Compositions and methods for treating disorders ameliorated by muscarinic receptor activation

文档序号:638821 发布日期:2021-05-11 浏览:15次 中文

阅读说明:本技术 用于治疗由毒蕈碱受体激活所缓解的障碍的组合物和方法 (Compositions and methods for treating disorders ameliorated by muscarinic receptor activation ) 是由 A·贝坦库特 B·罗兰德 R·蒂伯特 于 2019-09-27 设计创作,主要内容包括:本文提供了一种口服药物组合物,所述口服药物组合物包含多个具有核心的呫诺美林珠,所述核心包含呫诺美林或其盐;以及多个具有核心的曲司氯胺珠,所述核心包含曲司氯胺盐。(Provided herein is an oral pharmaceutical composition comprising a plurality of xanomeline beads having a core comprising xanomeline or a salt thereof; and a plurality of trospium beads having a core comprising a trospium salt.)

1. An oral pharmaceutical composition, comprising:

a plurality of xanomeline beads comprising xanomeline or a salt thereof; and

a plurality of trospium beads comprising a trospium salt.

2. The oral pharmaceutical composition of claim 1, wherein said plurality of xanomeline beads have a core comprising said xanomeline or a salt thereof.

3. The oral pharmaceutical composition of claim 1 or 2, wherein said plurality of trospium beads has a core comprising said trospium salt.

4. The oral pharmaceutical composition of any one of claims 1-3, wherein said xanomeline beads are from 0.425mm to 1.18mm in size.

5. The oral pharmaceutical composition of claim 4, wherein said xanomeline beads are from 0.6mm to 0.85mm in size.

6. The oral pharmaceutical composition of any one of claims 1 to 5, wherein said trospium bead is 0.425mm to 1.18mm in size.

7. The oral pharmaceutical composition of claim 6, wherein said trospium bead is from 0.6mm to 0.85mm in size.

8. The oral pharmaceutical composition of any one of claims 1 to 7, wherein said xanomeline beads contain about 2.5 times as much xanomeline as said trospium salt of said trospium beads.

9. The oral pharmaceutical composition of any one of claims 1 to 8, having a dissolution rate of said plurality of xanomeline beads and said plurality of trospium beads of greater than about 95% within about the first 45 minutes after the dosage form enters an aqueous solution.

10. The oral pharmaceutical composition of claim 9, having a dissolution rate greater than about 95% within about the first 20 minutes after the dosage form enters an aqueous solution.

11. The oral pharmaceutical composition of any one of claims 1 to 10, which when administered to a patient twice daily at 20mg of trospium for at least 7 days provides a mean C of trospiumMaximum of7850. + -. 3360 pg/mL.

12. The oral pharmaceutical composition of any one of claims 1 to 11, which when administered to a patient twice daily at 20mg of trospium for at least 7 days, provides a mean AUC0-1241900. + -. 15500 hr. pg/mL.

13. The oral pharmaceutical composition of any one of claims 1-12, wherein said xanomeline is xanomeline tartrate.

14. The oral pharmaceutical composition of claim 13, wherein said xanomeline bead comprises 30 to 80 wt.% xanomeline tartrate.

15. The oral pharmaceutical composition of claim 14, wherein said xanomeline bead comprises 66 wt.% xanomeline tartrate.

16. The oral pharmaceutical composition of any one of claims 1-15, wherein said xanomeline beads comprise 15 wt.% to 65 wt.% microcrystalline cellulose.

17. The oral pharmaceutical composition of claim 14, wherein said xanomeline beads comprise 33.5 wt.% microcrystalline cellulose.

18. The oral pharmaceutical composition of any one of claims 1-17, wherein said xanomeline bead comprises 0 wt.% to 2 wt.% talc.

19. The oral pharmaceutical composition of claim 18, wherein said xanomeline bead comprises 0.5 wt.% talc.

20. The oral pharmaceutical composition of any one of claims 1-12, wherein said xanomeline bead comprises 30 to 80 wt.% xanomeline tartrate, 15 to 65 wt.% microcrystalline cellulose, and 0 to 2 wt.% talc.

21. The oral pharmaceutical composition of claim 20, wherein said xanomeline bead comprises 66 wt.% xanomeline tartrate, 33.5 wt.% microcrystalline cellulose, and 0.5 wt.% talc.

22. The oral pharmaceutical composition of any one of claims 1 to 21, wherein said trospium salt is chlorinated trospium.

23. The oral pharmaceutical composition of claim 22, wherein said trospium bead comprises 8 to 35 wt.% chlorinated trospium.

24. The oral pharmaceutical composition of claim 23, wherein said trospium bead comprises 17.7 wt.% chlorinated trospium.

25. The oral pharmaceutical composition of any one of claims 1 to 24, wherein said trospium bead comprises 25 to 80 wt.% microcrystalline cellulose.

26. The oral pharmaceutical composition of claim 25, wherein said trospium bead comprises 46.8 wt.% microcrystalline cellulose.

27. The oral pharmaceutical composition of any one of claims 1 or 26, wherein said trospium bead comprises from 15 wt.% to 70 wt.% lactose monohydrate.

28. The oral pharmaceutical composition of claim 27, wherein said trospium bead comprises 35 wt.% lactose monohydrate.

29. The oral pharmaceutical composition of any one of claims 1 to 28, wherein said trospium bead comprises 0 to 2 wt.% talc.

30. The oral pharmaceutical composition of claim 29, wherein said trospium bead comprises 0.5 wt.% talc.

31. The oral pharmaceutical composition of claims 1 to 21, wherein said trospium bead comprises 8 to 35 wt.% chlorinated trospium, 25 to 80 wt.% microcrystalline cellulose, 15 to 70 wt.% lactose monohydrate, and 0 to 2 wt.% talc.

32. The oral pharmaceutical composition of claim 31, wherein said trospium bead comprises 17.7 wt.% chlorinated trospium, 46.8 wt.% microcrystalline cellulose, 35 wt.% lactose monohydrate, and 0.5 wt.% talc.

33. The oral pharmaceutical composition of any one of claims 1-32, further comprising a capsule comprising said plurality of xanomeline beads and said plurality of trospium beads.

34. An oral pharmaceutical composition, comprising:

a plurality of xanomeline beads having a size of 0.425mm to 1.18mm, and a core comprising 30 wt.% to 80 wt.% xanomeline tartrate, 15 wt.% to 65 wt.% microcrystalline cellulose, and 0 wt.% to 2 wt.% talc; and

a plurality of trospium beads having a size of 0.425mm to 1.18mm, and a core comprising 8 wt.% to 35 wt.% chlorinated trospium, 25 wt.% to 80 wt.% microcrystalline cellulose, 15 wt.% to 70 wt.% lactose monohydrate, and 0 wt.% to 2 wt.% talc;

a dissolution rate of the plurality of xanomeline beads and the plurality of trospimine beads within about the first 45 minutes after the dosage form enters the aqueous solution of greater than about 95%; and wherein when administered to a patient twice daily at 20mg of trospium for at least 7 days, provides a mean C of trospiumMaximum of7850. + -. 3360pg/mL, mean AUC0-1241900. + -. 15500 hr. pg/mL.

35. The oral pharmaceutical composition of claim 34, wherein said xanomeline beads are from 0.6mm to 0.85mm in size.

36. The oral pharmaceutical composition of claim 34 or 35, wherein said trospium bead is 0.6mm to 0.85mm in size.

37. The oral pharmaceutical composition of any one of claims 34 to 36, wherein said xanomeline beads contain about 2.5 times as much xanomeline as said trospium beads contain chlorinated trospium.

38. The oral pharmaceutical composition of any one of claims 34 to 37, having a dissolution rate of said xanomeline and said trospium of greater than about 95% within about the first 20 minutes after entry of the dosage form into an aqueous solution.

39. The oral pharmaceutical composition of any one of claims 34-38, wherein said xanomeline bead comprises 66 wt.% xanomeline tartrate, 33.5 wt.% microcrystalline cellulose, and 0.5 wt.% talc.

40. The oral pharmaceutical composition of any one of claims 34 to 39, wherein said trospium beads comprise 17.7 wt.% chlorinated trospium, 46.8 wt.% microcrystalline cellulose, 35 wt.% lactose monohydrate, and 0.5 wt.% talc.

41. The oral pharmaceutical composition of any one of claims 34 to 40, further comprising a capsule comprising said plurality of xanomeline beads and said plurality of trospium beads.

42. An oral pharmaceutical composition, comprising:

a capsule comprising a plurality of xanomeline beads and a plurality of trospimine beads;

the plurality of xanomeline beads have a size of 0.6mm to 0.85mm, and a core comprising 66 wt.% xanomeline tartrate, 33.5 wt.% microcrystalline cellulose, and 0.5 wt.% talc; and is

The plurality of trospium beads have a size of 0.6mm to 0.85mm, and a core comprising 17.7 wt.% chlorinated trospium, 46.8 wt.% microcrystalline cellulose, 35 wt.% lactose monohydrate, and 0.5 wt.% talc;

a dissolution rate of the plurality of xanomeline beads and the plurality of trospimine beads within about the first 20 minutes after the dosage form enters the aqueous solution of greater than about 95%; and is

Wherein when administered to a patient twice daily at 20mg of trospium for at least 7 days, provides a mean C of trospiumMaximum of7850. + -. 3360pg/mL, mean AUC0-1241900. + -. 15500 hr. pg/mL.

43. The oral pharmaceutical composition of any one of claims 1-42, wherein said capsule has a dosage intensity of 25mg xanomeline free base and 10mg trospium chloride.

44. The oral pharmaceutical composition of any one of claims 1-42, wherein said capsule has a dosage intensity of 50mg of xanomeline free base and 20mg of chlorinated trospium.

45. The oral pharmaceutical composition of claims 1-42, wherein said capsule has a dosage intensity of 50mg of xanomeline free base and 10mg of chlorinated trospium chloride.

46. The oral pharmaceutical composition of claims 1-42, wherein said capsule has a dosage intensity of 75mg of xanomeline free base and 10mg of chlorinated trospium chloride.

47. The oral pharmaceutical composition of claims 1-42, wherein said capsule has a dosage intensity of 75mg of xanomeline free base and 20mg of chlorinated trospium chloride.

48. The oral pharmaceutical composition of claims 1-42, wherein said capsule has a dosage intensity of 125mg xanomeline free base and 20mg trospium chloride.

49. The oral pharmaceutical composition of claims 1-42, wherein said capsule has a dosage intensity of 125mg xanomeline free base and 30mg trospium chloride.

50. The oral pharmaceutical composition of claims 1-42, wherein said capsule has a dosage intensity of 125mg xanomeline free base and 40mg trospium chloride.

51. The oral pharmaceutical composition of any of the preceding claims, wherein said xanomeline bead comprises less than 0.5 wt.% of 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium.

52. An oral pharmaceutical composition comprising xanomeline and/or a salt thereof and chlorinated trospium for treating a muscarinic disorder in a patient in need thereof, wherein when administered to said patient in need thereof is sufficient to provide an in vivo plasma profile comprising a 2 hour median xanomeline TmaxMaximum ofAnd 1 hour median trospium TMaximum of

53. The method of claim 52The oral pharmaceutical composition of claim, wherein the in vivo plasma profile further comprises a mean dose normalized Cmax of 48.5pg/mL/mg to 121.3pg/mL/mgMaximum ofAnd a mean dose normalized Cmax of trospimine from 156pg/mL/mg to 375pg/mL/mgMaximum of

54. The oral pharmaceutical composition of claim 52 or 53, wherein the in vivo plasma profile further comprises a mean dose normalized AUC for xanomeline of 263 hr-pg/mL/mg to 577 hr-pg/mL/mg0-12And a mean dose normalized AUC of trospium of 881 hr-pg/mL/mg to 2024 hr-pg/mL/mg0-12

55. A method of activating muscarinic receptors in a biological sample, which comprises contacting said biological sample with an oral pharmaceutical composition according to any one of claims 1 to 55.

56. A method of treating a disorder ameliorated by activation of a muscarinic receptor in a subject in need thereof, which comprises administering to a patient in need thereof an oral pharmaceutical composition of any one of claims 1 to 55.

57. A method of treating a disorder ameliorated by activation of a muscarinic receptor in a subject in need thereof, which comprises sequential or co-administration of an oral pharmaceutical composition according to any one of claims 1 to 55; and a second therapeutic agent.

58. The method of any one of claims 55 to 57, wherein the subject is a human.

59. The method of any one of claims 55-57, wherein the disorder is selected from schizophrenia, Alzheimer's disease, Parkinson's disease, depression, movement disorders, pain, drug addiction, tauopathy, and synucleinopathy.

60. The method of any one of claims 55-57, wherein the disorder is a neurodegenerative disease.

61. The method of any one of claims 55-57, wherein the disorder is a central nervous system disease.

62. The compound 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium.

63. An oral pharmaceutical composition comprising xanomeline and/or a salt thereof and less than 0.5 wt.% of 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium.

64. A method of making the oral pharmaceutical composition of any one of claims 1 to 55, said method comprising combining a strain comprising a plurality of xanomeline beads comprising xanomeline or a pharmaceutically acceptable salt thereof with a plurality of trospium beads comprising a trospium salt.

65. The method of claim 64 wherein said plurality of beads containing xanomeline or a pharmaceutically acceptable salt thereof comprise an antioxidant.

66. The method of claim 64 or 65, further comprising: the mixed beads were formulated into capsules.

67. The method of any one of claims 64 to 66, further comprising storing the oral pharmaceutical composition at a temperature of about 2 ℃ to about 8 ℃ prior to dispensing the oral pharmaceutical composition to the subject.

68. The method of claim 67, wherein after dispensing the oral pharmaceutical composition to the subject, the method further comprises storing the oral pharmaceutical composition at a temperature of about 20 ℃ to about 25 ℃.

Drawings

The present disclosure may be understood more readily by reference to the following detailed description taken in conjunction with the accompanying drawings, in which like reference numerals identify like structural elements. The drawings provide exemplary embodiments or aspects of the disclosure and do not limit the scope of the disclosure.

Figure 1 shows a stability schedule and protocol for the xanomeline/trospium capsules.

Fig. 2 is a Scanning Electron Microscope (SEM) image of 66% beads of xanomeline tartrate at 30x magnification showing bead sizes for xanomeline/trospium capsules ranging from 0.6mm to 0.85 mm.

Fig. 3 is an SEM image of chlorinated trospium chloride 17.7% beads at 30x magnification showing bead sizes for xanomeline/trospium capsules ranging from 0.6mm to 0.85 mm.

Figure 4 is a dissolution profile of 50/20mg capsules of xanomeline/trospium Cl, containing xanomeline beads and trospium Cl beads, measured at time 0, 1 month, 2 months, 3 months, and 6 months after storage at 40 ℃/75% RH and after storage at 25 ℃/60% RH for 3 months.

Figure 5 is a dissolution profile of 50/10mg capsules of xanomeline/trospium Cl, containing xanomeline beads and trospium Cl beads, measured at 0, 1,2, and 3 months after storage at 40 ℃/75% RH and at time after storage at 25 ℃/60% RH for 3 months.

Figure 6 shows stability data for 50/10mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, 6 months, and 9 months.

Figure 7 shows stability data for 50/10mg capsules of xanomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0, 3 months, and 6 months.

Figure 8 shows stability data for 50/10mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

Figure 9 is a dissolution of 50/10mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, 6 months, and 9 months.

FIG. 10 is a dissolution profile of 50/10mg capsules of xanomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0, 3 months, and 6 months.

FIG. 11 is a dissolution profile of 50/10mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

Figure 12 is a mass spectrum of materials associated with the xanomeline active pharmaceutical ingredient measured at times of 0, 3 months, 6 months, and 9 months for 50/10mg capsules of xanomeline/trospium chloride Cl.

