For the treatment of cachexia

文档序号:1145121 发布日期:2020-09-11 浏览:18次 中文

阅读说明:本技术 用于恶病质的治疗 (For the treatment of cachexia ) 是由 张领兵 张东旭 于 2018-01-15 设计创作,主要内容包括:提供了用于治疗恶病质以便降低慢性疾病的发病率和死亡率并且总体上改善治疗慢性疾病的有效性的方法和组合物。更具体地,本文提供了通过调节免疫系统和/或炎症的剂来治疗恶病质的方法和组合物。(Methods and compositions for treating cachexia to reduce morbidity and mortality of chronic disease and generally improve the effectiveness of treating chronic disease are provided. More specifically, provided herein are methods and compositions for treating cachexia by agents that modulate the immune system and/or inflammation.)

1. A method for treating an immune disorder characterized by (1) increased leukocyte counts, (2) increased neutrophil to leukocyte ratios, and (3) decreased T cell to leukocyte ratios, comprising administering to a subject in need thereof an effective amount of (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both.

2. A method for treating cachexia, said method comprising administering to a subject in need thereof an effective amount of an agent selected from the group consisting of: (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both.

3. The method of claim 1 or 2, wherein the immune disorder is further characterized by wasting symptoms.

4. The method of claim 3, wherein the wasting symptoms comprise a fluid corrected weight loss and at least three or more symptoms selected from the group consisting of: reduced muscle strength, fatigue, anorexia, low body mass index to fat reduction, and abnormal biochemistry as manifested by increased inflammatory markers, anemia, or low serum albumin, and any combination thereof.

5. The method of any one of claims 1, 3, or 4, wherein the immune disorder is induced by a chronic disease.

6. The method of claim 5, wherein the chronic disease is selected from the group consisting of: AIDS, chronic obstructive pulmonary disease, multiple sclerosis, congestive heart failure, tuberculosis, familial amyloid polyneuropathy, chronic kidney disease, and cystic fibrosis.

7. The method of claim 5, wherein the chronic disease is cancer.

8. The method of any one of claims 1-7, wherein the immunosuppressive agent is a pyrimidine nucleoside antimetabolite.

9. The method of claim 8, wherein the nucleoside antimetabolite is according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof,

wherein:

y is N or C;

r1 and R2 are independently selected from the group consisting of: hydrogen, aliphatic C2-C5-acyl, benzoyl and carboxy-C1-C3-alkylcarbonyl;

r3 is selected from the group consisting of: H. acyl esters of aliphatic C1-C24-acyl, benzoyl, carboxy-C1-C3-alkylcarbonyl, phosphate, alkylphosphate, phosphoramidate, alkylphosphate, 2, 3-dihydroxypropyl phosphate and 2, 3-dihydroxypropyl phosphate optionally containing 1-2 double bonds; and is

R4 is selected from the group consisting of: H. optionally substituted C1-C24 alkyl optionally containing 1-2 double bonds, optionally substituted aliphatic C1-C24 acyl optionally containing 1-2 double bonds, valinyl, leucyl, isoleucyl, aspartyl, benzoyl and 4-methoxybenzoyl.

10. The method of claim 8 or 9, wherein the pyrimidine nucleoside analog is cytarabine or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof.

11. The method of claim 10, wherein said pyrimidine nucleoside analog is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine.

12. The method of claim 11, wherein the pyrimidine nucleoside analog is cytarabine or cytarabine hydrochloride.

13. The method of any one of claims 1-12, wherein the anti-inflammatory agent is an NSAID.

14. The method of claim 13, wherein the NSAID is a non-selective COX inhibitor.

15. The method of claim 14, wherein the non-selective COX inhibitor is an acetic acid derivative.

16. The method of claim 15, wherein the acetic acid derivative is indomethacin, diclofenac, tolmetin, aceclofenac, sulindac, nabumetone, etodolac, ketorolac, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof.

17. The method of claim 16, wherein the acetic acid derivative is ketorolac or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof.

18. The method of claim 17, wherein said acetic acid derivative is a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug of ketorolac selected from the group consisting of: ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl esters, ketorolac piperazinyl alkyl esters, and ketorolac amides.

19. The method of claim 17, wherein the acetic acid derivative is according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof

Figure FDA0002562372200000031

Wherein:

x1 is selected from the group consisting of: H. a linear or branched, saturated or unsaturated C1-C20 aliphatic radical, optionally substituted by a C6-C10 aryl radical, -NHC6H5, -NHC6H5OCH3, -NHCH2C6H5, -NHCH26H4OCH3, -NHCH2COOCH3, -NHCH2CH2COOC2H5, -NHCH2(CH2)2CH3, -NHC6H10, -NHCH2CH2CH3, -NHCH (CH3)2, -NH (CH2)3OCH3, -NHCH2CH ═ CH2, optionally substituted talosyl, optionally substituted galactosyl, optionally substituted idosyl, optionally substituted gulosyl, optionally substituted mannosyl, optionally substituted glucosyl, optionally substituted altrose, optionally substituted allosyl, optionally substituted alkylpiperidinyl, optionally substituted piperazinyl, optionally substituted alkylpiperazinyl, optionally substituted morpholinyl, and optionally substituted alkylmorpholinyl.

20. The method of claim 19, wherein the acetic acid derivative according to formula III is formula V or a pharmaceutically acceptable salt, solvate, hydrate, or clathrate thereof

Figure FDA0002562372200000041

Wherein:

x1 is selected from the group consisting of: H. a linear or branched, saturated or unsaturated C1-C20 aliphatic radical, optionally substituted by a C6-C10 aryl radical, -NHC6H5, -NHC6H5OCH3, -NHCH2C6H5, -NHCH26H4OCH3, -NHCH2COOCH3, -NHCH2CH2COOC2H5, -NHCH2(CH2)2CH3, -NHC6H10, -NHCH2CH2CH3, -NHCH (CH3)2, -NH (CH2)3OCH3, -NHCH2CH ═ CH2, optionally substituted talosyl, optionally substituted galactosyl, optionally substituted idosyl, optionally substituted gulosyl, optionally substituted mannosyl, optionally substituted glucosyl, optionally substituted altrose, optionally substituted allosyl, optionally substituted alkylpiperidinyl, optionally substituted piperazinyl, optionally substituted alkylpiperazinyl, optionally substituted morpholinyl, and optionally substituted alkylmorpholinyl.

21. The process of claim 20, wherein the acetic acid derivative according to formula V is (R) -ketorolac.

22. The method of any one of claims 1-21, comprising administering to a subject in need thereof an effective amount of an immunosuppressive and anti-inflammatory agent.

23. The method of claim 22, wherein the immunosuppressive agent is according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, and the anti-inflammatory agent is according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof.

24. The method of claim 23, wherein the immunosuppressive agent is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine, and the anti-inflammatory agent is ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl ester, ketorolac piperazinyl alkyl ester, or ketorolac amide.

25. The method of claim 24, wherein the immunosuppressive agent is cytarabine or cytarabine hydrochloride and the anti-inflammatory agent is ketorolac or ketorolac tromethamine.

26. The method of any one of claims 22-25, wherein the immunosuppressive agent and the anti-inflammatory agent are administered in a molar ratio of about 20:1 to about 0.8: 1.

27. The method of any one of claims 22-26, wherein the immunosuppressive agent and the anti-inflammatory agent are administered in the same composition.

28. The method of any one of claims 22-27, wherein the immunosuppressive agent and the anti-inflammatory agent are administered in separate compositions.

29. The method of claim 22, wherein the immunosuppressive agent and the anti-inflammatory agent are administered via separate routes of administration.

30. The method of claim 22, wherein the immunosuppressive agent and the anti-inflammatory agent are administered within 24 hours of each other.

31. The method of any one of claims 1-30, wherein one or more of the immunosuppressive agent and the anti-inflammatory agent are administered orally.

32. The method of any one of claims 1-30, wherein one or more of the immunosuppressive agent and the anti-inflammatory agent are administered parenterally.

33. The method of any one of claims 1-32, wherein the method:

(a) effective to reduce the amount of leukocytes in the subject by at least about 10%;

(b) effectively reducing the neutrophil to leukocyte ratio of the subject by at least about 10%; and

(c) effectively increasing the T cell to leukocyte ratio of the subject by at least about 10%.

34. The method of any one of claims 1-33, wherein the method is effective to increase the subject's body weight by at least about 10%.

35. A composition comprising a therapeutically effective amount of formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, and an anti-inflammatory agent according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof.

36. The composition of claim 35, wherein the anti-inflammatory agent according to formula III is according to formula V or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof.

37. The composition of claim 36, wherein the anti-inflammatory agent according to formula V is (R) -ketorolac.

38. The composition of claims 35-37, wherein the immunosuppressive agent and the anti-inflammatory agent are present in the composition in a molar ratio of about 20:1 to about 0.8: 1.

39. The composition of any one of claims 35-38, wherein the immunosuppressive agent is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine, and the anti-inflammatory agent is ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl esters, ketorolac piperazinyl alkyl esters, or ketorolac amide.

40. The composition of claim 39, wherein the immunosuppressive agent is cytarabine or cytarabine hydrochloride and the anti-inflammatory agent is ketorolac or ketorolac tromethamine.

41. The composition of any one of claims 35-40, wherein the composition further comprises a therapeutically acceptable excipient.

42. The composition of any one of claims 35-41, wherein the composition is formulated for oral administration.

43. The composition of any one of claims 35-42, wherein the composition is formulated for parenteral administration.

44. A kit comprising a first dosage form comprising a therapeutically effective dose of an immunosuppressant according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer or clathrate thereof and a second dosage form comprising a therapeutically effective dose of an anti-inflammatory agent according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer or clathrate thereof.

45. The kit of claim 40, wherein the anti-inflammatory agent according to formula III is according to formula V or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof.

46. The kit of claim 41, wherein the anti-inflammatory agent according to formula V is (R) -ketorolac.

47. The kit of claim 44, wherein the immunosuppressive agent is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine, and the anti-inflammatory agent is ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl ester, ketorolac piperazinyl alkyl ester, or ketorolac amide.

48. The kit of claim 47, wherein the immunosuppressive agent is cytarabine, cytarabine hydrochloride, and the anti-inflammatory agent is ketorolac or ketorolac tromethamine.

49. The kit of any one of claims 44-48, wherein the dose ratio of the immunosuppressive agent and the anti-inflammatory agent in the kit is about 20:1 to about 0.8: 1.

50. The kit of any one of claims 40-49, wherein said first dosage form or said second dosage form is oral.

51. The kit of any one of claims 40-50, wherein the first dosage form or the second dosage form is parenteral.

52. A method for treating a subject suffering from a primary condition accompanied by cachexia, said method comprising alternately administering to a subject in need thereof: (i) an effective amount of an immunosuppressive agent according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, and an effective amount of an anti-inflammatory agent according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof; and (ii) an additional agent in an amount effective for treating the primary condition, wherein the primary condition is cancer.

53. The method of claim 52, wherein the immunosuppressive agent is cytarabine or cytarabine hydrochloride and the anti-inflammatory agent is ketorolac or ketorolac tromethamine.

54. The method of claim 52, wherein the anti-inflammatory agent according to formula III is formula V or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof.

55. The method of claim 54, wherein the anti-inflammatory agent according to formula V is (R) -ketorolac.

56. The method of claim 45 or 46, wherein the immunosuppressive agent and the anti-inflammatory agent are administered within 24 hours of each other.

57. The method of any one of claims 45-48, wherein (i) and (ii) are separated by a non-treatment interval of at least 1 week.

Background

Cachexia is a complex metabolic syndrome associated with an underlying disease and characterized by muscle loss (most commonly manifested as weight loss) with or without loss of fat mass (fatglass). Cachexia is distinguished from hunger, age-related loss of muscle mass, primary depression, malabsorption and hyperthyroidism and is associated with increased incidence due to disease. About half of all cancer patients show a syndrome of cachexia, which contributes to decreased treatment success (treatment success) and survival. Cancer cachexia patients experience a variety of complications such as reduced effectiveness of chemotherapy, mobility, and function of the muscular system (e.g., cardiovascular and respiratory systems).

Cachexia remains a largely underestimated and untreated condition. The prevalence of cachexia (prevalence) in cancer patients can reach up to 86% in the last 1-2 weeks of life, and 45% of cancer patients lose more than 10% of their original body weight in the course of their disease. Since death usually occurs after weight loss has reached 30% of the patient's historical body weight, cachexia is a contributing factor to a number of cancer deaths. To date, there is no single approved treatment for cachexia.

Summary of The Invention

In view of the foregoing, there is a need for methods and compositions for treating cachexia so as to reduce morbidity and mortality of chronic diseases and generally improve the effectiveness of treating chronic diseases.

In one aspect, the invention provides a method for treating an immune disorder characterized by (1) increased leukocyte count, (2) increased neutrophil to leukocyte ratio and (3) decreased T cell to leukocyte ratio, comprising administering to a subject in need thereof an effective amount of (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both. In one aspect, the present invention provides a method for treating cachexia, said method comprising administering to a subject in need thereof an effective amount of an agent selected from the group consisting of: (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both. In certain embodiments, the immune disorder is further characterized by wasting symptoms (wasting symptoms). In certain embodiments, the wasting symptoms comprise fluid-corrected weight loss and at least three or more symptoms selected from the group consisting of: reduced muscle strength, fatigue, anorexia, low fat-free mass index (fat-free mass index), and abnormal biochemistry as manifested by increased inflammatory markers, anemia, or low serum albumin, and any combination thereof. In certain embodiments, the immune disorder is induced by a chronic disease. In certain embodiments, the chronic disease is selected from the group consisting of: AIDS, chronic obstructive pulmonary disease, multiple sclerosis, congestive heart failure, tuberculosis, familial amyloid polyneuropathy (familial amyloid polyneuropathy), chronic kidney disease, and cystic fibrosis. In certain embodiments, the chronic disease is cancer. In certain embodiments, the immunosuppressive agent is a pyrimidine nucleoside antimetabolite. In certain embodiments, the nucleoside antimetabolite is according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof

Wherein:

y is N or C;

r1 and R2 are independently selected from the group consisting of: hydrogen, aliphatic C2-C5-acyl, benzoyl and carboxy-C1-C3-alkylcarbonyl;

r3 is selected from the group consisting of: H. acyl esters of aliphatic C1-C24-acyl, benzoyl, carboxy-C1-C3-alkylcarbonyl, phosphate, alkylphosphate, phosphoramidate, alkylphosphate, 2, 3-dihydroxypropyl phosphate and 2, 3-dihydroxypropyl phosphate optionally containing 1-2 double bonds; and is

R4 is selected from the group consisting of: H. optionally substituted C1-C24 alkyl optionally containing 1-2 double bonds, optionally substituted aliphatic C1-C24 acyl optionally containing 1-2 double bonds, valinyl, leucyl, isoleucyl, aspartyl, benzoyl and 4-methoxybenzoyl.

