/or PUFA mimetic and/or PUFA prodrug into a salt for use (e.g., as a pharmaceutically acceptable salt). For example, the use of salt formation as a means of modifying the properties of pharmaceutical compounds is well known. See Stahl et al, Handbook of pharmaceutical salts: Properties, selection and use (2002) Weinheim/Zurich: Wiley-VCH/VHCA; gould, Salt selection for basic drugs, Int J.Phann. (1986),33: 201-. Salt formation can be used to increase or decrease solubility, improve stability or toxicity, and reduce hygroscopicity of pharmaceutical products.

The salt formulation of PUFAs and/or PUFA esters and/or PUFA mimetics and/or PUFA prodrugs can include any of the PUFAs described herein.

It may not be necessary to replace all non-isotopically modified polyunsaturated fatty acids (e.g., non-deuterated PUFAs) with isotopically modified polyunsaturated fatty acids (e.g., deuterated PUFAs). In some embodiments, it is preferred to have sufficient isotopically modified PUFAs (e.g., D-PUFAs) in the membrane to prevent unmodified PUFAs (e.g., H-PUFAs) from sustaining an autoxidized chain reaction. During autoxidation, when one PUFA is oxidized and there is an unoxidized PUFA nearby, the unoxidized PUFA will be oxidized by the oxidized PUFA. This may also be referred to as autoxidation. In some cases, if low concentrations are present, such as "dilution" of the H-PUFA with D-PUFA in the membrane, this oxidation cycle may be interrupted by the distance separating the H-PUFA. In some embodiments, the isotopically modified PUFA is present in an amount sufficient to disrupt the autoxidative chain reaction. For example, to break the autoxidative chain reaction, an effective amount of isotopically modified PUFAs may be 1-60%, 5-50% or 15-35% of the total molecules of the same type in the membrane.

Pharmaceutical composition

Some embodiments include a pharmaceutical composition comprising: (a) a safe and therapeutically effective amount of a substituted compound as described herein; and (b) a pharmaceutically acceptable carrier, diluent, excipient, or combination thereof. In some embodiments, the substituted compounds are isotopically modified polyunsaturated acids (PUFAs) or esters, thioesters, amides, or other prodrugs thereof, or combinations thereof. In some further embodiments, the isotopically modified PUFA is 11,11-D2 linoleic acid or an ester thereof. In a particular embodiment, the isotopically modified PUFA is ethyl 11, 11-D2-linoleate.

The compounds useful as described above may be formulated into pharmaceutical compositions for the treatment of various conditions or disorders. Standard pharmaceutical formulation techniques, such as those disclosed in Remington's The Science and Practice of Pharmacy,21st Ed., Lippincott Williams & Wilkins (2005), are used, The entire contents of which are incorporated herein by reference.

In addition to selected compounds useful as described above, some embodiments include compositions comprising a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" as used herein refers to one or more compatible solid or liquid filler diluents or encapsulating substances suitable for mammalian administration. As used herein, the term "compatible" means that the components of the composition are capable of being mixed with the subject compound in a non-interactive manner, and with each other, which interaction would significantly reduce the pharmaceutical efficacy of the composition under ordinary use conditions. Of course, the pharmaceutically acceptable carriers must be of sufficiently high purity and sufficiently low toxicity to render them suitable for preferred administration to the animal, preferably a mammal, being treated.

Pharmaceutically acceptable carriers include, for example, solid or liquid fillers, diluents, hydrotropes, surfactants and encapsulating substances. Some examples of substances that can be used as pharmaceutically acceptable carriers or components thereof are sugars (e.g., lactose, glucose and sucrose); starches (e.g., corn starch and potato starch); cellulose and its derivatives (such as sodium carboxymethyl cellulose, ethyl cellulose and methyl cellulose); powdered gum tragacanth; malt; gelatin; talc powder; solid lubricants (e.g., stearic acid and magnesium stearate); calcium sulfate; vegetable oils (such as peanut oil, cottonseed oil, sesame oil, olive oil, corn oil, and cocoa butter); polyols (such as propylene glycol, glycerol, sorbitol, mannitol, and polyethylene glycol); alginic acid; emulsifiers (e.g., tween); wetting agents (such as sodium lauryl sulfide); a colorant; a flavoring agent; preparing tablets and stabilizing agents; an antioxidant; a preservative; pyrogen-free water; isotonic saline; and phosphate buffer solutions.

