Treatment of fatty liver disease

文档序号:411091 发布日期:2021-12-17 浏览:33次 中文

阅读说明:本技术 脂肪性肝病的治疗 (Treatment of fatty liver disease ) 是由 S·B·兰多 C·R·谢泼德 于 2020-01-08 设计创作,主要内容包括:本发明涉及一种在有需要的受试者中治疗脂肪性肝病的方法,该方法包括向该受试者施用治疗有效量的具有式(I)的化合物5’-N~(1)N~(2)N~(3)N~(4)N~(5)N~(6)-N~(7)N~(8)-dC~(m)-dG~(c)-N~(11)N~(12)N~(13)N~(14)N~(15)N~(16)-N~(17)N~(18)-3’。该脂肪性肝病可以是非酒精性脂肪性肝病,如单纯性脂肪性肝病或NASH。本发明还涉及脂联素作为生物标志物用于以下的用途:鉴定更有可能对使用具有式(I)的化合物5’-N~(1)N~(2)N~(3)N~(4)N~(5)N~(6)-N~(7)N~(8)-dC~(m)-dG~(c)-N~(11)N~(12)N~(13)N~(14)N~(15)N~(16)-N~(17)N~(18)-3’治疗有响应的受试者,以及用于评估该受试者在使用具有式(I)的化合物5’-N~(1)N~(2)N~(3)N~(4)N~(5)N~(6)-N~(7)N~(8)-dC~(m)-dG~(c)-N~(11)N~(12)N~(13)N~(14)N~(15)N~(16)-N~(17)N~(18)-3’治疗期间的响应。(The present invention relates to a method of treating fatty liver disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound 5' -N of formula (I) 1 N 2 N 3 N 4 N 5 N 6 ‑N 7 N 8 ‑dC m ‑dG c ‑N 11 N 12 N 13 N 14 N 15 N 16 ‑N 17 N 18 -3'. The fatty liver disease can be a non-alcoholic fatty liver disease, such as simple fatty liver disease or NASH. The invention also relates to the use of adiponectin as a biomarker for: identification of 5' -N more likely to be useful for the use of compounds of formula (I) 1 N 2 N 3 N 4 N 5 N 6 ‑N 7 N 8 ‑dC m ‑dG c ‑N 11 N 12 N 13 N 14 N 15 N 16 ‑N 17 N 18 -3 'treatment of a subject responsive thereto, and for assessing that the subject is using compound 5' -N having formula (I) 1 N 2 N 3 N 4 N 5 N 6 ‑N 7 N 8 ‑dC m ‑dG c ‑N 11 N 12 N 13 N 14 N 15 N 16 ‑N 17 N 18 -response during 3' treatment.)

1. A method of treating fatty liver disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound represented by formula (I):

5’-N1N2N3N4N5N6-N7N8-dCm-dGc-N11N12N13N14N15N16-N17N18-3’ (I)

or a pharmaceutically acceptable salt thereof

Wherein N is1To N6And N11To N16Each independently is a 2' -deoxyribonucleoside;

N7、N8、N17and N18Each independently is a ribonucleoside;

dCmis a 5-methyl-cytosine 2' -deoxynucleoside having the structural formula:

dGcis a 7-deaza-guanine 2' -deoxynucleoside having the following structural formula:

and is

Any two adjacent ribonucleosides are covalently linked by a phosphodiester linker or a phosphorothioate linker.

2. The method of claim 1, wherein the N is7Is a guanine 2' -methoxy-nucleoside represented by the following structural formula:

3. the method of claim 1 or claim 2, wherein N is8Is a uracil 2' -methoxy-nucleoside represented by the following structural formula:

4. the method of any one of claims 1-3, wherein N is17Is guanine 2' -methoxy-nucleoside.

5. The method of any one of claims 1-4, wherein N is18Is uracil 2' -methoxy-nucleoside.

6. The method of claim 1, wherein N is7Is guanine 2' -methoxy-nucleoside, N8Is uracil 2' -methoxy-nucleoside, N17Is guanine 2' -methoxy-nucleoside, and N18Is uracil 2' -methoxy-nucleoside.

7. The method of any one of claims 1-6, wherein N is1Is cytosine 2' -deoxynucleoside, N2Is thymine 2' -deoxynucleoside, N3Is adenine 2' -deoxynucleoside, N4Is thymine

2' -deoxynucleoside, N5Is cytosine 2' -deoxynucleoside, and N6Is thymine

2' -deoxynucleosides.

8. The method of any one of claims 1-7, wherein N is11Is thymine 2' -deoxynucleoside, N12Is thymine 2' -deoxynucleoside, N13Is cytosine 2' -deoxynucleoside, N14Is thymine

2' -deoxynucleoside, N15Is cytosine 2' -deoxynucleoside, and N16Is thymine

2' -deoxynucleosides.