In certain embodiments, the pyrimidine nucleoside analog is cytarabine or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the pyrimidine nucleoside analog is cytarabine, cytarabine hydrochloride, valyl cytarabine, Elacytarabine (lacytarabine) (CP-4055), cytarabine phosphoramidate, or Astarabine. In certain embodiments, the pyrimidine nucleoside analog is cytarabine or cytarabine hydrochloride. In certain embodiments, the anti-inflammatory agent is an NSAID. In certain embodiments, the NSAID is a non-selective COX inhibitor. In certain embodiments, the non-selective COX inhibitor is an acetic acid derivative. In certain embodiments, the acetic acid derivative is indomethacin (indomethacin), diclofenac (diclofenac), tolmetin (tolmetin), aceclofenac (aceclofenac), sulindac (sulindac), nabumetone (nabumetone), etodolac (etodolac), ketorolac (ketorolac), or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the acetic acid derivative is ketorolac or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the acetic acid derivative is a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug of ketorolac selected from the group consisting of: ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl esters, ketorolac piperazinyl alkyl esters, and ketorolac amides. In certain embodiments, the acetic acid derivative is according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof

Figure BDA0002562372210000031

Wherein:

x1 is selected from the group consisting of: H. linear or branched, saturated or unsaturated C1-C20 aliphatic group optionally substituted by C6-C10 aryl group, -NHC6H5, -NHC6H5OCH3, -NHCH2C6H5, -NHCH26H4OCH3, -NHCH2COOCH3, -NHCH2CH2COOC2H5, -NHCH2(CH2)2CH3, -NHC6H10, -NHCH2CH 3, -NHCH (CH3)2, -NH (CH2)3OCH3, -NHCH2CH ═ CH2, optionally substituted talosyl (talosyl), optionally substituted galactosyl, optionally substituted idosyl (idosyl), optionally substituted gulosyl (gulosyl), optionally substituted mannosyl, optionally substituted glucosyl, optionally substituted alterosyl (altosyl), optionally substituted axosyl (alsylosyl), optionally substituted piperazinyl, An optionally substituted morpholinyl group and an optionally substituted alkylmorpholinyl group.

In certain embodiments, the acetic acid derivative according to formula III is formula V or a pharmaceutically acceptable salt, solvate, hydrate or clathrate thereof

Figure BDA0002562372210000041

Wherein:

x1 is selected from the group consisting of: H. a linear or branched, saturated or unsaturated C1-C20 aliphatic radical, optionally substituted by a C6-C10 aryl radical, -NHC6H5, -NHC6H5OCH3, -NHCH2C6H5, -NHCH26H4OCH3, -NHCH2COOCH3, -NHCH2CH2COOC2H5, -NHCH2(CH2)2CH3, -NHC6H10, -NHCH2CH2CH3, -NHCH (CH3)2, -NH (CH2)3OCH3, -NHCH2CH ═ CH2, optionally substituted talosyl, optionally substituted galactosyl, optionally substituted idosyl, optionally substituted gulosyl, optionally substituted mannosyl, optionally substituted glucosyl, optionally substituted altrose, optionally substituted allosyl, optionally substituted alkylpiperidinyl, optionally substituted piperazinyl, optionally substituted alkylpiperazinyl, optionally substituted morpholinyl, and optionally substituted alkylmorpholinyl.

In certain embodiments, the acetic acid derivative according to formula V is (R) -ketorolac.

In certain embodiments, the methods further comprise administering to a subject in need thereof an effective amount of an immunosuppressive agent and an anti-inflammatory agent. In certain embodiments, the immunosuppressive agent is according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, and the anti-inflammatory agent is according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof. In certain embodiments, the immunosuppressive agent is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine, and the anti-inflammatory agent is ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl ester, ketorolac piperazinyl alkyl ester, or ketorolac amide. In certain embodiments, the immunosuppressive agent is cytarabine or cytarabine hydrochloride and the anti-inflammatory agent is ketorolac or ketorolac tromethamine. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered in a molar ratio of about 20:1 to about 0.8: 1. In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered in the same composition. In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered in separate compositions. In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered via separate routes of administration. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered within 24 hours of each other. In certain embodiments, one or more of the immunosuppressive and anti-inflammatory agents are administered orally. In certain embodiments, one or more of the immunosuppressive and anti-inflammatory agents are administered parenterally. In certain embodiments, the method comprises:

(a) effective to reduce the amount of leukocytes in the subject by at least about 10%;

(b) effectively reducing the neutrophil to leukocyte ratio of the subject by at least about 10%; and

(c) the subject's T cell to leukocyte ratio is effectively increased by at least about 10%. In certain embodiments, the method is effective to increase body weight of the subject by at least about 10%.

In one aspect, the present invention provides a composition comprising a therapeutically effective amount of formula I, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, and an anti-inflammatory agent according to formula III, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof. In certain embodiments, the anti-inflammatory agent according to formula III is according to formula V or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof. In certain embodiments, the anti-inflammatory agent according to formula V is (R) -ketorolac. In certain embodiments, the immunosuppressive and anti-inflammatory agents are present in the composition in a molar ratio of about 20:1 to about 0.8: 1. In certain embodiments, the immunosuppressive agent is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine, and the anti-inflammatory agent is ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl ester, ketorolac piperazinyl alkyl ester, or ketorolac amide. In certain embodiments, the immunosuppressive agent is cytarabine or cytarabine hydrochloride and the anti-inflammatory agent is ketorolac or ketorolac tromethamine. In certain embodiments, the composition further comprises a therapeutically acceptable excipient. In certain embodiments, the composition is formulated for oral administration. In certain embodiments, the composition is formulated for parenteral administration.

In one aspect, the invention provides a kit comprising a first dosage form comprising a therapeutically effective dose of an immunosuppressant according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer or clathrate thereof and a second dosage form comprising a therapeutically effective dose of an anti-inflammatory agent according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer or clathrate thereof. In certain embodiments, the anti-inflammatory agent according to formula III is according to formula V or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof. In certain embodiments, the anti-inflammatory agent according to formula V is (R) -ketorolac. In certain embodiments, the immunosuppressive agent is cytarabine, cytarabine hydrochloride, valyl cytarabine, elacytarabine (CP-4055), cytarabine phosphoramidate, or Astarabine, and the anti-inflammatory agent is ketorolac, ketorolac tromethamine, galactosylated ketorolac, ketorolac alkyl ester, ketorolac piperazinyl alkyl ester, or ketorolac amide. In certain embodiments, the immunosuppressive agent is cytarabine, cytarabine hydrochloride, and the anti-inflammatory agent is ketorolac or ketorolac tromethamine. In certain embodiments, the dose ratio of the immunosuppressive and anti-inflammatory agents in the kit is about 20:1 to about 0.8: 1. In certain embodiments, the first dosage form or the second dosage form is oral. In certain embodiments, the first dosage form or the second dosage form is parenteral.

In one aspect, the present invention provides a method for treating a subject suffering from a primary condition associated with cachexia, said method comprising administering alternately to a subject in need thereof: (i) an effective amount of an immunosuppressive agent according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, and an effective amount of an anti-inflammatory agent according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof; and (ii) an additional agent in an amount effective for treating the primary condition, wherein the primary condition is cancer. In certain embodiments, the immunosuppressive agent is cytarabine or cytarabine hydrochloride and the anti-inflammatory agent is ketorolac or ketorolac tromethamine. In certain embodiments, the anti-inflammatory agent according to formula III is formula V or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof. In certain embodiments, the anti-inflammatory agent according to formula V is (R) -ketorolac. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered within 24 hours of each other. In certain embodiments, (i) and (ii) are separated by a non-treatment interval of at least 1 week.

Is incorporated by reference

All publications, patents and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference.

Brief Description of Drawings

The novel features believed characteristic of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:

figure 1 shows that mice under cachexia have a significantly elevated total leukocyte count caused by increased Gr1+/CD11b + neutrophils and decreased CD3+ lymphocytes; (A) is a Balb/c mouse depicting cachexia relative to a Balb/c mouse that has not been subjected to an experiment (Balb/c mie), and (B) is a graph depicting the ratio of CD3+ lymphocytes and Gr1+/CD11B + neutrophils to total leukocytes in naive Balb/c mice versus cachectic Balb/c mice.

Figure 2 shows that (a) cytarabine, (B) ketorolac, and (C) a combination of cytarabine and ketorolac reversed the weight loss in cachectic mice; (A) is a graph showing the individual body weight of cachectic Balb/C mice injected with C26 tumor cells after treatment with 0.3mg cytarabine, (B) is a graph showing the individual body weight of cachectic Balb/C mice injected with C26 tumor cells after treatment with 0.1mg ketorolac, and (C) is a graph showing the average body weight of cachectic Balb/C mice injected with C26 tumor cells after treatment with 0.3mg cytarabine (●, n ═ 7), 0.1mg ketorolac (@, n ═ 7), or a combination (0.3mg cytarabine +0.1mg ketorolac, ■, n ═ 7) ± s.e.m. and the average body weight of untreated mice (═ 7).

FIG. 3 shows that the combination of (A) cytarabine and (B) cytarabine + ketorolac prolongs survival of cachectic mice; (A) is a Kaplan-Meier survival curve for C26 tumor cell injected cachectic Balb/C mice after two treatment cycles with 0.3mg cytarabine (●, n ═ 8) or PBS control (xxx, n ═ 9), and (B) is a Kaplan-Meier survival curve for C26 tumor cell injected cachectic Balb/C mice after two treatment cycles with 0.3mg cytarabine (●, n ═ 8), 0.1mg ketorolac (@, n ═ 9), combination (■, n ═ 8) or PBS control (n ═ 9).

Figure 4 demonstrates that cytarabine and ketorolac in combination reverse cancer cachexia in mice not by inhibiting tumor growth but by correcting underlying immune disorders; (A) mice inoculated with C26 tumor cells were treated with cytarabine: graphs of mean tumor size ± SD of 4 injections of ketorolac combination (6:2 cytarabine/ketorolac dose ratio), (B), (C), (D) and (E) are graphs of mice treated with cytarabine: graphs plotting multiple combinations of dose ratios of ketorolac versus total leukocyte population or T lymphocyte (CD3+) or neutrophil (Gr1+/CD11b +) ratios before and after treatment.

Figure 5 demonstrates that a higher ratio of cytarabine to ketorolac further improved survival in cachectic mice; FIG. 5 is a Kaplan-Meier survival curve for mice treated with cytarabine to ketorolac ratios of 3:1, 6:1, and 9:1 for 3 treatment cycles.

FIG. 6 shows that the leukocyte response to the cytarabine/ketorolac combination correlates with the potency of the cytarabine/ketorolac combination on cancer cachexia; (A) (B), (B) and (C) are depicted as 6:1(●, ■) or 9:1 (a. thaliana, t. cit.,) cytarabine: a graph of total leukocyte population (a), T-cell to leukocyte ratio (B) or neutrophil to leukocyte ratio (C) before combination treatment, after combination treatment and after recurrence of cachexia, and corresponding levels in cachexia-free mice (corresponding levels in cachexia-free mice are represented by reference lines) for ketorolac-treated individual mice. Figure 6 shows that recovery of these parameters is achieved after combination therapy.

Figure 7 shows cytarabine: ketorolac therapy can be used multiple times over the course of cancer therapy to counteract cachexia. Panels (a), (B) and (C) show graphs of the body weight of individual mice, wherein the time of cytarabine/ketorolac treatment is depicted by an arrow.

Figure 8 demonstrates that (R) -ketorolac alone can reverse weight loss and prolong survival in cachectic mice. (A) Is a graph showing the mean body weight ± s.e.m. of cachectic mice treated with 0.05mg (r) -ketorolac (●, n ═ 5) or PBS control (■, n ═ 5). (B) Is a Kaplan-Meier survival curve for cachexia mice treated with 0.05mg of (r) -ketorolac (●, n ═ 7) or PBS control (■, n ═ 5).

Detailed Description

I.Definition of

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

The terms "treat," "treating," or "treatment" as used herein may include alleviating, reducing, or alleviating a symptom of a disease or condition, preventing an additional symptom, inhibiting a disease or condition, e.g., prophylactically and/or therapeutically arresting the development of a disease or condition, alleviating a disease or condition, causing regression of a disease or condition, alleviating a condition caused by a disease or condition, or stopping a symptom of a disease or condition.

The term "primary disease" or "primary disease associated with cachexia" refers to a disease known in the medical literature to be associated with cachexia; in certain embodiments, the primary disease associated with cachexia is chronic/severe organ dysfunction (e.g., congestive heart failure), chronic infectious disease (e.g., AIDS), autoimmune disease (e.g., Crohn's disease), or cancer. As used herein, the term "treating a primary disease associated with cachexia" does not necessarily relate to regression of the primary disease, but rather to treating cachexia in a manner that facilitates treatment of the primary disease.

The term "therapeutically effective amount" or "effective amount" may generally refer to an amount (or dose) of a compound or other treatment that is minimally sufficient to prevent, reduce, treat, or eliminate the condition or risk thereof when administered to a subject in need of such compound or other treatment. In certain instances, the term "therapeutically effective amount" can refer to an amount of a compound or other treatment sufficient to have a prophylactic effect when administered to a subject. The therapeutically effective amount may vary; for example, it may vary depending on the condition of the subject, the weight and age of the subject, the severity of the disease condition, the mode of administration, and the like, all of which may be determined according to standard pharmacodynamic dosing models. The amount of the compound actually administered can be determined by a physician or caregiver depending on the relevant circumstances, including the condition to be treated, the chosen route of administration, the compound administered and its relative activity, the age, weight, response of the individual patient, the severity of the patient's symptoms, and the like.