An optional pharmaceutically active substance may be included which does not substantially interfere with the inhibitory activity of the compound. The amount of carrier employed in conjunction with the compound is sufficient to provide the actual amount of material used per unit dose of the compound administered. Techniques and compositions for preparing dosage forms useful in the methods described herein are described in the following references, all of which are incorporated herein by reference: modern pharmacy, 4 th edition, chapters 9 and 10 (Banker & Rhodes, editions, 2002); lieberman et al, pharmaceutical dosage form: tablet (1989); and Ansel, introduction to pharmaceutical dosage forms, 8 th edition (2004).

Various oral dosage forms can be used, including solid forms such as tablets, capsules, granules, and bulk powders. Tablets may be compressed, tablet milled, enteric coated, sugar coated, film coated, or multiple compressed, containing suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow inducing agents, and melting agents. Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules, and effervescent formulations reconstituted from effervescent granules, including suitable solvents, preservatives, emulsifiers, suspending agents, diluents, sweeteners, melting agents, colorants and flavoring agents.

Pharmaceutically acceptable carriers suitable for use in preparing unit dosage forms for oral administration are well known in the art. Tablets typically contain conventional pharmaceutically compatible adjuvants as inert diluents (e.g., calcium carbonate, sodium carbonate, mannitol, lactose and cellulose); binders (e.g., starch, gelatin, and sucrose); disintegrating agents (such as starch, alginic acid and croscarmellose); lubricants (e.g., magnesium stearate, stearic acid, and talc). Glidants such as silicon dioxide can be used to improve the flow characteristics of the powder mixture. Colorants (such as FD & C dyes) may be added to improve appearance. Sweetening agents and flavoring agents, such as aspartame, saccharin, menthol, peppermint, and fruit flavors, are useful adjuvants for chewable tablets. Capsules typically contain one or more of the above-described solid diluents. The choice of carrier component depends on secondary considerations (such as taste, cost and shelf stability) which are not critical and can be readily selected by one skilled in the art.

Oral compositions also include liquid solutions, emulsions, suspensions, and the like. Pharmaceutically acceptable carriers suitable for use in preparing such compositions are well known in the art. Typical carrier ingredients for syrups, elixirs, emulsions and suspensions include ethanol, glycerol, propylene glycol, polyethylene glycol, liquid sucrose, sorbitol and water. For suspensions, typical suspending agents include methylcellulose, sodium carboxymethylcellulose, AVICEL RC-591, gum tragacanth and sodium alginate; typical wetting agents include lecithin and polysorbate 80; typical preservatives include methyl paraben and sodium benzoate. Oral liquid compositions may also comprise one or more ingredients such as sweetening agents, flavoring agents and coloring agents as disclosed above.

Such compositions may also be coated by conventional means, usually with a pH or time dependent coating, to allow release of the compound of interest in the gastrointestinal tract in the vicinity of the desired topical application, or at different times to prolong the desired effect. Such dosage forms typically include, but are not limited to, one or more of cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropyl methylcellulose phthalate, ethyl cellulose, Eudragit coatings, waxes, and shellac.

The compositions described herein may optionally include other pharmaceutically active substances or supplements. For example, the pharmaceutical composition is administered with one or more antioxidants. In some embodiments, the antioxidant is selected from the group consisting of: coenzyme Q, idebenone, mitoxantrone, mitoquinol, vitamin E, vitamin C, and combinations thereof. In some such embodiments, the at least one antioxidant may be administered simultaneously with, prior to, or after administration of the substituted compounds described herein. In some embodiments, the antioxidant and the substituted compound are in a single dosage form. In some embodiments, the single dosage form is selected from the group consisting of: pills, tablets and capsules.

Those skilled in the art will appreciate that many and various modifications may be made without departing from the spirit of the disclosure. Accordingly, it should be clearly understood that the embodiments disclosed herein are illustrative only and are not intended to limit the scope of the present invention. Any reference mentioned herein is incorporated by reference in its entirety for the materials discussed herein.