9. The method of claim 1, wherein the oligonucleotide having formula (I) is represented by the following structural formula:

5’-dC-dT-dA-dT-dC-dT-(G-Rib2Me)-(U-Rib2Me)-dCm-dGc-dT-dT-dC-dT-dC-dT-(G-Rib2Me)-(U-Rib2Me)-3’ (II)

or a pharmaceutically acceptable salt thereof.

10. The method of claim 1, wherein the oligonucleotide having formula (I) is represented by the following structural formula:

5’-dC-Sp-dT-Sp-dA-Sp-dT-Sp-dC-Sp-dT-Sp-(G-Rib2Me)-Sp-(U-Rib2Me)-Sp-dCm-Sp-dGc-Sp-dT-Sp-dT-Sp-dC-Sp-dT-Sp-dC-Sp-dT-Sp-(G-Rib2Me)-Sp-(U-Rib2Me)-3’ (III)

or a pharmaceutically acceptable salt thereof, wherein Sp represents a phosphorothioate linker in the "Sp" configuration.

11. The method of any one of claims 1-10, wherein the oligonucleotide is in the form of a sodium salt.

12. The method of claim 1, wherein the oligonucleotide is Ballotora Pacticola (Bazlitoran).

13. The method of any one of claims 1-12, wherein the fatty liver disease is non-alcoholic fatty liver disease.

14. The method of claim 13, wherein the non-alcoholic fatty liver disease is non-alcoholic steatohepatitis.

15. Identification of fatty liver disease in a subject

5’-N1N2N3N4N5N6-N7N8-dCm-dGc-N11N12N13N14N15N16-N17N18-3’ (I)

Or a pharmaceutically acceptable salt thereof, wherein N is1To N6And N11To N16Each independently is a 2' -deoxyribonucleoside;

N7、N8、N17and N18Each independently is a ribonucleoside;

dCmis a 5-methyl-cytosine 2' -deoxynucleoside having the structural formula:

dGcis a 7-deaza-guanine 2' -deoxynucleoside having the following structural formula:

and is

Any two adjacent ribonucleosides are covalently linked by a phosphodiester linker or a phosphorothioate linker, the method comprising:

a) obtaining or providing a plasma sample from a subject having fatty liver disease;

b) measuring plasma adiponectin levels in the sample; and

c) comparing the plasma adiponectin levels to adiponectin control levels;

wherein the fatty liver disease is identified as more likely to be responsive to administration of the compound of formula (I) if the plasma adiponectin level is less than the control level.

16. The method of claim 15, wherein the control level of adiponectin is 38 μ g/mL.

17. An assessment of use of a compound having formula (I) in a subject in need thereof

5’-N1N2N3N4N5N6-N7N8-dCm-dGc-N11N12N13N14N15N16-N17N18-3’ (I)

Or a pharmaceutically acceptable salt thereof, for the treatment of fatty liver disease, wherein,

N1to N6And N11To N16Each independently is a 2' -deoxyribonucleoside;

N7、N8、N17and N18Each independently is a ribonucleoside;

dCmis a 5-methyl-cytosine 2' -deoxynucleoside having the structural formula:

dGcis a 7-deaza-guanine 2' -deoxynucleoside having the following structural formula:

and is

Any two adjacent ribonucleosides are covalently linked by a phosphodiester linker or a phosphorothioate linker for use in the treatment of fatty liver disease, the method comprising:

a) detecting adiponectin levels in a plasma sample of the subject at a first time point;

b) repeating step a) at least one subsequent time point after administration of the compound represented by formula (I); and

c) comparing the levels detected in step a) and step b), wherein an increase in adiponectin level relative to at least one subsequent subject plasma sample indicates that the compound having formula (I) treats fatty liver disease in the subject.

Background

Alcoholic and non-alcoholic fatty liver diseases (also known as hepatic steatosis) are a common liver disorder that occurs when lipids accumulate in hepatocytes. Lipid accumulation can cause cellular damage, sensitize the liver to further damage, and impair hepatic microvascular circulation. The etiology of fatty liver disease is associated with excessive alcohol consumption, metabolic disorders, eating conditions, exposure to certain chemicals and drugs, and pregnancy complications (e.g., preeclampsia).

Fatty liver disease is a major health burden worldwide. Nonalcoholic fatty liver disease (NAFLD) has a prevalence in the population of between 15% and 37% and is considered to be the most common liver disease worldwide. Furthermore, it is also believed that as many as 3% to 10% of obese children are affected by NAFLD. NAFLD can progress to more severe liver disease, such as nonalcoholic steatohepatitis (NASH), a condition characterized by hepatitis and liver damage, and is often accompanied by liver fibrosis or cirrhosis, which can further lead to end-stage liver disease and primary liver cancer. The prevalence of NASH in the general population is 3% to 10%.

Currently, there is no approved treatment for NAFLD and NASH. Generally, current therapies include healthy lifestyle and non-specific metabolic regulators.