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

The term "prodrug" is intended to indicate a compound that can be converted under physiological conditions or by solvolysis to the biologically active compounds described herein. Thus, the term "prodrug" refers to a precursor of a pharmaceutically acceptable biologically active compound. In certain aspects, the prodrug is inactive when administered to a subject, but is converted to an active compound in vivo, e.g., by hydrolysis. Prodrug compounds generally provide the advantages of solubility, histocompatibility, or delayed release in mammalian organisms (see, e.g., Bundgard, h., Design of produgs (1985), pages 7-9, pages 21-24 (Elsevier, Amsterdam); Higuchi, t. et al, "Pro-drugs as Novel delivery systems," (1987) a.c.s.symposium Series, volume 14; and Bioreversible Carriers in drug Design, Edward b.roche editions, American Pharmaceutical Association and pergammon Press), each of which is incorporated herein by reference in its entirety for the purpose of describing how a compound may be modified with a moiety. The term "prodrug" is also intended to include any covalently bonded carrier that releases the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrugs of active compounds as described herein are typically prepared by: the functional groups present in the active compound are modified in such a way that the modification is cleaved to the parent active compound in routine manipulation or in vivo. Prodrugs include compounds that: wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that cleaves to form a free hydroxy, free amino, or free sulfhydryl group, respectively, when a prodrug of the active compound is administered to a mammalian subject. Examples of prodrugs include, but are not limited to, acetate, formate, and benzoate derivatives of hydroxyl functional groups in the active compound, or acetamide, formamide, and benzamide derivatives of amine functional groups, and the like.

The term "salt" or "pharmaceutically acceptable salt" refers to salts derived from a variety of organic and inorganic counterions well known in the art. Salts include, for example, acid addition salts and base addition salts. The acid added to the compound to form an acid addition salt may be an organic acid or an inorganic acid. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, iodic acid, phosphoric acid, carbonic acid, and the like. Organic acids from which salts may be derived include, for example, acetic, propionic, glycolic, pyruvic, oxalic, maleic, malonic, malic, succinic, fumaric, tartaric, acid tartaric, citric, benzoic, cinnamic, mandelic, methanesulfonic (methanesulfonic), methanesulfonic (methylsulfonic), naphthalenesulfonic, camphorsulfonic, ethanedisulfonic (ethanedisulfonic), lauryl sulfuric, ethanesulfonic, p-toluenesulfonic, pamoic, glucoheptonic, gluconic, glucuronic, hippuric, isethionic (isethionic), lactic, lactobionic, salicylic, stearic, toluenesulfonic, phenylsulfonic acid, and the like. The base added to the compound to form a base addition salt may be an organic base or an inorganic base. In some cases, the salt may be a metal salt. In some cases, the salt may be an ammonium salt. Inorganic bases from which salts may be derived include, for example, sodium, potassium, lithium, ammonium, calcium, glycol amine, lysine, meglumine, magnesium, alcohol amine (olamine), tromethamine, iron, zinc, copper, manganese, aluminum, and the like. Organic bases from which salts can be derived include, for example, primary, secondary and tertiary amines, substituted amines (including naturally occurring substituted amines), cyclic amines, basic ion exchange resins, and the like.

The term "solvate" means a compound of the invention or a salt thereof that further comprises a stoichiometric or non-stoichiometric amount of a pharmaceutically acceptable solvent bound by non-covalent intermolecular forces. When the solvent is water, the solvate is a hydrate.

The term "clathrate" means a compound of the present invention or a salt thereof in the form of a crystal lattice (crystal lattice) comprising spaces (e.g., channels) having guest molecules (e.g., pharmaceutically acceptable solvents or water) trapped therein.

The term "stereoisomer" refers to a compound composed of the same atoms bonded by the same bonds but having different three-dimensional structures that are not interchangeable. The present disclosure contemplates a variety of stereoisomers and mixtures thereof and includes "enantiomers," which refers to two stereoisomers whose molecules are non-superimposable mirror images of each other. The optically active (+) and (-) isomers, the optically active (R) -and (S) -isomers, or the optically active (D) -and (L) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques such as chromatography and fractional crystallization. Conventional techniques for the preparation/separation of the individual enantiomers include chiral synthesis from suitable optically pure precursors, or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral High Pressure Liquid Chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless otherwise specified, it is intended that the compounds include both E and Z geometric isomers.

The term "cachexia" refers to a complex metabolic syndrome associated with an underlying disease and characterized by muscle loss with or without loss of fat mass. Prominent clinical features of cachexia are fluid-retention-corrected weight loss in adults or non-endocrine growth disorders in children. Anorexia, inflammation, insulin resistance (insulin resistance) and increased muscle proteolysis are frequently associated with cachexia. Cachexia is distinguished from hunger, age-related loss of muscle mass, primary depression, malabsorption and hyperthyroidism and is associated with increased morbidity. For cachexia in humans caused by cancer (e.g., tumors or cancers of a particular tissue type), agreed diagnostic criteria (see, e.g., Fearon et al, Lancet Oncol 2011; 12:489-95, which provides diagnostic criteria for cachexia and its stage) are that consumption has been shown (body mass index [ BMI ] based on current weight and height]<20kg/m2) Or a weight loss of greater than 5%, or greater than 2%, of individuals who have shown wasting (sarcopenia) based on skeletal muscle mass.

The term "white blood cells" or "leukocytes" refers to nucleated circulating cells of hematopoietic origin that are involved in protecting the body against both infectious diseases and foreign invaders. The term "leukocyte" encompasses a variety of subtypes of cells with different immune functions, including but not limited to granulocytes, monocytes and lymphocytes. The population of white blood cells in a blood sample from a subject can be determined by a variety of methods including counting whole diluted blood (white blood cells) directly on a hemocytometer, either manually or automatically (e.g., with a device such as Coulter S-Plus Jr), white blood cell separation by centrifugation followed by manual or automatic counting, or resuspending the blood in a red blood cell lysis buffer followed by manual or automatic counting. In certain embodiments, the white blood cells are measured in terms of absolute cell counts (e.g., the number of cells per volume of blood).

The term "T cell" refers to a subset of leukocytes (e.g., lymphocytes) that play a central role in cell-mediated immunity. T cells are distinguished from other lymphocytes (e.g., B cells) by the presence of T cell receptors on the surface of T cells, molecules responsible for recognizing antigen fragments that are peptides bound to Major Histocompatibility Complex (MHC) molecules. An exemplary method of differentiating T cells via immunoreactive methods (e.g., immunofluorescence, IHC, FACS) includes determining the presence of a co-receptor for CD3 on its surface (co-receptor). The term "T cell" encompasses multiple subtypes of cells with different immune functions, including but not limited to helper T cells (helper T-cells), cytotoxic T cells, memory T cells, regulatory T cells, natural killer T cells (natural killer T-cells), mucosa-associated invariant T cells (mucosally associated invariant T-cells), and gamma T cells. In the measurement of T cells, T cells may be expressed as absolute population size (e.g., cell count per volume of blood) or relative population size (e.g., pre-treatment versus post-treatment), as a fraction of total leukocytes, or as a ratio relative to a larger class (genus) encompassing a given cell (e.g., T cell to leukocyte ratio, or as a percentage of leukocytes for T cells).

The term "neutrophil" refers to a subset of highly motile leukocytes (e.g., granulocytes) that are dedicated to uptake of particulate material by phagocytosis and are capable of entering infected or inflamed tissue. Neutrophils can be subclassed (sub-classify) into types such as high density neutrophils and low density neutrophils. Like T cells, neutrophils can be distinguished from other leukocytes by measuring the presence of molecules on their surface by immunoreactive methods (e.g., immunofluorescence, IHC, FACS); however, the markers used differ between humans and mice. In mice, neutrophils can be differentiated, for example, by the presence of cell surface markers such as CD11b, Ly6G, and Gr 1. In humans, neutrophils can be differentiated, for example, by the presence of cell surface markers such as CD15 and CD16 and the absence of cell surface markers such as CD49 d. In the measurement of neutrophils, neutrophils may be expressed as absolute population size (e.g., cell count per volume of blood) or relative population size (e.g., pre-treatment versus post-treatment), as a fraction of total leukocytes, or as a ratio relative to a larger class encompassing a given cell (e.g., neutrophil to leukocyte ratio, or as a percentage of neutrophils).

The term "immunosuppressive agent" refers to a drug that inhibits or prevents the activity of the immune system. Immunosuppressive agents include, but are not limited to, classes such as cytostatics (e.g., alkylating agents, antimetabolites), drugs that act on immunophilin (immunophilin) (e.g., cyclosporine, tacrolimus, sirolimus), anti-lymphocyte antibodies, opioids (opioids), TNF binding agents, mycophenolate (mycophenolate), and sphingosine targeting agents (e.g., myriocin and fingolimod).

The term "anti-inflammatory agent" refers to a drug that reduces inflammation or swelling. Anti-inflammatory agents include, but are not limited to, classes such as NSAIDs (e.g., COX inhibitors such as aspirin and ibuprofen), steroidal anti-inflammatory drugs (e.g., corticosteroids), and anti-leukotrienes (anti-leukotrienes).

The term "NSAID" refers to nonsteroidal anti-inflammatory agents. NSAIDs are known to inhibit cyclooxygenase I and cyclooxygenase II (enzymes responsible for the biosynthesis of prostaglandins and certain related endocrine products). NSAIDs are known to be antipyretic, analgesic and anti-inflammatory. Exemplary NSAIDs can be found, for example, in The pharmaceutical Basis of Therapeutics, 9 th edition, Macmillan publishing Co.,1996, page 617-655.

II.Overview

The present disclosure describes a method of treating a subject suffering from a condition characterized by an immune disorder comprising one or more of the following, using (a) one or more immunosuppressive agents, (b) one or more anti-inflammatory agents, or (c) a combination of an immunological agent and an anti-inflammatory agent: (1) increased white blood cell count, (2) increased neutrophil to white blood cell ratio, and (3) decreased T cell to white blood cell ratio. In the case where a combination of an immunosuppressive agent and an anti-inflammatory agent is administered, the immunosuppressive agent and the anti-inflammatory agent may be administered simultaneously, separately, or sequentially. In certain embodiments, treatment of the underlying immune disorder provides a more direct and effective way to treat conditions believed to be caused by the underlying immune disorder, such as cachexia (and in particular cancer cachexia).

The present disclosure also describes methods of treating a subject afflicted with a primary condition associated with cachexia with an effective amount of (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both, in combination with an effective amount of an agent for treating the primary condition. In certain embodiments, the treatment of cachexia in conjunction with a primary condition enables the treatment of the primary condition with fewer treatment interruptions, side effects, and/or adverse consequences.

The present disclosure also provides compositions comprising one or more of an immunosuppressive agent and an anti-inflammatory agent. In some cases, the composition further comprises a pharmaceutically acceptable excipient.

III.Subject to be treated

A subject treated by the methods and compositions provided herein can have any of a number of chronic progressive diseases or can be suspected of having any of a number of chronic progressive diseases. Thus, the methods and compositions herein may include methods or compositions for treating a subject suffering from or suspected of suffering from a chronic disease and/or a progressive disease. Many chronic and/or progressive diseases are also accompanied by wasting syndromes, such as cachexia. Thus, the methods and compositions herein may include methods of treating subjects suffering from chronic and/or progressive diseases accompanied by wasting syndromes such as cachexia (see, e.g., Evans et al, Clinical Nutrition (2008)27, 793-.

The subject is preferably a human subject or patient, but in some cases may be a non-human subject (e.g., a non-human mammal). Examples of non-human mammals include, but are not limited to, non-human primates (e.g., apes, monkeys, and gorillas), rodents (e.g., mice, rats), cows, pigs, sheep, horses, dogs, cats, and rabbits.

The concurrence of cachexia with chronic and/or progressive disease poses significant complications for its treatment, as skeletal muscle dysfunction/loss places patients at increased risk of morbidity and mortality, even with refeeding (refeeding) and parenteral nutrition.

A major class of chronic and/or progressive diseases associated with cachexia are diseases or conditions involving severe organ dysfunction. Thus, in certain instances, a subject treated according to the methods herein has chronic obstructive pulmonary disease, congestive heart failure, chronic kidney disease, cystic fibrosis, or has suffered trauma. In certain embodiments, the subject has chronic obstructive pulmonary disease. In certain embodiments, the subject has congestive heart failure. In certain embodiments, the subject has chronic kidney disease. In certain embodiments, the subject has cystic fibrosis. In certain embodiments, the subject has suffered trauma.

Another major class of chronic and/or progressive diseases associated with cachexia is diseases with infectious etiology. In certain instances, a subject treated according to the methods herein has AIDS, sepsis, chronic infection, or tuberculosis. In certain embodiments, the subject has AIDS. In certain embodiments, the subject suffers from sepsis. In certain embodiments, the subject is suffering from a chronic infection. In certain embodiments, the subject has tuberculosis.

Other types of diseases associated with cachexia include autoimmune diseases (e.g., multiple sclerosis, rheumatoid arthritis, crohn's disease), amyloid diseases (e.g., familial amyloid polyneuropathy), and type I diabetes. In certain embodiments, a subject treated with a method or composition described herein has an autoimmune disease, an amyloid disease, or type I diabetes. In certain embodiments, the subject has multiple sclerosis. In certain embodiments, the subject has rheumatoid arthritis. In certain embodiments, the subject has crohn's disease. In certain embodiments, the subject has familial amyloid polyneuropathy. In certain embodiments, the subject has type I diabetes.

A major class of chronic progressive diseases associated with cachexia is neoplastic conditions, such as cancer. Cancer is a collection of related diseases characterized by uncontrolled proliferation of cells with the potential to metastasize throughout the body. Cancers can be classified into five broad categories, including, for example, malignancies, sarcomas, lymphomas, leukemias, and adenomas. Malignant tumors can be caused by cells that cover both internal and external parts of the body, such as the lungs, breast and colon. Sarcomas can be caused by cells located in bone, cartilage, fat, connective tissue, muscle, and other supportive tissues. Lymphomas can occur in lymph nodes and immune system tissues. Leukemias can occur in the bone marrow and accumulate in the bloodstream. Adenomas can occur in the thyroid, pituitary gland, adrenal gland and other glandular tissues.