Figure 13 is a mass spectrum of 50/10mg capsules of xanomeline/trospium Cl associated with chlorinated trospium active pharmaceutical ingredient measured over time periods of 0, 3, 6 and 9 months.

FIG. 14 is a detail of 50/10mg capsules of xanomeline/trospium Cl.

Figure 15 shows stability data for 50/20mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, and 6 months.

Figure 16 shows stability data for 50/20mg capsules of xanomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0 and 6 months.

Figure 17 shows stability data for 50/20mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

FIG. 18 is a dissolution of 50/20mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3, 6, and 9 months.

FIG. 19 is a dissolution profile of 50/20mg capsules of xanthomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0 and 6 months.

FIG. 20 is a dissolution profile of 50/20mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

Figure 21 is a mass spectrum of 50/20mg capsules of xanomeline/trospium Cl associated with the xanomeline active pharmaceutical ingredient measured at times of 0, 3 months, and 6 months.

Figure 22 is a mass spectrum associated with chlorinated trospium active pharmaceutical ingredient measured at 0, 3 and 6 months times for 50/20mg capsules of xanomeline/trospium Cl.

FIG. 23 is a detail of 50/20mg capsules of xanomeline/trospium Cl.

Figure 24 shows stability data for 75/10mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, and 6 months.

Figure 25 shows stability data for 75/10mg capsules of xanomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0 and 6 months.

Figure 26 shows stability data for 75/10mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

FIG. 27 is a dissolution of 75/10mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, and 6 months.

FIG. 28 is a dissolution profile of 75/10mg capsules of xanthomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0 and 6 months.

FIG. 29 is a dissolution profile of 75/10mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

Figure 30 is a mass spectrum of 75/10mg capsules of xanomeline/trospium Cl associated with the xanomeline active pharmaceutical ingredient measured at times of 0, 3 months, and 6 months.

Figure 31 is a mass spectrum associated with chlorinated trospium active pharmaceutical ingredient measured at 0, 3 and 6 months times for 75/10mg capsules of xanomeline/trospium Cl.

FIG. 32 is a detail of 75/10mg capsules of xanomeline/trospium Cl.

FIG. 33 is a dissolution of 75/20mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, and 6 months.

FIG. 34 is a dissolution of 75/20mg capsules of xanthomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0 and 6 months.

Figure 35 shows stability data for 75/20mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

FIG. 36 is a dissolution of 75/20mg capsules of xanomeline/trospium Cl stored at 25 ℃/60% RH and measured at times of 0, 3 months, and 6 months.

FIG. 37 is a dissolution profile of 75/20mg capsules of xanthomeline/trospium Cl stored at 30 ℃/65% RH and measured at times of 0 and 6 months.

FIG. 38 is a dissolution profile of 75/20mg capsules of xanomeline/trospium Cl stored at 40 ℃/75% RH and measured at times of 0, 3 months, and 6 months.

FIG. 39 is a mass spectrum of 75/20mg capsules of xanomeline/trospium Cl correlated with xanomeline active pharmaceutical ingredient measured at times of 0, 3 months, and 6 months.

Figure 40 is a mass spectrum associated with chlorinated trospium active pharmaceutical ingredient measured at 0, 3 and 6 months times for 75/20mg capsules of xanomeline/trospium Cl.

FIG. 41 is a detail of 75/20mg capsules of xanomeline/trospium Cl.

Figure 42 depicts the mean (± standard deviation) xanomeline pharmacokinetic concentrations on day 1 for two treatments of KarXT 50/20 per day for a group of all KAR-003 pharmacokinetic populations.

Figure 43 depicts the mean (± standard deviation) xanomeline pharmacokinetic concentrations on day 3 for all groups of KAR-003 pharmacokinetic populations treated by KarXT 50/20 twice daily.

Figure 44 depicts the mean (± standard deviation) xanomeline pharmacokinetic concentrations on day 7 for the group of all KAR-003 pharmacokinetic populations, by treatment with KarXT 50/20 twice daily.

Figure 45 depicts the mean (± standard deviation) xanomeline pharmacokinetic concentrations through treatment and follow-up for the KAR-003 pharmacokinetic population.

FIG. 46 depicts mean (. + -. standard deviation) xanomeline pharmacokinetic trough concentrations by treatment for the KAR-003 pharmacokinetic population.

Figure 47 depicts the mean (± standard deviation) trospimine pharmacokinetic concentrations on day 1 for both KAR 50/20 treatments per day for the group of all KAR-003 pharmacokinetic populations.

Figure 48 depicts the mean (± standard deviation) trospimine pharmacokinetic concentrations on day 3 by treatment for the KAR-003 pharmacokinetic population.

Figure 49 depicts the mean (± standard deviation) trospimine pharmacokinetic concentrations at day 7 by treatment for the KAR-003 pharmacokinetic population.

Figure 50 depicts the mean (± standard deviation) trospimine pharmacokinetic concentrations through treatment and follow-up for the KAR-003 pharmacokinetic population.

Figure 51 depicts mean (± standard deviation) trospimine pharmacokinetic trough concentrations through treatment and follow-up for KAR-003 pharmacokinetic populations.

Detailed Description

The articles "a" and "an" refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.

The terms "comprising" and "comprising" are inclusive and open-ended and mean that additional elements may be included.

The term "consisting of … …" limits the elements to those specified, except for impurities normally associated therewith.

The term "consisting essentially of … …" is intended to limit the elements to those specified, as well as those elements that do not materially affect the basic and novel characteristics of the material or step.

All ranges set forth herein include all possible subsets of ranges and any combination of ranges of such subsets. By default, ranges include the endpoints specified, and unless otherwise specified, each intervening value, to the extent that a value is provided, between the upper and lower limit of that range and any other stated or intervening value in that range, is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also considered part of the disclosure.

The term wt.% is the weight percentage of the core, enteric coating or total beads, for example as described above and below, based on the total weight. Unless otherwise indicated, wt.% is intended to describe weight percent on a dry basis (e.g., for the core after drying).

The term "controlled release" is defined as an extended release pattern of one or more drugs such that the drug is released over a period of time. The release kinetics of controlled release formulations may result in serum levels of the drug being measurable over a longer period of time than is possible after intravenous injection or after administration of an immediate release oral dosage form. Controlled release, slow release, sustained release, extended release and delayed release are defined herein as the same.

The term "including" means "including but not limited to". "include" and "include but are not limited to" are used interchangeably.

The term "mammal" is known in the art. Exemplary mammals include humans, primates, cows, pigs, dogs, cats, and rodents (e.g., mice and rats).

The terms "parenteral administration" and "parenterally administered" are art-recognized and refer to modes of administration other than enteral and topical administration, typically by injection. These modes include, but are not limited to, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.

A "patient," "subject," or "host" treated by the subject methods refers to a human or non-human mammal.

The term "pharmaceutically acceptable carrier" is art-recognized and refers to a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material, involved in carrying or transporting any subject composition or component thereof from one organ or portion of the body to another organ or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the subject composition and its components and not injurious to the patient. Some examples of materials that can be used as pharmaceutically acceptable carriers include sugars such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc powder; excipients, such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; ringer's solution; ethanol; a phosphate buffer solution; and other non-toxic compatible materials used in pharmaceutical formulations.

The term "pharmaceutically acceptable salts" is art-recognized and refers to salts prepared from relatively nontoxic acids or bases, including inorganic acids and bases as well as organic acids and bases, including, for example, those contained in the compositions of the present disclosure. Suitable non-toxic acids include inorganic and organic acids such as acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, itaconic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, hydrochloric, hydrobromic, phosphoric, and sulfuric acids, and the like.

The term "treating" is art-recognized and refers to curing and ameliorating at least one symptom of any condition or disorder.

In the jurisdictions where patenting methods for administration to humans is prohibited, the meaning of "administering" a composition to a human subject should be limited to prescribing a controlled substance that the human subject will self-administer by any technique (e.g., oral, inhalation, topical application, injection, insertion, etc.). Is intended to conform to the broadest reasonable interpretation of the law or law that defines patentable subject matter. In the jurisdictions where patenting of methods for administration to humans is not prohibited, the "administration" of a composition includes both the methods for administration to humans and the aforementioned activities.

The term "therapeutic agent" is art-recognized and refers to any chemical moiety that is a biologically, physiologically, or pharmacologically active substance that acts locally or systemically in a subject. Examples of therapeutic agents (also referred to as "drugs") are described in well-known literature references (e.g., Merck Index (Merck Index) (14 th edition), The Physicians' Desk Reference (64 th edition), and The Pharmacological Basis of Therapeutics (12 th edition)). Such therapeutic agents include, but are not limited to, drugs; a vitamin; a mineral supplement; substances for treating, preventing, diagnosing, curing or alleviating a disease or condition; substances that affect body structure or function, or prodrugs, become biologically active or more active after being placed in a physiological environment.

The term "psychotherapy" refers to non-drug therapy, wherein one of skill in the art uses a variety of techniques (which involve verbal and other interactions with the patient) to achieve positive therapeutic results. Such techniques include, but are not limited to, behavioral therapy, cognitive therapy, psychodynamic therapy, psychoanalytic therapy, group therapy, family counseling, art therapy, musical therapy, occupational therapy, humanistic therapy, presence therapy, super-personal therapy, customer-centric therapy (also known as human-centric therapy), lattice therapy (getaltherapy), biofeedback therapy, rational emotional behavior therapy, reality therapy, response-based therapy, sand-play therapy, state-dynamics therapy, hypnosis, and confirmatory therapy. Psychotherapy may involve a combination of two or more techniques. The therapist can select and adjust the technique according to the needs of each patient and the patient's response.

The term "muscarinic disorder" refers to any disease or condition which is ameliorated by the activation of the muscarinic system. Such diseases include diseases in which direct activation of muscarinic receptors themselves or inhibition of cholinesterase has produced a therapeutic effect.

The terms "schizophrenia-related disease" and "schizophrenia-related disorder" include, but are not limited to, schizoaffective disorder, psychosis, delusional disorder, psychosis associated with alzheimer's disease, psychosis associated with parkinson's disease, psychotic depression, bipolar disorder, psychotic bipolar disorder, huntington's disease, lewy body dementia, or any other disease with psychotic features.

The term "movement disorder" includes, but is not limited to, Gilles de la Tourette syndrome, Friedrich's ataxia, Huntington's chorea, restless legs syndrome and other diseases or disorders, the symptoms of which include excessive movement, itching and cramping.

The term "mood disorder" includes major depressive disorder, dysthymia, recurrent transient depression, minor depressive disorder, bipolar disorder, mania and anxiety.

The term "cognitive disorder" refers to a disease or disorder characterized by cognitive deficits (e.g., having abnormal working memory, ability to solve problems, etc.). Diseases include, but are not limited to, alzheimer's Disease, parkinson's Disease, dementia (including, but not limited to, AIDS-related dementia, vascular dementia, age-related dementia, dementia associated with lewy bodies, and idiopathic dementia), pick's Disease, tauopathies, synucleinopathies, confusion, cognitive deficits associated with fatigue, learning disorders, brain trauma, autism, age-related cognitive decline, and Cushing's Disease (cognitive disorders associated with autoimmune diseases).

The term "attention disorder" refers to a disease or condition characterized by an abnormal or reduced duration of attention. Diseases include, but are not limited to, Attention Deficit and Hyperactivity Disorder (ADHD), Attention Deficit Disorder (ADD), Dubowitz Syndrome (Dubowitz Syndrome), FG Syndrome, Down Syndrome, growth delay due to insulin-like growth factor I (IGF1) deficiency, hepatic encephalopathy Syndrome, and Strauss Syndrome (Strauss Syndrome).

The term "addictive disorder" refers to a disease or condition characterized by an addiction or drug dependence as defined in Diagnostic & Statistical Manual V (DSM-5). Such disorders are characterized by physical dependence on substances, withdrawal and tolerance. These substances include, but are not limited to, alcohol, cocaine, amphetamines, opioids, benzodiazepines, inhalants, nicotine, barbiturates, cocaine, and cannabis. Addictive disorders also include behaviors that the patient is forced or sustained despite significant negative consequences. For example, gambling addiction (gambling addiction or compulsive gambling) is recognized by those skilled in the art as an addictive behaviour which often has devastating consequences. In certain embodiments, the addictive behavior may be an internet gaming disorder (game addiction) as defined in DSM-5.

The term "pain" refers to physical distress or discomfort caused by a disease or injury. Pain is a subjective experience, and the perception of pain is part of the Central Nervous System (CNS). Usually, noxious (peripheral) stimuli are delivered to the CNS in advance, but pain is not always associated with nociception. There is a wide variety of clinical pain that results from different underlying pathophysiological mechanisms and requires different treatments. Three main types of clinical pain have been characterized: acute pain, chronic pain, and neuropathic pain.

Acute clinical pain may be due to inflammation or soft tissue injury. This type of pain is adaptive and has a biologically relevant alarm function and leaves the healing and repair of already damaged body parts undisturbed. The protective function is achieved by making the injured or inflamed area and surrounding tissue highly sensitive to all stimuli, so that contact with any external stimuli can be avoided. The neuronal mechanisms of such clinical pain are well known and pharmacological control of acute clinical pain is effective, for example via non-steroidal anti-inflammatory drugs (NSAIDs) through opioids, depending on the type and degree of pain sensation.

Chronic clinical pain manifests itself as persistent paresthesia that is caused by persistent peripheral lesions such as cancer or chronic inflammation (e.g. arthritis), or it may be independent of such triggering factors. Chronic pain, independent of the causative factor, is maladaptive, has no survival advantage, and generally has no effective treatment.

Neuropathic pain can be classified as peripheral or central. Peripheral neuropathic pain is caused by injury or infection of peripheral sensory nerves, while central neuropathic pain is caused by injury of the CNS or/and spinal cord. Both peripheral and central neuropathic pain can occur without significant initial nerve damage.

The term "activator" refers to a molecule described as an agonist, partial agonist, co-agonist, physiological agonist, potentiator, stimulant, allosteric potentiator, positive allosteric modulator, allosteric agonist, or a molecule that directly or indirectly increases receptor activity or signaling.

The term "inhibitor" refers to a molecule that is described as an antagonist, partial antagonist, competitive antagonist, noncompetitive antagonist, silent antagonist, inverse agonist, reversible antagonist, physiological antagonist, irreversible antagonist, inhibitor, reversible inhibitor, irreversible inhibitor, negative allosteric modulator, allosteric antagonist, or a molecule that directly or indirectly reduces receptor activity or signaling.

The term "maximum tolerated dose" refers to the maximum dose of a drug or therapeutic agent that a patient can take without the patient experiencing intolerable side effects. The maximum tolerated dose is usually determined empirically in clinical trials.

The term "muscarinic receptor" refers to a G protein-linked receptor that binds the neurotransmitter acetylcholine. To date, five subtypes of muscarinic receptors have been identified. "M1" refers to the subtype-muscarinic receptor. "M2" refers to the subtype two muscarinic receptors. "M3" refers to the subtype trimuscarinic receptor. "M4" refers to the subtype four muscarinic receptors. "M5" refers to the subtype pentamuscarinic receptor.

The term "antipsychotic" refers to a drug that reduces psychosis, hallucinations, or delusions. Antipsychotic agents include, but are not limited to, haloperidol, chlorpromazine fluphenazine, perphenazine, prochlorperazine, thioridazine, trifluoperazine, mesoridazine, prazosin, promazine, levopromazine, promethazine, pimazine, chlorprothixene, flupentixol, thiothixene, zulothiol, clozapine, olanzapine, risperidone, quetiapine, ziprasidone, amisulpride, asenapine, paliperidone, zotepine, aripiprazole, bifeprunox, and tetrabenazine.