Disclosure of Invention

The present invention relates to a method of treating fatty liver disease. The method comprises administering to a subject in need thereof a therapeutically effective amount of a compound having formula (I) or a pharmaceutically acceptable salt thereof.

In one embodiment, the fatty liver disease is non-alcoholic fatty liver disease (NAFLD).

In a specific embodiment, NAFLD is simple fatty liver disease.

In another embodiment, the NAFLD is nonalcoholic steatohepatitis (NASH).

In yet another embodiment, the fatty liver disease is Alcoholic Liver Disease (ALD).

In another embodiment, the present invention relates to the use of a compound having formula (I), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of fatty liver disease.

In yet another embodiment, the present invention relates to a compound having formula (I), or a pharmaceutically acceptable salt thereof, for use in treating fatty liver disease.

In yet another embodiment, the invention relates to a method of identifying the likelihood that a fatty liver disease in a subject will respond to administration of a compound having formula (I), wherein the method comprises: a) obtaining or providing a plasma sample from a subject having fatty liver disease;

b) measuring plasma adiponectin levels in the sample; and

c) comparing the plasma adiponectin levels to adiponectin control levels;

wherein the fatty liver disease is identified as more likely to be responsive to administration of the compound of formula (I) if the plasma adiponectin level is less than the control level.

In a specific embodiment, the control level of adiponectin is 38 μ g/mL.

In certain embodiments, the control level of adiponectin is 38 μ g/mL or less. For example, adiponectin levels are 38. mu.g/ml, 37. mu.g/ml, 36. mu.g/ml, 35. mu.g/ml, 34. mu.g/ml, 33. mu.g/ml, 32. mu.g/ml, 31. mu.g/ml, 30. mu.g/ml, 29. mu.g/ml, 28. mu.g/ml, 27. mu.g/ml, 26. mu.g/ml, 25. mu.g/ml, 24. mu.g/ml, 23. mu.g/ml, 22. mu.g/ml, 21. mu.g/ml, 20. mu.g/ml, 19. mu.g/ml, 18. mu.g/ml, 17. mu.g/ml, 16. mu.g/ml, 15. mu.g/ml, 14. mu.g/ml, 13. mu.g/ml, 12. mu.g/ml, 11. mu.g/ml, 10. mu.g/ml, 9. mu.g/ml, 8. mu.g/ml, 7. mu.g/ml, 6. mu.g/ml, 5. mu.g/ml, 4. mu.g/ml, 3. mu.g/ml, 2. mu.g/ml, 1. mu.g/ml, 0.5. mu.g/ml, 0.4. mu.g/ml, 0.3. mu.g/ml, 0.2. mu.g/ml or 0.1. mu.g/ml.

In additional embodiments, the present invention relates to a method of assessing the efficacy of a compound represented by formula (I) in treating fatty liver disease in a subject in need thereof, the method comprising:

a) detecting adiponectin levels in a plasma sample of the subject at a first time point;

b) repeating step a) at least one subsequent time point after administration of the compound represented by formula (I); and

c) comparing the levels detected in step a) and step b), wherein an increase in adiponectin level relative to at least one subsequent subject plasma sample indicates that the compound having formula (I) treats fatty liver disease in the subject.

Drawings

Fig. 1 is a graph showing the change in body weight of the control group (group 1) and the treatment group (group 2) during the treatment period.

Figure 2 is a structural formula representing the sodium salt of Ballitoran (also referred to herein as AVO 010).

Detailed Description

A description of example embodiments of the invention follows.

While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

The teachings of all patents, published applications, and references cited herein are incorporated by reference in their entirety.

Compounds for administration

The compounds described herein for treating fatty liver disease (e.g., NASH) are 18-mer 5 'to 3' oligonucleotides represented by the following structural formula (I):

5’-N1N2N3N4N5N6-N7N8-dCm-dGc-N11N12N13N14N15N16-N17N18-3' (I) or a pharmaceutically acceptable salt thereof.

In structural formula (I):

N1to N6And N11To N16Each independently is a 2' -deoxyribonucleoside;

N7、N8、N17and N18Each independently a ribonucleoside, e.g.,

2' -methoxy-ribonucleosides;

dCmis a 5-methyl-cytosine 2' -deoxynucleoside ("m") having the structural formula5C-dRib”、“m5Cyt-dRib', or "m5Cyt-dRibf”):

dGcIs a 7-deaza-guanine 2' -deoxynucleoside of the formula7G-dRib”、“c7Gua-dRib', or "c7Gua-Ribf”):

And is

Any two adjacent ribonucleosides are covalently linked by a phosphodiester linker or a phosphorothioate linker, as represented by the following structural formula:

it is understood that each phosphorothioate bond introduces a chiral center designated as "Sp" or "Rp". Unless otherwise indicated, compounds of the invention that include a phosphorothioate linker may include Sp or Rp stereoisomers. In one embodiment, all linkers in the compound of formula (I) are Sp phosphorothioate linkers.