In certain embodiments, the subject has or is suspected of having cancer or a tumorigenic syndrome. The cancer may include thymus cancer, brain cancer, lung cancer, skin cancer, eye cancer, oropharyngeal cancer, gastrointestinal cancer, pancreatic cancer, breast cancer, head cancer, neck cancer, kidney cancer, liver cancer, ovarian cancer, testicular cancer, gynecological cancer, thyroid cancer, AIDS-related cancer, or virus-induced cancer. Neoplastic conditions also include tumors (e.g., of the previously described cancer types), CNS tumors, and paraneoplastic neurological syndromes. In certain embodiments, the cancer is a thymus carcinoma. In certain embodiments, the cancer is a brain cancer (e.g., glioma, meningioma). In certain embodiments, the cancer is lung cancer (e.g., lung adenoma). In certain embodiments, the cancer is a skin cancer (e.g., squamous cell carcinoma). In certain embodiments, the cancer is an eye cancer (e.g., retinoblastoma, intraocular melanoma). In certain embodiments, the cancer is oral cancer or oropharyngeal cancer. In certain embodiments, the cancer is bladder cancer. In certain embodiments, the cancer is gastric cancer. In certain embodiments, the cancer is pancreatic cancer. In certain embodiments, the cancer is a gynecological cancer (e.g., cervical cancer, ovarian cancer). In certain embodiments, the cancer is head or neck cancer. In certain embodiments, the cancer is renal cancer. In certain embodiments, the cancer is prostate cancer. In certain embodiments, the cancer is testicular cancer. In certain embodiments, the cancer is AIDS-related cancer (e.g., Kaposi's sarcoma). In certain embodiments, the cancer is a virus-induced cancer (e.g., Burkitt's lymphoma, nasopharyngeal carcinoma caused by EBV). In certain embodiments, the tumorigenic syndrome is a CNS tumor. In certain embodiments, the tumorigenic condition is paraneoplastic neurological syndrome.

Other non-limiting examples of cancers that may be treated by the compositions or methods according to the invention include: acute lymphocytic leukemia, acute myelogenous leukemia, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphomas, anal cancers, appendiceal cancers, astrocytomas, basal cell carcinomas, bile duct cancers, bladder cancers, bone cancers, brain tumors such as cerebellar astrocytomas, cerebral astrocytomas/malignant gliomas, ependymomas, medulloblastomas, supratentorial primitive neuroectodermal tumors (supratentorial primary neuroectodermal tumors), visual acuity (Evoblastomas), neuroepithelial tumors, and the likeSensory pathway and hypothalamic glioma (visual pathway and hypothalamic glioma), breast cancer, bronchial adenoma, burkitt lymphoma, cancer of unknown primary origin, central nervous system lymphoma, cerebellar astrocytoma, cervical cancer, childhood cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, cutaneous T-cell lymphoma, desmoplastic small round cell tumor (desmoplastic small round cell tumor), endometrial cancer, ependymoma, esophageal cancer, Ewing's sarcoma, germ cell tumor, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (gastrointestintal tumor), glioma, hairy cell leukemia, heart cancer (heart cancer), hepatocellular (liver) cancer, hodgkin's lymphoma, hypopharyngeal lymphoma, melanoma, cervical cancer, pediatric lymphoma, cervical cancer, pediatric lymphoma, cervical cancer, islet cell carcinoma, kaposi's sarcoma, kidney cancer, larynx cancer (laryngel cancer), lip and oral cavity cancer, liposarcoma, liver cancer, lung cancer such as non-small cell lung cancer and small cell lung cancer, lymphoma, leukemia, macroglobulinemia, malignant fibrous histiocytoma of bone/osteosarcoma, medulloblastoma, melanoma, mesothelioma, metastatic squamous neck cancer with occult primary foci, oral cancer, multiple endocrine neoplasia syndrome, myelodysplastic syndrome, myeloid leukemia, nasal and paranasal sinus cancers, nasopharyngeal cancers, neuroblastoma, non-hodgkin's lymphoma, non-small cell lung cancer, oral cavity cancer (oral cancer), oropharyngeal cancer, malignant fibrous histiocytoma of osteosarcoma/bone, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, pancreatic cancer, pancreatic islet cells, paranasal sinus and nasal cavity cancer, parathyroid cancer, pancreatic islet cells, paranasal sinus and nasal cavity cancer, parathyroid cancer, thyroid cancer, pancreatic, Penile cancer, pharyngeal cancer, pheochromocytoma, pineal astrocytoma, pineal germ cell tumor, pituitary adenoma, pleuropulmonary blastoma, plasmacytoma formation, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell carcinoma, transitional cell carcinoma of renal pelvis and ureter, retinoblastoma, rhabdomyosarcoma, salivary gland carcinoma, sarcoma, skin carcinoma, merkel cell, small intestine carcinoma, soft tissue sarcoma, squamous cell carcinoma, stomach cancer, T-cell lymphoma, laryngeal carcinoma (throatancer), thymoma, thoracic carcinoma, thymic carcinoma, cervical carcinoma, bladder carcinomaAdenocarcinoma, thyroid carcinoma, trophoblastic tumors (gestational), cancer of unknown primary site, cancer of the urethra, sarcoma of the uterus, vaginal cancer, vulvar cancer, Waldenstrom's macroglobulinemia: (macrogolulinemia) and nephroblastoma (Wilms tumor).

The subject in need of treatment according to the methods and compositions provided herein can be male or female. The subject may include adults, adolescents, children, toddlers, infants, and newborns. Such subjects may have an age range that may include > 10 minutes of age, > 1 hour of age, > 1 day of age, > 1 month of age, > 2 months of age, > 6 months of age, > 1 year of age, > 2 years of age, > 5 years of age, > 10 years of age, > 15 years of age, > 18 years of age, > 25 years of age, > 35 years of age, > 45 years of age, > 55 years of age, > 65 years of age, > 80 years of age, < 70 years of age, < 60 years of age, < 50 years of age, < 40 years of age, < 30 years of age, < 20 years of age, or < 10 years of age. The subjects may have different genetic backgrounds, including different ethnic groups or genetically mixed populations.

IV.Therapeutic agents

The methods provided herein include administering a therapeutic agent (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) to a subject.

In certain embodiments, the methods or compositions provided herein relate to the administration of an immunosuppressive agent, either alone or in combination with an anti-inflammatory agent, or alone or comprising an immunosuppressive agent in combination with an anti-inflammatory agent. Immunosuppressive agents include several classes of agents that act by different mechanisms, including, but not limited to, cytostatics, drugs that act on immunoaffinity proteins (e.g., cyclosporine, rapamycin (rapamycin), sirolimus), antibodies that target immune cells (e.g., anti-lymphocyte antibodies, anti-thymocyte globulin, anti-lymphocyte globulin, anti-neutrophil granulocytes, etc.), opioids (e.g., fentanyl), TNF binding agents (e.g., TNF receptor analogs and anti-TNF α antibodies), mycophenolates (e.g., mycophenolic acid and mycophenolate mofetil), and sphingosine targeting agents (e.g., myriocin and fingolimod). In certain embodiments, the methods and compositions disclosed herein comprise the use of an immunosuppressive agent selected from the group consisting of: cytostatics, drugs acting on immunoaffinity proteins, anti-lymphocyte antibodies, opioids, TNF binding agents, mycophenolate mofetil and sphingosine targeting agents and any combination thereof.

Cytostatics are agents that prevent cell division and thereby inhibit immune activity by affecting the proliferation of immune cells (e.g., neutrophils, T cells, or B cells). Cytostatics can be grouped into two main categories: alkylating agents and antimetabolites. In certain embodiments, the methods and compositions disclosed herein comprise the use of an immunosuppressive agent selected from the group consisting of: an alkylating agent and an antimetabolite, and any combination thereof. In certain embodiments, the methods and compositions disclosed herein include the use of cytostatics (e.g., lymphocytostatics, leukocytostatics) that target the proliferation of neutrophils, T cells, B cells, or both.

Alkylating agents are reactive immunosuppressants that attach alkyl groups to DNA and thereby interfere with cell proliferation (e.g., cell proliferation of immune cells). The DNA alkylation characteristics of the alkylating agent may be accompanied by a number of structurally different moieties, including nitrogen/sulfur mustards, nitrosoureas, and organo-platinum compounds. In certain embodiments, the methods and compositions disclosed herein comprise the use of an immunosuppressive agent selected from the group consisting of: nitrogen/sulfur mustards, nitrosoureas, and organo-platinum compounds and any combination thereof. Nitrogen mustards/sulfur mustards include, for example, dichloromethyldiethylamine (mechlorrethamine), cyclophosphamide, Chlorambucil (chlembucil), Melphalan (Melphalan), and Ifosfamide (Ifosfamide). Nitrosoureas include, for example, Carmustine (Carmustine), Lomustine (Lomustine), arabinopyranosyl-N-methyl-N-nitrosourea, chlorouretocin (Chlorozotocin), ethylnitrosourea, fotemustine, Nimustine (Nimustine), N-nitroso-N-methylurea, Ranimustine (Ranimustine), Semustine (Semustine), and streptozotocin (streptacin), as well as any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof, and are notable for their ability to penetrate the blood-brain barrier. Organo-platinum compounds include, for example, Oxaliplatin (Oxaliplatin), Carboplatin (Carboplatin), Cisplatin (cissplatin) and Nedaplatin (Nedaplatin), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates and prodrugs thereof.

Antimetabolites are immunosuppressive agents that interfere with the synthesis of nucleic acids, and thus with cell proliferation (e.g., of immune cells). Antimetabolites include several classes of drugs with different structures and modes of action, including, but not limited to, folate antagonists and nucleotide analogs (purine analogs and pyrimidine analogs). In certain embodiments, the methods and compositions disclosed herein comprise the use of an immunosuppressive agent selected from the group consisting of: folic acid antagonists, nucleotide analogs, purine analogs, and pyrimidine analogs, and any combination thereof. Folate antagonists counteract the use or effect of folate (vitamin B9) in cells and thereby inhibit methyltransferases involved in serine, methionine, thymidine and purine biosynthesis, which are important for nucleic acid synthesis. Folic acid antagonists include compounds such as Methotrexate (Methotrexate) and Pemetrexed (Pemetrexed), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof. Purine analogs are compounds that have structural similarity to purine nitrogenous bases, which can interfere with nucleic acid synthesis by competing with endogenous purine bases and/or can counter-inhibit (retro-inhibit) enzymes responsible for purine nucleotide synthesis in cells. Purine analogs include, but are not limited to, Fludarabine (Fludarabine), Pentostatin (pentastatin), Cladribine (Cladribine), azathioprine, and mercaptopurine, and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof. In certain embodiments, the methods and compositions disclosed herein comprise the use of an immunosuppressive agent selected from the group consisting of: fludarabine, penstatin, cladribine, azathioprine and mercaptopurine and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates and prodrugs thereof. Pyrimidine analogs are compounds that have structural similarity to pyrimidine nitrogenous bases, which can interfere with nucleic acid synthesis by competing with endogenous pyrimidine bases and/or can counter-inhibit enzymes responsible for the synthesis of pyrimidine nucleotides in cells. Pyrimidine analogs include, but are not limited to, cytarabine, 5-fluorouracil, 5-fluorodeoxyuridine, Gemcitabine (Gemcitabine), Capecitabine (Capecitabine), Tegafur-uracil (Tegafur-uracil), Azacitidine (Azacitidine), and Decitabine (Decitabine) and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof (e.g., the free acid of cytarabine or a salt of cytarabine such as cytarabine hydrochloride). In certain embodiments, the methods and compositions disclosed herein comprise the use of an immunosuppressive agent selected from the group consisting of: cytarabine, 5-fluorouracil, 5-fluorodeoxyuridine, gemcitabine, capecitabine, tegafur-uracil, azacitidine and decitabine and any combination thereof, including any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates and prodrugs thereof. In particular, cytarabine encompasses many named pharmaceutically acceptable administration forms that may be used in the methods or compositions according to the invention described herein, including cytarabine, cytarabine-HCl, L-valyl-ara-C (valyl cytarabine), elacytarabine (CP-4055), CNDAC ((1- (2-C-cyano-2-deoxy-d-arabino-pentofuranosyl) cytosine), Sapacitabine, cytarabine phosphoramidate prodrugs (see, e.g., Tobias et al, molecular pharmaceuticals, 2004,1(2), p. 112 and 116), and Astaracitabine Icocitabine (CP-4055), CNDAC ((1- (2-C-cyano-2-deoxy β -d-arabino-pentofuranosyl) cytosine), Sapacitabine, Cytarabine phosphoramidate prodrugs (see, e.g., Tobias et al, Molecular pharmaceuticals, 2004,1(2), p. 112. 116), Fosteabine, Cytarabine octadecyl phosphate hydrate (Cytarabine ocfosfate hydrate), palmitoyl Cytarabine, adamantanoyl Cytarabine, Enocitabine (Enocitabase) and Astaribine, and any combination thereof.

In certain embodiments, the methods provided herein comprise administering or using an immunosuppressive agent (e.g., cytarabine derivative) according to formula I or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof

Wherein:

y is N or C;

r1 and R2 are independently selected from the group consisting of: hydrogen, aliphatic C2-C5-acyl, benzoyl and carboxy-C1-C3-alkylcarbonyl;

r3 is selected from the group consisting of: H. acyl esters of aliphatic C1-C24-acyl, benzoyl, carboxy-C1-C3-alkylcarbonyl, phosphate, alkylphosphate, phosphoramidate, alkylphosphate, 2, 3-dihydroxypropyl phosphate and 2, 3-dihydroxypropyl phosphate optionally containing 1-2 double bonds; and is

R4 is selected from the group consisting of: H. optionally substituted C1-C24 alkyl optionally containing 1-2 double bonds, optionally substituted aliphatic C1-C24 acyl optionally containing 1-2 double bonds, valinyl, leucyl, isoleucyl, aspartyl, benzoyl and 4-methoxybenzoyl.