Examples

Example 1

In 2017, 3 months, a fatal neurodegenerative disease, namely infantile neuraxial cord malnutrition (INAD), was studied. At the beginning of the study, the subject was a 2.5 year old female with gene mutations in both copies of the PLA2G6 gene, mostly unresponsive to stimuli, and with little ability to perform any activity. All normally developing milestone events previously acquired have been lost. Prognosis is the need for a feeding tube in the recent progression of the disease. The patient received a six month trial according to the Expanded Access (Association Use) protocol, taking 1.8g of D-PUFA (11, 11-D2-linoleic acid) twice daily. Since she was unable to swallow the pill, the soft gel containing the D-PUFA was placed in a warm liquid (water or milk), punctured with a fork, and the active oil was pressed into the liquid, which was then soaked in food or cookies and taken. The patient's condition improved from within one month after administration, and continued for one year after administration. Figure 1 shows a detailed list of milestone events of the missing development of the subject prior to drug treatment, and observations by the treating physician versus baseline assessment at the start of the trial. These results were recorded in videotape examinations at baseline, 1 month, 3 months, 6 months, 9 months, and 12 months.

Figure 1 summarizes the baseline status and the treatment status of the patient for one year (degree of impairment: (0) severely impaired, (+1) moderately impaired, (+2) slightly impaired or not impaired).

Within the first week, chronic constipation resolved. She regained adequate bulbar function 2 months after taking the medication, could stop the syringe from feeding liquid, and regained drinking from the child's straw cup.

After 4 months of administration, no drug-related adverse events of any type were reported. Plasma deuterated polyunsaturated fatty acid levels were stable for one and three months, accounting for about 44% of total linoleic acid (diet plus modification), indicating excellent uptake and absorption of therapeutic levels (> about 10-15%) of drug. In the first and third months, deuterated arachidonic acid was observed to be 0.6% and 3.0% of total arachidonic acid in the plasma, respectively. In addition, since the start of the trial, clinical examination at 3 months showed no progression of the disease. Importantly, clinical examination videos, medical records, and caregiver reports all reveal the following observations: high doses of 11, 11-D2-linoleic acid are safe and well tolerated in children 2 years of age; no progression of the disease in the subject (n-1); parents noted the following improvements at 4 months, including: regaining the ability to grip and hold the spoon; resume the ability to swallow using a straw cup (vs. syringe feed); the ability to eat (chew and swallow) food (e.g., bananas); the salivation is reduced by 99 percent, and the salivation is basically recovered to be normal; better muscle strength (grasping and lifting the rattle); new responses to verbal requests during treatment; and constipation resolved, the subject was completely normal (constipation is a symptom of PLA2G6 disease).

After 6 months, she had improved in the qualitative measures obtained in the video treatment session or examination, including eye tracking, response to verbal instructions, head control, lifting, and reaching to grasp the spoon (a lost skill). INAD disease is a strictly progressive disease that only worsens over time. Recovery of degenerative stabilization and missing milestone events indicates the effectiveness of the trial treatment in one severely affected INAD patient.

Stabilization of the progression of the missing developmental milestone event is a major advance in INAD treatment. In view of the clinical findings, these clear reversals indicate that the substituted compounds described herein are effective in treating patients suffering from or at risk of suffering from diseases or conditions associated with impaired phospholipase a2 group VI (PLA2G6) activity, particularly the stabilized PUFA (11, 11-D2-linoleic acid), in patients with classical INAD disease.

Example 2

In this example, case studies and findings of a single patient with late Tay-Sachs disease (LOTS) treated with ethyl 11, 11-D2-linoleate were reported.

Designing/method: ethyl 11, 11-D2-linoleate was administered to patients at a dose of 2.7g (bid) and subjected to periodic repeated assessments including baseline PK measurements, Activities of Daily Living (ADL), 25 foot walking time (25FWT) and 6 minute walking distance (6 MWD). In particular, ADL is measured on a scale of 0 to 5 in a group of 12 elements representing language, strength, coordination, and the like.

As a result: 11, 11-D2-linoleic acid (D-LA) was extended to 13, 13-D2-arachidonic acid (D-AA) and both deuterated PUFAs reached significant plasma levels and Red Blood Cell (RBC) membrane incorporation within 1 month after administration. ADL, 25FWT and 6MWD were also improved. No significant toxicity was found (table 1).

TABLE 1

And (4) conclusion: early signs of therapeutic efficacy were found to inhibit disease progression and some regression. In addition, 11,11-D2 ethyl linoleate was well tolerated and reported to be non-toxic.

37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:用于治疗房性心律失常的TASK-1抑制剂

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!