The term "anxiolytic" refers to a drug that reduces anxiety, fear, panic, or related sensations. Such drugs include, but are not limited to, benzodiazepines (e.g., alprazolam, methotrexate, clonazepam, lorazepam, diazepam, lorazepam), buspirone, barbiturates (e.g., amobarbital, pentobarbital, secobarbital, phenobarbital), and hydroxyzine.

The term "antidepressant" refers to a drug that alleviates depression and related conditions (e.g., mood disorders). Such agents include, but are not limited to, selective serotonin reuptake inhibitors (SSRIs, e.g., citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline), serotonin norepinephrine reuptake inhibitors (SNRIs, e.g., desvenlafaxine, duloxetine, milnacipran (milnacipram), venlafaxine, mianserin, mirtazapine, norepinephrine reuptake inhibitors (e.g., atomoxetine, mazindol, reboxetine, viloxazine, bupropion, tianeptine, agomelatine, tricyclic antidepressants (e.g., amitriptyline, clomipramine, doxepin, imipramine, trimipramine, desipramine, nortriptyline, protriptyline), and monoamine oxidase inhibitors (e.g., isocarboxazid, moclobemide, phenelzine, selegiline, prednisolone).

The term "sedative" or "tranquilizer" refers to a drug which induces somnolence, promotes fatigue or is craving to sleep or promotes an unconscious state. Such agents include, but are not limited to, benzodiazepines, barbiturates (e.g., amobarbital, pentobarbital, secobarbital, phenobarbital), dexzopiclone, zaleplon, zolpidem, and zopiclone.

Pharmaceutical composition

Early development of the muscarinic receptor agonist xanomeline as a monotherapy was terminated due to peripheral cholinergic side effects. The present disclosure provides a dosage form with dissolution kinetics that has more efficacious therapeutic effects on both active ingredients, enhanced pharmacokinetics for chlorinated trospium, and greater dose compliance. The present disclosure also provides dosage forms having different strengths and/or different ratios of the two active ingredients.

Provided herein is an oral pharmaceutical composition comprising a plurality of xanomeline beads comprising xanomeline or a salt thereof; and a plurality of trospimine beads comprising a trospimine salt. In certain embodiments, the plurality of xanomeline beads have a core comprising xanomeline or a salt thereof. In certain embodiments, the plurality of trospium beads has a core comprising a trospium salt.

In certain embodiments, a capsule shell comprising Hydroxypropylmethylcellulose (HPMC) contains separate populations of beads comprising xanomeline tartrate or trospium chloride, wherein the beads are of comparable size and rapidly release the active ingredient at a substantially similar rate. After dissolution of the capsule shell in the stomach, the beads may dissolve in the stomach and/or pass completely or partially completely through the pyloric valve into the duodenum, but the ratio of the two drugs, whether in dissolved or undissolved form, remains relatively constant in the gastrointestinal tract until the drug is absorbed.

The formulation of each bead allows substantially similar performance to be achieved from two active ingredients at different dosage ranges, wherein the active ingredients are released into the serum at substantially similar ratesAnd/or achieving substantially similar TMaximum of. In certain embodiments, a capsule comprising 50mg of xanomeline as the tartrate salt; and 10mg of trospium chloride. Because 50mg of xanomeline free base is equivalent to about 76mg of xanomeline tartrate, the ratio of active ingredients in this formulation is about 7.6 to 1.

The difference in the number of beads in the capsule increases the likelihood that the bead ratio will not remain substantially constant after the beads are released and dispersed. Thus, in certain embodiments, trospium beads are formulated at a lower drug load such that an effective dose of trospium and xanomeline are contained in approximately equal numbers of beads. In certain embodiments, the trospium and xanomeline beads are released at a substantially similar rate despite the differences in drug loading. For example, if the dissolution of the capsule is evaluated using a United States Pharmacopeia (USP) dissolution apparatus, the percentage of dissolved xanomeline is substantially equal to the percentage of dissolved chlorinated trospium, e.g., at 10 minutes, 20 minutes, or 30 minutes.

The medicament may also comprise one or more pharmaceutically acceptable salts. The medicament may comprise one or more pharmaceutically acceptable carriers. The medicament may be administered orally. The drug may be delivered orally using tablets, lozenges, liquids, emulsions, suspensions, drops, capsules, caplets or gel caps and other methods of oral administration known to those skilled in the art.

The drug may be in a dosage form that releases the drug immediately. In another embodiment, the drug may have a controlled release dosage form.

The drug may be in a dosage form using other controlled release formulation methods known to those skilled in the art.

In another embodiment, the drug is used in combination with one or more therapies (including psychotherapy and drugs). Therapeutic agents include, but are not limited to, antipsychotics, anxiolytics, antidepressants, sedatives, tranquilizers, analgesics, and other pharmacological interventions known to those of skill in the art. Therapeutic agents may belong to a class having more than one drug. For example, benzodiazepines can be considered anxiolytics, sedatives and tranquilizers.

Bead/core excipients

The beads and/or core may comprise one or more excipients. In one embodiment, the excipients include one or more fillers, binders, and surfactants. Other optional ingredients include, but are not limited to, glidants, lubricants, disintegrants, swelling agents, and antioxidants. The xanomeline or a pharmaceutically acceptable salt thereof and the trospium salt may be in different matrices within the same medicament.

The amount of xanomeline free base in the core may be at least 10 wt.%, or at least 15 wt.%, or at least 20 wt.%, or at least 25 wt.%, or at least 30 wt.%. For example, the amount of xanomeline tartrate can be at least 50 wt.%, or at least 55 wt.%, or at least 60 wt.%, or at least 65 wt.%, or at least 70 wt.%, or at least 75 wt.%, or at least 80 wt.%, or at least 85 wt.% of the core, e.g., in the range of from about 60 wt.% to about 90 wt.%, or from about 65 wt.% to about 85 wt.%. It is to be understood that all ranges including these values as endpoints are contemplated, for example, at least about 15 wt.% to about 90 wt.%, about 20 wt.% to about 85 wt.%, about 30 wt.% to about 85 wt.%, or about 50 wt.% to about 90 wt.%. In certain embodiments, the xanomeline bead comprises 30 wt.% to 80 wt.% xanomeline tartrate, e.g., 66 wt.% xanomeline tartrate.

The amount of trospium salt in the core may be at least 10 wt.%, or at least 15 wt.%, or at least 20 wt.%, or at least 25 wt.%, or at least 30 wt.%. For example, the amount of chlorinated trospium may be at least 50 wt.%, or at least 55 wt.%, or at least 60 wt.%, or at least 65 wt.%, or at least 70 wt.%, or at least 75 wt.%, or at least 80 wt.%, or at least 85 wt.% of the core, for example, in the range of about 60 wt.% to about 90 wt.%, or about 65 wt.% to about 85 wt.%. It is to be understood that all ranges including these values as endpoints are contemplated, for example, at least about 15 wt.% to about 90 wt.%, about 20 wt.% to about 85 wt.%, about 30 wt.% to about 85 wt.%, or about 50 wt.% to about 90 wt.%. In certain embodiments, the trospium is chlorinated trospium. In certain embodiments, the trospium bead comprises 8 to 35 wt.% chlorinated trospium, e.g., 17.7 wt.% chlorinated trospium.

In another embodiment, the matrix comprises a polymer, for example to modify the release profile of the active ingredient in the matrix. In another embodiment, the polymer comprises a water soluble polymer. In another embodiment, the water soluble polymer is selected from EudragitTMRL, polyvinyl alcohol, polyvinylpyrrolidone, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyethylene glycol, and mixtures thereof. In another embodiment, the polymer comprises a water insoluble polymer. In another embodiment, the water insoluble polymer is selected from EudragitTMRS, ethylcellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate), poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly (octadecyl acrylate), poly (ethylene), low density poly (ethylene), high density poly (ethylene), poly (propylene), poly (ethylene terephthalate), poly (vinyl isobutyl ether), poly (vinyl acetate), poly (vinyl chloride), Polyurethanes, and mixtures thereof.

Fillers include, but are not limited to, lactose, sucrose, glucose, starch, microcrystalline cellulose, superfine cellulose, mannitol, sorbitol, dibasic calcium phosphate, aluminum silicate, amorphous silicon dioxide, and sodium chloride, starch, and dibasic calcium phosphate dihydrate. In one embodiment, the filler, while absorbing water, is insoluble in water. In one embodiment, the filler is a spheronization aid. The spheronization aids may include one or more of crospovidone, carrageenan, chitosan, pectic acid, glycerides, beta-cyclodextrin (beta-CD), cellulose derivatives, microcrystalline cellulose, powdered cellulose, povidone crospovidone, and polyethylene oxide. In one embodiment, the filler comprises microcrystalline cellulose.

The amount of filler in the xanomeline core is not particularly limited. In embodiments, the amount of filler (e.g., microcrystalline cellulose) may be in the range of about 10 wt.% to about 70 wt.%, or about 16 wt.% to about 23 wt.%, or at least 19 wt.% or at least 19.5 wt.%, e.g., about 20 wt.%. In certain embodiments, the xanomeline beads comprise 15 to 65 wt.% microcrystalline cellulose, e.g., about 15 to 20 wt.%, about 20 to 25 wt.%, about 25 to 30 wt.%, about 30 to 35 wt.%, about 35 to 40 wt.%, about 40 to 45 wt.%, about 45 to 50 wt.%, about 50 to 55 wt.%, about 55 to 60 wt.%, or about 60 to 65 wt.%. In certain embodiments, the xanomeline beads comprise 33.5 wt.% microcrystalline cellulose.

The amount of filler in the trospium core is not particularly limited. In embodiments, the amount of filler (e.g., microcrystalline cellulose or lactose) may be in the range of about 10 wt.% to about 80 wt.%, or about 16 wt.% to about 23 wt.%, or at least 19 wt.% or at least 19.5 wt.%, e.g., about 20 wt.%. In certain embodiments, the trospium beads comprise 25 to 80 wt.% microcrystalline cellulose, e.g., about 25 to 30 wt.%, about 30 to 35 wt.%, about 35 to 40 wt.%, about 40 to 45 wt.%, about 45 to 50 wt.%, about 50 to 55 wt.%, about 55 to 60 wt.%, about 60 to 65 wt.%, about 65 to 70 wt.%, about 70 to 75 wt.%, or about 75 to 80 wt.%. In certain embodiments, trospium bead comprises 46.8 wt.% microcrystalline cellulose.

In certain embodiments, the trospium bead comprises 15 to 70 wt.% lactose monohydrate, e.g., about 15 to 20 wt.%, about 20 to 25 wt.%, about 25 to 30 wt.%, about 30 to 35 wt.%, about 35 to 40 wt.%, about 40 to 45 wt.%, about 45 to 50 wt.%, about 50 to 55 wt.%, about 55 to 60 wt.%, about 60 to 65 wt.%, or about 65 to 70 wt.%. In certain embodiments, trospium bead comprises 35 wt.% lactose monohydrate.

Binders include, but are not limited to, cellulose ethers, methyl cellulose, ethyl celluloseCellulose, hydroxyethyl cellulose, propyl cellulose, hydroxypropyl cellulose, low-substituted hydroxypropyl cellulose, hydroxypropyl methylcellulose (hypromellose, e.g. hypromellose 2910, Methocel)TME) Carboxymethylcellulose, starch, pregelatinized starch, gum arabic, gum tragacanth, gelatin, polyvinylpyrrolidone (povidone), crospovidone, sodium alginate, microcrystalline cellulose, and lower alkyl-substituted hydroxypropylcellulose. In one embodiment, the adhesive is selected from wet adhesives. In one embodiment, the binder is selected from cellulose ethers, such as hypromellose.

The amount of binder in the xanomeline core is not particularly limited. In embodiments, the amount of binder (e.g., hypromellose) may range from about 1 wt.% to about 10 wt.%, from about 2 wt.% to about 8 wt.%, or from about 4 wt.% to about 6 wt.%, e.g., about 5 wt.%.

The amount of binder in the trospium core is not particularly limited. In embodiments, the amount of binder (e.g., hypromellose) may range from about 1 wt.% to about 10 wt.%, from about 2 wt.% to about 8 wt.%, or from about 4 wt.% to about 6 wt.%, e.g., about 5 wt.%.

Surfactants include, but are not limited to, anionic surfactants including sodium lauryl sulfate, sodium deoxycholate, dioctyl sodium sulfosuccinate, and sodium stearyl fumarate, nonionic surfactants including polyoxyethylene ethers and polysorbate 80, and cationic surfactants including quaternary ammonium compounds. In one embodiment, the surfactant is selected from anionic surfactants, such as sodium lauryl sulfate.

The amount of surfactant in the xanomeline core, e.g., as a processing aid, is not particularly limited. In embodiments, the amount of surfactant (e.g., microcrystalline cellulose) may be in the range of about 0.1 wt.% to about 1 wt.%, about 0.2 wt.% to about 0.8 wt.%, or about 0.4 wt.% to about 0.6 wt.%, e.g., about 0.5 wt.%.

The amount of surfactant in the trospium core, e.g., as a processing aid, is not particularly limited. In embodiments, the amount of surfactant (e.g., sodium lauryl sulfate) may range from about 0.1 wt.% to about 1 wt.%, from about 0.2 wt.% to about 0.8 wt.%, or from about 0.4 wt.% to about 0.6 wt.%, e.g., about 0.5 wt.%.

Disintegrants include, but are not limited to, starch, croscarmellose sodium, carboxymethylcellulose calcium, crospovidone, and sodium starch glycolate, low substituted hydroxypropylcellulose, and hydroxypropylstarch.

Glidants include, but are not limited to, polyethylene glycol of various molecular weights, magnesium stearate, calcium silicate, fumed silica, magnesium carbonate, magnesium lauryl sulfate, aluminum stearate, stearic acid, palmitic acid, cetyl alcohol, stearyl alcohol and talc.

Lubricants include, but are not limited to, stearic acid, magnesium stearate, calcium stearate, aluminum stearate, and talc silicide. In certain embodiments, the xanomeline beads comprise 0 wt.% to 2 wt.% talc, e.g., 0.5 wt.% talc. In certain embodiments, trospium bead contains 0 to 2 wt.% talc, e.g., 0.5 wt.% talc.

In certain embodiments, the formulation further comprises one or more antioxidants. Examples of pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium hydrogensulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants such as ascorbyl palmitate, Butylated Hydroxyanisole (BHA), Butylated 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. In certain embodiments, the formulation comprises less than 1 wt.% antioxidant, e.g., 0.9 wt.%, 0.8 wt.%, 0.7 wt.%, 0.6 wt.%, 0.5 wt.%, 0.4 wt.%, 0.3 wt.%, 0.2 wt.%, 0.1 wt.%, 0.09 wt.%, 0.08 wt.%, 0.07 wt.%, 0.06 wt.%, 0.05 wt.%, 0.04 wt.%, 0.03 wt.%, 0.02 wt.%, or 0.01 wt.%. In certain embodiments, the formulation comprises about 0.05 wt.% BHT or 0.5 wt.% ascorbic acid. In certain embodiments, the antioxidant is present in the xanomeline core or the xanomeline bead.

In certain embodiments, the xanomeline beads comprise 30 to 80 wt.% xanomeline tartrate, 15 to 65 wt.% microcrystalline cellulose, and 0 to 2 wt.% talc. In certain embodiments, trospium bead contains 0.2 to 2 wt.% talc, e.g., 0.5 wt.% talc. In certain embodiments, the trospium bead comprises 8 to 35 wt.% chlorinated trospium, 25 to 80 wt.% microcrystalline cellulose, 15 to 70 wt.% lactose monohydrate, and 0.2 to 2 wt.% talc.