In certain embodiments, the anti-inflammatory agent according to formula I, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, is according to formula II:

wherein:

r4 and R5 are independently selected from H, aliphatic C1-C24 acyl optionally containing 1 to 2 double bonds and branched aliphatic C1-C24 acyl optionally containing 1 to 2 double bonds.

Examples of the synthesis, chemical structure and properties of other cytarabine analogs or related compounds can be found in, for example, US5641758A, US6316425B1, Molecular pharmaceuticals by Tobias et al, 2004,1(2), p.112-116 and Acta Pharmacol Sin by Cheon et al, 2 months 2007; 28(2) 268-72.

In certain embodiments, the immunosuppressive agent is one of the compounds provided in table 1, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof:

table 1: exemplary Cytarabine related Compounds

Figure BDA0002562372210000241

In certain embodiments, the methods provided herein involve administering an anti-inflammatory agent, either alone or in combination with an immunosuppressive agent, or using an anti-inflammatory agent, either alone or in combination with an immunosuppressive agent. Anti-inflammatory agents include drugs that reduce inflammation or swelling. Anti-inflammatory agents include, but are not limited to, classes such as steroidal anti-inflammatory drugs, NSAIDs, and anti-leukotrienes. In certain embodiments, the methods and compositions disclosed herein comprise the use of an anti-inflammatory agent selected from the group consisting of: steroidal anti-inflammatory drugs, NSAIDs, and anti-leukotrienes, and any combination thereof.

Steroidal anti-inflammatory drugs (e.g., corticosteroids) are analogs of natural steroid hormones, such as cortisol, that interact with the glucocorticoid receptor, altering gene transcription to induce (transactivation) or repress (transrepression) gene transcription in both inflammatory leukocytes and structural cells, such as epithelial cells, thereby inhibiting inflammation. In certain embodiments, the methods and compositions disclosed herein comprise the use of a corticosteroid. Many synthetic corticosteroids are available that are suitable for administration to a subject, such as prednisone (prednisone), prednisolone (prednisone), dexamethasone (dexamethasone), betamethasone (betamethasone), methylprednisolone, triamcinolone (triamcinolone), hydrocortisone (hydrocortisone), and cortisone (cortisone).

NSAIDs (nonsteroidal anti-inflammatory drugs) are drugs that exhibit antipyretic, analgesic and anti-inflammatory effects through their inhibition of Cyclooxygenase (COX) enzyme I and enzyme II. NSAIDs are classified by both their chemical structural features and their COX I/II selectivity; exemplary classes of NSAIDs that may be used in the methods and compositions disclosed herein include salicylates, propionic acid derivatives, acetic acid derivatives, enolic acid (Oxicam) derivatives, anthranilic acid (Fenamate) derivatives, selective COX-2 inhibitors, non-selective COX inhibitors, and sulfoanilines (sulfonanilides). As all NSAIDs incorporate a chiral center, NSAIDs as described herein specifically include both racemic and enantiomerically pure forms. Salicylates are derivatives of salicylic acid; exemplary salicylates that may be used in the methods and compositions disclosed herein include, but are not limited to, aspirin (acetylsalicylic acid), Diflunisal (Diflunisal) (Dolobid), salicylic acid, and Salsalate (disalicid), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof. The propionic acid derivative is an aromatic derivative of propionic acid; exemplary NSAID propionic acid derivatives that may be used in the methods and compositions disclosed herein include, but are not limited to, ibuprofen, Dexibuprofen (Dexibuprofen), Naproxen (Naproxen), Fenoprofen (Fenoprofen), ketoprofen, Dexketoprofen (Dexketoprofen), Flurbiprofen (Flurbiprofen), Oxaprozin (Oxaprozin), and Loxoprofen (Loxoprofen), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof. The acetic acid derivative is an aromatic derivative of acetic acid; exemplary NSAID acetic acid derivatives that may be used in the methods and compositions disclosed herein include, but are not limited to, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac, aceclofenac, and nabumetone, and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof (e.g., ketorolac or a salt form of ketorolac such as ketorolac tromethamine). Enolic acid NSAID derivatives that may be used in the methods and compositions disclosed herein include, but are not limited to, Piroxicam (Piroxicam), Meloxicam (Meloxicam), Tenoxicam (Tenoxicam), Droxicam (Droxicam), Lornoxicam (Lornoxicam), and Isoxicam (Isoxicam), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof. Anthranilic acid derivatives that may be used in the methods and compositions disclosed herein include, but are not limited to, Mefenamic acid (Mefenamic acid), meclofenamic acid (meclofenamic acid), Flufenamic acid (Flufenamic acid), and Tolfenamic acid (Tolfenamic acid), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof. Sulfoanilines that may be used in the methods and compositions disclosed herein include, but are not limited to, Nimesulide (Nimesulide) and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof.

The overlap with the structural classification of NSAIDs is the classification of NSAIDs according to COX selectivity; NSAIDs may also be classified as selective COX-2 inhibitors or non-selective COX inhibitors. Examples of selective COX-2 inhibitors that may be used in the methods and compositions disclosed herein include, but are not limited to, Celecoxib (Celecoxib), Rofecoxib (Rofecoxib), Valdecoxib (Valdecoxib), Parecoxib (Parecoxib), Lumiracoxib (Lumiracoxib), Etoricoxib (Etoricoxib), and non-Rofecoxib (Firocoxib), and any pharmaceutically acceptable salts, solvates, hydrates, stereoisomers, clathrates, and prodrugs thereof.

Also contemplated within these classes of NSAIDs are pharmaceutically acceptable salts, solvates, hydrates, stereoisomers (e.g., enantiomers), clathrates, and prodrugs of salicylates, propionic acid derivatives, acetic acid derivatives, enolic acid (Oxicam) derivatives, anthranilic acid (Fenamate) derivatives, selective COX-2 inhibitors, non-selective COX inhibitors, and sulfonanilides, and any combination thereof. In certain embodiments, pharmaceutically acceptable salts, solvates, hydrates, stereoisomers (e.g., enantiomers), clathrates, and prodrugs of salicylates, propionic acid derivatives, acetic acid derivatives, enolic acid (Oxicam) derivatives, anthranilic acid (Fenamate) derivatives, selective COX-2 inhibitors, non-selective COX inhibitors, and sulfonanilides are administered as a racemic mixture. In certain embodiments, the pharmaceutically acceptable salts, solvates, hydrates, stereoisomers (e.g., enantiomers), clathrates, and prodrugs of salicylates, propionic acid derivatives, acetic acid derivatives, enolic acid (Oxicam) derivatives, anthranilic acid (Fenamate) derivatives, selective COX-2 inhibitors, non-selective COX inhibitors, and sulfonanilides are administered as a single enantiomer (e.g., (R) -ketorolac, (S) -ketorolac).

In certain embodiments, the methods provided herein comprise administering or using an anti-inflammatory agent (e.g., a ketorolac derivative) according to formula III or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof

Wherein:

x1 is selected from the group consisting of: H. a linear or branched, saturated or unsaturated C1-C20 aliphatic radical, optionally substituted by a C6-C10 aryl radical, -NHC6H5, -NHC6H5OCH3, -NHCH2C6H5, -NHCH26H4OCH3, -NHCH2COOCH3, -NHCH2CH2COOC2H5, -NHCH2(CH2)2CH3, -NHC6H10, -NHCH2CH2CH3, -NHCH (CH3)2, -NH (CH2)3OCH3, -NHCH2CH ═ CH2, optionally substituted talosyl, optionally substituted galactosyl, optionally substituted idosyl, optionally substituted gulosyl, optionally substituted mannosyl, optionally substituted glucosyl, optionally substituted altrose, optionally substituted allosyl, optionally substituted alkylpiperidinyl, optionally substituted piperazinyl, optionally substituted alkylpiperazinyl, optionally substituted morpholinyl, and optionally substituted alkylmorpholinyl.

In certain embodiments, the anti-inflammatory agent (e.g., a ketorolac derivative) according to formula III, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, is according to formula IV:

wherein:

z is C, N or O, with the proviso that when Z is O, X2 is absent,

x2 is selected from the group consisting of: H. linear or branched, saturated or unsaturated C1-C20 aliphatic groups, linear or branched, saturated or unsaturated C1-C20 acyl groups, and linear C1-C20 aliphatic alcohols.

Examples of the synthesis, chemical structure and properties of other ketorolac derivatives may be found in, for example, US8551958B 2; US20060183786a 1; qandil et al, Drug Dev Ind pharm.2008, month 10; 34(10) 1054-63; int J Mol sci.2012, qandil; 17244 17274, Pawar et al, AAPS PharmSciTech.2015, 6 months; 16(3) 518-; kim et al, Int J pharm.2005, month 4, day 11; 293(1-2):193-202 and Roy et al, J Pharm Sci.1994, 11 months; 83(11) 1548-53.

In certain embodiments, the anti-inflammatory agent (e.g., ketorolac derivative) according to formula III, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, is according to formula V:

Figure BDA0002562372210000281

wherein:

x1 is selected from the group consisting of: H. a linear or branched, saturated or unsaturated C1-C20 aliphatic radical, optionally substituted by a C6-C10 aryl radical, -NHC6H5, -NHC6H5OCH3, -NHCH2C6H5, -NHCH26H4OCH3, -NHCH2COOCH3, -NHCH2CH2COOC2H5, -NHCH2(CH2)2CH3, -NHC6H10, -NHCH2CH2CH3, -NHCH (CH3)2, -NH (CH2)3OCH3, -NHCH2CH ═ CH2, optionally substituted talosyl, optionally substituted galactosyl, optionally substituted idosyl, optionally substituted gulosyl, optionally substituted mannosyl, optionally substituted glucosyl, optionally substituted altrose, optionally substituted allosyl, optionally substituted alkylpiperidinyl, optionally substituted piperazinyl, optionally substituted alkylpiperazinyl, optionally substituted morpholinyl, and optionally substituted alkylmorpholinyl.

In certain embodiments, the compound according to formula III or formula IV, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, is administered separately. In certain embodiments, a compound according to formula III or formula IV, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof, is administered in combination with an immunosuppressive agent (e.g., a nucleoside analog antimetabolite such as cytarabine, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate of formula I or formula II).

In certain embodiments, the anti-inflammatory agent is one of the compounds provided in table 2, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, or clathrate thereof:

table 2: exemplary ketorolac-related compounds

Figure BDA0002562372210000282

In certain embodiments, the methods or compositions provided herein comprise administering or comprising an immunosuppressant (e.g., cytostatic agent, such as a nucleoside analog) in combination with an anti-inflammatory agent (e.g., NSAID, such as an acetic acid derivative). The immunosuppressant and anti-inflammatory agent may be administered simultaneously, separately or sequentially. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of 0.8 (e.g., 0.8:1) to 1,000 (e.g., 1000: 1). In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered or present in the composition in a molar ratio of at least 0.8. In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered or present in the composition in a molar ratio of up to 1,000. In certain embodiments, the immunosuppressive and anti-inflammatory agents are selected from 1,000 to 500, 1,000 to 200, 1,000 to 154.8, 1,000 to 150, 1,000 to 100, 1,000 to 75, 1,000 to 50, 1,000 to 30, 1,000 to 25, 1,000 to 20, 1,000 to 15, 500 to 200, 500 to 154.8, 500 to 150, 500 to 100, 500 to 75, 500 to 50, 500 to 30, 500 to 25, 500 to 20, 500 to 15, 200 to 154.8, 200 to 150, 200 to 100, 200 to 75, 200 to 50, 200 to 30, 200 to 25, 200 to 20, 200 to 15, 154.8 to 150, 154.8 to 100, 154.8 to 75, 154.8 to 30, 154.8 to 25, 154.8 to 20, 154.8 to 15, 150 to 100, 150 to 150, 150 to 100, 150 to 50, 150 to 30, 150 to 20, 100 to 50, 100 to 20, 100 to 75, 100 to 50, 100 to 25, 100 to 50, 100 to 75, 100 to 50, 30 to 25, 30 to 20, 30 to 15, 25 to 20, 25 to 15, or 20 to 15, 15 to 10, 10 to 9, 10 to 8, 10 to 7, 10 to 6, 10 to 5, 10 to 4, 10 to 3, 10 to 2, 10 to 1.5, 10 to 1, 10 to 0.8, 9 to 8, 9 to 7, 9 to 6, 9 to 5, 9 to 4, 9 to 3, 9 to 2, 9 to 1.5, 9 to 1, 9 to 0.8, 8 to 7, 8 to 6, 8 to 5, 8 to 4, 8 to 3, 8 to 2, 8 to 1.5, 8 to 1, 8 to 0.8, 7 to 6, 7 to 5, 7 to 4, 7 to 3, 7 to 2, 7 to 1.5, 7 to 1, 7 to 0.8, 6 to 5, 6 to 4, 6 to 3, 6 to 2, 6 to 5, 6 to 1.5, 6 to 5, 1.5, 1 to 4, 5 to 5, 1.5 to 4, 1.5, 3 to 4, 5 to 5, 1.8 to 5, 3 to 5, 3 to 4.5, 5, 3 to 4.8 to 5, 5 to 5, 3 to 5, 1.8 to 5, 5 to 5, A molar ratio of 3 to 1.5, 3 to 1, 3 to 0.8, 2 to 1.5, 2 to 1, 2 to 0.8, 1.5 to 1, 1.5 to 0.8, or 1 to 0.8 is applied or present in the composition. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of 1,000, 500, 200, 154.8, 150, 100, 75, 50, 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1.5, 1, or 0.8.