In a first exemplary embodiment of the oligonucleotide of formula (I), N7Is a guanine 2' -methoxy-nucleoside ("G-Rib 2 Me", "Gua-Rib 2 Me", or "Gua-Ribf 2 Me") represented by the following structural formula:

in a second exemplary embodiment, N8Is a uracil 2' -methoxy-nucleoside ("U-Rib 2 Me", "Ura-Rib 2 Me", or "Ura-Ribf 2 Me") represented by the following structural formula:

in a third exemplary embodiment, N17Is guanine 2' -methoxy-nucleoside. In a fourth example embodiment, N18Is uracil 2' -methoxy-nucleoside.

In a fifth exemplary embodiment of the oligonucleotide of formula (I), N7Is guanine 2' -methoxy-nucleoside, N8Is uracil 2' -methoxy-nucleoside, N17Is guanine 2' -methoxy-nucleoside, and N18Is uracil 2' -methoxy-nucleoside.

In a sixth exemplary embodiment of the oligonucleotide of formula (I), N1Is a cytosine 2' -deoxynucleoside ("dC" or "dCyd"), N2Is thymine 2' -deoxynucleoside ("dT" or "dThd"), N3Is adenine 2' -deoxynucleoside ("dA" or "dAdo"), N4Is thymine 2' -deoxynucleoside, N5Is cytosine 2' -deoxynucleoside, and N6Is thymine 2' -deoxynucleoside. N is a radical of7、N8、N17And N18The values and exemplary values of (a) are as described above for the first to fifth exemplary embodiments of the oligonucleotide of formula (I).

In the presence of a knotIn a seventh exemplary embodiment of the oligonucleotides of the formula (I), N11Is thymine 2' -deoxynucleoside, N12Is thymine 2' -deoxynucleoside, N13Is cytosine 2' -deoxynucleoside, N14Is thymine 2' -deoxynucleoside, N15Is cytosine 2' -deoxynucleoside, and N16Is thymine 2' -deoxynucleoside. N is a radical of1To N8And N17And N18The values and exemplary values of (a) are as described above for the first to sixth exemplary embodiments of the oligonucleotide of formula (I).

In an eighth exemplary embodiment of the oligonucleotide having structural formula (I), the oligonucleotide is represented by the following structural formula:

5’-dC-dT-dA-dT-dC-dT-(G-Rib2Me)-(U-Rib2Me)-dCm-dGc-dT-dT-dC-dT-dC-dT-(G-Rib2Me)-(U-Rib2Me)-3’ (II)。

in a ninth exemplary embodiment of the oligonucleotide having structural formula (I), the oligonucleotide is represented by the following structural formula:

5’-dC-Sp-dT-Sp-dA-Sp-dT-Sp-dC-Sp-dT-Sp-(G-Rib2Me)-Sp-(U-Rib2Me)-Sp-dCm-Sp-dGc-Sp-dT-Sp-dT-Sp-dC-Sp-dT-Sp-dC-Sp-dT-Sp-(G-Rib2Me)-Sp-(U-Rib2Me)-3’ (III)

or a pharmaceutically acceptable salt thereof. In a particular aspect, the oligonucleotide represented by structural formula (III) is in the form of a sodium salt represented by structural formula (IIIA):

5’-dC-Sp-dT-Sp-dA-Sp-dT-Sp-dC-Sp-dT-Sp-(G-Rib2Me)-Sp-(U-Rib2Me)-Sp-dCm-Sp-dGc-Sp-dT-Sp-dT-Sp-dC-Sp-dT-Sp-dC-Sp-dT-Sp-(G-Rib2Me)-Sp-(U-Rib2Me)-3’·17Na+ (IIIA)。

in a tenth exemplary embodiment, the oligonucleotide used in the methods described herein is a sodium salt of an oligonucleotide represented by structural formula (III) and is commonly referred to in the art as barr-ton or IMO-8400. The molecular weight of the non-salt form of IMO-8400 is 5800.67g/mol and the molecular formula of the non-salt form of IMO-8400 is C179H233N52O101P17S17. Sodium salt of Balttoran (also referred to herein as AVO)010) Represented by the structural formula shown in fig. 2. The IUPAC name of a compound represented by the structural formula depicted in FIG. 2 is

5' -hydroxy-2 ' -deoxy-P-thiocytidinyl- (3 ' → 5 ') -2 ' -deoxy-P-thioxanthyl- (3 ' → 5 ') -2 ' -deoxy-P-thiocytidinyl- (3 ' → 5 ') -2 ' -deoxy-P-thioxanthyl- (3 ' → 5 ') -2 ' -O-methyl-P-thioxanthyl- (3 ' → 5 ') -2 ' -deoxy-P-thiothymidine Para-5-methyl-cytidylyl- (3 ' → 5 ') -2 ' -deoxy-P-thioxo-7-deaza-guanylyl- (3 ' → 5 ') -2 ' -deoxy-P-thioxanthylyl- (3 ' → 5 ') -2 ' -deoxy-P-thioxanthyl- (3 ' → 5 ') -2 ' -deoxy-P-thioxanthylyl- (3 ' → 5 ') -2 ' -O-methyl-P-thioxanthylyl - (3 '→ 5') -3 '-hydroxy-2' -O-methyluridine 17 sodium salt.