In certain embodiments, the xanthonomilin tartrate pharmaceutical beads comprise 66 wt.% of xanomeline tartrate, 33.5 wt.% microcrystalline cellulose, and 0.5 wt.% talc. In certain embodiments, the chlorinated trospium bead comprises 17.7 wt.% chlorinated trospium, 46.8 wt.% microcrystalline cellulose, 35 wt.% lactose monohydrate, and 0.5 wt.% talc. In this example, the tartaric acid xanomeline beads contain about 2.5 times the amount of chlorinated trospium contained by the chlorinated trospium beads.

Depending on the dosage requirements, capsules may be prepared with varying amounts of xanomeline tartrate beads and trospium chloride beads. In various embodiments, the capsule comprises 50mg of xanomeline and 10mg of chlorinated trospium, 50mg of xanomeline and 20mg of chlorinated trospium, 75mg of xanomeline and 10mg of chlorinated trospium, 75mg of xanomeline and 20mg of chlorinated trospium, 125mg of xanomeline and 30mg of chlorinated trospium, or 125mg of xanomeline and 40mg of chlorinated trospium. In certain embodiments, the capsule contains 25mg of xanomeline tartrate and 10mg of trospium chloride. In certain embodiments, the capsule contains 50mg of xanomeline tartrate and 10mg of trospium chloride. In certain embodiments, the capsule contains 50mg of xanomeline tartrate and 20mg of trospium chloride. In certain embodiments, the capsule contains 75mg of xanomeline tartrate and 10mg of trospium chloride. In certain embodiments, the capsule contains 75mg of xanomeline tartrate and 20mg of trospium chloride. In certain embodiments, the capsule contains 125mg of xanomeline tartrate and 20mg of trospium chloride. In certain embodiments, the capsule contains 125mg of xanomeline tartrate and 40mg of chlorinated trospium chloride.

In another embodiment, the medicament comprises 5mg to 700 mg of xanomeline. In one embodiment, the medicament comprises between 25mg and 300mg of xanomeline.

In another embodiment, the medicament comprises 1 mg to 400 mg of trospium chloride. In one embodiment, the medicament comprises 6.5 mg to 200mg of trospium chloride.

In one embodiment, the chlorinated trospium extended release agent is used as chlorinated trospium in a medicament. In another embodiment, the medicament comprises 1 mg to 400 mg of the tramadol ammonium chloride extended release agent. In one embodiment, the medicament comprises an extended release agent of trospium chloride in an amount from 6.5 mg to 200 mg.

In one embodiment, the medicament comprises 75mg or 225mg of xanomeline, and the same medicament comprises 20mg or 40mg of chlorinated trospium. In another embodiment, the drug comprises 75mg or 225mg of xanomeline, and the different drug to be co-administered comprises 20mg or 40mg of chlorinated trospium.

Pearl coating

In other embodiments, the beads may be coated with a functional or non-functional coating for aesthetic, handling, or stability. In certain embodiments, the beads may be coated with a pH sensitive coating so that they do not dissolve in the low pH of the stomach. Non-functional coatings may be used to maintain chemical separation between beads or for decorative reasons.

In another embodiment, the controlled release formulation comprises a semipermeable coating. The xanomeline and trospium in the same formulation may be in different coatings. In another embodiment, the xanomeline and the chlorinated trospium may be in different coatings in different formulations or dosing vehicles. In another embodiment, the semipermeable coating comprises a polymer. In another embodiment, the controlled release formulation comprises a matrix suspending xanomeline and chlorinated trospium.

In certain embodiments, the distribution of coating thickness may be stated in weight gain of the coating material based on the total weight of the coated beads. Thus, in one embodiment, the distribution of coating thickness is at least 2% based on the total weight of the coated beads. In another embodiment, the distribution of coating thickness is at least 3%. In another embodiment, the distribution of coating thickness is at least 4%. In another embodiment, the distribution of coating thickness is at least 5%. In another embodiment, the distribution of coating thickness is at least 6%. In another embodiment, the distribution of coating thickness is at least 7%. In another embodiment, the distribution of coating thickness is at least 8%. In another embodiment, the distribution of coating thickness is at least 9%. In another embodiment, the distribution of coating thickness is at least 10%. In another embodiment, the distribution of coating thickness is at least 11%. In another embodiment, the distribution of coating thickness is at least 12%. In another embodiment, the distribution of coating thickness is at least 13%. In another embodiment, the distribution of coating thickness is at least 14%.

For example, the coating thickness difference from bead to bead can be in the range of +/-1% -7% based on the total weight of the coated beads. The distribution of coating thickness may be from about 2% to about 14%, for example from about 3% to about 13%, from about 4% to about 12%, from about 5% to about 11%, from about 6% to about 10%, from about 7% to 9%, from about 3% to 14%, from about 4% to 13% or from 4% to about 12%, based on the weight of the coated bead.

In one embodiment, the absorption (area under the curve, AUC) of the dosage form is advantageously increased upon oral administration as compared to other dosage forms of xanomeline or chlorinated trospium. Without being bound by any theory, the increase in absorption is affected by dosage forms exhibiting a pseudo-extended release profile. The pseudo-extended release profile is influenced by one or more factors, including the distribution of coating thickness (when present), the distribution of beadlets, and beads having irregular bead shapes. For example, in embodiments in which the beads have a coating thickness profile, for beads having a relatively thin coating, the coating completely dissolves relatively quickly at the trigger pH to release the xanomeline and/or chlorinated trospium composition, and for beads having a relatively thin coating, the coating completely dissolves relatively quickly to release the xanomeline and/or chlorinated trospium compositionThick coated beads, the coating taking longer to completely dissolve and release the xanomeline and/or chlorinated trospium composition. In embodiments where the beads have a particle size distribution and/or an irregular bead shape, the intestinal transit time of the beads may vary due to bead size and/or shape such that the transit time until the coating dissolution pH is reached is varied, thus contributing to a pseudo-extended release profile. In another embodiment, the dosage form exhibits a substantially equivalent (e.g., bioequivalent) C when administered orally, with or without a capsule shellMaximum ofAnd/or AUC characteristics.

In certain embodiments, the dosage form provides a gradual and predictable absorption profile. In one embodiment, when administered orally, the dosage form TMaximum ofMore stable on a dose-to-dose basis because the beads are coated individually. Predictable, consistent TMaximum ofIs beneficial to realizing more consistent and continuous treatment effect. For example, process-related variations in coating thickness or other effects on coating dissolution affect only a portion of the dosage form of xanomeline and chlorinated trospium, and tend to result in pseudo-extended release behavior. In contrast, coated capsules containing xanomeline and microspheres of chlorinated trospium showed significant variability in absorption time between capsules.

In certain embodiments, an oral pharmaceutical composition comprising xanomeline and/or a salt thereof and chlorinated trospium for treating a muscarinic disorder in a patient in need thereof is sufficient to provide an in vivo plasma profile comprising a 2 hour median xanomeline T when administered to the patient in need thereofMaximum ofAnd 1 hour median trospium TMaximum of. In certain embodiments, the in vivo plasma profile further comprises a mean dose normalized Cmax of 48.5 to 121.3pg/mL/mgMaximum of. In certain embodiments, the in vivo plasma profile further comprises a mean dose normalized Cmax of trospimine from 156pg/mL/mg to 375pg/mL/mgMaximum of. In certain embodiments, the in vivo plasma profile further comprises a mean dose normalized AUC for xanomeline of 263 to 577 hr-pg/mL/mg0-12. In certain embodiments, in vivo plasma profilesFurther comprising a mean dose normalized AUC of trospium from 881 hr-pg/mL/mg to 2024 hr-pg/mL/mg0-12. In certain embodiments, the in vivo plasma profile further comprises a mean C of trospium at 7850 ± 3360pg/mLMaximum of. In certain embodiments, the in vivo plasma profile further comprises a mean AUC of 41900 ± 15500 hr-pg/mL0-12

In another embodiment, the dosage form exhibits advantageous storage stability, for example as measured by the amount of xanomeline present after storage and/or the total amount of related substances. Storage stability can be assessed under typical ambient conditions (e.g., 25 ℃ and 60% relative humidity) or after storage under accelerated stability conditions involving elevated temperature and/or humidity.

Unless otherwise indicated, it is contemplated that the dosage forms and methods include embodiments of any combination of one or more of the additional optional elements, features, and steps (including those shown in the figures and examples) described further below. References to beads and their properties apply equally to a collection of beads (e.g., a plurality of such beads). Likewise, references to a core and its properties apply equally to a set of cores (e.g., a plurality of such cores).

The enteric (gastro-resistant) coating material, e.g. a polymer, may be a polymer which is soluble in intestinal fluids at a pH level above that of the stomach, e.g. at a pH above 4.5 (e.g. in the small intestine), thereby allowing the active substance to be released in the region of the small intestine and not substantially in the upper part of the GI tract. In one embodiment, the enteric material begins to dissolve in an aqueous solution having a pH of about 4.5 to about 5.5. In another embodiment, the enteric material dissolves rapidly in an aqueous solution having a pH of about 5. In another embodiment, the enteric material dissolves rapidly in an aqueous solution having a pH of about 5.5.

For example, the pH sensitive material does not dissolve significantly until the dosage form is emptied from the stomach. The pH of the small intestine gradually increases from about 4.5 to about 6.5 in the duodenal bulb, and then to about 7.2 in the distal part of the small intestine (ileum). In order to provide predictable dissolution corresponding to a small intestinal transit time of about 3 hours (e.g., 2-3 hours) and allow for reproducible release therein, the coating should begin dissolution within the pH range of the duodenum and continue to dissolve within the pH range of the small intestine. Thus, the amount (thickness) of enteric coating should be sufficient to substantially dissolve in the small intestine (e.g., proximal and middle small intestine) within a transit time of about three hours.

Suitable enteric (gastro-resistant) materials include, but are not limited to, cross-linked polyvinylpyrrolidone; non-crosslinked polyvinylpyrrolidone; hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, cellulose acetate succinate; cellulose acetate phthalate, hydroxypropyl methyl cellulose acetate succinate, cellulose acetate trimellitate; starch acetate phthalate; polyvinyl acetate phthalate; a carboxymethyl cellulose; methyl cellulose phthalate; methyl cellulose succinate; methyl cellulose phthalate succinate; methyl cellulose phthalate half ester; ethyl cellulose succinate; a carboxymethyl amide; potassium methacrylate divinylbenzene copolymer; polyvinyl alcohol; polyoxyethylene glycol; polyethylene glycol; sodium alginate; galactomannan; a carboxypolymethylene group; sodium carboxymethyl starch; copolymers of acrylic acid and/or methacrylic acid with monomers selected from: methyl methacrylate, ethyl acrylate, butyl methacrylate, hexyl methacrylate, decyl methacrylate, lauryl methacrylate, phenyl methacrylate, methyl acrylate, isopropyl acrylate, isobutyl acrylate, or stearyl acrylate (e.g., Eudragit @)TMThe L and S series, including L100-55, L30D-55, L100, S100, L12.5, and S12.5, are available from Evonik Industries; polyvinyl acetate; fat; an oil; a wax; a fatty alcohol; shellac; a zein; gluten; ethyl acrylate-maleic anhydride copolymer; maleic anhydride-vinyl methyl ether copolymers; styrene-maleic acid copolymers; 2-ethyl-hexyl-acrylate maleic anhydride; crotonic acid-vinyl acetate copolymer; glutamic acid/glutamate copolymers; carboxymethylethylcellulose glyceryl monocaprylate; poly-arginine; poly (ethylene); poly (propylene); poly (ethylene oxide); poly (ethylene terephthalate); poly (vinyl isobutyl ether); poly (vinyl chloride); and polyurethanes. Can also useCombinations of enteric materials. In one embodiment, the enteric material dissolves rapidly at pH 5.5 and higher to provide rapid dissolution in the upper intestine. For example, the enteric material may be selected from copolymers of methacrylic acid and methyl methacrylate, and copolymers of methacrylic acid and ethyl acrylate. For example, the enteric polymer is poly (ethyl methacrylate-co-acrylate) 1:1 (Eudragit)TML30D-55 and EudragitTML 100-55)。

Other suitable examples of one or more enteric coatings include beeswax and glyceryl monostearate; beeswax, shellac and cellulose; and cetyl alcohol, mastic and shellac, and shellac and stearic acid; polyvinyl acetate and ethyl cellulose; and neutral copolymers of polymethacrylates (Eudragit)TML30D); a copolymer of methacrylic acid and methyl methacrylate, or a neutral copolymer of polymethacrylate containing a metal stearate. Such coatings include mixtures of fats and fatty acids, shellac and shellac derivatives, and cellulose acid phthalates, such as those having a free carboxyl component.

One or more plasticizers may be added to the enteric polymer to increase its flexibility and reduce brittleness, as is known in the art. Suitable plasticizers include, for example, butyl citrate, triethyl citrate, diethyl phthalate, dibutyl sebacate, polyethylene glycol (PEG, e.g., PEG6000), acetyl triethyl citrate, and triacetin. In one embodiment, the plasticizer is triethyl citrate. Although some enteric materials are flexible and do not require plasticizers, relatively brittle polymers (e.g., Eudragit @)TMType L/S, EudragitTMRL/RS and EudragitTMFS 30D) benefits from plasticizers, for example, in the range of 5 wt.% to 30 wt.%, about 8 wt.% to about 12 wt.% triethyl citrate based on the dry polymer mass with poly (ethyl methacrylate-co-acrylate) 1: 1.

In certain embodiments, the enteric coating comprises one or more anti-tack agents (detackifiers) to reduce the stickiness of the film and prevent clumping, as is known in the art. Suitable antisticking agents include, but are not limited to, talc, glyceryl monostearate, gas phaseSilicon dioxide (e.g. Aerosil)TM200) Precipitated silicas (e.g. Sipernat)TMPQ) and magnesium stearate. The anti-blocking agent can be used in any suitable amount, for example, in the range of about 10 wt.% to 100 wt.%, about 10 wt.% to about 50 wt.%, about 10 wt.% to about 30 wt.%, or about 15 wt.% to about 30 wt.%, based on the dry polymer mass. For example, in one embodiment, the range is 15 wt.% to about 30 wt.% based on dry polymer mass.

One or more surfactants may also be added to the enteric coating mixture to improve wetting of the substrate and/or stabilize the suspension, as is known in the art. Surfactants include polysorbate 80, sorbitan monooleate, and sodium lauryl sulfate, as well as other surfactants described herein.

The enteric coating may be formed by any suitable method. Coating methods include, for example, pan coating, fluid bed coating, and dry coating (e.g., hot dry coating and electrostatic dry coating). Pan coating and fluid bed coating using solvents are recognized methods. In liquid coating, the enteric material and optional excipients (e.g., pigments, plasticizers, anti-adherents) are mixed in an organic solvent or water to form a solution or dispersion. The coating solution or dispersion is sprayed into solid dosage forms in a pan coater or fluid bed dryer and dried by hot air. For example, in the Wurster fluid bed coating process, the coating fluid is sprayed from the bottom of the fluid bed apparatus. Alternatively, the coating fluid is applied by top spraying. In certain embodiments, a tangential spray is applied.

The amount of enteric material is sufficient to achieve the desired acid resistance and release characteristics. For example, in one embodiment, the amount of enteric coating meets USP <711> requirements (USP 36-NF 31) for a delayed release dosage form, thereby releasing less than 10.0 wt.% of the drug after 2 hours in 0.1N HCl. In certain embodiments, the formulation releases at least 80% of the active ingredient within 20 minutes in a pH 6.8 buffered solution, e.g., using the dissolution method of USP 36-NF 31 section <711 >.