In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of 0.8 (e.g., 0.8:1) to 10 (e.g., 10: 1). In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered or present in the composition in a molar ratio of at least 0.8. In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered or present in the composition in a molar ratio of up to 10. In certain embodiments, the immunosuppressive and anti-inflammatory agents are selected from 10 to 9, 10 to 8, 10 to 7, 10 to 6, 10 to 5, 10 to 4, 10 to 3, 10 to 2, 10 to 1.5, 10 to 1, 10 to 0.8, 9 to 8, 9 to 7, 9 to 6, 9 to 5, 9 to 4, 9 to 3, 9 to 2, 9 to 1.5, 9 to 1, 9 to 0.8, 8 to 7, 8 to 6, 8 to 5, 8 to 4, 8 to 3, 8 to 2, 8 to 1.5, 8 to 1, 8 to 0.8, 7 to 6, 7 to 5, 7 to 4, 7 to 3, 7 to 2, 7 to 1, 7 to 0.8, 6 to 5, 6 to 4, 6 to 3, 6 to 2, 6 to 1.5, 6 to 1, 6 to 0.8, 5 to 4, 5 to 5, 3 to 1.5, 1 to 5, 1 to 1.5, 3 to 4, 3 to 5, 3 to 5 to 1.8, 3 to 1.5, 3 to 4, 3 to 5, 3 to 1.8, A molar ratio of 2 to 1.5, 2 to 1, 2 to 0.8, 1.5 to 1, 1.5 to 0.8, or 1 to 0.8 is applied or present in the composition. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of 10, 9, 8, 7, 6, 5, 4, 3, 2, 1.5, 1, or 0.8.

In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of about 0.8 to about 10. In certain embodiments, the immunosuppressant and anti-inflammatory agent are administered or present in the composition in a molar ratio of at least about 0.8. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of up to about 10. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered in a range of about 10 to about 9, about 10 to about 8, about 10 to about 7, about 10 to about 6, about 10 to about 5, about 10 to about 4, about 10 to about 3, about 10 to about 2, about 10 to about 1.5, about 10 to about 1, about 10 to about 0.8, about 9 to about 8, about 9 to about 7, about 9 to about 6, about 9 to about 5, about 9 to about 4, about 9 to about 3, about 9 to about 2, about 9 to about 1.5, about 9 to about 1, about 9 to about 0.8, about 8 to about 7, about 8 to about 6, about 8 to about 5, about 8 to about 4, about 8 to about 3, about 8 to about 2, about 8 to about 1.5, about 8 to about 1, about 8 to about 0.8, about 7 to about 6, about 7 to about 5, about 7 to about 7, about 3 to about 2, about 8 to about 7.5, about 7 to about 7, about 1.8 to about 6, about 7, about 6, about 7 to about 7, about 1.5, about 6, about 1.8 to about 1, about 6, about 7 to about 7, about 6, about 3 to about 1.8 to about 1.5, about 1, A molar ratio of about 6 to about 3, about 6 to about 2, about 6 to about 1.5, about 6 to about 1, about 6 to about 0.8, about 5 to about 4, about 5 to about 3, about 5 to about 2, about 5 to about 1.5, about 5 to about 1, about 5 to about 0.8, about 4 to about 3, about 4 to about 2, about 4 to about 1.5, about 4 to about 1, about 4 to about 0.8, about 3 to about 2, about 3 to about 1.5, about 3 to about 1, about 3 to about 0.8, about 2 to about 1.5, about 2 to about 1, about 2 to about 0.8, about 1.5 to about 1, about 1.5 to about 0.8, or about 1 to about 0.8 is applied or present in the composition. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered or present in the composition in a molar ratio of about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, about 1.5, about 1, or about 0.8.

If administered separately, the immunosuppressant (e.g., cytostatic agent, such as a nucleoside analogue) and the anti-inflammatory agent (e.g., NSAID, such as an acetic acid derivative) are preferentially administered within 24 hours of each other. In certain embodiments, the immunosuppressive and anti-inflammatory agents are administered within 18 hours, 12 hours, 6 hours, or 2 hours of each other.

V.Therapeutic agent formulation/route of administration

The compositions according to the invention (e.g. anti-inflammatory agents, immunosuppressive agents or a combination of anti-inflammatory and immunosuppressive agents) may be administered via a variety of routes. In certain embodiments, the pharmaceutical composition is formulated for oral administration. In other embodiments, the pharmaceutical composition is formulated for injection. In certain embodiments, the pharmaceutical composition is formulated for topical administration.

In certain embodiments, a compound described herein (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent, also described herein as an "active agent") is formulated into a pharmaceutical composition. In a particular embodiment, the pharmaceutical compositions are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations which can be used pharmaceutically. The appropriate formulation depends on the route of administration chosen. Using any pharmaceutically acceptable technique, carrier and excipient suitable for formulating the pharmaceutical compositions described herein: remington The Science and Practice of pharmacy, 19 th edition (Easton, Pa.: Mack Publishing Company, 1995); hoover, John e., Remington's Pharmaceutical Sciences, Mack Publishing co, Easton, Pennsylvania 1975; liberman, h.a. and Lachman, l. editors, Pharmaceutical document Forms, Marcel Decker, New York, n.y., 1980; and Pharmaceutical document Forms and Drug Delivery Systems, seventh edition (Lippincott Williams & Wilkins 1999).

Oral administration forms

In certain embodiments, a composition as described herein is formulated as an oral dosage form. One or more compounds according to the invention (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent, also described herein as "active agents") are formulated by combining them, for example, with a pharmaceutically acceptable carrier or excipient. In various embodiments, the compounds according to the present invention are formulated in oral dosage forms, including, by way of example only, tablets, powders, granules, pills, dragees (dragees), capsules, liquids, whey, gels, syrups, elixirs, slurries (pourry), suspensions, emulsions and the like.

In certain embodiments, a pharmaceutical preparation comprising an active agent for oral use is obtained by: one or more solid excipients are mixed with one or more of the compounds described herein, the resulting mixture is optionally milled, and the mixture of granules is processed, if desired after addition of suitable auxiliaries, to obtain tablets, pills or dragee cores (dragee cores). In particular, suitable excipients are fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, such as, for example, corn starch, wheat starch, rice starch (rice starch), potato starch, gelatin, gum tragacanth, methyl cellulose, microcrystalline cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose; or others such as polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. In a particular embodiment, a disintegrant is optionally added. By way of example only, disintegrating agents include cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

In one embodiment, dosage forms, such as dragee cores, pills, and tablets, are provided with one or more suitable coatings. In particular embodiments, the concentrated sugar solution is used to coat the dosage form. The sugar solution optionally comprises further components such as, by way of example only, gum arabic, talc, polyvinylpyrrolidone, carbopol gel (carbopol gel), polyethylene glycol and/or titanium dioxide, lacquer solutions (lacquer solutions) and suitable organic solvents or solvent mixtures. Dyes and/or pigments are also optionally added to the coating for identification purposes. In addition, dyes and/or pigments are optionally used to characterize different combinations of active compound dosages.

In certain embodiments, a therapeutically effective amount of an active agent described herein is formulated into other solid oral dosage forms. Oral dosage forms include push-fit capsules (push fit capsules) made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. In particular embodiments, the push-fit capsules comprise the active ingredient mixed with one or more fillers. By way of example only, fillers include lactose, binders such as starch and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In other embodiments, soft capsules comprise one or more active compounds dissolved or suspended in a suitable liquid. By way of example only, suitable liquids include one or more fatty oils, liquid paraffin, or liquid polyethylene glycol. In addition, a stabilizer is optionally added.

In other embodiments, the active agents described herein are formulated as oral liquid dosage forms. Exemplary liquid preparations for oral use include solutions, emulsions, whey, syrups or suspensions containing one or more active ingredients in a suitable vehicle. Syrups are clear, viscous oral liquids containing a high concentration of sugar or other sweetener, in which the active agent is dissolved in a pharmaceutically acceptable vehicle. Suspensions consist of finely divided particles of the active agent suspended in a pharmaceutically acceptable vehicle in which the particles are poorly soluble. Oral emulsions comprise a liquid form of the active agent dispersed as droplets in a continuous phase of another immiscible vehicle with the aid of an emulsifier (e.g., sugars, gelatin, high molecular weight alcohols, wetting agents, colloidal clays, and the like).

In certain embodiments, the active agent is formulated as a semi-solid oral dosage form, such as a gel. Gel-or jelly-like formulations (jelly-gels) are particularly relevant for elderly patients with difficulty consuming other oral dosage forms or patients with difficulty swallowing. The gel is formed by adding the active agent to water, adding a low critical concentration (e.g., 0.5% -2.5%) of gelling agent, heating and cooling. Examples of suitable gelling agents include agar, gelatin, carrageenan, sodium caseinate, glycerogelatin (glycogelatin), fibroin (silk fibroin), gellan (gellan gum), alginate (kelgel), xyloglucan, gellan (gellan), and pectin.

Injectable/topical forms

In still other embodiments, the active agents described herein (e.g., anti-inflammatory agents, immunosuppressive agents, or a combination of anti-inflammatory and immunosuppressive agents) are formulated for parenteral injection, including formulations suitable for bolus injection (bolus injection) or continuous infusion. In particular embodiments, the formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers. In particular embodiments, the formulations are formulated for intravenous, intramuscular, subcutaneous, or intraperitoneal administration. Preservatives are optionally added to the injectable formulations. In still other embodiments, the pharmaceutical compositions are formulated as sterile suspensions, solutions, or emulsions in oily or aqueous vehicles in a form suitable for parenteral injection. Parenteral injection formulations optionally comprise formulating agents (formulations), such as suspending, stabilizing and/or dispersing agents. In a particular embodiment, the pharmaceutical formulation for parenteral administration comprises an aqueous solution of the active compound in water-soluble form. In other embodiments, the suspension of the active agent is prepared as a suitable oily injection suspension. Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example only, fatty oils such as sesame oil, or synthetic fatty acid esters such as ethyl oleate or triglycerides, or liposomes. In certain particular embodiments, the aqueous injection suspension comprises a substance that increases the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension contains suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, in other embodiments, the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.

Pharmaceutical compositions comprising any of the pharmaceutical agents described herein may be formulated for sustained or slow release, also known as timed release (timed release) or controlled release. Such compositions may generally be prepared using well-known techniques and administered by, for example, oral, rectal, intradermal or subcutaneous implantation, or by implantation at the desired target site. Sustained release formulations may comprise the compound dispersed in a carrier matrix and/or contained in a reservoir (reservoir) surrounded by a rate controlling membrane. Excipients used in such formulations are biocompatible and may also be biodegradable; preferably, the formulation provides a relatively constant level of active ingredient release. The amount of the pharmaceutical agent included in the sustained release formulation depends on the site of implantation, the rate and expected duration of release, and the nature of the condition, disease or disorder to be treated or prevented.

In still other embodiments, the active agent is administered topically. The compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, foams, whey, pastes, medicated sticks (sticks), balms (palms), creams, or ointments. Such pharmaceutical compositions optionally comprise solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.

VI.Method of treatment

The compounds and compositions described herein (e.g., anti-inflammatory agents, immunosuppressive agents, or a combination of anti-inflammatory and immunosuppressive agents, or a combination thereof) can be used in the manufacture of a medicament for preventing or treating cachexia or a cachexia-related disease or condition. Further, a method for treating any of the diseases or conditions described herein in a subject in need of such treatment comprises administering to the subject a pharmaceutical composition comprising at least one compound described herein (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent), or a pharmaceutically acceptable salt, a pharmaceutically acceptable prodrug, a pharmaceutically acceptable stereoisomer, or a pharmaceutically acceptable solvate thereof, in a therapeutically effective amount.

Time Schedule (Schedule)

In certain embodiments, one or more of the active agents (e.g., an anti-inflammatory agent as described herein, an immunosuppressive agent as described herein) are administered separately. In certain embodiments, separately administered active agents are administered in separate dosage units (e.g., pills, dragees, tablets). In certain embodiments, the separately administered active agents are administered via separate routes of administration. In certain embodiments, an active agent (immunosuppressive or anti-inflammatory agent) is administered. In certain embodiments, two of the active agents are administered separately. In certain embodiments, the separately administered active agents are not administered simultaneously.

In certain embodiments, the active agents that are not concurrently administered are administered within a defined window (window). In particular embodiments, the defined window is 48 hours, 36 hours, 24 hours, 12 hours, or 6 hours. In certain embodiments, one or more, two or more, or three of the active agents are administered within a defined window.

In certain embodiments, an active agent or composition thereof for treatment is administered for a particular treatment period. In certain embodiments, the active agent or composition thereof is administered for a long-term treatment period (e.g., for an extended period of time, including the entire duration of the patient's life) in order to alleviate or otherwise control or limit the symptoms of the disease or condition in the patient. The active agents or compositions thereof for treatment are administered on a specific schedule over a treatment period. In further embodiments, the active agent or composition thereof is administered once, twice, three times, or four times daily. In certain embodiments, the active agent or composition thereof is administered in the morning and in the evening. In certain embodiments, the active agent or composition thereof is administered once, twice, three times, or four times per week. In certain embodiments, the active agent or composition thereof is administered once, twice, three times, or four times per month. In certain embodiments, the active agent or composition thereof is administered daily for a particular period of time, such as three, four, five, six, or seven consecutive days.

In certain embodiments, the method comprises administering the pharmaceutical agent in at least two treatment cycles. In particular embodiments, the non-treatment interval may be at least about 2 weeks or between at least about 0.5 month to 12 months, such as at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, or at least about 12 months (i.e., 1 year). In certain embodiments, the non-treatment intervals are between 1 year and 2 years or between 1 year and 3 years or longer. In certain embodiments, each course of treatment is no longer than about 1 month, no longer than about 2 months, or no longer than about 3 months; or no more than 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, or 31 days.

In cases where the patient's condition does improve, administration of the compound may be given continuously, at the discretion of the physician; alternatively, the dose of drug administered may be temporarily reduced or temporarily suspended for a length of time (i.e., a "drug holiday"). The length of the drug holiday can vary between 2 days and 1 year, including, by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday can be from about 10% to about 100%, including by way of example only about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.

When an improvement in the patient's condition has occurred, a maintenance dose is administered as necessary. Subsequently, as the symptoms change, the dose or frequency of administration, or both, can be reduced to a level at which the improved disease, disorder, or condition (e.g., cancer, immune disorder, cachexia) is maintained. However, patients may require long-term intermittent treatment based on any recurrence of symptoms.

Dosage form

In certain embodiments, the conditions described herein for treatment (e.g., cancer, immune disorders, cachexia) are treated with an anti-inflammatory or immunosuppressive agent and a dose previously described for administration of the compound in a clinical trial or listed in the product package sufficient to achieve the anti-inflammatory or immunosuppressive effect. In addition, the dose of the anti-inflammatory agent or immunosuppressive agent can be determined by monitoring subject levels of biomarkers of inflammation (e.g., C-reactive protein, IL-6, neutrophil to leukocyte ratio, neutrophil lymphocyte ratio) and/or biomarkers of immunosuppression (e.g., soluble CD30, T cell proliferation by PCNA, regulatory T cell ratio, IL-2, TNF α).