As used herein, the term "ribonucleoside" refers to a compound having the following structural formula:

wherein the base can be any one of nitrogenous bases, such as a pyrimidine-derived base or a purine-derived base; and, for example, the nucleobases adenine (a), uracil (U), guanine (G), thymine (T), and cytosine (C), each of which may be optionally modified. Unless specifically indicated otherwise, ribonucleosides include a 2' -hydroxyl group. The 2 '-deoxyribonucleoside includes a-CH at the 2' position2-a group.

Fatty liver disease

Fatty liver disease occurs when excess fat accumulates in the liver. There are two main types of fatty liver disease. The first type of fatty liver disease is non-alcoholic fatty liver disease (NAFLD). The second type of fatty liver disease is alcoholic fatty liver disease (ALD).

NAFLD includes simple fatty liver and nonalcoholic steatohepatitis (NASH). Simple fatty liver means that there is an excess of fat in the liver, but no inflammation. NASH is much more severe than simple fatty liver. NASH means that there is fat and also inflammation in the liver, and in some cases, damage to liver cells. Inflammation and hepatocyte injury that accompany NASH can cause serious problems, such as liver fibrosis (scarring of the liver), cirrhosis (severe scarring of the liver), and liver cancer.

ALD includes alcoholic hepatitis and alcoholic cirrhosis.

In some embodiments, the methods described herein may further comprise improving the NAS score of the subject. NAS score can be improved by at least 30%.

A "therapeutically effective amount" of an oligonucleotide or a salt thereof as described herein is an amount that, when administered to a subject having a disease or disorder, has the intended therapeutic effect (e.g., reduces, ameliorates, alleviates, or eliminates one or more manifestations of the disease or disorder in the subject). A therapeutically effective amount of a compound having formula (I) may be from about 0.5mg/kg to about 50 mg/kg. For example, from about 0.5mg/kg to about 25mg/kg and 0.5mg/kg to about 20 mg/kg. Suitable dosages include about 0.75mg/kg, about 1.5mg/kg, about 3mg/kg, and about 6 mg/kg. In a particular embodiment, the dose is 3 mg/kg. In another embodiment, the dose is 3mg/kg and the dose is administered once per week.

As used herein, a method of treatment means ameliorating, preventing or alleviating the symptoms and/or effects associated with a disorder or condition. The disorder or condition is fatty liver disease. In one embodiment, the fatty liver disease is non-alcoholic fatty liver disease (NAFLD). In particular embodiments, the NAFLD is simple fatty liver (steatosis). In another embodiment, the NAFLD is nonalcoholic steatohepatitis (NASH).

Pharmaceutical composition/method of administration

The compositions and methods of the invention can be used to treat a subject in need thereof. In certain embodiments, the subject is a mammal (e.g., a human) or a non-mammal. When administered to a subject (e.g., a human), the composition or compound is preferably administered as a pharmaceutical composition comprising, for example, a compound of the invention and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions, such as water or physiological buffered saline, or other solvents or vehicles, such as glycols, glycerol, oils (such as olive oil), or injectable organic esters. In particular embodiments, when such pharmaceutical compositions are for human administration, particularly for invasive routes of administration (i.e., routes such as injection or implantation that circumvent transport or diffusion across epithelial barriers), the aqueous solution is pyrogen-free or substantially pyrogen-free. The excipient may be selected to achieve, for example, delayed release of the agent or to selectively target one or more cells, tissues or organs. The pharmaceutical compositions may be in dosage unit forms such as tablets, capsules (including discrete capsules and gelatin capsules), granules, lyophilizates for reconstitution, powders, solutions, syrups, suppositories, injections and the like. The composition can also be present in a transdermal delivery system (e.g., a skin patch). The composition may also be present in a solution suitable for topical administration (e.g., eye drops).

The pharmaceutically acceptable carrier may contain physiologically acceptable agents that act, for example, to stabilize a compound (e.g., a compound of the invention), to increase its solubility, or to increase its absorption. Such physiologically acceptable agents include, for example, carbohydrates (such as glucose, sucrose, or dextran), antioxidants (such as ascorbic acid or glutathione), chelating agents, low molecular weight proteins, or other stabilizers or excipients. The choice of a pharmaceutically acceptable carrier (including physiologically acceptable agents) depends, for example, on the route of administration of the composition. The formulation or pharmaceutical composition may be a self-emulsifying drug delivery system or a self-microemulsifying drug delivery system. The pharmaceutical compositions (formulations) may also be liposomes or other polymeric matrices into which, for example, the compounds of the invention may be incorporated. Liposomes (e.g., comprising phospholipids or other lipids) are non-toxic, physiologically acceptable and metabolizable carriers that are relatively simple to prepare and administer.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

As used herein, the phrase "pharmaceutically acceptable carrier" means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Some examples of materials that can be used as pharmaceutically acceptable carriers include: (1) sugars such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) ringer's solution; (19) ethanol; (20) a phosphate buffer solution; and (21) other non-toxic compatible substances used in pharmaceutical formulations.