In one embodiment, the enteric coating is present in an amount ranging from about 10% to 40%, or 25% to about 35% (as measured by weight gain compared to the uncoated particulate core), or ranging from about 25% to about 31% weight gain, about 27% to about 31% weight gain, or about 28.5% to about 31% weight gain, based on the weight of the uncoated particulate core.

The formulation may include a capsule shell in which the beads are placed. Soft and hard capsule shells are known. In one embodiment, the capsule shell is a hard capsule shell, such as a gelatin capsule shell or a plant-based hard capsule shell. In certain embodiments, the capsule shell comprises one or more enteric coatings described herein. During accelerated storage, gelatin capsules may collapse. Thus, in certain embodiments, the formulation may comprise a hydroxypropyl methylcellulose capsule shell.

Thus, for example, one embodiment that combines the various features described above includes a pharmaceutical dosage form comprising a plurality of xanomeline beads comprising a core comprising xanomeline tartrate, a filler (optionally microcrystalline cellulose), a binder (optionally hypromellose), and an enteric coating (optionally Eudragit) surrounding the coreTML30D-55), wherein the plurality of beads has a particle size distribution ranging from about 0.7mm to about 2.5mm, wherein the enteric coating ranges from about 20% to about 40% based on the weight of the strain core, and wherein the beads are located in a capsule shell.

Size and shape of the beads

The plurality of beads has a particle size distribution. The plurality of beads have a bead shape. When present, the plurality of beads has a distribution of coating thicknesses.

Beads with a particle size distribution show favorable pharmacokinetics. Without being bound by any theory, it is expected that pharmacokinetics are affected by a plurality of beads having a core size distribution.

In one embodiment, the beads have a particle size in the range of about 0.4mm to about 1.2mm, for example about 0.4mm to about 0.5mm, about 0.5mm to about 0.6mm, about 0.6mm to about 0.7mm, about 0.7mm to about 0.8mm, about 0.8mm to about 0.9mm, about 0.9mm to about 1.0mm, about 1.0mm to about 1.1mm, or about 1.1mm to about 1.2 mm. In certain embodiments, the xanomeline beads have a size of about 0.425mm to about 1.18 mm. In certain embodiments, the xanomeline beads have a size of about 0.6mm to about 0.85 mm. In certain embodiments, the trospium bead is about 0.425mm to about 1.18mm in size. In certain embodiments, the trospium bead is about 0.6mm to about 0.85mm in size.

The beads or bead mixtures can be used, for example, as suspensions, filled into capsules, compressed into tablets or filled into sachets. One or more types of modified release beads can be mixed together and encapsulated, or sprinkled on a subject's food for use. In certain embodiments, the oral solid dosage form may be any of these forms. In certain embodiments, the dosage form is a capsule.

As the particle size of the beads becomes too small, the variability of the active ingredient content increases. As the particle size becomes too large, the beads are too large for the drug label to be administered by sprinkling (on applesauce or other soft food (e.g., jelly) and swallowing without chewing, or by enteral feeding tube. Also, as the particle size increases, larger particles are more coated than smaller particles, resulting in a lower relative assay compared to smaller particles. To compensate, relatively more beads are required to meet the labeling intensity of each capsule. It becomes difficult or impossible to fill the capsule shell with particles large enough to meet the labeling strength of each capsule (e.g., 75mg of xanomeline free base strength is filled into a size 0 capsule).

In one embodiment, the beads are encapsulated, for example, with an encapsulation machine. Various capsule sizes can be adapted to the strength and fill weight of the target formulation. For fill weights ranging from about 15mg to about 630mg, the capsule size ranges from 00-5.

The beads may be sorted (e.g., by sieving) to a desired particle size. In certain embodiments, the particle size range is any of the particle size ranges described above with respect to the core, or a combination thereof. In one embodiment, the particle size range is the same as the particle size range of the uncoated core. For example, the beads may be sieved such that 5% or less by weight of the bead cores remain on a #12 mesh (1.68mm) screen and 10% or less by weight pass through a #20 mesh (0.84mm) screen.

Method for preparing

A method of preparing an oral pharmaceutical composition is provided which comprises combining a strain comprising a plurality of xanomeline beads comprising xanomeline or a pharmaceutically acceptable salt thereof with a plurality of trospimine beads comprising a trospium salt, such as, for example, trospium chloride. In certain embodiments, the method further comprises formulating the mixed beads into a capsule.

Also disclosed herein is a method of making a dosage form comprising coating a core comprising xanomeline or a pharmaceutically acceptable salt thereof and an excipient with an enteric polymer to form an enteric coating, and coating a core comprising trospium chloride or a pharmaceutically acceptable salt thereof and an excipient with an enteric polymer to form an enteric coating. Optionally, the core may be formed by a wet granulation process. Optionally, the beads are sorted (e.g., by sieving) to a desired particle size range prior to enteric coating, and optionally after enteric coating.

These beads can be made by various methods including, but not limited to, spheronizing the extruded wet mass and coating the inert core spheres in a fluidized bed. In certain embodiments, the beads are prepared by extrusion and spheronization.

The beads are formulated to be free-flowing and compatible with modern encapsulation equipment. In some embodiments, the beads are blended together to form a homogeneous mixture that can be filled into capsules in a single stage. In other embodiments, the beads are filled separately into the capsule using a two-stage capsule filler.

The core comprising the xanomeline or a pharmaceutically acceptable salt thereof may be formed by any suitable method. In one embodiment, the core is formed by granulating a mixture of xanomeline or a pharmaceutically acceptable salt thereof and an excipient and grinding to a desired particle size range. In another embodiment, the core may be formed by extruding and spheronizing a mixture of xanomeline or a pharmaceutically acceptable salt thereof and an excipient.

The core comprising the chlorinated trospium or a pharmaceutically acceptable salt thereof may be formed by any suitable method. In one embodiment, the core is formed by granulating a mixture of chlorinated trospium or a pharmaceutically acceptable salt thereof and excipients and grinding to a desired particle size range. In another embodiment, the core may be formed by extruding and spheronizing a mixture of chlorinated trospium or a pharmaceutically acceptable salt thereof and excipients.

Granulation methods may include, for example, fluid bed granulation, wet granulation, hot melt granulation, and spray congealing. Other methods include slugging (slugging) and rolling. The mixture to be granulated is first dry blended. The dry blended dry ingredients may be mixed with water prior to extrusion.

Extrusion and spheronization of a mixture of xanomeline or a pharmaceutically acceptable salt thereof and chlorinated trospium and excipients can provide a desired core having a particle size distribution and one or more other desired properties as described herein. In certain embodiments, shorter processing times may result in a more stable product. For example, reducing rounding reduces friction and associated heat. Reducing the time the product is exposed to air (when moist and/or before packaging) can also reduce oxidation. On the other hand, rapid processing by extrusion and spheronization can result in a poor quality product, e.g., such that a large portion of the bead cores fall outside the desired particle size range. The moisture absorbed by the spheronization aid (which changes over time) affects the spheronization characteristics of the beads.

Accordingly, in one embodiment, the moisture content of the granulation mixture prior to drying ranges from about 20 wt.% to about 40 wt.%, e.g., 25 wt.% to about 35 wt.%, about 28 wt.% to about 32 wt.%, at least about 28 wt.%, at least about 28.5, about 20 wt.% to about 40 wt.%, about 25 wt.% to about 35 wt.%, about 27 wt.% to about 31 wt.%, or about 28.5 wt.% to about 31 wt.%.

In certain embodiments, the wet mass may be held prior to extrusion, for example, to swell the spheronization aid with the granulation fluid. The holding time may be at least 15 minutes, such as at least 30 minutes, at least 45 minutes, or at least 60 minutes. In certain embodiments, the holding time ranges from about 15 minutes to about 120 minutes, such as from about 30 minutes to 100 minutes, or from 60 minutes to 90 minutes.

As described above with respect to the core, the method may include the steps of: the core is sorted (e.g., by sieving) prior to optional coating to retain the particles within a predetermined size range, e.g., a size range of about 0.7mm to about 2.8mm, e.g., about 0.7mm to about 2.5mm, about 0.8mm to about 1.7mm, or any range described herein.

As described above with respect to beads, the method may comprise the steps of: the beads are sorted (e.g., by sieving) after optional coating to retain the particles in a size range, e.g., a size range of about 0.7mm to about 2.8mm, e.g., about 0.7mm to about 2.5mm, or about 0.8mm to about 1.7mm, or any range described herein.

In the extrusion and spheronization processes, the following optional features may be used alone or in combination with one or more thereof. The water may be a granulating agent. Microcrystalline cellulose may be used as a spheronization aid in the core. Hypromellose may be included in the core as a binder. The extrusion screen size may be 1.0 mm. The friction plates of the rounder may be cross-hatched. The friction plate of the spheronizer may be cross-hatched with a gridline of at least about 3mm, or greater than about 3mm, or to as little as about 4mm, or greater than about 4mm, or in the range of about 3mm to about 7mm, or about 5 mm. The spheronization time may be less than about 5 minutes, or less than about 4 minutes, or less than about 3 minutes, or less than about 2 minutes, or up to 1 minute. The spheronized particles may include non-spherical particles (i.e., irregular shapes), for example, a substantial portion thereof, such as at least about 20 wt.%, at least about 30 wt.%, at least about 40 wt.%, at least about 50 wt.%, at least about 60 wt.%, or at least about 70 wt.%.

In certain embodiments, the pharmaceutical composition is stored with a desiccant, e.g., pharmaceutical grade silica gel, crystalline sodium silicate, potassium or calcium aluminosilicate, colloidal silica, anhydrous calcium sulfate, and the like.

In certain embodiments, the pharmaceutical composition is stored with an oxygen absorber.

In certain embodiments, the pharmaceutical composition is stored under a dry inert gas such as nitrogen, helium, argon, neon, xenon, krypton, or mixtures thereof.

In certain embodiments, the pharmaceutical composition is stored at a reduced pressure compared to the outside ambient air.

In certain embodiments, the pharmaceutical composition is stored at a reduced temperature (e.g., at refrigeration temperatures (e.g., 2 ℃ to 8 ℃). in certain embodiments, the pharmaceutical composition is stored with fewer impurities (e.g., impurity a) than when stored at 25 ℃.

In certain embodiments, the pharmaceutical composition is stored by the manufacturer, distributor, pharmacy, or hospital at a temperature of about 2 ℃ to about 8 ℃ prior to dispensing the oral pharmaceutical composition to a subject. In certain embodiments, after dispensing an oral pharmaceutical composition to a subject, the pharmaceutical composition is stored at a temperature of about 20 ℃ to about 25 ℃.

Also provided is a method of stabilizing a pharmaceutical dosage form or composition described herein, the method comprising storing the dosage form at a temperature of from about 2 ℃ to about 8 ℃.

In certain embodiments, a method for preparing a pharmaceutical dosage form comprising a strain of xanomeline comprises forming a wet mass comprising xanomeline tartrate and an excipient (optionally microcrystalline cellulose) having a moisture content in the range of about 20 wt.% to about 40 wt.%, extruding and spheronizing the wet mass comprising xanomeline tartrate and an excipient to form cores, sorting the cores to a target particle size range (optionally about 0.7mm to about 2.5mm), coating the sorted cores with a polymer to form beads comprising a core and a coating, and sorting the bead particles to a target particle size range (optionally about 0.7mm to about 2.5 mm).

In certain embodiments, a method for preparing a pharmaceutical dosage form comprising a trospium strain, the method comprising forming a wet mass comprising chlorinated trospium and an excipient (optionally microcrystalline cellulose) having a moisture content ranging from about 20 wt.% to about 40 wt.%, extruding, spheronizing and drying the wet mass comprising chlorinated trospium and an excipient to make a core, sorting the core to a target particle size range (optionally about 0.7mm to about 2.5mm), coating the sorted core with a polymer to form beads comprising a core and a coating, and sorting the bead particles to a target particle size range (optionally about 0.7mm to about 2.5 mm).

Purity of

Also provided is the compound 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium.

Also provided is a pharmaceutical composition comprising xanomeline and/or a salt thereof and less than 0.5 wt.% of 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium (impurity a). In certain embodiments, the pharmaceutical composition comprises less than 0.30 wt.% of impurity a, e.g., less than 0.25 wt.%, less than 0.20 wt.%, less than 0.15 wt.%, less than 0.14 wt.%, or less than 0.1 wt.%. Also provided is a pharmaceutical composition comprising xanomeline and/or a salt thereof and less than 0.15 wt.% of 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium (impurity a).

Also provided is an oral pharmaceutical composition comprising a plurality of xanomeline beads comprising xanomeline or a salt thereof and less than 0.5 wt.% of 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium; and a plurality of trospimine beads comprising a trospimine salt. Also provided is an oral pharmaceutical composition comprising a plurality of xanomeline beads comprising xanomeline or a salt thereof and less than 0.15 wt.% of 3- [ (4-hexyloxy) -1,2, 5-thiadiazol-3-yl ] -5-hydroxy-1-methylpyridin-1-ium; and a plurality of trospimine beads comprising a trospimine salt.

In certain embodiments, the pharmaceutical composition comprises less than 0.5 wt.% of impurity a after storage of the pharmaceutical composition for at least 3 months at 40 ℃ and 75% relative humidity.

In certain embodiments, the total impurities in the pharmaceutical compositions provided herein are no more than about 5% by weight, no more than about 4% by weight, no more than about 3% by weight, no more than about 2.5% by weight, no more than about 2% by weight, no more than about 1.5% by weight, no more than about 1% by weight, no more than about 0.5% by weight, or no more than about 0.1% by weight.

Method of treatment

Further provided is a method of activating a muscarinic receptor in a biological sample, said method comprising contacting said biological sample with any of the oral pharmaceutical compositions described herein. Also provided is a method of treating a disorder ameliorated by the activation of muscarinic receptors in a subject in need thereof, comprising administering to the subject in need thereof any of the oral pharmaceutical compositions described herein.

Although M1 and M4 muscarinic receptor activators are considered to be effective treatments for schizophrenia, activation of muscarinic receptors located outside the brain leads to side effects that preclude clinical use of xanomeline. For example, in phase I and subsequent trials, the muscarinic agonist xanomeline has unacceptable GI and other side effects associated with muscarinic receptor binding in the distal regions of the human body. By combining xanomeline with chlorinated trospium, the desired therapeutic effect can be achieved while reducing or eliminating side effects associated with activation of muscarinic receptors located outside the brain.

Tolerance of the muscarinic activator, xanomeline, can be improved by co-administration with the muscarinic antagonist, trospium chloride. The most common adverse events observed with xanomeline administration are nausea, vomiting, diarrhea, excessive sweating, and excessive salivation (so-called cholinergic adverse events). The disclosed compositions reduce the incidence of these adverse events in humans, demonstrating increased xanomeline tolerance.

In one embodiment, xanomeline is combined with chlorinated trospium to treat muscarinic disorders, alleviating symptoms of muscarinic activation in response to xanomeline production in living tissue found outside the brain. In one embodiment, such diseases or disorders include schizophrenia and diseases associated with schizophrenia, cognitive disorders in neurodegenerative diseases such as alzheimer's disease, and pain such as nociceptive pain or neuropathic pain. The combination of xanomeline and chlorinated trospium is a safer approach for treating diseases that respond to activation of muscarinic receptors.