In certain embodiments, the dose for therapeutic application is determined in a model organism (e.g., mouse, monkey, dog) and then used to calculate an appropriate dose for treatment in humans. Various methods are available for calculating human doses from dosing data determined in other organisms; examples include factor dose methods (dose by factor methods) which use an index of body surface area to account for metabolic rate differences and convert doses between animals and humans, and allometric scaling methods (where interchange of drug doses is based on normalization of dose to body surface area), both of which are described, for example, in Nair et al, J Basic Clin pharm.2016, 3 months-2016, 5 months; 7(2) 27-31.

In certain embodiments, the applied factor dose method comprises applying the following equation (equation 1) to calculate HED (human equivalent dose) using the level of observed adverse effects (NOAEL) of the drug as determined using animal experiments.

HED (mg/kg) ═ animal NOAEL mg/kg × (weight)Animal(s) production[kg]Weight/weightHuman being[kg])(1–0.67)

(equation 1)

In certain embodiments, applying the anisometric scale law method comprises applying the following equation (equation 2) to calculate HED (human equivalent dose) using the animal dose and a correction factor (Km) estimated by dividing the average body weight (kg) of the species by its body surface area (m 2).

HED (mg/kg) ═ animal dose (mg/kg) × (animal Km/human Km)

(equation 2)

Treatment of immune disorders

In one aspect, the disclosure herein provides a method for treating a condition characterized by an immune disorder, the method comprising administering to a subject in need thereof an effective amount of (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both. Immune disorders involve dysfunction of the activity of a particular immune process (e.g., hyperactivity or hyperactivity) or the manifestation of one or more components of the immune system (e.g., a particular type of cell). In certain embodiments, the immune disorder comprises at least one of: (1) an increased leukocyte count, (2) an increased neutrophil to leukocyte ratio, and (3) a decreased T cell to leukocyte ratio, or (4) any combination thereof. In other embodiments, the immune disorder comprises a particular subtype of the disorder of (2), (3), or (4), including but not limited to cytotoxic T cells, helper T cells, low density neutrophils, high density neutrophils, and any combination thereof. In certain embodiments, the immune disorder comprises cachexia. In certain embodiments, the immune disorder comprises cachexia that manifests as symptoms of the immune disorder. The immunosuppressive and anti-inflammatory agents can include any immunosuppressive and anti-inflammatory agent described herein (e.g., NSAIDs, cytostatics, antimetabolites, purine antimetabolites, etc.).

In certain embodiments, the agent is administered within a treatment cycle that includes a course of treatment followed by a non-treatment interval. The course of treatment administered refers herein to a limited time frame over which one or more doses of a pharmaceutical agent are administered over one or more days. The limited time frame may also be referred to herein as a treatment window.

In certain embodiments, provided herein is a method for treating an immune disorder (e.g., cachexia), and the method comprises administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) to a subject in need thereof in a manner sufficient to (a) reduce the amount of leukocytes in the subject, (b) reduce the neutrophil to leukocyte ratio in the subject, (c) increase the T cell to leukocyte ratio in the subject, (d) reverse weight loss in the subject/increase the weight of the subject, (e) extend the survival time of the subject, or (f) any combination thereof. In certain embodiments, the route of administration, dosage, or pharmaceutical form of the one or more agents may be adjusted to modify any one of phenotype (a) -phenotype (d), and any combination thereof.

In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the amount of leukocytes in a subject by about 10% to about 95%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the amount of leukocytes in a subject by at least about 10%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the amount of leukocytes in a subject by up to about 95%. In certain embodiments, the method comprises reducing the amount of leukocytes in a subject by about 10% to about 20%, about 10% to about 30%, about 10% to about 40%, about 10% to about 50%, about 10% to about 60%, about 10% to about 70%, about 10% to about 80%, about 10% to about 90%, about 10% to about 95%, about 20% to about 30%, about 20% to about 40%, about 20% to about 50%, about 20% to about 60%, about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 20% to about 95%, about 30% to about 40%, about 30% to about 50%, about 30% to about 60%, about 30% to about 70%, about 30% to about 80%, about 30% to about 90%, about 30% to about 95%, about 40% to about 50%, about 40% to about 60%, about 40% to about 70%, about 40% to about 80%, about 40% to about 90%, or a combination thereof, The immune disorder (e.g., cachexia) is treated by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner that treats the immune disorder (e.g., cachexia) from about 40% to about 95%, from about 50% to about 60%, from about 50% to about 80%, from about 50% to about 90%, from about 50% to about 95%, from about 60% to about 70%, from about 60% to about 80%, from about 60% to about 90%, from about 60% to about 95%, from about 70% to about 80%, from about 70% to about 90%, from about 70% to about 95%, from about 80% to about 90%, from about 80% to about 95%, or from about 90% to about 95%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the amount of leukocytes in a subject by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95%.

In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the neutrophil to leukocyte ratio of the subject by about 10% to about 95%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the neutrophil to leukocyte ratio of the subject by at least about 10%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the neutrophil to leukocyte ratio of the subject by up to about 95%. In certain embodiments, the method comprises reducing the neutrophil to leukocyte ratio in a subject by about 10% to about 20%, about 10% to about 30%, about 10% to about 40%, about 10% to about 50%, about 10% to about 60%, about 10% to about 70%, about 10% to about 80%, about 10% to about 90%, about 10% to about 95%, about 20% to about 30%, about 20% to about 40%, about 20% to about 50%, about 20% to about 60%, about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 20% to about 95%, about 30% to about 40%, about 30% to about 50%, about 30% to about 60%, about 30% to about 70%, about 30% to about 80%, about 30% to about 90%, about 30% to about 95%, about 40% to about 50%, about 40% to about 60%, about 40% to about 70%, about 40% to about 80%, or about 40% to about 80%, or about 80, One or more agents (e.g., anti-inflammatory agents, immunosuppressive agents, or a combination of anti-inflammatory agents and immunosuppressive agents) are administered to treat an immune disorder (e.g., cachexia) in a manner that about 40% to about 90%, about 40% to about 95%, about 50% to about 60%, about 50% to about 70%, about 50% to about 80%, about 50% to about 90%, about 50% to about 95%, about 60% to about 70%, about 60% to about 80%, about 60% to about 90%, about 60% to about 95%, about 70% to about 95%, about 80% to about 90%, about 80% to about 95%, or about 90% to about 95%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to reduce the neutrophil to leukocyte ratio of the subject by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95%.

In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by about 110% to about 170%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by at least about 110%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by up to about 170%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of a subject by about 110% to about 120%, about 110% to about 130%, about 110% to about 140%, about 110% to about 150%, about 110% to about 160%, about 110% to about 170%, about 120% to about 130%, about 120% to about 140%, about 120% to about 150%, about 120% to about 160%, about 120% to about 170%, about 130% to about 140%, about 130% to about 150%, about 130% to about 160%, about 130% to about 170%, about 140% to about 150%, about 140% to about 160%, about 140% to about 170%, about 150% to about 170%, or about 160% to about 170%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by about 110%, about 120%, about 130%, about 140%, about 150%, about 160%, or about 170%.

In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by about 5% to about 170%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by at least about 5%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by up to about 170%. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) increases the body weight of a subject by about 5% to about 25%, about 5% to about 50%, about 5% to about 75%, about 5% to about 100%, about 5% to about 125%, about 5% to about 150%, about 5% to about 170%, about 25% to about 50%, about 25% to about 75%, about 25% to about 100%, about 25% to about 125%, about 25% to about 150%, about 25% to about 170%, about 50% to about 75%, about 50% to about 100%, about 50% to about 125%, about 50% to about 150%, about 50% to about 170%, about 75% to about 100%, about 75% to about 125%, about 75% to about 150%, about 75% to about 170%, about 100% to about 125%, about 100% to about 150%, about 100% to about 170%, about 125% to about 150%, or a combination of an anti-inflammatory agent and an immunosuppressive agent, From about 125% to about 170% or from about 150% to about 170% is effective. In certain embodiments, administration of one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) is effective to increase the body weight of the subject by about 5%, about 25%, about 50%, about 75%, about 100%, about 125%, about 150%, or about 170%.

In certain embodiments, a method for treating an immune disorder (e.g., cachexia) comprising administering to a subject in need thereof a combination of an anti-inflammatory agent and an immunosuppressive agent (e.g., ketorolac and cytarabine) counteracts the side effects of a single administration (monoadministerate) of the immunosuppressive agent (e.g., cytarabine) alone. In certain embodiments, the side effect is a transient weight loss. In certain embodiments, the temporary weight loss that is offset is about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or 20% of the body weight before treatment. In certain embodiments, the counteracted transient weight loss persists for no more than 1 day, no more than 2 days, no more than 3 days, no more than 4 days, or no more than 5 days after treatment.

In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to prolong the survival time of a subject by about 10% to about 1,000%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to prolong the survival time of a subject by at least about 10%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to extend the survival time of a subject by up to about 100%. In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to extend the survival time of a subject by up to about 1,000%. In certain embodiments, the method comprises increasing survival of a subject by about 10% to about 20%, about 10% to about 30%, about 10% to about 40%, about 10% to about 50%, about 10% to about 60%, about 10% to about 70%, about 10% to about 80%, about 10% to about 90%, about 10% to about 100%, about 20% to about 30%, about 20% to about 40%, about 20% to about 50%, about 20% to about 60%, about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 20% to about 100%, about 30% to about 40%, about 30% to about 50%, about 30% to about 60%, about 30% to about 70%, about 30% to about 80%, about 30% to about 90%, about 30% to about 100%, about 40% to about 50%, about 40% to about 60%, about 40% to about 70%, about 40% to about 80%, about 40% to about 90%, or a combination thereof, About 40% to about 100%, about 50% to about 60%, about 50% to about 70%, about 50% to about 80%, about 50% to about 90%, about 50% to about 100%, about 60% to about 70%, about 60% to about 80%, about 60% to about 90%, about 60% to about 100%, about 70% to about 80%, about 70% to about 90%, about 70% to about 100%, about 80% to about 90%, about 80% to about 100%, about 90% to about 100%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 100% to about 300%, about 100% to about 500%, about 100% to about 1,000%, about 150% to about 200%, about 150% to about 300%, about 150% to about 500%, about 150% to about 1,000%, about 200% to about 250%, about 200% to about 300%, about 200% to about 500%, about 200% to about 200%, about 1,000%, about 250% to about 500%, about 200% to about 250% to about 300%, about 200% to about 500, One or more agents (e.g., anti-inflammatory agents, immunosuppressive agents, or a combination of anti-inflammatory and immunosuppressive agents) are administered from about 250% to about 1,000%, from about 300% to about 500%, from about 300% to about 1,000%, or from about 500% to about 1,000% to treat an immune disorder (e.g., cachexia). In certain embodiments, the methods comprise treating an immune disorder (e.g., cachexia) by administering one or more agents (e.g., an anti-inflammatory agent, an immunosuppressive agent, or a combination of an anti-inflammatory agent and an immunosuppressive agent) in a manner sufficient to prolong the survival time of a subject by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 100%, about 150%, about 200%, about 250%, about 300%, about 500%, or about 1,000%.

Treatment of conditions associated with cachexia

In one aspect, the disclosure herein provides a method for treating a subject afflicted with a primary condition associated with cachexia, the method comprising alternately administering to a subject in need thereof: an effective amount of (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both; and an effective amount of an agent for treating the primary condition. The immunosuppressive and anti-inflammatory agents can include any immunosuppressive and anti-inflammatory agent described herein (e.g., NSAIDs, cytostatics, antimetabolites, purine antimetabolites, etc.).

By "administered alternately" is meant that administration of the agent for treating cachexia and administration of the agent for treating the primary condition are in one-by-one form, optionally with a non-treatment period in between, and optionally repeated for a number of cycles. Alternatively, administration may comprise a variety of different embodiments. For example, the treatment for cachexia as disclosed herein and the treatment for the primary condition may be administered in any order. In certain embodiments, treatment for cachexia as disclosed herein is first administered for a period of time, followed by treatment for the primary condition for another period of time. In other embodiments, the agent for treating the primary condition is administered first for a period of time, followed by treatment for cachexia for another period of time. In certain embodiments, the two treatments are separated by a drug holiday in which no treatment is administered. The length of the drug holiday can vary between 1 day or 2 days and 1 year, including, by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.

In certain embodiments, the primary condition associated with cachexia is any chronic disease/severity disease known to cause cachexia, such as tumor, AIDS, sepsis, severe trauma, chronic obstructive pulmonary disease, multiple sclerosis, congestive heart failure, tuberculosis, familial amyloidotic neuropathy, chronic kidney disease, cystic fibrosis, or type I diabetes, and any combination thereof. In certain instances, the treatment regimens described herein (e.g., a combination of an immunosuppressive agent and an anti-inflammatory agent) improve the treatment of the primary condition. Administration of a regimen (e.g., a combination of an immunosuppressive agent and an anti-inflammatory agent) for treating a primary condition with cachexia can improve survival in response to treatment of the primary disease. When the treatment for the primary disease includes treatment periods alternating with non-treatment periods, administration of a regimen (e.g., a combination of an immunosuppressive agent and an anti-inflammatory agent) for treating the primary condition with cachexia can reduce the amount of time between treatment cycles of the primary disease. When the treatment for the primary disease involves alternating treatment cycles and non-treatment cycles, and the number of treatment cycles is limited by the toxicity of the drug used to treat the primary condition associated with cachexia, administration of a regimen (e.g., a combination of an immunosuppressive agent and an anti-inflammatory agent) for treating the primary condition associated with cachexia can reduce the number of treatment cycles of the primary disease that can be safely administered to a subject.