The pharmaceutical compositions (formulations) can be administered to a subject by any of a variety of routes of administration, including, for example, oral (e.g., drenches in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including dispersion and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingual); anal, rectal or vaginal (e.g., in pessaries, creams or foams); parenteral (including intramuscular, intravenous, subcutaneous, or intrathecal in, for example, sterile solutions or suspensions); transnasally; intraperitoneal administration; subcutaneous injection; transdermal (e.g., as a patch applied to the skin); and topically (e.g., as a cream, ointment, or spray applied to the skin, or as eye drops). The compounds may also be formulated for inhalation. In certain embodiments, the compound may simply be dissolved or suspended in sterile water.

The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form will vary depending upon the subject being treated, the particular mode of administration. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form is generally that amount of the compound which produces a therapeutic effect. Generally, this amount will range from about 1% to about 99% active ingredient, preferably from about 5% to about 70%, most preferably from about 10% to about 30%, within the one hundred percent range.

The method of preparing these formulations or compositions comprises the step of combining the active compound (such as a compound of the present invention) with a carrier and optionally one or more accessory ingredients. In general, formulations are prepared by uniformly and intimately bringing into association a compound of the invention with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.

Formulations of the invention suitable for oral administration may be in the form of capsules (including dispersion capsules and gelatin capsules), cachets, pills, tablets, lozenges (using a flavored base, usually sucrose and acacia or tragacanth), lyophilizates, powders, granules, or as a solution or suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as lozenges (using an inert base such as gelatin and glycerin, or sucrose and acacia) and/or as mouthwashes and the like, each containing a predetermined amount of a compound of the invention as an active ingredient. The compositions or compounds may also be administered as a bolus, electuary or paste.

To prepare solid dosage forms for oral administration (capsules (including dispersion and gelatin capsules), tablets, pills, dragees, powders, granules, and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers (such as sodium citrate or dicalcium phosphate) and/or any of the following: (1) fillers or extenders, such as starch, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption promoters, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) adsorbents such as kaolin and bentonite; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof; (10) complexing agents, such as modified and unmodified cyclodextrins; and (11) a colorant. In the case of capsules (including dispersion capsules and gelatin capsules), tablets and pills, the pharmaceutical compositions may also contain buffering agents. Solid compositions of a similar type may also be employed as fillers in soft-filled and hard-filled gelatin capsules using excipients such as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

Tablets may be prepared by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binders (for example, gelatin or hydroxypropylmethyl cellulose), lubricants, inert diluents, preservatives, disintegrating agents (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agents. Molded tablets may be prepared by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.

Tablets and other solid dosage forms of the pharmaceutical compositions, such as dragees, capsules (including both dispersion capsules and gelatin capsules), pills and granules, can optionally be scored or prepared with useful coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may also be formulated with, for example, hydroxypropylmethyl cellulose in varying proportions to provide slow or controlled release of the active ingredient therein to provide a desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water or some other sterile injectable medium immediately prior to use. Optionally, these compositions may also contain opacifying agents and may be of a composition that it releases the active ingredient or ingredients only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient may also be in microencapsulated form, if appropriate with one or more of the abovementioned excipients.

Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.

In addition to inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.

Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

Formulations of pharmaceutical compositions for rectal, vaginal, or urethral administration can be presented as a suppository, which can be prepared by mixing one or more active compounds with one or more suitable non-irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature but liquid at body temperature and will therefore melt in the rectum or vaginal cavity and release the active compound.

Formulations of pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, or oral spray, or oral ointment.

Alternatively or additionally, the composition may be formulated for delivery via a catheter, stent, wire, or other intraluminal device. Delivery via such devices may be particularly useful for delivery to the bladder, urethra, ureter, rectum, or small intestine.

Formulations suitable for vaginal administration also include pessary, tampon, cream, gel, paste, foam or spray formulations containing such carriers as are known in the art to be suitable.

Dosage forms for topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.

Ointments, pastes, creams and gels may contain, in addition to the active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to the active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. In addition, sprays can contain conventional propellants, such as chlorofluorocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

Transdermal patches have the additional advantage of controlled delivery of the compounds of the present invention to the body. Such dosage forms may be prepared by dissolving or dispersing the active compound in a suitable medium. Absorption enhancers may also be used to increase the transdermal flux of the compound. This flux rate can be controlled by providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.

As used herein, the phrase "parenteral administration and administered parenterally" means modes of administration other than enteral and topical administration, typically by injection, and includes, but is not limited to, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.