In another embodiment, xanomeline and chlorinated trospium treat mood disorders. In another embodiment, the xanomeline and the chlorinated trospium treat a movement disorder. In another embodiment, xanomeline and trospium chloride treat cognitive disorders, including enhancing cognitive function not associated with a particular pathology. In another embodiment, the xanomeline and the chlorinated trospium treat attention disorders. In another embodiment, xanomeline and chlorinated trospium are used to treat pain. In addition to disease treatment, increased attention can increase learning rates and reduce fatigue due to lack of sleep and circadian rhythm disturbances (e.g., jet lag). In another embodiment, xanomeline and chlorinated trospium are used to treat an addictive disorder.

In one embodiment, xanomeline is treated in combination with chlorinated trospium in an animal. In another embodiment, the animal is a mammal. In one embodiment, the mammal is a human.

In one embodiment, the chlorinated trospium chloride reduces the side effects associated with xanomeline. Such side effects include, but are not limited to, GI side effects, cardiac side effects, hyperhidrosis, and excessive salivation. Use of trospium with xanomeline allows for clinical use of xanomeline when xanomeline cannot be used clinically due to its side effects. In another embodiment, the use of chlorinated trospium along with xanomeline allows the xanomeline to achieve a higher maximum tolerated dose than it would otherwise be possible to achieve.

Various time and resource intensive methods have demonstrated the combined efficacy of xanomeline and chlorinated trospium. For example, animal models demonstrate the efficacy of new therapeutic agents for schizophrenia, including pharmacological models (e.g., ketamine models) and genetic models (e.g., DISC1 mice). Likewise, animal models (including rodents, dogs, and non-human primates) demonstrate a spectrum of side effects of pharmacological effects. Animal models are experimental alternatives to humans, but may suffer from deficiencies in physiological differences from human to animal, and thus may have limited predictive capabilities for human experimentation, particularly in relation to central nervous system disorders. Alternatively, the disclosed combinations can be tried in a human controlled clinical trial. Standard measures based on patient self-reporting can be used by those skilled in the art to assess various side effects, such as GI discomfort. As another example, objective physiological measurements (e.g., EKG) may be used by one skilled in the art. A standard set of measures to assess the symptoms of schizophrenia has also been developed, including the Brief Psychotic Rating Scale (BPRS), the positive and negative syndrome scale (PANSS), and the Clinical Global Impression (CGI). Typically, clinical trials are double-blind, with one group of patients receiving ineffective placebo and another group of patients receiving active intervention.

Prior to administration of the claimed combination, the patient may have a lead-in period of one to fourteen days during which the lead-in period of chlorinated trospium chloride is administered alone. In one embodiment, the chlorinated trospium is administered one or more dose cycles prior to administration of the xanomeline, to accumulate chlorinated trospium in the body, or in order to bring the chlorinated trospium to or near a steady state exposure level. This accumulation or higher exposure level of chlorinated trospium increases blockade of muscarinic receptors outside the brain and reduces adverse events when taking xanomeline. In another embodiment, the chlorinated trospium is administered one or more days prior to the xanomeline.

In one embodiment, the patient is administered 6 times xanomeline and chlorinated trospium during a 24 hour period. In another embodiment, the patient is administered xanomeline and chlorinated trospium 5 times during a 24 hour period. In another embodiment, the patient is administered xanomeline and chlorinated trospium 4 times during a 24 hour period. In one embodiment, the xanomeline and the chlorinated trospium are administered to the patient 3 times during a 24 hour period. In another embodiment, the xanomeline and the chlorinated trospium are administered to the patient twice during a 24 hour period. In another embodiment, the xanomeline and the chlorinated trospium are administered to the patient once during a 24 hour period.

In one embodiment, an extended release formulation of chlorinated trospium is used in combination with xanomeline. In another embodiment, the extended release of chlorinated trospium is administered to the patient one to five times during a 24 hour period. In one embodiment, the chlorinated trospium extended release agent is administered one to three times during a 24 hour period. In another embodiment, from 5 to 400 milligrams of the chlorinated trospium extended release agent is used during a 24-hour period. In one embodiment, from 20 to 200 milligrams of the chlorinated trospium extended release agent is used during a 24-hour period.

In one embodiment, 225mg of xanomeline and 40mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 100mg of xanomeline and 20mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 125mg of xanomeline and 20mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 125mg of xanomeline and 30mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 125mg of xanomeline and 40mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 200mg of xanomeline and 40mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 200mg of xanomeline and 80mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 250mg of xanomeline and 60mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 250mg of xanomeline and 80mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 300mg of xanomeline and 40mg of chlorinated trospium are administered to the patient during a 24 hour period. In another embodiment, 300mg of xanomeline and 80mg of chlorinated trospium are administered to the patient during a 24 hour period.

Treatment may be initiated with a small dose. Thereafter, the dosage may be increased in small increments until a balance between therapeutic effect and side effect is achieved. When treating a subject, the health of the patient may be monitored by measuring one or more relevant indicators at predetermined times during the treatment. Treatments, including composition, amount, time of application, and formulation, may be adjusted based on such monitoring. The patient may be re-evaluated periodically to determine improvement by measuring the same parameters. The disclosed compositions administered and possibly the time of administration may be adjusted based on these re-evaluations.

Examples of the invention

The following examples are provided for illustration and are not intended to limit the scope of the present disclosure.

Example 1 immediate Release beads

Beads of xanomeline tartrate (table 1) and trospium chloride (table 2) were prepared.

Table 1: xanomerine tartrate (66%) beads, talc-free

Composition (I) % w/w (dry basis) g/batch
Xanomerine tartrate 66 99
Microcrystalline cellulose 34 51
Purified water (30) (45)
Total amount: 100 150

removed during drying.

Table 2: chlorotrichloramine (17.7%) beads, talc free

Composition (I) % w/w (dry basis) g/batch
Chlorinated trospium chloride 17.7 17.7
Microcrystalline cellulose 35 35
Lactose monohydrate 47.3 47.3
Purified water (45) (45)
Total amount: 100 100

removed during drying.

The powder was sieved using a Quadro Comil 197 fitted with a 457 μm round mesh sieve, 0.2 inch shim at 1625rpm and mixed in a Hobart low shear mixer/granulator (model N-50) at a fixed speed of 60rpm for 2 minutes. The dry blending step is optional because the mixing homogeneity is driven by the subsequent wet granulation. The beads were sieved by hand through a 40 mesh (425 μm) sieve.

Wetting was performed in the Hobart (Hobart). Water was added using a Cole-Parmer peristaltic pump. The water addition rate (amount of water/time of dose) is a process variable.

The wet mass was extruded through a perforated screen (dome configuration) single screw extruder using an LCI Multi Granulator MG-55 at 30rpm (shaft speed). After wetting, the wet mass is extruded directly. Hold time, shaft speed and extrusion rate (load) are process variables.

The extrudate was placed in an LCI Marumerizer (spheronizer) QJ-230T equipped with a 2.0mm friction plate. The extrudate was spheronized at different plate speeds for a total of no more than 4 minutes. The rounding speed and time are process variables.

Using AeromaticTMThe stream-1 fluidized bed dried the beads at an inlet temperature of 60 ℃ until a moisture content of not more than 3% was obtained. Since the beads melt after a few minutes at 60 ℃, the beads are dried at 30 ℃.

The water content was assessed gravimetrically by Loss On Drying (LOD) using a mertler-Toledo model HR83 halogen moisture analyzer. The beads were heated at 105 ℃ until the weight loss rate dropped to less than or equal to 0.0% in 60 seconds.

Table 3: extrusion/spheronization process parameters

Example 2-extended immediate Release bead formulation

The beads of example 1, with and without talc, were expanded (tables 4-7). The extrusion/spheronization process parameters are shown in table 8.

Table 4: xanomerine tartrate (66%) without talc

Removed during drying.

Table 5: xanomerine tartrate (66%) with talc

Abbreviations: eur ═ european pharmacopeia, USP ═ united states pharmacopeia

Evaporated during processing and is therefore not included in the total weight

Table 6: chlorotrichloramine (17.7%) beads, talc free

Composition (I) % w/w (dry basis) g/batch
Chlorinated trospium chloride 17.7 88.7
Microcrystalline cellulose 35 175.0
Lactose monohydrate 47.3 236.3
Purified water (59) (295)
Total amount: 100 500

removed during drying.

Table 7: chloramine chloride (17.7%) beads, talc powder

Abbreviations: NF, ph, eur, european pharmacopeia, USP, united states pharmacopeia. Evaporation during treatment

Table 8: extrusion/spheronization process parameters

Example 3 Capsule stability and dissolution test

Capsules were produced by weighing the beads and filling into HPMC capsules manually. Using AccofilTMThe capsule filling machine manually encapsulates the beads, wherein the beads previously mixed with talc (0.5%) are filled in the capsules one by one, respectively, as shown in table 9.

Table 9: composition of xanomeline/chlorinated trospium capsules. Ingredients are listed in mg/capsule.

After drying, the beads were sieved through 16 mesh (1.18mm) and 40 mesh (0.425mm) sieves by shaking for 5 minutes. Beads between 1.18mm and 0.425mm in size of the sieve were retained for further analysis.

Using JSM-6010LV InTouchScope with a backscattered electron detector (BES)TM(JEOL Ltd, Tokyo, Japan) microscope, and the morphology and surface characteristics of the beads were examined by Scanning Electron Microscope (SEM). The samples were placed on metal stakes using double-sided carbon conductive tape. Images were obtained under low vacuum (60Pa) and 30 Xmagnification at an accelerating voltage of 20 kV.

Bulk density and bulk density were determined in duplicate using a USP <616> method using a bulk density tester (JV 1000, cobley Scientific). Bulk density is measured from the volume of a powder sample of known mass in a graduated cylinder. Bulk density was measured by mechanically tapping the cylinder until the volume no longer changed.

The flow properties of the powders were evaluated using the Carr's Compressibility Index (Carr's compressive Index) and the Hausner ratio (Hausner ratio), both derived using measurements of volume and bulk density, and the Carr's Compressibility Index (CI) was calculated using the volume and bulk density data when fitted to the following equation: compressibility index ═ (bulk density-bulk density)/bulk density × 100%. The hausner ratio (H) is calculated as the ratio of bulk to bulk density. The capsules were analyzed for appearance, assay, related substances, water content and dissolution. Figure 1 shows a stability schedule and protocol for the xanomeline/trospium capsules.

The beads further range in size from 0.6mm to 0.85 mm. Some beads exhibited similar morphological properties. Modifications in some other beads reduce the density of the beads and result in a rough surface and loss of sphericity. Scanning Electron Microscope (SEM) images of 66% beads of xanthomeline tartrate (fig. 2) and 17.7% beads of chlorinated trospium chloride (fig. 3) at 30x magnification showed that the beads were between 0.6mm and 0.85mm in size. These beads are used in xanomeline/trospium capsules. The Particle Size Distribution (PSD) of the beads was determined by mechanical sieving. As shown in table 10, most beads of both APIs ranged in size from 0.425mm to 1.18 mm.

Table 10: particle size distribution of beads by mechanical sieving

Table 11 shows the density and flow properties of the beads collected between 0.425mm to 1.18mm screens. Xanomeline tartrate and trospium chloride IR beads exhibit different density and flow properties, which may be critical when mixing bead systems.

Table 11: density and flow Properties of 0.425mm-1.18mm beads

The analysis in table 12 shows the following good results: the moisture content of the test and related materials, as well as 50mg of xanomeline and 20mg of chlorinated trospium capsules, were determined. The data in table 13 show that these attributes are retained during the storage stability study. Similar data for 50mg xanomeline capsules and 10mg chlorinated trospium capsules are provided in table 14. Dissolution data for both dosage forms are provided in tables 15 and 16. Additional tables showing stability for the xanomeline/chlorinated trospium formulation are shown in figures 6-41.

Table 12: analysis results

Table 13: stability of KarXT 50/20

Table 14: dissolution of KarXT 50/20

Table 15: measurement of KarXT 50/10 and related substances

Table 16: dissolution of KarXT 50/10

Subsequent testing showed that KarXT 50/10, 50/20, and 75/20 in hard shell capsules were stable at 25 ℃/60% RH for at least 12 months. According to the present data, a shelf life of 15 months at 25 ℃/60% RH is recommended.

The dissolution results indicate that the two compounds are released very quickly, which may improve their bioavailability, and that they are also released at comparable rates despite the large differences in composition between the two bead formulations. Both xanomeline and chlorinated trospium have low bioavailability, while rapid release can increase bioavailability by overwhelming the saturable processes that limit absorption into the general circulation.

During the stability study of the combination drug product, an unknown xanomeline impurity was observed with a relative retention time of about 1.09. During the test, impurities were first observed for the 50mg xanomeline/10 mg chlorinated trospium drug product at the three month time point and for the other three combination products at the initial time point, both of which occurred simultaneously. The impurity peak increases with both increased time and increased storage temperature. The impurity was not observed before the present study.

Preliminary studies indicate that the RRT 1.09 impurity is 3- [ (4-hexyloxy) -1,2, 5-thiadiazole-3-yl]-5-hydroxy-1-methylpyridin-1-ium (C)14H20N3O2S+,MW=294.1271Da):

The RRT 1.09 impurity is the hydroxylated form of Compound V (C)14H20N3OS+MW 278.1322Da), which is the penultimate intermediate with negative mutagenesis potential in the synthesis of xanomeline:

in order to reduce the existence of impurities, the storage temperature of the medicine is reduced. During packaging, the bottles were flushed with argon to minimize headspace oxygen. In certain embodiments, the xanomeline bead formulation is formulated with an antioxidant (e.g., 0.5 wt.% ascorbic acid or 0.05 wt.% BHT).

Example 4-KAR-001 phase I study of combination of Xanomerine and chlorinated trospium chloride

In normal healthy volunteers, a phase I, double-blind, randomized, multi-dose lead study of xanomeline alone was conducted, as compared to xanomeline and trospium chloride administered together. The primary objective of this study was to (1) evaluate the safety and tolerability of: 225mg of xanomeline per day and 40mg of chlorinated trospium per day for 7 days, as compared to 225mg of xanomeline per day alone for 7 days; and (2) determining whether addition of 40mg daily of trospium chloride (20mg BID) to 225mg daily of xanomeline (75mg TID) significantly reduced peripheral cholinergic side effects (nausea, diarrhea, vomiting, sweating, excessive salivation) over 7 days relative to 225mg daily of xanomeline alone. Table 17 lists the parameters of this study.

Table 17: parameters of the KAR-001 study

Seventy study subjects were randomized into groups, of which 68 study subjects received at least one evaluation on day 3 (i.e., the first day of xanomeline administration). Table 18 lists the demographics of the study subjects.

Table 18: demographics of the subject of the KAR-001 study

The most common adverse events with xanomeline are the so-called cholinergic adverse events of nausea, vomiting, diarrhea, excessive sweating and excessive salivation. In this study, co-administration of chlorinated trospium with xanomeline resulted in a 43% reduction in the incidence of cholinergic adverse events (statistically significant (p ═ 0.016)) compared to xanomeline co-administered with placebo. In the study's xanomeline + placebo group, 63% of the subjects reported at least one cholinergic adverse event, in contrast to only 34% of the study's xanomeline + chlorinated trospium group.

Furthermore, the incidence of each type of individual cholinergic adverse event in the subject administered the xanomeline + chlorinated trospium was also reduced in the study compared to the incidence in the subject administered the xanomeline + placebo. The reduction in the incidence of sweating was itself statistically significant, with an incidence of 20.0% in the xanomeline + chlorinated trospium group and a 48.5% reduction of 59% in the xanomeline + placebo group (p ═ 0.013).