In certain embodiments, in a method of treating a primary condition associated with cachexia, an effective amount of (i) an immunosuppressive agent, (ii) an anti-inflammatory agent, or both, is administered for at least two treatment cycles. In particular embodiments, the non-treatment interval may be at least about 2 weeks or between at least about 0.5 month to 12 months, such as at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, or at least about 12 months (i.e., 1 year). In certain embodiments, the non-treatment intervals are between 1 year and 2 years or between 1 year and 3 years or longer. In certain embodiments, each course of treatment is no longer than about 1 month, no longer than about 2 months, or no longer than about 3 months; or no more than 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, or 31 days.

In certain instances, a method of treating a primary condition associated with cachexia may comprise a number of alternating cycles of (a) treatment of cachexia and (b) treatment of the primary condition. In certain embodiments, the method of treating a primary condition associated with cachexia comprises 1 cycle to 10 cycles. In certain embodiments, the method of treating a primary condition associated with cachexia comprises at least 1 cycle. In certain embodiments, the method of treating a primary condition associated with cachexia comprises up to 10 cycles. In certain embodiments, the method of treating a primary condition associated with cachexia comprises 1 cycle to 2 cycles, 1 cycle to 3 cycles, 1 cycle to 4 cycles, 1 cycle to 5 cycles, 1 cycle to 6 cycles, 1 cycle to 7 cycles, 1 cycle to 8 cycles, 1 cycle to 9 cycles, 1 cycle to 10 cycles, 2 cycle to 3 cycles, 2 cycle to 4 cycles, 2 cycle to 5 cycles, 2 cycle to 6 cycles, 2 cycle to 7 cycles, 2 cycle to 8 cycles, 2 cycle to 9 cycles, 2 cycle to 10 cycles, 3 cycle to 4 cycles, 3 cycle to 5 cycles, 3 cycle to 6 cycles, 3 cycle to 7 cycles, 3 cycle to 8 cycles, 3 cycle to 9 cycles, 9 cycles to 9 cycles, 3 cycle to 4 cycles, 3 cycle to 5 cycles, 3 cycle to 6 cycles, 3 cycle to 7 cycles, 3 cycle to 8 cycles, 3 cycle to 9 cycles, or, 3 cycles to 10 cycles, 4 cycles to 5 cycles, 4 cycles to 6 cycles, 4 cycles to 7 cycles, 4 cycles to 8 cycles, 4 cycles to 9 cycles, 4 cycles to 10 cycles, 5 cycles to 6 cycles, 5 cycles to 7 cycles, 5 cycles to 8 cycles, 5 cycles to 9 cycles, 5 cycles to 10 cycles, 6 cycles to 7 cycles, 6 cycles to 8 cycles, 7 cycles to 9 cycles, 7 cycles to 10 cycles, 8 cycles to 9 cycles, 8 cycles to 10 cycles, or 9 cycles to 10 cycles. In certain embodiments, the method of treating a primary condition associated with cachexia comprises 1 cycle, 2 cycles, 3 cycles, 4 cycles, 5 cycles, 6 cycles, 7 cycles, 8 cycles, 9 cycles, or 10 cycles.

In certain embodiments, in a method of treating a primary condition associated with cachexia, an effective amount of an agent for treating the primary condition associated with cachexia is administered for at least two treatment cycles. In particular embodiments, the non-treatment interval may be at least about 2 weeks or between at least about 0.5 month to 12 months, such as at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, or at least about 12 months (i.e., 1 year). In certain embodiments, the non-treatment intervals are between 1 year and 2 years or between 1 year and 3 years or longer. In certain embodiments, each course of treatment is no longer than about 1 month, no longer than about 2 months, or no longer than about 3 months; or no more than 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, or 31 days.

The agent for treating the primary condition associated with cachexia may be any pharmaceutically acceptable agent approved for use in treating the primary condition associated with cachexia. In the case where the primary condition is a tumor or cancer, the pharmaceutically acceptable agent includes a chemotherapeutic regimen (e.g., a combination of agents) including, but not limited to, 7+3, ABVD, AC, BACOD, BEACOPP, BEP, CA, CAF, CAPOX, CAV, CBV, CHOEP, CEPP, ChlVPP/EVA, CHOP, R-CHOP, ClaPD, CMF, CMV, COP or CVP, COPP, CT or TC, CTD, CVAD, CVE, CYBORD, DA or DAC, DAT, DCEP, DHAP-R, DICE, DT-PACE, EC, ECF, EOX, EP, CH, EPOCCH-R, ESHAP-R, FAM, FAMTX, FCM, FCIAM-R, FCR, FM-R, FM-FL (also known as Mayo), LFFLAG-R, FLAG-FLA, FLAI, FLA-FLAM, GemOX, GEI, GEmOX, FOMCO, FO-OX, FO-FO, CAO, CAC, CA, IFL, IVA, m-BACOD, MACOP-B, MAID, MINE-R, MMM, MOPP, MVAC, MVP, NP, PACE, PCV, PEB, PEI, POMP, ProMACE-MOPP, ProMACE-CytaBOM, RdC, R-Benda, R-DHAP, R-FCM, R-ICE, RVD, Stanford V, TAC, TAD, TC or CT, TCH, Thal/Dex, TIP, EE-4A, DD-4A, VABCD, VAVAC, VAVAVAD, VAMP, Regimen I, VAPEC-B, VD-PACE, VIFUP, VIP, and VTD-PACE and any combination thereof. In the case where the primary condition is a tumor or cancer, the pharmaceutically acceptable agent includes one or more alkylating agents (e.g., cyclophosphamide, dichloromethyldiethylamine, chlorambucil, melphalan, Dacarbazine (Dacarbazine)), anthracyclines (e.g., Daunorubicin (Daunorubicin), Doxorubicin (Doxorubicin), Epirubicin (epiubicin), Idarubicin (Idarubicin), Mitoxantrone (Mitoxantrone), Valrubicin (Valrubicin)), cytoskeletal disruptors (e.g., Paclitaxel), Docetaxel (Docetaxel), Abraxane, Taxotere), epothilones (epothilone) (e.g., ixabepilone), histone deacetylase inhibitors (e.g., Vorinostat, roxide (romycin)), topoisomerase I inhibitors (e.g., Irinotecan inhibitor (Irinotecan), Topotecan (Etoposide)), Topotecan (Etoposide), and combinations thereof, Teniposide (Teniposide), Tafluposide), kinase inhibitors (e.g. Bortezomib (Bortezomib), Erlotinib (Erlotinib), Gefitinib (Gefitinib), Imatinib (Imatinib), Vemurafenib (Vemurafenib), Vismodegib (Vismodegib)), peptide antibiotics (e.g. Bleomycin (Bleomycin), actinomycin), retinoids (e.g. Tretinoin, Alitretinoin (alitretinitinin), Bexarotene (Bexarotene)) or vinca alkaloids or derivatives of vinca alkaloids (e.g. Vinblastine (vinastine), Vincristine (vincrisine), Vindesine (Vindesine), Vinorelbine (Vinorelbine)). In the case where the primary condition is a tumor or cancer, the pharmaceutically acceptable agent includes one or more approved chemotherapeutic agents, such as cyclophosphamide, ifosfamide, mesna (mesna), methotrexate, mitomycin, etoposide, irinotecan, mitoxantrone, epirubicin, fludarabine, G-CSF, amsacrine (amsacrine), folinic acid (leucovorin), topotecan, hydroxydaunomycin, vincristine, bleomycin, dacarbazine, procarbazine (procarbazine), thalidomide (thalidomide), fluorouracil, cisplatin, oxaliplatin, carboplatin, thioguanine (tioguanine), capecitabine, gemcitabine, vinorelbine, 6-mercaptopurine, fluorouracil, bendamustine (bendamustine), carmustine, chlorambucil, docetaxel, paclitaxel, bortezomib, and hydroxyurea, and any combination thereof.

In cases where the primary condition is an autoimmune disease (e.g. crohn's disease, rheumatoid arthritis, systemic lupus erythematosus), pharmaceutically acceptable agents include disease-modifying antirheumatic drugs (DMARDS, e.g. hydroxychloroquine, methotrexate, azathioprine) and anti-TNF agents (e.g. infliximab, adalimumab, certolizumab pegol, golimumab, etanercept).

In the case where the primary condition is AIDS, the pharmaceutically acceptable agent includes a nucleotide reverse transcriptase inhibitor (NRTI, e.g., Abacavir (Abacavir), Didanosine (Didanosine), Emtricitabine (Emtricitabine), Lamivudine (Lamivudine), Stavudine (Stavudine), Tenofovir (Tenofovir), Zalcitabine (Zalcitabine), Zidovudine (Zidovudine)), a protease inhibitor (PI, e.g., Amprenavir (Amprenavir), Atazanavir (Atazanavir), Fosamprenavir (Fosamprenavir), Indinavir (Indinavir), Lopinavir (Lopinavir), Ritonavir (Ritonavir), Saquinavir (Saquinavir)), and any combination thereof (e.g., highly active antiretroviral therapy/HAART).

In certain embodiments, the method of treating a primary condition associated with cachexia can extend the survival time of a subject by about 10% to about 1,000%. In certain embodiments, the method of treating a primary condition associated with cachexia can extend the survival time of a subject by at least about 10%. In certain embodiments, the method of treating a primary condition associated with cachexia can extend the survival time of a subject by up to about 100%. In certain embodiments, the method of treating a primary condition associated with cachexia can extend the survival time of a subject by up to about 1,000%. In certain embodiments, the method of treating a primary condition associated with cachexia may extend survival time of a subject by about 10% to about 20%, about 10% to about 30%, about 10% to about 40%, about 10% to about 50%, about 10% to about 60%, about 10% to about 70%, about 10% to about 80%, about 10% to about 90%, about 10% to about 100%, about 20% to about 30%, about 20% to about 40%, about 20% to about 50%, about 20% to about 60%, about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 20% to about 100%, about 30% to about 40%, about 30% to about 50%, about 30% to about 60%, about 30% to about 70%, about 30% to about 80%, about 30% to about 90%, about 30% to about 100%, about 40% to about 50%, about 40% to about 60%, about 40% to about 70%, about 40% to about 80%, or about 40% to about 80, About 40% to about 90%, about 40% to about 100%, about 50% to about 60%, about 50% to about 70%, about 50% to about 80%, about 50% to about 90%, about 50% to about 100%, about 60% to about 70%, about 60% to about 80%, about 60% to about 90%, about 60% to about 100%, about 70% to about 80%, about 70% to about 90%, about 70% to about 100%, about 80% to about 90%, about 80% to about 100%, about 90% to about 100%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 100% to about 300%, about 100% to about 500%, about 100% to about 1,000%, about 150% to about 200%, about 150% to about 250%, about 150% to about 500%, about 150% to about 1,000%, about 200% to about 250%, about 200% to about 300%, about 200% to about 200%, about 200% to about 500%, about 1,000%, about 250% to about 250%, about 200% to about 300%, about 300, About 250% to about 500%, about 250% to about 1,000%, about 300% to about 500%, about 300% to about 1,000%, or about 500% to about 1,000%. In certain embodiments, the method of treating a primary condition associated with cachexia may extend the survival time of a subject by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 250%, about 300%, about 500%, or about 1,000%.

Reagent kit

The invention also includes kits suitable for use in performing the methods of treatment described above (e.g., methods of treating an immune disorder or methods of treating a primary disorder associated with cachexia). In one embodiment, the kit comprises a dosage form comprising one or more immunosuppressive agents (e.g., purine nucleotide analogs) as described above in an amount sufficient to perform the methods of the invention. Preferably, the dosage form comprises an effective amount of an immunosuppressive agent to treat an immune disorder selected from the group consisting of: (1) increased leukocyte count, (2) increased neutrophil to leukocyte ratio, and (3) decreased T cell to leukocyte ratio, and any combination thereof. In another embodiment, a kit comprises a first dosage form comprising one or more immunosuppressive agents as described above and a second dosage form comprising one or more anti-inflammatory agents as described above in amounts sufficient to perform the methods of the invention. Preferably, the first dosage form and the second dosage form together comprise a therapeutically effective amount of an immunosuppressive agent and an anti-inflammatory agent for treating an immune disorder selected from the group consisting of: (1) increased leukocyte count, (2) increased neutrophil to leukocyte ratio, and (3) decreased T cell to leukocyte ratio, and any combination thereof. In certain embodiments, the one or more immunosuppressive agents are cytarabine or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the one or more anti-inflammatory agents are ketorolac or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the dosage form further comprises a pharmaceutically acceptable excipient.

In yet another embodiment, a kit comprises (i) a first dosage form comprising one or more immunosuppressive agents as described above and a second dosage form comprising one or more anti-inflammatory agents as described above in amounts sufficient to perform the methods of the invention (e.g., for treatment of an immune disorder); and (ii) a third dosage form comprising one or more agents for treating a primary condition associated with cachexia as described above. In a preferred embodiment, the first, second and third dosage forms comprise a therapeutically effective amount of their respective agents. In certain embodiments, the one or more immunosuppressive agents are cytarabine or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the one or more anti-inflammatory agents are ketorolac or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof. In certain embodiments, the one or more agents for treating a primary condition associated with cachexia are chemotherapeutic agents. In certain embodiments, the chemotherapeutic agent is selected from the group consisting of: cyclophosphamide, ifosfamide, mesna, methotrexate, mitomycin, etoposide, irinotecan, mitoxantrone, epirubicin, fludarabine, G-CSF, amsacrine, folinic acid, topotecan, hydroxydaunomycin, vincristine, bleomycin, dacarbazine, procarbazine, thalidomide, fluorouracil, cisplatin, oxaliplatin, carboplatin, thioguanine, capecitabine, gemcitabine, vinorelbine, 6-mercaptopurine, fluorouracil, bendamustine, carmustine, chlorambucil, docetaxel, paclitaxel, bortezomib, and hydroxyurea, and any combination thereof. In certain embodiments, the dosage form further comprises a pharmaceutically acceptable excipient.

The following non-limiting embodiments provide illustrative examples of the invention, but do not limit the scope of the invention:

embodiment 1. a method for treating an immune disorder characterized by (1) increased leukocyte count, (2) increased neutrophil to leukocyte ratio, and (3) decreased T cell to leukocyte ratio, comprising administering to a subject in need thereof an effective amount of an immunosuppressive agent, with the proviso that the immunosuppressive agent is not cytarabine or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, clathrate, or prodrug thereof.

50页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用作皮肤外观修饰剂的经表面反应的碳酸钙

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!