Pharmaceutical compositions suitable for parenteral administration comprise an active compound in combination with: one or more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted in a sterile injectable solution or dispersion immediately prior to use may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

Examples of suitable aqueous and nonaqueous carriers that can be used in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters (such as ethyl oleate). For example, suitable fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.

These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Prevention of the action of microorganisms can be ensured by including various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol sorbic acid, and the like). It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like in the compositions. In addition, prolonged absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

In some cases, in order to prolong the effect of the drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This can be achieved by using liquid suspensions of crystalline or amorphous materials that are poorly water soluble. The rate of absorption of the drug then depends on its rate of dissolution, which in turn may depend on crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is achieved by dissolving or suspending the drug in an oil vehicle.

Injectable depot forms are prepared by forming a microcapsule matrix of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer and the nature of the particular polymer used, the release rate of the drug can be controlled. Examples of other biodegradable polymers include poly (orthoesters) and poly (anhydrides). Injectable depot formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.

For use in the methods of the invention, the active compound may be administered per se or as a pharmaceutical composition containing, for example, 0.1% to 99.5% (more preferably, 0.5% to 90%) of the active ingredient in combination with a pharmaceutically acceptable carrier.

The method of introduction may also be provided by a rechargeable or biodegradable device. A variety of sustained release polymeric devices have been developed in recent years and have been tested in vivo for the controlled delivery of drugs, including protein biopharmaceuticals. A variety of biocompatible polymers (including hydrogels), including biodegradable and non-degradable polymers, can be used to form implants for the sustained release of compounds at specific target sites.

The actual dosage level of the active ingredient in the pharmaceutical composition can be varied to obtain an amount of the active ingredient effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without toxicity to the patient.

The selected dosage level will depend upon a variety of factors including the activity of the particular compound or combination of compounds or esters, salts or amides thereof employed, the route of administration, the time of administration, the rate of excretion of the particular compound or compounds being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound or compounds employed, the age, sex, weight, condition, general health and past medical history of the subject being treated, and like factors well known in the medical arts.

A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the required pharmaceutical composition. For example, a physician or veterinarian can begin administration of the pharmaceutical compositions and compounds at levels lower than required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. By "therapeutically effective amount" is meant a concentration of the compound sufficient to produce the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary depending on the weight, sex, age, and medical history of the subject. Other factors that affect an effective amount may include, but are not limited to, the severity of the condition in the subject, the disorder being treated, the stability of the compound, and, if desired, another type of therapeutic agent to be administered with the compound of the invention. A larger total dose may be delivered by multiple administrations of the agent. Methods for determining efficacy and dosage are known to those skilled in the art (Isselbacher et al (1996) Harrison's Principles of Internal Medicine 13ed. [ Harrison's Principles of Internal Medicine (13 th edition) ],1814-1882, which is incorporated herein by reference).

In general, a suitable daily dose of the active compound for use in the compositions and methods of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Generally such effective dosages will depend on the factors set forth above.

If desired, effective daily doses of the active compound may be administered separately as single, two, three, four, five, six or more sub-doses at appropriate intervals throughout the day, optionally in unit dosage forms. In certain embodiments of the invention, the active compound may be administered two or three times per day. In a specific embodiment, the active compound will be administered once daily.

In certain embodiments, the compounds of the present invention are administered alone or in combination with another class of therapeutic agents. As used herein, the phrase "co-administration" refers to any form of administration of two or more different therapeutic compounds that results in administration of a second compound while a previously administered therapeutic compound is still effective in vivo (e.g., two compounds are effective simultaneously in a subject, which may include a synergistic effect of the two compounds). For example, different therapeutic compounds may be administered simultaneously or sequentially in the same formulation or in separate formulations. In certain embodiments, the different therapeutic compounds may be administered sequentially over a period of one hour, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, or one week. Thus, a subject receiving such treatment may benefit from the combined effects of different therapeutic compounds.

In certain embodiments, the co-administration of a compound of the present invention with one or more additional therapeutic agents provides improved efficacy relative to each individual administration of the compound of the present invention or one or more additional therapeutic agents. In certain embodiments, the combined administration provides an additive effect, wherein additive effect refers to the sum of each effect of the compound of the invention and one or more additional therapeutic agents administered separately.

The invention includes the use of pharmaceutically acceptable salts of the compounds of the invention in the compositions and methods of the invention. By "pharmaceutically acceptable salt" is meant any non-toxic salt that is capable of providing, directly or indirectly, a compound of the invention upon administration to a recipient. A "pharmaceutically acceptable counterion" is an ionic moiety of a salt that is not toxic when released from the salt upon administration to a recipient. In certain embodiments, salts contemplated by the present invention include, but are not limited to, Na salts, Ca salts, K salts, Mg salts, Zn salts, or other metal salts. In a specific embodiment, the salt of the present invention (salt of the compound represented by formula (I)) is a sodium salt.

Wetting agents, emulsifying agents, and lubricating agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preserving and anti-oxidants may also be present in the composition.