The total cholinergic adverse event incidence for the xanomeline + trospium chloride group in this study was very similar to the 32% incidence reported in subjects using placebo + placebo during the two-day break-in period. Although these two data points did not occur at different times in the study, the fact that the incidence of cholinergic adverse events was comparable to placebo suggests that the 43% reduction in adverse events by chlorinated trospium may have approached the maximum reduction possible in this study.

Table 19 shows the incidence and number of cholinergic adverse events in the evaluable population of the study as follows, with all p values based on the chi-square test except for those labeled with a x, which are based on the fisher (Fishers) exact test.

Table 19: cholinergic adverse events

In addition to assessing whether the addition of chlorinated trospium chloride could improve the tolerability of the xanomeline, this study also provided data on the overall safety and tolerability of xanomeline + chlorinated trospium chloride. Table 20 shows that the combination was generally well tolerated without severe and critical adverse events, and that most adverse events were mild.

Table 20: tolerance to stress

The tolerance profile found in this study allows for continued combined studies of xanomeline and chlorinated trospium in the future.

Example 5-KAR-003 phase I study of KarXT (Xanomelin + trospium combination formulation)

This study was a phase 1, randomized, multi-dose, acclimatized hospitalization study to evaluate the safety and tolerability of KarXT in normal healthy volunteers between 18 and 60 years of age. Subjects signed an informed consent and received screening assessments from day-21 to day-1. Upon successful completion of all screening assessments, subjects returned to the study clinic on day 0 for baseline safety assessments and enrollment into the study, and each cohort was randomized into one of two treatment groups at 3: 1: KarXT or placebo. Subjects were assigned to 1 of 4 cohorts (cohorts 1,2, 3 or 4).

Study drug was administered BID on days 1 to 7. A combined dose formulation of xanomeline and trospium was used in all cohorts. All cohorts started with a 2 day lead-in of KarXT 50/20 BID (for subjects randomized to receive active treatment); after the 2-day lead-in period, the non-blind pharmacist distributed study medication to each subject according to the subject's random assignment for a 5-day assigned cohort for a total of 7 days of treatment. A matching placebo was administered throughout the study to maintain blindness. Sentinel groups were introduced into the study for cohorts 2 to 4 and were monitored for safety and tolerability by the Data Safety Evaluation Group (DSEG) so that approximately 30% of the proposed cohorts had received treatment and were subjected to safety assessments prior to dosing the remaining subjects of the cohort. Subjects and study clinic staff were blind to treatment. The Dose Selection Committee (DSC) was non-blind to determine the dose for subsequent treatment groups.

Serial blood samples were drawn on days 1, 3 and 7 for PK assessment of xanomeline and trospium. More blood was collected at regular intervals for monitoring the trough concentrations of salmeterol and trospium and clinical laboratory assessments. On day 1, saliva amounts were collected twice. Saliva volume was measured before dosing on day 1 and then daily (afternoon) on days 1 to 7 at approximately the same time each day to avoid diurnal variation. Other assessments include pupil size measurement and Bristol stool scale assessment. Subjects remained in the study clinic for the entire treatment period (7 days). After the safety assessment on day 8, the subjects were discharged from the study clinic and required a return for the final safety assessment about 14 days after study drug administration.

During the course of the study, after 2 days introduction of KarXT 50/20 BID (for subjects randomized to active treatment) in each cohort, subjects were dosed as follows:

in cohort 1, subjects completed KarXT 100/20 BID (total daily dose (TDD) of 200mg of xanomeline plus 40mg of trospium) or placebo dosing on days 3 to 7.

In cohort 2, the sentinel group (group 2 a) discontinued dosing after the 4 th morning dose. The subjects in cohort 2 had a dose of KarXT 150/20 BID (300mg xanomeline plus 40mg trospium TDD) or placebo. The dosing in cohort 2 was discontinued (as determined by DSEG with observed tolerability problems). Since DSC determined that further dosing with KarXT 150/20 BID in cohort 2 is unlikely to be well tolerated enough to warrant further development of this dose combination for the clinical population, the study proceeded to dosing in cohort 3 (group 3 a).

In cohort 3, the sentinel group (group 3 a) completed the 3 rd to 7 th day administration of KarXT 150/40BID (TDD with 300mg xanomeline plus 80mg trospium) or placebo. The second group in cohort 3 (group 3 b) discontinued dosing after the 5 th morning dose.

In cohort 4, the sentinel group (group 4 a), the second group (group 4 b) and the remaining group (group 4 c) completed the administration of KarXT 125/40 BID (TDD with 250mg xanomeline plus 80mg trospium) or placebo on days 3 to 7.

Ninety-six subjects were planned, 248 subjects were screened, and randomized69 subjects were allocated, 51 subjects completed the study, and 18 subjects discontinued the study. The population comprises healthy male and female subjects aged 18 to 60 years and having a body mass index of 18 to 40kg/m2. Subjects were excluded from the study if they had a history of irritable bowel syndrome or severe constipation requiring treatment within 6 months prior to screening. Subjects were also excluded from the study if they had a history of or presence of any disease or disorder, including psychiatric or neurological disorders, which researchers believe might compromise subject safety or study outcome efficacy. Table 21 summarizes the demographics and baseline characteristics by treatment group. The demographic and baseline characteristics were consistent between the safety and PK populations.

Table 21: summary of demographic and baseline characteristics by treatment group-safety population

Serial blood samples were collected from all subjects in each cohort on days 1, 3, and 7 prior to the morning dose, and at 1,2, 3, 4, 6, 8, 10, and 12 hours after the morning dose for evaluation of the PK of xanomeline and trospium. The PK parameters listed below were calculated from individual xanomeline and trospium concentration-time profiles by standard non-compartmental methods. Calculating CMaximum ofAnd dose normalization parameters of the area under the concentration-time curve (AUC) values. During the study period, additional blood samples were collected for monitoring the trough concentrations of xanomeline and trospium on days 2, 4, 5 and 6 and prior to discharge on day 8 prior to the morning dose.

Safety assessments include spontaneously reported adverse events, ECG, laboratory assessments, vital signs, salivary volume assessments, bristol stool scale, pupil size, and physical examination. Descriptive statistics (n, mean, standard deviation, median, minimum and maximum) summarize the continuous data by treatment group. Geometric Mean (GM), geometric percent coefficient of variation (CV%), quartiles, or boxplots are generated. Although no formal statistics are made, the count and frequency tabulate the classification measures.

Unless otherwise indicated, treatment groups are summarized below: KarXT 50/20 BID (summarized for adverse events and day 1 PK only), KarXT 100/20 BID, KarXT 125/40 BID, KarXT 150/20 BID, KarXT 150/40BID, and placebo (empty)Adding capsules andall cohorts placebo groups were combined). Safety assessments are based on spontaneously reported adverse events, ECG, laboratory assessments, and vital signs. Exploratory analysis was also performed on saliva volume, bristol stool volume, and pupil size.

Upon oral administration of the KAR-003 formulation at all doses, the xanomeline is well absorbed into the systemic circulation. A peak concentration of xanomeline was observed at a median time of 2 hours for all treatment groups and study days.

Median t of xanomeline between treatment groups and throughout the study day1/2The values are similar, indicating t1/2And not dose-dependent. Median value t1/2Ranging from 3.4 to 5.8 hours.

The GM xanomeline exposure was increased on day 3 from 100 to 150mg (when xanomeline was administered with 20mg trospium) or from 125 to 150mg (when administered with 40mg trospium) without dose scaling. Lower xanomeline exposure was observed following treatment with KarXT 150/40 compared to KarXT 125/40. Day 3 GM xanomeline was exposed when a 150mg dose of xanomeline was administered with 20mg and 40mg of trospium chloride (C)Maximum of,AUC0-lastAnd AUC0-12hr) Similarly. On day 7, when xanomeline was used with 40mg of trospium,the increase in GM xanomeline exposure was slightly more than the dose proportionately from 125mg to 150 mg.

Minimal to no xanomeline accumulation in plasma on days 3 to 7 after treatment with KarXT 100/20 BID and KarXT 125/40 BID; however, of the 4 subjects who completed the study, 3 had accumulated after administration of KarXT 150/40 BID. The average accumulation ratio of the KarXT 150/40BID group was 366.2% for RAUC, RCMaximum ofThe content was 445.4%.

Example 6-Xanomemerine pharmacokinetics of KAR-003 in comparison to KAR-001

Comparison of the xanomeline GM exposure between KAR-001(75mg xanomeline TID. + -. 20mg trospium BID) and the group of KarXT 100/20 BID from KAR-003 indicates CMaximum ofValue and AUC0-6hr(KAR-003) or AUC0-tauThe (KAR-001) value is greater in KAR-003 (days 3 and 7) than the corresponding exposure from KAR-001 (days 3 and 9). In both studies and two days (day 3 and day 9 for KAR-001 and day 3 and day 7 for KAR-003), a median T was observed at 2 hoursMaximum of. These data indicate that the KarXT formulation enhances xanomeline exposure.

After oral administration of the KarXT formulation at all doses, trospium is absorbed into the systemic circulation. A peak concentration of trospium was observed at a median time of 1.0 hours for all treatment groups and study days.

Median t of trospium chloride between treatment groups on day 31/2Values were similar, with values ranging from 4.1 to 4.8 hours. Median t of KarXT 100/20 BID (4.9 hours) and KarXT 125/40 BID (4.5 hours) treatment on day 71/2Similarly, but slightly longer for the KarXT 150/40BID group (7.1 hours).

When administered with 150mg of xanomeline, the increase in GM trospium exposure on day 3 was slightly less than the dose proportionately from 20mg to 40 mg. When a 20mg BID dose of trospium was administered with 100mg BID xanomeline, the GM trospium exposure was greater on day 3 (C) than 150mg BID xanomeline (C)Maximum of,AUC0-lastAnd AUC0-12hr). When the dosage of 40mg trospium BID is matched with 125mg xanomeline BID and 150mg xantheneThe GM trospium exposure on day 3 was similar when nomelin BID was administered together.

Trospium does not accumulate in plasma from day 3 to day 7 after administration of KarXT 100/20 BID, KarXT 125/40 BID, and KarXT 150/40 BID. For the KarXT 100/20 BID group, trospium accumulated in plasma from day 1 to day 7. The average day 7/day 1 accumulation ratio was 348.7% (RAUC) and 379.9% (RC)Maximum of)。

Comparison of the trospimine GM exposure of the KarXT 100/20 BID group from KAR-001 and from KAR-003 shows C from KAR-003Maximum ofAnd AUC0-12hrThe values were greater on two days (day 3 and day 9 for KAR-001, day 3 and day 7 for KAR-003) than the corresponding exposures from KAR-001. On two days in both studies, the median T of trospium was observedMaximum ofIs 1.0 hour. These data indicate that the KarXT formulation enhances trospium exposure.

All cohorts of KAR-003 began with a 2-day lead-in period of KarXT 50/20 BID (for subjects who randomly received KarXT). Figure 42 shows the mean (± SD) xanomeline PK concentrations, and table 22 summarizes the xanomeline PK parameters for all cohorts of PK populations for KarXT 50/20 BID treatment day 1. The samples collected prior to administration of the first dose of xanomeline on day 1 did not show measurable concentrations of xanomeline. The concentration of xanomeline was quantifiable (>50pg/mL) at all time points up to 12 hours after morning dose administration on day 1.

Table 22: xanomelin PK parameters on day 1 by KarXT 50/20 BID (all cohorts)

Feature(s) n Statistics of
CMaximum of(pg/mL) 53 1972.3(131.8)
TMaximum of(h) 53 2.0(1.0,8.0)
t1/2(h) 48 3.4(2.0,4.6)
AUC0-last(h*pg/mL) 53 10775.5(102.2)
AUC0-12hr(h*pg/mL) 52 10810.3(103.5)
AUC0-inf(h*pg/mL) 48 12836.1(97.7)

Figure 43 shows the mean (± SD) xanomeline PK concentrations produced by treatment on day 3 for the PK population, and table 23 summarizes these parameters. For all cohorts, the concentration of xanomeline in samples prior to morning dose of study drug administered on day 3 and at all time points from morning dose administration on day 3 to 12 hours was quantifiable, except for one subject whose xanomeline plasma concentration was at 12 hours post-dose<50.0 pg/mL. The variability range between subjects for T across the four treatment groupsMaximum ofIs 23.7% to 58.2% (CV%) for CMaximum ofIs 79.8% to 136.3% (geometric)CV%) for t1/2Is 21.6% to 26.3% (CV%) for AUC0-12hrIs 77.1% to 96.1% (geometric CV%). Median xanomeline T at day 3 for groups KarXT 100/20 BID, KarXT 125/40 BID, KarXT 150/20 BID, and KarXT 150/40BIDMaximum ofIs 2 hours. Across four treatment groups, single TMaximum ofValues range from 1.0 to 6.0 hours. In contrast to previous studies of KAR-001 (where the elimination phase has not been well characterized), t1/2Estimated in 51 out of 53 subjects. Median xanomeline t on day 3 across four treatment groups1/2Are similar in value. Median value t1/2Ranging from 3.4 to 4.3 hours. Across four treatment groups, single t1/2Values range from 2.4 to 8.6 hours.

Table 23: xanomerine PK parameters generated by treatment on day 3

Day 3 dose normalized GM exposure (dose normalized GM C) for xanomeline when BID is administered, due to the increase of xanomeline dose from 100mg (cohort 1) to 150mg (cohort 2) without changing the trospium dose (20mg) for xanomelineMaximum ofAnd dose normalized GM AUC0-lastAnd AUC0-12hr) And decreases. Similarly, since the xanomeline dose was increased from 125mg (cohort 4) to 150mg (cohort 3) without changing the trospium dose (40mg), the day 3 dose-normalized GM exposure was slightly lower for xanomeline (i.e., the xanomeline exposure after treatment with KarXT 150/40BID was lower compared to treatment with KarXT 125/40 BID). Comparison of the xanthomeline BID exposure of 150mg with either 20mg or 40mg of trospium BID administered, shows GM, C on day 3 for xanthomelineMaximum of、AUC0-lastAnd AUC0-12hrSimilarly.

Figure 44 shows the mean (± SD) xanomeline PK concentrations produced by treatment on day 7 for the PK population, and table 24 summarizes these parameters. For the KarXT 100/20 BID, KarXT 125/40 BID, and KarXT 150/40BID groups, the concentration of xanomeline in samples collected prior to the morning dose of study drug administered on day 7 and at all time points from the morning dose to 12 hours on day 7 was quantifiable. Across the KarXT 100/20 BID, KarXT 150/40BID and KarXT 125/40 BID groups, the variability range between subjects was for TMaximum ofIs 38.3% to 47.9% (CV%) for CMaximum ofIs 81.4% to 106.8% (geometric CV%) for t1/2Is 15.4% to 42.1% (CV%) for AUC0-12hrIs 45.2% to 71.2% (geometric CV%). Median xanomeline T at day 7 for groups KarXT 100/20 BID, KarXT 125/40 BID and KarXT 150/40BIDMaximum ofIs 2.0 hours. Across groups of KarXT 100/20 BID, KarXT 150/40BID and KarXT 125/40 BID, a single TMaximum ofThe value ranges from 0.0 to 6.0 hours. Median xanthometrine t at day 7 for groups KarXT 100/20 BID, KarXT 125/40 BID and KarXT 150/40BID1/2Are similar in value. Median value of xanomeline t1/2The range is 4.6 to 5.8 hours. Across groups of KarXT 100/20 BID, KarXT 150/40BID and KarXT 125/40 BID, a single t1/2The values range from 3.6 to 14.0 hours.

Table 24: xanomerine PK parameters generated by treatment on day 7

Day 7 dose normalized GM exposure (dose normalized GM C) for xanomeline due to an increase in xanomeline dose from 125mg (cohort 4) to 150mg (cohort 3) without changing the trospium dose (40mg) when BID was administered for KarXTMaximum of、AUC0-lastAnd AUC0-12hr) And (4) increasing.

Table 25 summarizes the xanomeline PK accumulation rates (day 7/day 3) by treatment of the PK population. Minimal to no xanomeline accumulation in plasma from day 3 to day 7 was based on the mean rate of xanomeline accumulation following treatment with KarXT 100/20 BID (cohort 1) and KarXT 125/40 BID (cohort 4). The average accumulation ratio of the KarXT 100/20 BID group was 133.4% for RAUC and for RCMaximum ofIs 130.5%, the average accumulation ratio of the KarXT 125/40 BID group is 143.9% for RAUC and 143.9% for RCMaximum ofIs 151.0%. Only one subject in the KarXT 100/20 BID group showed lower exposure on day 7 compared to day 3. In contrast, xanomeline slightly accumulated in three of the four subjects completing the study in the KarXT 150/40BID group. Another subject in the KarXT 150/40BID group showed similar exposure on days 3 and 7. The average accumulation ratio of the KarXT 150/40BID group was 366.2% (RAUC) and 445.4% (RC)Maximum of)。

Table 25: xanomerine PK accumulation rates produced by treatment (day 7/day 3)

Figure 45 compares the mean (± SD) xanomeline PK concentration-time profiles for treatment and visit (days) by PK population. Figure 46 shows mean (± SD) xanomeline PK trough concentrations treated by PK populations. Steady state was not assessed.

Comparison of the xanomeline GM exposure between KAR-001(75mg xanomeline TID. + -. 20mg trospium BID) (Table 23) and the KarXT 100/20 BID group from KAR-003 (Table 21) indicates that the C of the KarXT 100/20 BID group (KAR-003) on day 3Maximum ofValue and AUC0-6hr(KAR-003) or AUC0-tau(AUC from 0 to 6 hours) values (KAR-001) were 2.3 to 2.6 times greater than the corresponding exposure from KAR-001 on day 3.

Will be from KAR-0Comparison of the day 7 GM exposure of the KarXT 100/20 BID group at 03 (Table 22) with the day 9 GM exposure from xanomeline alone and the xanomeline + trospium exposure from KAR-001 (Table 23) shows that the value of the KarXT 100/20 BID group at 7 (KAR-003) is approximately 1.4 to 1.8 times greater than the corresponding exposure from KAR-001 at 9. Median T on days 3 and 7 of KAR-003 (Table 22) and 3 and 9 of KAR-001 (Table 23)Maximum ofIs 2.0 hours. These data indicate that the KAR-003 formulation provides adequate exposure and PK properties.

Table 26 summarizes the subset of KAR-003 xanomeline PK parameters for the KarXT 100/20 BID group at day 3 and day 7 for the PK population. Table 27 presents a summary of KAR-001 treatment on day 3 and day 9 parameters of the KAR-001 xanomeline PK for the PK population.

Table 26: subset of Xanomelin PK parameters on days 3 and 7 for KarXT 100/20 BID

Table 27: subsets of the Xanomelin PK parameters of KAR-001 at days 3 and 9

Figure 47 presents the mean (± SD) trospium PK concentrations on day 1 for KarXT 50/20 BID treatment (all cohorts) for the PK population, and table 28 summarizes these parameters. The samples collected prior to the administration of the first dose of trospium on day 1 did not show measurable concentrations of trospium. The concentration of trospium was quantifiable (>20pg/mL) at all time points up to 12 hours after morning dose administration on day 1.

Table 28: trarschloramine PK parameters on day 1 by KarXT 50/20 BID (all cohorts)

Figure 48 shows the mean (± SD) trospium PK concentrations produced by treatment on day 3 for the PK population, and table 29 summarizes these parameters. For all treatment groups, the concentration of trospium in samples collected before the morning dose of study drug administered on day 3 and at all time points from 3 morning dose administration to 12 hours was quantifiable, except for one subject, whose plasma concentration of trospium at 12 hours post-dose was quantifiable<20.0 pg/mL. The variability range between subjects for T across the four treatment groupsMaximum ofIs 0.0% to 83.0% (CV%) for CMaximum ofIs 54.8% to 80.7% (geometric CV%) for t1/2Is 9.1% to 34.0% (CV%) for AUC0-12hrIs 59.0% to 67.6% (geometric CV%).

Table 29: trospimine PK parameters generated by treatment on day 3

Median T of trospium chloramine on day 3 for groups KarXT 100/20 BID, KarXT 125/40 BID, KarXT 150/20 BID and KarXT 150/40BIDMaximum ofIs 1.0 hour. Across 4 treatment groups, single TMaximum ofValues range from 1.0 to 6.0 hours. Median trospiam t at day 3 across 4 treatment groups1/2Are similar in value; median value t1/2The range is 4.1 to 4.8 hours. Across 4 treatment groups, single t1/2Values range from 2.8 to 9.0 hours.

When BID administration was performed for KarXT, the day 3 dose normalized GM exposure for trospimine increased as the trospimine dose was increased from 20mg (cohort 2) to 40mg (cohort 3) without changing the xanomeline dose (150 mg). Comparison of trospium exposure at day 3 after administration of 20mg of trospium BID and either 100mg (cohort 1) or 150mg (cohort 2) of xanomeline BID indicates that a dose of 20mg of BID of trospium was used compared to 150mg of xanomeline BIDGM C of trospium chloride when s-chloramine is administered with 100mg xanomeline BIDMaximum of、AUC0-lastAnd AUC0-12hrAnd is larger.

Similarly, comparison of trospium exposure after administration of 40mg of trospium BID and 125mg (cohort 4) or 150mg (cohort 3) of xanomeline BID indicates that GM C of trospium was present when trospium was administered on day 3 with 125 and 150mg of xanomeline BIDMaximum of、AUC0-lastAnd AUC0-12hrSubstantially similar.

Figure 49 shows the mean (± SD) trospium PK concentrations produced by treatment on day 7 for the PK population, which parameters are summarized in table 30. For the KarXT 100/20 BID, KarXT 125/40 BID, and KarXT 150/40BID groups, the concentration of trospium in samples collected before the morning dose of study drug administered on day 7 and at all time points from the morning dose to 12 hours on day 7 was quantifiable. Across the KarXT 100/20 BID, KarXT 150/40BID and KarXT 125/40 BID groups, the variability range between subjects was for TMaximum ofIs 0.0% to 86.3% (CV%) for CMaximum ofIs 51.2% to 93.8% (geometric CV%) for t1/2Is 23.0% to 44.5% (CV%) for AUC0-12hrIs 59.4% to 76.7% (geometric CV%).

Table 30: trospimine PK parameters generated by treatment on day 7

Median T of trospium chloramine on day 7 for KarXT 100/20 BID, KarXT 125/40 BID and KarXT 150/40BID treatmentsMaximum ofIs 1.0 hour. Across groups of KarXT 100/20 BID, KarXT 150/40BID and KarXT 125/40 BID, a single TMaximum ofThe value ranges from 0.0 to 6.0 hours.

Day 7 trass for the KarXT 100/20 BID (4.9 hours) and KarXT 125/40 BID (4.5 hours) groupsMedian chloramine value t1/2Similarly. Median t of KarXT 150/40BID group1/2Was 7.1 hours. Across groups of KarXT 100/20 BID, KarXT 150/40BID and KarXT 125/40 BID, a single t1/2The value ranges from 3.1 to 11.9 hours.

As observed on day 3, comparison of the day 7 trospium exposure following administration of 40mg of trospium BID and 125mg (cohort 4) or 150mg (cohort 3) of xanomeline BID indicates that GM C of trospium when trospium is administered with 125 and 150mg of xanomeline BIDMaximum of、AUC0-Finally, theAnd AUC0-12hrSimilarly.

Table 31 summarizes the rate of accumulation of trospimine PK by treatment of the PK population (day 7/day 3; day 7/day 1). According to the average trospimine PK accumulation ratio, trospimine accumulated minimally in plasma from day 3 to day 7 after administration of KarXT 100/20 BID (cohort 1), with little or no accumulation after administration of KarXT 125/40 BID (cohort 4) and KarXT 150/40BID (cohort 3). Both subjects showed lower exposure at day 7 compared to day 3 in the KarXT 100/20 BID group.

The accumulation ratios varied greatly between subjects in the KarXT 125/40 BID and KarXT 150/20 BID groups from day 3 to day 7. The average accumulation ratio ranges from 108.6% to 141.4% for RAUC, and for RCMaximum ofIs from 111.0% to 135.8%. For the KarXT 100/20 BID group, trospium accumulated slightly in plasma from day 1 to day 7. All subjects showed higher exposure to trospium on day 7 than on day 1, except for one subject. The average accumulation ratio was 348.7% for RAUC and for RCMaximum ofIs 379.9%. The possible effect of an increased dose of xanomeline (50 mg BID to 100mg BID from day 3) on PK and trospium bioavailability cannot be excluded as this leads to an increased exposure from day 1 to day 7.

Table 31: rate of accumulation of trospium PK by treatment (day 7/day 3; day 7/day 1)

Figure 50 compares the mean (± SD) trospiam PK concentration-time profiles for treatment and visit (days) by PK population. Figure 51 shows the mean (± SD) trospiam PK trough concentrations by PK population treatment and visit (days). Steady state was not assessed.

Example 7-Trospium pharmacokinetics of KAR-003 in comparison with KAR-001

Comparing the trospium GM exposure on day 1 of KAR-001 (table 33) (first dose of trospium alone without any prior treatment) and KAR-003 (table 32) (first dose of xanomeline + trospium without prior treatment) showed about 2.1 to 2.5 times higher trospium exposure from KAR-003 than that obtained from KAR-001. Although the comparison of GM exposure on day 3 between studies was not a true head-to-head comparison (xanomeline dosing started only on day 3 of the KAR-003 study), the number of trospium doses and daily doses administered to the subjects were the same. The day 3 GM trospium exposure of KAR-003 (Table 32) was also about 2.4-3.3 times higher than that obtained from KAR-001 (Table 33). Comparison of the day 7 GM exposure from the KarXT 100/20 BID cohort (cohort 1) of KAR-003 (Table 32) with the day 9 GM exposure of the xanomeline + trospium group from KAR-001 (Table 33) again indicated that exposure was higher (approximately 3.5-4.3 fold higher) from KAR-001.

Median value of trospium T on days 3 and 7 for the group KaR-003, KarXT 100/20 BID and on days 3 and 9 for the group KAR-001, xanomeline + trospiumMaximum ofIs 1.0 hour. Median Trospium T on day 1 for the KarXT 50/20 BID group (KAR-003)Maximum ofLower (1.0 hours), in contrast to the median value T of trospium on day 1 for the trospium only group (KAR-001)Maximum ofWas 3.0 hours.

Table 32 summarizes the subset of KAR-003 trospium PK parameters for the PK population, on day 1 for KarXT 50/20 BID treatment (all cohorts) and on days 3 and 7 for KarXT 100/20 BID treatment. Table 33 summarizes the subset of KAR-001 trospiam PK parameters for the PK population on day 1 for trospiam treatment alone and on days 3 and 9 for xanomeline + trospiam treatment.

Table 32: subsets of the KAR-003 trospium PK parameters for KarXT 50/20 BID (all cohorts) at day 1 and KarXT 100/20 BID at day 3 and day 7

Table 33: subsets of the Trospiam PK parameters for KAR-001 on days 1, 3, and 9

Table 34 lists the incidence of cholinergic TEAEs by Systemic Organ Class (SOC) and preferences for the safety population in the KAR-001 study. The overall subject incidence of cholinergic TEAEs was similar between the xanomeline + trospium group (12[ 34.3% ] subjects), the KarXT 100/20 BID group (7[ 38.9% ] subjects) and the KarXT 125/40 BID group (6 [ 33.3% ] subjects) in KAR-001.

Table 34: KAR-001 cholinergic therapy-incidence of emergent adverse events (by systemic organ categories) and preference-safety group

The subject incidence of excessive salivation, hyperhidrosis, and diarrhea was higher in the xanomeline + trospium group in KAR-001 compared to the KarXT 100/20 BID and KarXT 125/40 BID groups. Salivation occurred in 25.7% of subjects in the xanomeline + trospium group of KAR-001, 5.6% of subjects in the KarXT 100/20 BID group and none of the subjects in the KarXT 125/40 BID group. Hyperhidrosis occurs in 20.0% of the xanomeline + trospium group of KAR-001, 5.6% of the KarXT 100/20 BID group and 11.1% of the KarXT 125/40 BID group. Diarrhea occurred in 5.7% of subjects in the xanomeline + trospium group of KAR-001, and no subjects in either the KarXT 100/20 BID group or the KarXT 125/40 BID group developed diarrhea.

There was no other significant trend for the xanomeline + trospium group in KAR-001 for nausea and vomiting compared to the KarXT 100/20 BID and KarXT 125/40 BID groups. Nausea occurred in 17.1% of the xanomeline + trospium group of KAR-001 and 22.2% of each of the KarXT 100/20 BID and KarXT 125/40 BID groups. Emesis occurred in 5.7% of the xanomeline + trospium group of KAR-001, 27.8% of the KarXT 100/20 BID group and 5.6% of the KarXT 125/40 BID group.

Following oral administration of the KAR-003 formulation at all doses, the xanomeline and trospium chloride were well absorbed into the systemic circulation. The PK results indicate that neither xanomeline nor trospium significantly affected the PK behavior of the other drug. The KAR-003 formulation provides enhanced blood levels of xanomeline and trospium compared to KAR-001 where the two compounds are administered separately.

No new safety signals were reported with the KarXT formulation. All TEAEs were mild or moderate in severity with no SAE or death. The subject incidence of excessive salivation, hyperhidrosis, and diarrhea was higher in the xanomeline + trospium group in KAR-001 compared to the KarXT 100/20 BID and KarXT 125/40 BID groups in KAR-003.

The foregoing description is given for clearness of understanding only, and no unnecessary limitations should be understood therefrom, as modifications within the scope of the disclosure may be apparent to those having ordinary skill in the art. Throughout the specification, when a composition is described as including components or materials, it is contemplated that the composition can also consist essentially of, or consist of, any combination of the listed components or materials, unless otherwise specified. Likewise, where a method is described as comprising steps, it is contemplated that the method can also consist essentially of, or consist of, any combination of the recited steps, unless otherwise specified. The present disclosure illustratively disclosed herein suitably may be practiced in the absence of any element or step which is not specifically disclosed herein.

Practice of the methods disclosed herein and the various steps thereof can be performed manually and/or with automation provided by electronic devices. Although the method has been described with reference to embodiments, persons of ordinary skill in the art will readily appreciate that other ways of performing the actions associated with the method may be used. For example, unless otherwise specified, the order of individual steps may be changed without departing from the scope or spirit of the method. In addition, some of the individual steps may be combined, omitted, or further subdivided into additional steps.

It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. All combinations of the embodiments with respect to chemical groups represented by the variables contained in the general formulae described herein are specifically contemplated by the present invention as if each combination were specifically and individually stated to the extent that such combinations comprise stable compounds (i.e., compounds that can be isolated, characterized, and tested for biological activity). In addition, all subcombinations of the chemical groups listed in the examples describing such variables, as well as all subcombinations of uses and medical indications described herein, are also specifically described in this disclosure as if each subcombination of chemical groups and subcombination of uses and medical indications were individually and explicitly recited herein.

All patents, publications, and references cited herein are incorporated by reference in their entirety. In the event of a conflict between the present disclosure and an incorporated patent, publication, or reference, the present disclosure controls.

107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:防治NITM 的药物组合物及其医药用途

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!