Examples of pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants such as ascorbyl palmitate, Butyl Hydroxyanisole (BHA), Butyl Hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents such as citric acid, ethylenediaminetetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. While other compounds or methods may be used in practice or testing, certain preferred methods are now described in the context of the following preparations and protocols.

Experimental methods

Efficacy of IMO-8400 in diet-induced NAFLD/NASH cynomolgus monkeys

The objective of this study was to determine the efficacy of IMO-8400 in diet-induced NAFLD/NASH male cynomolgus monkeys after Subcutaneous (SC) administration of IMO-8400 at 3mg/kg once a week for 12 consecutive weeks.

Twelve NAFLD/NASH male cynomolgus monkeys were identified based on liver histopathology and cfDNA levels. Animals were randomized into the following groups:

group 1: n-5; vehicle (sterile saline) was administered by subcutaneous injection once weekly for 12 consecutive weeks.

Group 2, n is 7; the IMO-8400 solution was administered by subcutaneous injection once a week for 12 consecutive weeks.

The results are presented in detail below, and include the following observations:

a) adiponectin levels: adiponectin levels were significantly increased at week 12 in 6 of 7 animals administered IMO-8400 compared to pre-dose (baseline) levels.

b) Weight: the mean animal body weight in group 1 (control group) was reduced by 3.27% and in group 2 (treatment group) by 10.87% at day 84 (end of treatment) compared to day 1.

c) NAS scoring: NAS scores of 4 out of 7 animals in group 2 (treatment group) were reduced by 2 points or more, which was clinically significant. Only 1 in 5 out of 1 animals in group 1 (control) had 2 points decreased.

Dose administration: animals were weighed before administration of daily doses to calculate the actual dose volume. Subcutaneous injections were made on the back of the animals using an approximately 26 gauge needle. All animals of group 1 received subcutaneous administration of sterile saline once a week for 12 weeks. Animals of group 2 received subcutaneous administration of IMO-8400 solution once a week for 12 weeks.

Plasma preparation: whole blood (approximately 10mL) was collected from overnight fasted animals on day 1 (pre-dose), day 42 and morning on day 84. Samples were collected into commercial vacuum tubes containing EDTA-K2 (as an anticoagulant). The collected blood samples were placed on wet ice and centrifuged at 3000g for 15 minutes at about 4 ℃ within 30 minutes of collection to obtain plasma. Plasma supernatants were stored at-60 ℃ prior to analysis.

Adiponectin levels were measured by the human HMW adiponectin/Acrp 30 Quantikine ELISA kit (batch No. P172678, R & D Systems).

And (6) analyzing.

The NAS score was evaluated by standardized criteria as follows: brunt EM, Kleiner DE, Wilson LA, Belt P, Neuschwander-Tetri BA; NASH Clinical Research Network (CRN) [ NASH Clinical Research Network (CRN) ] Nonalcoholic fatty liver disease (NAFLD) activity score and the histopathological diagnosis in NAFLD [ non-alcoholic fatty liver disease (NAFLD) activity score and histopathological diagnosis in NAFLD: has obvious clinical pathology significance ] Hepatology 2011; 53: 810-; and Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, Ferrell LD, Liu YC, Torbenson MS, Unalp-Arida A, Yeh M, McCullough AJ, Sanyal AJ; nonalcoholic Steatohepatitis Clinical Research Network [ non-alcoholic Steatohepatitis Clinical Research net ] Design and evaluation of a histological scoring system for Nonalcoholic fatty liver disease [ Design and validation of histological scoring system for non-alcoholic fatty liver disease ] Hepatology [ Hepatology ] 2005; 41:1313-1321.

The following table provides a scoring framework:

the results shown in table 1 below show the adiponectin levels in the control group (group 1) and the treatment group (group 2).

Table 1:

ng/mL base line Week 6 Week 12 Middle of delta Delta final
Control-1 8200 6370 9902 -1831 1702
Control-2 12928 10706 13554 -2222 626
Control-3 5520 5905 21442 385 15922
Control-4 33107 29111 21050 -3996 -12057
Control-5 23280 15411 23740 -7869 460
Treatment-1 4781 8173 8973 3392 4192
Treatment-2 16041 19257 28451 3216 12410
Treatment-3 54142 47556 54806 -6586 664
Treatment-4 6219 12574 24074 6355 17856
Treatment-5 4393 5127 8762 734 4369
Treatment-6 7947 11662 14661 3715 6714
Treatment-7 971 1255 5107 283 4136

Human level: 2-38. mu.g/ml (depending on sex and BMI)

The results shown in table 2 below show the NAS scores of the control group (group 1) and the treatment group (group 2).

Table 2:

in table 2, ST ═ steatosis, BAL ═ balloon-like lesion, and INF ═ inflammation

The results of the weight loss over the 12-week test period for the control group (group 1) and the treatment group (group 2) are shown in fig. 1.

23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:食管狭窄抑制剂

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!